51
|
Jiang L, Jones S, Jia X. Stem Cell Transplantation for Peripheral Nerve Regeneration: Current Options and Opportunities. Int J Mol Sci 2017; 18:ijms18010094. [PMID: 28067783 PMCID: PMC5297728 DOI: 10.3390/ijms18010094] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 12/26/2016] [Accepted: 12/27/2016] [Indexed: 12/21/2022] Open
Abstract
Peripheral nerve regeneration is a complicated process highlighted by Wallerian degeneration, axonal sprouting, and remyelination. Schwann cells play an integral role in multiple facets of nerve regeneration but obtaining Schwann cells for cell-based therapy is limited by the invasive nature of harvesting and donor site morbidity. Stem cell transplantation for peripheral nerve regeneration offers an alternative cell-based therapy with several regenerative benefits. Stem cells have the potential to differentiate into Schwann-like cells that recruit macrophages for removal of cellular debris. They also can secrete neurotrophic factors to promote axonal growth, and remyelination. Currently, various types of stem cell sources are being investigated for their application to peripheral nerve regeneration. This review highlights studies involving the stem cell types, the mechanisms of their action, methods of delivery to the injury site, and relevant pre-clinical or clinical data. The purpose of this article is to review the current point of view on the application of stem cell based strategy for peripheral nerve regeneration.
Collapse
Affiliation(s)
- Liangfu Jiang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China.
| | - Salazar Jones
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Xiaofeng Jia
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China.
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
- Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
52
|
Gao X, Salomon C, Freeman DJ. Extracellular Vesicles from Adipose Tissue-A Potential Role in Obesity and Type 2 Diabetes? Front Endocrinol (Lausanne) 2017; 8:202. [PMID: 28868048 PMCID: PMC5563356 DOI: 10.3389/fendo.2017.00202] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 08/02/2017] [Indexed: 12/16/2022] Open
Abstract
Adipose tissue plays a key role in the development of insulin resistance and its pathological sequelae, such as type 2 diabetes and non-alcoholic fatty liver disease. Dysfunction in the adipose tissue response to storing excess fatty acids as triglyceride can lead to adipose tissue inflammation and spillover of fatty acids from this tissue and accumulation of fatty acids as lipid droplets in ectopic sites, such as liver and muscle. Extracellular vesicles (EVs) are released from adipocytes and have been proposed to be involved in adipocyte/macrophage cross talk and to affect insulin signaling and transforming growth factor β expression in liver cells leading to metabolic disease. Furthermore EV produced by adipose tissue-derived mesenchymal stem cells (ADSC) can promote angiogenesis and cancer cell migration and have neuroprotective and neuroregenerative properties. ADSC EVs have therapeutic potential in vascular and neurodegenerative disease and may also be used to target specific functional miRNAs to cells. Obesity is associated with an increase in adipose-derived EV which may be related to the metabolic complications of obesity. In this review, we discuss our current knowledge of EV produced by adipose tissue and the potential impact of adipose tissue-derived EV on metabolic diseases associated with obesity.
Collapse
Affiliation(s)
- Xuan Gao
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Carlos Salomon
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women’s Hospital, The University of Queensland, Brisbane, QLD, Australia
- Faculty of Pharmacy, Department of Clinical Biochemistry and Immunology, University of Concepción, Concepción, Chile
- Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Ochsner Clinic Foundation, New Orleans, LA, United States
- Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Dilys J. Freeman
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
- *Correspondence: Dilys J. Freeman,
| |
Collapse
|
53
|
Abstract
PURPOSE OF REVIEW To summarize recent literature on basic stem cell research in erectile dysfunction in cavernous nerve injury, aging, diabetes, and Peyronie's disease and to provide a perspective on clinical translation of these cellular therapies. RECENT FINDINGS Stem cell research has been concentrated on mesenchymal stem (stromal) cells from bone marrow and adipose tissue. Application of both cell types has produced positive effects on erectile function in various animal models of erectile dysfunction. In acute animal models, such as cavernous nerve injury-induced erectile dysfunction and chemically induced Peyronie's disease, engraftment and differentiation have not been observed, and stem cells are believed to interact with the host tissue in a paracrine fashion, whereas in chronic disease models some evidence suggests both engraftment and paracrine factors may support improved function. Clinical trials are now investigating therapeutic efficacy of cellular therapy, whereas the first safety studies in humans have recently been published. SUMMARY Evidence from preclinical studies has established stem cells as a potential curative treatment for erectile dysfunction and early phase clinical trials are currently performed.
Collapse
|
54
|
Lo Furno D, Mannino G, Cardile V, Parenti R, Giuffrida R. Potential Therapeutic Applications of Adipose-Derived Mesenchymal Stem Cells. Stem Cells Dev 2016; 25:1615-1628. [PMID: 27520311 DOI: 10.1089/scd.2016.0135] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Stem cells are subdivided into two main categories: embryonic and adult stem cells. In principle, pluripotent embryonic stem cells might differentiate in any cell types of the organism, whereas the potential of adult stem cells would be more restricted. Although adult stem cells from bone marrow have been initially the most extensively studied, those derived from human adipose tissue have been lately more widely investigated, because of several advantages. First, they can be easily obtained in large amounts from subcutaneous adipose tissue, with minimal pain and morbidity for the patients during harvesting. In addition, they feature low immunogenicity and can differentiate not only in cells of mesodermal lineage (adipocytes, osteoblasts, chondrocytes and muscle cells), but also in cells of other germ layers, such as neural or epithelial cells. As their multilineage differentiation capabilities are increasingly highlighted, their possible use in cell-based regenerative medicine is now broadly explored. In fact, starting from in vitro observations, many studies have already entered the preclinical and clinical phases. In this review, because of our main scientific interest, adipogenic, osteogenic, chondrogenic, and neurogenic differentiation abilities of adipose-derived mesenchymal stem cells, as well as their possible therapeutic applications, are chiefly focused. In addition, their ability to differentiate toward muscle, epithelial, pancreatic, and hepatic cells is briefly reported.
Collapse
Affiliation(s)
- Debora Lo Furno
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania , Catania, Italy
| | - Giuliana Mannino
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania , Catania, Italy
| | - Venera Cardile
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania , Catania, Italy
| | - Rosalba Parenti
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania , Catania, Italy
| | - Rosario Giuffrida
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania , Catania, Italy
| |
Collapse
|
55
|
Lindstad T, Qu S, Sikkeland J, Jin Y, Kristian A, Mælandsmo GM, Collas P, Saatcioglu F. STAMP2 is required for human adipose-derived stem cell differentiation and adipocyte-facilitated prostate cancer growth in vivo. Oncotarget 2016; 8:91817-91827. [PMID: 29190878 PMCID: PMC5696144 DOI: 10.18632/oncotarget.11131] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 07/01/2016] [Indexed: 01/01/2023] Open
Abstract
Six Transmembrane Protein of Prostate 2 (STAMP2) has been implicated in both prostate cancer (PCa) and metabolic disease. STAMP2 has unique anti-inflammatory and pro-metabolic properties in mouse adipose tissue, but there is limited information on its role in human metabolic tissues. Using human adipose-derived stem cells (ASCs), we report that STAMP2 expression is dramatically upregulated during adipogenesis. shRNA-mediated STAMP2 knockdown in ASCs significantly suppresses adipogenesis and interferes with optimal expression of adipogenic genes and adipocyte metabolic function. Furthermore, ASC-derived adipocyte-mediated stimulation of prostate tumor growth in nude mice is significantly reduced upon STAMP2 knockdown in ASC adipocytes. These results suggest that STAMP2 is crucial for normal ASC conversion into adipocytes and their metabolic function, as well as their ability to facilitate PCa growth in vivo.
