1
|
Cai Z, Li Z, Wei Q, Yang F, Li T, Ke C, He Y, Wang J, Ni B, Lin M, Li L. MiR-24-3p regulates the differentiation of adipose-derived stem cells toward pericytes and promotes fat grafting vascularization. FASEB J 2023; 37:e22935. [PMID: 37086094 DOI: 10.1096/fj.202202037rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 03/29/2023] [Accepted: 04/11/2023] [Indexed: 04/23/2023]
Abstract
Adipose-derived stem cells (ADSCs) enhance fat graft survival by promoting neovascularization. The mechanism that promotes ADSCs differentiation toward pericytes was not known. We treated ADSCs with conditional medium (CM) from endothelial cells (ECs) or human recombinant transforming growth factor β (TGF-β) to induce differentiation into pericytes. Pericytes markers, including platelet-derived growth factor receptor β (PDGFRβ), alpha-smooth muscle actin (α-SMA), and desmin, were examined. Pericytes differentiation markers, migration, and their association with ECs were examined in ADSCs transfected with miR-24-3p mimics and inhibitors. Bioinformatics target prediction platforms and luciferase assays were used to investigate whether PDGFRβ was directly targeted by miR-24-3p. In vivo, fat mixed with ADSCs transfected with miR-24-3p mimics or inhibitors was implanted subcutaneously on the lower back region of nude mice. Fat grafts were harvested and analyzed at 2, 4, 6, and 8 weeks. Results showed that endogenous TGF-β derived from CM from EC or human recombinant TGF-β promoted migration, association with ECs, and induced expression of pericyte markers (PDGFRβ, α-SMA, Desmin) in ADSCs. MiR-24-3p directly targeted PDGFRβ in ADSCs by lucifer reporter assays. Inhibition of miR-24-3p promoted pericytes differentiation, migration, and association with ECs in ADSCs. Inhibition of miR-24-3p in ADSCs promoted survival, integrity, adipocyte viability, vascularization, pericytes association with ECs, and reduced fibrosis, whereas overexpression of miR-24-3p in ADSCs yielded the opposite results. Collectively, TGF-β released by ECs induced ADSCs differentiation toward pericytes through miR-24-3p. Downregulation of miR-24-3p in ADSCs induced survival, integrity, adipocyte viability, vascularization, pericytes association with ECs, and reduced fibrosis after fat grafting.
Collapse
Affiliation(s)
- Zhongming Cai
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Zihao Li
- Department of First Clinical Medical School, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Qing Wei
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Fangfang Yang
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Tian Li
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Chen Ke
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Yucang He
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Jingping Wang
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Binting Ni
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Ming Lin
- Department of Obstetrics and Gynecology, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Liqun Li
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| |
Collapse
|
2
|
Anudeep TC, Jeyaraman M, Muthu S, Rajendran RL, Gangadaran P, Mishra PC, Sharma S, Jha SK, Ahn BC. Advancing Regenerative Cellular Therapies in Non-Scarring Alopecia. Pharmaceutics 2022; 14:pharmaceutics14030612. [PMID: 35335987 PMCID: PMC8953616 DOI: 10.3390/pharmaceutics14030612] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/28/2022] [Accepted: 03/07/2022] [Indexed: 02/05/2023] Open
Abstract
Alopecia or baldness is a common diagnosis in clinical practice. Alopecia can be scarring or non-scarring, diffuse or patchy. The most prevalent type of alopecia is non-scarring alopecia, with the majority of cases being androgenetic alopecia (AGA) or alopecia areata (AA). AGA is traditionally treated with minoxidil and finasteride, while AA is treated with immune modulators; however, both treatments have significant downsides. These drawbacks compel us to explore regenerative therapies that are relatively devoid of adverse effects. A thorough literature review was conducted to explore the existing proven and experimental regenerative treatment modalities in non-scarring alopecia. Multiple treatment options compelled us to classify them into growth factor-rich and stem cell-rich. The growth factor-rich group included platelet-rich plasma, stem cell-conditioned medium, exosomes and placental extract whereas adult stem cells (adipose-derived stem cell-nano fat and stromal vascular fraction; bone marrow stem cell and hair follicle stem cells) and perinatal stem cells (umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs), Wharton jelly-derived MSCs (WJ-MSCs), amniotic fluid-derived MSCs (AF-MSCs), and placental MSCs) were grouped into the stem cell-rich group. Because of its regenerative and proliferative capabilities, MSC lies at the heart of regenerative cellular treatment for hair restoration. A literature review revealed that both adult and perinatal MSCs are successful as a mesotherapy for hair regrowth. However, there is a lack of standardization in terms of preparation, dose, and route of administration. To better understand the source and mode of action of regenerative cellular therapies in hair restoration, we have proposed the "À La Mode Classification". In addition, available evidence-based cellular treatments for hair regrowth have been thoroughly described.
Collapse
Affiliation(s)
- Talagavadi Channaiah Anudeep
- Department of Plastic Surgery, Topiwala National Medical College and BYL Nair Ch. Hospital, Mumbai 400008, India;
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, India; (M.J.); (S.M.); (S.K.J.)
- À La Mode Esthétique Studio, Mysuru 570011, India
- International Association of Stem Cell and Regenerative Medicine (IASRM), New Delhi 110092, India; (P.C.M.); (S.S.)
| | - Madhan Jeyaraman
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, India; (M.J.); (S.M.); (S.K.J.)
- International Association of Stem Cell and Regenerative Medicine (IASRM), New Delhi 110092, India; (P.C.M.); (S.S.)
- Department of Orthopaedics, Faculty of Medicine—Sri Lalithambigai Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai 600095, India
| | - Sathish Muthu
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, India; (M.J.); (S.M.); (S.K.J.)
- International Association of Stem Cell and Regenerative Medicine (IASRM), New Delhi 110092, India; (P.C.M.); (S.S.)
- Department of Orthopaedics, Government Medical College and Hospital, Dindigul 624304, India
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea;
| | - Prakash Gangadaran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea;
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- Correspondence: (P.G.); (B.-C.A.)
| | - Prabhu Chandra Mishra
- International Association of Stem Cell and Regenerative Medicine (IASRM), New Delhi 110092, India; (P.C.M.); (S.S.)
| | - Shilpa Sharma
- International Association of Stem Cell and Regenerative Medicine (IASRM), New Delhi 110092, India; (P.C.M.); (S.S.)
- Department of Paediatric Surgery, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, India; (M.J.); (S.M.); (S.K.J.)
- International Association of Stem Cell and Regenerative Medicine (IASRM), New Delhi 110092, India; (P.C.M.); (S.S.)
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea;
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- Correspondence: (P.G.); (B.-C.A.)
| |
Collapse
|
3
|
Jeyaraman M, Muthu S, Jain R, Satish AS, Garg N, Mishra PC, Swati K, Parkash A, Jha NK, Ojha S, Roychoudhury S, Kumar D, Ruokolainen J, Kamal MA, Kesari KK, Jha SK. Total Stromal Fraction (TSF) - Fortified Adipose Tissue-Derived Stem
Cells Source: An Emerging Regenerative Realm Against COVID-19 Induced
Pulmonary Compromise. CORONAVIRUSES 2022. [DOI: 10.2174/2666796702666210908151708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
:
The inception of the COVID-19 pandemic has jeopardized humanity with markedly dampening
of worldwide resources. The viral infection may present with varying signs and symptoms,
imitating pneumonia and seasonal flu. With a gradual course, this may progress and result in the
deadliest state of acute respiratory distress syndrome (ARDS) and acute lung injury (ALI). Moreover,
following recovery from the severe brunt of COVID-19 infection, interstitial portions of alveoli
have been found to undergo residual scarring and further to have compromised air exchange.
Such alterations in the lung microenvironment and associated systemic manifestations have been
recognized to occur due to the extensive release of cytokines. The mortality rate increases with advancing
age and in individuals with underlying co-morbidity. Presently, there is no availability of
specific antiviral therapy or any other definitive modality to counter this progressive worsening.
However, we believe principles and advancing cell-based therapy may prove fruitful in subjugating
such reported worsening in these patients. This article reviews eminent knowledge and relevant advancements
about the amelioration of lung damage due to COVID-19 infection using adipose tissue-
derived - total stromal fraction (TSF).
Collapse
Affiliation(s)
- Madhan Jeyaraman
- Department of Orthopaedics, School of Medical Sciences and Research, Sharda University, Greater Noida, Uttar
Pradesh, India
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida,
Uttar Pradesh, India
- International Association of Stem Cell and Regenerative Medicine (IASRM), New Delhi, India
- Orthopaedic Research Group, Coimbatore, Tamil Nadu, India
| | - Sathish Muthu
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida,
Uttar Pradesh, India
- International Association of Stem Cell and Regenerative Medicine (IASRM), New Delhi, India
- Orthopaedic Research Group, Coimbatore, Tamil Nadu, India
- Department of Orthopaedics, Government
Dindigul Medical College and Hospital, Dindigul, Tamil Nadu, India
| | - Rashmi Jain
- School of Medical Sciences and Research, Sharda
University, Greater Noida, Uttar Pradesh, India
| | - Ajay Shringeri Satish
- International Association of Stem Cell and Regenerative Medicine (IASRM), New Delhi, India
- Department of Orthopaedics, Sri Devaraj Urs Medical College,
Kolar, Karnataka, India
| | - Neha Garg
- International Association of Stem Cell and Regenerative Medicine (IASRM), New Delhi, India
- Department of Obstetrics & Gynaecology, Lady Hardinge Medical College, New Delhi, India
| | - Prabhu Chandra Mishra
- International Association of Stem Cell and Regenerative Medicine (IASRM), New Delhi, India
| | - Kumari Swati
- Department of Biotechnology, School of Life Science, Mahatma Gandhi Central University, Motihari, Bihar, India
| | - Anand Parkash
- Department of Biotechnology, School of Life Science, Mahatma Gandhi Central University, Motihari, Bihar, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida,
Uttar Pradesh, India
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab, Emirates
University, Abu Dhabi, United Arab Emirates
| | | | - Dhruv Kumar
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, India
| | - Janne Ruokolainen
- Department of Applied Physics, School of Science, Aalto University, Espoo, Finland
| | - Mohammad Amjad Kamal
- King Fahd Medical Research
Center, King Abdulaziz University, Jeddah, Saudi Arabia
- West China School of Nursing / Institutes for Systems Genetics,
Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University,
Chengdu 610041, Sichuan, China
- Enzymoics, Novel Global Community Educational Foundation, Hebersham, NSW
2770, Australia
| | - Kavindra Kumar Kesari
- Department of Applied Physics, School of Science, Aalto University, Espoo, Finland
- Enzymoics, Novel Global Community Educational Foundation, Hebersham, NSW
2770, Australia
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida,
Uttar Pradesh, India
- International Association of Stem Cell and Regenerative Medicine (IASRM), New Delhi, India
- Enzymoics, Novel Global Community Educational Foundation, Hebersham, NSW
2770, Australia
| |
Collapse
|
4
|
Jeyaraman M, Muthu S, Ganie PA. Does the Source of Mesenchymal Stem Cell Have an Effect in the Management of Osteoarthritis of the Knee? Meta-Analysis of Randomized Controlled Trials. Cartilage 2021; 13:1532S-1547S. [PMID: 32840122 PMCID: PMC8808923 DOI: 10.1177/1947603520951623] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
STUDY DESIGN Meta-analysis. OBJECTIVES To compare the efficacy and safety of bone marrow(BM)-derived mesenchymal stem cell(MSCs) and adipose-derived(AD) MSCs in the management of osteoarthritis of knee from randomized controlled trials(RCTs) available in the literature. MATERIALS AND METHODS We conducted electronic database searche from PubMed, Embase, and Cochrane Library till May 2020 for RCTs analyzing the efficacy and safety of MSCs in management of osteoarthritis of knee. Visual Analog Score(VAS) for Pain, Western Ontario McMaster Universities Osteoarthritis Index(WOMAC), Lysholm Knee Scale(Lysholm), Whole-Organ Magnetic Resonance Imaging Score(WORMS), Knee Osteoarthritis Outcome Score(KOOS), and adverse events were the outcomes analyzed. Analysis was performed in R platform using OpenMeta[Analyst] software. RESULTS Nineteen studies involving 811 patients were included for analysis. None of the studies compared the source of MSCs for osteoarthritis of knee and results were obtained by pooled data analysis of both sources. At 6 months, AD-MSCs showed significantly better VAS(P<0.001,P=0.069) and WOMAC(P=0.134,P=0.441) improvement than BM-MSCs, respectively, compared to controls. At 1 year, AD-MSCs outperformed BM-MSCs compared to their control in measures like WOMAC(P=0.007,P=0.150), KOOS(P<0.001;P=0.658), and WORMS(P<0.001,P=0.041), respectively. Similarly at 24 months, AD-MSCs showed significantly better Lysholm score(P=0.037) than BM-MSCs(P=0.807) although VAS improvement was better with BM-MSCs at 24 months(P<0.001). There were no significant adverse events with either of the MSCs compared to their controls. CONCLUSION Our analysis establishes the efficacy, safety, and superiority of AD-MSC transplantation, compared to BM-MSC, in the management of osteoarthritis of knee from available literature. Further RCTs are needed to evaluate them together with standardized doses.
