701
|
Manferdini C, Maumus M, Gabusi E, Paolella F, Grassi F, Jorgensen C, Fleury-Cappellesso S, Noël D, Lisignoli G. Lack of anti-inflammatory and anti-catabolic effects on basal inflamed osteoarthritic chondrocytes or synoviocytes by adipose stem cell-conditioned medium. Osteoarthritis Cartilage 2015; 23:2045-57. [PMID: 26521751 DOI: 10.1016/j.joca.2015.03.025] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 02/25/2015] [Accepted: 03/20/2015] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To define whether good manufacturing practice (GMP)-clinical grade adipose stem cell (ASC)-derived conditioned medium (CM) is as effective as GMP-ASC in modulating inflammatory and catabolic factors released by both osteoarthritis (OA) chondrocytes or synoviocytes. METHODS OA chondrocytes and synoviocytes were treated with ASC-CM or co-cultured with ASC. Inflammatory factors (IL6, CXCL1/GROα,CXCL8/IL8, CCL2/MCP-1, CCL3/MIP-1α and CCL5/RANTES) and proteinases, such as metalloproteinase (MMP13), a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS4, ADAMTS5) and their tissue metalloproteinase inhibitors (TIMP1, TIMP3) were evaluated by qRT-PCR or immunoassays. The involvement of prostaglandin E2 (PGE2) was also analyzed. RESULTS Most ASC-CM ratios tested did not decrease IL6, CCL2/MCP-1, CCL3/MIP1-α, CCL5/RANTES on basal inflamed chondrocytes or synoviocytes in contrast to what we found using ASC in co-culture. CXCL8/IL8 and CXCL1/GROα were not decreased by ASC-CM on synoviocytes but were only partially reduced on chondrocytes. Moreover, ASC-CM was less efficient both on basal inflamed OA chondrocytes and synoviocytes in reducing proteinases, such as MMP13, ADAMTS4, ADAMTS5 and increasing TIMP1 and TIMP3 compared to ASC in co-culture. The different ratios of ASC-CM contain lower amounts of PGE2 which were not sufficient to reduce inflammatory factors. CONCLUSIONS These data show that ASC-CM has a limited ability to decrease inflammatory and proteinases factors produced by OA chondrocytes or synoviocytes. ASC-CM is not sufficient to recapitulate the beneficial effect demonstrated using ASC in co-culture with inflamed OA chondrocytes and synoviocytes and shows that their use in clinical trials is fundamental to counteract OA progression.
Collapse
Affiliation(s)
- C Manferdini
- SC Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Istituto Ortopedico Rizzoli, Bologna 40136, Italy; Laboratorio RAMSES, Istituto Ortopedico Rizzoli, Bologna 40136, Italy.
| | - M Maumus
- Inserm U844, Hôpital Saint-Eloi, Montpellier F-34295, France; Université Montpellier 1, UFR de Médicine, Montpellier F-34967, France.
| | - E Gabusi
- Laboratorio RAMSES, Istituto Ortopedico Rizzoli, Bologna 40136, Italy.
| | - F Paolella
- Laboratorio RAMSES, Istituto Ortopedico Rizzoli, Bologna 40136, Italy.
| | - F Grassi
- Laboratorio RAMSES, Istituto Ortopedico Rizzoli, Bologna 40136, Italy.
| | - C Jorgensen
- Inserm U844, Hôpital Saint-Eloi, Montpellier F-34295, France; Université Montpellier 1, UFR de Médicine, Montpellier F-34967, France; Service d'Immuno-Rhumatologie Thérapeutique, Hopital Lapeyronie, Montpellier F-34295, France.
| | | | - D Noël
- Inserm U844, Hôpital Saint-Eloi, Montpellier F-34295, France; Université Montpellier 1, UFR de Médicine, Montpellier F-34967, France.
| | - G Lisignoli
- SC Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Istituto Ortopedico Rizzoli, Bologna 40136, Italy; Laboratorio RAMSES, Istituto Ortopedico Rizzoli, Bologna 40136, Italy.
| |
Collapse
|
702
|
Sayegh ET, Sandy JD, Virk MS, Romeo AA, Wysocki RW, Galante JO, Trella KJ, Plaas A, Wang VM. Recent Scientific Advances Towards the Development of Tendon Healing Strategies. ACTA ACUST UNITED AC 2015; 4:128-143. [PMID: 26753125 DOI: 10.2174/2211542004666150713190231] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
There exists a range of surgical and non-surgical approaches to the treatment of both acute and chronic tendon injuries. Despite surgical advances in the management of acute tears and increasing treatment options for tendinopathies, strategies frequently are unsuccessful, due to impaired mechanical properties of the treated tendon and/or a deficiency in progenitor cell activities. Hence, there is an urgent need for effective therapeutic strategies to augment intrinsic and/or surgical repair. Such approaches can benefit both tendinopathies and tendon tears which, due to their severity, appear to be irreversible or irreparable. Biologic therapies include the utilization of scaffolds as well as gene, growth factor, and cell delivery. These treatment modalities aim to provide mechanical durability or augment the biologic healing potential of the repaired tissue. Here, we review the emerging concepts and scientific evidence which provide a rationale for tissue engineering and regeneration strategies as well as discuss the clinical translation of recent innovations.
Collapse
Affiliation(s)
- Eli T Sayegh
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612
| | - John D Sandy
- Department of Biochemistry, Rush University Medical Center, Chicago, IL 60612
| | - Mandeep S Virk
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612
| | - Anthony A Romeo
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612
| | - Robert W Wysocki
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612
| | - Jorge O Galante
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612
| | - Katie J Trella
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612
| | - Anna Plaas
- Department of Rheumatology/Internal Medicine, Rush University Medical Center, Chicago, IL 60612
| | - Vincent M Wang
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612
| |
Collapse
|
703
|
Overcoming translational challenges - The delivery of mechanical stimuli in vivo. Int J Biochem Cell Biol 2015; 69:162-72. [PMID: 26482595 DOI: 10.1016/j.biocel.2015.10.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 10/11/2015] [Accepted: 10/12/2015] [Indexed: 01/22/2023]
Abstract
Despite major medical advances, non-union bone fractures and skeletal defects continue to place significant burden on the patient, the clinicians and the healthcare system as a whole. Current bone substitute approaches are still limited in effectiveness and to date no adequate bone substitute material has been developed for routine clinical application. Tissue engineering presents a novel approach to tackling this clinical burden and developing an acceptable solution for the treatment of skeletal defects. Over the past three decades the field has evolved to appreciate the key biological, material and physical parameters influencing the development of a cell-based tissue engineered therapy and to create associated technologies to exploit such parameters. In recent years a number of therapies have started progressing along the pre-clinical pipeline to build a case for regulatory approval and ultimately clinical adoption. However, little emphasis has been given to the translational challenges faced when moving from "bench-to-bedside". One particular challenge lies in the delivery of functional mechanical stimuli to implanted cell populations to activate and promote osteogenic activities. This review introduces novel bio-magnetic approaches to overcoming this challenge.
Collapse
|
704
|
Lee MW, Ryu S, Kim DS, Sung KW, Koo HH, Yoo KH. Strategies to improve the immunosuppressive properties of human mesenchymal stem cells. Stem Cell Res Ther 2015; 6:179. [PMID: 26445096 PMCID: PMC4596374 DOI: 10.1186/s13287-015-0178-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are of particular interest for the treatment of immune-related diseases because of their immunosuppressive capacities. However, few clinical trials of MSCs have yielded satisfactory results. A number of clinical trials using MSCs are currently in progress worldwide. Unfortunately, protocols and methods, including optimized culture conditions for the harvest of MSCs, have not been standardized. In this regard, complications in the ex vivo expansion of MSCs and MSC heterogeneity have been implicated in the failure of clinical trials. In this review, potential strategies to obtain MSCs with improved immunosuppressive properties and the potential roles of specific immunomodulatory genes, which are differentially upregulated in certain culture conditions, will be discussed.
Collapse
Affiliation(s)
- Myoung Woo Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 50 Irwon-Dong, Gangnam-Gu, Seoul, 135-710, Korea
| | - Somi Ryu
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 50 Irwon-Dong, Gangnam-Gu, Seoul, 135-710, Korea
| | - Dae Seong Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 50 Irwon-Dong, Gangnam-Gu, Seoul, 135-710, Korea
| | - Ki Woong Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 50 Irwon-Dong, Gangnam-Gu, Seoul, 135-710, Korea
| | - Hong Hoe Koo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 50 Irwon-Dong, Gangnam-Gu, Seoul, 135-710, Korea. .,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, 50 Irwon-Dong, Gangnam-Gu, Seoul, 135-710, Korea.
| | - Keon Hee Yoo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 50 Irwon-Dong, Gangnam-Gu, Seoul, 135-710, Korea. .,Department of Medical Device Management and Research, SAIHST, Sungkyunkwan University, 50 Irwon-Dong, Gangnam-Gu, Seoul, 135-710, Korea.
| |
Collapse
|
705
|
Harada Y, Nakasa T, Mahmoud EE, Kamei G, Adachi N, Deie M, Ochi M. Combination therapy with intra-articular injection of mesenchymal stem cells and articulated joint distraction for repair of a chronic osteochondral defect in the rabbit. J Orthop Res 2015; 33:1466-73. [PMID: 26174695 DOI: 10.1002/jor.22922] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 04/07/2015] [Indexed: 02/04/2023]
Abstract
The present study investigated intra-articular injection of bone-marrow-derived mesenchymal stem cells (MSCs) combined with articulated joint distraction as treatment for osteochondral defects. Large osteochondral defects were created in the weight-bearing area of the medial femoral condyle in rabbit knees. Four weeks after defect creation, rabbits were divided into six groups: control group, MSC group, distraction group, distraction + MSC group, temporary distraction group, and temporary distraction + MSC group. Groups with MSC received intra-articular injection of MSCs. Groups with distraction underwent articulated distraction arthroplasty. Groups with temporary distraction discontinued the distraction after 4 weeks. The rabbits were euthanized at 4, 8, and 12 weeks after treatment except temporary distraction groups which were euthanized at only 12 weeks. Histological scores in the distraction + MSC group were significantly better than in the control, MSC group or distraction group at 4 and 8 weeks, but showed no further improvement. At 12 weeks, the temporary distraction + MSC group showed the best results, demonstrating hyaline cartilage repair with regeneration of the osteochondral junction. In conclusion, joint distraction with intra-articular injection of MSCs promotes early cartilage repair, and compressive loading of the repair tissue after temporary distraction stimulates articular cartilage regeneration.
Collapse
Affiliation(s)
- Yohei Harada
- Department of Orthopaedic Surgery, Integrated Health Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Tomoyuki Nakasa
- Department of Orthopaedic Surgery, Integrated Health Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Elhussein Elbadry Mahmoud
- Department of Orthopaedic Surgery, Integrated Health Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Goki Kamei
- Department of Orthopaedic Surgery, Miyoshi Central Hospital, 531 Higashisakeyamachi, Miyoshi, Hiroshima, 728-8502, Japan
| | - Nobuo Adachi
- Department of Orthopaedic Surgery, Integrated Health Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Masataka Deie
- Department of Musculoskeletal Functional Research and Regeneration, Institute of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Mitsuo Ochi
- Department of Orthopaedic Surgery, Integrated Health Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| |
Collapse
|
706
|
Molinos M, Almeida CR, Caldeira J, Cunha C, Gonçalves RM, Barbosa MA. Inflammation in intervertebral disc degeneration and regeneration. J R Soc Interface 2015; 12:20141191. [PMID: 25673296 DOI: 10.1098/rsif.2014.1191] [Citation(s) in RCA: 256] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Intervertebral disc (IVD) degeneration is one of the major causes of low back pain, a problem with a heavy economic burden, which has been increasing in prevalence as populations age. Deeper knowledge of the complex spatial and temporal orchestration of cellular interactions and extracellular matrix remodelling is critical to improve current IVD therapies, which have so far proved unsatisfactory. Inflammation has been correlated with degenerative disc disease but its role in discogenic pain and hernia regression remains controversial. The inflammatory response may be involved in the onset of disease, but it is also crucial in maintaining tissue homeostasis. Furthermore, if properly balanced it may contribute to tissue repair/regeneration as has already been demonstrated in other tissues. In this review, we focus on how inflammation has been associated with IVD degeneration by describing observational and in vitro studies as well as in vivo animal models. Finally, we provide an overview of IVD regenerative therapies that target key inflammatory players.
Collapse
Affiliation(s)
- Maria Molinos
- Instituto de Engenharia Biomédica-INEB, Universidade do Porto, Porto, Portugal Instituto de Ciências Biomédicas Abel Salazar-ICBAS, Universidade do Porto, Porto, Portugal
| | - Catarina R Almeida
- Instituto de Engenharia Biomédica-INEB, Universidade do Porto, Porto, Portugal
| | - Joana Caldeira
- Instituto de Engenharia Biomédica-INEB, Universidade do Porto, Porto, Portugal Instituto de Patologia e Imunologia-IPATIMUP, Universidade do Porto, Porto, Portugal
| | - Carla Cunha
- Instituto de Engenharia Biomédica-INEB, Universidade do Porto, Porto, Portugal
| | - Raquel M Gonçalves
- Instituto de Engenharia Biomédica-INEB, Universidade do Porto, Porto, Portugal
| | - Mário A Barbosa
- Instituto de Engenharia Biomédica-INEB, Universidade do Porto, Porto, Portugal Instituto de Ciências Biomédicas Abel Salazar-ICBAS, Universidade do Porto, Porto, Portugal
| |
Collapse
|
707
|
Zeira O, Asiag N, Aralla M, Ghezzi E, Pettinari L, Martinelli L, Zahirpour D, Dumas MP, Lupi D, Scaccia S, Konar M, Cantile C. Adult autologous mesenchymal stem cells for the treatment of suspected non-infectious inflammatory diseases of the canine central nervous system: safety, feasibility and preliminary clinical findings. J Neuroinflammation 2015; 12:181. [PMID: 26415563 PMCID: PMC4587680 DOI: 10.1186/s12974-015-0402-9] [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: 03/13/2015] [Accepted: 09/22/2015] [Indexed: 01/29/2023] Open
Abstract
Background Non-infectious inflammatory diseases of the canine central nervous system (CNS) are common idiopathic disorders grouped under the term meningoencephalomyelitis of unknown origin (MUO). Ante mortem diagnosis is achieved via assessment of clinical signs, magnetic resonance imaging (MRI), and cerebrospinal fluid (CSF) analysis, but the definitive diagnosis needs histopathological examination. MUO are mostly considered as autoimmune CNS disorders, so that suppressing the immune reaction is the best management method for patients. Mesenchymal stem cells (MSCs) are under investigation to treat autoimmune and degenerative disorders due to their immunomodulatory and regenerative properties. This study aims to verify the safety, feasibility, and efficacy of MSCs treatment in canine idiopathic autoimmune inflammatory disorders of the CNS. Methods Eight dogs presented with acute onset and rapid progression of multifocal neurological signs were selected to the study. In all patients’ physical and neurological examinations, MRI and CSF analyses were performed. Clinical diagnosis in all cases was MUO. All selected dogs responded initially to immunosuppressive drugs (prednisone and a combination of prednisolone and cytosine arabinoside) but developed undesirable side effects. For all eight dogs, the owners considered euthanasia but accepted cell therapy as a last possibility. Autologous bone marrow MSCs (BMMSCs), isolated, cultured, and expanded, were administered by intrathecal (IT) injection in the cisterna magna intravenously (IV) and by intra-arterial (IA) injection in the right carotid artery. Adverse effects and clinical response were monitored for 6 months up to 2-year follow-up. Results The use of autologous BMMSCs in dogs with MUO was safe for IT, IV, and IA injections. No major short- or long-term adverse effects were registered. All the dogs presented early improvement in their general and neurological conditions, with particular effect on cervical pain. The group of dogs treated by IT+IA administration showed a shorter time of reaction to therapy compared to the group treated by IT+IV administration. Conclusions MSCs treatment in dogs affected by MOU is safe and feasible. A larger group of dogs is needed to confirm these results as well as CNS histology in order to better understand the underlying mechanisms.
