1
|
De Francesco F, Sbarbati A, Sierra LAQ, Zingaretti N, Sarmadian Z, Parodi PC, Ricci G, Riccio M, Mobasheri A. Anatomy, Histology, and Embryonic Origin of Adipose Tissue: Insights to Understand Adipose Tissue Homofunctionality in Regeneration and Therapies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1474:53-78. [PMID: 39107527 DOI: 10.1007/5584_2024_801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Preadipocytes are formed during the 14th and 16th weeks of gestation. White adipose tissue, in particular, is generated in specific areas and thereby assembles after birth, rapidly increasing following the propagation of adipoblasts, which are considered the preadipocyte cell precursors. The second trimester of gestation is a fundamental phase of adipogenesis, and in the third trimester, adipocytes, albeit small may be present within the main deposition areas. In the course of late gestation, adipose tissue develops in the foetus and promotes the synthesis of large amounts of uncoupling protein 1, in similar quantities relative to differentiated brown adipose tissue. In mammals, differentiation occurs in two functionally different types of adipose cells: white adipose cells resulting from lipid storage and brown adipose cells from increased metabolic energy consumption. During skeletogenesis, synovial joints develop through the condensation of mesenchymal cells, which forms an insertional layer of flattened cells that umlaut skeletal elements, by sharing the same origin in the development of synovium. Peri-articular fat pads possess structural similarity with body subcutaneous white adipose tissue; however, they exhibit a distinct metabolic function due to the micro-environmental cues in which they are embedded. Fat pads are an important component of the synovial joint and play a key role in the maintenance of joint homeostasis. They are also implicated in pathological states such as osteoarthritis.In this paper we explore the therapeutic potential of adipocyte tissue mesenchymal precursor-based stem cell therapy linking it back to the anatomic origin of adipose tissue.
Collapse
Affiliation(s)
- Francesco De Francesco
- Department of Reconstructive Surgery and Hand Surgery, AOU Ospedali Riuniti delle Marche, Ancona, Italy
| | - Andrea Sbarbati
- Department of Neuroscience, Biomedicine and Movement, Human Anatomy and Histology Section, University of Verona, Verona, Italy
| | | | - Nicola Zingaretti
- Department of Medical Area (DAME), Clinic of Plastic and Reconstructive Surgery, Academic Hospital of Udine, University of Udine, Udine, Italy
| | - Zahra Sarmadian
- Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Pier Camillo Parodi
- Department of Medical Area (DAME), Clinic of Plastic and Reconstructive Surgery, Academic Hospital of Udine, University of Udine, Udine, Italy
| | - Giulia Ricci
- Department of Experimental Medicine, Università Degli Studi Della Campania "Luigi Vanvitelli", Naples, Italy
| | - Michele Riccio
- Department of Reconstructive Surgery and Hand Surgery, AOU Ospedali Riuniti delle Marche, Ancona, Italy
| | - Ali Mobasheri
- Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland.
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania.
- Department of Joint Surgery, Sun Yat-sen University, Guangzhou, People's Republic of China.
- World Health Organization Collaborating Center for Public Health Aspects of Musculoskeletal Health and Aging, Université de Liège, Liège, Belgium.
| |
Collapse
|
2
|
Lau CS, Park SY, Ethiraj LP, Singh P, Raj G, Quek J, Prasadh S, Choo Y, Goh BT. Role of Adipose-Derived Mesenchymal Stem Cells in Bone Regeneration. Int J Mol Sci 2024; 25:6805. [PMID: 38928517 PMCID: PMC11204188 DOI: 10.3390/ijms25126805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Bone regeneration involves multiple factors such as tissue interactions, an inflammatory response, and vessel formation. In the event of diseases, old age, lifestyle, or trauma, bone regeneration can be impaired which could result in a prolonged healing duration or requiring an external intervention for repair. Currently, bone grafts hold the golden standard for bone regeneration. However, several limitations hinder its clinical applications, e.g., donor site morbidity, an insufficient tissue volume, and uncertain post-operative outcomes. Bone tissue engineering, involving stem cells seeded onto scaffolds, has thus been a promising treatment alternative for bone regeneration. Adipose-derived mesenchymal stem cells (AD-MSCs) are known to hold therapeutic value for the treatment of various clinical conditions and have displayed feasibility and significant effectiveness due to their ease of isolation, non-invasive, abundance in quantity, and osteogenic capacity. Notably, in vitro studies showed AD-MSCs holding a high proliferation capacity, multi-differentiation potential through the release of a variety of factors, and extracellular vesicles, allowing them to repair damaged tissues. In vivo and clinical studies showed AD-MSCs favoring better vascularization and the integration of the scaffolds, while the presence of scaffolds has enhanced the osteogenesis potential of AD-MSCs, thus yielding optimal bone formation outcomes. Effective bone regeneration requires the interplay of both AD-MSCs and scaffolds (material, pore size) to improve the osteogenic and vasculogenic capacity. This review presents the advances and applications of AD-MSCs for bone regeneration and bone tissue engineering, focusing on the in vitro, in vivo, and clinical studies involving AD-MSCs for bone tissue engineering.
Collapse
Affiliation(s)
- Chau Sang Lau
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
- Oral Health Academic Clinical Programme, Duke-NUS Medical School, Singapore 169857, Singapore
| | - So Yeon Park
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
| | - Lalith Prabha Ethiraj
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
- Oral Health Academic Clinical Programme, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Priti Singh
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
| | - Grace Raj
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
| | - Jolene Quek
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; (J.Q.); (Y.C.)
| | - Somasundaram Prasadh
- Center for Clean Energy Engineering, University of Connecticut, Storrs, CT 06269, USA;
| | - Yen Choo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; (J.Q.); (Y.C.)
| | - Bee Tin Goh
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
- Oral Health Academic Clinical Programme, Duke-NUS Medical School, Singapore 169857, Singapore
| |
Collapse
|
3
|
Liang W, Han M, Li G, Dang W, Wu H, Meng X, Zhen Y, Lin W, Ao R, Hu X, An Y. Perfusable adipose decellularized extracellular matrix biological scaffold co-recellularized with adipose-derived stem cells and L6 promotes functional skeletal muscle regeneration following volumetric muscle loss. Biomaterials 2024; 307:122529. [PMID: 38489911 DOI: 10.1016/j.biomaterials.2024.122529] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 02/02/2024] [Accepted: 03/08/2024] [Indexed: 03/17/2024]
Abstract
Muscle tissue engineering is a promising therapeutic strategy for volumetric muscle loss (VML). Among them, decellularized extracellular matrix (dECM) biological scaffolds have shown certain effects in restoring muscle function. However, researchers have inconsistent or even contradictory results on whether dECM biological scaffolds can efficiently regenerate muscle fibers and restore muscle function. This suggests that therapeutic strategies based on dECM biological scaffolds need to be further optimized and developed. In this study, we used a recellularization method of perfusing adipose-derived stem cells (ASCs) and L6 into adipose dECM (adECM) through vascular pedicles. On one hand, this strategy ensures sufficient quantity and uniform distribution of seeded cells inside scaffold. On the other hand, auxiliary L6 cells addresses the issue of low myogenic differentiation efficiency of ASCs. Subsequently, the treatment of VML animal experiments showed that the combined recellularization strategy can improve muscle regeneration and angiogenesis than the single ASCs recellularization strategy, and the TA of former had greater muscle contraction strength. Further single-nucleus RNA sequencing (snRNA-seq) analysis found that L6 cells induced ASCs transform into a new subpopulation of cells highly expressing Mki67, CD34 and CDK1 genes, which had stronger ability of oriented myogenic differentiation. This study demonstrates that co-seeding ASCs and L6 cells through vascular pedicles is a promising recellularization strategy for adECM biological scaffolds, and the engineered muscle tissue constructed based on this has significant therapeutic effects on VML. Overall, this study provides a new paradigm for optimizing and developing dECM-based therapeutic strategies.
Collapse
Affiliation(s)
- Wei Liang
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Meng Han
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Guan Li
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Wanwen Dang
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Huiting Wu
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Xiaoyu Meng
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Yonghuan Zhen
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Weibo Lin
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Rigele Ao
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Xiaoqing Hu
- Department of Sports Medicine, Peking University Third Hospital, Beijing, 100191, China.
| | - Yang An
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
4
|
Abdelbasset WK, Jasim SA, Bokov DO, Shalaby MN, Opulencia MJC, Thangavelu L, Alkadir OKA, Ansari MJ, Kzar HH, Al-Gazally ME. Polysaccharides, as biological macromolecule-based platforms in skeletal muscle tissue engineering: a systematic review. INT J POLYM MATER PO 2022. [DOI: 10.1080/00914037.2022.2090940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Walid Kamal Abdelbasset
- Department of Health and Rehabilitation Sciences, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Al Kharj, Saudi Arabia
- Department of Physical Therapy, Kasr Al-Aini Hospital, Cairo University, Giza, Egypt
| | - Saade Abdalkareem Jasim
- Medical Laboratory Techniques Department, Al-Maarif University College, Al-Anbar-Ramadi, Iraq
| | - Dmitry Olegovich Bokov
- Institute of Pharmacy, Sechenov First Moscow State Medical University, Moscow, Russia
- Federal Research Center of Nutrition, Biotechnology and Food Safety, Laboratory of Food Chemistry, Moscow, Russia
| | - Mohammed Nader Shalaby
- Biological Sciences and Sports Health Department, Faculty of Physical Education, Suez Canal University, Sheikh Zayed City, Egypt
| | | | - Lakshmi Thangavelu
- Department of Pharmacology, Center for Transdisciplinary Research, Saveetha Dental College, Saveetha Institute of Medical and Technical Science, Saveetha University, Chennai, India
| | | | - Mohammad Javed Ansari
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Hamzah H. Kzar
- College of Veterinary Medicine, Al Qasim Green University, Iraq
| | | |
Collapse
|
5
|
Ragni E, Viganò M, De Luca P, Pedrini E, de Girolamo L. Adipose-Derived Stem/Stromal Cells, Stromal Vascular Fraction, and Microfragmented Adipose Tissue. ORTHOBIOLOGICS 2022:47-61. [DOI: 10.1007/978-3-030-84744-9_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
6
|
Ferroni L, De Francesco F, Pinton P, Gardin C, Zavan B. Methods to isolate adipose tissue-derived stem cells. Methods Cell Biol 2022; 171:215-228. [DOI: 10.1016/bs.mcb.2022.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
7
|
Raghav PK, Mann Z, Ahlawat S, Mohanty S. Mesenchymal stem cell-based nanoparticles and scaffolds in regenerative medicine. Eur J Pharmacol 2021; 918:174657. [PMID: 34871557 DOI: 10.1016/j.ejphar.2021.174657] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 11/05/2021] [Accepted: 11/24/2021] [Indexed: 12/20/2022]
Abstract
Mesenchymal stem cells (MSCs) are adult stem cells owing to their regenerative potential and multilineage potency. MSCs have wide-scale applications either in their native cellular form or in conjugation with specific biomaterials as nanocomposites. Majorly, these natural or synthetic biomaterials are being used in the form of metallic and non-metallic nanoparticles (NPs) to encapsulate MSCs within hydrogels like alginate or chitosan or drug cargo loading into MSCs. In contrast, nanofibers of polymer scaffolds such as polycaprolactone (PCL), poly-lactic-co-glycolic acid (PLGA), poly-L-lactic acid (PLLA), silk fibroin, collagen, chitosan, alginate, hyaluronic acid (HA), and cellulose are used to support or grow MSCs directly on it. These MSCs based nanotherapies have application in multiple domains of biomedicine including wound healing, bone and cartilage engineering, cardiac disorders, and neurological disorders. This study focused on current approaches of MSCs-based therapies and has been divided into two major sections. The first section elaborates on MSC-based nano-therapies and their plausible applications including exosome engineering and NPs encapsulation. The following section focuses on the various MSC-based scaffold approaches in tissue engineering. Conclusively, this review mainly focused on MSC-based nanocomposite's current approaches and compared their advantages and limitations for building effective regenerative medicines.
