1
|
Qiu Z, Cai W, Liu Q, Liu K, Liu C, Yang H, Huang R, Li P, Zhao Q. Unravelling novel and pleiotropic genes for cannon bone circumference and bone mineral density in Yorkshire pigs. J Anim Sci 2024; 102:skae036. [PMID: 38330300 PMCID: PMC10914368 DOI: 10.1093/jas/skae036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 02/03/2024] [Indexed: 02/10/2024] Open
Abstract
Leg weakness is a prevalent health condition in pig farms. The augmentation of cannon bone circumference and bone mineral density can effectively improve limb strength in pigs and alleviate leg weakness. This study measured forelimb cannon bone circumference (fCBC) and rear limb cannon bone circumference (rCBC) using an inelastic tapeline and rear limb metatarsal area bone mineral density (raBMD) using a dual-energy X-ray absorptiometry bone density scanner. The samples of Yorkshire castrated boars were genotyped using a 50K single-nucleotide polymorphism (SNP) array. The SNP-chip data were imputed to the level of whole-genome sequencing data (iWGS). This study used iWGS data to perform genome-wide association studies and identified novel significant SNPs associated with fCBC on SSC6, SSC12, and SSC13, rCBC on SSC12 and SSC14, and raBMD on SSC7. Based on the high phenotypic and genetic correlations between CBC and raBMD, multi-trait meta-analysis was performed to identify pleiotropic SNPs. A significant potential pleiotropic quantitative trait locus (QTL) regulating both CBC and raBMD was identified on SSC15. Bayes fine mapping was used to establish the confidence intervals for these novel QTLs with the most refined confidence interval narrowed down to 56 kb (15.11 to 15.17 Mb on SSC12 for fCBC). Furthermore, the confidence interval for the potential pleiotropic QTL on SSC15 in the meta-analysis was narrowed down to 7.45 kb (137.55 to137.56 Mb on SSC15). Based on the biological functions of genes, the following genes were identified as novel regulatory candidates for different phenotypes: DDX42, MYSM1, FTSJ3, and MECOM for fCBC; SMURF2, and STC1 for rCBC; RGMA for raBMD. Additionally, RAMP1, which was determined to be located 23.68 kb upstream of the confidence interval of the QTL on SSC15 in the meta-analysis, was identified as a potential pleiotropic candidate gene regulating both CBC and raBMD. These findings offered valuable insights for identifying pathogenic genes and elucidating the genetic mechanisms underlying CBC and BMD.
Collapse
Affiliation(s)
- Zijian Qiu
- Key Laboratory in Nanjing for Evaluation and Utilization of Pigs Resources, Ministry of Agriculture and Rural Areas of China, Institute of Swine Science, Nanjing Agricultural University, Nanjing 210095, China
| | - Wenwu Cai
- Key Laboratory in Nanjing for Evaluation and Utilization of Pigs Resources, Ministry of Agriculture and Rural Areas of China, Institute of Swine Science, Nanjing Agricultural University, Nanjing 210095, China
| | - Qian Liu
- Key Laboratory in Nanjing for Evaluation and Utilization of Pigs Resources, Ministry of Agriculture and Rural Areas of China, Institute of Swine Science, Nanjing Agricultural University, Nanjing 210095, China
| | - Kaiyue Liu
- Key Laboratory in Nanjing for Evaluation and Utilization of Pigs Resources, Ministry of Agriculture and Rural Areas of China, Institute of Swine Science, Nanjing Agricultural University, Nanjing 210095, China
| | - Chenxi Liu
- Key Laboratory in Nanjing for Evaluation and Utilization of Pigs Resources, Ministry of Agriculture and Rural Areas of China, Institute of Swine Science, Nanjing Agricultural University, Nanjing 210095, China
| | - Huilong Yang
- Key Laboratory in Nanjing for Evaluation and Utilization of Pigs Resources, Ministry of Agriculture and Rural Areas of China, Institute of Swine Science, Nanjing Agricultural University, Nanjing 210095, China
| | - Ruihua Huang
- Key Laboratory in Nanjing for Evaluation and Utilization of Pigs Resources, Ministry of Agriculture and Rural Areas of China, Institute of Swine Science, Nanjing Agricultural University, Nanjing 210095, China
- Huaian Academy, Nanjing Agricultural University, Huaian 223005, China
| | - Pinghua Li
- Key Laboratory in Nanjing for Evaluation and Utilization of Pigs Resources, Ministry of Agriculture and Rural Areas of China, Institute of Swine Science, Nanjing Agricultural University, Nanjing 210095, China
- Huaian Academy, Nanjing Agricultural University, Huaian 223005, China
| | - Qingbo Zhao
- Key Laboratory in Nanjing for Evaluation and Utilization of Pigs Resources, Ministry of Agriculture and Rural Areas of China, Institute of Swine Science, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
2
|
Caruso M, Shuttle S, Amelse L, Elkhenany H, Schumacher J, Dhar MS. A pilot study to demonstrate the paracrine effect of equine, adult allogenic mesenchymal stem cells in vitro, with a potential for healing of experimentally-created, equine thoracic wounds in vivo. Front Vet Sci 2022; 9:1011905. [PMID: 36452146 PMCID: PMC9702339 DOI: 10.3389/fvets.2022.1011905] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 10/18/2022] [Indexed: 10/18/2023] Open
Abstract
Regenerative biological therapies using mesenchymal stem cells (MSCs) are being studied and used extensively in equine veterinary medicine. One of the important properties of MSCs is the cells' reparative effect, which is brought about by paracrine signaling, which results in the release of biologically active molecules, which in turn, can affect cellular migration and proliferation, thus a huge potential in wound healing. The objective of the current study was to demonstrate the in vitro and in vivo potentials of equine allogenic bone marrow-derived MSCs for wound healing. Equine bone marrow-derived MSCs from one allogenic donor horse were used. Equine MSCs were previously characterized for their in vitro proliferation, expression of cluster-of-differentiation markers, and trilineage differentiation. MSCs were first evaluated for their migration using an in vitro wound healing scratch assay, and subsequently, the conditioned medium was evaluated for their effect on human fibroblast proliferation. Subsequently, allogenic cells were intradermally injected into full-thickness, cutaneous thoracic wounds of 4 horses. Wound healing was assessed by using 3-D digital imaging and by measuring mRNA expression of pro-and anti-inflammatory markers for 30 days. Using human fibroblasts in an in vitro wound healing assay, we demonstrate a significantly higher healing in the presence of conditioned medium collected from proliferating MSCs than in the presence of medium containing fetal bovine serum. The in vitro effect of MSCs did not translate into a detectable effect in vivo. Nonetheless, we proved that molecularly characterized equine allogenic MSCs do not illicit an immunologic response. Investigations using MSCs derived from other sources (adipose tissue, umbilical cord), or a higher number of MSCs or a compromised animal model may be required to prove the efficacy of equine MSCs in wound healing in vivo.
Collapse
Affiliation(s)
- Michael Caruso
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Knoxville, TN, United States
| | - Shannon Shuttle
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Knoxville, TN, United States
| | - Lisa Amelse
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Knoxville, TN, United States
| | - Hoda Elkhenany
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Knoxville, TN, United States
- Department of Surgery, Faculty of Veterinary Medicine, Alexandria University, Alexandria, Egypt
| | - James Schumacher
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Knoxville, TN, United States
| | - Madhu S. Dhar
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Knoxville, TN, United States
- Department of Surgery, Faculty of Veterinary Medicine, Alexandria University, Alexandria, Egypt
| |
Collapse
|
3
|
Lv X, Wang L, Zou X, Huang S. Umbilical Cord Mesenchymal Stem Cell Therapy for Regenerative Treatment of Rheumatoid Arthritis: Opportunities and Challenges. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:3927-3936. [PMID: 34584402 PMCID: PMC8462093 DOI: 10.2147/dddt.s323107] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 08/26/2021] [Indexed: 12/25/2022]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease of unknown etiology with a high rate of disability. Traditional treatments for RA remain a challenging issue. For example, nonsteroidal anti-inflammatory drugs (NSAIDs) have no therapeutic effects on joint destruction, and the prominent side effects include gastrointestinal symptoms. RA is characterized by recurrence and bone attrition. Therefore, regenerative medicine and the use of umbilical cord mesenchymal stem cell (UC-MSC) therapies have recently emerged as potential options. UC-MSCs are multifunctional stem cells that are present in neonatal umbilical cord tissue and can differentiate into many kinds of cells, which have broad clinical application prospects in the tissue engineering of bone, cartilage, muscle, tendon, ligament, nerve, liver, endothelium, and myocardium. Moreover, UC-MSCs have advantages, such as convenient collection of materials and no ethical disputes; thus, these cells have attracted increasing attention from researchers. However, there are few clinical studies regarding UC-MSC therapy for RA. In this paper, we will review traditional drugs for RA treatment and then focus on UC-MSC therapy for RA, including preclinical and clinical UC-MSC applications for RA patients in the context of regenerative medicine. Finally, we will summarize the challenges and perspectives of UC-MSCs as a potential therapeutic strategy for RA. This review will help to design and discover more potent and efficacious treatments for RA patients and aid in advancing this class of cell therapy.