Collapse
Affiliation(s)
| | - Su Qu
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Jørgen Sikkeland
- Department of Biosciences, University of Oslo, Oslo, Norway.,Department of Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
| | - Yang Jin
- Department of Biosciences, University of Oslo, Oslo, Norway.,Department of Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
| | - Alexandr Kristian
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Gunhild M Mælandsmo
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Philippe Collas
- Institute of Basic Medical Sciences, Norwegian Center for Stem Cell Research, University of Oslo, Oslo, Norway
| | - Fahri Saatcioglu
- Department of Biosciences, University of Oslo, Oslo, Norway.,Department of Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
56
|
Zhang P, Li J, Qi Y, Zou Y, Liu L, Tang X, Duan J, Liu H, Zeng G. Vitamin C promotes the proliferation of human adipose-derived stem cells via p53-p21 pathway. Organogenesis 2016; 12:143-151. [PMID: 27231022 DOI: 10.1080/15476278.2016.1194148] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Although adipose-derived stem cells (ADSCs) have demonstrated a promising potential for the applications of cell-based therapy and regenerative medicine, excessive reactive oxygen species (ROS) are harmful to ADSCs cell survival and proliferation. Vitamin C is an important antioxidant, and is often added into culture media as an essential micronutrient. However, its roles on the proliferation of human ADSCs have not been studied. Therefore, in this study, human ADSCs were isolated, and detected by flow cytometry for the analysis of their cell surface antigens. Cell proliferation and cell cycle progression were measured with cell counting kit-8 assay and flow cytometry, respectively. Western blotting was used to detect the expression levels of cyclin E1, p53, p21, and CDK2 proteins. The effect of vitamin C pretreatment on the production of hydrogen peroxide (H2O2)-mediated ROS in the ADSCs was evaluated by flow cytometry. Our results indicated that vitamin C treatment significantly increased cell proliferation, and changed the cell cycle distribution of ADSCs by decreasing the percentage of G1 phase, and concurrently increased the percentage of S and G2/M phase. Western blot analysis indicated that vitamin C treatment up-regulated the expression levels of cyclin E1 and CDK2, but down-regulated p53 and p21 proteins expression, which contributed to cell proliferation and cell cycle progression. Vitamin C pretreatment significantly reduced the production of H2O2-induced ROS in the ADSCs. These findings suggest that vitamin C can promote the proliferation and cell cycle progression in the ADSCs possibly through regulation of p53-p21 signal pathway.
Collapse
Affiliation(s)
- Peihua Zhang
- a Institute of Plastic Surgery, Affiliated Hospital of Guangdong Medical University , Zhanjiang, Guangdong Province , China
| | - Jin Li
- a Institute of Plastic Surgery, Affiliated Hospital of Guangdong Medical University , Zhanjiang, Guangdong Province , China
| | - Yawei Qi
- a Institute of Plastic Surgery, Affiliated Hospital of Guangdong Medical University , Zhanjiang, Guangdong Province , China
| | - Yaqing Zou
- a Institute of Plastic Surgery, Affiliated Hospital of Guangdong Medical University , Zhanjiang, Guangdong Province , China
| | - Li Liu
- a Institute of Plastic Surgery, Affiliated Hospital of Guangdong Medical University , Zhanjiang, Guangdong Province , China
| | - Xudong Tang
- b Institute of Biochemistry and Molecular Biology, Guangdong Medical University , Zhanjiang, Guangdong Province , China
| | - Jianfeng Duan
- a Institute of Plastic Surgery, Affiliated Hospital of Guangdong Medical University , Zhanjiang, Guangdong Province , China
| | - Hongwei Liu
- c Department of Plastic Surgery , the First Affiliated Hospital of Jinan University, Key Laboratory for Regenerative Medicine, Ministry of Education , Guangzhou, Guangdong Province , China
| | - Guofang Zeng
- a Institute of Plastic Surgery, Affiliated Hospital of Guangdong Medical University , Zhanjiang, Guangdong Province , China
| |
Collapse
|
57
|
Pachón-Peña G, Serena C, Ejarque M, Petriz J, Duran X, Oliva-Olivera W, Simó R, Tinahones FJ, Fernández-Veledo S, Vendrell J. Obesity Determines the Immunophenotypic Profile and Functional Characteristics of Human Mesenchymal Stem Cells From Adipose Tissue. Stem Cells Transl Med 2016; 5:464-75. [PMID: 26956208 DOI: 10.5966/sctm.2015-0161] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 11/19/2015] [Indexed: 12/14/2022] Open
Abstract
UNLABELLED Adipose tissue is a major source of mesenchymal stem cells (MSCs), which possess a variety of properties that make them ideal candidates for regenerative and immunomodulatory therapies. Here, we compared the immunophenotypic profile of human adipose-derived stem cells (hASCs) from lean and obese individuals, and explored its relationship with the apparent altered plasticity of hASCs. We also hypothesized that persistent hypoxia treatment of cultured hASCs may be necessary but not sufficient to drive significant changes in mature adipocytes. hASCs were obtained from subcutaneous adipose tissue of healthy, adult, female donors undergoing abdominal plastic surgery: lean (n=8; body mass index [BMI]: 23±1 kg/m2) and obese (n=8; BMI: 35±5 kg/m2). Cell surface marker expression, proliferation and migration capacity, and adipogenic differentiation potential of cultured hASCs at two different oxygen conditions were studied. Compared with lean-derived hASCs, obese-derived hASCs demonstrated increased proliferation and migration capacity but decreased lipid droplet accumulation, correlating with a higher expression of human leukocyte antigen (HLA)-II and cluster of differentiation (CD) 106 and lower expression of CD29. Of interest, adipogenic differentiation modified CD106, CD49b, HLA-ABC surface protein expression, which was dependent on the donor's BMI. Additionally, low oxygen tension increased proliferation and migration of lean but not obese hASCs, which correlated with an altered CD36 and CD49b immunophenotypic profile. In summary, the differences observed in proliferation, migration, and differentiation capacity in obese hASCs occurred in parallel with changes in cell surface markers, both under basal conditions and during differentiation. Therefore, obesity is an important determinant of stem cell function independent of oxygen tension. SIGNIFICANCE The obesity-related hypoxic environment may have latent effects on human adipose tissue-derived mesenchymal stem cells (hASCs) with potential consequences in mature cells. This study explores the immunophenotypic profile of hASCs obtained from lean and obese individuals and its potential relationship with the altered plasticity of hASCs observed in obesity. In this context, an altered pattern of cell surface marker expression in obese-derived hASCs in both undifferentiated and differentiated stages is demonstrated. Differences in proliferation, migration, and differentiation capacity of hASCs from obese adipose tissue correlated with alterations in cell surface expression. Remarkably, altered plasticity observed in obese-derived hASCs was maintained in the absence of hypoxia, suggesting that these cells might be obesity conditioned.
