1
|
Tavares DF, Mano JF, Oliveira MB. Advances in abiotic tissue-based biomaterials: A focus on decellularization and devitalization techniques. Mater Today Bio 2025; 32:101735. [PMID: 40275948 PMCID: PMC12020859 DOI: 10.1016/j.mtbio.2025.101735] [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: 01/03/2025] [Revised: 03/14/2025] [Accepted: 04/05/2025] [Indexed: 04/26/2025] Open
Abstract
This Review explores the growing and diversifying field of tissue-derived abiotic constructs for tissue engineering applications, with main focus on decellularization and devitalization techniques and principles. Acellular fractions derived from biological tissues, such as the extracellular matrix (ECM), have long been considered a valuable approach for the generation of numerous scaffolds and more complex constructs. The removal of the cellular content has been considered essential to prevent the development of adverse immunological reactions. Nevertheless, the discovery of promising features of certain cellular components has sparked interest in the use of inactivated or devitalized cellular fractions for several applications, particularly in regenerative medicine and inflammation control. Devitalization has been described for several clinical applications, but remains poorly explored in terms of in vitro constructs compared to decellularization methods currently available. In this review, we present and critically evaluate a spectrum of approaches for the decellularization of whole-organs and in vitro constructs, and the most prevalent devitalization techniques, with a discussion on their implications on scaffolds composition, structure, and potentially therapeutic properties. Processing methodologies to achieve optimal cell-based abiotic materials and approaches for their effective characterization are described and discussed. The application of these materials in healthcare, with most focus on regenerative approaches and including examples of commercially available products, is also addressed.
Collapse
Affiliation(s)
- Diana F. Tavares
- Department of Chemistry, CICECO – Aveiro Institute of Materials. University of Aveiro., Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - João F. Mano
- Department of Chemistry, CICECO – Aveiro Institute of Materials. University of Aveiro., Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Mariana B. Oliveira
- Department of Chemistry, CICECO – Aveiro Institute of Materials. University of Aveiro., Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| |
Collapse
|
2
|
Villicana C, Su N, Yang A, Tong X, Lee HP, Ayushman M, Lee J, Tai M, Kim T, Yang F. Incorporating Bone-Derived ECM into Macroporous Microribbon Scaffolds Accelerates Bone Regeneration. Adv Healthc Mater 2025; 14:e2402138. [PMID: 39891301 DOI: 10.1002/adhm.202402138] [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: 06/10/2024] [Revised: 11/12/2024] [Indexed: 02/03/2025]
Abstract
Tissue-derived extracellular matrix (tdECM) hydrogels serve as effective scaffolds for tissue regeneration by promoting a regenerative immune response. While most tdECM hydrogels are nanoporous and tailored for soft tissue, macroporosity is crucial for bone regeneration. Yet, there's a shortage of macroporous ECM-based hydrogels for this purpose. The study aims to address this gap by developing a co-spinning technique to integrate bone-derived ECM (bECM) into gelatin-based, macroporous microribbon (µRB) scaffolds. The effect of varying doses of bECM on scaffold properties was characterized. In vitro studies revealed 15% bECM as optimal for promoting MSC osteogenesis and macrophage (Mφ) polarization. When implanted in a mouse critical-sized cranial bone defect model, 15% bECM with tricalcium phosphate (TCP) microparticles significantly accelerated bone regeneration and vascularization, filling over 55% of the void by week 2. Increasing bECM to 25% enhanced mesenchymal stem cell (MSC) recruitment and decreased M1 Mφ polarization but reduced overall bone formation and vascularization. The findings demonstrate co-spun gelatin/bECM hydrogels as promising macroporous scaffolds for robust endogenous bone regeneration, without the need for exogenous cells or growth factors. While this study focused on bone regeneration, this platform holds the potential for incorporating various tdECM into macroporous scaffolds for diverse tissue regeneration applications.
Collapse
Affiliation(s)
- Cassandra Villicana
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Ni Su
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Andrew Yang
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Xinming Tong
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Hung Pang Lee
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Manish Ayushman
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Jeehee Lee
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Michelle Tai
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Tayne Kim
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Fan Yang
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| |
Collapse
|
3
|
Jin X, Yoo H, Tran VVT, Yi C, Hong KY, Chang H. Efficacy and Safety of Cell-Assisted Acellular Adipose Matrix Transfer for Volume Retention and Regeneration Compared to Hyaluronic Acid Filler Injection. Aesthetic Plast Surg 2025; 49:1276-1289. [PMID: 39354227 PMCID: PMC11965223 DOI: 10.1007/s00266-024-04408-0] [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: 07/02/2024] [Accepted: 09/11/2024] [Indexed: 10/03/2024]
Abstract
BACKGROUND Cell-assisted acellular adipose matrix (AAM) transfer is a novel technique for soft tissue volume restoration, where AAM acts as a scaffold for tissue proliferation and promotes host cell migration, vascularization, and adipogenesis. This study aimed to evaluate the efficacy and safety of in vivo cell-assisted AAM transfer compared to hyaluronic acid (HA) filler injection. METHODS Human adipose tissue was used to manufacture AAM, and murine adipose-derived stem cells (ASCs) were prepared. Nude mice were divided into four groups: AAM transfer (AT), ASC-assisted AAM transfer (CAT), HA filler injection (HI), and ASC-assisted HA filler injection (CHI). Eight weeks post-transfer, in vivo graft volume/weight, histology, and gene expression were analyzed to assess efficacy and safety. RESULTS The AAM retained its three-dimensional scaffold structure without cellular components. AT/CAT showed lower volume retention than HA/CHA; however, CAT maintained a similar volume to HA. Histologically, adipogenesis and collagen formation were increased in AT/CAT compared to HA/CHA, with CAT showing the highest levels. CAT also demonstrated superior angiogenesis, adipogenesis, and gene expression (Vegf and Pparg), along with lower Il-6 expression, higher Il-10 expression, and reduced capsule formation, indicating better biocompatibility. CONCLUSIONS Cell-assisted AAM transfer is a promising technique for volume retention and tissue regeneration, offering a safe and effective alternative to HA filler injections. LEVEL OF EVIDENCE III This journal requires that authors assign a level of evidence to each article. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .
Collapse
Affiliation(s)
- Xian Jin
- Department of Plastic Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Department of Plastic and Reconstructive Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyokyung Yoo
- Department of Plastic and Reconstructive Surgery, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Vinh Vuong The Tran
- Department of Plastic and Reconstructive Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Chenggang Yi
- Department of Plastic Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Ki Yong Hong
- Department of Plastic and Reconstructive Surgery, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
| | - Hak Chang
- Department of Plastic and Reconstructive Surgery, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
| |
Collapse
|
4
|
Hou M, Tang J, Guo Y, Peng H, Liang B, Cheng Y, Zhang Z, Wei S, Yi C, Li H. T-PRP-DAT Gel: A Novel Material Promotes Adipose Tissue Regeneration. J Cosmet Dermatol 2025; 24:e70045. [PMID: 39963070 PMCID: PMC11833427 DOI: 10.1111/jocd.70045] [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: 11/12/2024] [Revised: 01/05/2025] [Accepted: 01/31/2025] [Indexed: 02/20/2025]
Abstract
BACKGROUND Decellularized adipose tissue (DAT) has emerged as a promising tissue-specific regenerative platform for soft tissue augmentation and reconstruction. Hydrogels are a widely used DAT scaffold format for their injectability and porous structure. While unstable structure and poor vascularization limit the adipose tissue regeneration of DAT gels, this yields significant clinical necessity for solutions to this problem. METHODS Based on collagen-fibrin interpenetrating, we developed an injectable thermosensitive DAT/PRP interpenetrating polymer network (t-DP gel/t-DPI) with the composition of DAT and temperature-controlled platelet-rich plasma (t-PRP). The same volume of t-DP gel and DAT gel were transplanted in a mouse model, and graft volume, weight, adipose tissue regeneration rate, and vascularization were compared. RESULTS The t-DPI showed reinforced stability for the interpenetrating polymer network (IPN) of collagen-fibrin and sustained release of growth factors from the t-PRP, resulting in improved graft volume, weight, adipose tissue regeneration, and vascularization. CONCLUSIONS Compared with traditional DAT gel, t-DP gel promotes adipose tissue regeneration by promoting angiogenesis and stability, and t-DP gel has great potential for future applications in the field of plastic surgery for its simple preparation and regeneration ability.