Collapse
Affiliation(s)
- Xiaolan Lv
- Department of Laboratory Medicine, Liuzhou Maternity and Child Healthcare Hospital, Liu Zhou, Guang Xi, People's Republic of China
| | - Liming Wang
- Shaanxi Jiuzhou Biomedical Science and Technology Group, Xi'an, Shaan Xi, People's Republic of China
| | - XiaoRong Zou
- Department of Hematology, 986 Hospital of Fourth Military Medical University, Xi'an, Shaan Xi, People's Republic of China
| | - Shigao Huang
- Faculty of Health Sciences, University of Macau, Macau, People's Republic of China
| |
Collapse
|
4
|
Kang MH, Park HM. Challenges of stem cell therapies in companion animal practice. J Vet Sci 2020; 21:e42. [PMID: 32476316 PMCID: PMC7263915 DOI: 10.4142/jvs.2020.21.e42] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 03/15/2020] [Accepted: 03/17/2020] [Indexed: 12/13/2022] Open
Abstract
Regenerative medicine using stem cells from various sources are emerging treatment modality in several refractory diseases in veterinary medicine. It is well-known that stem cells can differentiate into specific cell types, self-renew, and regenerate. In addition, the unique immunomodulatory effects of stem cells have made stem cell transplantation a promising option for treating a wide range of disease and injuries. Recently, the medical demands for companion animals have been rapidly increasing, and certain disease conditions require alternative treatment options. In this review, we focused on stem cell application research in companion animals including experimental models, case reports and clinical trials in dogs and cats. The clinical studies and therapeutic protocols were categorized, evaluated and summarized according to the organ systems involved. The results indicate that evidence for the effectiveness of cell-based treatment in specific diseases or organ systems is not yet conclusive. Nonetheless, stem cell therapy may be a realistic treatment option in the near future, therefore, considerable efforts are needed to find optimized cell sources, cell numbers and delivery methods in order to standardize treatment methods and evaluation processes.
Collapse
Affiliation(s)
- Min Hee Kang
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea
| | - Hee Myung Park
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea.
| |
Collapse
|
5
|
Amini-Nik S, Dolp R, Eylert G, Datu AK, Parousis A, Blakeley C, Jeschke MG. Stem cells derived from burned skin - The future of burn care. EBioMedicine 2018; 37:509-520. [PMID: 30409728 PMCID: PMC6284415 DOI: 10.1016/j.ebiom.2018.10.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 10/03/2018] [Accepted: 10/04/2018] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Thermal injuries affect millions of adults and children worldwide and are associated with high morbidity and mortality. The key determinant for the survival of burns is rapid wound healing. Large wounds exceed intrinsic wound-healing capacities, and the currently available coverage materials are insufficient due to lack of cellularity, availability or immunological rejection. METHODS Using the surgically debrided tissue, we isolated viable cells from burned skin. The isolated cells cultured in tissue culture dishes and characterized. FINDINGS We report here that debrided burned skin, which is routinely excised from patients and otherwise considered medical waste and unconsciously discarded, contains viable, undamaged cells which show characteristics of mesenchymal skin stem cells. Those cells can be extracted, characterized, expanded, and incorporated into created epidermal-dermal substitutes to promote wound healing in immune-compromised mice and Yorkshire pigs without adverse side effects. INTERPRETATION These findings are of paramount importance and provide an ideal cell source for autologous skin regeneration. Furthermore, this study highlights that skin contains progenitor cells resistant to thermal stress. FUND: Canadian Institutes of Health Research # 123336. CFI Leader's Opportunity Fund: Project # 25407 National Institutes of Health 2R01GM087285-05A1. EMHSeed: Fund: 500463, A generous donation from Toronto Hydro. Integra© Life Science Company provided the meshed bilayer Integra© for porcine experiments.
Collapse
Affiliation(s)
- Saeid Amini-Nik
- Sunnybrook Research Institute, Canada; Department of Laboratory Medicine and Pathobiology (LMP), University of Toronto, Canada; Division of Plastic and Reconstructive Surgery, Department of Surgery, Faculty of Medicine, University of Toronto, Canada.
| | - Reinhard Dolp
- Sunnybrook Research Institute, Canada; Institute of Medical Science, University of Toronto, Canada
| | - Gertraud Eylert
- Sunnybrook Research Institute, Canada; Institute of Medical Science, University of Toronto, Canada
| | | | | | | | - Marc G Jeschke
- Sunnybrook Research Institute, Canada; Institute of Medical Science, University of Toronto, Canada; Ross Tilley Burn Centre, Sunnybrook Health Sciences Centre, Canada; Division of Plastic and Reconstructive Surgery, Department of Surgery, Faculty of Medicine, University of Toronto, Canada.
| |
Collapse
|
6
|
Gadelkarim M, Abushouk AI, Ghanem E, Hamaad AM, Saad AM, Abdel-Daim MM. Adipose-derived stem cells: Effectiveness and advances in delivery in diabetic wound healing. Biomed Pharmacother 2018; 107:625-633. [DOI: 10.1016/j.biopha.2018.08.013] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 07/31/2018] [Accepted: 08/03/2018] [Indexed: 12/20/2022] Open
|
7
|
Yeo GC, Kosobrodova E, Kondyurin A, McKenzie DR, Bilek MM, Weiss AS. Plasma‐Activated Substrate with a Tropoelastin Anchor for the Maintenance and Delivery of Multipotent Adult Progenitor Cells. Macromol Biosci 2018; 19:e1800233. [DOI: 10.1002/mabi.201800233] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 08/19/2018] [Indexed: 01/05/2023]
Affiliation(s)
- Giselle C. Yeo
- Charles Perkins CentreUniversity of Sydney NSW 2006 Australia
- School of Life and Environmental SciencesUniversity of Sydney NSW 2006 Australia
- Bosch InstituteUniversity of Sydney NSW 2006 Australia
- The Cooperative Research Centre for Cell Therapy ManufacturingUniversity of South Australia City West Campus Adelaide SA 5000 Australia
| | - Elena Kosobrodova
- School of PhysicsUniversity of Sydney NSW 2006 Australia
- School of AerospaceMechanical and Mechatronic EngineeringUniversity of Sydney NSW 2006 Australia
- The Cooperative Research Centre for Cell Therapy ManufacturingUniversity of South Australia City West Campus Adelaide SA 5000 Australia
| | - Alexey Kondyurin
- School of PhysicsUniversity of Sydney NSW 2006 Australia
- The Cooperative Research Centre for Cell Therapy ManufacturingUniversity of South Australia City West Campus Adelaide SA 5000 Australia
| | - David R. McKenzie
- School of PhysicsUniversity of Sydney NSW 2006 Australia
- The Cooperative Research Centre for Cell Therapy ManufacturingUniversity of South Australia City West Campus Adelaide SA 5000 Australia
| | - Marcela M. Bilek
- Charles Perkins CentreUniversity of Sydney NSW 2006 Australia
- School of PhysicsUniversity of Sydney NSW 2006 Australia
- School of AerospaceMechanical and Mechatronic EngineeringUniversity of Sydney NSW 2006 Australia
- Australian Institute of Nanoscale Science and TechnologyUniversity of Sydney NSW 2006 Australia
- The Cooperative Research Centre for Cell Therapy ManufacturingUniversity of South Australia City West Campus Adelaide SA 5000 Australia
| | - Anthony S. Weiss
- Charles Perkins CentreUniversity of Sydney NSW 2006 Australia
- School of Life and Environmental SciencesUniversity of Sydney NSW 2006 Australia
- Bosch InstituteUniversity of Sydney NSW 2006 Australia
- The Cooperative Research Centre for Cell Therapy ManufacturingUniversity of South Australia City West Campus Adelaide SA 5000 Australia
| |
Collapse
|
8
|
Al-Jaibaji O, Swioklo S, Gijbels K, Vaes B, Figueiredo FC, Connon CJ. Alginate encapsulated multipotent adult progenitor cells promote corneal stromal cell activation via release of soluble factors. PLoS One 2018; 13:e0202118. [PMID: 30192833 PMCID: PMC6128465 DOI: 10.1371/journal.pone.0202118] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 07/28/2018] [Indexed: 01/26/2023] Open
Abstract
To reduce the increasing need for corneal transplantation, attempts are currently aiming to restore corneal clarity, one potent source of cells are multipotent adult progenitor cells (MAPC®). These cells release a powerful cocktail of paracrine factors that can guide wound healing and tissue regeneration. However, their role in corneal regeneration has been overlooked. Thus, we sought to explore the potential of combining the cytoprotective storage feature of alginate, with MAPC to generate a storable cell-laden gel for corneal wound healing. 72 hours following hypothermic storage, alginate encapsulation was shown to maintain MAPC viability at either 4 or 15°C. Encapsulated MAPC (2 x106 cells/mL) stored at 15°C presented the optimum temperature that allowed for cell recovery. These cells had the ability to reattach to tissue culture plastic whilst exhibiting normal phenotype and this was maintained in serum-free and xenobiotic-free medium. Furthermore, corneal stromal cells presented a significant decrease in scratch-wounds in the presence of alginate encapsulated MAPC compared to a no-cell control (p = 0.018). This study shows that immobilization of MAPC within an alginate hydrogel does not hinder their ability to affect a secondary cell population via soluble factors and that these effects are successfully retained following hypothermic storage.
