1
|
Hedemann N, Thomas A, Tribian N, Amler AK, Krüger S, Holthaus D, Huebbe P, Flörkemeier I, Weimer J, Maass N, Kloke L, Bauerschlag D, van Mackelenbergh MT. Light-based multi-material bioprinting of vascularised adipose tissue for breast fatty tissue engineering. Biofabrication 2025; 17:025034. [PMID: 39978067 DOI: 10.1088/1758-5090/adb890] [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: 05/06/2024] [Accepted: 02/20/2025] [Indexed: 02/22/2025]
Abstract
Reconstructive surgery following breast cancer ablation is a surgical gold standard, but current options comprising autologous fatty tissue transfer and artificial soft tissue implants are inferior. With the advent of powerful biofabrication technologies, researchers for the first time have the tools to engineer life-like tissues with the ultimate goal of clinical application. Here, we apply multi-material stereolithographic bioprinting together with a novel sacrificial biomaterial system to engineer complex fatty tissue constructs. Biomaterials, cellular composition and cultivation conditions of these constructs were designed to enablein vitrocreation of vascularised fatty tissue. Cells within the constructs showed an overall good survival (>93%), indicated by live-dead cell staining, over the entire cultivation period of 27 d. Adipose-derived stem cells were successfully differentiatedin situ, forming fat vesicles and expressing adipocyte markers PPARγ, FAPB4 and S100B. Additionally, secretion of adipokines leptin and adiponectin into culture supernatants increased significantly. Endothelial cells vascularised the constructs, creating macro- and microvascular structures within the printed channels and extending beyond with culture time. Moreover, cells invaded into the surrounding hydrogel. The engineered fatty tissue constructs could serve as a base to develop patient-specific tissue building blocks with the final goal to achieve an all-natural reconstruction of the breast.
Collapse
Affiliation(s)
- Nina Hedemann
- Department of Gynecology and Obstetrics, Kiel University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Alexander Thomas
- Technische Universität Berlin, Berlin, Germany
- Cellbricks GmbH, Berlin, Germany
| | - Nils Tribian
- Department of Gynecology and Obstetrics, Kiel University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | | | - Sandra Krüger
- Department of Pathology, Kiel University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - David Holthaus
- Department of Gynecology and Obstetrics, Kiel University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Patricia Huebbe
- Institute of Human Nutrition and Food Science, Christian-Albrechts-University, Kiel, Germany
| | - Inken Flörkemeier
- Department of Gynecology and Obstetrics, Kiel University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Jörg Weimer
- Department of Gynecology and Obstetrics, Kiel University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Nicolai Maass
- Department of Gynecology and Obstetrics, Kiel University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | | | - Dirk Bauerschlag
- Department of Gynecology and Obstetrics, Kiel University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Marion Tina van Mackelenbergh
- Department of Gynecology and Obstetrics, Kiel University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| |
Collapse
|
2
|
Kesharwani P, Alexander A, Shukla R, Jain S, Bisht A, Kumari K, Verma K, Sharma S. Tissue regeneration properties of hydrogels derived from biological macromolecules: A review. Int J Biol Macromol 2024; 271:132280. [PMID: 38744364 DOI: 10.1016/j.ijbiomac.2024.132280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 05/02/2024] [Accepted: 05/08/2024] [Indexed: 05/16/2024]
Abstract
The successful tissue engineering depends on the development of biologically active scaffolds that possess optimal characteristics to effectively support cellular functions, maintain structural integrity and aid in tissue regeneration. Hydrogels have emerged as promising candidates in tissue regeneration due to their resemblance to the natural extracellular matrix and their ability to support cell survival and proliferation. The integration of hydrogel scaffold into the polymer has a variable impact on the pseudo extracellular environment, fostering cell growth/repair. The modification in size, shape, surface morphology and porosity of hydrogel scaffolds has consequently paved the way for addressing diverse challenges in the tissue engineering process such as tissue architecture, vascularization and simultaneous seeding of multiple cells. The present review provides a comprehensive update on hydrogel production using natural and synthetic biomaterials and their underlying mechanisms. Furthermore, it delves into the application of hydrogel scaffolds in tissue engineering for cardiac tissues, cartilage tissue, adipose tissue, nerve tissue and bone tissue. Besides, the present article also highlights various clinical studies, patents, and the limitations associated with hydrogel-based scaffolds in recent times.
Collapse
Affiliation(s)
- Payal Kesharwani
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan, India; Institute of Pharmacy, Ram-Eesh Institute of Vocational and Technical Education Greater Noida, India
| | - Amit Alexander
- Department of Pharmaceuticals, National Institute of Pharmaceutical Education and Research, Guwahati, Assam, India
| | - Rahul Shukla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Lucknow, Uttar Pradesh, India
| | - Smita Jain
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan, India
| | - Akansha Bisht
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan, India
| | - Kajal Kumari
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan, India
| | - Kanika Verma
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan, India
| | - Swapnil Sharma
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan, India.
| |
Collapse
|
3
|
Xiong C, Yao W, Tao R, Yang S, Jiang W, Xu Y, Zhang J, Han Y. Application of Decellularized Adipose Matrix as a Bioscaffold in Different Tissue Engineering. Aesthetic Plast Surg 2024; 48:1045-1053. [PMID: 37726399 DOI: 10.1007/s00266-023-03608-4] [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: 06/22/2023] [Accepted: 08/10/2023] [Indexed: 09/21/2023]
Abstract
With the development of tissue engineering, the application of decellularized adipose matrix as scaffold material in tissue engineering has been intensively explored due to its wide source and excellent potential in tissue regeneration. Decellularized adipose matrix is a promising candidate for adipose tissue regeneration, while modification of decellularized adipose matrix scaffold can also allow it to transcend the limitations of adipose tissue source properties and applied to other tissue engineering fields, including cartilage and bone tissue engineering, neural tissue engineering, and skin tissue engineering. In this review, we summarized the development of the applications of decellularized adipose matrix in different tissue engineering and present future perspectives.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)
- Chenlu Xiong
- School of Medicine, Nankai University, Tianjin, China
- Department of Plastic and Reconstructive Surgery, The First Medical Centre, Chinese PLA General Hospital, 28 Fuxing Street, Beijing, 100853, China
| | - Wende Yao
- School of Medicine, Nankai University, Tianjin, China
- Department of Plastic and Reconstructive Surgery, The First Medical Centre, Chinese PLA General Hospital, 28 Fuxing Street, Beijing, 100853, China
| | - Ran Tao
- Department of Plastic and Reconstructive Surgery, The First Medical Centre, Chinese PLA General Hospital, 28 Fuxing Street, Beijing, 100853, China
| | - Sihan Yang
- School of Medicine, Nankai University, Tianjin, China
- Department of Plastic and Reconstructive Surgery, The First Medical Centre, Chinese PLA General Hospital, 28 Fuxing Street, Beijing, 100853, China
| | - Weiqian Jiang
- Department of Plastic and Reconstructive Surgery, The First Medical Centre, Chinese PLA General Hospital, 28 Fuxing Street, Beijing, 100853, China
| | - Yujian Xu
- Department of Plastic and Reconstructive Surgery, The First Medical Centre, Chinese PLA General Hospital, 28 Fuxing Street, Beijing, 100853, China
| | - Julei Zhang
- Department of Plastic and Reconstructive Surgery, The First Medical Centre, Chinese PLA General Hospital, 28 Fuxing Street, Beijing, 100853, China.
- Department of Burn and Plastic Surgery, The 980st Hospital of the PLA Joint Logistics Support Force, Hebei, China.
| | - Yan Han
- Department of Plastic and Reconstructive Surgery, The First Medical Centre, Chinese PLA General Hospital, 28 Fuxing Street, Beijing, 100853, China.
| |
Collapse
|
4
|
De Maeseneer T, Van Damme L, Aktan MK, Braem A, Moldenaers P, Van Vlierberghe S, Cardinaels R. Powdered Cross-Linked Gelatin Methacryloyl as an Injectable Hydrogel for Adipose Tissue Engineering. Gels 2024; 10:167. [PMID: 38534585 DOI: 10.3390/gels10030167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 02/18/2024] [Accepted: 02/20/2024] [Indexed: 03/28/2024] Open
Abstract
The tissue engineering field is currently advancing towards minimally invasive procedures to reconstruct soft tissue defects. In this regard, injectable hydrogels are viewed as excellent scaffold candidates to support and promote the growth of encapsulated cells. Cross-linked gelatin methacryloyl (GelMA) gels have received substantial attention due to their extracellular matrix-mimicking properties. In particular, GelMA microgels were recently identified as interesting scaffold materials since the pores in between the microgel particles allow good cell movement and nutrient diffusion. The current work reports on a novel microgel preparation procedure in which a bulk GelMA hydrogel is ground into powder particles. These particles can be easily transformed into a microgel by swelling them in a suitable solvent. The rheological properties of the microgel are independent of the particle size and remain stable at body temperature, with only a minor reversible reduction in elastic modulus correlated to the unfolding of physical cross-links at elevated temperatures. Salts reduce the elastic modulus of the microgel network due to a deswelling of the particles, in addition to triple helix denaturation. The microgels are suited for clinical use, as proven by their excellent cytocompatibility. The latter is confirmed by the superior proliferation of encapsulated adipose tissue-derived stem cells in the microgel compared to the bulk hydrogel. Moreover, microgels made from the smallest particles are easily injected through a 20G needle, allowing a minimally invasive delivery. Hence, the current work reveals that powdered cross-linked GelMA is an excellent candidate to serve as an injectable hydrogel for adipose tissue engineering.
Collapse
Affiliation(s)
- Tess De Maeseneer
- Soft Matter, Rheology and Technology, Department of Chemical Engineering, KU Leuven, Celestijnenlaan 200J Box 2424, 3001 Leuven, Belgium
| | - Lana Van Damme
- Polymer Chemistry & Biomaterials Group, Centre of Macromolecular Chemistry, Department of Organic and Macromolecular Chemistry, Ghent University (UGent), Krijgslaan 281, S4-Bis, 9000 Ghent, Belgium
| | - Merve Kübra Aktan
- Biomaterials and Tissue Engineering Research Group, Department of Materials Engineering, KU Leuven, Kasteelpark Arenberg 44 Box 2450, 3001 Leuven, Belgium
| | - Annabel Braem
- Biomaterials and Tissue Engineering Research Group, Department of Materials Engineering, KU Leuven, Kasteelpark Arenberg 44 Box 2450, 3001 Leuven, Belgium
| | - Paula Moldenaers
- Soft Matter, Rheology and Technology, Department of Chemical Engineering, KU Leuven, Celestijnenlaan 200J Box 2424, 3001 Leuven, Belgium
| | - Sandra Van Vlierberghe
- Polymer Chemistry & Biomaterials Group, Centre of Macromolecular Chemistry, Department of Organic and Macromolecular Chemistry, Ghent University (UGent), Krijgslaan 281, S4-Bis, 9000 Ghent, Belgium
| | - Ruth Cardinaels
- Soft Matter, Rheology and Technology, Department of Chemical Engineering, KU Leuven, Celestijnenlaan 200J Box 2424, 3001 Leuven, Belgium
- Processing and Performance of Materials, Department of Mechanical Engineering, TU Eindhoven, P.O. Box 513, 5600 MB Eindhoven, The Netherlands
| |
Collapse
|
5
|
Zhang S, Chen M, Geng Z, Liu T, Li S, Yu Q, Cao L, Liu D. Potential Application of Self-Assembled Peptides and Proteins in Breast Cancer and Cervical Cancer. Int J Mol Sci 2023; 24:17056. [PMID: 38069380 PMCID: PMC10706889 DOI: 10.3390/ijms242317056] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/22/2023] [Accepted: 11/30/2023] [Indexed: 12/18/2023] Open
Abstract
Ongoing research is gradually broadening the idea of cancer treatment, with attention being focused on nanoparticles to improve the stability, therapeutic efficacy, targeting, and other important metrics of conventional drugs and traditional drug delivery methods. Studies have demonstrated that drug delivery carriers based on biomaterials (e.g., protein nanoparticles and lipids) and inorganic materials (e.g., metal nanoparticles) have potential anticancer effects. Among these carriers, self-assembled proteins and peptides, which are highly biocompatible and easy to standardize and produce, are strong candidates for the preparation of anticancer drugs. Breast cancer (BC) and cervical cancer (CC) are two of the most common and deadly cancers in women. These cancers not only threaten lives globally but also put a heavy burden on the healthcare system. Despite advances in medical care, the incidence of these two cancers, particularly CC, which is almost entirely preventable, continues to rise, and the mortality rate remains steady. Therefore, there is still a need for in-depth research on these two cancers to develop more targeted, efficacious, and safe therapies. This paper reviews the types of self-assembling proteins and peptides (e.g., ferritin, albumin, and virus-like particles) and natural products (e.g., soy and paclitaxel) commonly used in the treatment of BC and CC and describes the types of drugs that can be delivered using self-assembling proteins and peptides as carriers (e.g., siRNAs, DNA, plasmids, and mRNAs). The mechanisms (including self-assembly) by which the natural products act on CC and BC are discussed. The mechanism of action of natural products on CC and BC and the mechanism of action of self-assembled proteins and peptides have many similarities (e.g., NF-KB and Wnt). Thus, natural products using self-assembled proteins and peptides as carriers show potential for the treatment of BC and CC.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Lingling Cao
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (S.Z.); (M.C.); (Z.G.); (T.L.); (S.L.); (Q.Y.)