Collapse
Affiliation(s)
- Pawan Kumar Raghav
- Stem Cell Facility, DBT Centre of Excellence for Stem Cell Research, All India Institute of Medical Sciences, New Delhi, 110029, India.
| | - Zoya Mann
- Stem Cell Facility, DBT Centre of Excellence for Stem Cell Research, All India Institute of Medical Sciences, New Delhi, 110029, India.
| | - Swati Ahlawat
- Stem Cell Facility, DBT Centre of Excellence for Stem Cell Research, All India Institute of Medical Sciences, New Delhi, 110029, India.
| | - Sujata Mohanty
- Stem Cell Facility, DBT Centre of Excellence for Stem Cell Research, All India Institute of Medical Sciences, New Delhi, 110029, India.
| |
Collapse
|
8
|
Chen H, Fei F, Li X, Nie Z, Zhou D, Liu L, Zhang J, Zhang H, Fei Z, Xu T. A structure-supporting, self-healing, and high permeating hydrogel bioink for establishment of diverse homogeneous tissue-like constructs. Bioact Mater 2021; 6:3580-3595. [PMID: 33869899 PMCID: PMC8024533 DOI: 10.1016/j.bioactmat.2021.03.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 03/06/2021] [Accepted: 03/06/2021] [Indexed: 12/17/2022] Open
Abstract
The ready-to-use, structure-supporting hydrogel bioink can shorten the time for ink preparation, ensure cell dispersion, and maintain the preset shape/microstructure without additional assistance during printing. Meanwhile, ink with high permeability might facilitate uniform cell growth in biological constructs, which is beneficial to homogeneous tissue repair. Unfortunately, current bioinks are hard to meet these requirements simultaneously in a simple way. Here, based on the fast dynamic crosslinking of aldehyde hyaluronic acid (AHA)/N-carboxymethyl chitosan (CMC) and the slow stable crosslinking of gelatin (GEL)/4-arm poly(ethylene glycol) succinimidyl glutarate (PEG-SG), we present a time-sharing structure-supporting (TSHSP) hydrogel bioink with high permeability, containing 1% AHA, 0.75% CMC, 1% GEL and 0.5% PEG-SG. The TSHSP hydrogel can facilitate printing with proper viscoelastic property and self-healing behavior. By crosslinking with 4% PEG-SG for only 3 min, the integrity of the cell-laden construct can last for 21 days due to the stable internal and external GEL/PEG-SG networks, and cells manifested long-term viability and spreading morphology. Nerve-like, muscle-like, and cartilage-like in vitro constructs exhibited homogeneous cell growth and remarkable biological specificities. This work provides not only a convenient and practical bioink for tissue engineering, targeted cell therapy, but also a new direction for hydrogel bioink development.
Collapse
Affiliation(s)
- Hongqing Chen
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
- Department of Neurosurgery, Central Theater General Hospital, Wuhan, 430010, People's Republic of China
| | - Fei Fei
- Department of Ophthalmology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Xinda Li
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
- Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, China
| | - Zhenguo Nie
- Department of Orthopedics, Fourth Medical Center of PLA General Hospital, 100048, Beijing, People's Republic of China
| | - Dezhi Zhou
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
- Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Libiao Liu
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
- Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Jing Zhang
- East China Institute of Digital Medical Engineering, Shangrao, 334000, People's Republic of China
| | - Haitao Zhang
- East China Institute of Digital Medical Engineering, Shangrao, 334000, People's Republic of China
| | - Zhou Fei
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Tao Xu
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
- Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
- Department of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, 518055, People's Republic of China
| |
Collapse
|
9
|
Dienes J, Browne S, Farjun B, Amaral Passipieri J, Mintz EL, Killian G, Healy KE, Christ GJ. Semisynthetic Hyaluronic Acid-Based Hydrogel Promotes Recovery of the Injured Tibialis Anterior Skeletal Muscle Form and Function. ACS Biomater Sci Eng 2021; 7:1587-1599. [PMID: 33660968 DOI: 10.1021/acsbiomaterials.0c01751] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Volumetric muscle loss (VML) injuries are characterized by a degree of tissue loss that exceeds the endogenous regenerative capacity of muscle, resulting in permanent structural and functional deficits. Such injuries are a consequence of trauma, as well as a host of congenital and acquired diseases and disorders. Despite significant preclinical research with diverse biomaterials, as well as early clinical studies with implantation of decellularized extracellular matrices, there are still significant barriers to more complete restoration of muscle form and function following repair of VML injuries. In fact, identification of novel biomaterials with more advantageous regenerative profiles is a critical limitation to the development of improved therapeutics. As a first step in this direction, we evaluated a novel semisynthetic hyaluronic acid-based (HyA) hydrogel that embodies material features more favorable for robust muscle regeneration. This HyA-based hydrogel is composed of an acrylate-modified HyA (AcHyA) macromer, an AcHyA macromer conjugated with the bsp-RGD(15) peptide sequence to enhance cell adhesion, a high-molecular-weight heparin to sequester growth factors, and a matrix metalloproteinase-cleavable cross-linker to allow for cell-dependent remodeling. In a well-established, clinically relevant rat tibialis anterior VML injury model, we report observations of robust functional recovery, accompanied by volume reconstitution, muscle regeneration, and native-like vascularization following implantation of the HyA-based hydrogel at the site of injury. These findings have important implications for the development and clinical application of the improved biomaterials that will be required for stable and complete functional recovery from diverse VML injuries.
Collapse
Affiliation(s)
- Jack Dienes
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Shane Browne
- Department of Bioengineering, University of California, Berkeley, Berkeley, California 94720, United States.,Department of Material Science and Engineering, University of California, Berkeley, Berkeley 94720, United States
| | - Bruna Farjun
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Juliana Amaral Passipieri
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Ellen L Mintz
- Pathology Department, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Grant Killian
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Kevin E Healy
- Department of Bioengineering, University of California, Berkeley, Berkeley, California 94720, United States.,Department of Material Science and Engineering, University of California, Berkeley, Berkeley 94720, United States
| | - George J Christ
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22908, United States.,Department of Orthopaedic Surgery, University of Virginia, Charlottesville, Virginia 22908, United States
| |
Collapse
|
10
|
Smith RJP, Faroni A, Barrow JR, Soul J, Reid AJ. The angiogenic potential of CD271+ human adipose tissue-derived mesenchymal stem cells. Stem Cell Res Ther 2021; 12:160. [PMID: 33653407 PMCID: PMC7927269 DOI: 10.1186/s13287-021-02177-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 01/19/2021] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Autologous fat grafting is often a crucial aspect of reconstructive and aesthetic surgeries, yet poor graft retention is a major issue with this technique. Enriching fat grafts with adipose tissue-derived mesenchymal stem cells (AD-MSCs) improves graft survival-however, AD-MSCs represent a heterogeneous population. Selection of subpopulations of AD-MSCs would allow the targeting of specific AD-MSCs that may benefit fat graft survival more than the general AD-MSC population. METHODS Human AD-MSCs were selected for the surface marker CD271 using magnetic-activated cell sorting and compared to the CD271 negative phenotype. These subpopulations were analysed for gene expression using Real-Time qPCR and RNA sequencing; surface marker characteristics using immunostaining; ability to form tubules when cultured with endothelial cells; and gene and protein expression of key angiogenic mediators when cultured with ex-vivo adipose tissue. RESULTS Human AD-MSCs with the surface marker CD271 express angiogenic genes at higher levels, and inflammatory genes at lower levels, than the CD271- AD-MSC population. A greater proportion of CD271+ AD-MSCs also possess the typical complement of stem cell surface markers and are more likely to promote effective neoangiogenesis, compared to CD271- AD-MSCs. CONCLUSION Enriching grafts with the CD271+ AD-MSC subpopulation holds potential for the improvement of reconstructive and aesthetic surgeries involving adipose tissue.
Collapse
Affiliation(s)
- Richard J P Smith
- Blond McIndoe Laboratories, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, UK
| | - Alessandro Faroni
- Blond McIndoe Laboratories, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, UK
| | - James R Barrow
- Blond McIndoe Laboratories, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, UK
| | - Jamie Soul
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Adam J Reid
- Blond McIndoe Laboratories, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, UK. .,Department of Plastic Surgery & Burns, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK.
| |
Collapse
|
11
|
De Francesco F. Editorial: Mesenchymal Stem Cells and Interactions With Scaffolds - Biomaterials in Regenerative Medicine: From Research to Translational Applications. Front Cell Dev Biol 2019; 7:193. [PMID: 31572725 PMCID: PMC6751261 DOI: 10.3389/fcell.2019.00193] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 08/28/2019] [Indexed: 12/31/2022] Open
Affiliation(s)
- Francesco De Francesco
- Hand Surgery Unit, Department of Plastic Reconstructive Surgery, Azienda Ospedaliero Universitaria Ospedali Riuniti, Ancona, Italy
| |
Collapse
|
12
|
Senesi L, De Francesco F, Farinelli L, Manzotti S, Gagliardi G, Papalia GF, Riccio M, Gigante A. Mechanical and Enzymatic Procedures to Isolate the Stromal Vascular Fraction From Adipose Tissue: Preliminary Results. Front Cell Dev Biol 2019; 7:88. [PMID: 31231649 PMCID: PMC6565890 DOI: 10.3389/fcell.2019.00088] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 05/09/2019] [Indexed: 12/30/2022] Open
Abstract
Adipose-derived MSCs (ASCs) and stromal vascular fraction (SVF) play an important role in regenerative medicine and in the treatment of osteoarthritis. ASCs extracted from lipoaspirates are a valuable cell source due to their abundance and accessibility. ASCs are retrieved from the aqueous fraction of the digested lipoaspirate. The aqueous fraction is known as SVF and includes, ASCs, endothelial precursor cells (EPCs), endothelial cells (ECs), macrophages, smooth muscle cells, lymphocytes, pericytes, as well as pre-adipocytes. To date, two types of techniques to isolate SVF have been proposed: enzymatic and mechanical. The enzymatic method is particularly indicated in SVF isolation since it disrupts the extracellular matrix (ECM) and the binding of adipocytes and other cells but is restricted by regulatory issues related to enzymatic procedures, especially within the European Community. Thus, making the search for alternative mechanical methods imperative. This study assesses the SVF harvested from subcutaneous abdominal fat via two different mechanical procedures and the standard enzymatic method to evaluate their eligibility in a clinical context. In particular, we analyze cell viability (at 0 and after 72 h) as well as the expression of cluster differentiation (CD) for each sample and the differentiation in adipocytic, chondrocytic, osteocytic linage. The mechanical procedures yielded no significant difference in cell viability and cluster differentiation pattern expression, even if enzymatic procedure still remain the "gold standard." We retain that clinical efficacy in treating ostheoarthrosis with SVF administration is probably related to his anti-inflammatory and immunoregulatory effect, rather than the ability to differentiate in specific cell lineage. However, further studies are required to support and improve our findings.
Collapse
Affiliation(s)
- Letizia Senesi
- Clinical Orthopaedics, Department of Clinical and Molecular Science, Università Politecnica delle Marche, Ancona, Italy
- Department of Plastic Reconstructive Surgery and Hand Surgery, Azienda Ospedaliero Universitaria Ospedali Riuniti, Ancona, Italy
| | - Francesco De Francesco
- Department of Plastic Reconstructive Surgery and Hand Surgery, Azienda Ospedaliero Universitaria Ospedali Riuniti, Ancona, Italy
- Regenerative Surgery, Research and Training Center, Lipofilling Academy, Ancona, Italy
| | - Luca Farinelli
- Clinical Orthopaedics, Department of Clinical and Molecular Science, Università Politecnica delle Marche, Ancona, Italy
| | - Sandra Manzotti
- Clinical Orthopaedics, Department of Clinical and Molecular Science, Università Politecnica delle Marche, Ancona, Italy
| | - Giulio Gagliardi
- Clinical Orthopaedics, Department of Clinical and Molecular Science, Università Politecnica delle Marche, Ancona, Italy
| | - Giuseppe Francesco Papalia
- Clinical Orthopaedics, Department of Clinical and Molecular Science, Università Politecnica delle Marche, Ancona, Italy
| | - Michele Riccio
- Department of Plastic Reconstructive Surgery and Hand Surgery, Azienda Ospedaliero Universitaria Ospedali Riuniti, Ancona, Italy
- Regenerative Surgery, Research and Training Center, Lipofilling Academy, Ancona, Italy
| | - Antonio Gigante
- Clinical Orthopaedics, Department of Clinical and Molecular Science, Università Politecnica delle Marche, Ancona, Italy
- Regenerative Surgery, Research and Training Center, Lipofilling Academy, Ancona, Italy
| |
Collapse
|
13
|
Hamada T, Matsubara H, Yoshida Y, Ugaji S, Nomura I, Tsuchiya H. Autologous adipose-derived stem cell transplantation enhances healing of wound with exposed bone in a rat model. PLoS One 2019; 14:e0214106. [PMID: 31083652 PMCID: PMC6513073 DOI: 10.1371/journal.pone.0214106] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 04/30/2019] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVES Soft tissue wounds with exposed bone often require extended healing times and can be associated with severe complications. We describe the ability of artificial dermis with autogenic adipose-derived stem cells (ADSCs) to promote the healing of wounds with exposed bone in a rat model. METHODS Adipose tissues harvested from the bilateral inguinal regions of Wistar rats were used as ADSCs. Rats were randomly divided into control and ADSC groups to investigate the efficacy of ADSC transplantation for wound healing (n = 20 per group). Soft tissue defects were created on the heads of the rats and were covered with artificial dermis with or without the seeded ADSCs. Specimens from these rats were evaluated using digital image analysis, histology, immunohistochemistry, cell labeling, and real-time reverse-transcription polymerase chain reaction (real-time RT-PCR). RESULTS The average global wound area was significantly smaller in the ADSC group than in the control group on days 3, 7, and 14 after surgery (p<0.05). After 14 days, the blood vessel density in the wound increased by 1.6-fold in the ADSC group compared with that in the control group (p<0.01). Real-time RT-PCR results showed higher Fgfb and Vegf expression levels at all time points, and higher Tgfb1 and Tgfb3 expression levels until 14 days after surgery in the ADSC group than in the control group (p<0.05). CONCLUSIONS In wounds with exposed bone, autogenic ADSCs can promote vascularization and wound healing. Use of this cell source has multiple benefits, including convenient clinical application and lack of ethical concerns.