Collapse
Affiliation(s)
- Gisela Pachón-Peña
- Hospital Universitari de Tarragona Joan XXIII, Institut d´Investigació Sanitària Pere Virgili Universitat Rovira i Virgili, Tarragona, Spain CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Carolina Serena
- Hospital Universitari de Tarragona Joan XXIII, Institut d´Investigació Sanitària Pere Virgili Universitat Rovira i Virgili, Tarragona, Spain CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Miriam Ejarque
- Hospital Universitari de Tarragona Joan XXIII, Institut d´Investigació Sanitària Pere Virgili Universitat Rovira i Virgili, Tarragona, Spain CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Jordi Petriz
- Institut de Recerca Contra la Leucemia Josep Carreras, Barcelona, Spain
| | - Xevi Duran
- Hospital Universitari de Tarragona Joan XXIII, Institut d´Investigació Sanitària Pere Virgili Universitat Rovira i Virgili, Tarragona, Spain CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - W Oliva-Olivera
- Hospital Universitario Virgen de la Victoria, Instituto de Investigaciones Biomédicas de Málaga (IBIMA), Universidad de Málaga, Málaga, Spain CIBER de Fisiopatología de Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Rafael Simó
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain Diabetes and Metabolism Research Unit, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Francisco J Tinahones
- Hospital Universitario Virgen de la Victoria, Instituto de Investigaciones Biomédicas de Málaga (IBIMA), Universidad de Málaga, Málaga, Spain CIBER de Fisiopatología de Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Sonia Fernández-Veledo
- Hospital Universitari de Tarragona Joan XXIII, Institut d´Investigació Sanitària Pere Virgili Universitat Rovira i Virgili, Tarragona, Spain CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Joan Vendrell
- Hospital Universitari de Tarragona Joan XXIII, Institut d´Investigació Sanitària Pere Virgili Universitat Rovira i Virgili, Tarragona, Spain CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
58
|
US stem cell clinics, patient safety, and the FDA. Trends Mol Med 2016; 21:271-3. [PMID: 25945404 DOI: 10.1016/j.molmed.2015.02.008] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 02/18/2015] [Accepted: 02/20/2015] [Indexed: 11/22/2022]
Abstract
Scholarship on patients accessing unproven stem cell interventions is dominated by research addressing 'stem cell tourism' to such countries as China, India, Mexico, and the Ukraine. However, clinics marketing 'adipose-derived mesenchymal stem cell treatments' are proliferating across the USA. These businesses typically claim to operate in compliance with federal regulations, but careful review of their commercial practices suggests that such clinics are marketing unapproved and noncompliant biological drugs.
Collapse
|
59
|
Haahr MK, Jensen CH, Toyserkani NM, Andersen DC, Damkier P, Sørensen JA, Lund L, Sheikh SP. Safety and Potential Effect of a Single Intracavernous Injection of Autologous Adipose-Derived Regenerative Cells in Patients with Erectile Dysfunction Following Radical Prostatectomy: An Open-Label Phase I Clinical Trial. EBioMedicine 2016; 5:204-10. [PMID: 27077129 PMCID: PMC4816754 DOI: 10.1016/j.ebiom.2016.01.024] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Revised: 01/12/2016] [Accepted: 01/18/2016] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Prostate cancer is the most common cancer in men, and radical prostatectomy (RP) often results in erectile dysfunction (ED) and a substantially reduced quality of life. The efficacy of current interventions, principal treatment with PDE-5 inhibitors, is not satisfactory and this condition presents an unmet medical need. Preclinical studies using adipose-derived stem cells to treat ED have shown promising results. Herein, we report the results of a human phase 1 trial with autologous adipose-derived regenerative cells (ADRCs) freshly isolated after a liposuction. METHODS Seventeen men suffering from post RP ED, with no recovery using conventional therapy, were enrolled in a prospective phase 1 open-label and single-arm study. All subjects had RP performed 5-18 months before enrolment, and were followed for 6 months after intracavernosal transplantation. ADRCs were analyzed for the presence of stem cell surface markers, viability and ability to differentiate. Primary endpoint was the safety and tolerance of the cell therapy while the secondary outcome was improvement of erectile function. Any adverse events were reported and erectile function was assessed by IIEF-5 scores. The study is registered with ClinicalTrials.gov, NCT02240823. FINDINGS Intracavernous injection of ADRCs was well-tolerated and only minor events related to the liposuction and cell injections were reported at the one-month evaluation, but none at later time points. Overall during the study period, 8 of 17 men recovered their erectile function and were able to accomplish sexual intercourse. Post-hoc stratification according to urinary continence status was performed. Accordingly, for continent men (median IIEFinclusion = 7 (95% CI 5-12), 8 out of 11 men recovered erectile function (IIEF6months = 17 (6-23)), corresponding to a mean difference of 0.57 (0.38-0.85; p = 0.0069), versus inclusion. In contrast, incontinent men did not regain erectile function (median IIEF1/3/6 months = 5 (95% CI 5-6); mean difference 1 (95% CI 0.85-1.18), p > 0.9999). INTERPRETATION In this phase I trial a single intracavernosal injection of freshly isolated autologous ADRCs was a safe procedure. A potential efficacy is suggested by a significant improvement in IIEF-5 scores and erectile function. We suggest that ADRCs represent a promising interventional therapy of ED following prostatectomy. FUNDING Danish Medical Research Council, Odense University Hospital and the Danish Cancer Society.
Collapse
Key Words
- ADRC, adipose-derived regenerative cells
- Adipose-derived regenerative cells
- Adipose-derived stem cells
- Adipose-derived stromal vascular fraction
- BMI, body mass index
- CFU-F, fibroblastoid colony forming units
- Cell therapy
- Clinical trial
- ED, erectile dysfunction
- EHS, erection hardness score
- Erectile dysfunction
- ICIQ-UI SF, incontinence questionnaire – urinary incontinence – short form questionnaire
- IIEF-5, international index of erectile function-5
- LUTS, lower urinary tract symptoms
- NSAID, nonsteroidal antiinflammatory drug
- PDE-5, phosphodiesterase-5
- RP, radical prostatectomy
- SVF, stromal vascular fraction
Collapse
Affiliation(s)
- Martha Kirstine Haahr
- Department of Urology, Odense University Hospital, Sdr. Boulevard 29, 5000 Odense C, Denmark; The Danish Centre for Regenerative Medicine (www.danishcrm.com); Odense University Hospital, Denmark; Clinical Institute, University of Southern Denmark, 5000 Odense C, Denmark
| | - Charlotte Harken Jensen
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark; The Danish Centre for Regenerative Medicine (www.danishcrm.com); Odense University Hospital, Denmark
| | - Navid Mohamadpour Toyserkani
- Department of Plastic Surgery, Odense University Hospital, Odense, Denmark; The Danish Centre for Regenerative Medicine (www.danishcrm.com); Odense University Hospital, Denmark; Clinical Institute, University of Southern Denmark, 5000 Odense C, Denmark
| | - Ditte Caroline Andersen
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark; The Danish Centre for Regenerative Medicine (www.danishcrm.com); Odense University Hospital, Denmark; Clinical Institute, University of Southern Denmark, 5000 Odense C, Denmark
| | - Per Damkier
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark; Clinical Institute, University of Southern Denmark, 5000 Odense C, Denmark
| | - Jens Ahm Sørensen
- Department of Plastic Surgery, Odense University Hospital, Odense, Denmark; The Danish Centre for Regenerative Medicine (www.danishcrm.com); Odense University Hospital, Denmark; Clinical Institute, University of Southern Denmark, 5000 Odense C, Denmark
| | - Lars Lund
- Department of Urology, Odense University Hospital, Sdr. Boulevard 29, 5000 Odense C, Denmark; The Danish Centre for Regenerative Medicine (www.danishcrm.com); Odense University Hospital, Denmark; Clinical Institute, University of Southern Denmark, 5000 Odense C, Denmark
| | - Søren Paludan Sheikh
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark; Institute of Molecular Medicine, University of Southern Denmark, Winsloewparken 21 3rd, 5000 Odense C, Denmark; The Danish Centre for Regenerative Medicine (www.danishcrm.com); Odense University Hospital, Denmark
| |
Collapse
|
60
|
Strickland LR, Collawn SS. Steroid atrophy scarring treated with fat grafting in a patient with complex regional pain syndrome: A case report. J COSMET LASER THER 2016; 18:165-9. [PMID: 26735938 DOI: 10.3109/14764172.2015.1114639] [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] [Indexed: 01/14/2023]
Abstract
Subcutaneous atrophy is a known complication of steroid injections. Excellent results with fat grafting for the treatment of steroid atrophy have been documented. However, the benefit of treating steroid-induced subcutaneous atrophy in an extremity diagnosed with complex regional pain syndrome (CRPS) has not been described. CRPS, known formerly as reflex sympathetic dystrophy or RSD, causalgia, or reflex neurovascular dystrophy, is a severe, progressive musculoskeletal pain syndrome characterized by pain which is disproportionate to the severity of the inciting event, edema, or skin changes. Common treatment modalities include pharmacotherapy, physical therapy, and nerve blocks-each therapy producing varying results. We present a literature review of CRPS and the case of a 15-year-old female who developed CRPS of the left lower leg after arthroscopic debridement with retrograde drilling of an osteochondral lesion. Steroid atrophy of the involved area following a saphenous nerve block complicated the patient's treatment course. The area of atrophy was treated with autologous fat grafting. Following the adipose injection procedure, the patient experienced almost complete resolution of her CPRS-associated pain symptoms, along with improved cosmetic appearance of the area.