Collapse
Affiliation(s)
- Olla Al-Jaibaji
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Stephen Swioklo
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | | | | | | | - Che J. Connon
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
- * E-mail:
| |
Collapse
|
9
|
Mesenchymal Stromal Cell Therapy for Pancreatitis: A Systematic Review. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:3250864. [PMID: 29743979 PMCID: PMC5878867 DOI: 10.1155/2018/3250864] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 12/31/2017] [Indexed: 12/19/2022]
Abstract
Background Based on animal studies, adult mesenchymal stromal cells (MSCs) are promising for the treatment of pancreatitis. However, the best type of this form of cell therapy and its mechanism of action remain unclear. Methods We searched the PubMed, Web of Science, Scopus, Google Scholar, and Clinical Trials.gov websites for studies using MSCs as a therapy for both acute and chronic pancreatitis published until September 2017. Results We identified 276 publications; of these publications, 18 met our inclusion criteria. In animal studies, stem cell therapy was applied more frequently for acute pancreatitis than for chronic pancreatitis. No clinical trials were identified. MSC therapy ameliorated pancreatic inflammation in acute pancreatitis and pancreatic fibrosis in chronic pancreatitis. Bone marrow and umbilical cord MSCs were the most frequently administered cell types. Due to the substantial heterogeneity among the studies regarding the type, source, and dose of MSCs used, conducting a meta-analysis was not feasible to determine the best type of MSCs. Conclusion The available data were insufficient for determining the best type of MSCs for the treatment of acute or chronic pancreatitis; therefore, clinical trials investigating the use of MSCs as therapy for pancreatitis are not warranted.
Collapse
|
10
|
Lee JH, Jeong JK, Park SY. AMPK Activation Mediated by Hinokitiol Inhibits Adipogenic Differentiation of Mesenchymal Stem Cells through Autophagy Flux. Int J Endocrinol 2018; 2018:2014192. [PMID: 30123258 PMCID: PMC6079415 DOI: 10.1155/2018/2014192] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 05/27/2018] [Accepted: 06/03/2018] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND AND PURPOSE Hinokitiol, a natural monopenoid present in the essential oil of Calocedrus formosana heartwood, exerts potent anticancer, anti-inflammatory, antibacterial, and neuroprotective effects on various cells. However, the antiobesity effect of hinokitiol on adipocytes is unclear. EXPERIMENTAL APPROACH In this study, we observed that hinokitiol affected the differentiation to adipocytes in mesenchymal stem cells (MSCs). Hinokitiol was treated with 3-isobutyl-1-methylxanthine, insulin, and dexamethasone to induce differentiation and maturing adipocytes in cultured MSCs. KEY RESULTS Hinokitiol treatment of MSCs decreased their differentiation to mature adipocytes and increased AMPK phosphorylation in a concentration-dependent manner. Moreover, we confirmed that the antiadipogenic effect of hinokitiol was associated with autophagy. The levels of LC3-II decreased and those of p62 increased in hinokitiol-treated MSCs. The treatment of hinokitiol-treated MSCs with the autophagy activator, rapamycin, restored the hinokitiol-induced decrease in the adipocyte differentiation of MSCs. The inhibition of AMPK phosphorylation also suppressed hinokitiol-mediated inhibition of autophagy and antiadipogenic effects. CONCLUSIONS AND IMPLICATIONS Taken together, these results indicated that AMPK activation and autophagy flux inhibition mediated by hinokitiol inhibited lipid accumulation and differentiation of MSCs to adipocytes and also suggest that differentiation of mesenchymal stem cells may be regulated by using the modulator of autophagy flux and AMPK signals including hinokitiol.
Collapse
Affiliation(s)
- Ju-Hee Lee
- Biosafety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, Jeonbuk 54596, Republic of Korea
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, 88 Dongnae-ro, Dong-gu, Daegu City 41061, Republic of Korea
| | - Jae-Kyo Jeong
- Biosafety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, Jeonbuk 54596, Republic of Korea
| | - Sang-Youel Park
- Biosafety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, Jeonbuk 54596, Republic of Korea
| |
Collapse
|
11
|
Xu J, Zgheib C, Hodges MM, Caskey RC, Hu J, Liechty KW. Mesenchymal stem cells correct impaired diabetic wound healing by decreasing ECM proteolysis. Physiol Genomics 2017; 49:541-548. [PMID: 28842435 DOI: 10.1152/physiolgenomics.00090.2016] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 08/22/2017] [Accepted: 08/22/2017] [Indexed: 12/26/2022] Open
Abstract
Impaired diabetic wound healing is associated with a dermal extracellular matrix protein profile favoring proteolysis; within the healing diabetic wound, this is represented by an increase in activated matrix metalloproteinase (MMPs). Treatment of diabetic wounds with mesenchymal stem cells (MSCs) has been shown to improve wound healing; however, there has not yet been an assessment of their ability to correct dysregulation of MMPs in diabetic wounds. Furthermore, there has been no prior assessment of the role of microRNA29b (miR-29b), an inhibitory regulatory molecule that targets MMP-9 mRNA. Using in vitro models of fibroblast coculture with MSCs and in vivo murine wound healing models, we tested the hypothesis that MSCs correct dysregulation of MMPs in a microRNA-29b-dependent mechanism. In this study, we first demonstrated that collagen I and III protein content is significantly reduced in diabetic wounds, and treatment with MSCs significantly improves collagen I content in both nondiabetic and diabetic wounds. We then found that MMP-9 gene expression and protein content were significantly upregulated in diabetic wounds, indicating elevated proteolysis. Treatment with MSCs resulted in a decrease in MMP-9 gene expression and protein content level in diabetic wounds 3 and 7 days after wounding. Zymographic analysis indicated that MSC treatment also decreased the amount of activated MMP-9 present in diabetic wounds. Furthermore, miR-29b expression was inversely associated with MMP-9 gene expression; miR-29b expression was decreased in diabetic wounds and diabetic fibroblast. Following treatment of diabetic wounds with MSCs, as well as in diabetic fibroblasts cocultured with MSCs, miR-29b was significantly increased. These findings suggest a potential mechanism through which MSCs enhance diabetic wound healing by improving collagen I content in diabetic wounds through decreasing MMP-9 expression and increasing miR-29b expression.