Collapse
Affiliation(s)
- Tomo Hamada
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Hidenori Matsubara
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
- * E-mail:
| | - Yasuhisa Yoshida
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Shuhei Ugaji
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Issei Nomura
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Hiroyuki Tsuchiya
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
14
|
Kundu B, Bastos ARF, Brancato V, Cerqueira MT, Oliveira JM, Correlo VM, Reis RL, Kundu SC. Mechanical Property of Hydrogels and the Presence of Adipose Stem Cells in Tumor Stroma Affect Spheroid Formation in the 3D Osteosarcoma Model. ACS APPLIED MATERIALS & INTERFACES 2019; 11:14548-14559. [PMID: 30943004 DOI: 10.1021/acsami.8b22724] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Osteosarcoma is one of the most common metastatic bone cancers, which results in significant morbidity and mortality. Unfolding of effectual therapeutic strategies against osteosarcoma is impeded because of the absence of adequate animal models, which can truly recapitulate disease biology of humans. Tissue engineering provides an opportunity to develop physiologically relevant, reproducible, and tunable in vitro platforms to investigate the interactions of osteosarcoma cells with its microenvironment. Adipose-derived stem cells (ASCs) are detected adjacent to osteosarcoma masses and are considered to have protumor effects. Hence, the present study focuses on investigating the role of reactive ASCs in formation of spheroids of osteosarcoma cells (Saos 2) within a three-dimensional (3D) niche, which is created using gellan gum (GG)-silk fibroin. By modifying the blending ratio of GG-silk, the optimum stiffness of the resultant hydrogels such as GG and GG75: S25 is obtained for cancer spheroid formation. This work indicates that the co-existence of cancer and stem cells can form a spheroid, the hallmark of cancer, only in particular microenvironment stiffness. The incorporation of fibrillar silk fibroin within the hydrophilic network of GG in GG75: S25 spongy-like hydrogels closely mimics the stiffness of commercially established cancer biomaterials (e.g., Matrigel, HyStem). The GG75: S25 hydrogel maintains the metabolically active construct for a longer time with elevated expression of osteopontin, osteocalcin, RUNX 2, and bone sialoprotein genes, the biomarkers of osteosarcoma, compared to GG. The GG75: S25 construct also exhibits intense alkaline phosphatase expression in immunohistochemistry compared to GG, indicating itspotentiality to serve as biomimetic niche to model osteosarcoma. Taken together, the GG-silk fibroin-blended spongy-like hydrogel is envisioned as an alternative low-cost platform for 3D cancer modeling.
Collapse
Affiliation(s)
- B Kundu
- I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics , University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine , AvePark, Zona Industrial da Gandra , Barco, Guimarães 4805-017 , Portugal
- ICVS/3B's-PT Government Associate Laboratory , Braga, Guimarães 4805-017 , Portugal
| | - A R F Bastos
- I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics , University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine , AvePark, Zona Industrial da Gandra , Barco, Guimarães 4805-017 , Portugal
- ICVS/3B's-PT Government Associate Laboratory , Braga, Guimarães 4805-017 , Portugal
| | - V Brancato
- I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics , University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine , AvePark, Zona Industrial da Gandra , Barco, Guimarães 4805-017 , Portugal
- ICVS/3B's-PT Government Associate Laboratory , Braga, Guimarães 4805-017 , Portugal
| | - M T Cerqueira
- I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics , University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine , AvePark, Zona Industrial da Gandra , Barco, Guimarães 4805-017 , Portugal
- ICVS/3B's-PT Government Associate Laboratory , Braga, Guimarães 4805-017 , Portugal
| | - J M Oliveira
- I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics , University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine , AvePark, Zona Industrial da Gandra , Barco, Guimarães 4805-017 , Portugal
- ICVS/3B's-PT Government Associate Laboratory , Braga, Guimarães 4805-017 , Portugal
- The Discoveries Centre for Regenerative and Precision Medicine , Headquarters at University of Minho , Avepark , Barco, Guimarães 4805-017 , Portugal
| | - V M Correlo
- I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics , University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine , AvePark, Zona Industrial da Gandra , Barco, Guimarães 4805-017 , Portugal
- ICVS/3B's-PT Government Associate Laboratory , Braga, Guimarães 4805-017 , Portugal
- The Discoveries Centre for Regenerative and Precision Medicine , Headquarters at University of Minho , Avepark , Barco, Guimarães 4805-017 , Portugal
| | - R L Reis
- I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics , University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine , AvePark, Zona Industrial da Gandra , Barco, Guimarães 4805-017 , Portugal
- ICVS/3B's-PT Government Associate Laboratory , Braga, Guimarães 4805-017 , Portugal
- The Discoveries Centre for Regenerative and Precision Medicine , Headquarters at University of Minho , Avepark , Barco, Guimarães 4805-017 , Portugal
| | - S C Kundu
- I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics , University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine , AvePark, Zona Industrial da Gandra , Barco, Guimarães 4805-017 , Portugal
- ICVS/3B's-PT Government Associate Laboratory , Braga, Guimarães 4805-017 , Portugal
| |
Collapse
|
15
|
Stanco D, Caprara C, Ciardelli G, Mariotta L, Gola M, Minonzio G, Soldati G. Tenogenic differentiation protocol in xenogenic-free media enhances tendon-related marker expression in ASCs. PLoS One 2019; 14:e0212192. [PMID: 30753235 PMCID: PMC6372228 DOI: 10.1371/journal.pone.0212192] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 01/29/2019] [Indexed: 12/03/2022] Open
Abstract
Adipose-derived stem cells (ASCs) are multipotent and immune-privileged mesenchymal cells, making them ideal candidates for therapeutic purposes to manage tendon disorders. Providing safe and regulated cell therapy products to patients requires adherence to good manufacturing practices. To this aim we investigated the in vitro tenogenic differentiation potential of ASCs using a chemically defined serum-free medium (SF) or a xenogenic-free human pooled platelet lysate medium (hPL) suitable for cell therapy and both supplemented with CTGF, TGFβ-3, BMP-12 and ascorbic acid (AA) soluble factors. Human ASCs were isolated from 4 healthy donors and they were inducted to differentiate until 14 days in both hPL and SF tenogenic media (hPL-TENO and SF-TENO). Cell viability and immunophenotype profile were analysed to evaluate mesenchymal stem cell (MSC) characteristics in both xenogenic-free media. Moreover, the expression of stemness and tendon-related markers upon cell differentiation by RT-PCR, protein staining and cytofluorimetric analysis were also performed. Our results showed the two xenogenic-free media well support cell viability of ASCs and maintain their MSC nature as demonstrated by their typical immunophenototype profile and by the expression of NANOG, OCT4 and Ki67 genes. Moreover, both hPL-TENO and SF-TENO expressed significant high levels of the tendon-related genes SCX, COL1A1, COL3A1, COMP, MMP3 and MMP13 already at early time points in comparison to the respective controls. Significant up-regulations in scleraxis, collagen and tenomodulin proteins were also demonstrated at in both differentiated SF and hPL ASCs. In conclusion, we demonstrated firstly the feasibility of both serum and xenogenic-free media tested to culture ASCs moving forward the GMP-compliant approaches for clinical scale expansion of human MSCs needed for therapeutical application of stem cells. Moreover, a combination of CTGF, BMP-12, TGFβ3 and AA factors strongly and rapidly induce human ASCs to differentiate into tenocyte-like cells.
Collapse
Affiliation(s)
- Deborah Stanco
- Swiss Stem Cell Foundation, Gentilino, Switzerland
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | | | - Gianluca Ciardelli
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | | | - Mauro Gola
- Swiss Stem Cell Foundation, Gentilino, Switzerland
| | | | - Gianni Soldati
- Swiss Stem Cell Foundation, Gentilino, Switzerland
- * E-mail:
| |
Collapse
|
16
|
Silva Garcia JM, Panitch A, Calve S. Functionalization of hyaluronic acid hydrogels with ECM-derived peptides to control myoblast behavior. Acta Biomater 2019; 84:169-179. [PMID: 30508655 DOI: 10.1016/j.actbio.2018.11.030] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 10/31/2018] [Accepted: 11/19/2018] [Indexed: 01/07/2023]
Abstract
Volumetric muscle loss (VML) occurs when skeletal muscle injury is too large for the body to fully self-repair. Typically, fibrotic tissue fills the void, which reduces muscle functionality and limb movement. Although a wide variety of natural and synthetic scaffolds have been studied with the purpose of providing the appropriate structural support, to date no scaffold has significantly restored muscle functionality after VML. Satellite cells, adult stem cells within the muscle capable of restoring smaller injuries, are sensitive to the stiffness and composition of the surrounding environment. Scaffolds that only address structural support are not sufficient to restore functionality and instead need to be designed to both promote satellite cell activation and prevent excessive fibroblast recruitment. The objective of this study was to design a scaffold that mimicked the regenerative environment and determine how the biomechanical properties differentially influence myogenic precursor and connective tissue cells. One of the main extracellular matrix (ECM) molecules upregulated during regeneration is hyaluronic acid (HA). Therefore, thiol-modified HA and poly(ethylene glycol) diacrylate hydrogels were generated and functionalized with peptides based on ECM known to influence regeneration, including fibronectin, laminin and tenascin-C. Scaffolds with different stiffness were created by varying HA content. The influence of HA stiffness and peptide functionalization on myogenic precursor and connective tissue cell proliferation, migration and gene expression was quantified. Our results indicated that HA hydrogels functionalized with the laminin peptide, IKVAV, show potential due to the enhanced promotion of myogenic cell behaviors including migration, proliferation and an increase in relevant transcription factors. STATEMENT OF SIGNIFICANCE: The goal of this study was to identify hyaluronic acid (HA) hydrogels with peptide and stiffness combinations that will direct muscle-derived cells towards regenerating phenotypes. While the interaction of skeletal muscle with RGD-functionalized HA hydrogels has been investigated, none of the other peptides described in this study had been used in the context of HA-based scaffolds and skeletal muscle-derived cells. Notably, the response of cells to variations in mechanics was dependent on ECM coating and lineage. The 3% HA functionalized with the laminin peptide, IKVAV, showed the most promise for future in vivo studies, as these hydrogels best promoted myoblast cell proliferation, attachment and spreading, enhanced migration over connective tissue cells and upregulated transcription factors associated with activated satellite cells.
Collapse
|
17
|
Smith RJ, Reid AJ. The potential of adipose-derived stem cell subpopulations in regenerative medicine. Regen Med 2018; 13:357-360. [PMID: 29979099 DOI: 10.2217/rme-2018-0030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Affiliation(s)
- Richard Jp Smith
- Blond McIndoe Laboratories, Division of Cell Matrix Biology & Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine & Health, University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PL, UK
| | - Adam J Reid
- Blond McIndoe Laboratories, Division of Cell Matrix Biology & Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine & Health, University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PL, UK.,Department of Plastic Surgery & Burns, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| |
Collapse
|
18
|
Tong J, Mou S, Xiong L, Wang Z, Wang R, Weigand A, Yuan Q, Horch RE, Sun J, Yang J. Adipose-derived mesenchymal stem cells formed acinar-like structure when stimulated with breast epithelial cells in three-dimensional culture. PLoS One 2018; 13:e0204077. [PMID: 30335754 PMCID: PMC6193614 DOI: 10.1371/journal.pone.0204077] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 08/31/2018] [Indexed: 02/08/2023] Open
Abstract
Lipotransfer has been applied in breast augmentation surgery for several years and the resident adipose-derived stem cells (ASCs) play an important role in enhancing fat graft survival. However, the interaction between ASCs and mammary epithelium is not fully understood. Many studies have shown that ASCs have a tumor-supportive effect in breast cancer. To the best of our knowledge, this is the first study on the effect of mammary epithelial cells on the human ASCs in 3D culture. ASCs were cultivated on matrigel in the conditioned medium (CM) prepared from a human breast epithelial cell line (HBL-100). The ASCs formed KRT18-positive acini-like structures after stimulation with breast epithelial cells. The expression of epithelial genes (CDH1 and KRT18) was up-regulated while the expression of mesenchymal specific genes (CDH2 and VIM) was down-regulated as determined by qRT-PCR. The stemness marker (CD29) and angiogenic factors (CD31 and VEGF) were also down-regulated as examined by immunofluorescence. In addition, the CM obtained from HBL-100 enhanced the migration and inhibited the adipogenic differentiation of ASCs. These results demonstrate that ASCs have the ability to transform into epithelial-like cells when cultured with mammary epithelial cells. Given these observations, we infer that ASCs have a positive effect on lipotransfer, not only due to their ability to secrete growth factors, but also due to their direct participation in the formation of new breast tissue.