Collapse
Affiliation(s)
- Leah R Strickland
- a Medicine, University of Alabama School of Medicine , Birmingham , AL , USA
| | - Sherry S Collawn
- b Plastic Surgery, University of Alabama at Birmingham , Birmingham , AL , USA
| |
Collapse
|
61
|
Allogeneic Platelet Releasate Preparations Derived via a Novel Rapid Thrombin Activation Process Promote Rapid Growth and Increased BMP-2 and BMP-4 Expression in Human Adipose-Derived Stem Cells. Stem Cells Int 2015; 2016:7183734. [PMID: 26823671 PMCID: PMC4707349 DOI: 10.1155/2016/7183734] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 10/01/2015] [Indexed: 01/03/2023] Open
Abstract
The administration of human adipose-derived stem cells (ASCs) represents a promising regenerative therapy for the treatment of orthopedic injuries. While ASCs can be easily isolated from liposuction-derived adipose tissue, most clinical applications will likely require in vitro culture expansion of these cells using nonxenogeneic components. In this study, platelet releasate was generated using a novel rapid thrombin activation method (tPR). ASCs grown in media supplemented with tPR proliferated much faster than ASCs grown in media supplemented with 10% fetal bovine serum. The cells also retained the ability to differentiate along chondrogenic, adipogenic, and osteogenic lineages. The tPR cultured ASCs displayed elevated expression of BMP-4 (5.7 ± 0.97-fold increase) and BMP-2 (4.7 ± 1.3-fold increase) and decreased expression of PDGF-B (4.0 ± 1.4-fold decrease) and FGF-2 (33 ± 9.0-fold decrease). No significant changes in expression were seen with TGF-β and VEGF. This pattern of gene expression was consistent across different allogeneic tPR samples and different ASC lines. The use of allogeneic rapidly activated tPR to culture ASCs is associated with both an increased cell yield and a defined gene expression profile making it an attractive option for cell expansion prior to cell-based therapy for orthopedic applications.
Collapse
|
62
|
Fernández-Trasancos Á, Guerola-Segura R, Paradela-Dobarro B, Álvarez E, García-Acuña JM, Fernández ÁL, González-Juanatey JR, Eiras S. Glucose and Inflammatory Cells Decrease Adiponectin in Epicardial Adipose Tissue Cells: Paracrine Consequences on Vascular Endothelium. J Cell Physiol 2015; 231:1015-23. [DOI: 10.1002/jcp.25189] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 09/08/2015] [Indexed: 12/12/2022]
Affiliation(s)
- Ángel Fernández-Trasancos
- Cardiology Group, Health Research Institute; University Clinical Hospital of Santiago de Compostela; Santiago de Compostela Spain
| | - Raquel Guerola-Segura
- Department of Cardiology, Coronary Care Unit; University Clinical Hospital of Santiago de Compostela; Santiago de Compostela Spain
| | - Beatriz Paradela-Dobarro
- Cardiology Group, Health Research Institute; University Clinical Hospital of Santiago de Compostela; Santiago de Compostela Spain
| | - Ezequiel Álvarez
- Cardiology Group, Health Research Institute; University Clinical Hospital of Santiago de Compostela; Santiago de Compostela Spain
| | - José María García-Acuña
- Cardiology Group, Health Research Institute; University Clinical Hospital of Santiago de Compostela; Santiago de Compostela Spain
- Department of Cardiology, Coronary Care Unit; University Clinical Hospital of Santiago de Compostela; Santiago de Compostela Spain
| | - Ángel Luis Fernández
- Department of Heart Surgery; University Clinical Hospital of Santiago de Compostela; Santiago de Compostela Spain
| | - José Ramón González-Juanatey
- Cardiology Group, Health Research Institute; University Clinical Hospital of Santiago de Compostela; Santiago de Compostela Spain
- Department of Cardiology, Coronary Care Unit; University Clinical Hospital of Santiago de Compostela; Santiago de Compostela Spain
| | - Sonia Eiras
- Cardiology Group, Health Research Institute; University Clinical Hospital of Santiago de Compostela; Santiago de Compostela Spain
| |
Collapse
|
63
|
Human Adipose-Derived Mesenchymal Stem Cells Modulate Experimental Autoimmune Arthritis by Modifying Early Adaptive T Cell Responses. Stem Cells 2015. [DOI: 10.1002/stem.2113] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
64
|
Turner LG. Federal Regulatory Oversight of US Clinics Marketing Adipose-Derived Autologous Stem Cell Interventions: Insights From 3 New FDA Draft Guidance Documents. Mayo Clin Proc 2015; 90:567-71. [PMID: 25939934 DOI: 10.1016/j.mayocp.2015.02.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Revised: 02/03/2015] [Accepted: 02/06/2015] [Indexed: 12/15/2022]
Affiliation(s)
- Leigh G Turner
- Center for Bioethics, School of Public Health, University of Minnesota, Minneapolis, MN.
| |
Collapse
|
65
|
Kingham PJ, Reid AJ, Wiberg M. Adipose-derived stem cells for nerve repair: hype or reality? Cells Tissues Organs 2015; 200:23-30. [PMID: 25825218 DOI: 10.1159/000369336] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2014] [Indexed: 11/19/2022] Open
Abstract
Peripheral nerve injury is a relatively commonly occurring trauma which seriously compromises the quality of life for many individuals. There is a major need to devise new treatment strategies, and one possible approach is to develop cellular therapies to bioengineer new nerve tissue and/or modulate the endogenous regenerative mechanisms within the peripheral nervous system. In this short review we describe how stem cells isolated from adipose tissue could be a suitable element of this approach. We describe the possible mechanisms through which the stem cells might exert a positive influence on peripheral nerve regeneration. These include their ability to differentiate into cells resembling Schwann cells and their secretion of a plethora of neurotrophic growth factors. We also review the literature describing the effects of these cells when tested using in vivo peripheral nerve injury models.