| | - Da Liu
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (S.Z.); (M.C.); (Z.G.); (T.L.); (S.L.); (Q.Y.)
| |
Collapse
|
6
|
King O, Pérez-Madrigal MM, Murphy ER, Hmayed AAR, Dove AP, Weems AC. 4D Printable Salicylic Acid Photopolymers for Sustained Drug Releasing, Shape Memory, Soft Tissue Scaffolds. Biomacromolecules 2023; 24:4680-4694. [PMID: 37747816 DOI: 10.1021/acs.biomac.3c00416] [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: 09/27/2023]
Abstract
3D printing of pharmaceuticals offers a unique opportunity for long-term, sustained drug release profiles for an array of treatment options. Unfortunately, this approach is often limited by physical compounding or processing limitations. Modification of the active drug into a prodrug compound allows for seamless incorporation with advanced manufacturing methods that open the door to production of complex tissue scaffold drug depots. Here we demonstrate this concept using salicylic acids with varied prodrug structures for control of physical and chemical properties. The role of different salicylic acid derivatives (salicylic acid, bromosalicylic allyl ester, iodosalicylic allyl ester) and linker species (allyl salicylate, allyl 2-(allyloxy)benzoate, allyl 2-(((allyloxy)carbonyl)oxy)benzoate) were investigated using thiol-ene cross-linking in digital light processing (DLP) 3D printing to produce porous prodrug tissue scaffolds containing more than 50% salicylic acid by mass. Salicylic acid photopolymer resins were all found to be highly reactive (solidification within 5 s of irradiation at λ = 405 nm), while the cross-linked solids display tunable thermomechanical behaviors with low glass transition temperatures (Tgs) and elastomeric behaviors, with the carbonate species displaying an elastic modulus matching that of adipose tissue (approximately 65 kPa). Drug release profiles were found to be zero order, sustained release based upon hydrolytic degradation of multilayered scaffolds incorporating fluorescent modeling compounds, with release rates tuned through selection of the linker species. Cytocompatibility in 2D and 3D was further demonstrated for all species compared to polycarbonate controls, as well as salicylic acid-containing composites (physical incorporation), over a 2-week period using murine fibroblasts. The use of drugs as the matrix material for solid prodrug tissue scaffolds opens the door to novel therapeutic strategies, longer sustained release profiles, and even reduced complications for advanced medicine.
Collapse
Affiliation(s)
- Olivia King
- Biomedical Engineering, Russ College of Engineering, Ohio University, Athens, Ohio 45701, United States
| | - Maria M Pérez-Madrigal
- School of Chemistry, University of Birmingham, Birmingham B15 2TT, U.K
- Departament d'Enginyeria Química, Campus Diagonal Besòs (EEBE), Universitat Politècnica de Catalunya, C/Eduard Maristany, 10-14, 08019, Barcelona, Spain
- Barcelona Research Center for Multiscale Science and Engineering, Campus Diagonal Besòs (EEBE), Universitat Politècnica de Catalunya, C/Eduard Maristany, 10-14, 08019, Barcelona, Spain
| | - Erin R Murphy
- Molecular and Cellular Biology Program, Ohio University, Athens, Ohio 45701, United States
- Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio 45701, United States
- Infectious and Tropical Diseases Institute, Ohio University, Athens, Ohio 45701, United States
| | | | - Andrew P Dove
- School of Chemistry, University of Birmingham, Birmingham B15 2TT, U.K
| | - Andrew C Weems
- Biomedical Engineering, Russ College of Engineering, Ohio University, Athens, Ohio 45701, United States
- School of Chemistry, University of Birmingham, Birmingham B15 2TT, U.K
- Molecular and Cellular Biology Program, Ohio University, Athens, Ohio 45701, United States
- Mechanical Engineering, Russ College of Engineering, Ohio Musculoskeletal and Neurological Institute, Ohio University, Athens, Ohio 45701, United States
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
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
|
10
|
LCAT-trial-24 weeks: Protocol for a clinical study to evaluate the safety of regenerative medicine and gene therapy by the autologous transplantation of human lecithin:cholesterol acyltransferase gene-transduced human pre-adipocytes. Contemp Clin Trials Commun 2022; 28:100946. [PMID: 35734220 PMCID: PMC9207543 DOI: 10.1016/j.conctc.2022.100946] [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: 01/21/2022] [Revised: 05/06/2022] [Accepted: 06/08/2022] [Indexed: 11/22/2022] Open
Abstract
Backgrounds Despite the absolute need for life-long treatment of inherited and genetic diseases, there has been little effort to develop such treatments for most of these conditions due to their rarity. Familial lecithin:cholesterol acyltransferase (LCAT) deficiency is recognized as one such orphan disease. We have been developing an adipocyte-based ex vivo gene therapy/regenerative medicine, a novel methodology that differs from the adeno-associated virus-mediated in vivo gene therapy or ex vivo gene-transduced hematopoietic cell therapy, to treat familial LCAT deficiency. Recently, a first-in-human (FIH) clinical study was conducted under the Act on Securement of Safety of Regenerative Medicine, wherein a patient with familial LCAT deficiency was treated. To obtain approval to put this treatment into practical use, a clinical trial has been designed with reference to the FIH clinical study. Methods An interventional, open-label, unblinded dose-escalation trial was planned, referring to previous FIH clinical study. The trial aims to evaluate the safety of the investigational product in relation to the characteristics of the investigational product (ex vivo gene/cell therapy product by retroviral vector-mediated LCAT gene transduction) using two doses, and the efficacy of the treatment will be evaluated exploratively. A total of three patients will be enrolled sequentially and followed for 24 weeks after administration. This study is designed as a multicenter trial, with Chiba University Hospital administering and evaluating the safety/efficacy of the investigational products at the prescribed visit. Conclusion This clinical trial is expected to facilitate the provision of lifelong treatment to many patients with LCAT deficiency. Trial registration number Japan Registry of Clinical Trials (jRCT2033200096). Familial LCAT deficiency is an orphan disease without any effective treatment. We have been developing a novel adipocyte-based ex vivo gene therapy that enables life-long enzyme replacement. This clinical study was designed to assess the dose-response and the safety in a limited number of orphan disease patients. The protocol includes a preclinical tumorigenicity test in immunodeficient (NSG) mice as one of the primary endpoints.
Collapse
|
11
|
Pan Y, Xiao Y, Hao Y, Shi K, Pan M, Qian Z. An injectable mPEG-PDLLA microsphere/PDLLA-PEG-PDLLA hydrogel composite for soft tissue augmentation. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2021.12.093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
12
|
Kim SH, Kim D, Cha M, Kim SH, Jung Y. The Regeneration of Large-Sized and Vascularized Adipose Tissue Using a Tailored Elastic Scaffold and dECM Hydrogels. Int J Mol Sci 2021; 22:ijms222212560. [PMID: 34830444 PMCID: PMC8624932 DOI: 10.3390/ijms222212560] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/12/2021] [Accepted: 11/17/2021] [Indexed: 12/03/2022] Open
Abstract
A dome-shaped elastic poly(l-lactide-co-caprolactone) (PLCL) scaffold with a channel and pore structure was fabricated by a combinative method of 3D printing technology and the gel pressing method (13 mm in diameter and 6.5 mm in thickness) for patient-specific regeneration. The PLCL scaffold was combined with adipose decellularized extracellular matrix (adECM) and heart decellularized extracellular matrix (hdECM) hydrogels and human adipose-derived stem cells (hADSCs) to promote adipogenesis and angiogenesis. These scaffolds had mechanical properties similar to those of native adipose tissue for improved tissue regeneration. The results of the in vitro real-time PCR showed that the dECM hydrogel mixture induces adipogenesis. In addition, the in vivo study at 12 weeks demonstrated that the tissue-engineered PLCL scaffolds containing the hydrogel mixture (hdECM/adECM (80:20)) and hADSCs promoted angiogenesis and adipose tissue formation, and suppressed apoptosis. Therefore, we expect that our constructs will be clinically applicable as material for the regeneration of patient-specific large-sized adipose tissue.
Collapse
Affiliation(s)
- Su Hee Kim
- Biomaterials Research Center, Korea Institute of Science and Technology (KIST), 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Korea; (S.H.K.); (D.K.)
- R&D Center, Medifab Co., Ltd., 70 Dusan-ro, Geumcheon-gu, Seoul 08584, Korea;
| | - Donghak Kim
- Biomaterials Research Center, Korea Institute of Science and Technology (KIST), 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Korea; (S.H.K.); (D.K.)
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Korea
| | - Misun Cha
- R&D Center, Medifab Co., Ltd., 70 Dusan-ro, Geumcheon-gu, Seoul 08584, Korea;
| | - Soo Hyun Kim
- Biomaterials Research Center, Korea Institute of Science and Technology (KIST), 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Korea; (S.H.K.); (D.K.)
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Korea
- Korea Institute of Science and Technology (KIST) Europe, Campus E 7.1, 66123 Saarbrücken, Germany
- Correspondence: (S.H.K.); (Y.J.)
| | - Youngmee Jung
- Biomaterials Research Center, Korea Institute of Science and Technology (KIST), 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Korea; (S.H.K.); (D.K.)
- School of Electrical and Electronic Engineering, YU-KIST Institute, Yonsei University, Seoul 03722, Korea
- Correspondence: (S.H.K.); (Y.J.)
| |
Collapse
|
13
|
Gordiienko IM, Gubar OS, Sulik R, Kunakh T, Zlatskiy I, Zlatska A. Empty nose syndrome pathogenesis and cell-based biotechnology products as a new option for treatment. World J Stem Cells 2021; 13:1293-1306. [PMID: 34630863 PMCID: PMC8474723 DOI: 10.4252/wjsc.v13.i9.1293] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/29/2021] [Accepted: 08/13/2021] [Indexed: 02/06/2023] Open
Abstract
Empty nose syndrome (ENS) is a rare complication that develops after partial or complete turbinectomy. The main feature of ENS is paradoxical nasal obstruction feeling despite objectively wide nasal airway. ENS pathogenesis is multifactorial and includes changes in laminar physiological airflow, disruption of mucosa functions and deficient neural sensation. This leads to the development of ENS symptomatology such as dyspnea, nasal dryness, nasal burning, nasal obstruction, feeling of suffocation and even comorbid psychiatric disorders that significantly impairs life quality. Specific effective treatment of ENS does not exist up to date. In this review we outline existing biomaterial for surgical reconstitution of nasal anatomy and discuss the perspective of stem cell-based technologies in ENS management. The main focus is directed to justification of rationality application of adult mesenchymal stem cells (MSCs) from different tissues origin and neural crest-derived stem cells (NCSCs) based on their intrinsic biological properties. MSCs transplantation may stimulate mucosa tissue regeneration via trophic factors secretion, direct transdifferentiation into epithelial cells and pronounced immunosuppressive effect. From the other hand, NCSCs based on their high neuroprotective properties may reconstitute nerve structure and functioning leading to normal sensation in ENS patients. We postulate that application of cell-based and tissue-engineered products can help to significantly improve ENS symptomatology only as complex approach aimed at reconstitution of nasal anatomy, recovery the nasal mucosa functionality and neural tissue sensation.