Collapse
Affiliation(s)
- Jing Tong
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shan Mou
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lingyun Xiong
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhenxing Wang
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rongrong Wang
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Annika Weigand
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital Erlangen, Friedrich Alexander University, Erlangen‐Nuernberg, FAU, Germany
| | - Quan Yuan
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Raymund E. Horch
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital Erlangen, Friedrich Alexander University, Erlangen‐Nuernberg, FAU, Germany
| | - Jiaming Sun
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- * E-mail: (JY); (JS)
| | - Jie Yang
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- * E-mail: (JY); (JS)
| |
Collapse
|
19
|
Gilbert-Honick J, Ginn B, Zhang Y, Salehi S, Wagner KR, Mao HQ, Grayson WL. Adipose-derived Stem/Stromal Cells on Electrospun Fibrin Microfiber Bundles Enable Moderate Muscle Reconstruction in a Volumetric Muscle Loss Model. Cell Transplant 2018; 27:1644-1656. [PMID: 30298751 PMCID: PMC6299198 DOI: 10.1177/0963689718805370] [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] [Indexed: 01/08/2023] Open
Abstract
Current treatment options for volumetric muscle loss (VML) are limited due to donor site morbidity, lack of donor tissue, and insufficient functional recovery. Tissue-engineered skeletal muscle grafts offer the potential to significantly improve functional outcomes. In this study, we assessed the potential pro-myogenic effects of human adipose-derived stem cells (ASCs) seeded onto electrospun uniaxially aligned fibrin hydrogel microfiber bundles. Although both uninduced and 5-azacytidine-induced ASCs exhibited alignment, elongation, and diffuse muscle marker expression when grown on microfiber bundles for 2 months in vitro, both groups failed to fully recapitulate myotube characteristics. To assess the muscle regeneration potential of ASCs in vivo, ASC-seeded fibrin microfiber bundles were implanted in a robust murine VML defect model. Minimal fibrosis was observed surrounding implanted acellular hydrogel fibers at 2 and 4 weeks, and fibers seeded with ASCs exhibited up to 4 times higher volume retention than acellular fibers. We observed increased numbers of cells positive for the regenerating muscle marker embryonic myosin and the mature muscle marker myosin heavy chain in ASC-seeded fibers compared with acellular fibers at 1 and 3 months post-transplantation. Regenerating muscle cells were closely associated with ASC-derived cells and in some cases had potentially fused with them. These findings demonstrate that despite failing to undergo myogenesis in vitro, ASCs combined with electrospun fibrin microfibers moderately increased muscle reconstruction in vivo compared with acellular fibers following a severe VML defect.
Collapse
Affiliation(s)
- Jordana Gilbert-Honick
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Brian Ginn
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA.,Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Yuanfan Zhang
- The Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD, USA.,Graduate Program in Cellular and Molecular Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Sara Salehi
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kathryn R Wagner
- The Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD, USA.,Graduate Program in Cellular and Molecular Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA.,Departments of Neurology and Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Hai-Quan Mao
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA.,Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Warren L Grayson
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA.,Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
20
|
Hasani S, Boroujeni ME, Aliaghaei A, Baghai K, Rostami A. Dopaminergic induction of human adipose-derived mesenchymal stem cells is accompanied by transcriptional activation of autophagy. Cell Biol Int 2018; 42:1688-1694. [DOI: 10.1002/cbin.11056] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Accepted: 09/16/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Sanaz Hasani
- Faculty of Medical Biotechnology; Department of Stem Cells and Regenerative Medicine; National Institute of Genetic Engineering and Biotechnology; Tehran Iran
| | - Mahdi Eskandarian Boroujeni
- Faculty of Medical Biotechnology; Department of Stem Cells and Regenerative Medicine; National Institute of Genetic Engineering and Biotechnology; Tehran Iran
| | - Abbas Aliaghaei
- Cell Biology and Anatomical Sciences; School of Medicine; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - Kaveh Baghai
- Basic and Molecular Epidemiology of Gastrointestinal Disorder Research center; Research institute for Gastroenterology and Liver Diseases; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - Amin Rostami
- Gastroenterology and Liver Disease Research Center; Research institute for Gastroenterology and Liver Diseases; Shahid Beheshti University of Medical Sciences; Tehran Iran
| |
Collapse
|
21
|
Scioli MG, Artuso S, D'Angelo C, Porru M, D’Amico F, Bielli A, Gentile P, Cervelli V, Leonetti C, Orlandi A. Adipose-derived stem cell-mediated paclitaxel delivery inhibits breast cancer growth. PLoS One 2018; 13:e0203426. [PMID: 30192811 PMCID: PMC6128546 DOI: 10.1371/journal.pone.0203426] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 08/21/2018] [Indexed: 01/20/2023] Open
Abstract
Breast cancer represents the main malignancy in women and autologous fat grafting is a diffuse procedure in the management of post-surgical breast defects causing patients’ psychosocial problems, with high costs for the public health. Recently, beneficial effects of fat grafting during post-surgical breast reconstruction have been amplified from the enrichment with human adipose-derived stem cells (ASCs) present in the stromal vascular fraction (SVF) of adult adipose tissue isolated during intraoperatory procedures. The major concern about the ASC enrichment during post-surgery breast reconstruction depends on their potential ability to release growth factors and hormones that can promote proliferation of residual or quiescent cancer cells, with the risk of de novo cancer development or recurrence. The recent description that adult stem cells primed in vitro may be vehicle for anti-cancer drug delivery offers a new vision concerning the role of ASCs in breast reconstruction after cancer surgery. Paclitaxel (PTX) is a chemotherapeutic agent acting as a microtubule-stabilizing drug inhibiting cancer cell mitotic activity. We optimized PTX loading and release in cultured ASCs and then analyzed the effects of PTX-loaded ASCs and their conditioned medium on CG5 breast cancer survival, proliferation and apoptosis in vitro, and inCG5 xenograft in vivo. We documented that ASCs can uptake and release PTX in vitro, with slight cytotoxic effects. Interestingly, PTX-loaded ASCs in co-culture, as well as conditioned medium alone, inhibited CG5 cell proliferation and survival in vitro and xenograft tumor growth in vivo. The antitumor effect of PTX-loaded ASCs may offer a new perspective concerning the use of ASCs during breast reconstruction becoming an additional local preventive chemotherapeutic agent against tumor recurrence. However, further experiments in vitro and in vivo are needed to collect more evidence confirming the efficacy and safety in cancer patients.
Collapse
Affiliation(s)
- Maria Giovanna Scioli
- Anatomic Pathology, Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Simona Artuso
- SAFU, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Carmen D'Angelo
- Unit of Oncogenomics and Epigenetics, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Manuela Porru
- SAFU, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Federico D’Amico
- Anatomic Pathology, Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Alessandra Bielli
- Anatomic Pathology, Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Pietro Gentile
- Plastic and Reconstructive Surgery, Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Valerio Cervelli
- Plastic and Reconstructive Surgery, Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Carlo Leonetti
- SAFU, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Augusto Orlandi
- Anatomic Pathology, Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
- Anatomic Pathology, Department of Biomedical Sciences, Catholic University Our lady of Good Counsel, Tirana, Albania
- * E-mail:
| |
Collapse
|
22
|
Deng C, Sun Y, Liu H, Wang W, Wang J, Zhang F. Selective adipogenic differentiation of human periodontal ligament stem cells stimulated with high doses of glucose. PLoS One 2018; 13:e0199603. [PMID: 29979705 PMCID: PMC6034828 DOI: 10.1371/journal.pone.0199603] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 06/11/2018] [Indexed: 02/07/2023] Open
Abstract
Periodontal tissue damage, accompanied by the degradation and destruction of periodontal tissue collagen, is one of the most clinically common complications and difficulty self-repair in patients with diabetes. Human periodontal ligament stem cells (PDLSC) are the undifferentiated mesenchymal cells that persist in the periodontal ligament after development of periodontal tissue and the ability of PDLSC osteogenic differentiation is responsible for repairing periodontal tissue defects. However, the reasons of high glucose environment in diabetic patients inhibiting PDLSC to repair periodontal tissues are unclear. To address these issues, we propose exposing PDLSC to high-sugar mimics the diabetic environment and investigating the activity of osteogenic differentiation and adipogenic differentiation of PDLSC. At the cellular level, high glucose can promote the adipogenic differentiation and inhibit osteogenic differentiation to decrease the self-repair ability of PDLSC in periodontal tissues. Mechanistically at the molecular level, these effects are elicited via regulating the mRNA and protein expression of C/EBPβ, PPAR-γ.
Collapse
Affiliation(s)
- Chao Deng
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University,Nanjing, China
- School of Stomatology, Wannan Medical College, Wuhu, China
| | - Yi Sun
- School of Stomatology, Wannan Medical College, Wuhu, China
| | - Hai Liu
- School of Stomatology, Wannan Medical College, Wuhu, China
| | - Wei Wang
- School of Stomatology, Wannan Medical College, Wuhu, China
| | - Jingmen Wang
- School of Stomatology, Wannan Medical College, Wuhu, China
| | - Feimin Zhang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University,Nanjing, China
- * E-mail:
| |
Collapse
|
23
|
Bērziņš U, Matise-VanHoutana I, Pētersone I, Dūrītis I, Ņikuļšins S, Bogdanova-Jātniece A, Kālis M, Svirskis Š, Skrastiņa D, Ezerta A, Kozlovska T. Characterisation and In Vivo Safety of Canine Adipose-Derived Stem Cells. PROCEEDINGS OF THE LATVIAN ACADEMY OF SCIENCES. SECTION B. NATURAL, EXACT, AND APPLIED SCIENCES. 2018; 72:160-171. [DOI: 10.2478/prolas-2018-0004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Abstract
The study characterises canine adipose-derived stem cells (cASCs) in comparison to human ASCs (hASCs) and tests their safety in a canine model after intravenous administration. cASCs from two dogs were cultured under hypoxic conditions in a medium supplemented with autologous serum. They were plastic adherent, spindle-shaped cells that expressed CD73, CD90, and CD44 but lacked CD45, CD14, HLA-DR, and CD34. cASCs differentiated toward adipogenic, osteogenic, and chondrogenic lineages, although adipogenic differentiation capacity was low. Blast transformation reaction demonstrated that these cells significantly suppress T-cell proliferation, and this ability is dose-dependent. Intravenous administration of a cell freezing medium, therapeutic dose of cASCs (2 × 106 live cells/kg), and five times higher dose of cASCs showed no significant side effects in two dogs. Microscopic tissue lesions were limited to only mild, non-specific changes. There were no signs of malignancy. The results of the study indicate that cASCs are similar to hASCs and are safe for therapeutic applications in a canine model. The proposed methodology for ASC preparation on a non-routine basis, which includes individually optimised cell culture conditions and offers risk-adapted treatment, could be used for future personalised off-the-shelf therapies, for example, in myocardial infarction or stroke.
Collapse
Affiliation(s)
- Uldis Bērziņš
- Latvian Biomedical Research and Study Centre , 1 Rātsupītes Str., Rīga , LV-1067 , Latvia
- Stem Cells Technologies Ltd. , Rīga , Latvia
| | - Ilze Matise-VanHoutana
- Faculty of Veterinary Medicine , Latvia University of Agriculture , 2 Lielā Str., Jelgava , LV-3001 , Latvia
| | - Ilze Pētersone
- Faculty of Veterinary Medicine , Latvia University of Agriculture , 2 Lielā Str., Jelgava , LV-3001 , Latvia
| | - Ilmārs Dūrītis
- Faculty of Veterinary Medicine , Latvia University of Agriculture , 2 Lielā Str., Jelgava , LV-3001 , Latvia
| | - Sergejs Ņikuļšins
- Children’s Clinical University Hospital , 45 Vienības gatve, Rīga , LV-1004 , Latvia
| | | | - Mārtiņš Kālis
- Augusts Kirhenšteins Institute of Microbiology and Virology , Rīga Stradiņš University , 5 Rātsupītes Str., Rīga , LV-1067 , Latvia
| | - Šimons Svirskis
- Augusts Kirhenšteins Institute of Microbiology and Virology , Rīga Stradiņš University , 5 Rātsupītes Str., Rīga , LV-1067 , Latvia
| | - Dace Skrastiņa
- Latvian Biomedical Research and Study Centre , 1 Rātsupītes Str., Rīga , LV-1067 , Latvia
| | - Agnese Ezerta
- Latvian Biomedical Research and Study Centre , 1 Rātsupītes Str., Rīga , LV-1067 , Latvia
| | - Tatjana Kozlovska
- Latvian Biomedical Research and Study Centre , 1 Rātsupītes Str., Rīga , LV-1067 , Latvia
| |
Collapse
|
24
|
Olimpio RMC, de Oliveira M, De Sibio MT, Moretto FCF, Deprá IC, Mathias LS, Gonçalves BM, Rodrigues BM, Tilli HP, Coscrato VE, Costa SMB, Mazeto GMFS, Fernandes CJC, Zambuzzi WF, Saraiva PP, Maria DA, Nogueira CR. Cell viability assessed in a reproducible model of human osteoblasts derived from human adipose-derived stem cells. PLoS One 2018; 13:e0194847. [PMID: 29641603 PMCID: PMC5895002 DOI: 10.1371/journal.pone.0194847] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 03/09/2018] [Indexed: 01/04/2023] Open
Abstract
Human adipose tissue-derived stem cells (hASCs) have been subjected to extensive investigation because of their self-renewal properties and potential to restore damaged tissues. In the literature, there are several protocols for differentiating hASCs into osteoblasts, but there is no report on the control of cell viability during this process. In this study, we used osteoblasts derived from hASCs of patients undergoing abdominoplasty. The cells were observed at the beginning and end of bone matrix formation, and the expression of proteins involved in this process, including alkaline phosphatase and osteocalcin, was assessed. RANKL, Osterix, Runx2, Collagen3A1, Osteopontin and BSP expression levels were analyzed using real-time PCR, in addition to a quantitative assessment of protein levels of the markers CD45, CD105, STRO-1, and Nanog, using immunofluorescence. Rhodamine (Rho123), cytochrome-c, caspase-3, P-27, cyclin D1, and autophagy cell markers were analyzed by flow cytometry to demonstrate potential cellular activity and the absence of apoptotic and tumor cell processes before and after cell differentiation. The formation of bone matrix, along with calcium nodules, was observed after 16 days of osteoinduction. The gene expression levels of RANKL, Osterix, Runx2, Collagen3A1, Osteopontin, BSP and alkaline phosphatase activity were also elevated after 16 days of osteoinduction, whereas the level of osteocalcin was higher after 21 days of osteoinduction. Our data also showed that the cells had a high mitochondrial membrane potential and a low expression of apoptotic and tumor markers, both before and after differentiation. Cells were viable after the different phases of differentiation. This proposed methodology, using markers to evaluate cell viability, is therefore successful in assessing different phases of stem cell isolation and differentiation.