Collapse
|
66
|
Yeh DC, Chan TM, Harn HJ, Chiou TW, Chen HS, Lin ZS, Lin SZ. Adipose Tissue-Derived Stem Cells in Neural Regenerative Medicine. Cell Transplant 2015; 24:487-92. [PMID: 25647067 DOI: 10.3727/096368915x686940] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Adipose tissue-derived stem cells (ADSCs) have two essential characteristics with regard to regenerative medicine: the convenient and efficient generation of large numbers of multipotent cells and in vitro proliferation without a loss of stemness. The implementation of clinical trials has prompted widespread concern regarding safety issues and has shifted research toward the therapeutic efficacy of stem cells in dealing with neural degeneration in cases such as stroke, amyotrophic lateral sclerosis, Parkinson's disease, Alzheimer's disease, Huntington's disease, cavernous nerve injury, and traumatic brain injury. Most existing studies have reported that cell therapies may be able to replenish lost cells and promote neuronal regeneration, protect neuronal survival, and play a role in overcoming permanent paralysis and loss of sensation and the recovery of neurological function. The mechanisms involved in determining therapeutic capacity remain largely unknown; however, this concept can still be classified in a methodical manner by citing current evidence. Possible mechanisms include the following: 1) the promotion of angiogenesis, 2) the induction of neuronal differentiation and neurogenesis, 3) reductions in reactive gliosis, 4) the inhibition of apoptosis, 5) the expression of neurotrophic factors, 6) immunomodulatory function, and 7) facilitating neuronal integration. In this study, several human clinical trials using ADSCs for neuronal disorders were investigated. It is suggested that ADSCs are one of the choices among various stem cells for translating into clinical application in the near future.
Collapse
Affiliation(s)
- Da-Chuan Yeh
- Department of Internal Medicine, China Medical University Beigan Hospital, Yunlin, Taiwan
| | - Tzu-Min Chan
- Department of Medical Education and Research, China Medical University Beigan Hospital, Yunlin, Taiwan
- Department of Medical Education and Research, China Medical University-An-Nan Hospital, Tainan, Taiwan
| | - Horng-Jyh Harn
- Department of Medicine, China Medical University, Taichung, Taiwan
- Department of Pathology, China Medical University Hospital, Taichung, Taiwan
| | - Tzyy-Wen Chiou
- Department of Life Science and Graduate Institute of Biotechnology, National Dong Hwa University, Hualien, Taiwan
| | - Hsin-Shui Chen
- School of Medicine, China Medical University, Taichung, Taiwan
- Department of Physical Medicine and Rehabilitation, China Medical University Beigang Hospital, Yunlin, Taiwan
| | - Zung-Sheng Lin
- Department of Surgery, China Medical University Beigan Hospital, Yunlin, Taiwan
| | - Shinn-Zong Lin
- Center for Neuropsychiatry, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Immunology, China Medical University, Taichung, Taiwan
- Department of Neurosurgery, China Medical University Beigan Hospital, Yunlin, Taiwan
- Department of Neurosurgery, Tainan Municipal An-Nan Hospital-China Medical University, Tainan, Taiwan
| |
Collapse
|
67
|
Zack-Williams SDL, Butler PE, Kalaskar DM. Current progress in use of adipose derived stem cells in peripheral nerve regeneration. World J Stem Cells 2015; 7:51-64. [PMID: 25621105 PMCID: PMC4300936 DOI: 10.4252/wjsc.v7.i1.51] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 09/25/2014] [Accepted: 10/29/2014] [Indexed: 02/06/2023] Open
Abstract
Unlike central nervous system neurons; those in the peripheral nervous system have the potential for full regeneration after injury. Following injury, recovery is controlled by schwann cells which replicate and modulate the subsequent immune response. The level of nerve recovery is strongly linked to the severity of the initial injury despite the significant advancements in imaging and surgical techniques. Multiple experimental models have been used with varying successes to augment the natural regenerative processes which occur following nerve injury. Stem cell therapy in peripheral nerve injury may be an important future intervention to improve the best attainable clinical results. In particular adipose derived stem cells (ADSCs) are multipotent mesenchymal stem cells similar to bone marrow derived stem cells, which are thought to have neurotrophic properties and the ability to differentiate into multiple lineages. They are ubiquitous within adipose tissue; they can form many structures resembling the mature adult peripheral nervous system. Following early in vitro work; multiple small and large animal in vivo models have been used in conjunction with conduits, autografts and allografts to successfully bridge the peripheral nerve gap. Some of the ADSC related neuroprotective and regenerative properties have been elucidated however much work remains before a model can be used successfully in human peripheral nerve injury (PNI). This review aims to provide a detailed overview of progress made in the use of ADSC in PNI, with discussion on the role of a tissue engineered approach for PNI repair.
Collapse
|
68
|
Chan TM, Harn HJ, Lin HP, Chiu SC, Lin PC, Wang HI, Ho LI, Chuu CP, Chiou TW, Hsieh AC, Chen YW, Ho WY, Lin SZ. The use of ADSCs as a treatment for chronic stroke. Cell Transplant 2015; 23:541-7. [PMID: 24816449 DOI: 10.3727/096368914x678409] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Stroke is one of the disorders for which clinically effective therapeutic modalities are most needed, and numerous ways have been explored to attempt to investigate their feasibilities. However, ischemic- or hemorrhagic-induced inflammatory neuron death causes irreversible injuries and infarction regions, and there are currently no truly effective drugs available as therapy. It is therefore urgent to be able to provide a fundamental treatment method to regenerate neuronal brain cells, and therefore, the use of stem cells for curing chronic stroke could be a major breakthrough development. In this review, we describe the features and classification of stroke and focus on the benefits of adipose tissue-derived stem cells and their applications in stroke animal models. The results show that cell-based therapies have resulted in significant improvements in neuronal behaviors and functions through different molecular mechanisms, and no safety problems have so far arisen after transplantation. Further, we propose a clinical possibility to create a homing niche by reducing the degree of invasive intracerebroventricular transplantation and combining it with continuous intravenous administration to achieve a complete cure.
Collapse
Affiliation(s)
- Tzu-Min Chan
- Center for Neuropsychiatry, China Medical University Hospital, Taichung, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
69
|
Efimenko AY, Kochegura TN, Akopyan ZA, Parfyonova YV. Autologous Stem Cell Therapy: How Aging and Chronic Diseases Affect Stem and Progenitor Cells. Biores Open Access 2015; 4:26-38. [PMID: 26309780 PMCID: PMC4497652 DOI: 10.1089/biores.2014.0042] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
During recent years different types of adult stem/progenitor cells have been successfully applied for the treatment of many pathologies, including cardiovascular diseases. The regenerative potential of these cells is considered to be due to their high proliferation and differentiation capacities, paracrine activity, and immunologic privilege. However, therapeutic efficacy of the autologous stem/progenitor cells for most clinical applications remains modest, possibly because of the attenuation of their regenerative potential in aged patients with chronic diseases such as cardiovascular diseases and metabolic disorders. In this review we will discuss the risk factors affecting the therapeutic potential of adult stem/progenitor cells as well as the main approaches to mitigating them using the methods of regenerative medicine.