Collapse
Affiliation(s)
- Madhan Jeyaraman
- Department of Orthopaedics, School of
Medical Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh,
India
| | - Sathish Muthu
- Government Hospital, Velayuthampalayam,
Karur, Tamil Nadu, India
- Sathish Muthu, Government Hospital,
Velayuthampalayam, Pugalur Road, Karur, Tamil Nadu 639117, India.
| | - Parvez Ahmad Ganie
- Department of Orthopaedics, School of
Medical Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh,
India
| |
Collapse
|
5
|
Sharma S, Muthu S, Jeyaraman M, Ranjan R, Jha SK. Translational products of adipose tissue-derived mesenchymal stem cells: Bench to bedside applications. World J Stem Cells 2021; 13:1360-1381. [PMID: 34786149 PMCID: PMC8567449 DOI: 10.4252/wjsc.v13.i10.1360] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 07/02/2021] [Accepted: 08/30/2021] [Indexed: 02/06/2023] Open
Abstract
With developments in the field of tissue engineering and regenerative medicine, the use of biological products for the treatment of various disorders has come into the limelight among researchers and clinicians. Among all the available biological tissues, research and exploration of adipose tissue have become more robust. Adipose tissue engineering aims to develop by-products and their substitutes for their regenerative and immunomodulatory potential. The use of biodegradable scaffolds along with adipose tissue products has a major role in cellular growth, proliferation, and differentiation. Adipose tissue, apart from being the powerhouse of energy storage, also functions as the largest endocrine organ, with the release of various adipokines. The progenitor cells among the heterogeneous population in the adipose tissue are of paramount importance as they determine the capacity of regeneration of these tissues. The results of adipose-derived stem-cell assisted fat grafting to provide numerous growth factors and adipokines that improve vasculogenesis, fat graft integration, and survival within the recipient tissue and promote the regeneration of tissue are promising. Adipose tissue gives rise to various by-products upon processing. This article highlights the significance and the usage of various adipose tissue by-products, their individual characteristics, and their clinical applications.
Collapse
Affiliation(s)
- Shilpa Sharma
- Department of Pediatric Surgery, All India Institute of Medical Sciences, New Delhi 110029, India
- Indian Stem Cell Study Group, Lucknow, Uttar Pradesh 226010, India
| | - Sathish Muthu
- Indian Stem Cell Study Group, Lucknow, Uttar Pradesh 226010, India
- Department of Orthopaedics, Government Medical College and Hospital, Dindigul, Tamil Nadu 624304, India
- Research Scholar, Department of Biotechnology, School of Engineering and Technology, Greater Noida, Sharda University, Uttar Pradesh 201306, India
| | - Madhan Jeyaraman
- Indian Stem Cell Study Group, Lucknow, Uttar Pradesh 226010, India
- Research Scholar, Department of Biotechnology, School of Engineering and Technology, Greater Noida, Sharda University, Uttar Pradesh 201306, India
- Department of Orthopaedics, School of Medical Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh 201306, India
| | - Rajni Ranjan
- Department of Orthopaedics, School of Medical Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh 201306, India
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh 201306, India
| |
Collapse
|
6
|
Functional Recovery after Intramyocardial Injection of Adipose-Derived Stromal Cells Assessed by Cardiac Magnetic Resonance Imaging. Stem Cells Int 2021; 2021:5556800. [PMID: 33976700 PMCID: PMC8087467 DOI: 10.1155/2021/5556800] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 04/02/2021] [Accepted: 04/15/2021] [Indexed: 12/22/2022] Open
Abstract
Aims A major clinical concern is the continuous increase in the number of patients diagnosed with advanced coronary artery disease, ischemic heart failure, and refractory angina, and one of the most promising treatment options for these conditions is stem cell-based therapy. The aim of this study was to assess the functional improvement following intramyocardial injection of adipose-derived stromal cells, using cardiac magnetic resonance. Methods and Results Thirteen patients with ischemic heart failure, reduced left ventricular ejection fraction, refractory angina, and who have been disqualified from any form of direct revascularization were enrolled in the study with transthoracic autologous adipose-derived stromal cell implantation. All patients underwent cardiac magnetic resonance prior to the procedure and after 12 months of follow-up. A significant increase in stroke volume (83.1 ± 8.5 mL vs 93.8 ± 13.8 mL, p = 0.025) and stroke volume index (43.3 ± 7.6 mL/m2 vs 48.7 ± 9.1 mL/m2, p = 0.019), a statistical trend toward an increase in left ventricle ejection fraction (36.7 ± 13.2 vs 39.7 ± 14.9, p = 0.052), and cardiac output improvement (5.0 ± 0.7 vs 5.5 ± 0.9, p = 0.073) was observed in the patient postprocedure. Enhanced relative regional thickening was noted in the segments with adipose-derived stromal cell implantation. Conclusions Intramyocardial adipose-derived stromal cell implantation is a promising therapeutic option for selected, symptomatic patients with ischemic heart failure, who have preserved myocardial viability despite being unsuitable for direct revascularization.
Collapse
|
7
|
Muthu S, Jeyaraman M, Jain R, Gulati A, Jeyaraman N, Prajwal GS, Mishra PC. Accentuating the sources of mesenchymal stem cells as cellular therapy for osteoarthritis knees-a panoramic review. Stem Cell Investig 2021; 8:13. [PMID: 34386542 PMCID: PMC8327191 DOI: 10.21037/sci-2020-055] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 04/25/2021] [Indexed: 02/05/2023]
Abstract
The large economic burden on the global health care systems is due to the increasing number of symptomatic osteoarthritis (OA) knee patients whereby accounting for greater morbidity and impaired functional quality of life. The recent developments and impulses in molecular and regenerative medicine have paved the way for inducing the biological active cells such as stem cells, bioactive materials, and growth factors towards the healing and tissue regenerative process. Mesenchymal stem cells (MSCs) act as a minimally invasive procedure that bridges the gap between pharmacological treatment and surgical treatment for OA. MSCs are the ideal cell-based therapy for treating disorders under a minimally invasive environment in conjunction with cartilage regeneration. Due to the worldwide recognized animal model for such cell-based therapies, global researchers have started using the various sources of MSCs towards cartilage regeneration. However, there is a lacuna in literature on the comparative efficacy and safety of various sources of MSCs in OA of the knee. Hence, the identification of a potential source for therapeutic use in this clinical scenario remains unclear. In this article, we compared the therapeutic effects of various sources of MSCs in terms of efficacy, safety, differentiation potential, durability, accessibility, allogenic preparation and culture expandability to decide the optimal source of MSCs for OA knee.
Collapse
Affiliation(s)
- Sathish Muthu
- Assistant Orthopaedic Surgeon, Government Hospital, Velayuthampalayam, Karur, Tamil Nadu, India
- International Association of Stemcell and Regenerative Medicine (IASRM), New Delhi, India
| | - Madhan Jeyaraman
- International Association of Stemcell and Regenerative Medicine (IASRM), New Delhi, India
- Department of Orthopaedics, School of Medical Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Rashmi Jain
- Department of Orthopaedics, School of Medical Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Arun Gulati
- Department of Orthopaedics, Kalpana Chawla Government Medical College & Hospital, Karnal, Haryana, India
| | - Naveen Jeyaraman
- International Association of Stemcell and Regenerative Medicine (IASRM), New Delhi, India
- Department of Orthopaedics, Kasturba Medical College, MAHE University, Manipal, Karnataka, India
| | | | - Prabhu Chandra Mishra
- International Association of Stemcell and Regenerative Medicine (IASRM), New Delhi, India
| |
Collapse
|
8
|
Liang T, Wen D, Zhong X, Jiang L, Zhu JJ, Gu Z. Therapeutic potential of adipose tissue. Sci Bull (Beijing) 2020; 65:1702-1704. [PMID: 36659240 DOI: 10.1016/j.scib.2020.06.037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Tingxizi Liang
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA; State Key Laboratory of Analytical Chemistry and Collaborative Innovation Center of Chemistry for Life Sciences, School of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, China; Jonsson Comprehensive Cancer Center, California NanoSystems Institute, and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| | - Di Wen
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, California NanoSystems Institute, and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| | - Xintong Zhong
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, California NanoSystems Institute, and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| | - Liping Jiang
- State Key Laboratory of Analytical Chemistry and Collaborative Innovation Center of Chemistry for Life Sciences, School of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, China.
| | - Jun-Jie Zhu
- State Key Laboratory of Analytical Chemistry and Collaborative Innovation Center of Chemistry for Life Sciences, School of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, China.
| | - Zhen Gu
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, California NanoSystems Institute, and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
9
|
Park Y, Lee YJ, Koh JH, Lee J, Min HK, Kim MY, Kim KJ, Lee SJ, Rhie JW, Kim WU, Park SH, Moon SH, Kwok SK. Clinical Efficacy and Safety of Injection of Stromal Vascular Fraction Derived from Autologous Adipose Tissues in Systemic Sclerosis Patients with Hand Disability: A Proof-Of-Concept Trial. J Clin Med 2020; 9:jcm9093023. [PMID: 32961802 PMCID: PMC7565930 DOI: 10.3390/jcm9093023] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 09/15/2020] [Accepted: 09/16/2020] [Indexed: 12/22/2022] Open
Abstract
Background: Stromal vascular fraction (SVF) has recently emerged as a potential therapeutic modality, due to its multipotent cellular components in tissue regeneration. Systemic sclerosis (SSc) is a progressive autoimmune disease that results in hand disability by skin fibrosis and microangiopathies. We performed an open-label study to investigate the efficacy and safety of SVF injection in SSc patients (Clinical Trial number: NCT03060551). Methods: We gathered 20 SSc patients with hand disability, planning for a 24-week follow-up period. SVF was extracted from autologous adipose tissues, processed by the closed system kit, and injected into each finger of SSc patients. We observed various efficacy and safety profiles at each follow-up visit. Results: Among the 20 initially enrolled patients, eighteen received SVF injection, and were completely followed-up for the whole study period. Patients received 3.61 × 106 mesenchymal stem cells into each finger on average. Skin fibrosis, hand edema, and quality of life were significantly improved, and 31.6% of active ulcers were healed at 24 weeks after injections. Semiquantitative results of nailfold capillary microscopy were ameliorated. There was no single serious adverse event related to the procedure. Conclusions: Injection of SVF derived from autologous adipose tissues is tolerable, and shows clinical efficacy in SSc patients.
Collapse
Affiliation(s)
- Youngjae Park
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (Y.P.); (J.L.); (W.-U.K.); (S.-H.P.)
| | - Yoon Jae Lee
- Department of Plastic Surgery, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 07345, Korea;
| | - Jung Hee Koh
- Division of Rheumatology, Department of Internal Medicine, Bucheon St. Mary’s Hospital, College of Medicine, the Catholic University of Korea, Bucheon 14647, Korea;
| | - Jennifer Lee
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (Y.P.); (J.L.); (W.-U.K.); (S.-H.P.)
| | - Hong-Ki Min
- Division of Rheumatology, Department of Internal Medicine, Konkuk University Medical Centre, Konkuk University School of Medicine, Seoul 05030, Korea;
| | - Moon Young Kim
- Division of Rheumatology, Department of Internal Medicine, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Incheon 21431, Korea;
| | - Ki Joo Kim
- Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (K.J.K.); (S.J.L.)
| | - Su Jin Lee
- Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (K.J.K.); (S.J.L.)
| | - Jong Won Rhie
- Department of Plastic Surgery, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
| | - Wan-Uk Kim
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (Y.P.); (J.L.); (W.-U.K.); (S.-H.P.)
| | - Sung-Hwan Park
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (Y.P.); (J.L.); (W.-U.K.); (S.-H.P.)
| | - Suk-Ho Moon
- Department of Plastic Surgery, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
- Correspondence: (S.-H.M.); (S.-K.K.)
| | - Seung-Ki Kwok
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (Y.P.); (J.L.); (W.-U.K.); (S.-H.P.)
- Correspondence: (S.-H.M.); (S.-K.K.)
| |
Collapse
|
10
|
Akakpo W, Schirmann A, Ferretti L, Ben-Naoum K, Carnicelli D, Graziana JP, Hupertan V, Madec FX, Marcelli F, Methorst C, Morel-Journel N, Savareux L, Terrier JE, Faix A, Huyghe E, Yiou R. [Biotherapies for erectile dysfunction and Peyronie's disease: Where are we now?]. Prog Urol 2020; 30:1000-1013. [PMID: 32826194 DOI: 10.1016/j.purol.2020.05.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/21/2020] [Accepted: 05/05/2020] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Clinical trials of cell therapy for erectile dysfunction (ED) and Peyronie's disease (PD) were recently conducted after preclinical studies. AIMS The aims of this study are to give an update on biotherapy for ED and PD and to describe the regulatory framework for these therapies. MATERIALS AND METHODS A literature review was performed through PubMed and Clinical.trials.gov addressing cell therapy for ED and PD and using following keywords "erectile dysfunction", "Peyronie's disease", "stem cell", and "platelet-rich plasma". RESULTS Preclinical studies in rodent models have shown the potential benefit of cell therapy for ED after radical prostatectomy or caused by metabolic diseases, and PD. The tissues used to obtain the therapeutic product were bone marrow, adipose tissue and blood (PRP, platelet-rich plasma). Mechanism of action was shown to be temporary and mainly paracrine. Four clinical trials were published concerning ED after radical prostatectomy and in diabetic patients and one for PD. Eleven clinical trials including three randomized trials are currently going on. Preclinical and preliminary clinical results suggested the possibility to improve spontaneous erectile function and response to pharmaceutical treatment in initially non-responder patients. This effect is mediated by an improvement of penile vascularization. A reduction of penile curvature without side effect was noted after injections into the plaque of PD patients. Most of these therapeutic strategies using autologous cells were considered as "Advanced Therapy Medicinal Products" with strict regulatory frameworks imposing heavy constraints, in particular in case of "substantial" modification of the cells. The regulatory framework remains unclear and more permissive for PRP and cell therapy processes with extemporaneous preparation/injection and no "substantial" modifications. CONCLUSIONS First results on cell therapy for ED and PD are promising. The regulatory framework can significantly change according to cell preparations and origins leading to various constraints. This regulatory framework is crucial to consider for the choice of the procedure.