Collapse
Affiliation(s)
- Mengmeng Hou
- Department of Plastic and Reconstructive SurgeryXijing Hospital, Fourth Military Medical UniversityXi'anChina
| | - Jiezhang Tang
- Department of Plastic and Reconstructive SurgeryXijing Hospital, Fourth Military Medical UniversityXi'anChina
| | - Yajie Guo
- Department of Plastic and Reconstructive SurgeryXijing Hospital, Fourth Military Medical UniversityXi'anChina
| | - Han Peng
- Department of Plastic and Reconstructive SurgeryXijing Hospital, Fourth Military Medical UniversityXi'anChina
| | - Baoyan Liang
- Department of Plastic and Reconstructive SurgeryXijing Hospital, Fourth Military Medical UniversityXi'anChina
| | - Yi Cheng
- Department of Plastic and Reconstructive SurgeryXijing Hospital, Fourth Military Medical UniversityXi'anChina
| | - Zhaoxiang Zhang
- Department of Plastic and Reconstructive SurgeryXijing Hospital, Fourth Military Medical UniversityXi'anChina
| | - Siming Wei
- Department of Plastic and Reconstructive SurgeryXijing Hospital, Fourth Military Medical UniversityXi'anChina
| | - Chenggang Yi
- The Second Affiliated Hospital of Zhejiang University College of MedicineHangzhouChina
| | - Huichen Li
- Department of Plastic and Reconstructive SurgeryXijing Hospital, Fourth Military Medical UniversityXi'anChina
| |
Collapse
|
5
|
Rahman E, Carruthers JDA, Rao P, Abu-Farsakh HN, Garcia PE, Ioannidis S, Sayed K, Philipp-Dormston WG, Mosahebi A, Webb WR. Regenerative Aesthetics: A Genuine Frontier or Just a Facet of Regenerative Medicine: A Systematic Review. Aesthetic Plast Surg 2025; 49:341-355. [PMID: 39198280 DOI: 10.1007/s00266-024-04287-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 07/24/2024] [Indexed: 09/01/2024]
Abstract
BACKGROUND Regenerative aesthetics claims to enhance cosmetic outcomes through advanced biological interventions like Stem cell and Exosome therapy, Polydeoxyribonucleotide (PDRN), Photobiomodulation, bioactive peptides and treatment for cellular senescence yet lacks substantial scientific and regulatory validation. OBJECTIVE To evaluate the scientific and clinical foundations of regenerative medicine techniques in non-surgical aesthetics and assess the legitimacy of regenerative aesthetics as a medical specialty. METHODS A systematic review was conducted according to PRISMA guidelines, searching databases including PubMed, Scopus, and Web of Science for studies published in the last ten years. We included 19 studies, comprising 14 randomized controlled trials (RCTs) and 5 prospective studies, focusing on interventions that purportedly use regenerative medicine principles in aesthetic applications. RESULTS The review highlights a prevalent gap in molecular and clinical evidence supporting the efficacy and safety of regenerative aesthetics. Despite the robust design of the included RCTs and prospective studies, there remains a significant lack of consistent, high-quality evidence proving the effectiveness of these interventions. Issues such as inadequate reporting, unclear molecular mechanisms, and absence of long-term safety data were common. CONCLUSION The field of regenerative aesthetics lacks the necessary scientific rigour and regulatory compliance to be recognized as a legitimate medical specialty. This review underscores the need for stringent scientific validation and regulatory oversight to ensure patient safety and treatment efficacy before these techniques can be recommended for clinical use. LEVEL OF EVIDENCE II This journal requires that authors assign a level of evidence to each article. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .
Collapse
Affiliation(s)
- Eqram Rahman
- Research and Innovation Hub, Innovation Aesthetics, London, WC2H 9JQ, UK
| | - Jean D A Carruthers
- Department of Ophthalmology, University of British Columbia, Vancouver, BC, Canada
| | - Parinitha Rao
- The Skin Address, Aesthetic Dermatology Practice, Bangalore, India
| | | | | | | | - Karim Sayed
- Nomi Oslo, Oslo, Norway
- University of South-Eastern Norway, Drammen, Norway
| | | | | | | |
Collapse
|
6
|
Khudyakov JI. Protein Sample Preparation for Bottom-Up, Label-Free Quantitative Proteomics of Adipose Tissue. Methods Mol Biol 2025; 2884:43-56. [PMID: 39715996 DOI: 10.1007/978-1-0716-4298-6_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2024]
Abstract
Adipose tissue (AT) is a complex, multifunctional endocrine organ that plays a significant role in animal evolution and human disease. Profiling of the proteome, or the set of proteins produced by a cell or tissue at a given time, can be used to explore the myriad functions of adipose tissue and understand its role in health and disease. The main challenges of adipose tissue proteomics include the high lipid and low protein content of the tissue and association of many proteins with lipid droplets. Here, we present a protocol for gel-free, label-free, bottom-up, relative quantitative proteomics of adipose tissue based on findings from the literature and our laboratory that yields reproducible protein and peptide identification rates while minimizing cost and processing time. This approach involves tissue homogenization, protein precipitation from homogenates, solubilization and denaturation of proteins in a buffer containing 5% sodium deoxycholate (SDC, an acid-insoluble detergent) and 5 mM tris(2-carboxyethyl)phosphine (TCEP, a reducing agent), alkylation with chloroacetamide, and in-solution tandem digestion with trypsin and Lys-C enzymes in the presence of 1% SDC. Acidification of peptides efficiently removes SDC prior to desalting and mass spectrometry. This method has been used successfully in our laboratory by both experienced researchers and those with limited technical backgrounds, including high school, undergraduate, and graduate students. We have identified >1500 proteins in adipose tissue of non-model mammals (e.g., blubber of marine mammals) spanning a dynamic range of 105 using this approach, including proteins of interest for comparative physiology such as adipokines, metabolic and antioxidant enzymes, lipid droplet proteins, metabolite transporters, and mitochondrial proteins, among others.
Collapse
Affiliation(s)
- Jane I Khudyakov
- Department of Biological Sciences, University of the Pacific, Stockton, CA, USA.
| |
Collapse
|
7
|
Schuster CR, Reiche E, Keller PR, Hu S, Soares V, Rahmayanti S, Suresh V, Harris TGW, Doloff JC, Tuffaha S, Coon D. Testosterone Promotes Nerve Tethering and Acellular Biomaterial Perineural Fibrosis in a Rat Wound Repair Model. Adv Wound Care (New Rochelle) 2024; 13:615-624. [PMID: 38775428 DOI: 10.1089/wound.2024.0043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2024] Open
Abstract
Objective: Nerve scarring after traumatic or iatrogenic exposure can lead to impaired function and pain. Nerve-adjacent biomaterials promoting a regenerative tissue response may help reduce perineural fibrosis. Our prior work suggests that testosterone may promote fibrotic skin scarring, but it is unknown how testosterone alters nerve fibrosis or shifts the response to biomaterials. Approach: Sterilized Lewis rats received either testosterone cypionate (+T) or placebo (-T) biweekly. Fifteen days later, wounds were created over the sciatic nerve and covered with an acellular matrix (AM) or closed via primary closure (PC). At day 42, force gauge testing measured the force required to mobilize the nerve, and wound tissue was analyzed. Results: Nerve mobilization force was greater in +T versus -T wounds (p < 0.01). Nerves tore before gliding in 60% of +T versus 6% of -T rats. Epidermal gap (p < 0.01), scar width (p < 0.01), and cross-sectional scar tissue area (p = 0.02) were greater in +T versus -T rats. +T versus -T rats expressed less Col-3 (p = 0.02) and CD68 (p = 0.02). Nerve mobilization force trended nonsignificantly higher for PC versus AM wounds and for +T versus -T wounds within the AM cohort. Innovation: Testosterone increases nerve tethering in the wound healing milieu, altering repair and immune cell balances. Conclusion: Testosterone significantly increases the force required to mobilize nerves in wound beds and elevates histological markers of scarring, suggesting that testosterone-induced inflammation may increase perineural adhesion. Testosterone may reduce the potential anti-tethering protective effect of AM. Androgen receptor antagonism may represent a therapeutic target to reduce scar-related nerve morbidity.