Collapse
Affiliation(s)
- Inna M Gordiienko
- Biotechnology Laboratory, Medical Company “Good Cells”, Kyiv 03115, Ukraine
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology NAS of Ukraine, Kyiv 03022, Ukraine
| | - Olga S Gubar
- Institute of Molecular Biology and Genetics NAS of Ukraine, Kyiv 03143, Ukraine
| | - Roman Sulik
- Biotechnology Laboratory, Medical Company “Good Cells”, Kyiv 03115, Ukraine
| | - Taras Kunakh
- Biotechnology Laboratory, Medical Company “Good Cells”, Kyiv 03115, Ukraine
| | - Igor Zlatskiy
- State Institute of Genetic and Regenerative Medicine, National Academy of Medical Sciences of Ukraine, Kyiv 04114, Ukraine
| | - Alona Zlatska
- Biotechnology Laboratory, Medical Company “Good Cells”, Kyiv 03115, Ukraine
- State Institute of Genetic and Regenerative Medicine, National Academy of Medical Sciences of Ukraine, Kyiv 04114, Ukraine
| |
Collapse
|
14
|
Engineering a 3D Vascularized Adipose Tissue Construct Using a Decellularized Lung Matrix. Biomimetics (Basel) 2021; 6:biomimetics6030052. [PMID: 34562876 PMCID: PMC8482279 DOI: 10.3390/biomimetics6030052] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/01/2021] [Accepted: 09/10/2021] [Indexed: 01/16/2023] Open
Abstract
Critically sized defects in subcutaneous white adipose tissue result in extensive disfigurement and dysfunction and remain a reconstructive challenge for surgeons; as larger defect sizes are correlated with higher rates of complications and failure due to insufficient vascularization following implantation. Our study demonstrates, for the first time, a method to engineer perfusable, pre-vascularized, high-density adipose grafts that combine patient-derived adipose cells with a decellularized lung matrix (DLM). The lung is one of the most vascularized organs with high flow, low resistance, and a large blood-alveolar interface separated by a thin basement membrane. For our work, the large volume capacity within the alveolar compartment was repurposed for high-density adipose cell filling, while the acellular vascular bed provided efficient graft perfusion throughout. Both adipocytes and hASCs were successfully delivered and remained in the alveolar space even after weeks of culture. While adipose-derived cells maintained their morphology and functionality in both static and perfusion DLM cultures, perfusion culture offered enhanced outcomes over static culture. Furthermore, we demonstrate that endothelial cells seamlessly integrate into the acellular vascular tree of the DLM with adipocytes. These results support that the DLM is a unique platform for creating vascularized adipose tissue grafts for large defect filling.
Collapse
|
15
|
Marew T, Birhanu G. Three dimensional printed nanostructure biomaterials for bone tissue engineering. Regen Ther 2021; 18:102-111. [PMID: 34141834 PMCID: PMC8178073 DOI: 10.1016/j.reth.2021.05.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 03/29/2021] [Accepted: 05/04/2021] [Indexed: 12/13/2022] Open
Abstract
The suffering from organ dysfunction due to damaged or diseased tissue/bone has been globally on the rise. Current treatment strategies for non-union bone defects include: the use of autografts, allografts, synthetic grafts and free vascularized fibular grafts. Bone tissue engineering has emerged as an alternative for fracture repair to satisfy the current unmet need of bone grafts and to alleviate the problems associated with autografts and allografts. The technology offers the possibility to induce new functional bone regeneration using synergistic combination of functional biomaterials (scaffolds), cells, and growth factors. Bone scaffolds are typically made of porous biodegradable materials that provide the mechanical support during repair and regeneration of damaged or diseased bone. Significant progress has been made towards scaffold materials for structural support, desired osteogenesis and angiogenesis abilities. Thanks for innovative scaffolds fabrication technologies, bioresorbable scaffolds with controlled porosity and tailored properties are possible today. Despite the presence of different bone scaffold fabrication methods, pore size, shape and interconnectivity have not yet been fully controlled in most of the methods. Moreover, scaffolds with tailored porosity for specific defects are still difficult to manufacture. Nevertheless, such scaffolds can be designed and fabricated using three dimensional (3D) printing approaches. 3D printing technology, as an advanced tissue scaffold fabrication method, offers the opportunity to produce complex geometries with distinct advantages. The technology has been used for the production of various types of bodily constructs such as blood vessels, vascular networks, bones, cartilages, exoskeletons, eyeglasses, cell cultures, tissues, organs and novel drug delivery devices. This review focuses on 3D printed scaffolds and their application in bone repair and regeneration. In addition, different classes of biomaterials commonly employed for the fabrication of 3D nano scaffolds for bone tissue engineering application so far are briefly discussed.
Collapse
Affiliation(s)
- Tesfa Marew
- Department of Pharmaceutics & Social Pharmacy, School of Pharmacy, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Gebremariam Birhanu
- Department of Pharmaceutics & Social Pharmacy, School of Pharmacy, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| |
Collapse
|
16
|
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
|
17
|
Cho W, Kim BS, Ahn M, Ryu YH, Ha D, Kong JS, Rhie J, Cho D. Flexible Adipose-Vascular Tissue Assembly Using Combinational 3D Printing for Volume-Stable Soft Tissue Reconstruction. Adv Healthc Mater 2021; 10:e2001693. [PMID: 33236508 DOI: 10.1002/adhm.202001693] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 11/05/2020] [Indexed: 12/12/2022]
Abstract
A new concept, assembling cell-laden tissue modules, is for the first time proposed for soft tissue engineering. Adipose-vascular tissue modules composed of a synthetic polymer-based substructure and customized bioinks using planar 3D cell printing are engineered. Such tissue modules are systematically assembled into a synthetic polymer-based module holder fabricated with rotational 3D printing, resulting in the development of a flexible and volumetric tissue assembly. Whereas most of the previous studies about the construction of adipose tissue are limited to hypoxia, poor vascularization, rapid resorption, and mismatch in mechanical properties, it is aimed to realize the construction of nonhypoxic, flexible, and volume-stable tissue assembly in this study. The significance of engineered tissue assembly is proven through various in vitro and in vivo evaluations. In particular, stable volume and remarkable neovascularization/adipogenesis are observed in the implanted assembly over four weeks. Interestingly, the size of newly formed lipid droplets and the remodeled morphology in the assembly are comparable to those in native adipose tissue. As far as it is known, this work is a first report suggesting a cell printing-based tissue assembly for functional reconstruction of soft tissue.
Collapse
Affiliation(s)
- Won‐Woo Cho
- Department of Mechanical Engineering Pohang University of Science and Technology (POSTECH) 77 Cheongam‐ro, Nam‐gu Pohang Kyungbuk 37673 Republic of Korea
- POSTECH‐Catholic Biomedical Engineering Institute POSTECH Pohang Kyungbuk 37673 Republic of Korea
| | - Byoung Soo Kim
- POSTECH‐Catholic Biomedical Engineering Institute POSTECH Pohang Kyungbuk 37673 Republic of Korea
- Future IT Innovation Laboratory POSTECH 77 Cheongam‐ro, Nam‐gu Pohang Kyungbuk 37673 Republic of Korea
| | - Minjun Ahn
- Department of Mechanical Engineering Pohang University of Science and Technology (POSTECH) 77 Cheongam‐ro, Nam‐gu Pohang Kyungbuk 37673 Republic of Korea
- POSTECH‐Catholic Biomedical Engineering Institute POSTECH Pohang Kyungbuk 37673 Republic of Korea
| | - Yeon Hee Ryu
- Department of Biomedicine and Health Sciences College of Medicine The Catholic University of Korea 222, Banpo‐daero Seoul 06591 Republic of Korea
| | - Dong‐Heon Ha
- Department of Mechanical Engineering Pohang University of Science and Technology (POSTECH) 77 Cheongam‐ro, Nam‐gu Pohang Kyungbuk 37673 Republic of Korea
- EDmicBio Inc. 26, Kyungheedae‐ro, Dongdaemun‐gu Seoul 02447 Republic of Korea
| | - Jeong Sik Kong
- POSTECH‐Catholic Biomedical Engineering Institute POSTECH Pohang Kyungbuk 37673 Republic of Korea
- School of Interdisciplinary Bioscience and Bioengineering POSTECH 77 Cheongam‐ro, Nam‐gu Pohang Kyungbuk 37673 Republic of Korea
| | - Jong‐Won Rhie
- Department of Plastic and Reconstructive Surgery Seoul St. Mary's Hospital College of Medicine The Catholic University of Korea 222, Banpo‐daero Seoul 06591 Republic of Korea
| | - Dong‐Woo Cho
- Department of Mechanical Engineering Pohang University of Science and Technology (POSTECH) 77 Cheongam‐ro, Nam‐gu Pohang Kyungbuk 37673 Republic of Korea
- POSTECH‐Catholic Biomedical Engineering Institute POSTECH Pohang Kyungbuk 37673 Republic of Korea
| |
Collapse
|
18
|
Liu W, Xie Y, Zheng Y, He W, Qiao K, Meng H. Regulatory science for hernia mesh: Current status and future perspectives. Bioact Mater 2021; 6:420-432. [PMID: 32995670 PMCID: PMC7490592 DOI: 10.1016/j.bioactmat.2020.08.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/16/2020] [Accepted: 08/23/2020] [Indexed: 11/25/2022] Open
Abstract
Regulatory science for medical devices aims to develop new tools, standards and approaches to assess the safety, effectiveness, quality and performance of medical devices. In the field of biomaterials, hernia mesh is a class of implants that have been successfully translated to clinical applications. With a focus on hernia mesh and its regulatory science system, this paper collected and reviewed information on hernia mesh products and biomaterials in both Chinese and American markets. The current development of regulatory science for hernia mesh, including its regulations, standards, guidance documents and classification, and the scientific evaluation of its safety and effectiveness was first reported. Then the research prospect of regulatory science for hernia mesh was discussed. New methods for the preclinical animal study and new tools for the evaluation of the safety and effectiveness of hernia mesh, such as computational modeling, big data platform and evidence-based research, were assessed. By taking the regulatory science of hernia mesh as a case study, this review provided a research basis for developing a regulatory science system of implantable medical devices, furthering the systematic evaluation of the safety and effectiveness of medical devices for better regulatory decision-making. This was the first article reviewing the regulatory science of hernia mesh and biomaterial-based implants. It also proposed and explained the concepts of evidence-based regulatory science and technical review for the first time.
Collapse
Affiliation(s)
- Wenbo Liu
- School of Material Science and Engineering, University of Science and Technology Beijing, 30 Xueyuan Road, Haidian District, Beijing, China
- Center for Medical Device Evaluation, National Medical Products Administration, Intellectual Property Publishing House Mansion, Qixiang Road, Haidian District, Beijing, China
| | - Yajie Xie
- School of Material Science and Engineering, University of Science and Technology Beijing, 30 Xueyuan Road, Haidian District, Beijing, China
| | - Yudong Zheng
- School of Material Science and Engineering, University of Science and Technology Beijing, 30 Xueyuan Road, Haidian District, Beijing, China
| | - Wei He
- School of Material Science and Engineering, University of Science and Technology Beijing, 30 Xueyuan Road, Haidian District, Beijing, China
| | - Kun Qiao
- School of Material Science and Engineering, University of Science and Technology Beijing, 30 Xueyuan Road, Haidian District, Beijing, China
| | - Haoye Meng
- School of Material Science and Engineering, University of Science and Technology Beijing, 30 Xueyuan Road, Haidian District, Beijing, China
| |
Collapse
|
19
|
Wang L, Zhang CG, Jia YL, Hu L. Tissue Inhibitor of Metalloprotease-1 (TIMP-1) Regulates Adipogenesis of Adipose-derived Stem Cells (ASCs) via the Wnt Signaling Pathway in an MMP-independent Manner. Curr Med Sci 2020; 40:989-996. [PMID: 33123912 DOI: 10.1007/s11596-020-2265-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 05/14/2020] [Indexed: 12/30/2022]
Abstract
Tissue inhibitor of metalloprotease-1 (TIMP-1) is a tissue inhibitor of matrix metalloproteinases (MMPs). It however exerts multiple effects on biological processes, such as cell growth, proliferation, differentiation and apoptosis, in an MMP-independent manner. This study aimed to examine the role of TIMP-1 in adipogenesis of adipose-derived stem cells (ASCs) and the underlying mechanism. We knocked down the TIMP-1 gene in ASCs through lentiviral vectors encoding TIMP-1 small interfering RNA (siRNA), and then found that the knockdown of TIMP-1 in ASCs promoted the adipogenic differentiation of stem cells and inhibited the Wnt/β-catenin signaling pathway in ASCs. We also noted that mutant TIMP-1 without the inhibitory activity on MMPs promoted the activation of Wnt/β-catenin pathway as well as the recombinant wild type TIMP-1 did, which indicated that the effect of TIMP-1 on Wnt/β-catenin pathway was MMP-independent. Our study suggested that TIMP-1 negatively regulated the adipogenesis of ASCs via the Wnt/β-catenin signaling pathway in an MMP-independent manner.