Collapse
Affiliation(s)
- Offer Zeira
- San Michele Veterinary Hospital, Via Primo Maggio 37, 26838, Tavazzano con Villavesco, Italy.
| | - Nimrod Asiag
- San Michele Veterinary Hospital, Via Primo Maggio 37, 26838, Tavazzano con Villavesco, Italy.
| | - Marina Aralla
- San Michele Veterinary Hospital, Via Primo Maggio 37, 26838, Tavazzano con Villavesco, Italy.
| | - Erica Ghezzi
- San Michele Veterinary Hospital, Via Primo Maggio 37, 26838, Tavazzano con Villavesco, Italy.
| | - Letizia Pettinari
- San Michele Veterinary Hospital, Via Primo Maggio 37, 26838, Tavazzano con Villavesco, Italy.
| | - Laura Martinelli
- San Michele Veterinary Hospital, Via Primo Maggio 37, 26838, Tavazzano con Villavesco, Italy.
| | - Daniele Zahirpour
- San Michele Veterinary Hospital, Via Primo Maggio 37, 26838, Tavazzano con Villavesco, Italy.
| | - Maria Pia Dumas
- San Michele Veterinary Hospital, Via Primo Maggio 37, 26838, Tavazzano con Villavesco, Italy.
| | - Davide Lupi
- San Michele Veterinary Hospital, Via Primo Maggio 37, 26838, Tavazzano con Villavesco, Italy.
| | - Simone Scaccia
- San Michele Veterinary Hospital, Via Primo Maggio 37, 26838, Tavazzano con Villavesco, Italy.
| | - Martin Konar
- San Michele Veterinary Hospital, Via Primo Maggio 37, 26838, Tavazzano con Villavesco, Italy.
| | - Carlo Cantile
- Department of Veterinary Sciences, University of Pisa, Viale delle Piagge 2, 56124, Pisa, Italy.
| |
Collapse
|
708
|
Ruiz M, Cosenza S, Maumus M, Jorgensen C, Noël D. Therapeutic application of mesenchymal stem cells in osteoarthritis. Expert Opin Biol Ther 2015; 16:33-42. [PMID: 26413975 DOI: 10.1517/14712598.2016.1093108] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
INTRODUCTION Osteoarthritis (OA) is a degenerative disease characterized by cartilage degradation and subchondral bone alterations. This disease represents a global public health problem whose prevalence is rapidly growing with the increasing aging of the population. With the discovery of mesenchymal stem cells (MSC) as possible therapeutic agents, their potential for repairing cartilage damage in OA is under investigation. AREAS COVERED Characterization of MSCs and their functional properties are mentioned with an insight into their trophic function and secretory profile. We present a special focus on the types of extracellular vesicles (EVs) that are produced by MSCs and their role in the paracrine activity of MSCs. We then discuss the therapeutic approaches that have been evaluated in pre-clinical models of OA and the results coming out from the clinical trials in patients with OA. EXPERT OPINION MSC-based therapy seems a promising approach for the treatment of patients with OA. Further research is still needed to demonstrate their efficacy in clinical trials using controlled, prospective studies. However, the emergence of MSC-derived EVs as possible therapeutic agents could be an alternative to cell-based therapy.
Collapse
Affiliation(s)
- Maxime Ruiz
- a 1 Inserm, U1183, CHRU Saint Eloi, Hôpital Saint-Eloi , 80 avenue Augustin Fliche, Montpellier, F-34295, France +33 4 67 33 04 73 ; +33 4 67 33 01 13 ; .,b 2 Université Montpellier, UFR de Médecine , Montpellier, F-34000, France
| | - Stella Cosenza
- a 1 Inserm, U1183, CHRU Saint Eloi, Hôpital Saint-Eloi , 80 avenue Augustin Fliche, Montpellier, F-34295, France +33 4 67 33 04 73 ; +33 4 67 33 01 13 ; .,b 2 Université Montpellier, UFR de Médecine , Montpellier, F-34000, France
| | - Marie Maumus
- a 1 Inserm, U1183, CHRU Saint Eloi, Hôpital Saint-Eloi , 80 avenue Augustin Fliche, Montpellier, F-34295, France +33 4 67 33 04 73 ; +33 4 67 33 01 13 ; .,b 2 Université Montpellier, UFR de Médecine , Montpellier, F-34000, France
| | - Christian Jorgensen
- a 1 Inserm, U1183, CHRU Saint Eloi, Hôpital Saint-Eloi , 80 avenue Augustin Fliche, Montpellier, F-34295, France +33 4 67 33 04 73 ; +33 4 67 33 01 13 ; .,b 2 Université Montpellier, UFR de Médecine , Montpellier, F-34000, France.,c 3 Hôpital Lapeyronie, Service d'immuno-Rhumatologie , Montpellier, F-34295, France
| | - Danièle Noël
- a 1 Inserm, U1183, CHRU Saint Eloi, Hôpital Saint-Eloi , 80 avenue Augustin Fliche, Montpellier, F-34295, France +33 4 67 33 04 73 ; +33 4 67 33 01 13 ; .,b 2 Université Montpellier, UFR de Médecine , Montpellier, F-34000, France.,c 3 Hôpital Lapeyronie, Service d'immuno-Rhumatologie , Montpellier, F-34295, France
| |
Collapse
|
709
|
Bionaz M, Monaco E, Wheeler MB. Transcription Adaptation during In Vitro Adipogenesis and Osteogenesis of Porcine Mesenchymal Stem Cells: Dynamics of Pathways, Biological Processes, Up-Stream Regulators, and Gene Networks. PLoS One 2015; 10:e0137644. [PMID: 26398344 PMCID: PMC4580618 DOI: 10.1371/journal.pone.0137644] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 07/27/2015] [Indexed: 12/20/2022] Open
Abstract
The importance of mesenchymal stem cells (MSC) for bone regeneration is growing. Among MSC the bone marrow-derived stem cells (BMSC) are considered the gold standard in tissue engineering and regenerative medicine; however, the adipose-derived stem cells (ASC) have very similar properties and some advantages to be considered a good alternative to BMSC. The molecular mechanisms driving adipogenesis are relatively well-known but mechanisms driving osteogenesis are poorly known, particularly in pig. In the present study we have used transcriptome analysis to unravel pathways and biological functions driving in vitro adipogenesis and osteogenesis in BMSC and ASC. The analysis was performed using the novel Dynamic Impact Approach and functional enrichment analysis. In addition, a k-mean cluster analysis in association with enrichment analysis, networks reconstruction, and transcription factors overlapping analysis were performed in order to uncover the coordination of biological functions underlining differentiations. Analysis indicated a larger and more coordinated transcriptomic adaptation during adipogenesis compared to osteogenesis, with a larger induction of metabolism, particularly lipid synthesis (mostly triglycerides), and a larger use of amino acids for synthesis of feed-forward adipogenic compounds, larger cell signaling, lower cell-to-cell interactions, particularly for the cytoskeleton organization and cell junctions, and lower cell proliferation. The coordination of adipogenesis was mostly driven by Peroxisome Proliferator-activated Receptors together with other known adipogenic transcription factors. Only a few pathways and functions were more induced during osteogenesis compared to adipogenesis and some were more inhibited during osteogenesis, such as cholesterol and protein synthesis. Up-stream transcription factor analysis indicated activation of several lipid-related transcription regulators (e.g., PPARs and CEBPα) during adipogenesis but osteogenesis was driven by inhibition of several up-stream regulators, such as MYC. Between MSCs the data indicated an ‘adipocyte memory’ in ASC with also an apparent lower immunogenicity compared to BMSC during differentiations. Overall the analysis allowed proposing a dynamic model for the adipogenic and osteogenic differentiation in porcine ASC and BMSC.
Collapse
Affiliation(s)
- Massimo Bionaz
- Laboratory of Stem Cell Biology and Engineering in the Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Elisa Monaco
- Laboratory of Stem Cell Biology and Engineering in the Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Matthew B. Wheeler
- Laboratory of Stem Cell Biology and Engineering in the Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- * E-mail:
| |
Collapse
|
710
|
Roubeix C, Godefroy D, Mias C, Sapienza A, Riancho L, Degardin J, Fradot V, Ivkovic I, Picaud S, Sennlaub F, Denoyer A, Rostene W, Sahel JA, Parsadaniantz SM, Brignole-Baudouin F, Baudouin C. Intraocular pressure reduction and neuroprotection conferred by bone marrow-derived mesenchymal stem cells in an animal model of glaucoma. Stem Cell Res Ther 2015; 6:177. [PMID: 26377305 PMCID: PMC4574127 DOI: 10.1186/s13287-015-0168-0] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 01/28/2015] [Accepted: 08/21/2015] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Glaucoma is a sight-threatening retinal neuropathy associated with elevated intraocular pressure (IOP) due to degeneration and fibrosis of the trabecular meshwork (TM). Glaucoma medications aim to reduce IOP without targeting the specific TM pathology, Bone-marrow mesenchymal stem cells (MSCs) are used today in various clinical studies. Here, we investigated the potential of MSCs therapy in an glaucoma-like ocular hypertension (OHT) model and decipher in vitro the effects of MSCs on primary human trabecular meshwork cells. METHODS Ocular hypertension model was performed by cauterization of 3 episcleral veins (EVC) of Long-Evans male rat eyes. MSCs were isolated from rat bone marrow, amplified in vitro and tagged with quantum dot nanocrystals. Animals were distributed as 1) MSCs group receiving 5.10(5)cells/6μl Minimum Essential Medium and 2) MEM group receiving 6μl MEM (n = 10 each). Injections were performed into the anterior chamber of 20 days-hypertensive eyes and IOP was monitored twice a week for 4 weeks. At the end of experiment, cell distribution in the anterior segment was examined in confocal microscopy on flat mounted corneas. Moreover, we tested in vitro effects of MSCs conditioned medium (MSC-CM) on primary human trabecular meshwork cells (hTM cells) using Akt activation, myosin phosphorylation and TGF-β2-dependent profibrotic phenotype in hTM cells. RESULTS We demonstrated a rapid and long-lasting in vivo effect of MSCs transplantation that significantly reduced IOP in hypertensive eyes induced by EVC. MSCs were located to the ciliary processes and the TM. Enumeration of RGCs on whole flat-mounted retina highlighted a protective effect of MSCs on RGCs death. In vitro, MSC-CM promotes: (i) hTM cells survival by activating the antiapoptotic pathway, Akt, (ii) hTM cells relaxation as analyzed by the decrease in myosin phosphorylation and (iii) inhibition of TGF-β2-dependent profibrotic phenotype acquisition in hTM cells. CONCLUSIONS MSCs injection in the ocular anterior chamber in a rat model of OHT provides neuroprotective effect in the glaucoma pathophysiology via TM protection. These results demonstrate that MSCs constitute promising tool for treating ocular hypertension and retinal cell degeneration.
Collapse
Affiliation(s)
- Christophe Roubeix
- INSERM, U968, Paris, F-75012, France.
- UPMC Université Paris 06, UMR_S 968, Institut de la Vision, Paris, F-75012, France.
- CNRS, UMR_7210, Paris, F-75012, France.
| | - David Godefroy
- INSERM, U968, Paris, F-75012, France.
- UPMC Université Paris 06, UMR_S 968, Institut de la Vision, Paris, F-75012, France.
- CNRS, UMR_7210, Paris, F-75012, France.
| | - Céline Mias
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM UMR 1048, 31432 Toulouse cedex 4, France, Toulouse, France.
| | - Anaïs Sapienza
- INSERM, U968, Paris, F-75012, France.
- UPMC Université Paris 06, UMR_S 968, Institut de la Vision, Paris, F-75012, France.
- CNRS, UMR_7210, Paris, F-75012, France.
| | - Luisa Riancho
- INSERM, U968, Paris, F-75012, France.
- UPMC Université Paris 06, UMR_S 968, Institut de la Vision, Paris, F-75012, France.
- CNRS, UMR_7210, Paris, F-75012, France.
| | - Julie Degardin
- INSERM, U968, Paris, F-75012, France.
- UPMC Université Paris 06, UMR_S 968, Institut de la Vision, Paris, F-75012, France.
- CNRS, UMR_7210, Paris, F-75012, France.
| | - Valérie Fradot
- INSERM, U968, Paris, F-75012, France.
- UPMC Université Paris 06, UMR_S 968, Institut de la Vision, Paris, F-75012, France.
- CNRS, UMR_7210, Paris, F-75012, France.
| | - Ivana Ivkovic
- INSERM, U968, Paris, F-75012, France.
- UPMC Université Paris 06, UMR_S 968, Institut de la Vision, Paris, F-75012, France.
- CNRS, UMR_7210, Paris, F-75012, France.
| | - Serge Picaud
- INSERM, U968, Paris, F-75012, France.
- UPMC Université Paris 06, UMR_S 968, Institut de la Vision, Paris, F-75012, France.
- CNRS, UMR_7210, Paris, F-75012, France.
| | - Florian Sennlaub
- INSERM, U968, Paris, F-75012, France.