Collapse
Affiliation(s)
- Calvin R Schuster
- Departments of Plastic and Reconstructive Surgery and Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Erik Reiche
- Division of Plastic Surgery, Brigham and Women's Hospital Harvard Medical School, Boston, Massachusetts, USA
| | - Patrick R Keller
- Departments of Plastic and Reconstructive Surgery and Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sophia Hu
- Division of Plastic Surgery, Brigham and Women's Hospital Harvard Medical School, Boston, Massachusetts, USA
| | - Vance Soares
- Departments of Plastic and Reconstructive Surgery and Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Siti Rahmayanti
- Division of Plastic Surgery, Brigham and Women's Hospital Harvard Medical School, Boston, Massachusetts, USA
| | - Visakha Suresh
- Departments of Plastic and Reconstructive Surgery and Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Thomas G W Harris
- Departments of Plastic and Reconstructive Surgery and Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Joshua C Doloff
- Departments of Plastic and Reconstructive Surgery and Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sami Tuffaha
- Departments of Plastic and Reconstructive Surgery and Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Devin Coon
- Division of Plastic Surgery, Brigham and Women's Hospital Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
8
|
Song JH, Kim SJ, Kwon S, Jeon SY, Park SE, Choi SJ, Oh SY, Jeon HB, Chang JW. Nervonic acid improves fat transplantation by promoting adipogenesis and angiogenesis. Int J Mol Med 2024; 54:108. [PMID: 39364738 PMCID: PMC11517738 DOI: 10.3892/ijmm.2024.5432] [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: 06/12/2024] [Accepted: 08/19/2024] [Indexed: 10/05/2024] Open
Abstract
Adipose tissue engraftment has become a promising strategy in the field of regenerative surgery; however, there are notable challenges associated with it, such as resorption of 50‑90% of the transplanted fat or cyst formation due to fat necrosis after fat transplantation. Therefore, identifying novel materials or methods to improve the engraftment efficiency is crucial. The present study investigated the effects of nervonic acid (NA), a monounsaturated very long‑chain fatty acid, on adipogenesis and fat transplantation, as well as its underlying mechanisms. To assess this, NA was used to treat cells during adipogenesis in vitro, and the expression levels of markers, including PPARγ and CEBPα, and signaling molecules were detected through reverse transcription‑quantitative PCR and western blotting. In addition, NA was mixed with fat grafts in in vivo fat transplantation, followed by analysis through Oil Red O staining, hematoxylin & eosin staining and immunohistochemistry. It was demonstrated that NA treatment accelerated adipogenesis through activation of the Akt/mTOR pathway and inhibition of Wnt signaling. NA treatment enriched the expression of Akt/mTOR signaling‑related genes, and increased the expression of genes involved in angiogenesis and fat differentiation in human mesenchymal stem cells (MSCs). Additionally, NA effectively improved the outcome of adipose tissue engraftment in mice. Treatment of grafts with NA at transplantation reduced the resorption of transplanted fat and increased the proportion of perilipin‑1+ adipocytes with a lower portion of vacuoles in mice. Moreover, the NA‑treated group exhibited a reduced pro‑inflammatory response and had more CD31+ vessel structures, which were relatively evenly distributed among viable adipocytes, facilitating successful engraftment. In conclusion, the present study demonstrated that NA may not only stimulate adipogenesis by regulating signaling pathways in human MSCs, but could improve the outcome of fat transplantation by reducing inflammation and stimulating angiogenesis. It was thus hypothesized that NA could serve as an adjuvant strategy to enhance fat engraftment in regenerative surgery.
Collapse
Affiliation(s)
- Jae Hoon Song
- Cell and Gene Therapy Institute, ENCell Co., Ltd., Seoul 06072, Republic of Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Sun Jeong Kim
- Cell and Gene Therapy Institute, ENCell Co., Ltd., Seoul 06072, Republic of Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Soojin Kwon
- Cell and Gene Therapy Institute, ENCell Co., Ltd., Seoul 06072, Republic of Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Su Yeon Jeon
- Cell and Gene Therapy Institute, ENCell Co., Ltd., Seoul 06072, Republic of Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Sang Eon Park
- Cell and Gene Therapy Institute, ENCell Co., Ltd., Seoul 06072, Republic of Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Suk-Joo Choi
- Department of Obstetrics and Gynecology, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Soo-Young Oh
- Department of Obstetrics and Gynecology, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Hong Bae Jeon
- Cell and Gene Therapy Institute, ENCell Co., Ltd., Seoul 06072, Republic of Korea
| | - Jong Wook Chang
- Cell and Gene Therapy Institute, ENCell Co., Ltd., Seoul 06072, Republic of Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul 06351, Republic of Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul 06355, Republic of Korea
| |
Collapse
|
9
|
Kim J, Tran VVT, Hong KY, Chang H. Comparison of Stored and Fresh Injectable Acellular Adipose Matrix in Soft Tissue Reconstruction in a Murine Model. Aesthetic Plast Surg 2024; 48:4546-4553. [PMID: 38913200 PMCID: PMC11588782 DOI: 10.1007/s00266-024-04175-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 05/30/2024] [Indexed: 06/25/2024]
Abstract
BACKGROUND We previously showed comparable volume effects of injections of acellular adipose matrix (AAM), an adipose tissue-derived extracellular matrix, and conventional fat grafting in a murine model. Thus, AAM could be a novel allogenic injectable product. However, its retention rate poses a concern, as repeated AAM injections may be required in some cases. This study investigated the biological properties and therapeutic value of stored AAM and compared them with those of fresh AAM, in a murine model. METHODS AAM was manufactured from fresh human abdominoplasty fat. Fresh and stored injectable AAM was prepared within 24 h and 3 months after generation, respectively. Either fresh or stored injectable AAM was injected into the scalp of athymic nude mice (0.2 mL/sample, n = 6 per group). After 8 weeks, graft retention was assessed through weight measurement, and histological analysis was performed, including immunofluorescence staining for CD31 and perilipin. RESULTS Retention rate was significantly reduced in the stored compared to the fresh injectable AAM group. Nevertheless, histological analysis revealed comparable inflammatory cell presence, with minimal capsule formation, in both groups. Adipogenesis occurred in both groups, with no significant difference in the blood vessel area (%) between groups. CONCLUSIONS Although the volume effects of stored AAM for soft tissue reconstruction were limited compared to those of fresh injectable AAM, stored AAM had similar capacity for adipogenesis and angiogenesis. This promising allogeneic injectable holds the potential to serve as an effective "off-the-shelf" alternative for repeated use within a 3-month storage period. NO LEVEL ASSIGNED This journal requires that authors assign a level of evidence to each submission to which Evidence-Based Medicine rankings are applicable. This excludes Review Articles, Book Reviews, and manuscripts that concern Basic Science, Animal Studies, Cadaver Studies, and Experimental Studies. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors https://link.springer.com/journal/00266 .