Collapse
Affiliation(s)
- Lu Wang
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chen-Guang Zhang
- Stomatological Hospital, Southern Medical University, Guangzhou, 510000, China
| | - Yu-Lin Jia
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Li Hu
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
20
|
Injectable and Cryopreservable MSC-Loaded PLGA Microspheres for Recovery from Chemically Induced Liver Damage. Macromol Res 2020. [DOI: 10.1007/s13233-020-8139-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
21
|
Fitzgerald SJ, Cobb JS, Janorkar AV. Comparison of the formation, adipogenic maturation, and retention of human adipose-derived stem cell spheroids in scaffold-free culture techniques. J Biomed Mater Res B Appl Biomater 2020; 108:3022-3032. [PMID: 32396702 PMCID: PMC8506838 DOI: 10.1002/jbm.b.34631] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 04/16/2020] [Accepted: 04/19/2020] [Indexed: 11/02/2023]
Abstract
While three-dimensional spheroids outperform traditional two-dimensional monolayer culture for human adipose-derived stem cells (hASCs), there is not a consensus on the most successful method for enhancing their adipogenic differentiation and minimizing the loss of physiologically relevant, fatty spheroids during culture. To this end, we compared three culture methods, namely, elastin-like polypeptide-polyethyleneimine (ELP-PEI) coated surfaces, ultra-low attachment static culture, and suspension culture for their ability to form and retain productive hASC spheroids. The ELP-PEI coatings used the ELP conjugated to two molecular weights of PEI (800 or 25,000 g/mol). FTIR spectroscopy, atomic force microscopy, and contact angle goniometry revealed that the ELP-PEI coatings had similar chemical structures, surface topography, and hydrophobicity. Time-lapse microscopy showed that increasing the PEI molecular weight resulted in smaller spheroids. Measurement of triglyceride content showed that the three methods induced comparable differentiation of hASCs toward the adipogenic lineage. DNA content and morphometric analysis revealed merging of spheroids to form larger spheroids in the ultra-low attachment static culture and suspension culture methods. In contrast, the retention of hASC spheroid sizes and numbers with a regular spheroid size (~100 μm) were best atop the ELP-PEI800 coatings. Overall, this research shows that the spheroid culture atop the ELP-PEI coatings is a suitable cell culture model for future studies involving long-term, three-dimensional culture of mature adipocytes derived from hASCs.
Collapse
Affiliation(s)
- Sarah J. Fitzgerald
- Biomedical Materials Science, School of Dentistry, University of Mississippi Medical Center, 2500 N State St, Jackson, MS 39216
| | - Jared S. Cobb
- Biomedical Materials Science, School of Dentistry, University of Mississippi Medical Center, 2500 N State St, Jackson, MS 39216
| | - Amol V. Janorkar
- Biomedical Materials Science, School of Dentistry, University of Mississippi Medical Center, 2500 N State St, Jackson, MS 39216
| |
Collapse
|
22
|
Anti-HER2 antibody therapy using gene-transduced adipocytes for HER2-positive breast cancer. Breast Cancer Res Treat 2020; 180:625-634. [PMID: 32124135 DOI: 10.1007/s10549-020-05581-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 02/19/2020] [Indexed: 10/24/2022]
Abstract
PURPOSE Although recent advances in molecular target therapy have improved the survival of breast cancer patients, high cost and frequent hospital visits result in both societal and individual burden. To reduce these problems, it has been proposed to produce antibodies in vivo. Here, we constructed gene-transduced human ceiling culture-derived proliferative adipocytes secreting anti-HER2 antibody (HER2-ccdPAs) and evaluated their ability to secrete antibody and mediate an anti-tumor effect. METHODS Plasmid lentivirus was used as a recipient for anti-HER2 antibody cDNA and transduced into human proliferative adipocyte. Secretory antibody expression was evaluated by ELISA and western blot. Specific binding of secretory antibody to HER2 was examined by immunofluorescence analysis. Direct and indirect anti-tumor effects of supernatants from HER2-ccdPAs were evaluated using BT474 (HER2+) and MDA-MB-231 (HER2-) breast cancer cell lines. Additionally, whether adipocyte differentiation affects antibody secretion was investigated using supernatant collected from different cell maturation states. RESULTS Anti-HER2 antibody was identified in the supernatant from HER2-ccdPAs and its production increased with the differentiation into mature adipocyte. Antibodies in supernatants from HER2-ccdPAs bound to HER2-positive breast cancer cells similar to trastuzumab. Supernatant from HER2-ccdPAs inhibited the proliferation of BT474 but not MDA-MB-231 cells, and downregulated AKT phosphorylation in BT474 cells compared with controls. Supernatants from HER2-ccdPAs also had an indirect anti-tumor effect on BT474 cells through ADCC. Additionally, Single inoculation of HER2-ccdPAs showed an anti-tumor effect in BT474 xenograft model. CONCLUSIONS HER2-ccdPAs might be useful for cell-based gene therapy. This system could be a platform for various antibody therapies.
Collapse
|
23
|
Nie J, Yi Y, Zhu Y. [Construction of tissue engineered adipose by human adipose tissue derived extracellular vesicle combined with decellularized adipose tissues scaffold]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2020; 34:226-233. [PMID: 32030956 DOI: 10.7507/1002-1892.201903064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Objective To explore the possibility of constructing tissue engineered adipose by adipose tissue derived extracellular vesicles (hAT-EV) combined with decellularized adipose tissue (DAT) scaffolds, and to provide a new therapy for soft tissue defects. Methods The adipose tissue voluntarily donated by the liposuction patient was divided into two parts, one of them was decellularized and observed by HE and Masson staining and scanning electron microscope (SEM). Immunohistochemical staining and Western blot detection for collagen type Ⅰ and Ⅳ and laminin were also employed. Another one was incubated with exosome-removed complete medium for 48 hours, then centrifuged to collect the medium and to obtain hAT-EV via ultracentrifugation. The morphology of hAT-EV was observed by transmission electron microscopy; the nanoparticle tracking analyzer (NanoSight) was used to analyze the size distribution; Western blot was used to analyse membrane surface protein of hAT-EV. Adipose derived stem cells (ADSCs) were co-cultured with PKH26 fluorescently labeled hAT-EV, confocal fluorescence microscopy was used to observe the uptake of hAT-EV by ADSCs. Oil red O staining was used to evaluate adipogenic differentiation after hAT-EV and ADSCs co-cultured for 15 days. The DAT was scissored and then injected into the bilateral backs of 8 C57 mice (6-week-old). In experimental group, 0.2 mL hAT-EV was injected weekly, and 0.2 mL PBS was injected weekly in control group. After 12 weeks, the mice were sacrificed, and the new fat organisms on both sides were weighed. The amount of new fat was evaluated by HE and peri-lipoprotein immunofluorescence staining to evaluate the ability of hAT-EV to induce adipogenesis in vivo. Results After acellularization of adipose tissue, HE and Masson staining showed that DAT was mainly composed of loosely arranged collagen with no nucleus; SEM showed that no cells and cell fragments were found in DAT, and thick fibrous collagen bundles could be seen; immunohistochemical staining and Western blot detection showed that collagen type Ⅰ and Ⅳ and laminin were retained in DAT. It was found that hAT-EV exhibited a spherical shape of double-layer envelope, with high expressions of CD63, apoptosis-inducible factor 6 interacting protein antibody, tumor susceptibility gene 101, and the particle size of 97.9% hAT-EV ranged from 32.67 nmto 220.20 nm with a peak at 91.28 nm. Confocal fluorescence microscopy and oil red O staining showed that hAT-EV was absorbed by ADSCs and induced adipogenic differentiation. In vivo experiments showed that the wet weight of fat new organisms in the experimental group was significantly higher than that in the control group ( t=2.278, P=0.048). HE staining showed that the structure of lipid droplets in the experimental group was more than that in the control group, and the collagen content in the control group was higher than that in the experimental group. The proportion of new fat in the experimental group was significantly higher than that in the control group ( t=4.648, P=0.017). Conclusion DAT carrying hAT-EV can be used as a new method to induce adipose tissue regeneration and has a potential application prospect in the repair of soft tissue defects.
Collapse
Affiliation(s)
- Jiaying Nie
- Department of Plastic Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang Jiangxi, 330006, P.R.China
| | - Yangyan Yi
- Department of Plastic Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang Jiangxi, 330006, P.R.China
| | - Yuanzheng Zhu
- Department of Plastic Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang Jiangxi, 330006, P.R.China
| |
Collapse
|
24
|
|
25
|
Kolodziej M, Strauss S, Lazaridis A, Bucan V, Kuhbier JW, Vogt PM, Könneker S. Influence of glucose and insulin in human adipogenic differentiation models with adipose-derived stem cells. Adipocyte 2019; 8:254-264. [PMID: 31280651 PMCID: PMC6768274 DOI: 10.1080/21623945.2019.1636626] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Autologous fat grafting represents an attractive source for tissue engineering applications in the field of reconstructive medicine. However, in adipogenic differentiation protocols for human adipose-derived stem cells, the concentration of glucose and insulin varies considerably. With the intent to gain maximum tissue augmentation, we focused on the late phase of adipogenesis. In this study, we modified the differentiation protocol for adipose-derived stem cells by prolongation of the induction period and the application highly concentrated glucose and insulin. Human adipose-derived stem cells were isolated from subcutaneous depots and differentiated in a standard induction medium for the first two weeks, followed by two weeks with varying glucose and insulin concentrations. Morphological changes assessed using Oil-Red-O staining were examined for corresponding alterations in the expression of the adipogenic markers peroxisome proliferator-activated receptor gamma (PPARγ) and lipoprotein lipase (LPL). Furthermore, glucose and lactate levels in conditioned media were monitored over the period of differentiation. We found high-glucose media increasing the level of lipid accumulation and the size of single droplets whereas insulin significantly showed a dose-dependent negative effect on fat storage. However, whereas high glucose stimulated PPARγ transcription, expression levels in insulin-treated cells remained constant. Results permit assumptions that a high-glucose medium intensifies the degree of differentiation in mature adipocytes providing conditions to promote graft volume while we have identified highly concentrated insulin treatment as an inhibitor of lipid storage in the late adipogenic differentiation.
Collapse
Affiliation(s)
- Michaela Kolodziej
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, Hanover, Germany
| | - Sarah Strauss
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, Hanover, Germany
| | - Andrea Lazaridis
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, Hanover, Germany
| | - Vesna Bucan
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, Hanover, Germany
| | - Jörn W. Kuhbier
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, Hanover, Germany
| | - Peter M. Vogt
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, Hanover, Germany
| | - Sören Könneker
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, Hanover, Germany
| |
Collapse
|
26
|
Pullulan/Poly(Vinyl Alcohol) Composite Hydrogels for Adipose Tissue Engineering. MATERIALS 2019; 12:ma12193220. [PMID: 31581444 PMCID: PMC6804089 DOI: 10.3390/ma12193220] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 09/25/2019] [Accepted: 09/28/2019] [Indexed: 01/10/2023]
Abstract
Composite hydrogels based on pullulan (HP) and poly(vinyl alcohol) (PVA) were both prepared by simple chemical crosslinking with sodium trimethaphosphate (STMP) or by dual crosslinking (simultaneously chemical crosslinking with STMP and physical crosslinking by freeze-thaw technique). The resulting hydrogels and cryogels were designed for tissue engineering applications. PVA, with two different molecular weights (47,000 and 125,000 g/mol; PVA47 and PVA125, respectively), as well as different P/PVA weight ratios were tested. The physico-chemical characterization of the hydrogels was performed by FTIR spectroscopy and scanning electron microscopy (SEM). The swelling kinetics, dissolution behavior, and degradation profiles in simulated physiological conditions (phosphate buffer at pH 7.4) were investigated. Pullulan concentration and the crosslinking method had significant effects on the pore size, swelling ratio, and degradation profiles. Cryogels exhibit lower swelling capacities than the conventional hydrogels but have better stability against hydrolitic degradation. Biocompatibility of the hydrogels was also investigated by both MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) and LDH (lactaten dehydrogenase) assay. The MTT and LDH assays proved that dual crosslinked HP/PVA125 (75:25, w/w) scaffolds are more biocompatible and promote to a greater extent the adhesion and proliferation of L929 murine fibroblast cells than chemically crosslinked HP/PVA47 (50/50, w/w) scaffolds. Moreover, the HP/PVA125 cryogel had the best ability for the adipogenic differentiation of cells. The overall results demonstrated that the HP/PVA composite hydrogels or cryogels are suitable biomaterials for tissue engineering applications.