Collapse
Affiliation(s)
- W Akakpo
- Service d'urologie, université Pierre et Marie Curie, hôpital universitaire de la Pitié-Salpêtrière, 75013 Paris, France
| | - A Schirmann
- Service d'urologie, hôpitaux universitaires Henri-Mondor, CHU Henri-Mondor, AP-HP, 51, avenue du Maréchal-de-Lattre-de-Tassigny, 94010 Créteil, France
| | - L Ferretti
- Service d'urologie, hôpital d'instruction des armées Robert-Picqué, Villenave-d'Ornon, France
| | - K Ben-Naoum
- Service d'urologie, CHU de Nîmes, Nîmes, France
| | - D Carnicelli
- Service d'urologie, CH de Chambery, Chambery, France
| | - J-P Graziana
- Clinique mutualiste de la porte de l'Orient, Lorient, France
| | - V Hupertan
- Cabinet médical Paris Batignolles, Paris, France
| | - F X Madec
- Service d'urologie, CHU de Nantes, Nantes, France
| | - F Marcelli
- Service d'urologie, CHRU de Lille, Lille, France
| | - C Methorst
- Service d'urologie, CH des Quatre Villes, Saint-Cloud, France
| | | | - L Savareux
- Hôpital privé de La Châtaignerie, Beaumont, France
| | | | - A Faix
- Clinique mutualiste Beau-Soleil, Montpellier, France
| | - E Huyghe
- Service d'urologie, CHU de Toulouse, Toulouse, France
| | - R Yiou
- Service d'urologie, hôpitaux universitaires Henri-Mondor, CHU Henri-Mondor, AP-HP, 51, avenue du Maréchal-de-Lattre-de-Tassigny, 94010 Créteil, France.
| |
Collapse
|
11
|
Czapla J, Cichoń T, Pilny E, Jarosz-Biej M, Matuszczak S, Drzyzga A, Krakowczyk Ł, Smolarczyk R. Adipose tissue-derived stromal cells stimulated macrophages-endothelial cells interactions promote effective ischemic muscle neovascularization. Eur J Pharmacol 2020; 883:173354. [PMID: 32663541 DOI: 10.1016/j.ejphar.2020.173354] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/02/2020] [Accepted: 07/08/2020] [Indexed: 12/31/2022]
Abstract
Neovascularization, the process of new blood vessels formation in response to hypoxia induced signals, is an essential step during wound healing or ischemia repair. It follows as a cascade of consecutive events leading to new blood vessels formation and their subsequent remodeling to a mature and functional state, enabling tissue regeneration. Any disruption in consecutive stages of neovascularization can lead to chronic wounds or impairment of tissue repair. In the study we try to explain the biological basis of accelerated blood vessels formation in ischemic tissue after adipose tissue-derived stromal cells (ADSCs) administration. Experiments were performed on mouse models of hindlimb ischemia. We have evaluated the level of immune cells (neutrophils, macrophages) infiltration. The novelty of our work was the assessment of bone marrow-derived stem/progenitor cells (BMDCs) infiltration and their contribution to the neovascularization process in ischemic tissue. We have noticed that ADSCs regulated immune response and affected the kinetics and ratio of macrophages population infiltrating ischemic tissue. Our research revealed that ADSCs promoted changes in the morphology of infiltrating macrophages and their tight association with forming blood vessels. We assume that recruited macrophages may take over the role of pericytes and stabilize the new blood vessel or even differentiate into endothelial cells, which in consequence can accelerate vascular formation upon ADSCs administration. Our findings indicate that administration of ADSCs into ischemic muscle influence spatio-temporal distribution of infiltrating cells (macrophages, neutrophils and BMDCs), which are involved in each step of vascular formation, promoting effective ischemic tissue neovascularization.
Collapse
Affiliation(s)
- Justyna Czapla
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-101, Gliwice, Poland.
| | - Tomasz Cichoń
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-101, Gliwice, Poland
| | - Ewelina Pilny
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-101, Gliwice, Poland; Department of Organic Chemistry, Biochemistry and Biotechnology, Silesian University of Technology, Księdza Marcina Strzody 9 Street, 44-100, Gliwice, Poland
| | - Magdalena Jarosz-Biej
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-101, Gliwice, Poland
| | - Sybilla Matuszczak
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-101, Gliwice, Poland
| | - Alina Drzyzga
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-101, Gliwice, Poland
| | - Łukasz Krakowczyk
- Department of Oncologic and Reconstructive Surgery, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15 Street, 44-101, Gliwice, Poland
| | - Ryszard Smolarczyk
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-101, Gliwice, Poland
| |
Collapse
|
12
|
Sun Y, Chen S, Zhang X, Pei M. Significance of Cellular Cross-Talk in Stromal Vascular Fraction of Adipose Tissue in Neovascularization. Arterioscler Thromb Vasc Biol 2020; 39:1034-1044. [PMID: 31018663 DOI: 10.1161/atvbaha.119.312425] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Adult stem cell-based therapy has been regarded as a promising treatment for tissue ischemia because of its ability to promote new blood vessel formation. Bone marrow-derived mesenchymal stem cells are the most used angiogenic cells for therapeutic neovascularization, yet the side effects and low efficacy have limited their clinical application. Adipose stromal vascular fraction is an easily accessible, heterogeneous cell system comprised of endothelial, stromal, and hematopoietic cell lineages, which has been shown to spontaneously form robust, patent, and functional vasculatures in vivo. However, the characteristics of each cell population and their specific roles in neovascularization remain an area of ongoing investigation. In this review, we summarize the functional capabilities of various stromal vascular fraction constituents during the process of neovascularization and attempt to analyze whether the cross-talk between these constituents generates a synergetic effect, thus contributing to the development of new potential therapeutic strategies to promote neovascularization.
Collapse
Affiliation(s)
- Yuan Sun
- From the Department of Vascular Surgery, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Jiangsu, China (Y.S., X.Z.); Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics (Y.S., M.P.), Exercise Physiology (M.P.), and WVU Cancer Institute, Robert C. Byrd Health Sciences Center (M.P.), West Virginia University, Morgantown; and Department of Orthopaedics, Chengdu Military General Hospital, Chengdu, Sichuan, China (S.C.)
| | - Song Chen
- From the Department of Vascular Surgery, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Jiangsu, China (Y.S., X.Z.); Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics (Y.S., M.P.), Exercise Physiology (M.P.), and WVU Cancer Institute, Robert C. Byrd Health Sciences Center (M.P.), West Virginia University, Morgantown; and Department of Orthopaedics, Chengdu Military General Hospital, Chengdu, Sichuan, China (S.C.)
| | - Xicheng Zhang
- From the Department of Vascular Surgery, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Jiangsu, China (Y.S., X.Z.); Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics (Y.S., M.P.), Exercise Physiology (M.P.), and WVU Cancer Institute, Robert C. Byrd Health Sciences Center (M.P.), West Virginia University, Morgantown; and Department of Orthopaedics, Chengdu Military General Hospital, Chengdu, Sichuan, China (S.C.)
| | - Ming Pei
- From the Department of Vascular Surgery, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Jiangsu, China (Y.S., X.Z.); Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics (Y.S., M.P.), Exercise Physiology (M.P.), and WVU Cancer Institute, Robert C. Byrd Health Sciences Center (M.P.), West Virginia University, Morgantown; and Department of Orthopaedics, Chengdu Military General Hospital, Chengdu, Sichuan, China (S.C.)
| |
Collapse
|
13
|
Arderiu G, Peña E, Aledo R, Juan-Babot O, Crespo J, Vilahur G, Oñate B, Moscatiello F, Badimon L. MicroRNA-145 Regulates the Differentiation of Adipose Stem Cells Toward Microvascular Endothelial Cells and Promotes Angiogenesis. Circ Res 2019; 125:74-89. [PMID: 31219744 DOI: 10.1161/circresaha.118.314290] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
RATIONALE Adipose-derived stem cells (ASCs) are a potential adult mesenchymal stem cell source for restoring endothelial function in ischemic tissues. However, the mechanism that promotes ASCs differentiation toward endothelial cells (ECs) is not known. OBJECTIVE To investigate the mechanisms of ASCs differentiation into ECs. METHODS AND RESULTS ASCs were isolated from clinical lipoaspirates and cultured with DMEM or endothelial cell-conditioned medium. Endothelial cell-conditioned medium induced downregulation of miR-145 in ASCs and promoted endothelial differentiation. We identified bFGF (basic fibroblast growth factor) released by ECs as inducer of ASCs differentiation through receptor-induced AKT (protein kinase B) signaling and phosphorylation of FOXO1 (forkhead box protein O1) suppressing its transcriptional activity and decreasing miR-145 expression. Blocking bFGF-receptor or PI3K/AKT signaling in ASCs increased miR-145 levels. Modulation of miR-145 in ASCs, using a miR-145 inhibitor, regulated their differentiation into ECs: increasing proliferation, migration, inducing expression of EC markers (VE-cadherin, VEGFR2 [vascular endothelial growth factor receptor 2], or VWF [von Willebrand Factor]), and tube-like formation. Furthermore, in vivo, downregulation of miR-145 in ASCs enhanced angiogenesis in subcutaneously implanted plugs in mice. In a murine hindlimb ischemia model injection of ASCs with downregulated miR-145 induced collateral flow and capillary formation evidenced by magnetic resonance angiography. Next, we identified ETS1 (v-ets avian erythroblastosis virus E26 oncogene homolog 1) as the target of miR-145. Upregulation of miR-145 in ASCs, by mimic miR-145, suppressed ETS1 expression and consequently abolished EC differentiation and the angiogenic properties of endothelial cell-conditioned medium-preconditioned ASCs; whereas, overexpression of ETS1 reversed the abrogated antiangiogenic capacity of miR-145. ETS1 overexpression induced similar results to those obtained with miR-145 knockdown. CONCLUSIONS bFGF released by ECs induces ASCs differentiation toward ECs through miR-145-regulated expression of ETS1. Downregulation of miR-145 in ASCs induce vascular network formation in ischemic muscle.
Collapse
Affiliation(s)
- Gemma Arderiu
- From the Cardiovascular-Program ICCC, IR-Hospital Santa Creu i Sant Pau, IIBSantPau Barcelona, Spain (G.A., E.P., R.A., O.J.-B., J.C., G.V., B.O., L.B.)
| | - Esther Peña
- From the Cardiovascular-Program ICCC, IR-Hospital Santa Creu i Sant Pau, IIBSantPau Barcelona, Spain (G.A., E.P., R.A., O.J.-B., J.C., G.V., B.O., L.B.)
- Ciber CV, Instituto Carlos III, Madrid, Spain (E.P., R.A., G.V., L.B.)
| | - Rosa Aledo
- From the Cardiovascular-Program ICCC, IR-Hospital Santa Creu i Sant Pau, IIBSantPau Barcelona, Spain (G.A., E.P., R.A., O.J.-B., J.C., G.V., B.O., L.B.)
- Ciber CV, Instituto Carlos III, Madrid, Spain (E.P., R.A., G.V., L.B.)
| | - Oriol Juan-Babot
- From the Cardiovascular-Program ICCC, IR-Hospital Santa Creu i Sant Pau, IIBSantPau Barcelona, Spain (G.A., E.P., R.A., O.J.-B., J.C., G.V., B.O., L.B.)
| | - Javier Crespo
- From the Cardiovascular-Program ICCC, IR-Hospital Santa Creu i Sant Pau, IIBSantPau Barcelona, Spain (G.A., E.P., R.A., O.J.-B., J.C., G.V., B.O., L.B.)
| | - Gemma Vilahur
- From the Cardiovascular-Program ICCC, IR-Hospital Santa Creu i Sant Pau, IIBSantPau Barcelona, Spain (G.A., E.P., R.A., O.J.-B., J.C., G.V., B.O., L.B.)
- Ciber CV, Instituto Carlos III, Madrid, Spain (E.P., R.A., G.V., L.B.)
| | - Blanca Oñate
- From the Cardiovascular-Program ICCC, IR-Hospital Santa Creu i Sant Pau, IIBSantPau Barcelona, Spain (G.A., E.P., R.A., O.J.-B., J.C., G.V., B.O., L.B.)
| | | | - Lina Badimon
- From the Cardiovascular-Program ICCC, IR-Hospital Santa Creu i Sant Pau, IIBSantPau Barcelona, Spain (G.A., E.P., R.A., O.J.-B., J.C., G.V., B.O., L.B.)
- Ciber CV, Instituto Carlos III, Madrid, Spain (E.P., R.A., G.V., L.B.)
| |
Collapse
|
14
|
Di Stefano AB, Massihnia D, Grisafi F, Castiglia M, Toia F, Montesano L, Russo A, Moschella F, Cordova A. Adipose tissue, angiogenesis and angio-MIR under physiological and pathological conditions. Eur J Cell Biol 2019; 98:53-64. [DOI: 10.1016/j.ejcb.2018.11.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Revised: 11/28/2018] [Accepted: 11/29/2018] [Indexed: 01/06/2023] Open
|
15
|
Del Papa N, Di Luca G, Andracco R, Zaccara E, Maglione W, Pignataro F, Minniti A, Vitali C. Regional grafting of autologous adipose tissue is effective in inducing prompt healing of indolent digital ulcers in patients with systemic sclerosis: results of a monocentric randomized controlled study. Arthritis Res Ther 2019; 21:7. [PMID: 30616671 PMCID: PMC6322261 DOI: 10.1186/s13075-018-1792-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 12/11/2018] [Indexed: 12/14/2022] Open
Abstract
Background A randomized controlled trial (RCT) was performed to confirm preliminary uncontrolled data indicating that regional adipose tissue (AT) grafting (G) is effective in inducing ischemic digital ulcer (IDU) healing in patients with systemic sclerosis (SSc). Patients and methods SSc patients with IDUs were randomized to be blindly treated with AT-G or a sham procedure (SP). AT-G consisted of injection, at the base of the finger with the IDU, of 0.5–1 ml AT after centrifugation of fat aspirate. The SP consisted of false liposuction and local injection of saline solution. The primary endpoint was to compare the cumulative prevalence of healed IDUs in the two groups within the following 8 weeks. Results AT-G and the SP were carried out in 25 and 13 patients, respectively. The two groups were comparable for age, gender, disease duration, and SSc subtypes. IDU healing was observed in 23/25 and 1/13 patients treated with AT-G and the SP, respectively (p < 0.0001). The 12 patients who received the unsuccessful SP underwent a rescue AT-G. In all of them, IDU healing was observed after 8 weeks of observation. It was noticeable that in the AT-G-treated patients a significant reduction of pain intensity (measured by visual analogue scale) was recorded after 4 and 8 weeks (p < 0.0001 in all cases). Similarly, a significant increase of capillary numbers in the affected finger was recorded by nailfold videocapillaroscopy after 4 and 8 weeks (p < 0.0001 in both cases). Conclusion This RCT strongly confirms that AT-G is effective in inducing IDU healing in SSc patients. Trial registration ClinicalTrials.gov, NCT03406988. Registered retrospectively on 25 January 2018.