- UPMC Université Paris 06, UMR_S 968, Institut de la Vision, Paris, F-75012, France.
- CNRS, UMR_7210, Paris, F-75012, France.
| | - Alexandre Denoyer
- INSERM, U968, Paris, F-75012, France.
- UPMC Université Paris 06, UMR_S 968, Institut de la Vision, Paris, F-75012, France.
- CNRS, UMR_7210, Paris, F-75012, France.
- Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, INSERM-DHOS CIC 503, Paris, F-75012, France.
| | - William Rostene
- INSERM, U968, Paris, F-75012, France.
- UPMC Université Paris 06, UMR_S 968, Institut de la Vision, Paris, F-75012, France.
- CNRS, UMR_7210, Paris, F-75012, France.
| | - José Alain Sahel
- INSERM, U968, Paris, F-75012, France.
- UPMC Université Paris 06, UMR_S 968, Institut de la Vision, Paris, F-75012, France.
- CNRS, UMR_7210, Paris, F-75012, France.
- Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, INSERM-DHOS CIC 503, Paris, F-75012, France.
| | - Stéphane Melik Parsadaniantz
- INSERM, U968, Paris, F-75012, France.
- UPMC Université Paris 06, UMR_S 968, Institut de la Vision, Paris, F-75012, France.
- CNRS, UMR_7210, Paris, F-75012, France.
| | - Françoise Brignole-Baudouin
- INSERM, U968, Paris, F-75012, France.
- UPMC Université Paris 06, UMR_S 968, Institut de la Vision, Paris, F-75012, France.
- CNRS, UMR_7210, Paris, F-75012, France.
- Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, INSERM-DHOS CIC 503, Paris, F-75012, France.
- University Paris Descartes, Sorbonne Paris Cité, Paris, F-75006, France.
- Faculté de Pharmacie de Paris, University Paris Descartes, Sorbonne Paris Cité, Paris, F-75006, France.
| | - Christophe Baudouin
- INSERM, U968, Paris, F-75012, France.
- UPMC Université Paris 06, UMR_S 968, Institut de la Vision, Paris, F-75012, France.
- CNRS, UMR_7210, Paris, F-75012, France.
- Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, INSERM-DHOS CIC 503, Paris, F-75012, France.
- Department of Ophthalmology, Hôpital Ambroise Pare, AP HP, Boulogne, F-92100, France.
- University Versailles St Quentin en Yvelines, Montigny-Le-Bretonneux, F-78180, France.
| |
Collapse
|
711
|
Skyler JS, Fonseca VA, Segal KR, Rosenstock J. Allogeneic Mesenchymal Precursor Cells in Type 2 Diabetes: A Randomized, Placebo-Controlled, Dose-Escalation Safety and Tolerability Pilot Study. Diabetes Care 2015; 38:1742-9. [PMID: 26153271 PMCID: PMC4542273 DOI: 10.2337/dc14-2830] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 06/09/2015] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To assess the safety, tolerability, and feasibility of adult allogeneic bone marrow-derived mesenchymal precursor cells (MPCs) in type 2 diabetes inadequately controlled with metformin either alone or with one additional oral antidiabetic agent. RESEARCH DESIGN AND METHODS The study was a dose-escalating randomized placebo-controlled trial assessing one intravenous (IV) infusion of MPCs (rexlemestrocel-L; Mesoblast Inc.) 0.3 × 10(6)/kg (n = 15), 1.0 × 10(6)/kg (n = 15), or 2.0 × 10(6)/kg (n = 15) or placebo (n = 16). Study duration was 12 weeks. RESULTS Subjects (21 women, 40 men) with a mean ± SD baseline HbA1c 8.3 ± 1.0% (67 ± 10.9 mmol/mol), BMI 33.5 ± 5.5 kg/m(2), and diabetes duration 10.1 ± 6.0 years were enrolled at 18 U.S. sites. No acute adverse events (AEs) were associated with infusion. No serious AEs, serious hypoglycemia AEs, or discontinuations due to AEs over 12 weeks were found. No subjects developed donor-specific anti-HLA antibodies or became sensitized. The safety profile was comparable among treatment groups. Compared with placebo, a single IV infusion of rexlemestrocel-L reduced HbA1c at all time points after week 1. The adjusted least squares mean ± SE dose-related differences in HbA1c from placebo in the rexlemestrocel-L groups ranged from -0.1 ± 0.2% (-1.1 ± 2.2 mmol/mol) to -0.4 ± 0.2% (4.4 ± 2.2 mmol/mol) at 8 weeks and from 0.0 ± 0.25% to -0.3 ± 0.25% (-3.3 ± -2.7 mmol/mol) at 12 weeks (P < 0.05 for 2.0 × 10(6)/kg dose at 8 weeks). The clinical target HbA1c <7% (53 mmol/mol) was achieved by 33% (5 of 15) of the subjects who received the 2.0 × 10(6)/kg dose vs. 0% of those who received placebo (P < 0.05). CONCLUSIONS This short-term study demonstrates the safety and feasibility of up to 246 million MPCs in subjects with type 2 diabetes.
Collapse
Affiliation(s)
- Jay S Skyler
- Diabetes Research Institute, University of Miami, Miami, FL
| | | | | | - Julio Rosenstock
- Dallas Diabetes and Endocrine Center at Medical City, Dallas, TX
| | | |
Collapse
|
712
|
Kim SB, Joo J, Hong YB, Choi BO. Ultrasonography-guided transplantation facilitates perineural delivery of stem cells. Anim Cells Syst (Seoul) 2015. [DOI: 10.1080/19768354.2015.1053521] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
713
|
da Silva Meirelles L, Malta TM, de Deus Wagatsuma VM, Palma PVB, Araújo AG, Ribeiro Malmegrim KC, Morato de Oliveira F, Panepucci RA, Silva WA, Kashima Haddad S, Covas DT. Cultured Human Adipose Tissue Pericytes and Mesenchymal Stromal Cells Display a Very Similar Gene Expression Profile. Stem Cells Dev 2015; 24:2822-40. [PMID: 26192741 DOI: 10.1089/scd.2015.0153] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) are cultured cells that can give rise to mature mesenchymal cells under appropriate conditions and secrete a number of biologically relevant molecules that may play an important role in regenerative medicine. Evidence indicates that pericytes (PCs) correspond to mesenchymal stem cells in vivo and can give rise to MSCs when cultured, but a comparison between the gene expression profiles of cultured PCs (cPCs) and MSCs is lacking. We have devised a novel methodology to isolate PCs from human adipose tissue and compared cPCs to MSCs obtained through traditional methods. Freshly isolated PCs expressed CD34, CD140b, and CD271 on their surface, but not CD146. Both MSCs and cPCs were able to differentiate along mesenchymal pathways in vitro, displayed an essentially identical surface immunophenotype, and exhibited the ability to suppress CD3(+) lymphocyte proliferation in vitro. Microarray expression data of cPCs and MSCs formed a single cluster among other cell types. Further analyses showed that the gene expression profiles of cPCs and MSCs are extremely similar, although MSCs differentially expressed endothelial cell (EC)-specific transcripts. These results confirm, using the power of transcriptomic analysis, that PCs give rise to MSCs and suggest that low levels of ECs may persist in MSC cultures established using traditional protocols.
Collapse
Affiliation(s)
- Lindolfo da Silva Meirelles
- 1 Center for Cell-Based Therapy (CEPID/FAPESP), Regional Center for Hemotherapy of Ribeirão Preto, University of São Paulo , Ribeirão Preto, Brazil .,2 Laboratory for Stem Cells and Tissue Engineering, PPGBioSaúde, Lutheran University of Brazil , Canoas, Brazil
| | - Tathiane Maistro Malta
- 1 Center for Cell-Based Therapy (CEPID/FAPESP), Regional Center for Hemotherapy of Ribeirão Preto, University of São Paulo , Ribeirão Preto, Brazil
| | - Virgínia Mara de Deus Wagatsuma
- 1 Center for Cell-Based Therapy (CEPID/FAPESP), Regional Center for Hemotherapy of Ribeirão Preto, University of São Paulo , Ribeirão Preto, Brazil
| | - Patrícia Viana Bonini Palma
- 1 Center for Cell-Based Therapy (CEPID/FAPESP), Regional Center for Hemotherapy of Ribeirão Preto, University of São Paulo , Ribeirão Preto, Brazil
| | - Amélia Goes Araújo
- 3 Laboratory of Large-Scale Functional Biology (LLSFBio), Regional Center for Hemotherapy of Ribeirão Preto, University of São Paulo , Ribeirão Preto, Brazil
| | | | - Fábio Morato de Oliveira
- 1 Center for Cell-Based Therapy (CEPID/FAPESP), Regional Center for Hemotherapy of Ribeirão Preto, University of São Paulo , Ribeirão Preto, Brazil
| | - Rodrigo Alexandre Panepucci
- 3 Laboratory of Large-Scale Functional Biology (LLSFBio), Regional Center for Hemotherapy of Ribeirão Preto, University of São Paulo , Ribeirão Preto, Brazil
| | - Wilson Araújo Silva
- 1 Center for Cell-Based Therapy (CEPID/FAPESP), Regional Center for Hemotherapy of Ribeirão Preto, University of São Paulo , Ribeirão Preto, Brazil .,5 Department of Genetics, School of Medicine of Ribeirão Preto, University of São Paulo , Ribeirão Preto, Brazil
| | - Simone Kashima Haddad
- 1 Center for Cell-Based Therapy (CEPID/FAPESP), Regional Center for Hemotherapy of Ribeirão Preto, University of São Paulo , Ribeirão Preto, Brazil
| | - Dimas Tadeu Covas
- 1 Center for Cell-Based Therapy (CEPID/FAPESP), Regional Center for Hemotherapy of Ribeirão Preto, University of São Paulo , Ribeirão Preto, Brazil .,6 Department of Clinical Medicine, School of Medicine of Ribeirão Preto, University of São Paulo , Ribeirão Preto, Brazil
| |
Collapse
|
714
|
Chung E. Stem-cell-based therapy in the field of urology: a review of stem cell basic science, clinical applications and future directions in the treatment of various sexual and urinary conditions. Expert Opin Biol Ther 2015; 15:1623-32. [DOI: 10.1517/14712598.2015.1075504] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
715
|
Busser H, Najar M, Raicevic G, Pieters K, Velez Pombo R, Philippart P, Meuleman N, Bron D, Lagneaux L. Isolation and Characterization of Human Mesenchymal Stromal Cell Subpopulations: Comparison of Bone Marrow and Adipose Tissue. Stem Cells Dev 2015; 24:2142-57. [PMID: 26086188 DOI: 10.1089/scd.2015.0172] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Preparations of mesenchymal stromal cells (MSCs) are generally obtained from unfractionated tissue cells, resulting in heterogeneous cell mixtures. Several markers were proposed to enrich these cells, but the majority of these markers are defined for bone marrow (BM). Moreover, the surface markers of freshly isolated MSCs also differ from those of cultured MSCs in addition to a phenotypic variation depending on the MSC source. For tissue engineering applications, it is crucial to start with a well-defined cell population. In this study, we performed immunomagnetic selections with five single surface markers to isolate MSC subpopulations from BM and adipose tissue (AT): CD271, SUSD2, MSCA-1, CD44, and CD34. We determined the phenotype, the clonogenicity, the proliferation, the differentiation capacity, and the immunoregulatory profile of the subpopulations obtained in comparison with unselected cells. We showed that native BM-MSCs can be enriched from the positive fractions of MSCA-1, SUSD2, and CD271 selections. In contrast, we observed that SUSD2 and MSCA-1 were unable to identify MSCs from AT, meaning they are not expressed in situ. Only the CD34(+) selection successfully isolated MSCs from AT. Interestingly, we observed that CD271 selection can define AT cell subsets with particular abilities, but only in lipoaspiration samples and not in abdominoplasty samples. Importantly, we found a population of clear CD34(+) fresh BM-MSCs displaying different properties. A single marker-based selection for MSC enrichment should be more advantageous for cell therapy and would enable the standardization of efficient and safe therapeutic intervention through the use of a well-identified and homogeneous cell population.
Collapse
Affiliation(s)
- Hélène Busser
- 1 Laboratory of Clinical Cell Therapy, Jules Bordet Institute, Université Libre de Bruxelles , Brussels, Belgium
| | - Mehdi Najar
- 1 Laboratory of Clinical Cell Therapy, Jules Bordet Institute, Université Libre de Bruxelles , Brussels, Belgium
| | - Gordana Raicevic
- 1 Laboratory of Clinical Cell Therapy, Jules Bordet Institute, Université Libre de Bruxelles , Brussels, Belgium
| | - Karlien Pieters
- 1 Laboratory of Clinical Cell Therapy, Jules Bordet Institute, Université Libre de Bruxelles , Brussels, Belgium
| | - Rafael Velez Pombo
- 2 Plastic, Aesthetic and Reconstructive Surgery, Iris South Hospitals (HIS) , Joseph Bracops Site, Brussels, Belgium
| | - Pierre Philippart
- 3 Department of Stomatology and Maxillo-Facial Surgery, Iris South Hospitals (HIS) , Joseph Bracops Site, Brussels, Belgium
| | | | - Dominique Bron
- 4 Hematology, Jules Bordet Institute , Brussels, Belgium
| | - Laurence Lagneaux
- 1 Laboratory of Clinical Cell Therapy, Jules Bordet Institute, Université Libre de Bruxelles , Brussels, Belgium
| |
Collapse
|
716
|
Bottlenecks in the Efficient Use of Advanced Therapy Medicinal Products Based on Mesenchymal Stromal Cells. Stem Cells Int 2015; 2015:895714. [PMID: 26273307 PMCID: PMC4530293 DOI: 10.1155/2015/895714] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 03/05/2015] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) have been established as promising candidate sources of universal donor cells for cell therapy due to their contributions to tissue and organ homeostasis, repair, and support by self-renewal and multidifferentiation, as well as by their anti-inflammatory, antiproliferative, immunomodulatory, trophic, and proangiogenic properties. Various diseases have been treated by MSCs in animal models. Additionally, hundreds of clinical trials related to the potential benefits of MSCs are in progress. However, although all MSCs are considered suitable to exert these functions, dissimilarities have been found among MSCs derived from different tissues. The same levels of efficacy and desired outcomes have not always been achieved in the diverse studies that have been performed thus far. Moreover, autologous MSCs can be affected by the disease status of patients, compromising their use. Therefore, collecting information regarding the characteristics of MSCs obtained from different sources and the influence of the host (patient) medical conditions on MSCs is important for assuring the safety and efficacy of cell-based therapies. This review provides relevant information regarding factors to consider for the clinical application of MSCs.