Collapse
|
27
|
Chun SY, Lim JO, Lee EH, Han MH, Ha YS, Lee JN, Kim BS, Park MJ, Yeo M, Jung B, Kwon TG. Preparation and Characterization of Human Adipose Tissue-Derived Extracellular Matrix, Growth Factors, and Stem Cells: A Concise Review. Tissue Eng Regen Med 2019; 16:385-393. [PMID: 31413942 DOI: 10.1007/s13770-019-00199-7] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 06/06/2019] [Indexed: 01/19/2023] Open
Abstract
Background Human adipose tissue is routinely discarded as medical waste. However, this tissue may have valuable clinical applications since methods have been devised to effectively isolate adipose-derived extracellular matrix (ECM), growth factors (GFs), and stem cells. In this review, we analyze the literature that devised these methods and then suggest an optimal method based on their characterization results. Methods Methods that we analyze in this article include: extraction of adipose tissue, decellularization, confirmation of decellularization, identification of residual active ingredients (ECM, GFs, and cells), removal of immunogens, and comparing structural/physiological/biochemical characteristics of active ingredients. Results Human adipose ECMs are composed of collagen type I-VII, laminin, fibronectin, elastin, and glycosaminoglycan (GAG). GFs immobilized in GAG include basic fibroblast growth factor (bFGF), transforming growth factor beta 1(TGF-b1), insulin like growth factor 1 (IGF-1), vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF), BMP4 (bone morphogenetic protein 4), nerve growth factor (NGF), hepatocyte growth factor (HGF), and epithermal growth factor (EGF). Stem cells in the stromal-vascular fraction display mesenchymal markers, self-renewal gene expression, and multi-differentiation potential. Conclusion Depending on the preparation method, the volume, biological activity, and physical properties of ECM, GFs, and adipose tissue-derived cells can vary. Thus, the optimal preparation method is dependent on the intended application of the adipose tissue-derived products.
Collapse
Affiliation(s)
- So Young Chun
- 1BioMedical Research Institute, Joint Institute for Regenerative Medicine, Kyungpook National University Hospital, Daegu, 41940 Republic of Korea
| | - Jeong Ok Lim
- 1BioMedical Research Institute, Joint Institute for Regenerative Medicine, Kyungpook National University Hospital, Daegu, 41940 Republic of Korea
| | - Eun Hye Lee
- 2Department of Pathology, School of Medicine, Kyungpook National University, Daegu, 41944 Republic of Korea
| | - Man-Hoon Han
- 2Department of Pathology, School of Medicine, Kyungpook National University, Daegu, 41944 Republic of Korea
| | - Yun-Sok Ha
- 3Department of Urology, School of Medicine, Kyungpook National University, Daegu, 41944 Republic of Korea
| | - Jun Nyung Lee
- 3Department of Urology, School of Medicine, Kyungpook National University, Daegu, 41944 Republic of Korea
| | - Bum Soo Kim
- 3Department of Urology, School of Medicine, Kyungpook National University, Daegu, 41944 Republic of Korea
| | - Min Jeong Park
- 4Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Cheombok-ro 80, Dong-gu, Daegu, 41061 Republic of Korea
| | - MyungGu Yeo
- 4Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Cheombok-ro 80, Dong-gu, Daegu, 41061 Republic of Korea
| | - Bongsu Jung
- 4Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Cheombok-ro 80, Dong-gu, Daegu, 41061 Republic of Korea
| | - Tae Gyun Kwon
- 3Department of Urology, School of Medicine, Kyungpook National University, Daegu, 41944 Republic of Korea
- 5Department of Urology, Kyungpook National University Chilgok Hospital, 807 Hoguk-ro, Buk-gu, Daegu, 41404 Republic of Korea
| |
Collapse
|
28
|
Contessi Negrini N, Tarsini P, Tanzi MC, Farè S. Chemically crosslinked gelatin hydrogels as scaffolding materials for adipose tissue engineering. J Appl Polym Sci 2018. [DOI: 10.1002/app.47104] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- N. Contessi Negrini
- Department of ChemistryMaterials and Chemical Engineering “G. Natta”, Politecnico di Milano Piazza Leonardo da Vinci 32, 20133, Milan Italy
- National Interuniversity Consortium of Materials Science and Technology (INSTM), via Giuseppe Giusti 9, 50121 Florence Italy
| | - P. Tarsini
- Department of ChemistryMaterials and Chemical Engineering “G. Natta”, Politecnico di Milano Piazza Leonardo da Vinci 32, 20133, Milan Italy
| | - M. C. Tanzi
- National Interuniversity Consortium of Materials Science and Technology (INSTM), via Giuseppe Giusti 9, 50121 Florence Italy
| | - S. Farè
- Department of ChemistryMaterials and Chemical Engineering “G. Natta”, Politecnico di Milano Piazza Leonardo da Vinci 32, 20133, Milan Italy
- National Interuniversity Consortium of Materials Science and Technology (INSTM), via Giuseppe Giusti 9, 50121 Florence Italy
| |
Collapse
|
29
|
Ghosh U, Ning S, Wang Y, Kong YL. Addressing Unmet Clinical Needs with 3D Printing Technologies. Adv Healthc Mater 2018; 7:e1800417. [PMID: 30004185 DOI: 10.1002/adhm.201800417] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 05/29/2018] [Indexed: 01/04/2023]
Abstract
Recent advances in 3D printing have enabled the creation of novel 3D constructs and devices with an unprecedented level of complexity, properties, and functionalities. In contrast to manufacturing techniques developed for mass production, 3D printing encompasses a broad class of fabrication technologies that can enable 1) the creation of highly customized and optimized 3D physical architectures from digital designs; 2) the synergistic integration of properties and functionalities of distinct classes of materials to create novel hybrid devices; and 3) a biocompatible fabrication approach that facilitates the creation and cointegration of biological constructs and systems. This progress report describes how these capabilities can potentially address a myriad of unmet clinical needs. First, the creation of 3D-printed prosthetics to regain lost functionalities by providing structural support for skeletal and tubular organs is highlighted. Second, novel drug delivery strategies aided by 3D-printed devices are described. Third, the advancement of medical research heralded by 3D-printed tissue/organ-on-chips systems is discussed. Fourth, the developments of 3D-printed tissue and organ regeneration are explored. Finally, the potential for seamless integration of engineered organs with active devices by leveraging the versatility of multimaterial 3D printing is envisioned.
Collapse
Affiliation(s)
- Udayan Ghosh
- Department of Mechanical Engineering; University of Utah; 1495 E 100 S (1550 MEK) Salt Lake City UT 84112 USA
| | - Shen Ning
- Boston University School of Medicine; Boston University; 72 E Concord St Boston MA 02118 USA
| | - Yuzhu Wang
- Department of Mechanical Engineering; University of Utah; 1495 E 100 S (1550 MEK) Salt Lake City UT 84112 USA
| | - Yong Lin Kong
- Department of Mechanical Engineering; University of Utah; 1495 E 100 S (1550 MEK) Salt Lake City UT 84112 USA
| |
Collapse
|
30
|
Generation of Gellan Gum-Based Adipose-Like Microtissues. Bioengineering (Basel) 2018; 5:bioengineering5030052. [PMID: 29954069 PMCID: PMC6163196 DOI: 10.3390/bioengineering5030052] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/12/2018] [Accepted: 06/21/2018] [Indexed: 11/16/2022] Open
Abstract
Adipose tissue is involved in many physiological processes. Therefore, the need for adipose tissue-like analogues either for soft tissue reconstruction or as in vitro testing platforms is undeniable. In this work, we explored the natural features of gellan gum (GG) to recreate injectable stable adipose-like microtissues. GG hydrogel particles with different percentages of polymer (0.5%, 0.75%, 1.25%) were developed and the effect of obtained mechanical properties over the ability of hASCs to differentiate towards the adipogenic lineage was evaluated based on the expression of the early (PPARγ) and late (FABP4) adipogenic markers, and on lipids formation and accumulation. Constructs were cultured in adipogenic induction medium up to 21 days or for six days in induction plus nine days in maintenance media. Overall, no significant differences were observed in terms of hASCs adipogenic differentiation within the range of Young’s moduli between 2.7 and 12.9 kPa. The long-term (up to six weeks) stability of the developed constructs supported its application in soft tissue reconstruction. Moreover, their ability to function as adipose-like microtissue models for drug screening was demonstrated by confirming its sensitivity to TNFα and ROCK inhibitor, respectively involved in the repression and induction of the adipogenic differentiation.
Collapse
|
31
|
Whitehead AK, Barnett HH, Caldorera-Moore ME, Newman JJ. Poly (ethylene glycol) hydrogel elasticity influences human mesenchymal stem cell behavior. Regen Biomater 2018; 5:167-175. [PMID: 29942649 PMCID: PMC6007362 DOI: 10.1093/rb/rby008] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 04/03/2018] [Accepted: 04/04/2018] [Indexed: 12/17/2022] Open
Abstract
Coordinated investigations into the interactions between biologically mimicking (biomimetic) material constructs and stem cells advance the potential for the regeneration and possible direct replacement of diseased cells and tissues. Any clinically relevant therapies will require the development and optimization of methods that mass produce fully functional cells and tissues. Despite advances in the design and synthesis of biomaterial scaffolds, one of the biggest obstacles facing tissue engineering is understanding how specific extracellular cues produced by biomaterial scaffolds influence the proliferation and differentiation of various cell sources. Matrix elasticity is one such tailorable property of synthetic scaffolds that is known to differ between tissues. Here, we investigate the interactions between an elastically tailorable polyethylene glycol (PEG)-based hydrogel platform and human bone marrow-derived mesenchymal stem cells (hMSCs). For these studies, two different hydrogel compositions with elastic moduli in the ranges of 50-60 kPa and 8-10 kPa were implemented. Our findings demonstrate that the different elasticities in this platform can produce changes in hMSC morphology and proliferation, indicating that the platform can be implemented to produce changes in hMSC behavior and cell state for a broad range of tissue engineering and regenerative applications. Furthermore, we show that the platform's different elasticities influence stem cell differentiation potential, particularly when promoting stem cell differentiation toward cell types from tissues with stiffer elasticity. These findings add to the evolving and expanding library of information on stem cell-biomaterial interactions and opens the door for continued exploration into PEG-based hydrogel scaffolds for tissue engineering and regenerative medicine applications.
Collapse
|
32
|
Mahoney CM, Imbarlina C, Yates CC, Marra KG. Current Therapeutic Strategies for Adipose Tissue Defects/Repair Using Engineered Biomaterials and Biomolecule Formulations. Front Pharmacol 2018; 9:507. [PMID: 29867506 PMCID: PMC5966552 DOI: 10.3389/fphar.2018.00507] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 04/27/2018] [Indexed: 01/01/2023] Open
Abstract
Tissue engineered scaffolds for adipose restoration/repair has significantly evolved in recent years. Patients requiring soft tissue reconstruction, caused by defects or pathology, require biomaterials that will restore void volume with new functional tissue. The gold standard of autologous fat grafting (AFG) is not a reliable option. This review focuses on the latest therapeutic strategies for the treatment of adipose tissue defects using biomolecule formulations and delivery, and specifically engineered biomaterials. Additionally, the clinical need for reliable off-the-shelf therapies, animal models, and challenges facing current technologies are discussed.