Collapse
Affiliation(s)
| | | | - Romina Andracco
- U.O.C. Day Hospital Reumatologia, ASST G. Pini-CTO, Milan, Italy
| | - Eleonora Zaccara
- U.O.C. Day Hospital Reumatologia, ASST G. Pini-CTO, Milan, Italy
| | - Wanda Maglione
- U.O.C. Day Hospital Reumatologia, ASST G. Pini-CTO, Milan, Italy
| | | | - Antonina Minniti
- U.O.C. Day Hospital Reumatologia, ASST G. Pini-CTO, Milan, Italy
| | - Claudio Vitali
- Sections of Rheumatology, Villa S. Giuseppe, Como and Casa di Cura di Lecco, Lecco, Italy
| |
Collapse
|
16
|
Nahar S, Nakashima Y, Miyagi-Shiohira C, Kinjo T, Toyoda Z, Kobayashi N, Saitoh I, Watanabe M, Noguchi H, Fujita J. Cytokines in adipose-derived mesenchymal stem cells promote the healing of liver disease. World J Stem Cells 2018; 10:146-159. [PMID: 30631390 PMCID: PMC6325075 DOI: 10.4252/wjsc.v10.i11.146] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 09/07/2018] [Accepted: 10/11/2018] [Indexed: 02/06/2023] Open
Abstract
Adipose-derived mesenchymal stem cells (ADSCs) are a treatment cell source for patients with chronic liver injury. ADSCs are characterized by being harvested from the patient's own subcutaneous adipose tissue, a high cell yield (i.e., reduced immune rejection response), accumulation at a disease nidus, suppression of excessive immune response, production of various growth factors and cytokines, angiogenic effects, anti-apoptotic effects, and control of immune cells via cell-cell interaction. We previously showed that conditioned medium of ADSCs promoted hepatocyte proliferation and improved the liver function in a mouse model of acute liver failure. Furthermore, as found by many other groups, the administration of ADSCs improved liver tissue fibrosis in a mouse model of liver cirrhosis. A comprehensive protein expression analysis by liquid chromatography with tandem mass spectrometry showed that the various cytokines and chemokines produced by ADSCs promote the healing of liver disease. In this review, we examine the ability of expressed protein components of ADSCs to promote healing in cell therapy for liver disease. Previous studies demonstrated that ADSCs are a treatment cell source for patients with chronic liver injury. This review describes the various cytokines and chemokines produced by ADSCs that promote the healing of liver disease.
Collapse
Affiliation(s)
- Saifun Nahar
- Department of Infectious, Respiratory, and Digestive Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan
| | - Yoshiki Nakashima
- Department of Regenerative Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan
| | - Chika Miyagi-Shiohira
- Department of Regenerative Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan
| | - Takao Kinjo
- Department of Basic Laboratory Sciences, School of Health Sciences in the Faculty of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan
| | - Zensei Toyoda
- Department of Basic Laboratory Sciences, School of Health Sciences in the Faculty of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan
| | | | - Issei Saitoh
- Division of Pediatric Dentistry, Graduate School of Medical and Dental Science, Niigata University, Niigata 951-8514, Japan
| | - Masami Watanabe
- Department of Urology, Okayama Univer sity Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Hirofumi Noguchi
- Department of Regenerative Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan.
| | - Jiro Fujita
- Department of Infectious, Respiratory, and Digestive Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan
| |
Collapse
|
17
|
Nahar S, Nakashima Y, Miyagi-Shiohira C, Kinjo T, Kobayashi N, Saitoh I, Watanabe M, Noguchi H, Fujita J. A Comparison of Proteins Expressed between Human and Mouse Adipose-Derived Mesenchymal Stem Cells by a Proteome Analysis through Liquid Chromatography with Tandem Mass Spectrometry. Int J Mol Sci 2018; 19:E3497. [PMID: 30404232 PMCID: PMC6274862 DOI: 10.3390/ijms19113497] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 10/27/2018] [Accepted: 11/04/2018] [Indexed: 12/20/2022] Open
Abstract
Adipose-derived mesenchymal stem cells (ADSCs) have become a common cell source for cell transplantation therapy. Clinical studies have used ADSCs to develop treatments for tissue fibrosis, such as liver cirrhosis and pulmonary fibroma. The need to examine and compare basic research data using clinical research data derived from mice and humans is expected to increase in the future. Here, to better characterize the cells, the protein components expressed by human ADSCs used for treatment, and mouse ADSCs used for research, were comprehensively analyzed by liquid chromatography with tandem mass spectrometry. We found that 92% (401 type proteins) of the proteins expressed by ADSCs in humans and mice were consistent. When classified by the protein functions in a gene ontology analysis, the items that differed by >5% between human and mouse ADSCs were "biological adhesion, locomotion" in biological processes, "plasma membrane" in cellular components, and "antioxidant activity, molecular transducer activity" in molecular functions. Most of the listed proteins were sensitive to cell isolation processes. These results show that the proteins expressed by human and murine ADSCs showed a high degree of correlation.
Collapse
Affiliation(s)
- Saifun Nahar
- Department of Infectious, Respiratory and Digestive Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan.
| | - Yoshiki Nakashima
- Department of Regenerative Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan.
| | - Chika Miyagi-Shiohira
- Department of Regenerative Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan.
| | - Takao Kinjo
- Department of Basic Laboratory Sciences, School of Health Sciences in the Faculty of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan.
| | | | - Issei Saitoh
- Division of Pediatric Dentistry, Graduate School of Medical and Dental Science, Niigata University, Niigata 951-8514, Japan.
| | - Masami Watanabe
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan.
| | - Hirofumi Noguchi
- Department of Regenerative Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan.
| | - Jiro Fujita
- Department of Infectious, Respiratory and Digestive Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan.
| |
Collapse
|
18
|
Kiaie N, Aghdam RM, Tafti SHA, Gorabi AM. Stem Cell-Mediated Angiogenesis in Tissue Engineering Constructs. Curr Stem Cell Res Ther 2018; 14:249-258. [PMID: 30394215 DOI: 10.2174/1574888x13666181105145144] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 10/09/2018] [Accepted: 10/31/2018] [Indexed: 11/22/2022]
Abstract
Angiogenesis has always been a concern in the field of tissue engineering. Poor vascularization of engineered constructs is a problem for the clinical success of these structures. Among the various methods employed to induce angiogenesis, stem cells provide a promising tool for the future. The present review aims to present the application of stem cells in the induction of angiogenesis. Additionally, it summarizes recent advancements in stem cell-mediated angiogenesis of different tissue engineering constructs.
Collapse
Affiliation(s)
- Nasim Kiaie
- School of Metallurgy & Materials Engineering, College of Engineering, University of Tehran, Tehran, Iran.,Department of Tissue Engineering, Amirkabir University of Technology, Tehran 15875, Iran
| | - Rouhollah M Aghdam
- School of Metallurgy & Materials Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Seyed H Ahmadi Tafti
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Armita M Gorabi
- Department of Basic and Clinical Research, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
19
|
A Comparison of the Preservation of Mouse Adipose Tissue-Derived Mesenchymal Stem Cells Using the University of Wisconsin Solution and Hank's Balanced Salt Solution. Stem Cells Int 2018; 2018:1625464. [PMID: 30258463 PMCID: PMC6146634 DOI: 10.1155/2018/1625464] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 07/17/2018] [Accepted: 07/30/2018] [Indexed: 02/01/2023] Open
Abstract
Preservation of adipose tissue before the isolation of cells is one of the most important steps in maintaining the cell viability of adipose tissue-derived mesenchymal stem cells (ADSCs) for clinical use. Hank's balanced salt solution (HBSS) is one of the main ADSC preservation solutions used clinically. However, this step is known to lead to decreased cell viability. The University of Wisconsin (UW) solution is recognized by transplant physicians as an excellent organ preservation solution. We aimed to investigate the effectiveness of UW solution in preservation of the viability of ADSCs. We collected adipose tissue from the inguinal fat pad of mice and compared preservation in UW solution and HBSS overnight by measuring cell viability after isolation. We found that the number of viable cells harvested per gram of adipose tissue mass was higher in UW solution- than HBSS-preserved tissue.
Collapse
|
20
|
Nakashima Y, Nahar S, Miyagi-Shiohira C, Kinjo T, Kobayashi N, Saitoh I, Watanabe M, Fujita J, Noguchi H. A Liquid Chromatography with Tandem Mass Spectrometry-Based Proteomic Analysis of Cells Cultured in DMEM 10% FBS and Chemically Defined Medium Using Human Adipose-Derived Mesenchymal Stem Cells. Int J Mol Sci 2018; 19:ijms19072042. [PMID: 30011845 PMCID: PMC6073410 DOI: 10.3390/ijms19072042] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 07/09/2018] [Accepted: 07/11/2018] [Indexed: 02/07/2023] Open
Abstract
Human adipose-derived mesenchymal stem cells (hADSCs) are representative cell sources for cell therapy. Classically, Dulbecco's Modified Eagle's medium (DMEM) containing 10% fetal bovine serum (FBS) has been used as culture medium for hADSCs. A chemically defined medium (CDM) containing no heterologous animal components has recently been used to produce therapeutic hADSCs. However, how the culture environment using a medium without FBS affects the protein expression of hADSC is unclear. We subjected hADSCs cultured in CDM and DMEM (10% FBS) to a protein expression analysis by tandem mass spectrometry liquid chromatography and noted 98.2% agreement in the proteins expressed by the CDM and DMEM groups. We classified 761 proteins expressed in both groups by their function in a gene ontology analysis. Thirty-one groups of proteins were classified as growth-related proteins in the CDM and DMEM groups, 16 were classified as antioxidant activity-related, 147 were classified as immune system process-related, 557 were involved in biological regulation, 493 were classified as metabolic process-related, and 407 were classified as related to stimulus responses. These results show that the trend in the expression of major proteins related to the therapeutic effect of hADSCs correlated strongly in both groups.
Collapse
Affiliation(s)
- Yoshiki Nakashima
- Department of Regenerative Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan.
| | - Saifun Nahar
- Department of Infectious, Respiratory, and Digestive Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan.
| | - Chika Miyagi-Shiohira
- Department of Regenerative Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan.
| | - Takao Kinjo
- Department of Basic Laboratory Sciences, School of Health Sciences in the Faculty of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan.
| | | | - Issei Saitoh
- Division of Pediatric Dentistry, Graduate School of Medical and Dental Science, Niigata University, Niigata 951-8514, Japan.
| | - Masami Watanabe
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan.
| | - Jiro Fujita
- Department of Infectious, Respiratory, and Digestive Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan.
| | - Hirofumi Noguchi
- Department of Regenerative Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan.
| |
Collapse
|
21
|
Oliva-Olivera W, Coín-Aragüez L, Lhamyani S, Salas J, Gentile AM, Romero-Zerbo SY, Zayed H, Valderrama J, Tinahones FJ, El Bekay R. Differences in the neovascular potential of thymus versus subcutaneous adipose-derived stem cells from patients with myocardial ischaemia. J Tissue Eng Regen Med 2018; 12:e1772-e1784. [PMID: 29024495 DOI: 10.1002/term.2585] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 07/19/2017] [Accepted: 10/03/2017] [Indexed: 12/13/2022]
Abstract
Adipose tissue-derived multipotent mesenchymal cells (ASCs) participate in the information of blood vessels under hypoxic conditions. It is probable that the susceptibility of ASCs to the influence of age and ageing-associated pathologies compromises their therapeutic effectiveness depending on the adipose tissue depot. Our aim was to examine the neovascular potential under hypoxic conditions of ASCs-derived from thymic (thymASCs) and subcutaneous (subASCs) adipose tissue from 39 subjects with and without type 2 diabetes mellitus (T2DM) and of different ages who were undergoing coronary bypass surgery. We confirmed a significant decrease in the percentage of CD34+ CD31- CD45- subASCs in the cell yield of subASCs and in the survival of cultured endothelial cells in the medium conditioned by the hypox-subASCs with increasing patient age, which was not observed in thymASCs. Whereas the length of the tubules generated by hypox-subASCs tended to correlate negatively with patient age, tubule formation capacity of the hypoxic thymASCs increased significantly. Compared with subASCs, thymASCs from subjects over age 65 and without T2DM showed higher cell yield, tubule formation capacity, vascular endothelial growth factor secretion levels, and ability to promote endothelial cell survival in their conditioned medium. Deterioration in subASCs neovascular potential relative to thymASCs derived from these subjects was accompanied by higher expression levels of NOX4 mRNA and fibrotic proteins. Our results indicate that thymASCs from patients over age 65 and without T2DM have a higher angiogenic potential than those from the other patient groups, suggesting they may be a good candidate for angiogenic therapy in subjects undergoing coronary bypass surgery.