Collapse
|
717
|
Carmelo JG, Fernandes-Platzgummer A, Diogo MM, da Silva CL, Cabral JMS. A xeno-free microcarrier-based stirred culture system for the scalable expansion of human mesenchymal stem/stromal cells isolated from bone marrow and adipose tissue. Biotechnol J 2015; 10:1235-47. [DOI: 10.1002/biot.201400586] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 04/19/2015] [Accepted: 07/01/2015] [Indexed: 12/12/2022]
|
718
|
Zavan B, De Francesco F, D'Andrea F, Ferroni L, Gardin C, Salzillo R, Nicoletti G, Ferraro GA. Persistence of CD34 Stem Marker in Human Lipoma: Searching for Cancer Stem Cells. Int J Biol Sci 2015; 11:1127-39. [PMID: 26327807 PMCID: PMC4551749 DOI: 10.7150/ijbs.11946] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 05/05/2015] [Indexed: 01/19/2023] Open
Abstract
Background: Lipomas are benign solid tumours that develop in soft tissues with origin in mesenchymal progenitors. Macroscopically, they appear as soft-elastic nodules, varying in volume from a few millimiters to several centimetres and can enlarge progressively. Although they are usually asymptomatic, they can cause symptoms due to nerve or vessel compression. Microscopically they appear as fibrous connective tissue stroma with embedded adipocytes, and absence of inflammation. Up to now no characterisation of stem cell population present in this tissue has been performed. Methods: Cytofluorimetric, biological and molecular biology analyses have been performed in order to test superficial cell markers and gene expression profile related to stemness and apoptotic activity of cells present in lipoma tissues compared to those of adipose tissue's cells. Results: Our results confirmed that CD34+ cells in lipoma were present around small adipocytes, showing several altered biological activity such as proliferation, apoptotis and stemness. Conclusions: The data emerging from the comparison of the lipoma cells and normal adipose tissue, suggests the presence of cell precursors involved in the development of the lipoma. This hypothesis requires further investigation and may indicate new thresholds in the study of benign tumour pathogenesis.
Collapse
Affiliation(s)
- Barbara Zavan
- 2. Department of Biomedical Sciences, University of Padua, Ugo Bassi, 58/B, 35131, Padua (Italy)
| | - Francesco De Francesco
- 1. Multidisciplinary Department of Medical-Surgical and Dental Specialties, Second University of Naples, L. De Crecchio, 6, 80138, Naples (Italy)
| | - Francesco D'Andrea
- 1. Multidisciplinary Department of Medical-Surgical and Dental Specialties, Second University of Naples, L. De Crecchio, 6, 80138, Naples (Italy)
| | - Letizia Ferroni
- 2. Department of Biomedical Sciences, University of Padua, Ugo Bassi, 58/B, 35131, Padua (Italy)
| | - Chiara Gardin
- 2. Department of Biomedical Sciences, University of Padua, Ugo Bassi, 58/B, 35131, Padua (Italy)
| | - Rosa Salzillo
- 1. Multidisciplinary Department of Medical-Surgical and Dental Specialties, Second University of Naples, L. De Crecchio, 6, 80138, Naples (Italy)
| | - Gianfranco Nicoletti
- 1. Multidisciplinary Department of Medical-Surgical and Dental Specialties, Second University of Naples, L. De Crecchio, 6, 80138, Naples (Italy)
| | - Giuseppe A Ferraro
- 1. Multidisciplinary Department of Medical-Surgical and Dental Specialties, Second University of Naples, L. De Crecchio, 6, 80138, Naples (Italy)
| |
Collapse
|
719
|
Andreeva E, Andrianova I, Rylova J, Gornostaeva A, Bobyleva P, Buravkova L. Proinflammatory interleukins' production by adipose tissue-derived mesenchymal stromal cells: the impact of cell culture conditions and cell-to-cell interaction. Cell Biochem Funct 2015; 33:386-93. [PMID: 26179154 DOI: 10.1002/cbf.3125] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 05/22/2015] [Accepted: 06/08/2015] [Indexed: 11/11/2022]
Abstract
The impact of culture conditions and interaction with activated peripheral blood mononuclear cells on the interleukin (IL) gene expression profile and proinflammatory IL-6 and IL-8 production by adipose-derived stromal cells (ASCs) was investigated. A microarray analysis revealed a wide range of IL genes either under standard (20%) or hypoxic (5%) O2 concentrations, some highly up-regulated at hypoxia. IL-6 and IL-8 production was inversely dependent on cell culture density. In early (first-third) passages, IL-6 and IL-8 concentration was higher at 20% O2 and in late (8th-12th) passages under 5% O2. Interaction between ASCs and mononuclear cells in indirect setting was accompanied with a significant decrease of IL-6 and did not result in the elevation of IL-8 concentration. Thereby, the production of proinflammatory interleukins (IL-6 and IL-8) may be affected by the ASC intrinsic features (density in culture, and duration of expansion), as well as by microenvironmental factors, such as hypoxia and the presence of blood-borne cells. These data are important for elucidating ASC paracrine activity regulation in vitro. They would also be on demand for optimisation of the cell therapy protocols, based on the application of ASC biologically active substances. SIGNIFICANCE PARAGRAPH: Ex vivo expansion is widely used for increasing the number of adipose-derived stromal cells (ASCs) and improving of their quality. The present study was designed to elucidate the particular factors influencing the interleukin production in ASCs. The presented data specified the parameters (i.e. cell density, duration of cultivation, hypoxia, etc.) that should be taken in mind when ASCs are intended to be used in protocols implying their paracrine activity. These data would be of considerable interest for researchers and clinicians working in the biomedical science.
Collapse
Affiliation(s)
- Elena Andreeva
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| | - Irina Andrianova
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia.,Institute of Experimental Cardiology, Russian Cardiology Research Center, Moscow, Russia
| | - Julia Rylova
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| | | | - Polina Bobyleva
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| | - Ludmila Buravkova
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
720
|
Mesenchymal stromal cells derived from various tissues: Biological, clinical and cryopreservation aspects. Cryobiology 2015; 71:181-97. [PMID: 26186998 DOI: 10.1016/j.cryobiol.2015.07.003] [Citation(s) in RCA: 235] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 07/13/2015] [Indexed: 12/11/2022]
Abstract
Originally isolated from bone marrow, mesenchymal stromal cells (MSCs) have since been obtained from various fetal and post-natal tissues and are the focus of an increasing number of clinical trials. Because of their tremendous potential for cellular therapy, regenerative medicine and tissue engineering, it is desirable to cryopreserve and bank MSCs to increase their access and availability. A remarkable amount of research and resources have been expended towards optimizing the protocols, freezing media composition, cooling devices and storage containers, as well as developing good manufacturing practices in order to ensure that MSCs retain their therapeutic characteristics following cryopreservation and that they are safe for clinical use. Here, we first present an overview of the identification of MSCs, their tissue sources and the properties that render them suitable as a cellular therapeutic. Next, we discuss the responses of cells during freezing and focus on the traditional and novel approaches used to cryopreserve MSCs. We conclude that viable MSCs from diverse tissues can be recovered after cryopreservation using a variety of freezing protocols, cryoprotectants, storage periods and temperatures. However, alterations in certain functions of MSCs following cryopreservation warrant future investigations on the recovery of cells post-thaw followed by expansion of functional cells in order to achieve their full therapeutic potential.
Collapse
|
721
|
Thakkar UG, Trivedi HL, Vanikar AV, Dave SD. Co-infusion of insulin-secreting adipose tissue-derived mesenchymal stem cells and hematopoietic stem cells: novel approach to management of type 1 diabetes mellitus. Int J Diabetes Dev Ctries 2015. [DOI: 10.1007/s13410-015-0409-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
722
|
Mesenchymal stromal cell implantation for stimulation of long bone healing aggravates Staphylococcus aureus induced osteomyelitis. Acta Biomater 2015; 21:165-77. [PMID: 25805108 DOI: 10.1016/j.actbio.2015.03.019] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 03/05/2015] [Accepted: 03/17/2015] [Indexed: 01/08/2023]
Abstract
Large bone defects requiring long-term osteosynthetic stabilization or repeated surgeries show a considerable rate of infection. Mesenchymal stromal cells (MSCs) have been successfully used to enhance bone regeneration, but their powerful immunomodulatory effects may impose an enhanced risk for osteomyelitis development. In order to unravel whether implantation of MSCs aggravates a simultaneous bone infection, a hydrogel-supported osteomyelitis ostectomy model was developed in which rats received a femoral bone defect with rigid plate-fixation. After fibrin-assisted transfer of Staphylococcus aureus (SA), effects of MSC implantation on osteomyelitis development were quantified over 3-4 weeks. All SA-infected animals developed an acute local osteomyelitis with significantly increased blood neutrophil count, abscess formation and bone destruction. MSC-treatment of infected defects aggravated osteomyelitis according to a significantly elevated osteomyelitis score and enhanced distal bone loss with spongy alteration of cortical bone architecture. Increased attraction of macrophages, osteoclasts and regulation of pro- and anti-inflammatory mediators were potential MSC actions. Overall trophic actions of MSCs implanted into non-sterile bone defects may enhance an infection and/or exacerbate osteomyelitis. Studies on antibiotic carrier augmentation or antibiotic treatment are warranted to decide whether MSC implantation is a safe and promising therapy for orthopedic implant-stabilized bone defects at high risk for development of infection.
Collapse
|
723
|
de Morais SDBV, da Silva LEV, Lataro RM, Silva CAA, de Oliveira LFL, de Carvalho EEV, Simões MV, da Silva Meirelles L, Fazan R, Salgado HC. Mesenchymal Stem Cells Improve Heart Rate Variability and Baroreflex Sensitivity in Rats with Chronic Heart Failure. Stem Cells Dev 2015; 24:2181-92. [PMID: 26059001 DOI: 10.1089/scd.2014.0573] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Heart failure induced by myocardial infarct (MI) attenuates the heart rate variability (HRV) and baroreflex sensitivity, which are important risk factors for life-threatening cardiovascular events. Therapies with mesenchymal stem cells (MSCs) have shown promising results after MI. However, the effects of MSCs on hemodynamic (heart rate and arterial pressure) variability and baroreflex sensitivity in chronic heart failure (CHF) following MI have not been evaluated thus far. Male Wistar rats received MSCs or saline solution intravenously 1 week after ligation of the left coronary artery. Control (noninfarcted) rats were also evaluated. MI size was assessed using single-photon emission computed tomography (SPECT). The left ventricular ejection fraction (LVEF) was evaluated using radionuclide ventriculography. Four weeks after MSC injection, the animals were anesthetized and instrumented for chronic ECG recording and catheters were implanted in the femoral artery to record arterial pressure. Arterial pressure and HRVs were determined in time and frequency domain (spectral analysis) while HRV was also examined using nonlinear methods: DFA (detrended fluctuation analysis) and sample entropy. The initial MI size was the same among all infarcted rats but was reduced by MSCs. CHF rats exhibited increased myocardial interstitial collagen and sample entropy combined with the attenuation of the following cardiocirculatory parameters: DFA indices, LVEF, baroreflex sensitivity, and HRV. Nevertheless, MSCs hampered all these alterations, except the LVEF reduction. Therefore, 4 weeks after MSC therapy was applied to CHF rats, MI size and myocardial interstitial fibrosis decreased, while baroreflex sensitivity and HRV improved.
Collapse
Affiliation(s)
| | | | - Renata Maria Lataro
- 1 Department of Physiology, Medical School of Ribeirao Preto, University of Sao Paulo , Ribeirão Preto, Brazil
| | - Carlos Alberto Aguiar Silva
- 1 Department of Physiology, Medical School of Ribeirao Preto, University of Sao Paulo , Ribeirão Preto, Brazil
| | | | | | - Marcus Vinicius Simões
- 2 Department of Internal Medicine, Medical School of Ribeirao Preto, University of Sao Paulo , Ribeirão Preto, Brazil
| | - Lindolfo da Silva Meirelles
- 3 Graduate Program in Cellular and Molecular Biology Applied to Health, Lutheran University of Brazil , Ribeirão Preto, Brazil
| | - Rubens Fazan
- 1 Department of Physiology, Medical School of Ribeirao Preto, University of Sao Paulo , Ribeirão Preto, Brazil
| | - Helio Cesar Salgado
- 1 Department of Physiology, Medical School of Ribeirao Preto, University of Sao Paulo , Ribeirão Preto, Brazil
| |
Collapse
|
724
|
Huang W, Lv B, Zeng H, Shi D, Liu Y, Chen F, Li F, Liu X, Zhu R, Yu L, Jiang X. Paracrine Factors Secreted by MSCs Promote Astrocyte Survival Associated With GFAP Downregulation After Ischemic Stroke via p38 MAPK and JNK. J Cell Physiol 2015; 230:2461-75. [DOI: 10.1002/jcp.24981] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 02/23/2015] [Accepted: 03/02/2015] [Indexed: 12/16/2022]
Affiliation(s)
- Weiyi Huang
- The National Key Clinic Specialty; The Neurosurgery Institute of Guangdong Province; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration; Department of Neurosurgery, Zhujiang Hospital; Southern Medical University; Guangzhou China
| | - Bingke Lv
- The National Key Clinic Specialty; The Neurosurgery Institute of Guangdong Province; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration; Department of Neurosurgery, Zhujiang Hospital; Southern Medical University; Guangzhou China
| | - Huijun Zeng
- The National Key Clinic Specialty; The Neurosurgery Institute of Guangdong Province; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration; Department of Neurosurgery, Zhujiang Hospital; Southern Medical University; Guangzhou China
| | - Dandan Shi
- Department of Anatomy; Key Laboratory of Construction and Detection of Guangdong Province; Southern Medical University; Guangzhou China
| | - Yi Liu
- The National Key Clinic Specialty; The Neurosurgery Institute of Guangdong Province; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration; Department of Neurosurgery, Zhujiang Hospital; Southern Medical University; Guangzhou China
| | - Fanfan Chen
- The National Key Clinic Specialty; The Neurosurgery Institute of Guangdong Province; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration; Department of Neurosurgery, Zhujiang Hospital; Southern Medical University; Guangzhou China
| | - Feng Li
- The National Key Clinic Specialty; The Neurosurgery Institute of Guangdong Province; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration; Department of Neurosurgery, Zhujiang Hospital; Southern Medical University; Guangzhou China
| | - Xinghui Liu
- Department of Anatomy; Key Laboratory of Construction and Detection of Guangdong Province; Southern Medical University; Guangzhou China
| | - Rong Zhu
- Department of Anatomy; Key Laboratory of Construction and Detection of Guangdong Province; Southern Medical University; Guangzhou China
| | - Lei Yu
- Department of Anatomy; Key Laboratory of Construction and Detection of Guangdong Province; Southern Medical University; Guangzhou China
| | - Xiaodan Jiang
- The National Key Clinic Specialty; The Neurosurgery Institute of Guangdong Province; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration; Department of Neurosurgery, Zhujiang Hospital; Southern Medical University; Guangzhou China
| |
Collapse
|
725
|
Wang WZ, Yao XD, Huang XJ, Li JQ, Xu H. Effects of TGF-β1 and alginate on the differentiation of rabbit bone marrow-derived mesenchymal stem cells into a chondrocyte cell lineage. Exp Ther Med 2015; 10:995-1002. [PMID: 26622428 DOI: 10.3892/etm.2015.2584] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2014] [Accepted: 05/01/2015] [Indexed: 01/21/2023] Open
Abstract
The aim of the present study was to investigate the effect of a three-dimensional (3D) culture system of sodium alginate gel on the directional differentiation induction of bone marrow-derived mesenchymal stem cells (BMSCs) into chondrocytes, as well as the in vitro gene transfection technique. The biological characteristics of the passage and proliferation of rabbit BMSCs were investigated under conditions of in vitro monolayer and 3D culture of sodium alginate gel. Transforming growth factor (TGF)-β1 gene recombinant adenoviral cosmid vectors and the recombinant adenoviral vector Ad.TGF-β1 were constructed, and the effect of Ad.TGF-β1 transfection on the differentiation of BMSCs into chondrocytes was investigated. The whole bone marrow rinsing method was used to obtain, separate and purify the rabbit BMSCs, and the in vitro monolayer and 3D culture of sodium alginate gel were thus successfully and stably established. A safe, stable and efficient method of constructing Ad.TGF-β1 TGF-β1 gene recombinant adenoviral vectors was established. Following TGF-β1 transfection, BMSCs were able to continuously secrete significantly increased amounts of specific extracellular matrix components of chondrocytes, such as collagen II and proteoglycans. Furthermore, the effects in the post-gene transfection 3D culture group were found to be enhanced compared with those in the monolayer culture group. In conclusion, the 3D culture system of sodium alginate gel and in vitro gene transfection exhibited significant inductive effects on differentiation, which could be used to promote BMSCs to differentiate into chondrocytes.