Collapse
Affiliation(s)
- Christopher M Mahoney
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
| | - Cayla Imbarlina
- Department of Biology, Carlow University, Pittsburgh, PA, United States
| | - Cecelia C Yates
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Health Promotion and Development, School of Nursing, University of Pittsburgh, Pittsburgh, PA, United States.,McGowan Institute for Regenerative Medicine, Pittsburgh, PA, United States
| | - Kacey G Marra
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States.,McGowan Institute for Regenerative Medicine, Pittsburgh, PA, United States.,Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
33
|
Tanataweethum N, Zelaya A, Yang F, Cohen RN, Brey EM, Bhushan A. Establishment and characterization of a primary murine adipose tissue-chip. Biotechnol Bioeng 2018; 115:1979-1987. [PMID: 29689639 DOI: 10.1002/bit.26711] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 04/02/2018] [Accepted: 04/09/2018] [Indexed: 12/11/2022]
Abstract
Better experimental models are needed to enhance our understanding of metabolic regulation which is seen in obesity and metabolic disorders, such as type 2 diabetes. In vitro models based on microfluidics enable physiological representations of tissues with several advantages over conventional culture systems, such as perfused flow to better mimic the physiological environment. Although cell lines such as 3T3-L1 have been incorporated in microfluidic devices, murine primary preadipocytes have not been differentiated and maintained for long-term monitoring in these culture systems. We describe the differentiation of these cells into white adipose depots on a perfused microfluidic chip. We compare the effects of shear flow on these cells, and show with a direct comparison of high/low shear conditions that direct shear is detrimental to the viability of preadipocytes. We further develop a dual-chamber microfluidic chip that enables perfusion while at the same time protects the cells from direct fluidic shear. We show that the dual-layer microfluidic device enables long-term culture of cells and allows stimulation of cells through perfusion-we can culture, differentiate, and maintain the differentiated adipose tissue for over multiple weeks in the device. Both triglycerides and lipolytic glycerol production increased significantly by several folds during differentiation. After successful differentiation, the adipocytes had upregulated expression of leptin and adiponectin, which are important makers of the final stage of adipogenic differentiation. In conclusion, the dual-layer microfluidic device incorporated with primary adipocytes improves the understanding of adipose differentiation under dynamic conditions and is positioned to serve as a disease model for studying obesity and other metabolic disorders.
Collapse
Affiliation(s)
- Nida Tanataweethum
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois
| | - Adelyn Zelaya
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois
| | - Feipeng Yang
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois
| | - Ronald N Cohen
- Section of Endocrinology, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Eric M Brey
- Department of Biomedical Engineering, University of Texas at San Antonio, San Antonio, Texas
| | - Abhinav Bhushan
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois
| |
Collapse
|
34
|
Zou J, Wang W, Neffe AT, Xu X, Li Z, Deng Z, Sun X, Ma N, Lendlein A. Adipogenic differentiation of human adipose derived mesenchymal stem cells in 3D architectured gelatin based hydrogels (ArcGel). Clin Hemorheol Microcirc 2018; 67:297-307. [PMID: 28885199 DOI: 10.3233/ch-179210] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Polymeric matrices mimicking multiple functions of the ECM are expected to enable a material induced regeneration of tissues. Here, we investigated the adipogenic differentiation of human adipose derived mesenchymal stem cells (hADSCs) in a 3D architectured gelatin based hydrogel (ArcGel) prepared from gelatin and L-lysine diisocyanate ethyl ester (LDI) in an one-step process, in which the formation of an open porous morphology and the chemical network formation were integrated. The ArcGel was designed to support adipose tissue regeneration with its 3D porous structure, high cell biocompatibility, and mechanical properties compatible with human subcutaneous adipose tissue. The ArcGel could support initial cell adhesion and survival of hADSCs. Under static culture condition, the cells could migrate into the inner part of the scaffold with a depth of 840±120 μm after 4 days, and distributed in the whole scaffold (2 mm in thickness) within 14 days. The cells proliferated in the scaffold and the fold increase of cell number after 7 days of culture was 2.55±0.08. The apoptotic rate of hADSCs in the scaffold was similar to that of cells maintained on tissue culture plates. When cultured in adipogenic induction medium, the hADSCs in the scaffold differentiated into adipocytes with a high efficiency (93±1%). Conclusively, this gelatin based 3D scaffold presented high cell compatibility for hADSC cultivation and differentiation, which could serve as a potential implant material in clinical applications for adipose tissue reparation and regeneration.
Collapse
Affiliation(s)
- Jie Zou
- Institute of Biomaterial Science and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany.,Institute of Chemistry and Biochemistry, Freie Universität, Berlin, Germany
| | - Weiwei Wang
- Institute of Biomaterial Science and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| | - Axel T Neffe
- Institute of Biomaterial Science and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany.,Helmholtz Virtual Institute "Multifunctional Biomaterials in Medicine", Teltow, Germany
| | - Xun Xu
- Institute of Biomaterial Science and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany.,Institute of Chemistry and Biochemistry, Freie Universität, Berlin, Germany
| | - Zhengdong Li
- Institute of Biomaterial Science and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany.,Institute of Chemistry and Biochemistry, Freie Universität, Berlin, Germany
| | - Zijun Deng
- Institute of Biomaterial Science and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany.,Institute of Chemistry and Biochemistry, Freie Universität, Berlin, Germany
| | - Xianlei Sun
- Institute of Biomaterial Science and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany.,Institute of Biochemistry and Biology, Universität Potsdam, Potsdam, Germany
| | - Nan Ma
- Institute of Biomaterial Science and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany.,Institute of Chemistry and Biochemistry, Freie Universität, Berlin, Germany.,Helmholtz Virtual Institute "Multifunctional Biomaterials in Medicine", Teltow, Germany
| | - Andreas Lendlein
- Institute of Biomaterial Science and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany.,Institute of Chemistry and Biochemistry, Freie Universität, Berlin, Germany.,Helmholtz Virtual Institute "Multifunctional Biomaterials in Medicine", Teltow, Germany.,Institute of Biochemistry and Biology, Universität Potsdam, Potsdam, Germany
| |
Collapse
|
35
|
Kuss M, Kim J, Qi D, Wu S, Lei Y, Chung S, Duan B. Effects of tunable, 3D-bioprinted hydrogels on human brown adipocyte behavior and metabolic function. Acta Biomater 2018; 71:486-495. [PMID: 29555462 DOI: 10.1016/j.actbio.2018.03.021] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 03/07/2018] [Accepted: 03/08/2018] [Indexed: 12/16/2022]
Abstract
Obesity and its related health complications cause billions of dollars in healthcare costs annually in the United States, and there are yet to be safe and long-lasting anti-obesity approaches. Using brown adipose tissue (BAT) is a promising approach, as it uses fats for energy expenditure. However, the effect of the microenvironment on human thermogenic brown adipogenesis and how to generate clinically relevant sized and functioning BAT are still unknown. In our current study, we evaluated the effects of endothelial growth medium exposure on brown adipogenesis of human brown adipose progenitors (BAP). We found that pre-exposing BAP to angiogenic factors promoted brown adipogenic differentiation and metabolic activity. We further 3D bioprinted brown and white adipose progenitors within hydrogel-based bioink with controllable physicochemical properties and evaluated the cell responses in 3D bioprinted environments. We used soft, stiff, and stiff-porous constructs to encapsulate the cells. All three types had high cell viability and allowed for varying levels of function for both white and brown adipocytes. We found that the soft hydrogel constructs promoted white adipogenesis, while the stiff-porous hydrogel constructs improved both white and brown adipogenesis and were the optimal condition for promoting brown adipogenesis. Consistently, stiff-porous hydrogel constructs showed higher metabolic activities than stiff hydrogel constructs, as assessed by 2-deoxy glucose uptake (2-DOG) and oxygen consumption rate (OCR). These findings show that the physicochemical environments affect the brown adipogenesis and metabolic function, and further tuning will be able to optimize their functions. Our results also demonstrate that 3D bioprinting of brown adipose tissues with clinically relevant size and metabolic activity has the potential to be a viable option in the treatment of obesity and type 2 diabetes. STATEMENT OF SIGNIFICANCE One promising strategy for the treatment or prevention of obesity-mediated health complications is augmenting brown adipose tissues (BAT), which is a specialized fat that actively dissipate energy in the form of heat and maintain energy balance. In this study, we determined how pre-exposing human brown adipose progenitors (BAP) to angiogenic factors in 2D and how bioprinted microenvironments in 3D affected brown adipogenic differentiation and metabolic activity. We demonstrated that white and brown adipogenesis, and thermogenesis were regulated by tuning the bioprintable matrix stiffness and construct structure. This study not only unveils the interaction between BAP and 3D physiological microenvironments, but also presents a novel tissue engineered strategy to manage obesity and other related metabolic disorders.
Collapse
|
36
|
Strategic Design and Fabrication of Biomimetic 3D Scaffolds: Unique Architectures of Extracellular Matrices for Enhanced Adipogenesis and Soft Tissue Reconstruction. Sci Rep 2018; 8:5696. [PMID: 29632328 PMCID: PMC5890269 DOI: 10.1038/s41598-018-23966-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 03/23/2018] [Indexed: 01/16/2023] Open
Abstract
The higher rate of soft tissue impairment due to lumpectomy or other trauma greatly requires the restoration of the irreversibly lost subcutaneous adipose tissues. The nanofibers fabricated by conventional electrospinning provide only a superficial porous structure due to its sheet like 2D structure and thereby hinder the cell infiltration and differentiation throughout the scaffolds. Thus we developed a novel electrospun 3D membrane using the zwitterionic poly (carboxybetaine-co-methyl methacrylate) co-polymer (CMMA) through electrostatic repulsion based electrospinning for soft tissue engineering. The inherent charges in the CMMA will aid the nanofiber to directly transform into a semiconductor and thereby transfer the immense static electricity from the grounded collector and will impart greater fluffiness to the scaffolds. The results suggest that the fabricated 3D nanofiber (CMMA 3NF) scaffolds possess nanofibers with larger inter connected pores and less dense structure compared to the conventional 2D scaffolds. The CMMA 3NF exhibits significant cues of soft tissue engineering such as enhanced biocompatibility as well as the faster regeneration of cells. Moreover the fabricated 3D scaffolds greatly assist the cells to develop into its stereoscopic topographies with an enhanced adipogenic property.
Collapse
|
37
|
Kuroda M, Saito Y, Aso M, Yokote K. A Novel Approach to the Treatment of Plasma Protein Deficiency: Ex Vivo-Manipulated Adipocytes for Sustained Secretion of Therapeutic Proteins. Chem Pharm Bull (Tokyo) 2018; 66:217-224. [PMID: 29491255 DOI: 10.1248/cpb.c17-00786] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Despite the critical need for lifelong treatment of inherited and genetic diseases, there are no developmental efforts for most such diseases due to their rarity. Recent progress in gene therapy, including the approvals of two products (Glybera and Strimvelis) that may provide patients with sustained effects, has shed light on the development of gene therapy products. Most gene therapy products are based on either adeno-associated virus-mediated in vivo gene transfer to target tissues or administration of ex vivo gene-transduced hematopoietic cells. In such circumstances, there is room for different approaches to provide clinicians with other therapeutic options through a variety of principles based on studies not only to gain an understanding of the pathological mechanisms of diseases, but also to understand the physiological functions of target tissues and cells. In this review, we summarize recent progress in gene therapy-mediated enzyme replacement and introduce a different approach using adipocytes to enable lifelong treatment for intractable plasma protein deficiencies.
Collapse
Affiliation(s)
- Masayuki Kuroda
- Center for Advanced Medicine, Chiba University Hospital, Chiba University
| | | | | | - Koutaro Yokote
- Clinical Cell Biology and Medicine, Chiba University Graduate School of Medicine, Chiba University
| |
Collapse
|
38
|
Griessl M, Buchberger AM, Regn S, Kreutzer K, Storck K. Uncultivated stromal vascular fraction is equivalent to adipose-derived stem and stromal cells on porous polyurethrane scaffolds forming adipose tissue in vivo. Laryngoscope 2018; 128:E206-E213. [PMID: 29446455 DOI: 10.1002/lary.27124] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 01/09/2018] [Accepted: 01/16/2018] [Indexed: 12/21/2022]
Abstract
OBJECTIVES/HYPOTHESIS To find an alternative approach to contemporary techniques in tissue augmentation and reconstruction, tissue engineering strategies aim to involve adipose-derived stem and stromal cells (ASCs) harboring a strong differentiation potential into various tissue types such as bone, cartilage, and fat. STUDY DESIGN Animal research. METHODS The stromal vascular fraction (SVF) was used directly as a cell source to provide a potential alternative to contemporary ASC-based adipose tissue engineering. Seeded in TissuCol fibrin, we applied ASCs or SVF cells to porous, degradable polyurethane (PU) scaffolds. RESULTS We successfully demonstrated the in vivo generation of volume-stable, well-vascularized PU-based constructs containing host-derived mature fat pads. Seeded human stem cells served as modulators of host-cell migration rather than differentiating themselves. We further demonstrated that preliminary culture of SVF cells was not necessary. CONCLUSIONS Our results bring adipose tissue engineering, together with automated processing devices, closer to clinical applicability. The time-consuming and cost-intensive culture and induction of the ASCs is not necessary. LEVEL OF EVIDENCE NA. Laryngoscope, 128:E206-E213, 2018.