Collapse
Affiliation(s)
- Jaewoo Kim
- Department of Plastic and Reconstructive Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Plastic and Reconstructive Surgery, SMG-SNU Boramae Medical Center, Seoul, Republic of Korea
| | - Vinh Vuong The Tran
- Department of Plastic and Reconstructive Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
- Hi-Tech Center, Vinmec Healthcare System, Hanoi, Vietnam
| | - Ki Yong Hong
- Department of Plastic and Reconstructive Surgery, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
| | - Hak Chang
- Department of Plastic and Reconstructive Surgery, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
| |
Collapse
|
10
|
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
|
11
|
Kim J, Tran VVT, Hong KY, Chang H. Effect of Injectable Acellular Adipose Matrix on Soft Tissue Reconstruction in a Murine Model. Aesthetic Plast Surg 2024; 48:2210-2219. [PMID: 38499876 PMCID: PMC11150185 DOI: 10.1007/s00266-024-03924-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 02/09/2024] [Indexed: 03/20/2024]
Abstract
BACKGROUND The extracellular matrix isolated from adipose tissue, known as acellular adipose matrix (AAM), represents a novel biomaterial. AAM functions as a scaffold that not only supports stem cell proliferation and differentiation but also induces adipogenesis and angiogenesis. This study aims to investigate the volumetric effects and microenvironmental changes associated with injectable AAM in comparison to conventional fat grafting. METHODS AAM was manufactured from fresh human abdominoplasty fat using a mechanically modified method and then transformed into an injectable form. Lipoaspirate was harvested employing the Coleman technique. A weight and volume study was conducted on athymic nude mice by injecting either injectable AAM or lipoaspirate into the scalp (n=6 per group). After eight weeks, graft retention was assessed through weight measurement and volumetric analysis using micro-computed tomography (micro-CT) scanning. Histological analysis was performed using immunofluorescence staining for perilipin and CD31. RESULTS Injectable AAM exhibited similar weight and volume effects in murine models. Histological analysis revealed comparable inflammatory cell presence with minimal capsule formation when compared to conventional fat grafts. Adipogenesis occurred in both AAM-injected and conventional fat graft models, with no significant difference in the blood vessel area (%) between the two. CONCLUSIONS In summary, injectable AAM demonstrates effectiveness comparable to conventional fat grafting concerning volume effects and tissue regeneration in soft tissue reconstruction. This promising allogeneic injectable holds the potential to serve as a safe and effective "Off-the-Shelf" alternative in both aesthetic and reconstructive clinical practices. NO LEVEL ASSIGNED This journal requires that authors assign a level of evidence to each submission to which Evidence-Based Medicine rankings are applicable. This excludes Review Articles, Book Reviews, and manuscripts that concern Basic Science, Animal Studies, Cadaver Studies, and Experimental Studies. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .
Collapse
Affiliation(s)
- Jaewoo Kim
- Department of Plastic and Reconstructive Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Plastic and Reconstructive Surgery, SMG-SNU Boramae Medical Center, Seoul, Republic of Korea
| | - Vinh Vuong The Tran
- Department of Plastic and Reconstructive Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
- Hi-Tech Center, Vinmec Healthcare System, Hanoi, Vietnam
| | - Ki Yong Hong
- Department of Plastic and Reconstructive Surgery, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, 03080, Seoul, Republic of Korea.
| | - Hak Chang
- Department of Plastic and Reconstructive Surgery, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, 03080, Seoul, Republic of Korea.
| |
Collapse
|
12
|
Yang X, Jin L, Xu M, Liu X, Tan Z, Liu L. Adipose tissue reconstruction facilitated with low immunogenicity decellularized adipose tissue scaffolds. Biomed Mater 2024; 19:035023. [PMID: 38518362 DOI: 10.1088/1748-605x/ad3705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 03/22/2024] [Indexed: 03/24/2024]
Abstract
There is currently an urgent need to develop engineered scaffolds to support new adipose tissue formation and facilitate long-term maintenance of function and defect repair to further generate prospective bioactive filler materials capable of fulfilling surgical needs. Herein, adipose regeneration methods were optimized and decellularized adipose tissue (DAT) scaffolds with good biocompatibility were fabricated. Adipose-like tissues were reconstructed using the DAT and 3T3-L1 preadipocytes, which have certain differentiation potential, and the regenerative effects of the engineered adipose tissuesin vitroandin vivowere explored. The method improved the efficiency of adipose removal from tissues, and significantly shortened the time for degreasing. Thus, the DAT not only provided a suitable space for cell growth but also promoted the proliferation, migration, and differentiation of preadipocytes within it. Following implantation of the constructed adipose tissuesin vivo, the DAT showed gradual degradation and integration with surrounding tissues, accompanied by the generation of new adipose tissue analogs. Overall, the combination of adipose-derived extracellular matrix and preadipocytes for adipose tissue reconstruction will be of benefit in the artificial construction of biomimetic implant structures for adipose tissue reconstruction, providing a practical guideline for the initial integration of adipose tissue engineering into clinical medicine.
Collapse
Affiliation(s)
- Xun Yang
- Department of Traumatic Orthopedics, Shenzhen Second People's Hospital, The First Affiliated Hospital, Shenzhen University, Shenzhen 518028, People's Republic of China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, People's Republic of China
| | - Lijuan Jin
- Institute of Shenzhen, Hunan University, Shenzhen 518000, People's Republic of China
- Greater Bay Area Institute for Innovation, Hunan University, Guangzhou 511300, People's Republic of China
| | - Miaomiao Xu
- College of Biology, Hunan University, Changsha 410082, People's Republic of China
| | - Xiao Liu
- Greater Bay Area Institute for Innovation, Hunan University, Guangzhou 511300, People's Republic of China
| | - Zhikai Tan
- Institute of Shenzhen, Hunan University, Shenzhen 518000, People's Republic of China
- Greater Bay Area Institute for Innovation, Hunan University, Guangzhou 511300, People's Republic of China
- College of Biology, Hunan University, Changsha 410082, People's Republic of China
| | - Lijun Liu
- Department of Traumatic Orthopedics, Shenzhen Second People's Hospital, The First Affiliated Hospital, Shenzhen University, Shenzhen 518028, People's Republic of China
| |
Collapse
|
13
|
Xie C, Xu J, Wang X, Jiang S, Zheng Y, Liu Z, Jia Z, Jia Z, Lu X. Smart Hydrogels for Tissue Regeneration. Macromol Biosci 2024; 24:e2300339. [PMID: 37848181 DOI: 10.1002/mabi.202300339] [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: 07/25/2023] [Revised: 10/06/2023] [Indexed: 10/19/2023]
Abstract
The rapid growth in the portion of the aging population has led to a consequent increase in demand for biomedical hydrogels, together with an assortment of challenges that need to be overcome in this field. Smart hydrogels can autonomously sense and respond to the physiological/pathological changes of the tissue microenvironment and continuously adapt the response according to the dynamic spatiotemporal shifts in conditions. This along with other favorable properties, make smart hydrogels excellent materials for employing toward improving the precision of treatment for age-related diseases. The key factor during the smart hydrogel design is on accurately identifying the characteristics of natural tissues and faithfully replicating the composition, structure, and biological functions of these tissues at the molecular level. Such hydrogels can accurately sense distinct physiological and external factors such as temperature and biologically active molecules, so they may in turn actively and promptly adjust their response, by regulating their own biological effects, thereby promoting damaged tissue repair. This review summarizes the design strategies employed in the creation of smart hydrogels, their response mechanisms, as well as their applications in field of tissue engineering; and concludes by briefly discussing the relevant challenges and future prospects.