Collapse
Affiliation(s)
- Wan-Zong Wang
- Department of Orthopedics, Fuzhou General Hospital of Nanjing Command, PLA, Fuzhou, Fujian 350025, P.R. China
| | - Xiao-Dong Yao
- Department of Orthopedics, Fuzhou General Hospital of Nanjing Command, PLA, Fuzhou, Fujian 350025, P.R. China
| | - Xiao-Jin Huang
- Department of Orthopedics, Fuzhou General Hospital of Nanjing Command, PLA, Fuzhou, Fujian 350025, P.R. China
| | - Jin-Quan Li
- Department of Orthopedics, Fuzhou General Hospital of Nanjing Command, PLA, Fuzhou, Fujian 350025, P.R. China
| | - Hao Xu
- Department of Orthopedics, Fuzhou General Hospital of Nanjing Command, PLA, Fuzhou, Fujian 350025, P.R. China
| |
Collapse
|
726
|
Choi EW, Shin IS, Song JW, Yun TW, Yang J, Choi KS, Seong JK. Transplantation of Adipose Tissue-Derived Mesenchymal Stem Cells Prevents the Development of Lupus Dermatitis. Stem Cells Dev 2015; 24:2041-51. [PMID: 25941899 DOI: 10.1089/scd.2015.0021] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
MRL/lpr mice spontaneously develop high titers of anti-dsDNA antibodies and symptoms such as glomerular nephritis and organ weight gain. They also develop spontaneous skin inflammation similar to the cutaneous lesions common in human lupus erythematosus. This study aimed to compare the effects of long-term serial administration of human adipose tissue-derived mesenchymal stem cells (ASCs), CTLA4Ig-overexpressing ASCs, and cyclophosphamide treatment in MRL/lpr mice. MRL/lpr mice were divided into saline (C), cyclophosphamide (Y), ASC early (E), ASC late (L), and CTLA4Ig-overexpressing ASC (CT) treatment groups. Background-matched control MRL/MPJ mice treated with saline (N) were also compared. The treatment period was 5-23 weeks, except for the L group (15-23 weeks). Blood and tissue samples were collected when the mice were 24 weeks old. Organ weight, anti-dsDNA antibodies, urine protein, skin and kidney histologic abnormalities, and trabecular bone volume were evaluated. The Y group showed the greatest decrease in anti-dsDNA antibodies, organ weight, degree of kidney inflammation and glomerular infiltration of C3, and incidence rate of severe proteinuria; the E, L, and CT treatment groups showed better results than the C group. ASC transplantation reduced anti-dsDNA antibody levels significantly. Mice treated with ASCs or CTLA4Ig-ASCs starting from the early disease stage did not show dermatitis upon gross examination; they demonstrated significant improvement in hyperkeratosis, acanthosis, and inflammatory cell infiltration scores in histopathology. Micro-CT analysis revealed that cyclophosphamide treatment significantly decreased bone volume and increased bone spacing in the trabecular bone. Thus, we found that ASC and CTLA4-ASC treatments prevent lupus dermatitis development in MRL/lpr mice without adverse effects.
Collapse
Affiliation(s)
- Eun Wha Choi
- 1 Laboratory Animal Research Center, Samsung Biomedical Research Institute , Seoul, Republic of Korea.,2 School of Medicine, Sungkyunkwan University , Seoul, Republic of Korea
| | - Il Seob Shin
- 3 Biostar Stem Cell Research Center, K-STEMCELL , Seoul, Republic of Korea
| | - Ji Woo Song
- 1 Laboratory Animal Research Center, Samsung Biomedical Research Institute , Seoul, Republic of Korea
| | - Tae Won Yun
- 1 Laboratory Animal Research Center, Samsung Biomedical Research Institute , Seoul, Republic of Korea
| | - Jehoon Yang
- 1 Laboratory Animal Research Center, Samsung Biomedical Research Institute , Seoul, Republic of Korea.,2 School of Medicine, Sungkyunkwan University , Seoul, Republic of Korea
| | - Kyu-Sil Choi
- 1 Laboratory Animal Research Center, Samsung Biomedical Research Institute , Seoul, Republic of Korea.,2 School of Medicine, Sungkyunkwan University , Seoul, Republic of Korea
| | - Je Kyung Seong
- 4 Laboratory of Developmental Biology and Genomics, BK21 Program for Veterinary Science, Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University , Seoul, Republic of Korea
| |
Collapse
|
727
|
Tseng TC, Tao L, Hsieh FY, Wei Y, Chiu IM, Hsu SH. An Injectable, Self-Healing Hydrogel to Repair the Central Nervous System. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2015; 27:3518-24. [PMID: 25953204 DOI: 10.1002/adma.201500762] [Citation(s) in RCA: 394] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 04/04/2015] [Indexed: 05/24/2023]
Abstract
An injectable, self-healing hydrogel (≈1.5 kPa) is developed for healing nerve-system deficits. Neurosphere-like progenitors proliferate in the hydrogel and differentiate into neuron-like cells. In the zebrafish injury model, the central nervous system function is partially rescued by injection of the hydrogel and significantly rescued by injection of the neurosphere-laden hydrogel. The self-healing hydrogel may thus potentially repair the central nervous system.
Collapse
Affiliation(s)
- Ting-Chen Tseng
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, 10617, Taiwan, R.O.C
| | - Lei Tao
- The Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, 100084, P. R. China
| | - Fu-Yu Hsieh
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, 10617, Taiwan, R.O.C
| | - Yen Wei
- The Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, 100084, P. R. China
| | - Ing-Ming Chiu
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli County, 35053, Taiwan, R.O.C
| | - Shan-hui Hsu
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, 10617, Taiwan, R.O.C
| |
Collapse
|
728
|
Aquapuncture Using Stem Cell Therapy to Treat Mdx Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:132706. [PMID: 26074983 PMCID: PMC4444575 DOI: 10.1155/2015/132706] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 04/19/2015] [Indexed: 01/29/2023]
Abstract
Duchenne muscular dystrophy (DMD) occurs due to genetic mutations that lead to absence or decrease of dystrophin protein generating progressive muscle degeneration. Cell therapy using mesenchymal stem cell (MSC) has been described as a treatment to DMD. In this work, MSC derived from deciduous teeth, called stem cells from human exfoliated deciduous teeth (SHED), were injected in acupoint as an alternative therapy to minimize muscle degeneration in twenty-two mdx mice. The treatment occurred three times with intervals of 21 days, and animals were analyzed four times: seven days prior treatment (T-7); 10 days after first treatment (T10); 10 days after second treatment (T31); and 10 days after third treatment (T52). Animals were evaluated by wire test for estimate strength and blood was collected to perform a creatinine phosphokinase analysis. After euthanasia, cranial tibial muscles were collected and submitted to histological and immunohistochemistry analyses. Treated groups presented improvement of strength and reduced creatinine phosphokinase levels. Also, a slight dystrophin increase was observed in tibial cranial muscle when aquapuncture was associated SHED. All therapies have minimized muscle degeneration, but the association of aquapuncture with SHED appears to have better effect, reducing muscle damage, suggesting a therapeutic value.
Collapse
|
729
|
Arora S, Saha S, Roy S, Das M, Jana SS, Ta M. Role of Nonmuscle Myosin II in Migration of Wharton's Jelly-Derived Mesenchymal Stem Cells. Stem Cells Dev 2015; 24:2065-77. [PMID: 25923805 DOI: 10.1089/scd.2015.0095] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
It is the promise of regeneration and therapeutic applications that has sparked an interest in mesenchymal stem cells (MSCs). Following infusion, MSCs migrate to sites of injury or inflammation by virtue of their homing property. To exert optimal clinical benefits, systemically delivered MSCs need to migrate efficiently and in adequate numbers to pathological areas in vivo. However, underlying molecular mechanisms responsible for MSC migration are still not well understood. The Wharton's jelly (WJ) of the umbilical cord is an attractive source of MSCs for stem cell therapy because of its abundant availability and painless collection. In this study, we attempted to identify the role of nonmuscle myosin II (NMII), if any, in the migration of WJ-derived MSCs (WJ-MSCs). Expression of NMII isoforms, NMIIA, and NMIIB was observed both at RNA and protein levels in WJ-MSCs. Inhibition of NMII or its regulator ROCK, by pharmacological inhibitors, resulted in significant reduction in the migration of WJ-MSCs as confirmed by the scratch migration assay and time-lapse microscopy. Next, trying to dissect the role of each NMII isoform in migration of WJ-MSCs, we found that siRNA-mediated downregulation of NMIIA, but not NMIIB expression, led to cells failing to retract their trailing edge and losing cell-cell cohesiveness, while exhibiting a nondirectional migratory pathway. Migration, moreover, is also dependent on optimal affinity adhesion, which would allow rapid attachment and release of cells and, hence, can be influenced by extracellular matrix (ECM) and adhesion molecules. We demonstrated that inhibition of NMII and more specifically NMIIA resulted in increased gene expression of ECM and adhesion molecules, which possibly led to stronger adhesions and, hence, decreased migration. Therefore, these data suggest that NMII acts as a regulator of cell migration and adhesion in WJ-MSCs.
Collapse
Affiliation(s)
- Sneha Arora
- 1 Indian Institute of Science Education and Research Kolkata , Mohanpur, India
| | - Shekhar Saha
- 2 Indian Association for the Cultivation of Science , Kolkata, India
| | - Saheli Roy
- 1 Indian Institute of Science Education and Research Kolkata , Mohanpur, India
| | - Madhurima Das
- 1 Indian Institute of Science Education and Research Kolkata , Mohanpur, India
| | - Siddhartha S Jana
- 2 Indian Association for the Cultivation of Science , Kolkata, India
| | - Malancha Ta
- 1 Indian Institute of Science Education and Research Kolkata , Mohanpur, India
| |
Collapse
|
730
|
Qiu X, Zhang Y, Zhao X, Zhang S, Wu J, Guo H, Hu Y. Enhancement of endothelial differentiation of adipose derived mesenchymal stem cells by a three-dimensional culture system of microwell. Biomaterials 2015; 53:600-8. [DOI: 10.1016/j.biomaterials.2015.02.115] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 02/26/2015] [Accepted: 02/27/2015] [Indexed: 12/15/2022]
|
731
|
Mutlu L, Hufnagel D, Taylor HS. The endometrium as a source of mesenchymal stem cells for regenerative medicine. Biol Reprod 2015; 92:138. [PMID: 25904012 PMCID: PMC4652610 DOI: 10.1095/biolreprod.114.126771] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 12/11/2014] [Accepted: 04/09/2015] [Indexed: 12/21/2022] Open
Abstract
Stem cell therapies have opened new frontiers in medicine with the possibility of regenerating lost or damaged cells. Embryonic stem cells, induced pluripotent stem cells, hematopoietic stem cells, and mesenchymal stem cells have been used to derive mature cell types for tissue regeneration and repair. However, the endometrium has emerged as an attractive, novel source of adult stem cells that are easily accessed and demonstrate remarkable differentiation capacity. In this review, we summarize our current understanding of endometrial stem cells and their therapeutic potential in regenerative medicine.