Collapse
Affiliation(s)
- Junwang Xu
- Laboratory for Fetal and Regenerative Biology, University of Colorado Denver - Anschutz Medical Campus, University of Colorado School of Medicine, Aurora, Colorado.,Department of Surgery, Children's Hospital Colorado, Aurora, Colorado; and
| | - Carlos Zgheib
- Laboratory for Fetal and Regenerative Biology, University of Colorado Denver - Anschutz Medical Campus, University of Colorado School of Medicine, Aurora, Colorado.,Department of Surgery, Children's Hospital Colorado, Aurora, Colorado; and
| | - Maggie M Hodges
- Laboratory for Fetal and Regenerative Biology, University of Colorado Denver - Anschutz Medical Campus, University of Colorado School of Medicine, Aurora, Colorado.,Department of Surgery, Children's Hospital Colorado, Aurora, Colorado; and
| | - Robert C Caskey
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Junyi Hu
- Laboratory for Fetal and Regenerative Biology, University of Colorado Denver - Anschutz Medical Campus, University of Colorado School of Medicine, Aurora, Colorado.,Department of Surgery, Children's Hospital Colorado, Aurora, Colorado; and
| | - Kenneth W Liechty
- Laboratory for Fetal and Regenerative Biology, University of Colorado Denver - Anschutz Medical Campus, University of Colorado School of Medicine, Aurora, Colorado; .,Department of Surgery, Children's Hospital Colorado, Aurora, Colorado; and
| |
Collapse
|
12
|
Berry E, Liu Y, Chen L, Guo AM. Eicosanoids: Emerging contributors in stem cell-mediated wound healing. Prostaglandins Other Lipid Mediat 2016; 132:17-24. [PMID: 27825971 DOI: 10.1016/j.prostaglandins.2016.11.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 09/29/2016] [Accepted: 11/03/2016] [Indexed: 12/16/2022]
Abstract
Eicosanoids are bioactive lipid products primarily derived from the oxidation of arachidonic acid (AA). The individual contributions of eicosanoids and stem cells to wound healing have been of great interest. This review focuses on how stem cells work in concert with eicosanoids to create a beneficial environment in the wound bed and in the promotion of wound healing. Stem cells contribute to wound healing through modulating inflammation, differentiating into skin cells or endothelial cells, and exerting paracrine effects by releasing various potent growth factors. Eicosanoids have been shown to stimulate proliferation, migration, homing, and differentiation of stem cells, all of which contribute to the process of wound healing. Increasing evidence has shown that eicosanoids improve wound healing through increasing stem cell densities, stimulating differentiation, and enhancing the angiogenic properties of stem cells. Chronic wounds have become a major problem in health care. Therefore, research regarding the effects of stem cells and eicosanoids in the promotion wound healing is of great importance.
Collapse
Affiliation(s)
- Elizabeth Berry
- Department of Pharmacology, School of Medicine, New York Medical College, Valhalla, NY 10595 United States
| | - Yanzhou Liu
- Department of Pharmacology, School of Medicine, New York Medical College, Valhalla, NY 10595 United States; Department of Pharmacology, School of Medicine, Wuhan University, Wuhan, 430071, People's Republic of China
| | - Li Chen
- State Key Lab of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Austin M Guo
- Department of Pharmacology, School of Medicine, New York Medical College, Valhalla, NY 10595 United States; Department of Pharmacology, School of Medicine, Wuhan University, Wuhan, 430071, People's Republic of China.
| |
Collapse
|
13
|
Deubiquitinase MYSM1 Is Essential for Normal Bone Formation and Mesenchymal Stem Cell Differentiation. Sci Rep 2016; 6:22211. [PMID: 26915790 PMCID: PMC4768166 DOI: 10.1038/srep22211] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 02/09/2016] [Indexed: 12/14/2022] Open
Abstract
Deubiquitinase MYSM1 has been shown to play a critical role in hematopoietic cell differentiation and hematopoietic stem cell (HSC) maintenance. Mesenchymal stem cells (MSCs) are multipotent stromal cells within the bone marrow. MSCs are progenitors to osteoblasts, chondrocytes, adipocytes, and myocytes. Although, MSCs have been extensively studied, the roles of MYSM1 in these cells remain unclear. Here we describe the function of MYSM1 on MSC maintenance and differentiation. In this report, we found that Mysm1−/− mice had a lower bone mass both in long bone and calvaria compared with their control counterpart. Preosteoblasts from Mysm1−/− mice did not show changes in proliferation or osteogenesis when compared to WT mice. Conversely, Mysm1−/− MSCs showed enhanced autonomous differentiation and accelerated adipogenesis. Our results demonstrate that MYSM1 plays a critical role in MSC maintenance and differentiation. This study also underscores the biological significance of deubiquitinase activity in MSC function. Mysm1 may represent a potential therapeutic target for controlling MSC lineage differentiation, and possibly for the treatment of metabolic bone diseases such as osteoporosis.
Collapse
|
14
|
G-CSF Administration after the Intraosseous Infusion of Hypertonic Hydroxyethyl Starches Accelerating Wound Healing Combined with Hemorrhagic Shock. BIOMED RESEARCH INTERNATIONAL 2016; 2016:5317630. [PMID: 26989687 PMCID: PMC4773547 DOI: 10.1155/2016/5317630] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 01/13/2016] [Accepted: 01/21/2016] [Indexed: 12/04/2022]
Abstract
Objective. To evaluate the therapeutic effects of G-CSF administration after intraosseous (IO) resuscitation in hemorrhagic shock (HS) combined with cutaneous injury rats. Methods. The rats were randomly divided into four groups: (1) HS with resuscitation (blank), (2) HS with resuscitation + G-CSF (G-CSF, 200 μg/kg body weight, subcutaneous injection), (3) HS with resuscitation + normal saline solution injection (normal saline), and (4) HS + G-CSF injection without resuscitation (Unres/G-CSF). To estimate the treatment effects, the vital signs of alteration were first evaluated, and then wound closure rates and homing of MSCs and EPCs to the wound skins and vasculogenesis were measured. Besides, inflammation and vasculogenesis related mRNA expressions were also examined. Results. IO infusion hypertonic hydroxyethyl starch (HHES) exhibited beneficial volume expansion roles and G-CSF administration accelerated wound healing 3 days ahead of other groups under hemorrhagic shock. Circulating and the homing of MSCs and EPCs at wound skins were significantly elevated at 6 h after G-CSF treatment. Inflammation was declined since 3 d while angiogenesis was more obvious in G-CSF treated group on day 9. Conclusions. These results suggested that the synergistical application of HHES and G-CSF has life-saving effects and is beneficial for improving wound healing in HS combined with cutaneous injury rats.
Collapse
|
15
|
Stem Cells for Cutaneous Wound Healing. BIOMED RESEARCH INTERNATIONAL 2015; 2015:285869. [PMID: 26137471 PMCID: PMC4468276 DOI: 10.1155/2015/285869] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 03/20/2015] [Indexed: 01/08/2023]
Abstract
Optimum healing of a cutaneous wound involves a well-orchestrated cascade of biological and molecular processes involving cell migration, proliferation, extracellular matrix deposition, and remodelling. When the normal biological process fails for any reason, this healing process can stall resulting in chronic wounds. Wounds are a growing clinical burden on healthcare systems and with an aging population as well as increasing incidences of obesity and diabetes, this problem is set to increase. Cell therapies may be the solution. A range of cell based approaches have begun to cross the rift from bench to bedside and the supporting data suggests that the appropriate administration of stem cells can accelerate wound healing. This review examines the main cell types explored for cutaneous wound healing with a focus on clinical use. The literature overwhelmingly suggests that cell therapies can help to heal cutaneous wounds when used appropriately but we are at risk of clinical use outpacing the evidence. There is a need, now more than ever, for standardised methods of cell characterisation and delivery, as well as randomised clinical trials.