Collapse
Affiliation(s)
- Chaoming Xie
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Jie Xu
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Xinyi Wang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Shengxi Jiang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Yujia Zheng
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Zexin Liu
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Zhuo Jia
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Zhanrong Jia
- The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong, 523000, China
| | - Xiong Lu
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| |
Collapse
|
14
|
Bhar B, Ranta P, Samudrala PK, Mandal BB. Omentum Extracellular Matrix-Silk Fibroin Hydroscaffold Promotes Wound Healing through Vascularization and Tissue Remodeling in the Diabetic Rat Model. ACS Biomater Sci Eng 2024; 10:1090-1105. [PMID: 38275123 DOI: 10.1021/acsbiomaterials.3c01877] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2024]
Abstract
Nonhealing diabetic wounds are often associated with significant mortality and cause economic and clinical burdens to the healthcare system. Herein, a biomimetic hydroscaffold is developed using omentum tissue-derived decellularized-extracellular matrix (dECM) and silk fibroin (SF) proteins that associate the behavior of a collagenous fibrous scaffold and a hydrogel to reproduce all aspects of the provisional skin tissue matrix. The chemical cross-linker-free in situ gelation property of the two types of SF proteins from Bombyx mori and Antheraea assamensis ensures the adherence of dECM with surrounding tissue on the wound bed, circumventing further suturing. The physicochemical and mechanical properties of the composite hydroscaffold (SF-dECM) were thoroughly evaluated. The hydroscaffolds were found to support the growth and proliferation of human dermal fibroblasts and influence the angiogenic potential of endothelial cells under in vitro conditions. Furthermore, the healing efficacy of the composites was evaluated by generating full-thickness wounds on a streptozotocin-induced diabetic rat model. The presence of dECM components in the composite facilitated the rate of wound closure, granulation tissue formation, and re-epithelialization by providing intrinsic cues to advance the inflammatory stage and stimulating angiogenesis. Collectively, as an off-the-shelf wound dressing requiring only a single topical administration, the SF-dECM hydroscaffold is a promising, cost-effective dressing for the management of chronic diabetic wounds.
Collapse
Affiliation(s)
- Bibrita Bhar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Priyanka Ranta
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical and Educational Research Guwahati, Guwahati, Assam 781101, India
| | - Pavan Kumar Samudrala
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical and Educational Research Guwahati, Guwahati, Assam 781101, India
| | - Biman B Mandal
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| |
Collapse
|
15
|
Zhou Z, Pang Y, Ji J, He J, Liu T, Ouyang L, Zhang W, Zhang XL, Zhang ZG, Zhang K, Sun W. Harnessing 3D in vitro systems to model immune responses to solid tumours: a step towards improving and creating personalized immunotherapies. Nat Rev Immunol 2024; 24:18-32. [PMID: 37402992 DOI: 10.1038/s41577-023-00896-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2023] [Indexed: 07/06/2023]
Abstract
In vitro 3D models are advanced biological tools that have been established to overcome the shortcomings of oversimplified 2D cultures and mouse models. Various in vitro 3D immuno-oncology models have been developed to mimic and recapitulate the cancer-immunity cycle, evaluate immunotherapy regimens, and explore options for optimizing current immunotherapies, including for individual patient tumours. Here, we review recent developments in this field. We focus, first, on the limitations of existing immunotherapies for solid tumours, secondly, on how in vitro 3D immuno-oncology models are established using various technologies - including scaffolds, organoids, microfluidics and 3D bioprinting - and thirdly, on the applications of these 3D models for comprehending the cancer-immunity cycle as well as for assessing and improving immunotherapies for solid tumours.
Collapse
Affiliation(s)
- Zhenzhen Zhou
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Haidian District, Beijing, China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, China
- Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Beijing, China
| | - Yuan Pang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Haidian District, Beijing, China.
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, China.
- Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Beijing, China.
| | - Jingyuan Ji
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Haidian District, Beijing, China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, China
- Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Beijing, China
| | - Jianyu He
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Haidian District, Beijing, China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, China
- Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Beijing, China
| | - Tiankun Liu
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Haidian District, Beijing, China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, China
- Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Beijing, China
| | - Liliang Ouyang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Haidian District, Beijing, China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, China
- Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Beijing, China
| | - Wen Zhang
- Department of Immunology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Chaoyang District, Beijing, China
| | - Xue-Li Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhi-Gang Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kaitai Zhang
- State Key Laboratory of Molecular Oncology, Department of Aetiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Chaoyang District, Beijing, China
| | - Wei Sun
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Haidian District, Beijing, China.
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, China.
- Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Beijing, China.
- Department of Mechanical Engineering, Drexel University, Philadelphia, PA, USA.
| |
Collapse
|
16
|
Yang B, Rutkowski N, Elisseeff J. The foreign body response: emerging cell types and considerations for targeted therapeutics. Biomater Sci 2023; 11:7730-7747. [PMID: 37904536 DOI: 10.1039/d3bm00629h] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2023]
Abstract
The foreign body response (FBR) remains a clinical challenge in the field of biomaterials due to its ability to elicit a chronic and sustained immune response. Modulating the immune response to materials is a modern paradigm in tissue engineering to enhance repair while limiting fibrous encapsulation and implant isolation. Though the classical mediators of the FBR are well-characterized, recent studies highlight that our understanding of the cell types that shape the FBR may be incomplete. In this review, we discuss the emerging role of T cells, stromal-immune cell interactions, and senescent cells in the biomaterial response, particularly to synthetic materials. We emphasize future studies that will deepen the field's understanding of these cell types in the FBR, with the goal of identifying therapeutic targets that will improve implant integration. Finally, we briefly review several considerations that may influence our understanding of the FBR in humans, including rodent models, aging, gut microbiota, and sex differences. A better understanding of the heterogeneous host cell response during the FBR can enable the design and development of immunomodulatory materials that favor healing.
Collapse
Affiliation(s)
- Brenda Yang
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.
| | - Natalie Rutkowski
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.
| | - Jennifer Elisseeff
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.
- Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
17
|
Hu Y, Lu H, Yuan X, Yang Z, Gao Q, Qi Z. The histologic reaction and permanence of hyaluronic acid gel, calcium hydroxylapatite microspheres, and extracellular matrix bio gel. J Cosmet Dermatol 2023; 22:2685-2691. [PMID: 37082836 DOI: 10.1111/jocd.15767] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/24/2023] [Accepted: 03/28/2023] [Indexed: 04/22/2023]
Abstract
BACKGROUND The filling materials on the beauty market can be classified into three types: natural biological materials, synthetic polymer materials, and composites containing bioactive substances. However, comparative experimental data is lacking to compare their biological responses and permanence. AIMS The main object of this study was to evaluate the biological response of these three types of fillers to provide a theoretical basis for clinical application. METHODS Six-week-old female mice were injected subcutaneously with hyaluronic acid (HA) gel, calcium hydroxylapatite (CaHA) microspheres, and extracellular matrix (ECM) bio gel to observe the body reaction and permanence. At 1, 4, 8, and 16 weeks, the test sites were excised and analyzed by histopathology and proteomics. RESULTS Extracellular matrix had a minimal foreign body response. HA had a good volume effect at the early stage but the volume retention rate was lower than CaHA in the long term. CaHA could stimulate neo-collagen formation. CONCLUSION This study has proven the effectiveness and safety of these fillers and could provide clinical guidance for the plastic surgeon.