Collapse
Affiliation(s)
- Michael Griessl
- Department of ENT, Head and Neck Surgery, Technical University of Munich, Munich, Germany
| | - Anna-Maria Buchberger
- Department of ENT, Head and Neck Surgery, Technical University of Munich, Munich, Germany
| | - Sybille Regn
- Department of ENT, Head and Neck Surgery, Technical University of Munich, Munich, Germany
| | - Kilian Kreutzer
- Department of Maxillofacial Surgery, University Clinic of the Charité Berlin, Berlin, Germany
| | - Katharina Storck
- Department of ENT, Head and Neck Surgery, Technical University of Munich, Munich, Germany
| |
Collapse
|
39
|
Mahoney CM, Kelmindi-Doko A, Snowden MJ, Peter Rubin J, Marra KG. Adipose derived delivery vehicle for encapsulated adipogenic factors. Acta Biomater 2017; 58:26-33. [PMID: 28532902 DOI: 10.1016/j.actbio.2017.05.046] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 05/17/2017] [Accepted: 05/18/2017] [Indexed: 10/19/2022]
Abstract
Hydrogels derived from adipose tissue extracellular matrix (AdECM) have shown potential in the ability to generate new adipose tissue in vivo. To further enhance adipogenesis, a composite adipose derived delivery system (CADDS) containing single- and double-walled dexamethasone encapsulated microspheres (SW and DW Dex MS) has been developed. Previously, our laboratory has published the use of Dex MS as an additive to enhance adipogenesis and angiogenesis in adipose tissue grafts. In the current work, AdECM and CADDS are extensively characterized, in addition to conducting in vitro cell culture analysis. Study results indicate the AdECM used for the CADDS has minimal cellular and lipid content allowing for gelation of its collagen structure under physiological conditions. Adipose-derived stem cell (ASC) culture studies confirmed biocompatibility with the CADDS, and adipogenesis was increased in experimental groups containing the hydrogel scaffold. In vitro studies of AdECM hydrogel containing microspheres demonstrated a controlled release of dexamethasone from SW and DW formulations. The delivery of Dex MS via an injectable hydrogel scaffold combines two biologically responsive components to develop a minimally, invasive, off-the-shelf biomaterial for adipose tissue engineering. STATEMENT OF SIGNIFICANCE Scientists and doctors have yet to develop an off-the-shelf product for patients with soft tissue defects. Recently, the use of adipose derived extracellular matrix (adECM) to generate new adipose tissue in vivo has shown great promise but individually, adECM still has limitations in terms of volume and consistency. The current work introduces a novel composite off-the-shelf construct comprised of an adECM-based hydrogel and dexamethasone encapsulated microspheres (Dex MS). The hydrogel construct serves not only as an injectable protein-rich scaffold but also a delivery system for the Dex MS for non-invasive application to the defect site. The methods and results presented are a progressive step forward in the field of adipose tissue engineering.
Collapse
|
40
|
Tang Q, Chen C, Wang X, Li W, Zhang Y, Wang M, Jing W, Wang H, Guo W, Tian W. Botulinum toxin A improves adipose tissue engraftment by promoting cell proliferation, adipogenesis and angiogenesis. Int J Mol Med 2017; 40:713-720. [PMID: 28731141 PMCID: PMC5547983 DOI: 10.3892/ijmm.2017.3073] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 07/06/2017] [Indexed: 02/05/2023] Open
Abstract
Adipose tissue engraftment has become a well-established therapy in plastic and reconstructive surgery used to restore age-related or injury-related soft tissue loss. However, the unpredictable absorption rates limit its further application. Some clinicians have noted that more optimal aesthetic results are achieved when botulinum toxin A (BoNTA) is applied prior to adipose tissue grafting. In the present study, we transplanted allogeneic adipose tissue treated with or without BoNTA in SD rats in vivo. We subsequently evaluated the survival rate (weight, volume, apoptosis and cellular integrity) and revascularization of the adipose tissue. The results revealed that BoNTA improved the long-term weight and volume retention of the graft, and preserved cellular integrity. BoNTA significantly increased the expression levels of CD31 and vascular endothelial growth factor (VEGF), suggesting enhanced vasodilation and endothelial cell proliferation. In vitro, adipose-derived stem cells (ASCs) were isolated, identified and induced to proliferate and differentiate with or without BoNTA. Furthermore, to evaluate the proliferative, adipogenic and angiogenic ability of the ASCs, CCK-8 assay and Oil Red O staining were conducted. Gene and protein expression levels were analyzed by RT-qPCR and western blot analysis. The results revealed that 8×10−2 U/ml BoNTA as the optimal dose increased ASC proliferation and adipogenic differentiation capacity, as well as the expression level of the key cytokine of angiogenesis. On the whole, our findings indicate that BoNTA improves adipose tissue engraftment and promotes ASC regeneration, which could benefit future clinical applications.
Collapse
Affiliation(s)
- Qi Tang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| | - Chang Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| | - Xiaqi Wang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| | - Wei Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| | - Yan Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| | - Muyao Wang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| | - Wei Jing
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| | - Hang Wang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| | - Weihua Guo
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| | - Weidong Tian
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| |
Collapse
|
41
|
Self-Assembling RADA16-I Peptide Hydrogel Scaffold Loaded with Tamoxifen for Breast Reconstruction. BIOMED RESEARCH INTERNATIONAL 2017; 2017:3656193. [PMID: 28691024 PMCID: PMC5485292 DOI: 10.1155/2017/3656193] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 04/11/2017] [Accepted: 05/03/2017] [Indexed: 12/20/2022]
Abstract
More and more breast cancer patients prefer autologous fat tissue transfer following lumpectomy to maintain perfect female characteristics. However, the outcome was not satisfactory due to the transplanted fat absorption. In this study, we prepared two RADA16-I peptide scaffolds with and without tamoxifen. Both scaffolds were transparent, porous, and hemisphere-shaped. The hADSCs isolated from liposuction were attached to the scaffold. The growth inhibition of the hADSCs induced by TAM in 2-demensional (2D) culture was higher than that in TAM-loaded hydrogel scaffold 3D culture (P < 0.05); however, the same outcomes were not observed in MCF-7 cells. Correspondingly, the apoptosis of the hADSCs induced by TAM was significantly increased in 2D culture compared to that in scaffold 3D culture (P < 0.05). Yet the outcomes of the aoptosis in MCF-7 were contrary. Apoptosis-related protein Bcl-2 was involved in the process. In vivo experiments showed that both scaffolds formed a round mass after subcutaneous implantation and it retained its shape after being pressed slightly. The implantation had no effect on the weight and activity of the animals. The results suggested that TAM-loaded RADA16-I hydrogel scaffolds both provide support for hADSCs cells attachment/proliferation and retain cytotoxic effect on MCF-7 cells, which might be a promising therapeutic breast tissue following lumpectomy.
Collapse
|
42
|
Visscher LE, Cheng M, Chhaya M, Hintz ML, Schantz JT, Tran P, Ung O, Wong C, Hutmacher DW. Breast Augmentation and Reconstruction from a Regenerative Medicine Point of View: State of the Art and Future Perspectives. TISSUE ENGINEERING PART B-REVIEWS 2017; 23:281-293. [PMID: 28437235 DOI: 10.1089/ten.teb.2016.0303] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Breast reconstruction and augmentation are very common procedures, yet the prevailing current methods utilize silicone implants that may have significant local complications requiring reoperation. Lipofillling is increasingly used to contour and is considered safe, however, its utility is limited by significant volume loss. A new approach could offer an alternative and increase the scope of patient choice. A small number of teams around the world are investigating a breast tissue engineering (TE) paradigm. Conventional breast TE concepts are based on seeding a scaffold with the patients' own stem cells. However, the clinical viability of many of these approaches is limited by their costs in relevant volumes. In this article the state of the art of tissue-engineered breast reconstruction is reviewed and future perspectives are presented and discussed.
Collapse
Affiliation(s)
- Luke E Visscher
- 1 Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology , Australia .,2 School of Medicine, University of Queensland , Brisbane, Australia
| | - Matthew Cheng
- 1 Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology , Australia .,3 Plastic and Reconstructive Surgery Unit, Princess Alexandra Hospital , Woolloongabba, Australia
| | - Mohit Chhaya
- 1 Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology , Australia
| | - Madeline L Hintz
- 1 Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology , Australia
| | - Jan-Thorsten Schantz
- 4 Department of Plastic and Hand Surgery, Klinikum rechts der Isar, Technische Universität München , München, Germany
| | - Phong Tran
- 1 Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology , Australia
| | - Owen Ung
- 2 School of Medicine, University of Queensland , Brisbane, Australia .,5 Surg 1, Breast Endocrine Unit, Royal Brisbane and Women's Hospital , Herston, Brisbane, Australia
| | - Clement Wong
- 2 School of Medicine, University of Queensland , Brisbane, Australia .,5 Surg 1, Breast Endocrine Unit, Royal Brisbane and Women's Hospital , Herston, Brisbane, Australia
| | - Dietmar W Hutmacher
- 1 Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology , Australia .,6 ARC Centre in Additive Biomanufacturing, Queensland University of Technology, Brisbane, Australia
| |
Collapse
|
43
|
Turner PA, Gurumurthy B, Bailey JL, Elks CM, Janorkar AV. Adipogenic Differentiation of Human Adipose-Derived Stem Cells Grown as Spheroids. Process Biochem 2017; 59:312-320. [PMID: 28966553 DOI: 10.1016/j.procbio.2017.02.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Understanding the process of adipogenesis is critical if suitable therapeutics for obesity and related metabolic diseases are to be found. The current study presents proof of feasibility of creating a 3-D spheroid model using human adipose-derived stem cells (hASCs) and their subsequent adipogenic differentiation. hASC spheroids were formed atop an elastin-like polypeptide-polyethyleneimine (ELP-PEI) surface and differentiated using an adipogenic cocktail. Spheroids were matured in the presence of dietary fatty acids (linoleic or oleic acid) and evaluated based on functional markers including intracellular protein, CD36 expression, triglyceride accumulation, and PPAR-γ gene expression. Spheroid size was found to increase as the hASCs matured in the adipocyte maintenance medium, though the fatty acid treatment generally resulted in smaller spheroids compared to control. A stable protein content over the 10-day maturation period indicated contact-inhibited proliferation as well as minimal loss of spheroids during culture. Spheroids treated with fatty acids showed greater amounts of intracellular triglyceride content and greater expression of the key adipogenic gene, PPAR-γ. We also demonstrated that 3-D spheroids outperformed 2-D monolayer cultures in adipogenesis. We then compared the adipogenesis of hASC spheroids to that in 3T3-L1 spheroids and found that the triglyceride accumulation was less profound in hASC spheroids than that in 3T3-L1 adipocytes, correlated with smaller average spheroids, suggesting a relatively slower differentiation process. Taken together, we have shown the feasibility of adipogenic differentiation of patient-derived hASC spheroids, which with further development, may help elucidate key features in the adipogenesis process.
Collapse
Affiliation(s)
- Paul A Turner
- Department of Biomedical Materials Science, School of Dentistry, University of Mississippi Medical Center, 2500 N State Street, Jackson, MS
| | - Bhuvaneswari Gurumurthy
- Department of Biomedical Materials Science, School of Dentistry, University of Mississippi Medical Center, 2500 N State Street, Jackson, MS
| | - Jennifer L Bailey
- Matrix Biology Laboratory, Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA
| | - Carrie M Elks
- Matrix Biology Laboratory, Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA
| | - Amol V Janorkar
- Department of Biomedical Materials Science, School of Dentistry, University of Mississippi Medical Center, 2500 N State Street, Jackson, MS
| |
Collapse
|
44
|
Rodriguez MJ, Brown J, Giordano J, Lin SJ, Omenetto FG, Kaplan DL. Silk based bioinks for soft tissue reconstruction using 3-dimensional (3D) printing with in vitro and in vivo assessments. Biomaterials 2017; 117:105-115. [PMID: 27940389 PMCID: PMC5180454 DOI: 10.1016/j.biomaterials.2016.11.046] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Revised: 11/15/2016] [Accepted: 11/24/2016] [Indexed: 02/07/2023]
Abstract
In the field of soft tissue reconstruction, custom implants could address the need for materials that can fill complex geometries. Our aim was to develop a material system with optimal rheology for material extrusion, that can be processed in physiological and non-toxic conditions and provide structural support for soft tissue reconstruction. To meet this need we developed silk based bioinks using gelatin as a bulking agent and glycerol as a non-toxic additive to induce physical crosslinking. We developed these inks optimizing printing efficacy and resolution for patient-specific geometries that can be used for soft tissue reconstruction. We demonstrated in vitro that the material was stable under physiological conditions and could be tuned to match soft tissue mechanical properties. We demonstrated in vivo that the material was biocompatible and could be tuned to maintain shape and volume up to three months while promoting cellular infiltration and tissue integration.