Collapse
Affiliation(s)
- Anastasia Yu. Efimenko
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russian Federation
| | - Tatiana N. Kochegura
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russian Federation
| | - Zhanna A. Akopyan
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russian Federation
| | - Yelena V. Parfyonova
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russian Federation
- Laboratory of Angiogenesis, Russian Cardiology Research and Production Complex, Moscow, Russian Federation
| |
Collapse
|
70
|
Dzhoyashvili NA, Efimenko AY, Kochegura TN, Kalinina NI, Koptelova NV, Sukhareva OY, Shestakova MV, Akchurin RS, Tkachuk VA, Parfyonova YV. Disturbed angiogenic activity of adipose-derived stromal cells obtained from patients with coronary artery disease and diabetes mellitus type 2. J Transl Med 2014; 12:337. [PMID: 25491476 PMCID: PMC4268805 DOI: 10.1186/s12967-014-0337-4] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Accepted: 11/21/2014] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Multipotent mesenchymal stem/stromal cells (MSC) including adipose-derived stromal cells (ADSC) have been successfully applied for cardiovascular diseases treatment. Their regenerative potential is considered due to the multipotency, paracrine activity and immunologic privilege. However, therapeutic efficacy of autologous MSC for myocardial ischemia therapy is modest. We analyzed if ADSC properties are attenuated in patients with chronic diseases such as coronary artery disease (CAD) and diabetes mellitus type 2 (T2DM). METHODS AND RESULTS ADSC were isolated from subcutaneous fat tissue of patients without established cardiovascular diseases and metabolic disorders (control group, n = 19), patients with CAD only (n = 32) and patients with CAD and T2DM (n = 28). ADSC phenotype (flow cytometry) was CD90(+)/CD73(+)/CD105(+)/CD45(-)/CD31(-) and they were capable of adipogenic and osteogenic differentiation. ADSC morphology and immunophenotype were similar for all patients, but ADSC from patients with CAD and T2DM had higher proliferation activity and shorter telomeres compared to control patients. ADSC conditioned media stimulated capillary-like tubes formation by endothelial cells (EA.hy926), but this effect significantly decreased for patients with CAD (p = 0.03) and with CAD + T2DM (p = 0.017) compared to the control group. Surprisingly we revealed significantly higher secretion of some pro-angiogenic factors (ELISA) by ADSC: vascular endothelial growth factor (VEGF) and hepatocyte growth factor (HGF) for patients with CAD and HGF and placental growth factor (PlGF) for patients with CAD + T2DM. Among angiogenesis inhibitors such as thrombospondin-1, endostatin and plasminogen activator inhibitor-1 (PAI-1) level of PAI-1 in ADSC conditioned media was significantly higher for patients with CAD and CAD + T2DM compared to the control group (p < 0.01). Inhibition of PAI-1 in ADSC conditioned media by neutralizing antibodies partially restored ADSC angiogenic activity (p = 0.017). CONCLUSIONS ADSC angiogenic activity is significantly declined in patients with CAD and T2DM, which could restrict the effectiveness of autologous ADSC cell therapy in these cohorts of patients. This impairment might be due to the disturbance in coordinated network of pro- and anti-angiogenic growth factors secreted by ADSC. Changes in ADSC secretome differ between patients with CAD and T2DM and further investigation are necessary to reveal the MSC-involved mechanisms of cardiovascular and metabolic diseases and develop novel approaches to their correction using the methods of regenerative medicine.
Collapse
Affiliation(s)
- Nina A Dzhoyashvili
- Russian Cardiology Research and Production Complex, Moscow, Russian Federation. .,Faculty of Medicine, Lomonosov Moscow State University, Lomonosovsky av. 31/5, Moscow, 119192, Russian Federation.
| | - Anastasia Yu Efimenko
- Faculty of Medicine, Lomonosov Moscow State University, Lomonosovsky av. 31/5, Moscow, 119192, Russian Federation.
| | - Tatiana N Kochegura
- Faculty of Medicine, Lomonosov Moscow State University, Lomonosovsky av. 31/5, Moscow, 119192, Russian Federation.
| | - Natalia I Kalinina
- Faculty of Medicine, Lomonosov Moscow State University, Lomonosovsky av. 31/5, Moscow, 119192, Russian Federation.
| | - Natalia V Koptelova
- Faculty of Medicine, Lomonosov Moscow State University, Lomonosovsky av. 31/5, Moscow, 119192, Russian Federation.
| | - Olga Yu Sukhareva
- Institute of Diabetes Mellitus, Endocrinology Research Centre, Moscow, Russian Federation.
| | - Marina V Shestakova
- Institute of Diabetes Mellitus, Endocrinology Research Centre, Moscow, Russian Federation.
| | - Renat S Akchurin
- Russian Cardiology Research and Production Complex, Moscow, Russian Federation.
| | - Vsevolod A Tkachuk
- Russian Cardiology Research and Production Complex, Moscow, Russian Federation. .,Faculty of Medicine, Lomonosov Moscow State University, Lomonosovsky av. 31/5, Moscow, 119192, Russian Federation.
| | - Yelena V Parfyonova
- Russian Cardiology Research and Production Complex, Moscow, Russian Federation. .,Faculty of Medicine, Lomonosov Moscow State University, Lomonosovsky av. 31/5, Moscow, 119192, Russian Federation.
| |
Collapse
|
71
|
Okura H, Soeda M, Morita M, Fujita M, Naba K, Ito C, Ichinose A, Matsuyama A. Therapeutic potential of human adipose tissue-derived multi-lineage progenitor cells in liver fibrosis. Biochem Biophys Res Commun 2014; 456:860-5. [PMID: 25490388 DOI: 10.1016/j.bbrc.2014.11.122] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Accepted: 11/26/2014] [Indexed: 01/16/2023]
Abstract
INTRODUCTION Liver fibrosis is characterized by excessive accumulation of extracellular matrix. In a mouse model of liver fibrosis, systemic injection of bone marrow mesenchymal stem cells (BM-MSCs) was considered to rescue the diseased phenotype. The aim of this study was to assess the effectiveness of human adipose tissue-derived multi-lineage progenitor cells (hADMPCs) in improving liver fibrosis. METHODS AND RESULTS hADMPCs were isolated from subcutaneous adipose tissues of healthy volunteers and expanded. Six week-old male nude mice were treated with carbon tetra-chloride (CCl4) by intraperitoneal injection twice a week for 6 weeks, followed by a tail vein injection of hADMPCs or placebo control. After 6 more weeks of CCl4 injection (12 weeks in all), nude mice with hADMPCs transplants exhibited a significant reduction in liver fibrosis, as evidenced by Sirius Red staining, compared with nude mice treated with CCl4 for 12 weeks without hADMPCs transplants. Moreover, serum glutamic pyruvate transaminase and total bilirubin levels in hADMPCs-treated nude mice were lower levels than those in placebo controls. Production of fibrinolytic enzyme MMPs from hADMPCs were examined by ELISA and compared to that from BM-MSCs. MMP-2 levels in the culture media were not significantly different, whereas those of MMP-3 and -9 of hADMPCs were higher than those by BM-MSCs. CONCLUSION These results showed the mode of action and proof of concept of systemic injection of hADMPCs, which is a promising therapeutic intervention for the treatment of patients with liver fibrosis.
Collapse
Affiliation(s)
- Hanayuki Okura
- Platform of Therapeutics for Rare Disease, National Institute of Biomedical Innovation, 5-5-2-602 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan; The Center for Medical Engineering and Informatics, Osaka University, 2-2 Yamada-oka, Suita, Osaka 565-0879, Japan
| | - Mayumi Soeda
- Platform of Therapeutics for Rare Disease, National Institute of Biomedical Innovation, 5-5-2-602 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Mitsuko Morita
- Platform of Therapeutics for Rare Disease, National Institute of Biomedical Innovation, 5-5-2-602 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Maiko Fujita
- Platform of Therapeutics for Rare Disease, National Institute of Biomedical Innovation, 5-5-2-602 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Kyoko Naba
- Platform of Therapeutics for Rare Disease, National Institute of Biomedical Innovation, 5-5-2-602 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Chiyoko Ito
- Platform of Therapeutics for Rare Disease, National Institute of Biomedical Innovation, 5-5-2-602 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Akihiro Ichinose
- Department of Plastic Surgery, Kobe University Hospital, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, Japan
| | - Akifumi Matsuyama
- Platform of Therapeutics for Rare Disease, National Institute of Biomedical Innovation, 5-5-2-602 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan; The Center for Medical Engineering and Informatics, Osaka University, 2-2 Yamada-oka, Suita, Osaka 565-0879, Japan.