Collapse
Affiliation(s)
- Yuling Hu
- The 16th Department of Plastic Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.33 Badachu Road, Shijingshan District, Beijing, 100144, China
| | - Haibin Lu
- The 16th Department of Plastic Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.33 Badachu Road, Shijingshan District, Beijing, 100144, China
| | - Xihang Yuan
- The 16th Department of Plastic Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.33 Badachu Road, Shijingshan District, Beijing, 100144, China
| | - Zhenyu Yang
- The 16th Department of Plastic Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.33 Badachu Road, Shijingshan District, Beijing, 100144, China
| | - Qiuni Gao
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Zuoliang Qi
- The 16th Department of Plastic Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.33 Badachu Road, Shijingshan District, Beijing, 100144, China
| |
Collapse
|
18
|
Major G, Longoni A, Simcock J, Magon NJ, Harte J, Bathish B, Kemp R, Woodfield T, Lim KS. Clinical Applicability of Visible Light-Mediated Cross-linking for Structural Soft Tissue Reconstruction. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300538. [PMID: 37424046 PMCID: PMC10502829 DOI: 10.1002/advs.202300538] [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: 01/25/2023] [Revised: 05/28/2023] [Indexed: 07/11/2023]
Abstract
Visible light-mediated cross-linking has utility for enhancing the structural capacity and shape fidelity of laboratory-based polymers. With increased light penetration and cross-linking speed, there is opportunity to extend future applications into clinical spheres. This study evaluated the utility of a ruthenium/sodium persulfate photocross-linking system for increasing structural control in heterogeneous living tissues as an example, focusing on unmodified patient-derived lipoaspirate for soft tissue reconstruction. Freshly-isolated tissue is photocross-linked, then the molar abundance of dityrosine bonds is measured using liquid chromatography tandem mass spectrometry and the resulting structural integrity assessed. The cell function and tissue survival of photocross-linked grafts is evaluated ex vivo and in vivo, with tissue integration and vascularization assessed using histology and microcomputed tomography. The photocross-linking strategy is tailorable, allowing progressive increases in the structural fidelity of lipoaspirate, as measured by a stepwise reduction in fiber diameter, increased graft porosity and reduced variation in graft resorption. There is an increase in dityrosine bond formation with increasing photoinitiator concentration, and tissue homeostasis is achieved ex vivo, with vascular cell infiltration and vessel formation in vivo. These data demonstrate the capability and applicability of photocrosslinking strategies for improving structural control in clinically-relevant settings, potentially achieving more desirable patient outcomes using minimal manipulation in surgical procedures.
Collapse
Affiliation(s)
- Gretel Major
- Department of Orthopaedic Surgery and Musculoskeletal MedicineCentre for Bioengineering & NanomedicineUniversity of OtagoChristchurch8011New Zealand
| | - Alessia Longoni
- Department of Orthopaedic Surgery and Musculoskeletal MedicineCentre for Bioengineering & NanomedicineUniversity of OtagoChristchurch8011New Zealand
| | - Jeremy Simcock
- Department of SurgeryUniversity of OtagoChristchurch8011New Zealand
| | - Nicholas J Magon
- Centre for Free Radical ResearchDepartment of Pathology and Biomedical ScienceUniversity of OtagoChristchurch8011New Zealand
| | - Jessica Harte
- Jacqui Wood Cancer CentreDivision of Cellular MedicineNinewells Hospital and Medical SchoolUniversity of DundeeDundeeScotlandDD2 1GZUK
| | - Boushra Bathish
- Department of Orthopaedic Surgery and Musculoskeletal MedicineCentre for Bioengineering & NanomedicineUniversity of OtagoChristchurch8011New Zealand
- Jacqui Wood Cancer CentreDivision of Cellular MedicineNinewells Hospital and Medical SchoolUniversity of DundeeDundeeScotlandDD2 1GZUK
| | - Roslyn Kemp
- Department of Microbiology and ImmunologyUniversity of OtagoDunedin9016New Zealand
| | - Tim Woodfield
- Department of Orthopaedic Surgery and Musculoskeletal MedicineCentre for Bioengineering & NanomedicineUniversity of OtagoChristchurch8011New Zealand
| | - Khoon S Lim
- Department of Orthopaedic Surgery and Musculoskeletal MedicineCentre for Bioengineering & NanomedicineUniversity of OtagoChristchurch8011New Zealand
- Light‐Activated Biomaterials GroupSchool of Medical SciencesUniversity of SydneySydney2006Australia
| |
Collapse
|
19
|
Ziegler ME, Khabaz K, Khoshab N, Halaseh FF, Chnari E, Chen S, Baldi P, Evans GRD, Widgerow AD. Combining Allograft Adipose and Fascia Matrix as an Off-the-Shelf Scaffold for Adipose Tissue Engineering Stimulates Angiogenic Responses and Activates a Proregenerative Macrophage Profile in a Rodent Model. Ann Plast Surg 2023; 91:294-300. [PMID: 37489973 DOI: 10.1097/sap.0000000000003587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/26/2023]
Abstract
OBJECTIVE Bioscaffolds for treating soft tissue defects have limitations. As a bioscaffold, allograft adipose matrix (AAM) is a promising approach to treat soft tissue defects. Previously, we revealed that combining superficial adipose fascia matrix with AAM, components of the hypodermis layer of adipose tissue, improved volume retention, adipogenesis, and angiogenesis in rats 8 weeks after it was implanted compared with AAM alone. Here, we modified the fascia matrix and AAM preparation, examined the tissue over 18 weeks, and conducted a deeper molecular investigation. We hypothesized that the combined matrices created a better scaffold by triggering angiogenesis and proregenerative signals. METHODS Human AAM and fascia matrix were implanted (4 [1 mL] implants/animal) into the dorsum of male Fischer rats (6-8 weeks old; ~140 g) randomly as follows: AAM, fascia, 75/25 (AAM/fascia), 50/50, and 50/50 + hyaluronic acid (HA; to improve extrudability) (n = 4/group/time point). After 72 hours, as well as 1, 3, 6, 9, 12, and 18 weeks, graft retention was assessed by a gas pycnometer. Adipogenesis (HE), angiogenesis (CD31), and macrophage infiltration (CD80 and CD163) were evaluated histologically at all time points. The adipose area and M1/M2 macrophage ratio were determined using ImageJ. RNA sequencing (RNA-seq) and bioinformatics were conducted to evaluate pathway enrichments. RESULTS By 18 weeks, the adipose area was 2365% greater for 50/50 HA (281.6 ± 21.6) than AAM (11.4 ± 0.9) (P < 0.001). The M1/M2 macrophage ratio was significantly lower for 50/50 HA (0.8 ± 0.1) than AAM (0.9 ± 0.1) at 6 weeks (16%; P < 0.05). This inversely correlated with adipose area (r = -0.6; P > 0.05). The RNA-seq data revealed that upregulated adipogenesis, angiogenesis, and macrophage-induced tissue regeneration genes were temporally different between the groups. CONCLUSIONS Combining the fascia matrix with AAM creates a bioscaffold with an improved retention volume that supports M2 macrophage-mediated angiogenesis and adipogenesis. This bioscaffold is worthy of further investigation.