Collapse
|
16
|
Nishikori Y, Shiota N, Okunishi H. The role of mast cells in cutaneous wound healing in streptozotocin-induced diabetic mice. Arch Dermatol Res 2014; 306:823-35. [PMID: 25218083 DOI: 10.1007/s00403-014-1496-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 07/25/2014] [Accepted: 08/29/2014] [Indexed: 12/24/2022]
Abstract
Mast cells (MCs) reside in cutaneous tissue, and an increment of MCs is suggested to induce vascular regression in the process of wound healing. To clarify participation of MCs in diabetic cutaneous wound healing, we created an excisional wound on diabetic mice 4 weeks after streptozotocin injections and subsequently investigated the healing processes for 49 days, comparing them with control mice. The rate of wound closure was not markedly different between the diabetic and control mice. In the proliferative phase at days 7 and 14, neovascularization in the wound was weaker in diabetic mice than in control mice. In the remodeling phase at day 21 and afterward, rapid vascular regression occurred in control mice; however, neovascularization was still observed in diabetic mice where the number of vessels in granulation tissues was relatively higher than in control mice. In the remodeling phase of the control mice, MCs within the wound began to increase rapidly and resulted in considerable accumulation, whereas the increment of MCs was delayed in diabetic mice. In addition, the number of fibroblast growth factor (FGF)- or vascular endothelial growth factor (VEGF)-immunopositive hypertrophic fibroblast-like spindle cells and c-Kit-positive/VEGFR2-positive/FcεRIα-negative endothelial progenitor cells (EPCs) were higher in diabetic wounds. In conclusion, neovascularization in the proliferative phase and vascular regression in the remodeling phase were impaired in diabetic mice. The delayed increment of MCs and sustained angiogenic stimuli by fibroblast-like spindle cells and EPCs may inhibit vascular regression in the remodeling phase and impair the wound-healing process in diabetic mice.
Collapse
Affiliation(s)
- Yoriko Nishikori
- Department of Pharmacology, Shimane University School of Medicine, 89-1 Enya-cho, Izumo, Shimane, 693-8501, Japan,
| | | | | |
Collapse
|
17
|
Battiwalla M, Barrett AJ. Bone marrow mesenchymal stromal cells to treat complications following allogeneic stem cell transplantation. TISSUE ENGINEERING PART B-REVIEWS 2014; 20:211-7. [PMID: 24410434 DOI: 10.1089/ten.teb.2013.0566] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) is a technologically complicated procedure that represents the only cure for many hematologic malignancies. However, HSCT is often complicated by life-threatening toxicities related to the chemo-radiation conditioning regimen, poor engraftment of donor HSCs, the hyperinflammatory syndrome of graft-versus-host disease (GVHD), infection risks from immunosuppression, and end-organ damage. Bone marrow stromal cells (MSCs), also known as "mesenchymal stromal cells," not only play a nurturing role in the hematopoietic microenvironment but also can differentiate into other cell types of mesenchymal origin. MSCs are poorly immunogenic, and they can modulate immunological responses through interactions with a wide range of innate and adaptive immune cells to reduce inflammation. They are easily expanded ex vivo and after infusion, home to sites of injury and inflammation to promote tissue repair. Despite promising early trial results in HSCT with significant responses that have translated into survival benefits, there have been significant barriers to successful commercialization as an off-the-shelf therapy. Current efforts with MSCs in the HSCT setting are geared toward determining the factors determining potency, understanding the precise mechanisms of action in human HSCT, knowing their kinetics and fate, optimizing dose and schedule, incorporating biomarkers as response surrogates, addressing concerns about safety, optimizing clinical trial design, and negotiating the uncharted regulatory landscape for licensable cellular therapy.
Collapse
Affiliation(s)
- Minoo Battiwalla
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health , Bethesda, Maryland
| | | |
Collapse
|
18
|
Peng LH, Niu J, Zhang CZ, Yu W, Wu JH, Shan YH, Wang XR, Shen YQ, Mao ZW, Liang WQ, Gao JQ. TAT conjugated cationic noble metal nanoparticles for gene delivery to epidermal stem cells. Biomaterials 2014; 35:5605-18. [PMID: 24736021 DOI: 10.1016/j.biomaterials.2014.03.062] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Accepted: 03/21/2014] [Indexed: 01/30/2023]
Abstract
Most nonviral gene delivery systems are not efficient enough to manipulate the difficult-to-transfect cell types, including non-dividing, primary, neuronal or stem cells, due to a lack of an intrinsic capacity to enter the membrane and nucleus, release its DNA payload, and activate transcription. Noble metal nanoclusters have emerged as a fascinating area of widespread interest in nanomaterials. Herein, we report the synthesis of the TAT peptide conjugated cationic noble metal nanoparticles (metal NPs@PEI-TAT) as highly efficient carriers for gene delivery to stem cells. The metal NPs@PEI-TAT integrate the advantages of metal NPs and peptides: the presence of metal NPs can effectively decrease the cytotoxicity of cationic molecules, making it possible to apply them in biological systems, while the cell penetrating peptides are essential for enhanced cellular and nucleus entry to achieve high transfection efficiency. Our studies provide strong evidence that the metal NPs@PEI-TAT can be engineered as gene delivery agents for stem cells and subsequently enhance their directed differentiation for biomedical application.
Collapse
Affiliation(s)
- Li-Hua Peng
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Jie Niu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Chen-Zhen Zhang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Wei Yu
- Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, PR China
| | - Jia-He Wu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Ying-Hui Shan
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Xia-Rong Wang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - You-Qing Shen
- Center for Bionanoengineering and State Key Laboratory of Chemical Engineering, Zhejiang University, Hangzhou, PR China
| | - Zheng-Wei Mao
- Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, PR China.
| | - Wen-Quan Liang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Jian-Qing Gao
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China.
| |
Collapse
|
19
|
Rizzi SC, Upton Z, Bott K, Dargaville TR. Recent advances in dermal wound healing: biomedical device approaches. Expert Rev Med Devices 2014; 7:143-54. [DOI: 10.1586/erd.09.57] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
20
|
Degen KE, Gourdie RG. Embryonic wound healing: a primer for engineering novel therapies for tissue repair. ACTA ACUST UNITED AC 2013; 96:258-70. [PMID: 23109321 DOI: 10.1002/bdrc.21019] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Scar is the default tissue repair used by the body in response to most injuries-a response that occurs in wounds ranging in seriousness from minor skin cuts to complete severance of the spinal cord. By contrast, before the third trimester of pregnancy embryonic mammals tend to heal without scarring due to a variety of mechanisms and factors that are uniquely in operation during development in utero. The goal of tissue engineering is to develop safe and clinically effective biological substitutes that restore, maintain, or improve tissue function in patients. This review provides a comparative overview of wound healing during development and maturation and seeks to provide a perspective on just how much the embryo may be able teach us in the engineering of new therapies for tissue repair.