Collapse
Affiliation(s)
- Regiane M. C. Olimpio
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
- * E-mail:
| | - Miriane de Oliveira
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Maria T. De Sibio
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Fernanda C. F. Moretto
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Igor C. Deprá
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Lucas S. Mathias
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Bianca M. Gonçalves
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Bruna M. Rodrigues
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Helena P. Tilli
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Virgínia E. Coscrato
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Sarah M. B. Costa
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Gláucia M. F. S. Mazeto
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Célio J. C. Fernandes
- Institute of Biosciences, Department of Chemistry and Biochemistry, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Willian F. Zambuzzi
- Institute of Biosciences, Department of Chemistry and Biochemistry, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Patrícia P. Saraiva
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Durvanei A. Maria
- Biochemistry and Biophysics Laboratory, Butantan Institute, São Paulo, São Paulo, Brazil
| | - Célia R. Nogueira
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| |
Collapse
|
25
|
Wang YH, Wu JY, Kong SC, Chiang MH, Ho ML, Yeh ML, Chen CH. Low power laser irradiation and human adipose-derived stem cell treatments promote bone regeneration in critical-sized calvarial defects in rats. PLoS One 2018; 13:e0195337. [PMID: 29621288 PMCID: PMC5886537 DOI: 10.1371/journal.pone.0195337] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 03/20/2018] [Indexed: 01/18/2023] Open
Abstract
Both stem cell therapy and physical treatments have been shown to be beneficial in accelerating bone healing. However, the efficacy of combined treatment with stem cells and physical stimuli for large bone defects remains uncertain. The aim of this study was to evaluate the bone regeneration effects of low-power laser irradiation (LPLI) and human adipose-derived stem cell (ADSC) treatments during fracture repair using a comparative rat calvarial defect model. We evaluated the viability of human ADSCs, which were cultured on a porous PLGA scaffold using an MTS assay. The critical-sized calvarial bone defect rats were divided into 4 groups: control group, LPLI group, ADSC group, and ADSC+LPLI group. Bone formation was evaluated using micro-CT. New bone formation areas and osteogenic factor expression levels were then examined by histomorphological analysis and immunohistochemical staining. Our data showed that PLGA had no cytotoxic effect on human ADSCs. Micro-CT analyses revealed that both the LPLI and ADSC groups showed improved calvarial bone defect healing compared to the control group. In addition, the ADSC+LPLI group showed significantly increased bone volume at 16 weeks after surgery. The area of new bone formation ranked as follows: control group < LPLI group < ADSC group < ADSC+LPLI group. There were significant differences between the groups. In addition, both ADSC and ADSC+LPLI groups showed strong signals of vWF expression. ADSC and LPLI treatments improved fracture repair in critical-sized calvarial defects in rats. Importantly, the combined treatment of ADSCs and LPLI further enhances the bone healing process.
Collapse
Affiliation(s)
- Yan-Hsiung Wang
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jyun-Yi Wu
- Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Fresenius Kabi Taiwan Ltd, Taipei, Taiwan
| | - Su Chii Kong
- Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Min-Hsuan Chiang
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Mei-Ling Ho
- Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Ming-Long Yeh
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
- Medical Device Innovation Center, National Cheng Kung University, Tainan, Taiwan
| | - Chia-Hsin Chen
- Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Physical Medicine and Rehabilitation, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Physical Medicine and Rehabilitation, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- * E-mail:
| |
Collapse
|
26
|
Du Y, Ge J, Li Y, Ma PX, Lei B. Biomimetic elastomeric, conductive and biodegradable polycitrate-based nanocomposites for guiding myogenic differentiation and skeletal muscle regeneration. Biomaterials 2018; 157:40-50. [DOI: 10.1016/j.biomaterials.2017.12.005] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 12/05/2017] [Indexed: 02/08/2023]
|
27
|
Goodarzi P, Alavi-Moghadam S, Sarvari M, Tayanloo Beik A, Falahzadeh K, Aghayan H, Payab M, Larijani B, Gilany K, Rahim F, Adibi H, Arjmand B. Adipose Tissue-Derived Stromal Cells for Wound Healing. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1119:133-149. [PMID: 29858972 DOI: 10.1007/5584_2018_220] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Skin as the outer layer covers the body. Wounds can affect this vital organ negatively and disrupt its functions. Wound healing as a biological process is initiated immediately after an injury. This process consists of three stages: inflammation, proliferation, remodeling. Generally, these three stages occur continuously and timely. However, some factors such as infection, obesity and diabetes mellitus can interfere with these stages and impede the normal healing process which results in chronic wounds. Financial burden on both patients and health care systems, negative biologic effect on the patient's general health status and reduction in quality of life are a number of issues which make chronic wounds as a considerable challenge. During recent years, along with advances in the biomedical sciences, various surgical and non-surgical therapeutic methods have been suggested. All of these suggested treatments have their own advantages and disadvantages. Recently, cell-based therapies and regenerative medicine represent promising approaches to wound healing. Accordingly, several types of mesenchymal stem cells have been used in both preclinical and clinical settings for the treatment of wounds. Adipose-derived stromal cells are a cost-effective source of mesenchymal stem cells in wound management which can be easily harvest from adipose tissues through the less invasive processes with high yield rates. In addition, their ability to secrete multiple cytokines and growth factors, and differentiation into skin cells make them an ideal cell type to use in wound treatment. This is a concise overview on the application of adipose-derived stromal cells in wound healing and their role in the treatment of chronic wounds.
Collapse
Affiliation(s)
- Parisa Goodarzi
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Alavi-Moghadam
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Sarvari
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Akram Tayanloo Beik
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Khadijeh Falahzadeh
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamidreza Aghayan
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Moloud Payab
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Kambiz Gilany
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Fakher Rahim
- Health Research Institute, Thalassemia and Hemoglobinopathy Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hossein Adibi
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran. .,Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
28
|
Go and stop signals for glial regeneration. Curr Opin Neurobiol 2017; 47:182-187. [PMID: 29126016 PMCID: PMC6419527 DOI: 10.1016/j.conb.2017.10.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 08/16/2017] [Accepted: 10/10/2017] [Indexed: 12/22/2022]
Abstract
The regenerative response of ensheating glia to central nervous system (CNS) injury involves proliferation and differentiation, axonal re-enwrapment and some recovery of behaviour. Understanding this limited response could enable the enhancement of it. In Drosophila, the glial progenitor state is maintained by Notch, an activator of cell division and Prospero (Pros), a repressor. Injury provokes the activation of NFκB and up-regulation of Kon-tiki (Kon), driving cell proliferation. Homeostatic switch-off comes about as two negative feedback loops involving Pros terminate the response. Importantly, the functions of the kon and pros homologues NG2 and prox1, respectively, are conserved in mammalian NG2 glia. Controlling these genes is key for therapeutic manipulation of progenitors and stem cells to promote regeneration of the damaged CNS.
Collapse
|
29
|
La Gatta A, Ricci G, Stellavato A, Cammarota M, Filosa R, Papa A, D’Agostino A, Portaccio M, Delfino I, De Rosa M, Schiraldi C. Hyaluronan hydrogels with a low degree of modification as scaffolds for cartilage engineering. Int J Biol Macromol 2017; 103:978-989. [DOI: 10.1016/j.ijbiomac.2017.05.091] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 03/30/2017] [Accepted: 05/16/2017] [Indexed: 12/21/2022]
|
30
|
Bioinductive Scaffolds—Powerhouses of Skeletal Muscle Tissue Engineering. CURRENT PATHOBIOLOGY REPORTS 2017. [DOI: 10.1007/s40139-017-0151-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
31
|
Lepore M, Portaccio M, Delfino I, Sironi L, La Gatta A, D'Agostino A, Izzo E, Schiraldi C. Physico-optical properties of a crosslinked hyaluronic acid scaffold for biomedical applications. J Appl Polym Sci 2017. [DOI: 10.1002/app.45243] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Maria Lepore
- Dipartimento di Medicina Sperimentale; Università della Campania “Luigi Vanvitelli; Naples I-80123 Italy
| | - Marianna Portaccio
- Dipartimento di Medicina Sperimentale; Università della Campania “Luigi Vanvitelli; Naples I-80123 Italy
| | - Ines Delfino
- Dipartimento di Scienze Ecologiche e Biologiche; Università della Tuscia; Viterbo I-01100 Italy
| | - Laura Sironi
- Dipartimento di Fisica “G. Occhialini”; Università degli Studi di Milano-Bicocca; Milano I-20126 Italy
| | - Annalisa La Gatta
- Dipartimento di Medicina Sperimentale; Università della Campania “Luigi Vanvitelli; Naples I-80123 Italy
| | - Antonella D'Agostino
- Dipartimento di Medicina Sperimentale; Università della Campania “Luigi Vanvitelli; Naples I-80123 Italy
| | - E. Izzo
- Dipartimento di Medicina Sperimentale; Università della Campania “Luigi Vanvitelli; Naples I-80123 Italy
| | - Chiara Schiraldi
- Dipartimento di Medicina Sperimentale; Università della Campania “Luigi Vanvitelli; Naples I-80123 Italy
| |
Collapse
|
32
|
Barba M, Di Taranto G, Lattanzi W. Adipose-derived stem cell therapies for bone regeneration. Expert Opin Biol Ther 2017; 17:677-689. [PMID: 28374644 DOI: 10.1080/14712598.2017.1315403] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Cell-based therapies exploit the heterogeneous and self-sufficient biological environment of stem cells to restore, maintain and improve tissue functions. Adipose-derived stem cells (ASCs) are, to this aim, promising cell types thanks to advantageous isolation procedures, growth kinetics, plasticity and trophic properties. Specifically, bone regeneration represents a suitable, though often challenging, target setting to test and apply ASC-based therapeutic strategies. Areas covered: ASCs are extremely plastic and secrete bioactive peptides that mediate paracrine functions, mediating their trophic actions in vivo. Numerous preclinical studies demonstrated that ASCs improve bone healing. Clinical trials are ongoing to validate the clinical feasibility of these approaches. This review is intended to define the state-of-the-art on ASCs, encompassing the biological features that make them suitable for bone regenerative strategies, and to provide an update on existing preclinical and clinical applications. Expert opinion: ASCs offer numerous advantages over other stem cells in terms of feasibility of clinical translation. Data obtained from in vivo experimentation are encouraging, and clinical trials are ongoing. More robust validations are thus expected to be achieved during the next few years, and will likely pave the way to optimized patient-tailored treatments for bone regeneration.