Collapse
Affiliation(s)
- Wilfredo Oliva-Olivera
- Department of Clinical Endocrinology and Nutrition, Institute of Biomedical Research of Málaga (IBIMA), Clinical Hospital of Málaga (Virgen de la Victoria), University of Málaga (UMA), Malaga, Spain.,CIBER-The Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Malaga, Spain
| | - Leticia Coín-Aragüez
- Department of Clinical Endocrinology and Nutrition, Institute of Biomedical Research of Málaga (IBIMA), Clinical Hospital of Málaga (Virgen de la Victoria), University of Málaga (UMA), Malaga, Spain.,CIBER-The Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Malaga, Spain
| | | | - Julián Salas
- Cardiovascular Surgery Department, Carlos Haya University Hospital, Malaga, Spain
| | | | - Silvana-Yanina Romero-Zerbo
- Unidad de Gestión Clínica Intercentros de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga/Universidad de Málaga, Malaga, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Malaga, Spain
| | - Hatem Zayed
- Biomedical Sciences Program, Health Sciences Department, College of Arts and Sciences, Qatar University, Doha, Qatar
| | - Jf Valderrama
- Cardiovascular Surgery Department, Carlos Haya University Hospital, Malaga, Spain
| | - Francisco José Tinahones
- Department of Clinical Endocrinology and Nutrition, Institute of Biomedical Research of Málaga (IBIMA), Clinical Hospital of Málaga (Virgen de la Victoria), University of Málaga (UMA), Malaga, Spain.,CIBER-The Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Malaga, Spain
| | - Rajaa El Bekay
- CIBER-The Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Malaga, Spain.,Unidad de Gestión Clínica Intercentros de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga/Universidad de Málaga, Malaga, Spain
| |
Collapse
|
22
|
Bora P, Majumdar AS. Adipose tissue-derived stromal vascular fraction in regenerative medicine: a brief review on biology and translation. Stem Cell Res Ther 2017; 8:145. [PMID: 28619097 PMCID: PMC5472998 DOI: 10.1186/s13287-017-0598-y] [Citation(s) in RCA: 312] [Impact Index Per Article: 44.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Adipose/fat tissue provides an abundant source of stromal vascular fraction (SVF) cells for immediate administration and can also give rise to a substantial number of cultured, multipotent adipose-derived stromal cells (ADSCs). Recently, both SVF and ADSCs have gained wide-ranging translational significance in regenerative medicine. Initially used for cosmetic breast enhancement, this mode of treatment has found use in many diseases involving immune disorders, tissue degeneration, and ischaemic conditions. In this review, we try to address several important aspects of this field, outlining the biology, technology, translation, and challenges related to SVF- and ADSC-based therapies. Starting from the basics of SVF and ADSC isolation, we touch upon recently developed technologies, addressing elements of novel methods and devices under development for point-of-care isolation of SVF. Characterisation of SVF cells and ADSCs is also an evolving area and we look into unusual expression of CD34 antigen as an interesting marker for such purposes. Based on reports involving different cells of the SVF, we draw a potential mode of action, focussing on angiogenesis since it involves multiple cells, unlike immunomodulation which is governed predominantly by ADSCs. We have looked into the latest research, experimental therapies, and clinical trials which are utilising SVF/ADSCs in conditions such as multiple sclerosis, Crohn’s disease, peripheral neuropathy, osteoarthritis, diabetic foot ulcer, and so forth. However, problems have arisen with regards to the lack of proper regulatory guidelines for such therapies and, since the introduction of US Food and Drug Administration draft guidelines and the Reliable and Effective Growth for Regenerative Health Options that Improve Wellness (REGROW) Act, the debate became more public with regards to safe and efficacious use of these cells.
Collapse
Affiliation(s)
- Pablo Bora
- Stempeutics Research Private Limited, Akshay Tech Park, # 72&73, 2nd Floor, EPIP Zone, Phase 1, Whitefield, Bangalore, 560066, India.,Present Address: Department of Molecular Biology & Genetics, Faculty of Science, Jihočeská univerzita v Českých Budějovicích (University of South Bohemia), Branišovská 31, 37005, České Budějovice, Czech Republic
| | - Anish S Majumdar
- Stempeutics Research Private Limited, Akshay Tech Park, # 72&73, 2nd Floor, EPIP Zone, Phase 1, Whitefield, Bangalore, 560066, India.
| |
Collapse
|
23
|
|
24
|
Shen Y, Zuo S, Wang Y, Shi H, Yan S, Chen D, Xiao B, Zhang J, Gong Y, Shi M, Tang J, Kong D, Lu L, Yu Y, Zhou B, Duan SZ, Schneider C, Funk CD, Yu Y. Thromboxane Governs the Differentiation of Adipose-Derived Stromal Cells Toward Endothelial Cells In Vitro and In Vivo. Circ Res 2016; 118:1194-207. [PMID: 26957525 DOI: 10.1161/circresaha.115.307853] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2015] [Accepted: 03/08/2016] [Indexed: 12/30/2022]
Abstract
RATIONALE Autologous adipose-derived stromal cells (ASCs) offer great promise as angiogenic cell therapy for ischemic diseases. Because of their limited self-renewal capacity and pluripotentiality, the therapeutic efficacy of ASCs is still relatively low. Thromboxane has been shown to play an important role in the maintenance of vascular homeostasis. However, little is known about the effects of thromboxane on ASC-mediated angiogenesis. OBJECTIVE To explore the role of the thromboxane-prostanoid receptor (TP) in mediating the angiogenic capacity of ASCs in vivo. METHODS AND RESULTS ASCs were prepared from mouse epididymal fat pads and induced to differentiate into endothelial cells (ECs) by vascular endothelial growth factor. Cyclooxygenase-2 expression, thromboxane production, and TP expression were upregulated in ASCs on vascular endothelial growth factor treatment. Genetic deletion or pharmacological inhibition of TP in mouse or human ASCs accelerated EC differentiation and increased tube formation in vitro, enhanced angiogenesis in in vivo Matrigel plugs and ischemic mouse hindlimbs. TP deficiency resulted in a significant cellular accumulation of β-catenin by suppression of calpain-mediated degradation in ASCs. Knockdown of β-catenin completely abrogated the enhanced EC differentiation of TP-deficient ASCs, whereas inhibition of calpain reversed the suppressed angiogenic capacity of TP re-expressed ASCs. Moreover, TP was coupled with Gαq to induce calpain-mediated suppression of β-catenin signaling through calcium influx in ASCs. CONCLUSION Thromboxane restrained EC differentiation of ASCs through TP-mediated repression of the calpain-dependent β-catenin signaling pathway. These results indicate that TP inhibition could be a promising strategy for therapy utilizing ASCs in the treatment of ischemic diseases.
Collapse
Affiliation(s)
- Yujun Shen
- From the Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (Y.S., S.Z., Y.W., S.Y., D.C., B.X., J.Z., Y.G., M.S., J.T., D.K., L.L., Y.Y., B.Z., S.-Z.D., Y.Y.); Department of Nutrition, The NO.2 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China (Y.S., H.S.); Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, AREA Science Park, Trieste, Italy (C.S.); Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, Udine, Italy (C.S.); and Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.F.)
| | - Shengkai Zuo
- From the Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (Y.S., S.Z., Y.W., S.Y., D.C., B.X., J.Z., Y.G., M.S., J.T., D.K., L.L., Y.Y., B.Z., S.-Z.D., Y.Y.); Department of Nutrition, The NO.2 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China (Y.S., H.S.); Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, AREA Science Park, Trieste, Italy (C.S.); Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, Udine, Italy (C.S.); and Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.F.)
| | - Yuanyang Wang
- From the Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (Y.S., S.Z., Y.W., S.Y., D.C., B.X., J.Z., Y.G., M.S., J.T., D.K., L.L., Y.Y., B.Z., S.-Z.D., Y.Y.); Department of Nutrition, The NO.2 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China (Y.S., H.S.); Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, AREA Science Park, Trieste, Italy (C.S.); Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, Udine, Italy (C.S.); and Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.F.)
| | - Hongfei Shi
- From the Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (Y.S., S.Z., Y.W., S.Y., D.C., B.X., J.Z., Y.G., M.S., J.T., D.K., L.L., Y.Y., B.Z., S.-Z.D., Y.Y.); Department of Nutrition, The NO.2 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China (Y.S., H.S.); Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, AREA Science Park, Trieste, Italy (C.S.); Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, Udine, Italy (C.S.); and Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.F.)
| | - Shuai Yan
- From the Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (Y.S., S.Z., Y.W., S.Y., D.C., B.X., J.Z., Y.G., M.S., J.T., D.K., L.L., Y.Y., B.Z., S.-Z.D., Y.Y.); Department of Nutrition, The NO.2 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China (Y.S., H.S.); Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, AREA Science Park, Trieste, Italy (C.S.); Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, Udine, Italy (C.S.); and Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.F.)
| | - Di Chen
- From the Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (Y.S., S.Z., Y.W., S.Y., D.C., B.X., J.Z., Y.G., M.S., J.T., D.K., L.L., Y.Y., B.Z., S.-Z.D., Y.Y.); Department of Nutrition, The NO.2 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China (Y.S., H.S.); Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, AREA Science Park, Trieste, Italy (C.S.); Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, Udine, Italy (C.S.); and Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.F.)
| | - Bing Xiao
- From the Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (Y.S., S.Z., Y.W., S.Y., D.C., B.X., J.Z., Y.G., M.S., J.T., D.K., L.L., Y.Y., B.Z., S.-Z.D., Y.Y.); Department of Nutrition, The NO.2 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China (Y.S., H.S.); Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, AREA Science Park, Trieste, Italy (C.S.); Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, Udine, Italy (C.S.); and Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.F.)
| | - Jian Zhang
- From the Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (Y.S., S.Z., Y.W., S.Y., D.C., B.X., J.Z., Y.G., M.S., J.T., D.K., L.L., Y.Y., B.Z., S.-Z.D., Y.Y.); Department of Nutrition, The NO.2 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China (Y.S., H.S.); Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, AREA Science Park, Trieste, Italy (C.S.); Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, Udine, Italy (C.S.); and Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.F.)
| | - Yanjun Gong
- From the Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (Y.S., S.Z., Y.W., S.Y., D.C., B.X., J.Z., Y.G., M.S., J.T., D.K., L.L., Y.Y., B.Z., S.-Z.D., Y.Y.); Department of Nutrition, The NO.2 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China (Y.S., H.S.); Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, AREA Science Park, Trieste, Italy (C.S.); Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, Udine, Italy (C.S.); and Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.F.)
| | - Maohua Shi
- From the Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (Y.S., S.Z., Y.W., S.Y., D.C., B.X., J.Z., Y.G., M.S., J.T., D.K., L.L., Y.Y., B.Z., S.-Z.D., Y.Y.); Department of Nutrition, The NO.2 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China (Y.S., H.S.); Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, AREA Science Park, Trieste, Italy (C.S.); Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, Udine, Italy (C.S.); and Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.F.)
| | - Juan Tang
- From the Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (Y.S., S.Z., Y.W., S.Y., D.C., B.X., J.Z., Y.G., M.S., J.T., D.K., L.L., Y.Y., B.Z., S.-Z.D., Y.Y.); Department of Nutrition, The NO.2 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China (Y.S., H.S.); Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, AREA Science Park, Trieste, Italy (C.S.); Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, Udine, Italy (C.S.); and Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.F.)
| | - Deping Kong
- From the Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (Y.S., S.Z., Y.W., S.Y., D.C., B.X., J.Z., Y.G., M.S., J.T., D.K., L.L., Y.Y., B.Z., S.-Z.D., Y.Y.); Department of Nutrition, The NO.2 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China (Y.S., H.S.); Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, AREA Science Park, Trieste, Italy (C.S.); Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, Udine, Italy (C.S.); and Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.F.)
| | - Luheng Lu
- From the Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (Y.S., S.Z., Y.W., S.Y., D.C., B.X., J.Z., Y.G., M.S., J.T., D.K., L.L., Y.Y., B.Z., S.-Z.D., Y.Y.); Department of Nutrition, The NO.2 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China (Y.S., H.S.); Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, AREA Science Park, Trieste, Italy (C.S.); Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, Udine, Italy (C.S.); and Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.F.)
| | - Yu Yu
- From the Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (Y.S., S.Z., Y.W., S.Y., D.C., B.X., J.Z., Y.G., M.S., J.T., D.K., L.L., Y.Y., B.Z., S.-Z.D., Y.Y.); Department of Nutrition, The NO.2 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China (Y.S., H.S.); Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, AREA Science Park, Trieste, Italy (C.S.); Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, Udine, Italy (C.S.); and Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.F.)
| | - Bin Zhou
- From the Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (Y.S., S.Z., Y.W., S.Y., D.C., B.X., J.Z., Y.G., M.S., J.T., D.K., L.L., Y.Y., B.Z., S.-Z.D., Y.Y.); Department of Nutrition, The NO.2 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China (Y.S., H.S.); Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, AREA Science Park, Trieste, Italy (C.S.); Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, Udine, Italy (C.S.); and Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.F.)
| | - Sheng-Zhong Duan
- From the Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (Y.S., S.Z., Y.W., S.Y., D.C., B.X., J.Z., Y.G., M.S., J.T., D.K., L.L., Y.Y., B.Z., S.-Z.D., Y.Y.); Department of Nutrition, The NO.2 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China (Y.S., H.S.); Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, AREA Science Park, Trieste, Italy (C.S.); Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, Udine, Italy (C.S.); and Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.F.)
| | - Claudio Schneider
- From the Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (Y.S., S.Z., Y.W., S.Y., D.C., B.X., J.Z., Y.G., M.S., J.T., D.K., L.L., Y.Y., B.Z., S.-Z.D., Y.Y.); Department of Nutrition, The NO.2 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China (Y.S., H.S.); Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, AREA Science Park, Trieste, Italy (C.S.); Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, Udine, Italy (C.S.); and Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.F.)
| | - Colin D Funk
- From the Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (Y.S., S.Z., Y.W., S.Y., D.C., B.X., J.Z., Y.G., M.S., J.T., D.K., L.L., Y.Y., B.Z., S.-Z.D., Y.Y.); Department of Nutrition, The NO.2 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China (Y.S., H.S.); Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, AREA Science Park, Trieste, Italy (C.S.); Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, Udine, Italy (C.S.); and Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.F.)