Collapse
Affiliation(s)
- Mary E Ziegler
- From the Center for Tissue Engineering, UC Irvine Department of Plastic Surgery, Orange, CA
| | - Kameel Khabaz
- From the Center for Tissue Engineering, UC Irvine Department of Plastic Surgery, Orange, CA
| | - Nima Khoshab
- From the Center for Tissue Engineering, UC Irvine Department of Plastic Surgery, Orange, CA
| | - Faris F Halaseh
- From the Center for Tissue Engineering, UC Irvine Department of Plastic Surgery, Orange, CA
| | | | | | | | - Gregory R D Evans
- From the Center for Tissue Engineering, UC Irvine Department of Plastic Surgery, Orange, CA
| | - Alan D Widgerow
- From the Center for Tissue Engineering, UC Irvine Department of Plastic Surgery, Orange, CA
| |
Collapse
|
20
|
Feng J, Fu S, Luan J. Selection of Mechanical Fragmentation Methods Based on Enzyme-Free Preparation of Decellularized Adipose-Derived Matrix. Bioengineering (Basel) 2023; 10:758. [PMID: 37508785 PMCID: PMC10376183 DOI: 10.3390/bioengineering10070758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/20/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND The decellularized adipose-derived matrix (DAM) has emerged as a promising biomaterial for inducing adipose tissue regeneration. Various methods have been employed to produce DAM, among which the enzyme-free method is a relatively recent preparation technique. The mechanical fragmentation step plays a crucial role in determining the efficacy of the enzyme-free preparation. METHODS The adipose tissue underwent fragmentation through the application of ultrasonication, homogenization, and freeze ball milling. This study compared the central temperature of the mixture immediately following crushing, the quantity of oil obtained after centrifugation, and the thickness of the middle layer. Fluorescence staining was utilized to compare the residual cell activity of the broken fat in the middle layer, while electron microscopy was employed to assess the integrity and properties of the adipocytes among the three methods. The primary products obtained through the three methods were subsequently subjected to processing using the enzyme-free method DAM. The assessment of degreasing and denucleation of DAM was conducted through HE staining, oil red staining, and determination of DNA residues. Subsequently, the ultrasonication-DAM (U-DAM) and homogenation-DAM (H-DAM) were implanted bilaterally on the back of immunocompromised mice, and a comparative analysis of their adipogenic and angiogenic effects in vivo was performed. RESULTS Oil discharge following ultrasonication and homogenization was significantly higher compared to that observed after freeze ball milling (p < 0.001), despite the latter exhibiting the lowest center temperature (p < 0.001). The middle layer was found to be thinnest after ultrasonication (p < 0.001), and most of the remaining cells were observed to be dead following fragmentation. Except for DAM obtained through freeze ball milling, DAM obtained through ultrasonication and homogenization could be completely denucleated and degreased. In the in vivo experiment, the first adipocytes were observed in U-DAM as early as 1 week after implantation, but not in H-DAM. After 8 weeks, a significant number of adipocytes were regenerated in both groups, but the U-DAM group demonstrated a more efficient adipose regeneration than in H-DAM (p = 0.0057). CONCLUSIONS Ultrasonication and homogenization are effective mechanical fragmentation methods for breaking down adipocytes at the initial stage, enabling the production of DAM through an enzyme-free method that facilitates successful regeneration of adipose tissues in vivo. Furthermore, the enzyme-free method, which is based on the ultrasonication pre-fragmentation approach, exhibits superior performance in terms of denucleation, degreasing, and the removal of non-adipocyte matrix components, thereby resulting in the highest in vivo adipogenic induction efficiency.
Collapse
Affiliation(s)
- Jiayi Feng
- Breast Plastic and Reconstructive Surgery Center, Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100144, China
| | - Su Fu
- Breast Plastic and Reconstructive Surgery Center, Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100144, China
| | - Jie Luan
- Breast Plastic and Reconstructive Surgery Center, Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100144, China
| |
Collapse
|
21
|
Morrison RA, Brookes S, Puls TJ, Cox A, Gao H, Liu Y, Voytik-Harbin SL. Engineered collagen polymeric materials create noninflammatory regenerative microenvironments that avoid classical foreign body responses. Biomater Sci 2023; 11:3278-3296. [PMID: 36942875 PMCID: PMC10152923 DOI: 10.1039/d3bm00091e] [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: 01/18/2023] [Accepted: 02/26/2023] [Indexed: 03/23/2023]
Abstract
The efficacy and longevity of medical implants and devices is largely determined by the host immune response, which extends along a continuum from pro-inflammatory/pro-fibrotic to anti-inflammatory/pro-regenerative. Using a rat subcutaneous implantation model, along with histological and transcriptomics analyses, we characterized the tissue response to a collagen polymeric scaffold fabricated from polymerizable type I oligomeric collagen (Oligomer) in comparison to commercial synthetic and collagen-based products. In contrast to commercial biomaterials, no evidence of an immune-mediated foreign body reaction, fibrosis, or bioresorption was observed with Oligomer scaffolds for beyond 60 days. Oligomer scaffolds were noninflammatory, eliciting minimal innate inflammation and immune cell accumulation similar to sham surgical controls. Genes associated with Th2 and regulatory T cells were instead upregulated, implying a novel pathway to immune tolerance and regenerative remodeling for biomaterials.
Collapse
Affiliation(s)
- Rachel A Morrison
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA.
| | - Sarah Brookes
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA.
| | | | - Abigail Cox
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA
| | - Hongyu Gao
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yunlong Liu
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sherry L Voytik-Harbin
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA.
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
22
|
Khan RL, Khraibi AA, Dumée LF, Corridon PR. From waste to wealth: Repurposing slaughterhouse waste for xenotransplantation. Front Bioeng Biotechnol 2023; 11:1091554. [PMID: 36815880 PMCID: PMC9935833 DOI: 10.3389/fbioe.2023.1091554] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 01/23/2023] [Indexed: 02/05/2023] Open
Abstract
Slaughterhouses produce large quantities of biological waste, and most of these materials are underutilized. In many published reports, the possibility of repurposing this form of waste to create biomaterials, fertilizers, biogas, and feeds has been discussed. However, the employment of particular offal wastes in xenotransplantation has yet to be extensively uncovered. Overall, viable transplantable tissues and organs are scarce, and developing bioartificial components using such discarded materials may help increase their supply. This perspective manuscript explores the viability and sustainability of readily available and easily sourced slaughterhouse waste, such as blood vessels, eyes, kidneys, and tracheas, as starting materials in xenotransplantation derived from decellularization technologies. The manuscript also examines the innovative use of animal stem cells derived from the excreta to create a bioartificial tissue/organ platform that can be translated to humans. Institutional and governmental regulatory approaches will also be outlined to support this endeavor.
Collapse
Affiliation(s)
- Raheema L. Khan
- Department of Immunology and Physiology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Ali A. Khraibi
- Department of Immunology and Physiology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Ludovic F. Dumée
- Department of Chemical Engineering, College of Engineering, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Research and Innovation Center on CO2 and Hydrogen (RICH), Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Peter R. Corridon
- Department of Immunology and Physiology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Healthcare Engineering Innovation Center, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| |
Collapse
|
23
|
Yu Y, Shen H, Wang X, Gibril ME, Kong F, Wang S. Spherical nanoparticle-modified bacterial cellulose drives SH−SY5Y cell differentiation and inhibits bacterial proliferation. Process Biochem 2022. [DOI: 10.1016/j.procbio.2022.10.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
24
|
Zhong JX, Raghavan P, Desai TA. Harnessing Biomaterials for Immunomodulatory-Driven Tissue Engineering. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2022; 9:224-239. [PMID: 37333620 PMCID: PMC10272262 DOI: 10.1007/s40883-022-00279-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/08/2022] [Accepted: 09/07/2022] [Indexed: 11/29/2022]
Abstract
Abstract The immune system plays a crucial role during tissue repair and wound healing processes. Biomaterials have been leveraged to assist in this in situ tissue regeneration process to dampen the foreign body response by evading or suppressing the immune system. An emerging paradigm within regenerative medicine is to use biomaterials to influence the immune system and create a pro-reparative microenvironment to instigate endogenously driven tissue repair. In this review, we discuss recent studies that focus on immunomodulation of innate and adaptive immune cells for tissue engineering applications through four biomaterial-based mechanisms of action: biophysical cues, chemical modifications, drug delivery, and sequestration. These materials enable augmented regeneration in various contexts, including vascularization, bone repair, wound healing, and autoimmune regulation. While further understanding of immune-material interactions is needed to design the next generation of immunomodulatory biomaterials, these materials have already demonstrated great promise for regenerative medicine. Lay Summary The immune system plays an important role in tissue repair. Many biomaterial strategies have been used to promote tissue repair, and recent work in this area has looked into the possibility of doing repair by tuning. Thus, we examined the literature for recent works showcasing the efficacy of these approaches in animal models of injuries. In these studies, we found that biomaterials successfully tuned the immune response and improved the repair of various tissues. This highlights the promise of immune-modulating material strategies to improve tissue repair.