Collapse
Affiliation(s)
- Marta Barba
- a Institute of Anatomy and Cell Biology , Università Cattolica del Sacro Cuore , Rome , Italy
| | - Giuseppe Di Taranto
- b Department of Plastic, Reconstructive and Aesthetic Surgery , University of Rome "Sapienza" , Policlinico Umberto I, Rome , Italy
| | - Wanda Lattanzi
- a Institute of Anatomy and Cell Biology , Università Cattolica del Sacro Cuore , Rome , Italy
| |
Collapse
|
33
|
Human adipose stem cell differentiation is highly affected by cancer cells both in vitro and in vivo: implication for autologous fat grafting. Cell Death Dis 2017; 8:e2568. [PMID: 28102844 PMCID: PMC5386348 DOI: 10.1038/cddis.2016.308] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 08/29/2016] [Accepted: 09/02/2016] [Indexed: 12/14/2022]
Abstract
Recent studies showed that mesenchymal stem cells derived from adipose tissue can promote tumour progression, raising some concerns regarding their use in regenerative medicine. In this context, we co-cultured either SAOS2 osteosarcoma or MCF7 breast cancer cells with human adipose stem cells (hASCs), in order to evaluate potential effects of cancer cells on hASCs differentiation, in vitro and in vivo. In this study we observed that both SAOS2 and MCF7 cell lines induced an increase in hASCs proliferation, compared to hASCs alone, but, surprisingly, neither changes in the expression of CD90, CD29, CD324 and vimentin, nor variations in the Twist and Slug mRNAs were detectable. Noteworthy, SAOS2 and MCF7 cells induced in hASCs an upregulation of CD34 expression and stemness genes, including OCT3/4, Nanog, Sox2 and leptin, and a decrease in angiogenic factors, including CD31, PDGFα, PDGFRα, PDGFRβ and VEGF. SMAD and pSMAD2/3 increased only in hASCs alone. After 21 days of co-culture, hASCs differentiated both in adipocytes and endothelial cells. Moreover, co-injection of MCF7 cells with hASCs led to the formation of a highly vascularized tumour. Taken together our findings suggest that mesenchymal stem cells, under tumour cell induction, do not differentiate in vitro or facilitate the angiogenesis of the tumour in vivo, thus opening interesting new scenarios in the relationship between cancer and stem cells. These findings may also lead to greater caution, when managing autologous fat grafts in cancer patients.
Collapse
|
34
|
Feng C, Hu J, Liu C, Liu S, Liao G, Song L, Zeng X. Association of 17-β Estradiol with Adipose-Derived Stem Cells: New Strategy to Produce Functional Myogenic Differentiated Cells with a Nano-Scaffold for Tissue Engineering. PLoS One 2016; 11:e0164918. [PMID: 27783699 PMCID: PMC5081199 DOI: 10.1371/journal.pone.0164918] [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: 07/26/2016] [Accepted: 10/03/2016] [Indexed: 01/01/2023] Open
Abstract
The increased incidence of stress urinary incontinence (SUI) in postmenopausal women has been proposed to be associated with a reduction in the level of 17-β estradiol (E2). E2 has also been shown to enhance the multi-differentiation ability of adipose-derived stem cells (ASCs) in vitro. However, studies on the potential value of E2 for tissue engineering in SUI treatment are rare. In the present study, we successfully fabricated myogenically differentiated ASCs (MD-ASCs), which were seeded onto a Poly(l-lactide)/Poly(e-caprolactone) electrospinning nano-scaffold, and incorporated E2 into the system, with the aim of improving the proliferation and myogenic differentiation of ASCs. ASCs were collected from the inguinal subcutaneous fat of rats. The proliferation and myogenic differentiation of ASCs, as well as the nano-scaffold biocompatibility of MD-ASCs, with or without E2 supplementation, were investigated. We demonstrated that E2 incorporation enhanced the proliferation of ASCs in vitro, and the most optimal concentration was 10-9 M. E2 also led to modulation of the MD-ASCs phenotype toward a concentrated type with smooth muscle-inductive medium. The expression of early (alpha-smooth muscle actin), mid (calponin), and late-stage (myosin heavy chain) contractile markers in MD-ASCs was enhanced by E2 during the different differentiation stages. Furthermore, the nano-scaffold was biocompatible with MD-ASCs, and cell proliferation was significantly enhanced by E2. Taken together, these results demonstrate that E2 can enhance the proliferation and myogenic differentiation of ASCs and can be used to construct a biocompatible cell/nano-scaffold. These scaffolds with desirable differentiation cells show promising applications for tissue engineering.
Collapse
Affiliation(s)
- Chunxiang Feng
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jinqian Hu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chang Liu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shiliang Liu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Guiying Liao
- School of Material Science and Chemistry Engineering, China University of Geosciences (Wuhan), Wuhan, Hubei, China
| | - Linjie Song
- School of Material Science and Chemistry Engineering, China University of Geosciences (Wuhan), Wuhan, Hubei, China
| | - Xiaoyong Zeng
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- * E-mail:
| |
Collapse
|
35
|
Hyaluronic Acid (HA) Scaffolds and Multipotent Stromal Cells (MSCs) in Regenerative Medicine. Stem Cell Rev Rep 2016; 12:664-681. [DOI: 10.1007/s12015-016-9684-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
36
|
Ostrovidov S, Shi X, Sadeghian RB, Salehi S, Fujie T, Bae H, Ramalingam M, Khademhosseini A. Stem Cell Differentiation Toward the Myogenic Lineage for Muscle Tissue Regeneration: A Focus on Muscular Dystrophy. Stem Cell Rev Rep 2016; 11:866-84. [PMID: 26323256 DOI: 10.1007/s12015-015-9618-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Skeletal muscle tissue engineering is one of the important ways for regenerating functionally defective muscles. Among the myopathies, the Duchenne muscular dystrophy (DMD) is a progressive disease due to mutations of the dystrophin gene leading to progressive myofiber degeneration with severe symptoms. Although current therapies in muscular dystrophy are still very challenging, important progress has been made in materials science and in cellular technologies with the use of stem cells. It is therefore useful to review these advances and the results obtained in a clinical point of view. This article focuses on the differentiation of stem cells into myoblasts, and their application in muscular dystrophy. After an overview of the different stem cells that can be induced to differentiate into the myogenic lineage, we introduce scaffolding materials used for muscular tissue engineering. We then described some widely used methods to differentiate different types of stem cell into myoblasts. We highlight recent insights obtained in therapies for muscular dystrophy. Finally, we conclude with a discussion on stem cell technology. We discussed in parallel the benefits brought by the evolution of the materials and by the expansion of cell sources which can differentiate into myoblasts. We also discussed on future challenges for clinical applications and how to accelerate the translation from the research to the clinic in the frame of DMD.
Collapse
Affiliation(s)
- Serge Ostrovidov
- WPI-Advanced Institute for Materials Research, Tohoku University, Sendai, 980-8577, Japan
| | - Xuetao Shi
- National Engineering Research Center for Tissue Restoration and Reconstruction & School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510640, People's Republic of China
| | - Ramin Banan Sadeghian
- WPI-Advanced Institute for Materials Research, Tohoku University, Sendai, 980-8577, Japan
| | - Sahar Salehi
- WPI-Advanced Institute for Materials Research, Tohoku University, Sendai, 980-8577, Japan
| | - Toshinori Fujie
- Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo, 162-8480, Japan
| | - Hojae Bae
- College of Animal Bioscience and Technology, Department of Bioindustrial Technologies, Konkuk University, Hwayang-dong, Kwangjin-gu, Seoul, 143-701, Republic of Korea
| | - Murugan Ramalingam
- WPI-Advanced Institute for Materials Research, Tohoku University, Sendai, 980-8577, Japan
- Christian Medical College Bagayam Campus, Centre for Stem Cell Research, Vellore, 632002, India
| | - Ali Khademhosseini
- WPI-Advanced Institute for Materials Research, Tohoku University, Sendai, 980-8577, Japan.
- College of Animal Bioscience and Technology, Department of Bioindustrial Technologies, Konkuk University, Hwayang-dong, Kwangjin-gu, Seoul, 143-701, Republic of Korea.
- Division of Biomedical Engineering, Department of Medicine, Harvard Medical School, Biomaterials Innovation Research Center, Brigham and Women's Hospital, Boston, MA, 02139, USA.
- Division of Health Sciences and Technology, Harvard-Massachusetts Institute of Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA.
- Department of Physics, King Abdulaziz University, Jeddah, 21569, Saudi Arabia.
| |
Collapse
|
37
|
Akpancar S, Tatar O, Turgut H, Akyildiz F, Ekinci S. The Current Perspectives of Stem Cell Therapy in Orthopedic Surgery. ARCHIVES OF TRAUMA RESEARCH 2016; 5:e37976. [PMID: 28144608 PMCID: PMC5253188 DOI: 10.5812/atr.37976] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 06/16/2016] [Accepted: 06/23/2016] [Indexed: 12/26/2022]
Abstract
CONTEXT Musculoskeletal injuries may be painful, troublesome, life limiting and also one of the global health problems. There has been considerable amount of interest during the past two decades to stem cells and tissue engineering techniques in orthopedic surgery, especially to manage special and compulsive injuries within the musculoskeletal system. EVIDENCE ACQUISITION The aim of this study was to present a literature review regarding the most recent progress in stem cell procedures and current indications in orthopedics clinical care practice. The Medline and PubMed library databases were searched for the articles related with stem cell procedures in the field of orthopedic surgery and additionally the reference list of each article was also included to provide a comprehensive evaluation. RESULTS Various sources of stem cells have been studied for orthopedics clinical care practice. Stem cell therapy has successfully used for major orthopedic procedures in terms of bone-joint injuries (fractures-bone defects, nonunion, and spinal injuries), osteoarthritis-cartilage defects, ligament-tendon injuries, femoral head osteonecrosis and osteogenesis imperfecta. Stem cells have also used in bone tissue engineering in combining with the scaffolds and provided faster and better healing of tissues. CONCLUSIONS Large amounts of preclinical studies have been made of stem cells and there is an increasing interest to perform these studies within the human population but preclinical studies are insufficient; therefore, much more and efficient studies should be conducted to evaluate the efficacy and safety of stem cells.
Collapse
Affiliation(s)
- Serkan Akpancar
- Department of Orthopedic Surgery, Gulhane Military Hospital, Ankara, Turkey
- Corresponding author: Serkan Akpancar, Department of Orthopedic Surgery, Gulhane Military Medicine Academy, Ankara, Turkey. Tel: +90-5443229700, Fax: +90-3124045500, E-mail:
| | - Oner Tatar
- Department of Orthopedic Surgery, Air Force Academy Kasımpaşa Military Hospital, Istanbul, Turkey
| | - Hasan Turgut
- Department of Orthopedic Surgery, Bursa Military Hospital, Bursa, Turkey
| | - Faruk Akyildiz
- Department of Orthopedic Surgery, Gulhane Military Hospital, Ankara, Turkey
| | - Safak Ekinci
- Department of Orthopedic Surgery, Agri Military Hospital, Agri, Turkey
| |
Collapse
|
38
|
Trumbull A, Subramanian G, Yildirim-Ayan E. Mechanoresponsive musculoskeletal tissue differentiation of adipose-derived stem cells. Biomed Eng Online 2016; 15:43. [PMID: 27103394 PMCID: PMC4840975 DOI: 10.1186/s12938-016-0150-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 03/24/2016] [Indexed: 02/06/2023] Open
Abstract
Musculoskeletal tissues are constantly under mechanical strains within their microenvironment. Yet, little is understood about the effect of in vivo mechanical milieu strains on cell development and function. Thus, this review article outlines the in vivo mechanical environment of bone, muscle, cartilage, tendon, and ligaments, and tabulates the mechanical strain and stress in these tissues during physiological condition, vigorous, and moderate activities. This review article further discusses the principles of mechanical loading platforms to create physiologically relevant mechanical milieu in vitro for musculoskeletal tissue regeneration. A special emphasis is placed on adipose-derived stem cells (ADSCs) as an emerging valuable tool for regenerative musculoskeletal tissue engineering, as they are easily isolated, expanded, and able to differentiate into any musculoskeletal tissue. Finally, it highlights the current state-of-the art in ADSCs-guided musculoskeletal tissue regeneration under mechanical loading.
Collapse
Affiliation(s)
- Andrew Trumbull
- Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH, 43606, USA
| | - Gayathri Subramanian
- Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH, 43606, USA
| | - Eda Yildirim-Ayan
- Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH, 43606, USA. .,Department of Orthopaedic Surgery, University of Toledo Medical Center, Toledo, OH, 43614, USA.
| |
Collapse
|
39
|
Advances in Adipose-Derived Stem Cells Isolation, Characterization, and Application in Regenerative Tissue Engineering. Stem Cells Int 2016. [PMID: 26981130 DOI: 10.1155/+2016/3206807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Obesity is a complex, multifactorial disease that has been extensively researched in recent times. Obesity is characterized by excess deposition of adipose tissue in response to surplus energy. Despite the negative connotations of adipose tissue (AT), it serves as a critical endocrine organ. Adipose tissue is a source of several adipokines and cytokines which have been deemed important for both normal metabolic function and disease formation. The discoveries of metabolically active brown AT in adult humans and adipose tissue derived stem cells (ADSC) have been key findings in the past decade with potential therapeutic implications. ADSCs represent an enticing pool of multipotent adult stem cells because of their noncontroversial nature, relative abundance, ease of isolation, and expandability. A decade and a half since the discovery of ADSCs, the scientific community is still working to uncover their therapeutic potential in a wide range of diseases. In this review, we provide an overview of the recent developments in the field of ADSCs and examine their potential use in transplantation and cell-based therapies for the regeneration of diseased organs and systems. We also hope to provide perspective on how to best utilize this readily available, powerful pool of stem cells in the future.