| | - Ying Yu
- From the Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (Y.S., S.Z., Y.W., S.Y., D.C., B.X., J.Z., Y.G., M.S., J.T., D.K., L.L., Y.Y., B.Z., S.-Z.D., Y.Y.); Department of Nutrition, The NO.2 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China (Y.S., H.S.); Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, AREA Science Park, Trieste, Italy (C.S.); Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, Udine, Italy (C.S.); and Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.F.).
| |
Collapse
|
25
|
Kang T, Jones TM, Naddell C, Bacanamwo M, Calvert JW, Thompson WE, Bond VC, Chen YE, Liu D. Adipose-Derived Stem Cells Induce Angiogenesis via Microvesicle Transport of miRNA-31. Stem Cells Transl Med 2016; 5:440-50. [PMID: 26933040 PMCID: PMC4798737 DOI: 10.5966/sctm.2015-0177] [Citation(s) in RCA: 168] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 11/19/2015] [Indexed: 12/25/2022] Open
Abstract
UNLABELLED Cell secretion is an important mechanism for stem cell-based therapeutic angiogenesis, along with cell differentiation to vascular endothelial cells or smooth muscle cells. Cell-released microvesicles (MVs) have been recently implicated to play an essential role in intercellular communication. The purpose of this study was to explore the potential effects of stem cell-released MVs in proangiogenic therapy. We observed for the first time that MVs were released from adipose-derived stem cells (ASCs) and were able to increase the migration and tube formation of human umbilical vein endothelial cells (HUVECs). Endothelial differentiation medium (EDM) preconditioning of ASCs upregulated the release of MVs and enhanced the angiogenic effect of the released MVs in vitro. RNA analysis revealed that microRNA was enriched in ASC-released MVs and that the level of microRNA-31 (miR-31) in MVs was notably elevated upon EDM-preconditioning of MV-donor ASCs. Further studies exhibited that miR-31 in MVs contributed to the migration and tube formation of HUVECs, microvessel outgrowth of mouse aortic rings, and vascular formation of mouse Matrigel plugs. Moreover, factor-inhibiting HIF-1, an antiangiogenic gene, was identified as the target of miR-31 in HUVECs. Our findings provide the first evidence that MVs from ASCs, particularly from EDM-preconditioned ASCs, promote angiogenesis and the delivery of miR-31 may contribute the proangiogenic effect. SIGNIFICANCE This study provides the evidence that microvesicles (MVs) from adipose-derived stem cells (ASCs), particularly from endothelial differentiation medium (EDM)-preconditioned ASCs, promote angiogenesis. An underlying mechanism of the proangiogenesis may be the delivery of microRNA-31 via MVs from ASCs to vascular endothelial cells in which factor-inhibiting HIF-1 is targeted and suppressed. The study findings reveal the role of MVs in mediating ASC-induced angiogenesis and suggest a potential MV-based angiogenic therapy for ischemic diseases.
Collapse
Affiliation(s)
- Ting Kang
- Division of Cardiology, The First Affiliated Hospital, Nanchang University, Nanchang, People's Republic of China Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - Tia M Jones
- Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - Clayton Naddell
- Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - Methode Bacanamwo
- Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - John W Calvert
- Division of Cardiothoracic Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Winston E Thompson
- Department of Physiology, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - Vincent C Bond
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - Y Eugene Chen
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Dong Liu
- Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, Georgia, USA Department of Physiology, Morehouse School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
26
|
Szöke K, Reinisch A, Østrup E, Reinholt FP, Brinchmann JE. Autologous cell sources in therapeutic vasculogenesis: In vitro and in vivo comparison of endothelial colony-forming cells from peripheral blood and endothelial cells isolated from adipose tissue. Cytotherapy 2015; 18:242-52. [PMID: 26669908 DOI: 10.1016/j.jcyt.2015.10.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 08/22/2015] [Accepted: 10/14/2015] [Indexed: 01/19/2023]
Abstract
BACKGROUND AIMS Autologous endothelial cells are promising alternative angiogenic cell sources in trials of therapeutic vasculogenesis, in the treatment of vascular diseases and in the field of tissue engineering. A population of endothelial cells (ECs) with long-term proliferative capability, endothelial colony-forming cells (ECFCs), can be isolated from human peripheral blood. ECFCs are considered an endothelial precursor population. They can be expanded in cell factories in sufficient numbers for clinical applications, but because the number of isolated primary ECs is low, the culture period required may be long. Another EC population that is easily available in the autologous setting and may be expanded in vitro through several population doublings are ECs from adipose tissue (AT-ECs). METHODS Through extensive comparisons using whole-genome microarray analysis, morphology, phenotype and functional assays, we wanted to evaluate the potential of these EC populations for use in clinical neovascularization. RESULTS Global gene expression profiling of ECFCs, AT-ECs and the classical EC population, human umbilical vein ECs, showed that the EC populations clustered as unique populations, but very close to each other. By cell surface phenotype and vasculogenic potential in vitro and in vivo, we also found the ECFCs to be extremely similar to AT-ECs. CONCLUSIONS These properties, together with easy access in the autologous setting, suggest that both AT-ECs and ECFCs may be useful in trials of therapeutic neovascularization. However, AT-ECs may be a more practical alternative for obtaining large quantities of autologous ECs.
Collapse
Affiliation(s)
- Krisztina Szöke
- Department of Immunology and Norwegian Center for Stem Cell Research, Oslo University Hospital, Rikshospitalet, Oslo, Norway.
| | - Andreas Reinisch
- Stem Cell Research Unit, Department of Hematology and Stem Cell Transplantation, University Clinic of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Esben Østrup
- Department of Immunology and Norwegian Center for Stem Cell Research, Oslo University Hospital, Rikshospitalet, Oslo, Norway; Department of Biomaterials, Institute of Clinical Dentistry, University of Oslo, Oslo, Norway
| | - Finn P Reinholt
- Institute of Pathology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Jan E Brinchmann
- Department of Immunology and Norwegian Center for Stem Cell Research, Oslo University Hospital, Rikshospitalet, Oslo, Norway; Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| |
Collapse
|
27
|
Horikoshi-Ishihara H, Tobita M, Tajima S, Tanaka R, Oshita T, Tabata Y, Mizuno H. Coadministration of adipose-derived stem cells and control-released basic fibroblast growth factor facilitates angiogenesis in a murine ischemic hind limb model. J Vasc Surg 2015; 64:1825-1834.e1. [PMID: 26597457 DOI: 10.1016/j.jvs.2015.09.054] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 09/18/2015] [Indexed: 01/30/2023]
Abstract
OBJECTIVE Adipose-derived stem cells (ASCs) have angiogenic potential owing to their differentiation into endothelial cells and their release of angiogenic growth factors to elicit paracrine effects. In addition, control-released basic fibroblast growth factor (bFGF) sustained with a gelatin hydrogel also supports effective angiogenesis. We sought to determine if coadministration of ASCs and control-released bFGF into murine ischemic limbs facilitates angiogenesis. METHODS Levels of growth factors in the conditioned media of ASCs cultured with or without control-released bFGF were measured by enzyme-linked immunosorbent assays. A murine ischemic hind limb model was generated and intramuscularly injected with the following: gelatin hydrogel (group 1), a high number of ASCs (group 2), control-released bFGF (group 3), a small number of ASCs and control-released bFGF (group 4), and a high number of ASCs and control-released bFGF (group 5). Macroscopic and microscopic vascular changes were evaluated until day 7 by laser Doppler perfusion imaging and histologic analyses, respectively. RESULTS Secretion of hepatocyte growth factor, vascular endothelial growth factor, and transforming growth factor-β1 was enhanced by control-released bFGF. Vascular improvement was achieved in groups 4 and 5 according to laser Doppler perfusion imaging. Hematoxylin and eosin staining and CD31 immunohistochemical staining demonstrated an increase in the vascular density, vessel diameter, and thickness of vessel walls in groups 4 and 5. Cells positively stained for CD146, α-smooth muscle actin, and transforming growth factor-β1 were observed around vessel walls in groups 4 and 5. CONCLUSIONS These findings suggest that coadministration of ASCs and control-released bFGF facilitates angiogenesis in terms of vessel maturation in a murine ischemic hind limb model.
Collapse
Affiliation(s)
- Hisako Horikoshi-Ishihara
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Morikuni Tobita
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Satoshi Tajima
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Rica Tanaka
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Takashi Oshita
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Yasuhiko Tabata
- Department of Biomaterials, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Hiroshi Mizuno
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan.
| |
Collapse
|
28
|
Del Papa N, Di Luca G, Sambataro D, Zaccara E, Maglione W, Gabrielli A, Fraticelli P, Moroncini G, Beretta L, Santaniello A, Sambataro G, Ferraresi R, Vitali C. Regional Implantation of Autologous Adipose Tissue-Derived Cells Induces a Prompt Healing of Long-Lasting Indolent Digital Ulcers in Patients with Systemic Sclerosis. Cell Transplant 2015; 24:2297-305. [DOI: 10.3727/096368914x685636] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Digital ulcers (DUs) are a rather frequent and invalidating complication in systemic sclerosis (SSc), often showing a very slow or null tendency to heal, in spite of the commonly used systemic and local therapeutic procedures. Recently, stem cell therapy has emerged as a new approach to accelerate wound healing. In the present study, we have tentatively treated long-lasting and poorly responsive to traditional therapy SSc-related DUs by implantation of autologous adipose tissue-derived cell (ATDC) fractions. Fifteen patients with SSc having a long-lasting DU in only one fingertip who were unresponsive to intensive systemic and local treatment were enrolled in the study. The grafting procedure consisted of the injection, at the basis of the corresponding finger, of 0.5-1 ml of autologous ATDC fractions, separated by centrifugation of adipose tissue collected through liposuction from subcutaneous abdominal fat. Time to heal after the procedure was the primary end point of the study, while reduction of pain intensity and of analgesic consumption represented a secondary end point. Furthermore, the posttherapy variation of the number of capillaries, observed in the nailfold video capillaroscopy (NVC) exam and of the resistivity in the digit arteries, measured by high-resolution echocolor-Doppler, were also taken into account. A rather fast healing of the DUs was reached in all of the enrolled patients (mean time to healing 4.23 weeks; range 2-7 weeks). A significant reduction of pain intensity was observed after a few weeks ( p < 0.001), while the number of capillaries was significantly increased at 3- and 6-month NVC assessment ( p < 0.0001 in both cases). Finally, a significant after-treatment reduction of digit artery resistivity was also recorded ( p < 0.0001). Even with the limitations related to the small number of patients included and to the open-label design of the study, the observed strongly favorable outcome suggests that local grafting with ATDCs could represent a promising option for the treatment of SSc-related DUs unresponsive to more consolidated therapies.
Collapse
Affiliation(s)
| | | | | | - Eleonora Zaccara
- U.O.C. Day Hospital Reumatologia, Ospedale G. Pini, Milano, Italy
| | - Wanda Maglione
- U.O.C. Day Hospital Reumatologia, Ospedale G. Pini, Milano, Italy
| | - Armando Gabrielli
- U.O. Clinica Medica, Dipartimento di Medicina Interna, Ospedali Riuniti, Ancona, Italy
| | - Paolo Fraticelli
- U.O. Clinica Medica, Dipartimento di Medicina Interna, Ospedali Riuniti, Ancona, Italy
| | - Gianluca Moroncini
- U.O. Clinica Medica, Dipartimento di Medicina Interna, Ospedali Riuniti, Ancona, Italy
| | - Lorenzo Beretta
- Centro di Riferimento per le Malattie Autoimmuni Sistemiche, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico di Milano, Milano, Italy
| | - Alessandro Santaniello
- Centro di Riferimento per le Malattie Autoimmuni Sistemiche, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico di Milano, Milano, Italy
| | - Gianluca Sambataro
- Dipartimento Medicina Interna e Reumatologia, Campus Biomedico, Roma, Italy
| | - Roberto Ferraresi
- Laboratorio Emodinamica Periferica Interventistica, Humanitas Gavazzeni, Bergamo, Italy
| | - Claudio Vitali
- Servizio di Reumatologia, Istituto San Giuseppe, Anzano del Parco, Como, Italy
| |
Collapse
|
29
|
Jung S, Kleineidam B, Kleinheinz J. Regenerative potential of human adipose-derived stromal cells of various origins. J Craniomaxillofac Surg 2015; 43:2144-51. [PMID: 26541747 DOI: 10.1016/j.jcms.2015.10.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 08/22/2015] [Accepted: 10/01/2015] [Indexed: 12/12/2022] Open
Abstract
In regenerative concepts, the potential of adult stem cells holds great promise concerning an individualized therapeutic approach. These cells provide renewable progenitor cells to replace aged tissue, and play a significant role in tissue repair and regeneration. In this investigation, the characteristics of different types of adipose tissue are analysed systematically with special attention to their proliferation and differentiation potential concerning the angiogenic and osteogenic lineage. Tissue samples from subcutaneous, visceral, and omental fat were processed according to standard procedures. The cells were characterized and cultivated under suitable conditions for osteogenic and angiogenic cell culture. The development of the different cell cultures as well as their differentiation were analysed morphologically and immunohistochemically from cell passages P1 to P12. Harvesting and isolation of multipotent cells from all three tissue types could be performed reproducibly. The cultivation of these cells under osteogenic conditions led to a morphological and immunohistochemical differentiation; mineralization could be detected. The most stable results were observed for the cells of subcutaneous origin. An osteogenic differentiation from adipose-derived cells from all analysed fatty tissues can be achieved easily and reproducibly. In therapeutic concepts including angiogenic regeneration, adipose-derived cells from subcutaneous tissue provide the optimal cellular base.