Collapse
Affiliation(s)
- Katherine E Degen
- School of Biomedical Engineering Science, Virginia Tech, Blacksburg, USA
| | | |
Collapse
|
21
|
Kim JW, Lee JH, Lyoo YS, Jung DI, Park HM. The effects of topical mesenchymal stem cell transplantation in canine experimental cutaneous wounds. Vet Dermatol 2013; 24:242-e53. [PMID: 23432413 PMCID: PMC3618380 DOI: 10.1111/vde.12011] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2012] [Indexed: 12/26/2022]
Abstract
Background Adult stem cells have been widely investigated in bioengineering approaches for tissue repair therapy. We evaluated the clinical value and safety of the application of cultured bone marrow-derived allogenic mesenchymal stem cells (MSCs) for treating skin wounds in a canine model. Hypothesis Topical allogenic MSC transplantation can accelerate the closure of experimental full-thickness cutaneous wounds and attenuate local inflammation. Animals Adult healthy beagle dogs (n = 10; 3–6 years old; 7.2–13.1 kg) were studied. Methods Full-thickness skin wounds were created on the dorsum of healthy beagles, and allogenic MSCs were injected intradermally. The rate of wound closure and the degree of collagen production were analysed histologically using haematoxylin and eosin staining and trichrome staining. The degree of cellular proliferation and angiogenesis was evaluated by immunocytochemistry using proliferating cell nuclear antigen-, vimentin- and α-smooth muscle actin-specific antibodies. Local mRNA expression levels of interleukin-2, interferon-γ, basic fibroblast growth factor and matrix metalloproteinase-2 were evaluated by RT-PCR. Results Compared with the vehicle-treated wounds, MSC-treated wounds showed more rapid wound closure and increased collagen synthesis, cellular proliferation and angiogenesis. Moreover, MSC-treated wounds showed decreased expression of pro-inflammatory cytokines (interleukin-2 and interferon-γ) and wound healing-related factors (basic fibroblast growth factor and matrix metalloproteinase-2). Conclusion and clinical importance Topical transplantation of MSCs results in paracrine effects on cellular proliferation and angiogenesis, as well as modulation of local mRNA expression of several factors related to cutaneous wound healing. Résumé Resumen Zusammenfassung 摘要 要約
Collapse
Affiliation(s)
- Ju-Won Kim
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, South Korea
| | | | | | | | | |
Collapse
|
22
|
Hanson SE, Kleinbeck KR, Cantu D, Kim J, Bentz ML, Faucher LD, Kao WJ, Hematti P. Local delivery of allogeneic bone marrow and adipose tissue-derived mesenchymal stromal cells for cutaneous wound healing in a porcine model. J Tissue Eng Regen Med 2013; 10:E90-E100. [PMID: 23418160 DOI: 10.1002/term.1700] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Revised: 12/09/2012] [Accepted: 12/20/2012] [Indexed: 12/18/2022]
Abstract
Wound healing remains a major challenge in modern medicine. Bone marrow- (BM) and adipose tissue- (AT) derived mesenchymal stromal/stem cells (MSCs) are of great interest for tissue reconstruction due to their unique immunological properties and regenerative potential. The purpose of this study was to characterize BM and AT-MSCs and evaluate their effect when administered in a porcine wound model. MSCs were derived from male Göttingen Minipigs and characterized according to established criteria. Allogeneic BM- or AT-MSCs were administered intradermally (1 x 10(6) cells) into partial-thickness wounds created on female animals, and covered with Vaseline® gauze or fibrin in a randomized pattern. Animals were euthanized at 7, 10, 14 and 21 days. Tissues were analyzed visually for healing and by microscopic examination for epidermal development and remodelling. Polymerase chain reaction (PCR) was used to detect the presence of male DNA in the specimens. All wounds were healed by 14 days. MSC-injected wounds were associated with improved appearance and faster re-epithelialization compared to saline controls. Evaluation of rete ridge depth and architecture showed that MSC treatment promoted a faster rate of epidermal maturation. Male DNA was detected in all samples at days 7 and 10, suggesting the presence of MSCs. We showed the safety, feasibility and potential efficacy of local injection of allogeneic BM- and AT-MSCs for treatment of wounds in a preclinical model. Our data in this large animal model support the potential use of BM- and AT-MSC for treatment of cutaneous wounds through modulation of healing and epithelialization.
Collapse
Affiliation(s)
- Summer E Hanson
- Division of Plastic and Reconstructive Surgery, Department of Surgery, UW-Madison, School of Medicine and Public Health.,Department of Surgery, UW-Madison, School of Medicine and Public Health.,Department of Biomedical Engineering, UW-Madison, College of Engineering
| | | | - David Cantu
- Division of Pharmaceutical Sciences, UW-Madison
| | - Jaeyhup Kim
- Department of Medicine, UW-Madison, School of Medicine and Public Health
| | - Michael L Bentz
- Division of Plastic and Reconstructive Surgery, Department of Surgery, UW-Madison, School of Medicine and Public Health.,Department of Surgery, UW-Madison, School of Medicine and Public Health
| | - Lee D Faucher
- Department of Surgery, UW-Madison, School of Medicine and Public Health
| | - W John Kao
- Division of Pharmaceutical Sciences, UW-Madison.,Department of Surgery, UW-Madison, School of Medicine and Public Health.,Department of Biomedical Engineering, UW-Madison, College of Engineering
| | - Peiman Hematti
- Department of Medicine, UW-Madison, School of Medicine and Public Health.,University of Wisconsin Carbone Cancer Center, UW-Madison, School of Medicine and Public Health, Madison, WI 53705, USA
| |
Collapse
|
23
|
Ferretti C, Borsari V, Falconi M, Gigante A, Lazzarini R, Fini M, Di Primio R, Mattioli-Belmonte M. Human periosteum-derived stem cells for tissue engineering applications: the role of VEGF. Stem Cell Rev Rep 2012; 8:882-90. [PMID: 22622690 DOI: 10.1007/s12015-012-9374-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) are promising tools for studying the mechanisms of development and for the regeneration of injured tissues. Correct selection of the MSCs source is crucial in order to obtain a more efficient treatment and, in this respect Periosteum-Derived Cells (PDPCs) may represent an interesting alternative to bone marrow MSCs for osteochondral tissue regeneration. In the present study we have isolated and characterized a MSCs population from the periosteum of human adult donors. PDPCs were expanded under specific culture conditions that prevent fibroblast contamination and support the maintenance of their undifferentiated phenotype. We show, for the first time, that PDPCs expresses VEGF receptor (Flt1 and KDR/Flk1) proteins and that they were similar to bone marrow Multipotent Adult Progenitor Cells (MAPCs). Since the latter are able to differentiate into endothelial cells, we tested the possible PDPCs commitment toward an endothelial phenotype in view of bone tissue engineering approaches that takes into account not only bone formation but also vascularization. PDPCs were treated with two different VEGF concentrations for 7 and 15 days and, alternatively, with the supernatant of human primary osteoblasts. Differently from MAPCs our PDPCs were unable to differentiate into endothelial cells after their in vitro VEGF treatment. On the contrary, growth factor stimulation induces PDPCs differentiation toward osteoblasts. We concluded that in PDPCs the presence of VEGF receptors is related to different cross-talk between osteogenesis and angiogenesis that could involve in situ PDPCs recruitment.
Collapse
Affiliation(s)
- C Ferretti
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Via Tonto 10/a, 60126 Ancona, Italy
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Xia Z, Zhang C, Zeng Y, Wang T, Ai G. Transplantation of BMSCs expressing hVEGF165 /hBD3 promotes wound healing in rats with combined radiation-wound injury. Int Wound J 2012; 11:293-303. [PMID: 23137415 DOI: 10.1111/j.1742-481x.2012.01090.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
The combined radiation-wound injury is a refractory wound with decreased number or dysfunction of repairing cells and growth factors. This remains a challenge in clinical practice. The object of this study is to evaluate the therapeutic efficacy of a combination of human vascular endothelial growth factor 165 (hVEGF(165)) and human beta-defensin 3 (hBD3) in the treatment of such wounds. A plasmid-carrying hVEGF(165) gene and hBD3 gene was used to transfect rat bone-marrow-derived mesenchymal stem cells (BMSCs). The supernatant from the modified BMSCs significantly promoted the proliferation and cell migration of human endothelial cells and it also inhibited the growth of bacteria and fungus, demonstrating the successful expression of the transfected genes. The hVEGF(165)/hBD3-modified BMSCs were then injected into the sites of combined radiation-wound injury on rats. It demonstrated that wound-healing time was shortened significantly in the treated rats. The granulation tissue formation/maturation, skin appendage regeneration and collagen deposition were also improved significantly. Strong expression of hVEGF(165) and hBD3 was detected in the wound surface at early stage of the healing. The results indicate that topical transplantation of hVEGF(165)/hBD3-modified BMSCs promoted wound healing, and this gene therapy strategy presents a promising approach in the treatment of refractory wounds such as the combined radiation-wound injury.