Collapse
Affiliation(s)
- María J Rodriguez
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | - Joseph Brown
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | - Jodie Giordano
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | - Samuel J Lin
- Division of Plastic and Reconstructive Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | | | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA.
| |
Collapse
|
45
|
Venugopal B, Fernandez FB, Harikrishnan VS, John A. Post implantation fate of adipogenic induced mesenchymal stem cells on Type I collagen scaffold in a rat model. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2017; 28:28. [PMID: 28108956 DOI: 10.1007/s10856-016-5838-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 12/24/2016] [Indexed: 06/06/2023]
Abstract
Regenerative medicine via its application in soft tissue reconstruction through novel methods in adipose tissue engineering (ATE) has gained remarkable attention and investment despite simultaneous reports on clinical incidence of graft resorption and impaired vascularization. The underlying malaise here once identified may play a critical role in optimizing implant function. Our work attempts to determine the fate of donor cells and the implant in recipient micro environment using adipose-derived mesenchymal stem cells (ASCs) on a type I collagen sponge, an established scaffold for ATE. Cell components within the construct were identified 21 days post implantation to delineate cell survival, proliferation & terminal roles in vivo. ASC's are multipotent, while collagen type I is a natural extra cellular matrix component. Commercially available bovine type I collagen was characterized for its physiochemical properties and cyto-compatibility. Nile red staining of induced ASCs identified red globular structures in cell cytoplasm indicating oil droplet accumulation. Similarly, in vivo implantation of the cell seeded collagen construct in rat model for 21 days in the dorsal muscle, showed genesis of chicken wire network of fat-like cells, which was demonstrated histologically using a variety of staining techniques. Furthermore, fluorescent in situ hybridization (FISH) technique established the efficiency of transplantation wherein the male donor cells with labeled Y chromosome was identified 21 days post implantation from female rat model. Retrieved samples at 21 days indicated adipogenesis in situ, with donor cells highlighted via FISH. The study provides an insight to stem cells in ATE from genesis to functionalization.
Collapse
Affiliation(s)
- Balu Venugopal
- Division of Tissue Culture, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, India
| | - Francis B Fernandez
- Transmission Electron Microscopy Lab, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, India
| | - V S Harikrishnan
- Division of Laboratory Animal Science, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, India
| | - Annie John
- Transmission Electron Microscopy Lab, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, India.
| |
Collapse
|
46
|
Pyrintegrin Induces Soft Tissue Formation by Transplanted or Endogenous Cells. Sci Rep 2017; 7:36402. [PMID: 28128224 PMCID: PMC5269584 DOI: 10.1038/srep36402] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 09/16/2016] [Indexed: 12/11/2022] Open
Abstract
Focal adipose deficiency, such as lipoatrophy, lumpectomy or facial trauma, is a formidable challenge in reconstructive medicine, and yet scarcely investigated in experimental studies. Here, we report that Pyrintegrin (Ptn), a 2,4-disubstituted pyrimidine known to promote embryonic stem cells survival, is robustly adipogenic and induces postnatal adipose tissue formation in vivo of transplanted adipose stem/progenitor cells (ASCs) and recruited endogenous cells. In vitro, Ptn stimulated human adipose tissue derived ASCs to differentiate into lipid-laden adipocytes by upregulating peroxisome proliferator-activated receptor (PPARγ) and CCAAT/enhancer-binding protein-α (C/EBPα), with differentiated cells increasingly secreting adiponectin, leptin, glycerol and total triglycerides. Ptn-primed human ASCs seeded in 3D-bioprinted biomaterial scaffolds yielded newly formed adipose tissue that expressed human PPARγ, when transplanted into the dorsum of athymic mice. Remarkably, Ptn-adsorbed 3D scaffolds implanted in the inguinal fat pad had enhanced adipose tissue formation, suggesting Ptn’s ability to induce in situ adipogenesis of endogenous cells. Ptn promoted adipogenesis by upregulating PPARγ and C/EBPα not only in adipogenesis induction medium, but also in chemically defined medium specifically for osteogenesis, and concurrently attenuated Runx2 and Osx via BMP-mediated SMAD1/5 phosphorylation. These findings suggest Ptn’s novel role as an adipogenesis inducer with a therapeutic potential in soft tissue reconstruction and augmentation.
Collapse
|
47
|
Kelmendi-Doko A, Rubin JP, Klett K, Mahoney C, Wang S, Marra KG. Controlled dexamethasone delivery via double-walled microspheres to enhance long-term adipose tissue retention. J Tissue Eng 2017; 8:2041731417735402. [PMID: 29051810 PMCID: PMC5638157 DOI: 10.1177/2041731417735402] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 09/13/2017] [Indexed: 11/26/2022] Open
Abstract
Current materials used for adipose tissue reconstruction have critical shortcomings such as suboptimal volume retention, donor-site morbidity, and poor biocompatibility. The aim of this study was to examine a controlled delivery system of dexamethasone to generate stable adipose tissue when mixed with disaggregated human fat in an athymic mouse model for 6 months. The hypothesis that the continued release of dexamethasone from polymeric microspheres would enhance both adipogenesis and angiogenesis more significantly when compared to the single-walled microsphere model, resulting in long-term adipose volume retention, was tested. Dexamethasone was encapsulated within single-walled poly(lactic-co-glycolic acid) microspheres (Dex SW MS) and compared to dexamethasone encapsulated in a poly(lactic-co-glycolic acid) core surrounded by a shell of poly-l-lactide. The double-walled polymer microsphere system in the second model was developed to create a more sustainable drug delivery process. Dexamethasone-loaded poly(lactic-co-glycolic acid) microspheres (Dex SW MS) and dexamethasone-loaded poly(lactic-co-glycolic acid)/poly-l-lactide double-walled microspheres (Dex DW MS) were prepared using single and double emulsion/solvent techniques. In vitro release kinetics were determined. Two doses of each type of microsphere were examined; 50 and 27 mg of Dex MS and Dex DW MS were mixed with 0.3 mL of human lipoaspirate. Additionally, 50 mg of empty MS and lipoaspirate-only controls were examined. Samples were analyzed grossly and histologically after 6 months in vivo. Mass and volume were measured; dexamethasone microsphere-containing samples demonstrated greater adipose tissue retention compared to the control group. Histological analysis, including hematoxylin and eosin and CD31 staining, indicated increased vascularization (p < 0.05) within the Dex MS-containing samples. Controlled delivery of adipogenic factors, such as dexamethasone via polymer microspheres, significantly affects adipose tissue retention by maintaining healthy tissue formation and vascularization. Dex DW MS provide an improved model to former Dex SW MS, resulting in notably longer release time and, consequently, larger volumes of adipose retained in vivo. The use of microspheres, specifically double-walled, as vehicles for controlled drug delivery of adipogenic factors therefore present a clinically relevant model of adipose retention that has the potential to greatly improve soft tissue repair.
Collapse
Affiliation(s)
- Arta Kelmendi-Doko
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - J Peter Rubin
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Katarina Klett
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Christopher Mahoney
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sheri Wang
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kacey G Marra
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
48
|
|
49
|
Gomathysankar S, Halim AS, Yaacob NS, Noor NM, Mohamed M. Compatibility of Porous Chitosan Scaffold with the Attachment and Proliferation of human Adipose-Derived Stem Cells In Vitro. J Stem Cells Regen Med 2016. [PMID: 28096632 PMCID: PMC5227107 DOI: 10.46582/jsrm.1202012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Adipose-derived stem cells (ASCs) have potential applications in the repair and regeneration of various tissues and organs. The use of various scaffold materials as an excellent template for mimicking the extracellular matrix to induce the attachment and proliferation of different cell types has always been of interest in the field of tissue engineering because ideal biomaterials are in great demand. Chitosan, a marine polysaccharide, have wide clinical applications and it acts as a promising scaffold for cell migration and proliferation. ASCs, with their multi-differentiation potential, and chitosan, with its great biocompatibility with ASCs, were investigated in the present study. ASCs were isolated and were characterized by two different methods: immunocytochemistry and flow cytometry, using the mesenchymal stem cell markers CD90, CD105, CD73 and CD29. The ASCs were then induced to differentiate into adipogenic, osteogenic and chondrogenic lineages. These ASCs were incorporated into a porous chitosan scaffold (PCS), and their structural morphology was studied using a scanning electron microscope and hematoxylin and eosin staining. The proliferation rate of the ASCs on the PCS was assessed using a PrestoBlue viability assay. The results indicated that the PCS provides an excellent template for the adhesion and proliferation of ASCs. Thus, this study revealed that PCS is a promising biomaterial for inducing the proliferation of ASCs, which could lead to successful tissue reconstruction in the field of tissue engineering.
Collapse
Affiliation(s)
- Sankaralakshmi Gomathysankar
- Reconstructive Sciences Unit, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Ahmad Sukari Halim
- Reconstructive Sciences Unit, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Nik Soriani Yaacob
- Department of Chemical Pathology, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang kerian, Kelantan, Malaysia
| | - Norhayati Mohd Noor
- Reconstructive Sciences Unit, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Mohaini Mohamed
- Reconstructive Sciences Unit, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| |
Collapse
|
50
|
Kim DW, Kim EJ, Kim EN, Sung MW, Kwon TK, Cho YW, Kwon SK. Human Adipose Tissue Derived Extracellular Matrix and Methylcellulose Hydrogels Augments and Regenerates the Paralyzed Vocal Fold. PLoS One 2016; 11:e0165265. [PMID: 27768757 PMCID: PMC5074505 DOI: 10.1371/journal.pone.0165265] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 10/07/2016] [Indexed: 12/15/2022] Open
Abstract
Vocal fold paralysis results from various etiologies and can induce voice changes, swallowing complications, and issues with aspiration. Vocal fold paralysis is typically managed using injection laryngoplasty with fat or synthetic polymers. Injection with autologous fat has shown excellent biocompatibility. However, it has several disadvantages such as unpredictable resorption rate, morbidities associated with liposuction procedure which has to be done in operating room under general anesthesia. Human adipose-derived extracellular matrix (ECM) grafts have been reported to form new adipose tissue and have greater biostability than autologous fat graft. Here, we present an injectable hydrogel that is constructed from adipose tissue derived soluble extracellular matrix (sECM) and methylcellulose (MC) for use in vocal fold augmentation. Human sECM derived from adipose tissue was extracted using two major steps—ECM was isolated from human adipose tissue and was subsequently solubilized. Injectable sECM/MC hydrogels were prepared by blending of sECM and MC. Sustained vocal fold augmentation and symmetric vocal fold vibration were accomplished by the sECM/MC hydrogel in paralyzed vocal fold which were confirmed by laryngoscope, histology and a high-speed imaging system. There were increased number of collagen fibers and fatty granules at the injection site without significant inflammation or fibrosis. Overall, these results indicate that the sECM/MC hydrogel can enhance vocal function in paralyzed vocal folds without early resorption and has potential as a promising material for injection laryngoplasty for stable vocal fold augmentation which can overcome the shortcomings of autologous fat such as unpredictable duration and morbidity associated with the fat harvest.
Collapse
Affiliation(s)
- Dong Wook Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Eun Ji Kim
- Department of Chemical Engineering, Hanyang University, Ansan, Gyeonggi-do 426–791, Republic of Korea
| | - Eun Na Kim
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Myung Whun Sung
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Tack-Kyun Kwon
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yong Woo Cho
- Department of Chemical Engineering, Hanyang University, Ansan, Gyeonggi-do 426–791, Republic of Korea
- * E-mail: (SKK); (YWC)
| | - Seong Keun Kwon
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
- * E-mail: (SKK); (YWC)
| |
Collapse
|