Collapse
Affiliation(s)
- Susanne Jung
- Department of Cranio-Maxillofacial Surgery, Research Unit Vascular Biology of Oral Structures (VABOS), University Hospital Muenster, Germany.
| | - Benedikt Kleineidam
- Department of Cranio-Maxillofacial Surgery, Research Unit Vascular Biology of Oral Structures (VABOS), University Hospital Muenster, Germany
| | - Johannes Kleinheinz
- Department of Cranio-Maxillofacial Surgery, Research Unit Vascular Biology of Oral Structures (VABOS), University Hospital Muenster, Germany
| |
Collapse
|
30
|
Damous LL, Nakamuta JS, Carvalho AETSD, Carvalho KC, Soares JM, Simões MDJ, Krieger JE, Baracat EC. Does adipose tissue-derived stem cell therapy improve graft quality in freshly grafted ovaries? Reprod Biol Endocrinol 2015; 13:108. [PMID: 26394676 PMCID: PMC4580300 DOI: 10.1186/s12958-015-0104-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 09/11/2015] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND A major concern in ovarian transplants is substantial follicle loss during the initial period of hypoxia. Adipose tissue-derived stem cells (ASCs) have been employed to improve angiogenesis when injected into ischemic tissue. This study evaluated the safety and efficacy of adipose tissue-derived stem cells (ASCs) therapy in the freshly grafted ovaries 30 days after injection. METHODS Rat ASCs (rASCs) obtained from transgenic rats expressing green fluorescent protein (GFP)-(5 × 10(4) cells/ovary) were injected in topic (intact) or freshly grafted ovaries of 30 twelve-week-old adult female Wistar rats. The whole ovary was grafted in the retroperitoneum without vascular anastomosis, immediately after oophorectomy. Vaginal smears were performed daily to assess the resumption of the estrous cycle. Estradiol levels, grafts morphology and follicular viability and density were analyzed. Immunohistochemistry assays were conducted to identify and quantify rASC-GFP(+), VEGF tissue expression, apoptosis (cleaved caspase-3 and TUNEL), and cell proliferation (Ki-67). Quantitative gene expression (qPCR) for VEGF-A, Bcl2, EGF and TGF-β1 was evaluated using RT-PCR and a double labeling immunofluorescence assay for GFP and Von Willebrand Factor (VWF) was performed. RESULTS Grafted ovaries treated with rASC-GFP(+) exhibited earlier resumption of the estrous phase (p < 0.05), increased VEGF-A expression (11-fold in grafted ovaries and 5-fold in topic ovaries vs. control) and an increased number of blood vessels (p < 0.05) in ovarian tissue without leading to apoptosis or cellular proliferation (p > 0.05). Estradiol levels were similar among groups (p > 0.05). rASC-GFP(+) were observed in similar quantities in the topic and grafted ovaries (p > 0.05), and double-labeling for GFP and vWF was observed in both injected groups. CONCLUSION rASC therapy in autologous freshly ovarian grafts could be feasible and safe, induces earlier resumption of the estrous phase and enhances blood vessels in rats. This pilot study may be useful in the future for new researches on frozen-thawed ovarian tissue.
Collapse
Affiliation(s)
- Luciana L Damous
- Laboratório de Ginecologia Estrutural e Molecular (LIM-58), Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Dr Arnaldo av 455, 2nd floor, room 2113, Pacaembu, 01246-903, São Paulo, Brazil.
| | - Juliana S Nakamuta
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (Incor), Faculdade de Medicina da Universidade de São Paulo, Dr Enéas de Carvalho Aguiar Av 44, 10th floor, Cerqueira Cesar, 05403-000, São Paulo, Brazil.
| | - Ana E T Saturi de Carvalho
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (Incor), Faculdade de Medicina da Universidade de São Paulo, Dr Enéas de Carvalho Aguiar Av 44, 10th floor, Cerqueira Cesar, 05403-000, São Paulo, Brazil.
| | - Katia Candido Carvalho
- Laboratório de Ginecologia Estrutural e Molecular (LIM-58), Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Dr Arnaldo av 455, 2nd floor, room 2113, Pacaembu, 01246-903, São Paulo, Brazil.
| | - José Maria Soares
- Laboratório de Ginecologia Estrutural e Molecular (LIM-58), Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Dr Arnaldo av 455, 2nd floor, room 2113, Pacaembu, 01246-903, São Paulo, Brazil.
| | - Manuel de Jesus Simões
- Department of Morphology and Genetics, Universidade Federal de São Paulo (UNIFESP), Botucatu St 740. Ed. Lemos Torres, 2nd floor, Vila Clementino, 04023-009, São Paulo, Brazil.
| | - José Eduardo Krieger
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (Incor), Faculdade de Medicina da Universidade de São Paulo, Dr Enéas de Carvalho Aguiar Av 44, 10th floor, Cerqueira Cesar, 05403-000, São Paulo, Brazil.
| | - Edmund Chada Baracat
- Laboratório de Ginecologia Estrutural e Molecular (LIM-58), Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Dr Arnaldo av 455, 2nd floor, room 2113, Pacaembu, 01246-903, São Paulo, Brazil.
| |
Collapse
|
31
|
Kato Y, Iwata T, Morikawa S, Yamato M, Okano T, Uchigata Y. Allogeneic Transplantation of an Adipose-Derived Stem Cell Sheet Combined With Artificial Skin Accelerates Wound Healing in a Rat Wound Model of Type 2 Diabetes and Obesity. Diabetes 2015; 64:2723-34. [PMID: 25795216 DOI: 10.2337/db14-1133] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Accepted: 03/16/2015] [Indexed: 11/13/2022]
Abstract
One of the most common complications of diabetes is diabetic foot ulcer. Diabetic ulcers do not heal easily due to diabetic neuropathy and reduced blood flow, and nonhealing ulcers may progress to gangrene, which necessitates amputation of the patient's foot. This study attempted to develop a new cell-based therapy for nonhealing diabetic ulcers using a full-thickness skin defect in a rat model of type 2 diabetes and obesity. Allogeneic adipose-derived stem cells (ASCs) were harvested from the inguinal fat of normal rats, and ASC sheets were created using cell sheet technology and transplanted into full-thickness skin defects in Zucker diabetic fatty rats. The results indicate that the transplantation of ASC sheets combined with artificial skin accelerated wound healing and vascularization, with significant differences observed 2 weeks after treatment. The ASC sheets secreted large amounts of several angiogenic growth factors in vitro, and transplanted ASCs were observed in perivascular regions and incorporated into the newly constructed vessel structures in vivo. These results suggest that ASC sheets accelerate wound healing both directly and indirectly in this diabetic wound-healing model. In conclusion, allogeneic ASC sheets exhibit potential as a new therapeutic strategy for the treatment of diabetic ulcers.
Collapse
Affiliation(s)
- Yuka Kato
- Diabetic Center, Tokyo Women's Medical University School of Medicine, Tokyo, Japan Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, Japan
| | - Takanori Iwata
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, Japan
| | - Shunichi Morikawa
- Department of Anatomy and Developmental Biology, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | - Masayuki Yamato
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, Japan
| | - Teruo Okano
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, Japan
| | - Yasuko Uchigata
- Diabetic Center, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| |
Collapse
|
32
|
Damous LL, Nakamuta JS, Saturi de Carvalho AET, Carvalho KC, Soares-Jr JM, Simões MDJ, Krieger JE, Baracat EC. Scaffold-based delivery of adipose tissue-derived stem cells in rat frozen-thawed ovarian autografts: preliminary studies in a rat model. J Assist Reprod Genet 2015; 32:1285-94. [PMID: 26206456 PMCID: PMC4554376 DOI: 10.1007/s10815-015-0527-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Accepted: 06/30/2015] [Indexed: 12/24/2022] Open
Abstract
PURPOSE This study aimed to evaluate whether a gelatin-based Gelfoam sponge is feasible as a scaffold for adipose tissue-derived stem cell (ASC) therapy in rat frozen-thawed ovarian autografts. METHODS Two sets of studies were performed. The in vitro set evaluated ASCs' viability in the Gelfoam scaffold at different times of co-culturing (after 24, 48, 72, 96, and 120 h). The in vivo set used 20 12-week-old adult female Wistar rats. Frozen-thawed ovarian grafts were treated with ASCs delivered in Gelfoam scaffolds immediately after an autologous retroperitoneal transplant (ASCs-GS, n = 10). The controls received Gelfoam with a culture medium (GS, n = 10). Assessment of graft quality was conducted by vaginal smears (until euthanasia on the 30th postoperative day), histological analyses, follicular density, and viability and fibrosis. Immunohistochemical staining for VEGF-A expression, vascular network (vWF), apoptosis (caspase-3 and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL)), cell proliferation (Ki-67), and hormone receptors (estrogen and progesterone) were performed. RESULTS The cells remained viable in Gelfoam for up to 120 h of co-culturing. The graft morphology was similar among the groups. ASC therapy promoted the earlier resumption of the estrous phase (GS 16.6 ± 3 vs. ASCs-GS 12.8 ± 1.3 days) and enhanced estrogen receptors compared with the controls (p < 0.05) without interfering with the quantity and viability of the ovarian follicles, fibrosis, endothelial cells, VEGF immunoexpression, apoptosis, or cell proliferation (p > 0.05). CONCLUSION The Gelfoam scaffold could be a feasible and safe non-invasive technique for ASC delivery in the treatment of frozen-thawed ovarian autografts. Future studies should evaluate the real benefit of this treatment on the survival and endocrine activity of the graft.
Collapse
Affiliation(s)
- Luciana Lamarão Damous
- />Gynecology Discipline, Laboratory of Structural and Molecular Gynecology (LIM-58), Faculdade de Medicina da Universidade de São Paulo, Dr Arnaldo av 455, 2nd floor, room 2113, Pacaembu, São Paulo Brazil 01246-903
- />Galvão Bueno St, 499. Bloco A. Apto31, Liberdade, São Paulo Brazil 01506-000
| | - Juliana Sanajotti Nakamuta
- />Laboratory of Genetics and Molecular Cardiology, Heart Institute (Incor), Faculdade de Medicina da Universidade de São Paulo, Dr Enéas de Carvalho Aguiar Av 44, 10th floor, Cerqueira Cesar, São Paulo Brazil 05403-000
| | - Ana Elisa Teofilo Saturi de Carvalho
- />Laboratory of Genetics and Molecular Cardiology, Heart Institute (Incor), Faculdade de Medicina da Universidade de São Paulo, Dr Enéas de Carvalho Aguiar Av 44, 10th floor, Cerqueira Cesar, São Paulo Brazil 05403-000
| | - Kátia Cândido Carvalho
- />Gynecology Discipline, Laboratory of Structural and Molecular Gynecology (LIM-58), Faculdade de Medicina da Universidade de São Paulo, Dr Arnaldo av 455, 2nd floor, room 2113, Pacaembu, São Paulo Brazil 01246-903
| | - José Maria Soares-Jr
- />Gynecology Discipline, Laboratory of Structural and Molecular Gynecology (LIM-58), Faculdade de Medicina da Universidade de São Paulo, Dr Arnaldo av 455, 2nd floor, room 2113, Pacaembu, São Paulo Brazil 01246-903
| | - Manuel de Jesus Simões
- />Department of Morphology and Genetics, Universidade Federal de São Paulo (UNIFESP), Botucatu St 740. Ed. Lemos Torres, 2nd floor, Vila Clementino, São Paulo, Brazil 04023-009
| | - José Eduardo Krieger
- />Laboratory of Genetics and Molecular Cardiology, Heart Institute (Incor), Faculdade de Medicina da Universidade de São Paulo, Dr Enéas de Carvalho Aguiar Av 44, 10th floor, Cerqueira Cesar, São Paulo Brazil 05403-000
| | - Edmund C. Baracat
- />Gynecology Discipline, Laboratory of Structural and Molecular Gynecology (LIM-58), Faculdade de Medicina da Universidade de São Paulo, Dr Arnaldo av 455, 2nd floor, room 2113, Pacaembu, São Paulo Brazil 01246-903
| |
Collapse
|
33
|
Development of a System and Method for Automated Isolation of Stromal Vascular Fraction from Adipose Tissue Lipoaspirate. Stem Cells Int 2015; 2015:109353. [PMID: 26167182 PMCID: PMC4475713 DOI: 10.1155/2015/109353] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 02/27/2015] [Accepted: 04/09/2015] [Indexed: 01/08/2023] Open
Abstract
Autologous fat grafting for soft tissue reconstruction is challenged by unpredictable long-term graft survival. Fat derived stromal vascular fraction (SVF) is gaining popularity in tissue reconstruction as SVF-enriched fat grafts demonstrate improved engraftment. SVF also has potential in regenerative medicine for remodeling of ischemic tissues by promoting angiogenesis. Since SVF cells do not require culture expansion, attempts are being made to develop automated devices to isolate SVF at the point of care. We report development of a closed, automated system to process up to 500 mL lipoaspirate using cell size-dependent filtration technology. The yield of SVF obtained by automated tissue digestion and filtration (1.17 ± 0.5 × 105 cells/gram) was equivalent to that obtained by manual isolation (1.15 ± 0.3 × 105; p = 0.8), and the viability of the cells isolated by both methods was greater than 90%. Cell composition included CD34+CD31− adipose stromal cells, CD34+CD31+ endothelial progenitor cells, and CD34−CD31+ endothelial cells, and their relative percentages were equivalent to SVF isolated by the manual method. CFU-F capacity and expression of angiogenic factors were also comparable with the manual method, establishing proof-of-concept for fully automated SVF isolation, suitable for use in reconstructive surgeries and regenerative medicine applications.