Collapse
Affiliation(s)
- Levent Mutlu
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut
| | - Demetra Hufnagel
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut
| | - Hugh S Taylor
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
732
|
Hayashi Y, Murakami M, Kawamura R, Ishizaka R, Fukuta O, Nakashima M. CXCL14 and MCP1 are potent trophic factors associated with cell migration and angiogenesis leading to higher regenerative potential of dental pulp side population cells. Stem Cell Res Ther 2015; 6:111. [PMID: 26021377 PMCID: PMC4488059 DOI: 10.1186/s13287-015-0088-z] [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: 02/13/2015] [Revised: 02/13/2015] [Accepted: 05/05/2015] [Indexed: 12/19/2022] Open
Abstract
Introduction The release of trophic factors from mesenchymal stem cells (MSCs) is critical for tissue regeneration. A systematic investigation of the regenerative potential of trophic factors from different MSCs, however, has not been performed. Thus, in the present study, the regenerative potential of conditioned medium (CM) from dental pulp, bone marrow, and adipose tissue-derived CD31− side population (SP) cells from an individual source was compared in an ectopic tooth transplantation model. Methods The tooth root transplantation in an ectopic site model was used for investigation of the regenerative potential and trophic effects in vivo. Either pulp CD31− SP cell populations (1×106 cells) at the third to fourth passage or 5 μg/ml of CM from dental pulp, bone marrow, and adipose stem cells from four different individuals were injected into the root with collagen TE. Each root was transplanted subcutaneously in 5-week-old severe combined immunodeficiency mice. Each root with surrounding tissue was harvested for histology on days 7, 21, and 28 and for Western blot analysis and real-time reverse transcription-polymerase chain reaction (RT-PCR) analysis on day 28. Furthermore, the trophic factors responsible for the regenerative potential were identified as the upregulated genes present in pulp CD31− SP cells when compared with the genes in both bone marrow and adipose CD31− SP cells by using microarray analysis, real-time RT-PCR, and Western blot analysis. Results Transplantation of pulp CM yielded increased volume of pulp regeneration, more bromodeoxyuridine (BrdU)-positive migrated cells, and fewer caspase 3-positive cells in the regenerated pulp compared with the others. Pulp CM also demonstrated significantly increased cell migration, anti-apoptosis, and angiogenesis in C2C12 cells. Higher expression of CXCL14 and MCP1 in pulp SP cells suggested candidate trophic factors. The stimulatory effects on both migration and angiogenesis of CXCL14 and MCP1 were demonstrated in vitro. In the regenerated tissue, BrdU-positive migrated cells expressed CXCR4 and CCR2, receptors for CXCL14 and MCP1, respectively. Conclusions The higher regenerative potential of pulp SP cells may be due to potent trophic factors, including CXCL14 and MCP1, which promote migration and angiogenesis.
Collapse
Affiliation(s)
- Y Hayashi
- Department of Dental Regenerative Medicine, Center of Advanced Medicine for Dental and Oral Diseases, National Center for Geriatrics and Gerontology, Research Institute, Morioka 7-430, Obu, Aichi, 474-8511, Japan. .,Department of Pediatric Dentistry, School of Dentistry, Aichi-Gakuin University suemoridouri 2-11, Nagoya, Aichi, 464-8651, Japan.
| | - M Murakami
- Department of Dental Regenerative Medicine, Center of Advanced Medicine for Dental and Oral Diseases, National Center for Geriatrics and Gerontology, Research Institute, Morioka 7-430, Obu, Aichi, 474-8511, Japan.
| | - R Kawamura
- Department of Dental Regenerative Medicine, Center of Advanced Medicine for Dental and Oral Diseases, National Center for Geriatrics and Gerontology, Research Institute, Morioka 7-430, Obu, Aichi, 474-8511, Japan. .,Department of Gerodontology, School of Dentistry, Aichi-Gakuin University suemoridouri 2-11, Nagoya, Aichi, 464-8651, Japan.
| | - R Ishizaka
- Department of Pediatric Dentistry, School of Dentistry, Aichi-Gakuin University suemoridouri 2-11, Nagoya, Aichi, 464-8651, Japan.
| | - O Fukuta
- Department of Pediatric Dentistry, School of Dentistry, Aichi-Gakuin University suemoridouri 2-11, Nagoya, Aichi, 464-8651, Japan.
| | - M Nakashima
- Department of Dental Regenerative Medicine, Center of Advanced Medicine for Dental and Oral Diseases, National Center for Geriatrics and Gerontology, Research Institute, Morioka 7-430, Obu, Aichi, 474-8511, Japan.
| |
Collapse
|
733
|
Gray A, Maguire T, Schloss R, Yarmush ML. Identification of IL-1β and LPS as optimal activators of monolayer and alginate-encapsulated mesenchymal stromal cell immunomodulation using design of experiments and statistical methods. Biotechnol Prog 2015; 31:1058-70. [PMID: 25958832 DOI: 10.1002/btpr.2103] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 04/23/2015] [Indexed: 12/13/2022]
Abstract
Induction of therapeutic mesenchymal stromal cell (MSC) function is dependent upon activating factors present in diseased or injured tissue microenvironments. These functions include modulation of macrophage phenotype via secreted molecules including prostaglandin E2 (PGE2). Many approaches aim to optimize MSC-based therapies, including preconditioning using soluble factors and cell immobilization in biomaterials. However, optimization of MSC function is usually inefficient as only a few factors are manipulated in parallel. We utilized fractional factorial design of experiments to screen a panel of 6 molecules (lipopolysaccharide [LPS], polyinosinic-polycytidylic acid [poly(I:C)], interleukin [IL]-6, IL-1β, interferon [IFN]-β, and IFN-γ), individually and in combinations, for the upregulation of MSC PGE2 secretion and attenuation of macrophage secretion of tumor necrosis factor (TNF)-α, a pro-inflammatory molecule, by activated-MSC conditioned medium (CM). We used multivariable linear regression (MLR) and analysis of covariance to determine differences in functions of optimal factors on monolayer MSCs and alginate-encapsulated MSCs (eMSCs). The screen revealed that LPS and IL-1β potently activated monolayer MSCs to enhance PGE2 production and attenuate macrophage TNF-α. Activation by LPS and IL-1β together synergistically increased MSC PGE2, but did not synergistically reduce macrophage TNF-α. MLR and covariate analysis revealed that macrophage TNF-α was strongly dependent on the MSC activation factor, PGE2 level, and macrophage donor but not MSC culture format (monolayer versus encapsulated). The results demonstrate the feasibility and utility of using statistical approaches for higher throughput cell analysis. This approach can be extended to develop activation schemes to maximize MSC and MSC-biomaterial functions prior to transplantation to improve MSC therapies.
Collapse
Affiliation(s)
- Andrea Gray
- Dept. of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854
| | - Timothy Maguire
- Dept. of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854
| | - Rene Schloss
- Dept. of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854
| | - Martin L Yarmush
- Dept. of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854
| |
Collapse
|
734
|
Abbah SA, Spanoudes K, O'Brien T, Pandit A, Zeugolis DI. Assessment of stem cell carriers for tendon tissue engineering in pre-clinical models. Stem Cell Res Ther 2015; 5:38. [PMID: 25157898 PMCID: PMC4056691 DOI: 10.1186/scrt426] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Tendon injuries are prevalent and problematic, especially among young and otherwise healthy individuals. The inherently slow innate healing process combined with the inevitable scar tissue formation compromise functional recovery, imposing the need for the development of therapeutic strategies. The limited number of low activity/reparative capacity tendon-resident cells has directed substantial research efforts towards the exploration of the therapeutic potential of various stem cells in tendon injuries and pathophysiologies. Severe injuries require the use of a stem cell carrier to enable cell localisation at the defect site. The present study describes advancements that injectable carriers, tissue grafts, anisotropically orientated biomaterials, and cell-sheets have achieved in preclinical models as stem cell carriers for tendon repair.
Collapse
|
735
|
Pipino C, Pandolfi A. Osteogenic differentiation of amniotic fluid mesenchymal stromal cells and their bone regeneration potential. World J Stem Cells 2015; 7:681-690. [PMID: 26029340 PMCID: PMC4444609 DOI: 10.4252/wjsc.v7.i4.681] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 01/16/2015] [Accepted: 02/11/2015] [Indexed: 02/06/2023] Open
Abstract
In orthopedics, tissue engineering approach using stem cells is a valid line of treatment for patients with bone defects. In this context, mesenchymal stromal cells of various origins have been extensively studied and continue to be a matter of debate. Although mesenchymal stromal cells from bone marrow are already clinically applied, recent evidence suggests that one may use mesenchymal stromal cells from extra-embryonic tissues, such as amniotic fluid, as an innovative and advantageous resource for bone regeneration. The use of cells from amniotic fluid does not raise ethical problems and provides a sufficient number of cells without invasive procedures. Furthermore, they do not develop into teratomas when transplanted, a consequence observed with pluripotent stem cells. In addition, their multipotent differentiation ability, low immunogenicity, and anti-inflammatory properties make them ideal candidates for bone regenerative medicine. We here present an overview of the features of amniotic fluid mesenchymal stromal cells and their potential in the osteogenic differentiation process. We have examined the papers actually available on this regard, with particular interest in the strategies applied to improve in vitro osteogenesis. Importantly, a detailed understanding of the behavior of amniotic fluid mesenchymal stromal cells and their osteogenic ability is desirable considering a feasible application in bone regenerative medicine.
Collapse
|
736
|
Human Amnion-Derived Mesenchymal Stem Cell Transplantation Ameliorates Liver Fibrosis in Rats. Transplant Direct 2015; 1:e16. [PMID: 27500218 DOI: 10.1097/txd.0000000000000525] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 04/24/2015] [Indexed: 12/19/2022] Open
Abstract
UNLABELLED Mesenchymal stem cells (MSCs) are a valuable cell source in regenerative medicine. Recently, several studies have shown that MSCs can be easily isolated from human amnion. In this study, we investigated the therapeutic effect of transplantation of human amnion-derived MSCs (hAMSCs) in rats with liver fibrosis. METHODS Liver fibrosis was induced by an intraperitoneal injection of 2 mL/kg of 50% carbon tetrachloride twice a week for 6 weeks. At 3 weeks, hAMSCs (1 × 10(6) cells) were transplanted intravenously. Rats were sacrificed at 7 weeks, and histological analyses and quantitative reverse-transcription polymerase chain reaction were performed. In vitro experiments were conducted to investigate the effect of hAMSCs on the activation of Kupffer cells. RESULTS Transplantation of hAMSCs significantly reduced the fibrotic area, deposition of type-I collagen, the number of α-smooth muscle actin-positive hepatic stellate cells, and CD68-positive Kupffer cells in the livers. messenger RNA expression of α-smooth muscle actin and tissue inhibitor of metalloproteinase-1 was significantly decreased and the expression of matrix metalloproteinase-9 and hepatocyte growth factor was significantly increased in the liver of hAMSC-treated rats. Transplantation of hAMSCs at 3 weeks plus 5 weeks did not have an additive effect. In vitro experiments demonstrated that Kupffer cell activation induced by lipopolysaccharide was significantly decreased by culturing with conditioned medium obtained from hAMSCs. CONCLUSIONS Transplantation of hAMSCs provided significant improvement in a rat model of liver fibrosis, possibly through the inhibition of Kupffer cell and hepatic stellate cell activation. hAMSCs may be a potential new treatment for liver fibrosis.
Collapse
|
737
|
Wang J, Zhao Y, Wu X, Yin S, Chuai Y, Wang A. The utility of human fallopian tube mucosa as a novel source of multipotent stem cells for the treatment of autologous reproductive tract injury. Stem Cell Res Ther 2015; 6:98. [PMID: 25994820 PMCID: PMC4474566 DOI: 10.1186/s13287-015-0094-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 09/03/2014] [Accepted: 05/12/2015] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION Fallopian tube, which is normally discarded in surgical procedures, has proven to be a source of mesenchymal stem cells (MSCs) with increasing evidence. However, fallopian tube mucosa, which can be acquired via non-invasive procedures, is a previously unknown source of MSCs. In the present study, we explored the existence of MSCs in the human fallopian tube mucosa and also compared multipotent stem cells derived from fallopian tubes and fallopian tube mucosa according to their biological characteristics and therapeutic potential for treatment of autologous reproductive tract injury. METHODS Cells isolated from human fallopian tubes and fallopian tube mucosa were expanded and characterised by flow cytometry. The proliferative capacity of both cell types was measured by performing colony-forming unit-fibroblast and Cell Counting Kit-8 assays. Both cell types underwent in vitro adipogenic, chondrogenic, and osteogenic differentiation. The expression of osteocyte-, adipocyte-, and chondrocyte-related genes in the differentiated cell lineages was assessed by reverse transcription-polymerase chain reaction. The secretion of growth factors and immunomodulatory cytokines by both cell types were measured by enzyme-linked immunosorbent assays. RESULTS We found that MSCs existed in the fallopian tube mucosa. The comparison between human fallopian tube MSCs (hFTMSCs) and human fallopian tube mucosa MSCs (hFMMSCs) showed that hFTMSCs had a stronger proliferative capacity and shorter duplication time than hFMMSCs. Both cell types could be differentiated into adipocytes, osteoblasts, or chondrocytes in vitro. Real-time polymerase chain reaction analysis demonstrated that hFTMSCs displayed increased expression of osteogenic-specific genes compared with hFMMSCs, but the two types of cells showed no significant increase in the mRNA expression of adipogenic-specific or chondrogenic-specific genes. hFMMSCs and hFTMSCs robustly produced a variety of growth factors and immunomodulatory cytokines. CONCLUSIONS Human fallopian tube mucosa is a novel source of multipotent cells. hFMMSCs demonstrated stronger proliferative capacity and superior secretion of growth factors and immunomodulatory cytokines than hFTMSCs, making the former a better source of stem cells for the treatment of autologous reproductive tract injury. Compared with fallopian tube, fallopian tube mucosa has more wide-ranging applications and can be used to carry out autologous transplantation.