| |
Collapse
|
72
|
Davis TA, Anam K, Lazdun Y, Gimble JM, Elster EA. Adipose-derived stromal cells promote allograft tolerance induction. Stem Cells Transl Med 2014; 3:1444-50. [PMID: 25411475 DOI: 10.5966/sctm.2014-0131] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Amputations and unsalvageable injuries with devastating tissue loss are common in the combat wounded. Reconstructive transplantation in the civilian setting using vascular composite allotransplants (VCAs) with multiple tissues (skin, muscle, nerve, bone) combined with long-term multidrug immunosuppression has been encouraging. However, skin rejection remains a critical complication. Adipose-derived stromal/stem cells (ASCs) are easily obtained from normal individuals in high numbers, precluding ex vivo expansion. The reparative function and paracrine immunomodulatory capacity of ASCs has gained considerable attention. The present study investigated whether ASCs facilitate long-term skin allograft survival. ASCs were isolated from fresh human subcutaneous adipose lipoaspirate. Full-thickness skin grafts from BALB/c mice were transplanted onto the dorsal flanks of C57BL/6 mice treated with five doses of anti-CD4/CD8 monoclonal antibodies (10 mg/kg) on days 0, +2, +5, +7, and +14 relative to skin grafting. A single nonmyeloablative low dose of busulfan (5 mg/kg) was given on day +5. Seven days after skin transplantation, ASCs (3×10(6)) were infused i.v. with or without donor bone marrow cells (BMCs; 5×10(5)). ASC+BMC coinfusion with minimal conditioning led to stable lymphoid and myeloid macrochimerism, deletion of alloreactive T cells, expansion of regulatory T cells, and long-term allograft survival (>200 days). ASCs constitutively produced high levels of anti-inflammatory/immunoregulatory factors such as prostaglandin E2, indoleamine 2,3-dioxygenase, APO-1/Fas (CD95), and programmed cell death-1 ligand-2. These findings serve as a foundation for developing a translational advanced VCA protocol, embodying both ASCs and low-dose donor BMCs, in nonhuman primates, with the goal of enhancing functional outcomes and eliminating the complications associated with long-term immunosuppression.
Collapse
Affiliation(s)
- Thomas A Davis
- Regenerative Medicine Department, Operational and Undersea Medicine Directorate, Naval Medical Research Center, Silver Spring, Maryland, USA; Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA; Department of Medicine and Surgery Center for Stem Cell Research & Regenerative Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA; LaCell, LLC, New Orleans, Louisiana, USA
| | - Khairul Anam
- Regenerative Medicine Department, Operational and Undersea Medicine Directorate, Naval Medical Research Center, Silver Spring, Maryland, USA; Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA; Department of Medicine and Surgery Center for Stem Cell Research & Regenerative Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA; LaCell, LLC, New Orleans, Louisiana, USA
| | - Yelena Lazdun
- Regenerative Medicine Department, Operational and Undersea Medicine Directorate, Naval Medical Research Center, Silver Spring, Maryland, USA; Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA; Department of Medicine and Surgery Center for Stem Cell Research & Regenerative Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA; LaCell, LLC, New Orleans, Louisiana, USA
| | - Jeffrey M Gimble
- Regenerative Medicine Department, Operational and Undersea Medicine Directorate, Naval Medical Research Center, Silver Spring, Maryland, USA; Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA; Department of Medicine and Surgery Center for Stem Cell Research & Regenerative Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA; LaCell, LLC, New Orleans, Louisiana, USA
| | - Eric A Elster
- Regenerative Medicine Department, Operational and Undersea Medicine Directorate, Naval Medical Research Center, Silver Spring, Maryland, USA; Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA; Department of Medicine and Surgery Center for Stem Cell Research & Regenerative Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA; LaCell, LLC, New Orleans, Louisiana, USA
| |
Collapse
|
73
|
Kredel LI, Siegmund B. Adipose-tissue and intestinal inflammation - visceral obesity and creeping fat. Front Immunol 2014; 5:462. [PMID: 25309544 PMCID: PMC4174117 DOI: 10.3389/fimmu.2014.00462] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 09/10/2014] [Indexed: 12/18/2022] Open
Abstract
Obesity has become one of the main threats to health worldwide and therefore gained increasing clinical and economic significance as well as scientific attention. General adipose-tissue accumulation in obesity is associated with systemically increased pro-inflammatory mediators and humoral and cellular changes within this compartment. These adipose-tissue changes and their systemic consequences led to the concept of obesity as a chronic inflammatory state. A pathognomonic feature of Crohn’s disease (CD) is creeping fat (CF), a locally restricted hyperplasia of the mesenteric fat adjacent to the inflamed segments of the intestine. The precise role of this adipose-tissue and its mediators remains controversial, and ongoing work will have to define whether this compartment is protecting from or contributing to disease activity. This review aims to outline specific cellular changes within the adipose-tissue, occurring in either obesity or CF. Hence the potential impact of adipocytes and resident immune cells from the innate and adaptive immune system will be discussed for both diseases. The second part focuses on the impact of generalized adipose-tissue accumulation in obesity, respectively on the locally restricted form in CD, on intestinal inflammation and on the closely related integrity of the mucosal barrier.
Collapse
Affiliation(s)
- Lea I Kredel
- Gastroenterology, Rheumatology, Infectious Diseases, Medical Department I, Charité - Universitätsmedizin Berlin , Berlin , Germany
| | - Britta Siegmund
- Gastroenterology, Rheumatology, Infectious Diseases, Medical Department I, Charité - Universitätsmedizin Berlin , Berlin , Germany
| |
Collapse
|
74
|
Salamon A, Adam S, Rychly J, Peters K. Long-term tumor necrosis factor treatment induces NFκB activation and proliferation, but not osteoblastic differentiation of adipose tissue-derived mesenchymal stem cells in vitro. Int J Biochem Cell Biol 2014; 54:149-62. [PMID: 25066315 DOI: 10.1016/j.biocel.2014.07.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 07/16/2014] [Accepted: 07/17/2014] [Indexed: 01/08/2023]
Abstract
The pro-inflammatory cytokine tumor necrosis factor (TNF) is well known to induce differentiation of bone matrix-resorbing osteoclasts from hematopoietic stem cells. However, the impact of TNF on differentiation of bone matrix-forming osteoblasts from mesenchymal stem cells (MSC) was only fragmentarily studied so far. Therefore, we investigated what impact long-term TNF treatment has on osteoblastic differentiation of MSC isolated from the adipose tissue (ASC) in vitro. In summary, we found continuous TNF exposure to induce the nuclear factor of kappa B pathway in ASC as well as secretion of the pro-inflammatory chemokine interleukin 8, but not the mitogen-activated protein kinase and the apoptosis pathway in ASC. Moreover, TNF neither induced nor inhibited osteoblastic differentiation of ASC, but strongly increased their proliferation rate. In that manner, pro-inflammatory conditions in vivo may generate significantly increased numbers of progenitor cells, and ASC especially, in conjunction with external stimuli, may contribute to the events of ectopic ossification observed in chronic inflammatory diseases. The substantiation of the translation of our in vitro findings to the disease context encourages further in vivo studies.