Collapse
Affiliation(s)
- Zhangquan Xia
- Department of Stomatology, Southwest Hospital, Third Military Medical University, Chongqing, ChinaDepartment of Stomatology, No 291 Hospital of the People's Liberation Army, Baotou, ChinaDepartment of Radiation Medicine, Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing, China
| | | | | | | | | |
Collapse
|
25
|
Nie C, Zhang G, Yang D, Liu T, Liu D, Xu J, Zhang J. Targeted delivery of adipose-derived stem cells via acellular dermal matrix enhances wound repair in diabetic rats. J Tissue Eng Regen Med 2012; 9:224-35. [PMID: 23038677 DOI: 10.1002/term.1622] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Revised: 07/04/2012] [Accepted: 08/25/2012] [Indexed: 12/31/2022]
Affiliation(s)
- Chunlei Nie
- Department of Head and Neck Surgery; Third Affiliated Hospital of Harbin Medical University; People's Republic of China
- Tissue Repair and Engineering Laboratory; Harbin Medical University; People's Republic of China
| | - Guoyou Zhang
- Department of Dermatology; University of Lübeck; Germany
- Department of Hand and Plastic Surgery; Second Affiliated Hospital of Wenzhou Medical College; Zhejiang People's Republic of China
| | - Daping Yang
- Tissue Repair and Engineering Laboratory; Harbin Medical University; People's Republic of China
- Department of Plastic Surgery; Second Affiliated Hospital of Harbin Medical University; People's Republic of China
| | - Tong Liu
- Department of Head and Neck Surgery; Third Affiliated Hospital of Harbin Medical University; People's Republic of China
| | - Dan Liu
- Department of Plastic Surgery; Second Affiliated Hospital of Harbin Medical University; People's Republic of China
| | - Jin Xu
- Department of Cell Biology; Harbin Medical University; People's Republic of China
| | - Jiewu Zhang
- Department of Head and Neck Surgery; Third Affiliated Hospital of Harbin Medical University; People's Republic of China
| |
Collapse
|
26
|
Shohara R, Yamamoto A, Takikawa S, Iwase A, Hibi H, Kikkawa F, Ueda M. Mesenchymal stromal cells of human umbilical cord Wharton's jelly accelerate wound healing by paracrine mechanisms. Cytotherapy 2012; 14:1171-81. [PMID: 22900957 DOI: 10.3109/14653249.2012.706705] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND AIMS Mesenchymal stromal cells (MSC) can be isolated from the perivascular connective tissue of umbilical cords, called Wharton's jelly. These human umbilical cord perivascular cells (HUCPVC) might provide therapeutic benefits when treating skeletal or cutaneous malformations in neonatal patients. METHODS HUCPVC were isolated, and their proliferation rate, marker expression and multilineage differentiation potential determined. HUCPVC or their conditioned medium (HUCPVC-CM) was injected into the excisional wound of a mouse splinted-wound model. The effects of the treatment on wound closure were examined by morphohistochemical and gene expression analyses. RESULTS HUCPVC expressed typical MSC markers and could differentiate into osteoblastic and adipogenic lineages. HUCPVC transplanted into the mouse wound accelerated wound closure. Immunohistologic analysis showed that the HUCPVC accelerated wound healing by enhancing collagen deposition and angiogenesis via paracrine mechanisms. Furthermore, treatment with HUCPVC-CM alone significantly enhanced wound closure. HUCPVC-CM increased the number of anti-inflammatory M2 macrophages expressing resistin-like molecule (RELM)-α/CD11b and promoted neovessel maturation. Quantitative polymerase chain reaction (PCR) analysis showed that HUCPVC-CM increased the expression of tissue-repairing cytokines interleukin (IL)-10, transforming growth factor (TGF)-β1, vascular endothelial growth factor (VEGF)-1 and angiopoietin-1 at the healing wound. CONCLUSIONS Our results show that HUCPVC promotes wound healing via multifaceted paracrine mechanisms. Together with their ability to differentiate into the osteogenic linage, HUCPVC may provide significant therapeutic benefits for treating wounds in neonatal patients.
Collapse
Affiliation(s)
- Ryutaro Shohara
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, Nagoya 466 – 8550, Japan
| | | | | | | | | | | | | |
Collapse
|
27
|
Connolly EC, Freimuth J, Akhurst RJ. Complexities of TGF-β targeted cancer therapy. Int J Biol Sci 2012; 8:964-78. [PMID: 22811618 PMCID: PMC3399319 DOI: 10.7150/ijbs.4564] [Citation(s) in RCA: 269] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Accepted: 06/23/2012] [Indexed: 02/07/2023] Open
Abstract
Many advanced tumors produce excessive amounts of Transforming Growth Factor-β (TGF-β) which, in normal epithelial cells, is a potent growth inhibitor. However, in oncogenically activated cells, the homeostatic action of TGF-β is often diverted along alternative pathways. Hence, TGF-β signaling elicits protective or tumor suppressive effects during the early growth-sensitive stages of tumorigenesis. However, later in tumor development when carcinoma cells become refractory to TGF-β-mediated growth inhibition, the tumor cell responds by stimulating pathways with tumor progressing effects. At late stages of malignancy, tumor progression is driven by TGF-β overload. The tumor microenvironment is a target of TGF-β action that stimulates tumor progression via pro-tumorigenic effects on vascular, immune, and fibroblastic cells. Bone is one of the richest sources of TGF-β in the body and a common site for dissemination of breast cancer metastases. Osteoclastic degradation of bone matrix, which accompanies establishment and growth of metastases, triggers further release of bone-derived TGF-β. This leads to a vicious positive feedback of tumor progression, driven by ever increasing levels of TGF-β released from both the tumor and bone matrix. It is for this reason, that pharmaceutical companies have developed therapeutic agents that block TGF-β signaling. Nonetheless, the choice of drug design and dosing strategy can affect the efficacy of TGF-β therapeutics. This review will describe pre-clinical and clinical data of four major classes of TGF-β inhibitor, namely i) ligand traps, ii) antisense oligonucleotides, iii) receptor kinase inhibitors and iv) peptide aptamers. Long term dosing strategies with TGF-β inhibitors may be ill-advised, since this class of drug has potentially highly pleiotropic activity, and development of drug resistance might potentiate tumor progression. Current paradigms for the use of TGF-β inhibitors in oncology have therefore moved towards the use of combinatorial therapies and short term dosing, with considerable promise for the clinic.
Collapse
Affiliation(s)
- Erin C. Connolly
- 1. UCSF Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, California 94143-0512, USA
| | - Julia Freimuth
- 1. UCSF Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, California 94143-0512, USA
| | - Rosemary J. Akhurst
- 1. UCSF Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, California 94143-0512, USA
- 2. Department of Anatomy, University of California at San Francisco, California 94143-0512, USA
| |
Collapse
|
28
|
Dewi D, Ishii H, Haraguchi N, Nishikawa S, Kano Y, Fukusumi T, Ohta K, Miyazaki S, Ozaki M, Sakai D, Satoh T, Nagano H, Doki Y, Mori M. Reprogramming of gastrointestinal cancer cells. Cancer Sci 2012; 103:393-9. [PMID: 22151786 DOI: 10.1111/j.1349-7006.2011.02184.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Cell reprogramming reverts cells to multipotent, preprogrammed states by re-establishing epigenetic markers. It can also induce considerable malignant phenotype modification. Because key events in cancer relapse and metastasis, including epithelial-mesenchymal transition phenotypes, are regulated primarily by reversible and transient epigenetic modifications rather than the accumulation of irreversible and stable genetic abnormalities, studying dynamic mechanisms regulating these biological processes is important. Transcription factors for induced pluripotent stem cells and non-coding microRNAs allow pluripotent phenotype induction. We present the current knowledge of the possible applications of cell reprogramming in reducing aggressive phenotype expression, which can induce tumor cell hibernation and maintain appropriate phenotypes, thereby minimizing relapse and metastasis after surgical resection of gastrointestinal cancer.