Collapse
|
34
|
Hart ML, Izeta A, Herrera-Imbroda B, Amend B, Brinchmann JE. Cell Therapy for Stress Urinary Incontinence. TISSUE ENGINEERING PART B-REVIEWS 2015; 21:365-76. [PMID: 25789845 DOI: 10.1089/ten.teb.2014.0627] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Urinary incontinence (UI) is the involuntary loss of urine and is a common condition in middle-aged and elderly women and men. Stress urinary incontinence (SUI) is caused by leakage of urine when coughing, sneezing, laughing, lifting, and exercise, even standing leads to increased intra-abdominal pressure. Other types of UI also exist such as urge incontinence (also called overactive bladder), which is a strong and unexpected sudden urge to urinate, mixed forms of UI that result in symptoms of both urge and stress incontinence, and functional incontinence caused by reduced mobility, cognitive impairment, or neuromuscular limitations that impair mobility or dexterity. However, for many SUI patients, there is significant loss of urethral sphincter muscle due to degeneration of tissue, the strain and trauma of pregnancy and childbirth, or injury acquired during surgery. Hence, for individuals with SUI, a cell-based therapeutic approach to regenerate the sphincter muscle offers the advantage of treating the cause rather than the symptoms. We discuss current clinically relevant cell therapy approaches for regeneration of the external urethral sphincter (striated muscle), internal urethral sphincter (smooth muscle), the neuromuscular synapse, and blood supply. The use of mesenchymal stromal/stem cells is a major step in the right direction, but they may not be enough for regeneration of all components of the urethral sphincter. Inclusion of other cell types or biomaterials may also be necessary to enhance integration and survival of the transplanted cells.
Collapse
Affiliation(s)
- Melanie L Hart
- 1 Clinical Research Group KFO 273, Department of Urology, University of Tübingen , Tübingen, Germany
| | - Ander Izeta
- 2 Tissue Engineering Laboratory, Instituto Biodonostia, Hospital Universitario Donostia , San Sebastian, Spain
| | | | - Bastian Amend
- 4 Department of Urology, University of Tübingen , Tuebingen, Germany
| | - Jan E Brinchmann
- 5 Department of Immunology, Oslo University Hospital, Oslo, Norway
- 6 Norwegian Center for Stem Cell Research, Institute of Basic Medical Sciences, University of Oslo , Oslo, Norway
| |
Collapse
|
35
|
Damous LL, Nakamuta JS, de Carvalho AETS, Soares JM, de Jesus Simões M, Krieger JE, Baracat EC. Adipose tissue-derived stem cell therapy in rat cryopreserved ovarian grafts. Stem Cell Res Ther 2015; 6:57. [PMID: 25889829 PMCID: PMC4416311 DOI: 10.1186/s13287-015-0068-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 01/27/2015] [Accepted: 03/25/2015] [Indexed: 01/14/2023] Open
Abstract
The preliminary results of ovarian transplantation in clinical practice are encouraging. However, the follicular depletion caused by ischemic injury is a main concern and is directly related to short-term graft survival. Cell therapy with adipose tissue-derived stem cells (ASCs) could be an alternative to induce early angiogenesis in the graft. This study aimed to evaluate ASCs therapy in rat cryopreserved ovarian grafts. A single dose of rat ASC (rASCs) or vehicle was injected into the bilateral cryopreserved ovaries of twelve adult female rats immediately after an autologous transplant. Daily vaginal smears were performed for estrous cycle evaluation until euthanasia on postoperative day 30. Follicle viability, graft morphology and apoptosis were assessed. No differences were found with respect to estrous cycle resumption and follicle viability (P > 0.05). However, compared with the vehicle-treated grafts, the morphology of the ASCs-treated grafts was impaired, with diffuse atrophy and increased apoptosis (P < 0.05). ASCs direct injected in the stroma of rat cryopreserved ovarian grafts impaired its morphology although may not interfere with the functional resumption on short-term. Further investigations are necessary to evaluated whether it could compromise their viability in the long-term.
Collapse
Affiliation(s)
- Luciana Lamarão Damous
- Gynecology Discipline, Laboratory of Structural and Molecular Gynecology (LIM-58), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Dr Arnaldo av 455, 2nd floor, room 2113, Pacaembu, São Paulo, 01246-903, Brazil. .,, Galvão Bueno St, 499, Bloco A. Apto31, Liberdade, São Paulo, 01506-000, Brazil.
| | - Juliana Sanajotti Nakamuta
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (Incor), Faculdade de Medicina da Universidade de São Paulo, Dr Enéas de Carvalho Aguiar Av 44, 10th floor, Cerqueira Cesar, São Paulo, 05403-000, Brazil.
| | - Ana Elisa Teófilo Saturi de Carvalho
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (Incor), Faculdade de Medicina da Universidade de São Paulo, Dr Enéas de Carvalho Aguiar Av 44, 10th floor, Cerqueira Cesar, São Paulo, 05403-000, Brazil.
| | - José Maria Soares
- Gynecology Discipline, Laboratory of Structural and Molecular Gynecology (LIM-58), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Dr Arnaldo av 455, 2nd floor, room 2113, Pacaembu, São Paulo, 01246-903, Brazil.
| | - Manuel de Jesus Simões
- Department of Morphology and Genetics, Universidade Federal de São Paulo (UNIFESP), Botucatu St 740. Ed. Lemos Torres, 2nd floor, Vila Clementino, São Paulo, 04023-009, Brazil.
| | - José Eduardo Krieger
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (Incor), Faculdade de Medicina da Universidade de São Paulo, Dr Enéas de Carvalho Aguiar Av 44, 10th floor, Cerqueira Cesar, São Paulo, 05403-000, Brazil.
| | - Edmund C Baracat
- Gynecology Discipline, Laboratory of Structural and Molecular Gynecology (LIM-58), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Dr Arnaldo av 455, 2nd floor, room 2113, Pacaembu, São Paulo, 01246-903, Brazil.
| |
Collapse
|
36
|
Heo SC, Shin WC, Lee MJ, Kim BR, Jang IH, Choi EJ, Lee JS, Kim JH. Periostin accelerates bone healing mediated by human mesenchymal stem cell-embedded hydroxyapatite/tricalcium phosphate scaffold. PLoS One 2015; 10:e0116698. [PMID: 25775460 PMCID: PMC4361583 DOI: 10.1371/journal.pone.0116698] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 12/12/2014] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Periostin, an extracellular matrix protein, is expressed in bone, more specifically, the periosteum and periodontal ligaments, and plays a key role in formation and metabolism of bone tissues. Human adipose tissue-derived mesenchymal stem cells (hASCs) have been reported to differentiate into osteoblasts and stimulate bone repair. However, the role of periostin in hASC-mediated bone healing has not been clarified. In the current study, we examined the effect of periostin on bone healing capacity of hASCs in a critical size calvarial defect model. METHODS AND RESULTS Recombinant periostin protein stimulated migration, adhesion, and proliferation of hASCs in vitro. Implantation of either hASCs or periostin resulted in slight, but not significant, stimulation of bone healing, whereas co-implantation of hASCs together with periostin further potentiated bone healing. In addition, the number of Ki67-positive proliferating cells was significantly increased in calvarial defects by co-implantation of both hASCs and periostin. Consistently, proliferation of administered hASCs was stimulated by co-implantation with periostin in vivo. In addition, co-delivery of hASCs with periostin resulted in markedly increased numbers of CD31-positive endothelial cells and α-SMA-positive arterioles in calvarial defects. CONCLUSIONS These results suggest that recombinant periostin potentiates hASC-mediated bone healing by stimulating proliferation of transplanted hASCs and angiogenesis in calvarial defects.
Collapse
Affiliation(s)
- Soon Chul Heo
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 626-870, Gyeongsangnam-do, Republic of Korea
| | - Won Chul Shin
- Department of Orthopaedic Surgery, School of Medicine, Pusan National University, Yangsan 626-870, Gyeongsangnam-do, Republic of Korea
| | - Mi Jeong Lee
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 626-870, Gyeongsangnam-do, Republic of Korea
| | - Ba Reun Kim
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 626-870, Gyeongsangnam-do, Republic of Korea
| | - Il Ho Jang
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 626-870, Gyeongsangnam-do, Republic of Korea
| | - Eun-Jung Choi
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 626-870, Gyeongsangnam-do, Republic of Korea
| | - Jung Sub Lee
- Department of Orthopaedic Surgery, School of Medicine, Pusan National University, Yangsan 626-870, Gyeongsangnam-do, Republic of Korea
- * E-mail: (JHK); (JSL)
| | - Jae Ho Kim
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 626-870, Gyeongsangnam-do, Republic of Korea
- Research Institute of Convergence Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 626-770, Gyeongsangnam-do, Republic of Korea
- * E-mail: (JHK); (JSL)
| |
Collapse
|
37
|
Wakabayashi K, Hamada C, Kanda R, Nakano T, Io H, Horikoshi S, Tomino Y. Adipose-derived mesenchymal stem cells transplantation facilitate experimental peritoneal fibrosis repair by suppressing epithelial–mesenchymal transition. J Nephrol 2014; 27:507-14. [DOI: 10.1007/s40620-014-0133-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 08/12/2014] [Indexed: 12/30/2022]
|
38
|
Yamamoto M, Rafii S, Rabbany SY. Scaffold biomaterials for nano-pathophysiology. Adv Drug Deliv Rev 2014; 74:104-14. [PMID: 24075835 DOI: 10.1016/j.addr.2013.09.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 09/11/2013] [Accepted: 09/20/2013] [Indexed: 01/20/2023]
Abstract
This review is intended to provide an overview of tissue engineering strategies using scaffold biomaterials to develop a vascularized tissue engineered construct for nano-pathophysiology. Two primary topics are discussed. The first is the biological or synthetic microenvironments that regulate cell behaviors in pathological conditions and tissue regeneration. Second is the use of scaffold biomaterials with angiogenic factors and/or cells to realize vascularized tissue engineered constructs for nano-pathophysiology. These topics are significantly overlapped in terms of three-dimensional (3-D) geometry of cells and blood vessels. Therefore, this review focuses on neovascularization of 3-D scaffold biomaterials induced by angiogenic factors and/or cells. The novel strategy of this approach in nano-pathophysiology is to utilize the vascularized tissue engineered construct as a tissue model to predict the distribution and subsequent therapeutic efficacy of a drug delivery system with different physicochemical and biological properties.
Collapse
Affiliation(s)
- Masaya Yamamoto
- Department of Biomaterials, Institute for Frontier Medical Sciences, Kyoto University, 53 Kawara-cho Shogoin, Sakyo-ku, Kyoto 606-8507, Japan.
| | - Shahin Rafii
- Ansary Stem Cell Institute, Department of Genetic Medicine, Weill Cornell Medical College, 1300 York Ave., New York, NY 10065, USA
| | - Sina Y Rabbany
- Ansary Stem Cell Institute, Department of Genetic Medicine, Weill Cornell Medical College, 1300 York Ave., New York, NY 10065, USA; Bioengineering Program, Hofstra University, 110 Weed Hall, Hempstead, NY 11549, USA
| |
Collapse
|
39
|
Navarro A, Marín S, Riol N, Carbonell-Uberos F, Miñana MD. Human adipose tissue-resident monocytes exhibit an endothelial-like phenotype and display angiogenic properties. Stem Cell Res Ther 2014; 5:50. [PMID: 24731246 PMCID: PMC4055093 DOI: 10.1186/scrt438] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 04/04/2014] [Indexed: 01/01/2023] Open
Abstract
Introduction Adipose tissue has the unique property of expanding throughout adult life, and angiogenesis is required for its growth. However, endothelial progenitor cells contribute minimally to neovascularization. Because myeloid cells have proven to be angiogenic, and monocytes accumulate in expanding adipose tissue, they might contribute to vascularization. Methods The stromal vascular fraction (SVF) cells from human adipose tissue were magnetically separated according to CD45 or CD14 expression. Adipose-derived mesenchymal stromal cells (MSCs) were obtained from SVF CD45- cells. CD14+ monocytes were isolated from peripheral blood (PB) mononuclear cells and then cultured with SVF-derived MSCs. Freshly isolated or cultured cells were characterized with flow cytometry; the conditioned media were analyzed for the angiogenic growth factors, angiopoietin-2 (Ang-2), vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), hepatocyte growth factor (HGF), granulocyte colony-stimulating factor (G-CSF), and granulocyte macrophage colony-stimulating factor (GM-CSF) with Luminex Technology; their angiogenic capacity was determined in an in vivo gelatinous protein mixture (Matrigel) plug angiogenesis assay. Results CD45+ hematopoietic cells within the SVF contain CD14+ cells that co-express the CD34 progenitor marker and the endothelial cell antigens VEGF receptor 2 (VEGFR2/KDR), VEGFR1/Flt1, and Tie2. Co-culture experiments showed that SVF-derived MSCs promoted the acquisition of KDR and Tie-2 in PB monocytes. MSCs secreted significant amounts of Ang-2 and HGF, but minimal amounts of bFGF, G-CSF, or GM-CSF, whereas the opposite was observed for SVF CD14+ cells. Additionally, SVF CD14+ cells secreted significantly higher levels of VEGF and bFGF than did MSCs. Culture supernatants of PB monocytes cultured with MSCs contained significantly higher concentrations of VEGF, HGF, G-CSF, and GM-CSF than did the supernatants from cultures without MSCs. Quantitative analysis of angiogenesis at 14 days after implantation demonstrated that neovascularization of the implants containing SVF CD14+ cells or PB monocytes previously co-cultured with MSCs was 3.5 or 2 times higher than that observed in the implants with SVF-derived MSCs. Moreover, immunofluorescence of Matrigel sections revealed that SVF CD14+ cells differentiated into endothelial cells and contributed to vascular endothelium. Conclusions The results from this study suggest that adipose tissue-resident monocytes should contribute to tissue vascularization. Because SVF CD14+ cells were more efficient in inducing angiogenesis than SVF-derived MSCs, and differentiated into vascular endothelial cells, they may constitute a new cell source for cell-based therapeutic angiogenesis.
Collapse
|
40
|
|