Collapse
|
40
|
Adipose-Derived Stem Cells for Tissue Engineering and Regenerative Medicine Applications. Stem Cells Int 2016; 2016:6737345. [PMID: 27057174 PMCID: PMC4761677 DOI: 10.1155/2016/6737345] [Citation(s) in RCA: 179] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 01/02/2016] [Accepted: 01/03/2016] [Indexed: 02/05/2023] Open
Abstract
Adipose-derived stem cells (ASCs) are a mesenchymal stem cell source with properties of self-renewal and multipotential differentiation. Compared to bone marrow-derived stem cells (BMSCs), ASCs can be derived from more sources and are harvested more easily. Three-dimensional (3D) tissue engineering scaffolds are better able to mimic the in vivo cellular microenvironment, which benefits the localization, attachment, proliferation, and differentiation of ASCs. Therefore, tissue-engineered ASCs are recognized as an attractive substitute for tissue and organ transplantation. In this paper, we review the characteristics of ASCs, as well as the biomaterials and tissue engineering methods used to proliferate and differentiate ASCs in a 3D environment. Clinical applications of tissue-engineered ASCs are also discussed to reveal the potential and feasibility of using tissue-engineered ASCs in regenerative medicine.
Collapse
|
41
|
Advances in Adipose-Derived Stem Cells Isolation, Characterization, and Application in Regenerative Tissue Engineering. Stem Cells Int 2016; 2016:3206807. [PMID: 26981130 PMCID: PMC4766348 DOI: 10.1155/2016/3206807] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 12/14/2015] [Accepted: 01/14/2016] [Indexed: 12/22/2022] Open
Abstract
Obesity is a complex, multifactorial disease that has been extensively researched in recent times. Obesity is characterized by excess deposition of adipose tissue in response to surplus energy. Despite the negative connotations of adipose tissue (AT), it serves as a critical endocrine organ. Adipose tissue is a source of several adipokines and cytokines which have been deemed important for both normal metabolic function and disease formation. The discoveries of metabolically active brown AT in adult humans and adipose tissue derived stem cells (ADSC) have been key findings in the past decade with potential therapeutic implications. ADSCs represent an enticing pool of multipotent adult stem cells because of their noncontroversial nature, relative abundance, ease of isolation, and expandability. A decade and a half since the discovery of ADSCs, the scientific community is still working to uncover their therapeutic potential in a wide range of diseases. In this review, we provide an overview of the recent developments in the field of ADSCs and examine their potential use in transplantation and cell-based therapies for the regeneration of diseased organs and systems. We also hope to provide perspective on how to best utilize this readily available, powerful pool of stem cells in the future.
Collapse
|
42
|
Development of Synthetic and Natural Materials for Tissue Engineering Applications Using Adipose Stem Cells. Stem Cells Int 2016; 2016:5786257. [PMID: 26977158 PMCID: PMC4764745 DOI: 10.1155/2016/5786257] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 01/09/2016] [Accepted: 01/12/2016] [Indexed: 12/12/2022] Open
Abstract
Adipose stem cells have prominent implications in tissue regeneration due to their abundance and relative ease of harvest from adipose tissue and their abilities to differentiate into mature cells of various tissue lineages and secrete various growth cytokines. Development of tissue engineering techniques in combination with various carrier scaffolds and adipose stem cells offers great potential in overcoming the existing limitations constraining classical approaches used in plastic and reconstructive surgery. However, as most tissue engineering techniques are new and highly experimental, there are still many practical challenges that must be overcome before laboratory research can lead to large-scale clinical applications. Tissue engineering is currently a growing field of medical research; in this review, we will discuss the progress in research on biomaterials and scaffolds for tissue engineering applications using adipose stem cells.
Collapse
|
43
|
Vériter S, André W, Aouassar N, Poirel HA, Lafosse A, Docquier PL, Dufrane D. Human Adipose-Derived Mesenchymal Stem Cells in Cell Therapy: Safety and Feasibility in Different "Hospital Exemption" Clinical Applications. PLoS One 2015; 10:e0139566. [PMID: 26485394 PMCID: PMC4615620 DOI: 10.1371/journal.pone.0139566] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 09/15/2015] [Indexed: 02/07/2023] Open
Abstract
Based on immunomodulatory, osteogenic, and pro-angiogenic properties of adipose-derived stem cells (ASCs), this study aims to assess the safety and efficacy of ASC-derived cell therapies for clinical indications. Two autologous ASC-derived products were proposed to 17 patients who had not experienced any success with conventional therapies: (1) a scaffold-free osteogenic three-dimensional graft for the treatment of bone non-union and (2) a biological dressing for dermal reconstruction of non-healing chronic wounds. Safety was studied using the quality control of the final product (genetic stability, microbiological/mycoplasma/endotoxin contamination) and the in vivo evaluation of adverse events after transplantation. Feasibility was assessed by the ability to reproducibly obtain the final ASC-based product with specific characteristics, the time necessary for graft manufacturing, the capacity to produce enough material to treat the lesion, the surgical handling of the graft, and the ability to manufacture the graft in line with hospital exemption regulations. For 16 patients (one patient did not undergo grafting because of spontaneous bone healing), in-process controls found no microbiological/mycoplasma/endotoxin contamination, no obvious deleterious genomic anomalies, and optimal ASC purity. Each type of graft was reproducibly obtained without significant delay for implantation and surgical handling was always according to the surgical procedure and the implantation site. No serious adverse events were noted for up to 54 months. We demonstrated that autologous ASC transplantation can be considered a safe and feasible therapy tool for extreme clinical indications of ASC properties and physiopathology of disease.
Collapse
Affiliation(s)
- Sophie Vériter
- Endocrine Cell Therapy, Centre of Tissular and Cellular Therapy, Cliniques Universitaires Saint Luc, Brussels, Belgium
| | - Wivine André
- Endocrine Cell Therapy, Centre of Tissular and Cellular Therapy, Cliniques Universitaires Saint Luc, Brussels, Belgium
| | - Najima Aouassar
- Endocrine Cell Therapy, Centre of Tissular and Cellular Therapy, Cliniques Universitaires Saint Luc, Brussels, Belgium
| | - Hélène Antoine Poirel
- Center for Human Genetics, Cliniques Universitaires Saint Luc, Université catholique de Louvain, Brussels, Belgium
| | - Aurore Lafosse
- Plastic Surgery, Cliniques Universitaires Saint Luc, Brussels, Belgium
| | | | - Denis Dufrane
- Endocrine Cell Therapy, Centre of Tissular and Cellular Therapy, Cliniques Universitaires Saint Luc, Brussels, Belgium
- * E-mail:
| |
Collapse
|
44
|
Xu Y, Li Z, Li X, Fan Z, Liu Z, Xie X, Guan J. Regulating myogenic differentiation of mesenchymal stem cells using thermosensitive hydrogels. Acta Biomater 2015; 26:23-33. [PMID: 26277379 DOI: 10.1016/j.actbio.2015.08.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 07/30/2015] [Accepted: 08/11/2015] [Indexed: 01/02/2023]
Abstract
Stem cell therapy has potential to regenerate skeletal muscle tissue in ischemic limb. However, the delivered stem cells experience low rate of myogenic differentiation. Employing injectable hydrogels as stem cell carriers may enhance the myogenic differentiation as their modulus may be tailored to induce the differentiation. Yet current approaches used to manipulate hydrogel modulus often simultaneously vary other properties that also affect stem cell differentiation, such as chemical structure, composition and water content. Thus it is challenging to demonstrate the decoupled effect of hydrogel modulus on stem cell differentiation. In this report, we decoupled the hydrogel modulus from chemical structure, composition, and water content using injectable and thermosensitive hydrogels. The hydrogels were synthesized from N-isopropylacrylamide (NIPAAm), acrylic acid (AAc), and degradable macromer 2-hydroxyethyl methacrylate-oligomer [oligolatide, oligohydroxybutyrate, or oligo(trimethylene carbonate)]. We found that using the same monomer composition and oligomer chemical structure but different oligomer length can independently vary hydrogel modulus. Rat bone marrow mesenchymal stem cells (MSCs) were encapsulated in the hydrogels with elastic expansion moduli of 11, 20, and 40 kPa, respectively. After 14 days of culture, significant myogenic differentiation was achieved for the hydrogel with elastic expansion modulus of 20 kPa, as judged from both the gene and protein expression. In addition, MSCs exhibited an elastic expansion modulus-dependent proliferation rate. The most significant proliferation was observed in the hydrogel with elastic expansion modulus of 40 kPa. These results demonstrate that the developed injectable and thermosensitive hydrogels with suitable modulus has the potential to deliver stem cells into ischemic limb for enhanced myogenic differentiation and muscle regeneration. STATEMENT OF SIGNIFICANCE Stem cell therapy for skeletal muscle regeneration in ischemic limb experiences low rate of myogenic differentiation. Employing injectable hydrogels as stem cell carriers may enhance the myogenic differentiation as hydrogel modulus may be modulated to induce the differentiation. Yet current approaches used to modulate hydrogel modulus may simultaneously vary other properties that also affect stem cell myogenic differentiation, such as chemistry, composition and water content. In this report, we decoupled the hydrogel modulus from chemistry, composition, and water content using injectable and thermosensitive hydrogels. We found that mesenchymal stem cells best differentiated into myogenic lineage in the hydrogel with elastic modulus of 20 kPa.
Collapse
Affiliation(s)
- Yanyi Xu
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, United States
| | - Zhenqing Li
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, United States
| | - Xiaofei Li
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, United States
| | - Zhaobo Fan
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, United States
| | - Zhenguo Liu
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, United States
| | - Xiaoyun Xie
- Department of Gerontology, Tongji Hospital, Tongji University, Shanghai, China
| | - Jianjun Guan
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, United States; Tongji Hospital, Tongji University, Shanghai, China.
| |
Collapse
|
45
|
Abstract
We have witnessed a rapid expansion of in vitro characterization and differentiation of adipose-derived stem cells, with increasing translation to both in vivo models and a breadth of clinical specialties. However, an appreciation of the truly heterogeneous nature of this unique stem cell group has identified a need to more accurately delineate subpopulations by any of a host of methods, to include functional properties or surface marker expression. Cells selected for improved proliferative, differentiative, angiogenic or ischemia-resistant properties are but a few attributes that could prove beneficial for targeted treatments or therapies. Optimizing cell culture conditions to permit re-introduction to patients is critical for clinical translation.
Collapse
Affiliation(s)
- Kavan S Johal
- Blond McIndoe Laboratories, Institute of Inflammation & Repair, School of Medicine, University of Manchester, M13 9PT, UK
| | | | | |
Collapse
|
46
|
Shang X, Luo Z, Wang X, Jaeblon T, Marymont JV, Dong Y. Deletion of RBPJK in Mesenchymal Stem Cells Enhances Osteogenic Activity by Up-Regulation of BMP Signaling. PLoS One 2015; 10:e0135971. [PMID: 26285013 PMCID: PMC4540435 DOI: 10.1371/journal.pone.0135971] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 07/28/2015] [Indexed: 11/18/2022] Open
Abstract
Recently we have demonstrated the importance of RBPjk-dependent Notch signaling in the regulation of mesenchymal stem cell (MSC) differentiation during skeletogenesis both in vivo and in vitro. Here we further performed RBPJK loss-of-function experiments to demonstrate for the first time that RBPJK deficient MSC shows enhanced differentiation and osteogenesis acts via up-regulation of the BMP signaling. In the present study, we first compared the spontaneous and osteogenic differentiation in normal and recombination signal binding protein for immunoglobulin kappa J region (RBPJK) deficient human bone marrow-derived mesenchymal stem cells (MSCs). It was found that RBPJK highly expressed in fresh isolated MSCs and its expression was progressing down-regulated during spontaneous differentiation and even greater in osteogenic media inducted differentiation. Deletion of RBPJK in MSCs not only enhances cell spontaneous differentiation, but also significantly accelerates condition media inducted osteogenic differentiation by showing enhanced alkaline phosphatase (ALP) activity, Alizarin red staining, gene expression of Runx2, Osteopontin (OPN), Type I collagen (COL1a1) in culture. Additionally, BMP signaling responsive reporter activity and phosphor-smad1/5/8 expression were also significantly increased upon removal of RBPJK in MSCs. These data proved that inhibition of Notch signaling in MSCs promotes cell osteogenic differentiation by up-regulation of BMP signaling, and RBPJK deficient MSC maybe a better cell population for cell-based bone tissue engineering.