Collapse
Affiliation(s)
- Jiaojiao Wang
- Department for Gynaecology and Obstetrics, Navy General Hospital, PLA, Fuchengmen Road, No.6, Beijing, 100048, China.
| | - Yong Zhao
- Department for Gynaecology and Obstetrics, Navy General Hospital, PLA, Fuchengmen Road, No.6, Beijing, 100048, China.
| | - Xiaoyun Wu
- Jing-Meng Stem Cell Technology CO., Ltd, Shangdi East Road,No.5-2, Beijing, 100048, China.
| | - Shande Yin
- Department for Gynaecology and Obstetrics, Navy General Hospital, PLA, Fuchengmen Road, No.6, Beijing, 100048, China.
| | - Yunhai Chuai
- Department for Gynaecology and Obstetrics, Navy General Hospital, PLA, Fuchengmen Road, No.6, Beijing, 100048, China.
| | - Aiming Wang
- Department for Gynaecology and Obstetrics, Navy General Hospital, PLA, Fuchengmen Road, No.6, Beijing, 100048, China.
| |
Collapse
|
738
|
Lan YW, Choo KB, Chen CM, Hung TH, Chen YB, Hsieh CH, Kuo HP, Chong KY. Hypoxia-preconditioned mesenchymal stem cells attenuate bleomycin-induced pulmonary fibrosis. Stem Cell Res Ther 2015; 6:97. [PMID: 25986930 PMCID: PMC4487587 DOI: 10.1186/s13287-015-0081-6] [Citation(s) in RCA: 169] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 03/19/2015] [Accepted: 04/21/2015] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION Idiopathic pulmonary fibrosis is a progressive diffuse parenchymal lung disorder of unknown etiology. Mesenchymal stem cell (MSC)-based therapy is a novel approach with great therapeutic potential for the treatment of lung diseases. Despite demonstration of MSC grafting, the populations of engrafted MSCs have been shown to decrease dramatically 24 hours post-transplantation due to exposure to harsh microenvironments. Hypoxia is known to induce expression of cytoprotective genes and also secretion of anti-inflammatory, anti-apoptotic and anti-fibrotic factors. Hypoxic preconditioning is thought to enhance the therapeutic potency and duration of survival of engrafted MSCs. In this work, we aimed to prolong the duration of survival of engrafted MSCs and to enhance the effectiveness of idiopathic pulmonary fibrosis transplantation therapy by the use of hypoxia-preconditioned MSCs. METHODS Hypoxic preconditioning was achieved in MSCs under an optimal hypoxic environment. The expression levels of cytoprotective factors and their biological effects on damaged alveolar epithelial cells or transforming growth factor-beta 1-treated fibroblast cells were studied in co-culture experiments in vitro. Furthermore, hypoxia-preconditioned MSCs (HP-MSCs) were intratracheally instilled into bleomycin-induced pulmonary fibrosis mice at day 3, and lung functions, cellular, molecular and pathological changes were assessed at 7 and 21 days after bleomycin administration. RESULTS The expression of genes for pro-survival, anti-apoptotic, anti-oxidant and growth factors was upregulated in MSCs under hypoxic conditions. In transforming growth factor-beta 1-treated MRC-5 fibroblast cells, hypoxia-preconditioned MSCs attenuated extracellular matrix production through paracrine effects. The pulmonary respiratory functions significantly improved for up to 18 days of hypoxia-preconditioned MSC treatment. Expression of inflammatory factors and fibrotic factor were all downregulated in the lung tissues of the hypoxia-preconditioned MSC-treated mice. Histopathologic examination observed a significant amelioration of the lung fibrosis. Several LacZ-labeled MSCs were observed within the lungs in the hypoxia-preconditioned MSC treatment groups at day 21, but no signals were detected in the normoxic MSC group. Our data further demonstrated that upregulation of hepatocyte growth factor possibly played an important role in mediating the therapeutic effects of transplanted hypoxia-preconditioned MSCs. CONCLUSION Transplantation of hypoxia-preconditioned MSCs exerted better therapeutic effects in bleomycin-induced pulmonary fibrotic mice and enhanced the survival rate of engrafted MSCs, partially due to the upregulation of hepatocyte growth factor.
Collapse
Affiliation(s)
- Ying-Wei Lan
- Division of Biotechnology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan, Republic of China.
| | - Kong-Bung Choo
- Department of Preclinical Sciences, Faculty of Medicine and Health Sciences and Centre for Stem Cell Research, Universiti Tunku Abdul Rahman, Selangor, Malaysia.
| | - Chuan-Mu Chen
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan, Republic of China.
- Agricultural Biotechnology Center, National Chung Hsing University, Taichung, Taiwan, Republic of China.
- Rong-Hsing Translational Medicine Center, National Chung Hsing University, Taichung, Taiwan, Republic of China.
| | - Tsai-Hsien Hung
- Division of Biotechnology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan, Republic of China.
| | - Young-Bin Chen
- Institute of Biotechnology, National Taiwan University, Taichung, Taiwan, Republic of China.
| | - Chung-Hsing Hsieh
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan, Republic of China.
- Department of Thoracic Medicine, St Paul's Hospital, Taoyuan, Taiwan, Republic of China.
- Department of Thoracic Medicine, Ton-Yen General Hospital, Hsinchu, Taiwan, Republic of China.
| | - Han-Pin Kuo
- Department of Thoracic Medicine, Pulmonary Disease Research Center, Chang Gung Memorial Hospital, Taipei, Taiwan, Republic of China.
- Department of Medicine, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan, Republic of China.
| | - Kowit-Yu Chong
- Division of Biotechnology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan, Republic of China.
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan, Republic of China.
- Molecular Medicine Research Center, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan, Republic of China.
| |
Collapse
|
739
|
Iser IC, Bracco PA, Gonçalves CEI, Zanin RF, Nardi NB, Lenz G, Battastini AMO, Wink MR. Mesenchymal stem cells from different murine tissues have differential capacity to metabolize extracellular nucleotides. J Cell Biochem 2015; 115:1673-82. [PMID: 24802095 DOI: 10.1002/jcb.24830] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 03/29/2014] [Accepted: 05/02/2014] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) have shown a great potential for cell-based therapy and many different therapeutic purposes. Despite the recent advances in the knowledge of MSCs biology, their biochemical and molecular properties are still poorly defined. Ecto-nucleoside triphosphate diphosphohydrolases (E-NTPDases) and ecto-5'-nucleotidase (eNT/CD73) are widely expressed enzymes that hydrolyze extracellular nucleotides, generating an important cellular signaling cascade. Currently, studies have evidenced the relationship between the purinergic system and the development, maintenance, and differentiation of stem cells. The objective of this study is to identify the NTPDases and eNT/CD73 and compare the levels of nucleotide hydrolysis on MSCs isolated from different murine tissues (bone marrow, lung, vena cava, kidney, pancreas, spleen, skin, and adipose tissue). MSCs from all tissues investigated expressed the ectoenzymes at different levels. In MSCs from pancreas and adipose tissue, the hydrolysis of triphosphonucleosides was significantly higher when compared to the other cells. The diphosphonucleosides were hydrolyzed at a higher rate by MSC from pancreas when compared to MSC from other tissues. The differential nucleotide hydrolysis activity and enzyme expression in these cells suggests that MSCs play different roles in regulating the purinergic system in these tissues. Overall MSCs are an attractive adult-derived cell population for therapies, however, the fact that ecto-nucleotide metabolism can affect the microenvironment, modulating important events, such as immune response, makes the assessment of this metabolism an important part of the characterization of MSCs to be applied therapeutically.
Collapse
Affiliation(s)
- Isabele C Iser
- Departamento de Ciências Básicas da Saúde e Laboratório de Biologia Celular, Universidade Federal de Ciências da Saúde de Porto Alegre-UFCSPA, Porto Alegre, RS, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
740
|
Caliari-Oliveira C, Yaochite JNU, Ramalho LNZ, Palma PVB, Carlos D, Cunha FDQ, De Souza DA, Frade MAC, Covas DT, Malmegrim KCR, Oliveira MC, Voltarelli JC. Xenogeneic Mesenchymal Stromal Cells Improve Wound Healing and Modulate the Immune Response in an Extensive Burn Model. Cell Transplant 2015; 25:201-15. [PMID: 25955320 DOI: 10.3727/096368915x688128] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Major skin burns are difficult to treat. Patients often require special care and long-term hospitalization. Besides specific complications associated with the wounds themselves, there may be impairment of the immune system and of other organs. Mesenchymal stromal cells (MSCs) are a recent therapeutic alternative to treat burns, mainly aiming to accelerate the healing process. Several MSC properties favor their use as therapeutic approach, as they promote angiogenesis, stimulate regeneration, and enhance the immunoregulatory function. Moreover, since patients with extensive burns require urgent treatment and because the expansion of autologous MSCs is a time-consuming process, in this present study we chose to evaluate the therapeutic potential of xenogeneic MSCs in the treatment of severe burns in rats. MSCs were isolated from mouse bone marrow, expanded in vitro, and intradermally injected in the periphery of burn wounds. MSC-treated rats presented higher survival rates (76.19%) than control animals treated with PBS (60.86%, p < 0.05). In addition, 60 days after the thermal injury, the MSC-treated group showed larger proportion of healed areas within the burn wounds (90.81 ± 5.05%) than the PBS-treated group (76.11 ± 3.46%, p = 0.03). We also observed that CD4(+) and CD8(+) T cells in spleens and in damaged skin, as well as the percentage of neutrophils in the burned area, were modulated by MSC treatment. Plasma cytokine (TGF-β, IL-10, IL-6, and CINC-1) levels were also altered in the MSC-treated rats, when compared to controls. Number of injected GFP(+) MSCs progressively decreased over time, and 60 days after injection, few MSCs were still detected in the skin of treated animals. This study demonstrates the therapeutic effectiveness of intradermal application of MSCs in a rat model of deep burns, providing basis for future regenerative therapies in patients suffering from deep burn injuries.
Collapse
Affiliation(s)
- Carolina Caliari-Oliveira
- Department of Biochemistry and Immunology, Basic and Applied Immunology Program, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
741
|
Dooley LM, Abdalmula A, Washington EA, Kaufman C, Tudor EM, Ghosh P, Itescu S, Kimpton WG, Bailey SR. Effect of mesenchymal precursor cells on the systemic inflammatory response and endothelial dysfunction in an ovine model of collagen-induced arthritis. PLoS One 2015; 10:e0124144. [PMID: 25950840 PMCID: PMC4423911 DOI: 10.1371/journal.pone.0124144] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 03/10/2015] [Indexed: 02/07/2023] Open
Abstract
Background and Aim Mesenchymal precursor cells (MPC) are reported to possess immunomodulatory properties that may prove beneficial in autoimmune and other inflammatory conditions. However, their mechanism of action is poorly understood. A collagen-induced arthritis model has been previously developed which demonstrates local joint inflammation and systemic inflammatory changes. These include not only increased levels of inflammatory markers, but also vascular endothelial cell dysfunction, characterised by reduced endothelium-dependent vasodilation. This study aimed to characterise the changes in systemic inflammatory markers and endothelial function following the intravenous administration of MPC, in the ovine model. Methods Arthritis was induced in sixteen adult sheep by administration of bovine type II collagen into the hock joint following initial sensitisation. After 24h, sheep were administered either 150 million allogeneic ovine MPCs intravenously, or saline only. Fibrinogen and serum amyloid-A were measured in plasma to assess systemic inflammation, along with pro-inflammatory and anti-inflammatory cytokines. Animals were necropsied two weeks following arthritis induction. Coronary and digital arterial segments were mounted in a Mulvaney-Halpern wire myograph. The relaxant response to endothelium-dependent and endothelium-independent vasodilators was used to assess endothelial dysfunction. Results and Conclusion Arthritic sheep treated with MPC demonstrated a marked spike in plasma IL-10, 24h following MPC administration. They also showed significantly reduced plasma levels of the inflammatory markers, fibrinogen and serum amyloid A, and increased HDL. Coronary arteries from RA sheep treated with MPCs demonstrated a significantly greater maximal relaxation to bradykinin when compared to untreated RA sheep (253.6 ± 17.1% of pre-contracted tone vs. 182.3 ± 27.3% in controls), and digital arteries also demonstrated greater endothelium-dependent vasodilation. This study demonstrated that MPCs given intravenously are able to attenuate systemic inflammatory changes associated with a monoarthritis, including the development of endothelial dysfunction.
Collapse
Affiliation(s)
- Laura M. Dooley
- Faculty of Veterinary Science, The University of Melbourne, Parkville, Victoria, Australia
| | - Anwar Abdalmula
- Faculty of Veterinary Science, The University of Melbourne, Parkville, Victoria, Australia
| | | | - Claire Kaufman
- Faculty of Veterinary Science, The University of Melbourne, Parkville, Victoria, Australia
| | - Elizabeth M. Tudor
- Faculty of Veterinary Science, The University of Melbourne, Parkville, Victoria, Australia
| | - Peter Ghosh
- Mesoblast Ltd, Melbourne, Victoria, Australia
| | | | - Wayne G. Kimpton
- Faculty of Veterinary Science, The University of Melbourne, Parkville, Victoria, Australia
| | - Simon R. Bailey
- Faculty of Veterinary Science, The University of Melbourne, Parkville, Victoria, Australia
- * E-mail:
| |
Collapse
|
742
|
Cryopreservation of microencapsulated murine mesenchymal stem cells genetically engineered to secrete erythropoietin. Int J Pharm 2015; 485:15-24. [DOI: 10.1016/j.ijpharm.2015.02.047] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 02/17/2015] [Indexed: 01/06/2023]
|
743
|
Interleukin-17 and its implication in the regulation of differentiation and function of hematopoietic and mesenchymal stem cells. Mediators Inflamm 2015; 2015:470458. [PMID: 25999667 PMCID: PMC4427009 DOI: 10.1155/2015/470458] [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: 01/12/2015] [Revised: 03/24/2015] [Accepted: 03/24/2015] [Indexed: 12/21/2022] Open
Abstract
Adult stem cells have a great potential applicability in regenerative medicine and cell-based therapies. However, there are still many unresolved issues concerning their biology, and the influence of the local microenvironment on properties of stem cells has been increasingly recognized. Interleukin (IL-) 17, as a cytokine implicated in many physiological and pathological processes, should be taken into consideration as a part of a regulatory network governing tissue-associated stem cells' fate. This review is focusing on the published data on the effects of IL-17 on the properties and function of hematopoietic and mesenchymal stem cells and trying to discuss that IL-17 achieves many of its roles by acting on adult stem cells.
Collapse
|
744
|
Zou Z, Cai Y, Chen Y, Chen S, Liu L, Shen Z, Zhang S, Xu L, Chen Y. Bone marrow-derived mesenchymal stem cells attenuate acute liver injury and regulate the expression of fibrinogen-like-protein 1 and signal transducer and activator of transcription 3. Mol Med Rep 2015; 12:2089-97. [PMID: 25901902 DOI: 10.3892/mmr.2015.3660] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 02/27/2015] [Indexed: 01/16/2023] Open
Abstract
In recent years, bone marrow-derived mesenchymal stem cells (BMSCs) have been demonstrated to exert extensive therapeutic effects on acute liver injury; however, the underlying mechanisms of these effects have remained to be elucidated. The present study focused on the potential anti-apoptotic and pro-regenerative effects of BMSCs in D-galactosamine (D-Gal) and lipopolysaccharide (LPS)-induced acute liver injury in rats. An experimental rat acute liver injury model was established by intraperitoneal injection of D-Gal (400 mg/kg) and LPS (80 μg/kg). BMSCs and an identical volume of saline were administered via the caudal vein 2 h after the D-Gal and LPS challenge. Subsequently, the serum samples were collected to detect the levels of alanine aminotransferase and aspartate aminotransferase. Hematoxylin and eosin staining, terminal deoxynucleotidyl transferase-mediated nick-end labeling assay and immunohistochemical staining were performed to determine apoptosis, regeneration and histological changes of liver sections. Western blotting and reverse transcription-quantitative polymerase chain reaction were performed to detect the protein and mRNA expression levels of fibrinogen-like-protein 1 (FGL1), phosphorylated signal transducer and activator of transcription 3 (p-STAT3), STAT3 and B-cell lymphoma 2 (Bcl-2) and Bcl-2 associated X protein (Bax) in liver tissue samples. The results indicated that intravenous transplantation of BMSCs significantly decreased the levels of alanine aminotransferase and aspartate aminotransferase, and reduced hepatocellular necrosis and inflammatory cell infiltration. Additionally, a terminal deoxynucleotidyl transferase-mediated nick-end labeling assay and immunohistochemical staining revealed that BMSC treatment reduced hepatocyte apoptosis and enhanced liver regeneration. Furthermore, Bcl-2 expression was increased, whilst the protein expression of Bax was reduced. The expression of FGL1 and p-STAT3 were elevated concurrently with the improvement of liver function. These results demonstrated that BMSCs may provide a promising potential agent for the prevention of acute liver injury via inhibition of hepatocyte apoptosis and acceleration of liver regeneration. The mechanism may be, a least in part, a consequence of the upregulation of FGL1 expression and the induction of STAT3 phosphorylation.