Collapse
Affiliation(s)
- Justin X. Zhong
- UC Berkeley – UCSF Graduate Program in Bioengineering, San Francisco, CA 94143 USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94143 USA
| | - Preethi Raghavan
- UC Berkeley – UCSF Graduate Program in Bioengineering, San Francisco, CA 94143 USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94143 USA
| | - Tejal A. Desai
- UC Berkeley – UCSF Graduate Program in Bioengineering, San Francisco, CA 94143 USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94143 USA
- Department of Bioengineering, University of California, Berkeley, CA 94720 USA
- School of Engineering, Brown University, Providence, RI 02912 USA
| |
Collapse
|
25
|
Inflammation-mediated matrix remodeling of extracellular matrix-mimicking biomaterials in tissue engineering and regenerative medicine. Acta Biomater 2022; 151:106-117. [PMID: 35970482 DOI: 10.1016/j.actbio.2022.08.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 06/30/2022] [Accepted: 08/08/2022] [Indexed: 12/12/2022]
Abstract
Extracellular matrix (ECM)-mimicking biomaterials are considered effective tissue-engineered scaffolds for regenerative medicine because of their biocompatibility, biodegradability, and bioactivity. ECM-mimicking biomaterials preserve natural microstructures and matrix-related bioactive components and undergo continuous matrix remodeling upon transplantation. The interaction between host immune cells and transplanted ECM-mimicking biomaterials has attracted considerable attention in recent years. Transplantation of biomaterials may initiate injuries and early pro-inflammation reactions characterized by infiltration of neutrophils and M1 macrophages. Pro-inflammation reactions may lead to degradation of the transplanted biomaterial and drive the matrix into a fetal-like state. ECM degradation leads to the release of matrix-related bioactive components that act as signals for cell migration, proliferation, and differentiation. In late stages, pro-inflammatory cells fade away, and anti-inflammatory cells emerge, which involves macrophage polarization to the M2 phenotype and leukocyte activation to T helper 2 (Th2) cells. These anti-inflammatory cells interact with each other to facilitate matrix deposition and tissue reconstruction. Deposited ECM molecules serve as vital components of the mature tissue and influence tissue homeostasis. However, dysregulation of matrix remodeling results in several pathological conditions, such as aggressive inflammation, difficult healing, and non-functional fibrosis. In this review, we summarize the characteristics of inflammatory responses in matrix remodeling after transplantation of ECM-mimicking biomaterials. Additionally, we discuss the intrinsic linkages between matrix remodeling and tissue regeneration. STATEMENT OF SIGNIFICANCE: Extracellular matrix (ECM)-mimicking biomaterials are effectively used as scaffolds in tissue engineering and regenerative medicine. However, dysregulation of matrix remodeling can cause various pathological conditions. Here, the review describes the characteristics of inflammatory responses in matrix remodeling after transplantation of ECM-mimicking biomaterials. Additionally, we discuss the intrinsic linkages between matrix remodeling and tissue regeneration. We believe that understanding host immune responses to matrix remodeling of transplanted biomaterials is important for directing effective tissue regeneration of ECM-mimicking biomaterials. Considering the close relationship between immune response and matrix remodeling results, we highlight the need for studies of the effects of clinical characteristics on matrix remodeling of transplanted biomaterials.
Collapse
|
26
|
Liu K, He Y, Lu F. Research Progress on the Immunogenicity and Regeneration of Acellular Adipose Matrix: A Mini Review. Front Bioeng Biotechnol 2022; 10:881523. [PMID: 35733521 PMCID: PMC9207478 DOI: 10.3389/fbioe.2022.881523] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 05/18/2022] [Indexed: 11/13/2022] Open
Abstract
Acellular adipose matrix (AAM) has received increasing attention for soft tissue reconstruction, due to its abundant source, high long-term retention rate and in vivo adipogenic induction ability. However, the current decellularization methods inevitably affect native extracellular matrix (ECM) properties, and the residual antigens can trigger adverse immune reactions after transplantation. The behavior of host inflammatory cells mainly decides the regeneration of AAM after transplantation. In this review, recent knowledge of inflammatory cells for acellular matrix regeneration will be discussed. These advancements will inform further development of AAM products with better properties.
Collapse
|
27
|
Hayes DJ, Gimble JM. Developing a clinical grade human adipose decellularized biomaterial. BIOMATERIALS AND BIOSYSTEMS 2022; 7:100053. [PMID: 36824487 PMCID: PMC9934471 DOI: 10.1016/j.bbiosy.2022.100053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/18/2022] [Accepted: 05/24/2022] [Indexed: 11/29/2022] Open
Abstract
While tissue engineering investigators have appreciated adipose tissue as a repository of stromal/stem cells, they are only now beginning to see its value as a decellularized tissue resource. Independent academic investigators have successfully extracted lipid, genomic DNA and proteins from human fat to create a decellularized extracellular matrix enriched in collagen, glycoproteins, and proteoglycans. Pre-clinical studies have validated its compatibility with stromal/stem cells and its ability to support adipogenesis in vitro and in vivo in both small (murine) and large (porcine) subcutaneous implant models. Furthermore, Phase I safety clinical trials have injected decellularized human adipose tissue scaffolds in human volunteers without incident for periods of up to 127 days. This commentary takes an opinionated look at the under-appreciated but potential benefits of obesity as an increasingly available biomaterial resource.
Collapse
Affiliation(s)
- Daniel J. Hayes
- Department of Biomedical Engineering, Pennsylvania State University, State College, PA, USA
| | - Jeffrey M Gimble
- Obatala Sciences Inc., New Orleans, LA, USA,Corresponding author
| |
Collapse
|
28
|
Moore EM, Maestas DR, Cherry CC, Garcia JA, Comeau HY, Davenport Huyer L, Kelly SH, Peña AN, Blosser RL, Rosson GD, Elisseeff JH. Biomaterials direct functional B cell response in a material-specific manner. SCIENCE ADVANCES 2021; 7:eabj5830. [PMID: 34851674 PMCID: PMC8635437 DOI: 10.1126/sciadv.abj5830] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 10/13/2021] [Indexed: 05/13/2023]
Abstract
B cells are an adaptive immune target of biomaterials development in vaccine research but, despite their role in wound healing, have not been extensively studied in regenerative medicine. To probe the role of B cells in biomaterial scaffold response, we evaluated the B cell response to biomaterial materials implanted in a muscle wound using a biological extracellular matrix (ECM), as a reference for a naturally derived material, and synthetic polyester polycaprolactone (PCL), as a reference for a synthetic material. In the local muscle tissue, small numbers of B cells are present in response to tissue injury and biomaterial implantation. The ECM materials induced mature B cells in lymph nodes and antigen presentation in the spleen. The synthetic PCL implants resulted in prolonged B cell presence in the wound and induced an antigen-presenting phenotype. In summary, the adaptive B cell immune response to biomaterial induces local, regional, and systemic immunological changes.
Collapse
Affiliation(s)
- Erika M. Moore
- Department of Materials Science and Engineering, University of Florida, Gainesville, FL, USA
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - David R. Maestas
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Chris C. Cherry
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jordan A. Garcia
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Hannah Y. Comeau
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Locke Davenport Huyer
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Sean H. Kelly
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Alexis N. Peña
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Richard L. Blosser
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Gedge D. Rosson
- Division of Plastic Surgery, Department of Surgery, Johns Hopkins University, Baltimore, MD, USA
| | - Jennifer H. Elisseeff
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|