Collapse
Affiliation(s)
- Xifu Shang
- Department of Orthopaedic Surgery, Anhui Provincial Hospital, Hefei, Anhui, China
| | - Zhengliang Luo
- Department of Orthopaedic Surgery, Anhui Provincial Hospital, Hefei, Anhui, China
| | - Xudong Wang
- Department of Oral and Craniomaxillofacial Surgery, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Todd Jaeblon
- Department of Orthopaedic Surgery, Louisiana State University Health Sciences Center, Shreveport, LA, United States of America
| | - John V. Marymont
- Department of Orthopaedic Surgery, Louisiana State University Health Sciences Center, Shreveport, LA, United States of America
| | - Yufeng Dong
- Department of Orthopaedic Surgery, Louisiana State University Health Sciences Center, Shreveport, LA, United States of America
- * E-mail:
| |
Collapse
|
47
|
Zavan B, De Francesco F, D'Andrea F, Ferroni L, Gardin C, Salzillo R, Nicoletti G, Ferraro GA. Persistence of CD34 Stem Marker in Human Lipoma: Searching for Cancer Stem Cells. Int J Biol Sci 2015; 11:1127-39. [PMID: 26327807 PMCID: PMC4551749 DOI: 10.7150/ijbs.11946] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 05/05/2015] [Indexed: 01/19/2023] Open
Abstract
Background: Lipomas are benign solid tumours that develop in soft tissues with origin in mesenchymal progenitors. Macroscopically, they appear as soft-elastic nodules, varying in volume from a few millimiters to several centimetres and can enlarge progressively. Although they are usually asymptomatic, they can cause symptoms due to nerve or vessel compression. Microscopically they appear as fibrous connective tissue stroma with embedded adipocytes, and absence of inflammation. Up to now no characterisation of stem cell population present in this tissue has been performed. Methods: Cytofluorimetric, biological and molecular biology analyses have been performed in order to test superficial cell markers and gene expression profile related to stemness and apoptotic activity of cells present in lipoma tissues compared to those of adipose tissue's cells. Results: Our results confirmed that CD34+ cells in lipoma were present around small adipocytes, showing several altered biological activity such as proliferation, apoptotis and stemness. Conclusions: The data emerging from the comparison of the lipoma cells and normal adipose tissue, suggests the presence of cell precursors involved in the development of the lipoma. This hypothesis requires further investigation and may indicate new thresholds in the study of benign tumour pathogenesis.
Collapse
Affiliation(s)
- Barbara Zavan
- 2. Department of Biomedical Sciences, University of Padua, Ugo Bassi, 58/B, 35131, Padua (Italy)
| | - Francesco De Francesco
- 1. Multidisciplinary Department of Medical-Surgical and Dental Specialties, Second University of Naples, L. De Crecchio, 6, 80138, Naples (Italy)
| | - Francesco D'Andrea
- 1. Multidisciplinary Department of Medical-Surgical and Dental Specialties, Second University of Naples, L. De Crecchio, 6, 80138, Naples (Italy)
| | - Letizia Ferroni
- 2. Department of Biomedical Sciences, University of Padua, Ugo Bassi, 58/B, 35131, Padua (Italy)
| | - Chiara Gardin
- 2. Department of Biomedical Sciences, University of Padua, Ugo Bassi, 58/B, 35131, Padua (Italy)
| | - Rosa Salzillo
- 1. Multidisciplinary Department of Medical-Surgical and Dental Specialties, Second University of Naples, L. De Crecchio, 6, 80138, Naples (Italy)
| | - Gianfranco Nicoletti
- 1. Multidisciplinary Department of Medical-Surgical and Dental Specialties, Second University of Naples, L. De Crecchio, 6, 80138, Naples (Italy)
| | - Giuseppe A Ferraro
- 1. Multidisciplinary Department of Medical-Surgical and Dental Specialties, Second University of Naples, L. De Crecchio, 6, 80138, Naples (Italy)
| |
Collapse
|
48
|
De Francesco F, Ricci G, D'Andrea F, Nicoletti GF, Ferraro GA. Human Adipose Stem Cells: From Bench to Bedside. TISSUE ENGINEERING PART B-REVIEWS 2015; 21:572-84. [PMID: 25953464 DOI: 10.1089/ten.teb.2014.0608] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Stem cell-based therapies for repair and regeneration of different tissues are becoming more important in the treatment of several diseases. Adult stem cells currently symbolize the most available source of cell progenitors for tissue engineering and repair and can be harvested using minimally invasive procedures. Moreover, mesenchymal stem cells (MSCs), the most widely used stem cells in stem cell-based therapies, are multipotent progenitors, with capability to differentiate into cartilage, bone, connective, muscle, and adipose tissue. So far, bone marrow has been regarded as the main source of MSCs. To date, human adult adipose tissue may be the best suitable alternative source of MSCs. Adipose stem cells (ASCs) can be largely extracted from subcutaneous human adult adipose tissue. A large number of studies show that adipose tissue contains a biologically and clinically interesting heterogeneous cell population called stromal vascular fraction (SVF). The SVF may be employed directly or cultured for selection and expansion of an adherent population, so called adipose-derived stem cells (ASCs). In recent years, literature based on data related to SVF cells and ASCs has augmented considerably: These studies have demonstrated the efficacy and safety of SVF cells and ASCs in vivo in animal models. On the basis of these observations, in several countries, various clinical trials involving SVF cells and ASCs have been permitted. This review aims at summarizing data regarding either ASCs cellular biology or ASCs-based clinical trials and at discussing the possible future clinical translation of ASCs and their potentiality in cell-based tissue engineering.
Collapse
Affiliation(s)
- Francesco De Francesco
- 1 Multidisciplinary Department of Medical-Surgical and Dental Specialties, Second University of Naples , Naples, Italy
| | - Giulia Ricci
- 2 Department of Experimental Medicine, Second University of Naples , Naples, Italy
| | - Francesco D'Andrea
- 1 Multidisciplinary Department of Medical-Surgical and Dental Specialties, Second University of Naples , Naples, Italy
| | - Giovanni Francesco Nicoletti
- 1 Multidisciplinary Department of Medical-Surgical and Dental Specialties, Second University of Naples , Naples, Italy
| | - Giuseppe Andrea Ferraro
- 1 Multidisciplinary Department of Medical-Surgical and Dental Specialties, Second University of Naples , Naples, Italy
| |
Collapse
|
49
|
Tijore A, Wen F, Lam CRI, Tay CY, Tan LP. Modulating human mesenchymal stem cell plasticity using micropatterning technique. PLoS One 2014; 9:e113043. [PMID: 25401734 PMCID: PMC4234627 DOI: 10.1371/journal.pone.0113043] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 10/18/2014] [Indexed: 12/21/2022] Open
Abstract
In our previous work, we have reported that enforced elongation of human mesenchymal stem cells (hMSCs) through micropatterning promoted their myocardial lineage commitment. However, whether this approach is robust enough to retain the commitment when subsequently subjected to different conditions remains unsolved. This de-differentiation, if any, would have significant implication on the application of these myocardial-like hMSCs either as tissue engineered product or in stem cell therapy. Herein, we investigated the robustness of micropatterning induced differentiation by evaluating the retention of myocardial differentiation in patterned hMSCs when challenged with non-myocardial differentiation cues. Altogether, we designed four groups of experiments; 1) Patterned hMSCs cultured in normal growth medium serving as a positive control; 2) Patterned hMSCs cultured in normal growth medium for 14 days followed by osteogenic and adipogenic media for next 7 days (to study the robustness of the effect of micropatterning); 3) Patterned hMSCs (initially grown in normal growth medium for 14 days) trypsinized and recultured in different induction media for next 7 days (to study the robustness of the effect of micropatterning without any shape constrain) and 4) Patterned hMSCs cultured in osteogenic and adipogenic media for 14 days (to study the effects of biochemical cues versus biophysical cues). It was found that hMSCs that were primed to commit to myocardial lineage (Groups 2 and 3) were able to maintain myocardial lineage commitment despite subsequent culturing in osteogenic and adipogenic media. However, for hMSCs that were not primed (Group 4), the biochemical cues seem to dominate over the biophysical cue in modulating hMSCs differentiation. It demonstrates that cell shape modulation is not only capable of inducing stem cell differentiation but also ensuring the permanent lineage commitment.
Collapse
Affiliation(s)
- Ajay Tijore
- Division of Materials Technology, School of Materials Science and Engineering, Nanyang Technological University, Singapore, Singapore
| | - Feng Wen
- Division of Materials Technology, School of Materials Science and Engineering, Nanyang Technological University, Singapore, Singapore
| | - Chee Ren Ivan Lam
- Division of Materials Technology, School of Materials Science and Engineering, Nanyang Technological University, Singapore, Singapore
| | - Chor Yong Tay
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Singapore, Singapore
| | - Lay Poh Tan
- Division of Materials Technology, School of Materials Science and Engineering, Nanyang Technological University, Singapore, Singapore
- * E-mail:
| |
Collapse
|
50
|
Dossena M, Bedini G, Rusmini P, Giorgetti E, Canazza A, Tosetti V, Salsano E, Sagnelli A, Mariotti C, Gellera C, Navone SE, Marfia G, Alessandri G, Corsi F, Parati EA, Pareyson D, Poletti A. Human adipose-derived mesenchymal stem cells as a new model of spinal and bulbar muscular atrophy. PLoS One 2014; 9:e112746. [PMID: 25392924 PMCID: PMC4231043 DOI: 10.1371/journal.pone.0112746] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 10/13/2014] [Indexed: 01/08/2023] Open
Abstract
Spinal and bulbar muscular atrophy (SBMA) or Kennedy's disease is an X-linked CAG/polyglutamine expansion motoneuron disease, in which an elongated polyglutamine tract (polyQ) in the N-terminal androgen receptor (ARpolyQ) confers toxicity to this protein. Typical markers of SBMA disease are ARpolyQ intranuclear inclusions. These are generated after the ARpolyQ binds to its endogenous ligands, which promotes AR release from chaperones, activation and nuclear translocation, but also cell toxicity. The SBMA mouse models developed so far, and used in preclinical studies, all contain an expanded CAG repeat significantly longer than that of SBMA patients. Here, we propose the use of SBMA patients adipose-derived mesenchymal stem cells (MSCs) as a new human in vitro model to study ARpolyQ toxicity. These cells have the advantage to express only ARpolyQ, and not the wild type AR allele. Therefore, we isolated and characterized adipose-derived MSCs from three SBMA patients (ADSC from Kennedy's patients, ADSCK) and three control volunteers (ADSCs). We found that both ADSCs and ADSCKs express mesenchymal antigens, even if only ADSCs can differentiate into the three typical cell lineages (adipocytes, chondrocytes and osteocytes), whereas ADSCKs, from SBMA patients, showed a lower growth potential and differentiated only into adipocyte. Moreover, analysing AR expression on our mesenchymal cultures we found lower levels in all ADSCKs than ADSCs, possibly related to negative pressures exerted by toxic ARpolyQ in ADSCKs. In addition, with proteasome inhibition the ARpolyQ levels increased specifically in ADSCKs, inducing the formation of HSP70 and ubiquitin positive nuclear ARpolyQ inclusions. Considering all of this evidence, SBMA patients adipose-derived MSCs cultures should be considered an innovative in vitro human model to understand the molecular mechanisms of ARpolyQ toxicity and to test novel therapeutic approaches in SBMA.
Collapse
Affiliation(s)
- Marta Dossena
- Cellular Neurobiology Laboratory, Unit of Cerebrovascular Disease, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Gloria Bedini
- Cellular Neurobiology Laboratory, Unit of Cerebrovascular Disease, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Paola Rusmini
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Centro Interdipartimentale sulle Malattie Neurodegenerative, Università degli Studi di Milano, Milan, Italy
| | - Elisa Giorgetti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Centro Interdipartimentale sulle Malattie Neurodegenerative, Università degli Studi di Milano, Milan, Italy
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, 48109, United States of America
| | - Alessandra Canazza
- Cellular Neurobiology Laboratory, Unit of Cerebrovascular Disease, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Valentina Tosetti
- Cellular Neurobiology Laboratory, Unit of Cerebrovascular Disease, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Ettore Salsano
- Clinic of Central and Peripheral Degenerative Neuropathies Unit, Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Anna Sagnelli
- Clinic of Central and Peripheral Degenerative Neuropathies Unit, Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Caterina Mariotti
- Unit of Genetics of Neurodegenerative and Metabolic Diseases, Department of Diagnostic and Applied Technology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Cinzia Gellera
- Unit of Genetics of Neurodegenerative and Metabolic Diseases, Department of Diagnostic and Applied Technology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Stefania Elena Navone
- Cellular Neurobiology Laboratory, Unit of Cerebrovascular Disease, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Giovanni Marfia
- Cellular Neurobiology Laboratory, Unit of Cerebrovascular Disease, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Giulio Alessandri
- Cellular Neurobiology Laboratory, Unit of Cerebrovascular Disease, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Fabio Corsi
- Surgery Division, Department of Clinical Sciences, University of Milan, “Luigi Sacco” Hospital, Milan, Italy
| | - Eugenio Agostino Parati
- Cellular Neurobiology Laboratory, Unit of Cerebrovascular Disease, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Davide Pareyson
- Clinic of Central and Peripheral Degenerative Neuropathies Unit, Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
- * E-mail: (DP); (AP)
| | - Angelo Poletti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Centro Interdipartimentale sulle Malattie Neurodegenerative, Università degli Studi di Milano, Milan, Italy
- * E-mail: (DP); (AP)
| |
Collapse
|