Collapse
Affiliation(s)
- Zhuolin Zou
- Department of Infectious Disease, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Yijing Cai
- Department of Infectious Disease, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Yi Chen
- Department of Infectious Disease, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Si Chen
- Department of Infectious Disease, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Liyuan Liu
- Department of Infectious Disease, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Zhonghai Shen
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Sainan Zhang
- Department of Infectious Disease, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Lanman Xu
- Department of Infectious Disease, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Yongping Chen
- Department of Infectious Disease, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
745
|
Human Umbilical Cord Mesenchymal Stem Cells Inhibit the Function of Allogeneic Activated Vγ9Vδ2 T Lymphocytes In Vitro. BIOMED RESEARCH INTERNATIONAL 2015; 2015:317801. [PMID: 25984529 PMCID: PMC4423519 DOI: 10.1155/2015/317801] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 03/21/2015] [Accepted: 03/22/2015] [Indexed: 01/01/2023]
Abstract
Background. Human umbilical cord mesenchymal stem cells (UC-MSCs) can regulate the function of immune cells. However, whether and how UC-MSCs can modulate the function of Vγ9Vδ2 T cells has not been fully understood. Methods. The PBMCs or Vγ9Vδ2 T cells were activated and expanded with pamidronate (PAM) and interleukin-2 (IL-2) with or without the presence UC-MSCs. The effects of UC-MSCs on the proliferation, cytokine expression, and cytotoxicity of Vγ9Vδ2 T cells were determined by flow cytometry. The effects of UC-MSCs on Fas-L, TRAIL-expressing Vγ9Vδ2 T cells, and Vγ9Vδ2 T cell apoptosis were determined by flow cytometry. Results. UC-MSCs inhibited Vγ9Vδ2 T cell proliferation in a dose-dependent but cell-contact independent manner. Coculture with UC-MSCs reduced the frequency of IFNγ+ but increased granzyme B+ Vγ9Vδ2 T cells. UC-MSCs inhibited the cytotoxicity of Vγ9Vδ2 T cells against influenza virus H1N1 infected A549 cells and also reduced the frequency of Fas-L+, TRAIL+ Vγ9Vδ2 T cells but failed to modulate the apoptosis of Vγ9Vδ2 T cells. Conclusions. These results indicated that UC-MSCs efficiently suppressed the proliferation and cytotoxicity of Vγ9Vδ2 T cells and modulated their cytokine production. Fas-L and TRAIL were involved in the regulation. Cell contact and apoptosis of Vγ9Vδ2 T cells were not necessary for the inhibition.
Collapse
|
746
|
Li CY, Wu XY, Tong JB, Yang XX, Zhao JL, Zheng QF, Zhao GB, Ma ZJ. Comparative analysis of human mesenchymal stem cells from bone marrow and adipose tissue under xeno-free conditions for cell therapy. Stem Cell Res Ther 2015; 6:55. [PMID: 25884704 PMCID: PMC4453294 DOI: 10.1186/s13287-015-0066-5] [Citation(s) in RCA: 293] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2014] [Revised: 11/24/2014] [Accepted: 03/25/2015] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION Mesenchymal stem cells (MSCs) are promising candidates for cell-based therapies. Human platelet lysate represents an efficient alternative to fetal bovine serum for clinical-scale expansion of MSCs. Different media used in culture processes should maintain the biological characteristics of MSCs during multiple passages. However, bone marrow-derived MSCs and adipose tissue-derived MSCs have not yet been directly compared with each other under human platelet lysate conditions. This study aims to conduct a direct head-to-head comparison of the biological characteristics of the two types of MSCs under human platelet lysate-supplemented culture conditions for their ability to be used in regenerative medicine applications. METHODS The bone marrow- and adipose tissue-derived MSCs were cultured under human platelet lysate conditions and their biological characteristics evaluated for cell therapy (morphology, immunophenotype, colony-forming unit-fibroblast efficiency, proliferation capacity, potential for mesodermal differentiation, secreted proteins, and immunomodulatory effects). RESULTS Under human platelet lysate-supplemented culture conditions, bone marrow- and adipose tissue-derived MSCs exhibited similar fibroblast-like morphology and expression patterns of surface markers. Adipose tissue-derived MSCs had greater proliferative potential than bone marrow-derived MSCs, while no significantly difference in colony efficiency were observed between the two types of cells. However, bone marrow-derived MSCs possessed higher capacity toward osteogenic and chondrogenic differentiation compared with adipose tissue-derived MSCs, while similar adipogenic differentiation potential wase observed between the two types of cells. There were some differences between bone marrow- and adipose tissue-derived MSCs for several secreted proteins, such as cytokine (interferon-γ), growth factors (basic fibroblast growth factor, hepatocyte growth factor, and insulin-like growth factor-1), and chemokine (stem cell-derived factor-1). Adipose tissue-derived MSCs had more potent immunomodulatory effects than bone marrow-derived MSCs. CONCLUSIONS Adipose tissue-derived MSCs have biological advantages in the proliferative capacity, secreted proteins (basic fibroblast growth factor, interferon-γ, and insulin-like growth factor-1), and immunomodulatory effects, but bone marrow-derived MSCs have advantages in osteogenic and chondrogenic differentiation potential and secreted proteins (stem cell-derived factor-1 and hepatocyte growth factor); these biological advantages should be considered systematically when choosing the MSC source for specific clinical application.
Collapse
Affiliation(s)
- Chun-yu Li
- China Military Institute of Chinese Medicine, 302 Military Hospital, Beijing, 100039, China. .,School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 610000, China.
| | - Xiao-yun Wu
- Beijing Institute of Life Science Translational Medicine Research Center, Beijing, 100085, China.
| | - Jia-bei Tong
- Shandong Medicinal Biotechnology Centre, Shandong Academy of Medical Sciences, Jinan, 250000, China.
| | - Xin-xin Yang
- School of Pharmacy, Changchun University of Traditional Chinese Medicine, Changsha, 410208, China.
| | - Jing-li Zhao
- Jilin Vocational College of Industry and Technology, Jilin, 132013, China.
| | - Quan-fu Zheng
- China Military Institute of Chinese Medicine, 302 Military Hospital, Beijing, 100039, China. .,School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 610000, China.
| | - Guo-bin Zhao
- The First Affiliated Hospital, Hebei North University, Zhangjiakou, 075000, China.
| | - Zhi-jie Ma
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China.
| |
Collapse
|
747
|
Thakkar UG, Trivedi HL, Vanikar AV, Dave SD. Insulin-secreting adipose-derived mesenchymal stromal cells with bone marrow-derived hematopoietic stem cells from autologous and allogenic sources for type 1 diabetes mellitus. Cytotherapy 2015; 17:940-7. [PMID: 25869301 DOI: 10.1016/j.jcyt.2015.03.608] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 02/24/2015] [Indexed: 01/24/2023]
Abstract
BACKGROUND AIMS Stem cell therapy (SCT) is now the up-coming therapeutic modality for treatment of type 1 diabetes mellitus (T1DM). METHODS Our study was a prospective, open-labeled, two-armed trial for 10 T1DM patients in each arm of allogenic and autologous adipose-derived insulin-secreting mesenchymal stromal cells (IS-AD-MSC)+bone marrow-derived hematopoietic stem cell (BM-HSC) infusion. Group 1 received autologous SCT: nine male patients and one female patient; mean age, 20.2 years, disease duration 8.1 years; group 2 received allogenic SCT: six male patients and four female patients, mean age, 19.7 years and disease duration, 7.9 years. Glycosylated hemoglobin (HbA1c) was 10.99%; serum (S.) C-peptide, 0.22 ng/mL and insulin requirement, 63.9 IU/day in group 1; HbA1c was 11.93%, S.C-peptide, 0.028 ng/mL and insulin requirement, 57.55 IU/day in group 2. SCs were infused into the portal+thymic circulation and subcutaneous tissue under non-myelo-ablative conditioning. Patients were monitored for blood sugar, S.C-peptide, glutamic acid decarboxylase antibodies and HbA1c at 3-month intervals. RESULTS Group 1 received mean SCs 103.14 mL with 2.65 ± 0.8 × 10(4) ISCs/kg body wt, CD34+ 0.81% and CD45-/90+/73+, 81.55%. Group 2 received mean SCs 95.33 mL with 2.07 ± 0.67 × 10(4) ISCs/kg body wt, CD34+ 0.32% and CD45-/90+/73+ 54.04%. No untoward effect was observed with sustained improvement in HbA1c and S.C-peptide in both groups with a decrease in glutamic acid decarboxylase antibodies and reduction in mean insulin requirement. CONCLUSIONS SCT is a safe and viable treatment option for T1DM. Autologous IS-AD-MSC+ BM-HSC co-infusion offers better long-term control of hyperglycemia as compared with allogenic SCT.
Collapse
Affiliation(s)
- Umang G Thakkar
- Department of Regenerative Medicine and Stem Cell Therapy, G.R. Doshi and K.M. Mehta Institute of Kidney Diseases & Research Centre, Dr H.L. Trivedi Institute of Transplantation Sciences, Gujarat, India.
| | - Hargovind L Trivedi
- Department of Regenerative Medicine and Stem Cell Therapy, G.R. Doshi and K.M. Mehta Institute of Kidney Diseases & Research Centre, Dr H.L. Trivedi Institute of Transplantation Sciences, Gujarat, India; Department of Nephrology and Transplantation Medicine, G.R. Doshi and K.M. Mehta Institute of Kidney Diseases & Research Centre, Dr H.L. Trivedi Institute of Transplantation Sciences, Gujarat, India
| | - Aruna V Vanikar
- Department of Regenerative Medicine and Stem Cell Therapy, G.R. Doshi and K.M. Mehta Institute of Kidney Diseases & Research Centre, Dr H.L. Trivedi Institute of Transplantation Sciences, Gujarat, India; Department of Pathology, Laboratory Medicine, Transfusion Services and Immunohematology, G.R. Doshi and K.M. Mehta Institute of Kidney Diseases & Research Centre, Dr H.L. Trivedi Institute of Transplantation Sciences, Gujarat, India
| | - Shruti D Dave
- Department of Pathology, Laboratory Medicine, Transfusion Services and Immunohematology, G.R. Doshi and K.M. Mehta Institute of Kidney Diseases & Research Centre, Dr H.L. Trivedi Institute of Transplantation Sciences, Gujarat, India
| |
Collapse
|
748
|
Abstract
Tendon injuries are common and present a clinical challenge to orthopedic surgery mainly because these injuries often respond poorly to treatment and require prolonged rehabilitation. Therapeutic options used to repair ruptured tendons have consisted of suture, autografts, allografts, and synthetic prostheses. To date, none of these alternatives has provided a successful long-term solution, and often the restored tendons do not recover their complete strength and functionality. Unfortunately, our understanding of tendon biology lags far behind that of other musculoskeletal tissues, thus impeding the development of new treatment options for tendon conditions. Hence, in this review, after introducing the clinical significance of tendon diseases and the present understanding of tendon biology, we describe and critically assess the current strategies for enhancing tendon repair by biological means. These consist mainly of applying growth factors, stem cells, natural biomaterials and genes, alone or in combination, to the site of tendon damage. A deeper understanding of how tendon tissue and cells operate, combined with practical applications of modern molecular and cellular tools could provide the long awaited breakthrough in designing effective tendon-specific therapeutics and overall improvement of tendon disease management.
Collapse
|
749
|
Minteer DM, Marra KG, Rubin JP. Adipose stem cells: biology, safety, regulation, and regenerative potential. Clin Plast Surg 2015; 42:169-79. [PMID: 25827561 DOI: 10.1016/j.cps.2014.12.007] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
This article discusses adipose-derived stem cell (ASC) biology, describes the current knowledge in the literature for the safety and regulation of ASCs, and provides a brief overview of the regenerative potential of ASCs. It is not an exhaustive listing of all available clinical studies or every study applying ASCs in tissue engineering and regenerative medicine, but is an objective commentary of these topics.
Collapse
Affiliation(s)
- Danielle M Minteer
- Department of Bioengineering, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Kacey G Marra
- Department of Bioengineering, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA; Department of Plastic Surgery, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, PA 15213, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA 15213, USA
| | - J Peter Rubin
- Department of Bioengineering, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA; Department of Plastic Surgery, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, PA 15213, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA 15213, USA.
| |
Collapse
|
750
|
Lomas A, Ryan C, Sorushanova A, Shologu N, Sideri A, Tsioli V, Fthenakis G, Tzora A, Skoufos I, Quinlan L, O'Laighin G, Mullen A, Kelly J, Kearns S, Biggs M, Pandit A, Zeugolis D. The past, present and future in scaffold-based tendon treatments. Adv Drug Deliv Rev 2015; 84:257-77. [PMID: 25499820 DOI: 10.1016/j.addr.2014.11.022] [Citation(s) in RCA: 150] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 11/08/2014] [Accepted: 11/12/2014] [Indexed: 02/07/2023]
Abstract
Tendon injuries represent a significant clinical burden on healthcare systems worldwide. As the human population ages and the life expectancy increases, tendon injuries will become more prevalent, especially among young individuals with long life ahead of them. Advancements in engineering, chemistry and biology have made available an array of three-dimensional scaffold-based intervention strategies, natural or synthetic in origin. Further, functionalisation strategies, based on biophysical, biochemical and biological cues, offer control over cellular functions; localisation and sustained release of therapeutics/biologics; and the ability to positively interact with the host to promote repair and regeneration. Herein, we critically discuss current therapies and emerging technologies that aim to transform tendon treatments in the years to come.
Collapse
|