Collapse
Affiliation(s)
- Achim Salamon
- Department of Cell Biology, Rostock University Medical Center, Schillingallee 69, D-18057 Rostock, Germany.
| | - Stefanie Adam
- Department of Cell Biology, Rostock University Medical Center, Schillingallee 69, D-18057 Rostock, Germany
| | - Joachim Rychly
- Department of Cell Biology, Rostock University Medical Center, Schillingallee 69, D-18057 Rostock, Germany
| | - Kirsten Peters
- Department of Cell Biology, Rostock University Medical Center, Schillingallee 69, D-18057 Rostock, Germany
| |
Collapse
|
75
|
Salamon A, van Vlierberghe S, van Nieuwenhove I, Baudisch F, Graulus GJ, Benecke V, Alberti K, Neumann HG, Rychly J, Martins JC, Dubruel P, Peters K. Gelatin-Based Hydrogels Promote Chondrogenic Differentiation of Human Adipose Tissue-Derived Mesenchymal Stem Cells In Vitro. MATERIALS 2014; 7:1342-1359. [PMID: 28788517 PMCID: PMC5453082 DOI: 10.3390/ma7021342] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 01/24/2014] [Accepted: 01/27/2014] [Indexed: 12/22/2022]
Abstract
Due to the weak regeneration potential of cartilage, there is a high clinical incidence of articular joint disease, leading to a strong demand for cartilaginous tissue surrogates. The aim of this study was to evaluate a gelatin-based hydrogel for its suitability to support chondrogenic differentiation of human mesenchymal stem cells. Gelatin-based hydrogels are biodegradable, show high biocompatibility, and offer possibilities to introduce functional groups and/or ligands. In order to prove their chondrogenesis-supporting potential, a hydrogel film was developed and compared with standard cell culture polystyrene regarding the differentiation behavior of human mesenchymal stem cells. Cellular basis for this study were human adipose tissue-derived mesenchymal stem cells, which exhibit differentiation potential along the adipogenic, osteogenic and chondrogenic lineage. The results obtained show a promotive effect of gelatin-based hydrogels on chondrogenic differentiation of mesenchymal stem cells in vitro and therefore encourage subsequent in vivo studies.
Collapse
Affiliation(s)
- Achim Salamon
- Department of Cell Biology, Rostock University Medical Center, Schillingallee 69, Rostock D-18057,
Germany; E-Mails: (F.B.); (V.B.); (J.R.); (K.P.)
- Authors to whom correspondence should be addressed; E-Mails: (A.S.); (S.V.)
| | - Sandra van Vlierberghe
- Polymer Chemistry and Biomaterials Group, Gent University, Krijgslaan 281, Building S4, Gent BE-9000,
Belgium; E-Mails: (I.N.); (G.-J.G.); (P.D.)
- Authors to whom correspondence should be addressed; E-Mails: (A.S.); (S.V.)
| | - Ine van Nieuwenhove
- Polymer Chemistry and Biomaterials Group, Gent University, Krijgslaan 281, Building S4, Gent BE-9000,
Belgium; E-Mails: (I.N.); (G.-J.G.); (P.D.)
| | - Frank Baudisch
- Department of Cell Biology, Rostock University Medical Center, Schillingallee 69, Rostock D-18057,
Germany; E-Mails: (F.B.); (V.B.); (J.R.); (K.P.)
| | - Geert-Jan Graulus
- Polymer Chemistry and Biomaterials Group, Gent University, Krijgslaan 281, Building S4, Gent BE-9000,
Belgium; E-Mails: (I.N.); (G.-J.G.); (P.D.)
| | - Verena Benecke
- Department of Cell Biology, Rostock University Medical Center, Schillingallee 69, Rostock D-18057,
Germany; E-Mails: (F.B.); (V.B.); (J.R.); (K.P.)
| | - Kristin Alberti
- Department of Cell Biology, Rostock University Medical Center, Schillingallee 69, Rostock D-18057,
Germany; E-Mails: (F.B.); (V.B.); (J.R.); (K.P.)
| | | | - Joachim Rychly
- Department of Cell Biology, Rostock University Medical Center, Schillingallee 69, Rostock D-18057,
Germany; E-Mails: (F.B.); (V.B.); (J.R.); (K.P.)
| | - José C. Martins
- NMR and Structure Analysis Research Group, Gent University, Krijgslaan 281, Building S4, Gent BE-9000,
Belgium; E-Mail:
| | - Peter Dubruel
- Polymer Chemistry and Biomaterials Group, Gent University, Krijgslaan 281, Building S4, Gent BE-9000,
Belgium; E-Mails: (I.N.); (G.-J.G.); (P.D.)
| | - Kirsten Peters
- Department of Cell Biology, Rostock University Medical Center, Schillingallee 69, Rostock D-18057,
Germany; E-Mails: (F.B.); (V.B.); (J.R.); (K.P.)
| |
Collapse
|
76
|
Salamon A, Jonitz-Heincke A, Adam S, Rychly J, Müller-Hilke B, Bader R, Lochner K, Peters K. Articular cartilage-derived cells hold a strong osteogenic differentiation potential in comparison to mesenchymal stem cells in vitro. Exp Cell Res 2013; 319:2856-65. [PMID: 24055981 DOI: 10.1016/j.yexcr.2013.09.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Revised: 09/09/2013] [Accepted: 09/11/2013] [Indexed: 12/21/2022]
Abstract
Cartilaginous matrix-degenerative diseases like osteoarthritis (OA) are characterized by gradual cartilage erosion, and also by increased presence of cells with mesenchymal stem cell (MSC) character within the affected tissues. Moreover, primary chondrocytes long since are known to de-differentiate in vitro and to be chondrogenically re-differentiable. Since both findings appear to conflict with each other, we quantitatively assessed the mesenchymal differentiation potential of OA patient cartilage-derived cells (CDC) towards the osteogenic and adipogenic lineage in vitro and compared it to that of MSC isolated from adipose tissue (adMSC) of healthy donors. We analyzed expression of MSC markers CD29, CD44, CD105, and CD166, and, following osteogenic and adipogenic induction in vitro, quantified their expression of osteogenic and adipogenic differentiation markers. Furthermore, CDC phenotype and proliferation were monitored. We found that CDC exhibit an MSC CD marker expression pattern similar to adMSC and a similar increase in proliferation rate during osteogenic differentiation. In contrast, the marked reduction of proliferation observed during adipogenic differentiation of adMSC was absent in CDC. Quantification of differentiation markers revealed a strong osteogenic differentiation potential for CDC, however almost no capacity for adipogenic differentiation. Since in the pathogenesis of OA, cartilage degeneration coincides with high bone turnover rates, the high osteogenic differentiation potential of OA patient-derived CDC may affect clinical therapeutic regimens aiming at autologous cartilage regeneration in these patients.
Collapse
Affiliation(s)
- Achim Salamon
- Department of Cell Biology, Rostock University Medical Center, Schillingallee 69, D-18057 Rostock, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
77
|
Maki S, Kadokawa H. Increased ectopic fat cells in the longitudinal muscularis layer of the oviduct isthmus in obese Japanese Black cows. Anim Sci J 2013; 85:207-12. [PMID: 23981058 DOI: 10.1111/asj.12110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 06/06/2013] [Indexed: 11/29/2022]
Abstract
In obese humans, mesenchymal stem cells differentiate to become ectopic fat cells in muscles. These ectopic fat cells inhibit the contraction of vascular smooth muscles. Stem cells have been recently identified in the human oviduct, a structure important in reproduction. We therefore investigated the number of Oil Red O (ORO)-positive cells in the oviducts of control Japanese Black cows (n = 6; body condition score [BCS], 3.0 on a 5-point scale) compared to those with diet-induced obesity (n = 5; BCS, 4.0). We stained the ampulla and isthmus collected on the second day after ovulation with ORO and then counted the positive cells in each layer in 10 cross-sections of the ampulla or isthmus. The obese group (23.4 ± 3.4 in the 10 sections) had larger numbers of ORO-positive cells in the longitudinal muscularis of the isthmus (P < 0.05) than did the control group (15.0 ± 2.4). ORO-positive cells were also observed in all other layers of the isthmus and ampulla; however, the number of cells in these layers did not differ significantly between obese cows and controls. Whether this observed increase in ORO-positive cells in the oviducts of obese cows affects their reproduction warrants further study.
Collapse
Affiliation(s)
- Sachiko Maki
- Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | | |
Collapse
|