Collapse
Affiliation(s)
- DyahLaksmi Dewi
- Department of Frontier Science for Cancer and Chemotherapy, Graduate School of Medicine, Osaka University, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Connolly EC, Akhurst RJ. The complexities of TGF-β action during mammary and squamous cell carcinogenesis. Curr Pharm Biotechnol 2011; 12:2138-49. [PMID: 21619543 PMCID: PMC3520605 DOI: 10.2174/138920111798808284] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2010] [Revised: 01/28/2011] [Accepted: 01/31/2011] [Indexed: 12/19/2022]
Abstract
Many advanced tumors produce excess amounts of Transforming Growth Factor-β (TGF-β), which is a potent growth inhibitor of normal epithelial cells. However, in tumors its homeostatic action on cells can be diverted along several alternative pathways. Thus, TGF-β signaling has been reported to elicit a preventative or tumor suppressive effect during the earlier stages of tumorigenesis, but later in tumor development, when carcinoma cells become refractory to TGF-β-mediated growth inhibition, response to TGF-β signaling elicits predominantly tumor progressing effects. This is not a simple switch from suppression to progression, but more like a rheostat, involving multiple complementary and antagonizing activities that slowly tip the balance from one to the other. This review will focus on the multiple activities of TGF-β in regulation of two epithelial tumor types, namely squamous cell carcinoma and breast cancer. Basic findings in current mouse models of cancer are presented, as well as a discussion of the complicating issue of outcome of altered TGFβ signaling depending on genetic variability between mouse strains. This review also discusses the role TGF-β within the tumor microenvironment particularly its ability to polarize the microenvironment towards a pro-tumorigenic milieu.
Collapse
Affiliation(s)
- Erin C. Connolly
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, California 94143-0512. USA
| | - Rosemary J. Akhurst
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, California 94143-0512. USA
- Department of Anatomy, University of California San Francisco, California 94143-0512. USA
| |
Collapse
|
30
|
Plaas A, Velasco J, Gorski DJ, Li J, Cole A, Christopherson K, Sandy JD. The relationship between fibrogenic TGFβ1 signaling in the joint and cartilage degradation in post-injury osteoarthritis. Osteoarthritis Cartilage 2011; 19:1081-90. [PMID: 21624477 DOI: 10.1016/j.joca.2011.05.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Revised: 05/05/2011] [Accepted: 05/07/2011] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To review the literature on modulation of chondrocyte activities in the osteoarthritic joint, and to discuss these changes in relation to established hard and soft tissue repair paradigms, with an emphasis on transforming growth factor beta (TGFβ1)-mediated signaling which can promote either a chondrogenic or fibrogenic phenotype. METHODS Papers addressing the close relationship between repair in general, and the specific post-injury response of joint tissues are summarized. Different interpretations of the role of TGFβ1 in the emergence of an "osteoarthritic" chondrocyte are compared and the phenotypic plasticity of "reparative" progenitor cells is examined. Lastly, emerging data on a central role for A-Disintegrin-And-Metalloproteinase-with-Thrombospondin-like-Sequences-5 (ADAMTS5) activity in modulating TGFβ1 signaling through activin receptor-like kinase 1 (ALK1) and activin receptor-like kinase 5 (ALK5) pathways is discussed. RESULTS The review illustrates how a transition from ALK5-mediated fibrogenic signaling to ALK1-mediated chondrogenic signaling in joint cells represents the critical transition from a non-reparative to a reparative cell phenotype. Data from cell and in vivo studies illustrates the mechanism by which ablation of ADAMTS5 activity allows the transition to reparative chondrogenesis. Multiple large gene expression studies of normal and osteoarthritis (OA) human cartilages (CAs) also support an important role for TGFβ1-mediated pro-fibrogenic activities during disease progression. CONCLUSIONS We conclude that progressive articular CA damage in post-injury OA results primarily from biomechanical, cell biologic and mediator changes that promote a fibroblastic phenotype in joint cells. Since ADAMTS5 and TGFβ1 appear to control this process, agents which interfere with their activities may not only enhance endogenous CA repair in vivo, but also improve the properties of tissue-engineered CA for implantation.
Collapse
Affiliation(s)
- A Plaas
- Department of Internal Medicine (Rheumatology), Rush University Medical Center, Chicago, IL, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Maharlooei MK, Bagheri M, Solhjou Z, Jahromi BM, Akrami M, Rohani L, Monabati A, Noorafshan A, Omrani GR. Adipose tissue derived mesenchymal stem cell (AD-MSC) promotes skin wound healing in diabetic rats. Diabetes Res Clin Pract 2011; 93:228-234. [PMID: 21632142 DOI: 10.1016/j.diabres.2011.04.018] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Accepted: 04/25/2011] [Indexed: 12/21/2022]
Abstract
AIMS Stem cells are a new hope to ameliorate impaired diabetic wound healing. The purpose of this study was to evaluate the effect of adipose tissue derived mesenchymal stem cells (AD-MSCs) on wound healing in a diabetic rat model. METHODS Twenty-six rats became diabetic by a single intraperitoneal injection of streptozotocin. Six rats served as non-diabetic (non-DM). Diabetic rats were divided into two equal groups randomly; control and treatment. Six weeks later, a full-thickness circular excisional wound was created on the dorsum of each rat. AD-MSCs were injected intra-dermally around the wounds of treatment group. PBS was applied to control and non-DM groups. The wound area was measured every other day. After wound healing completion, full thickness skin samples were taken from the wound sites for evaluation of volume density of collagen fibers, length and volume density of vessels, and numerical density of fibroblasts by stereological methods. RESULTS AD-MSCs accelerated wound healing rate in diabetic rats, but did not increase length and volume density of the vessels and volume density of the collagen fibers. AD-MSCs decreased the numerical density of fibroblasts. CONCLUSIONS We concluded that AD-MSCs enhances diabetic wound healing rate probably by other mechanisms rather than enhancing angiogenesis or accumulating collagen fibers.
Collapse
Affiliation(s)
- Mohsen Khosravi Maharlooei
- Cell and Molecular Medicine Research Group, Student Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mansooreh Bagheri
- Cell and Molecular Medicine Research Group, Student Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Zhabiz Solhjou
- Cell and Molecular Medicine Research Group, Student Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Behnam Moein Jahromi
- Cell and Molecular Medicine Research Group, Student Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Majid Akrami
- Resident of General Surgery, Department of Surgery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Lili Rohani
- Laboratory for Stem Cell Research, Department of Anatomy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Monabati
- Department of Pathology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Noorafshan
- Histomorphometry & Stereology Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Gholamhossein Ranjbar Omrani
- Endocrine and Metabolism Research Centre, Department of Internal Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
32
|
Heo SC, Jeon ES, Lee IH, Kim HS, Kim MB, Kim JH. Tumor necrosis factor-α-activated human adipose tissue-derived mesenchymal stem cells accelerate cutaneous wound healing through paracrine mechanisms. J Invest Dermatol 2011; 131:1559-67. [PMID: 21451545 DOI: 10.1038/jid.2011.64] [Citation(s) in RCA: 141] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Human adipose tissue-derived mesenchymal stem cells (ASCs) stimulate regeneration of injured tissues by secretion of various cytokines and chemokines. Wound healing is mediated by multiple steps including inflammation, epithelialization, neoangiogenesis, and proliferation. To explore the paracrine functions of ASCs on regeneration of injured tissues, cells were treated with tumor necrosis factor-α (TNF-α), a key inflammatory cytokine, and the effects of TNF-α-conditioned medium (CM) on tissue regeneration were determined using a rat excisional wound model. We demonstrated that TNF-α CM accelerated wound closure, angiogenesis, proliferation, and infiltration of immune cells into the cutaneous wound in vivo. To assess the role of proinflammatory cytokines IL-6 and IL-8, which are included in TNF-α CM, IL-6 and IL-8 were depleted from TNF-α CM using immunoprecipitation. Depletion of IL-6 or IL-8 largely attenuated TNF-α CM-stimulated wound closure, angiogenesis, proliferation, and infiltration of immune cells. These results suggest that TNF-α-activated ASCs accelerate cutaneous wound healing through paracrine mechanisms involving IL-6 and IL-8.
Collapse
Affiliation(s)
- Soon Chul Heo
- Medical Research Center for Ischemic Tissue Regeneration, The Medical Research Institute, School of Medicine, Pusan National University, Gyeongsangnam-do, Republic of Korea
| | | | | | | | | | | |
Collapse
|
33
|
Hendrickx B, Vranckx JJ, Luttun A. Cell-Based Vascularization Strategies for Skin Tissue Engineering. TISSUE ENGINEERING PART B-REVIEWS 2011; 17:13-24. [DOI: 10.1089/ten.teb.2010.0315] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Benoit Hendrickx
- Center for Molecular and Vascular Biology, Katholieke Universiteit Leuven, Leuven, Belgium
- Laboratory of Plastic Surgery and Tissue Engineering Research, Department of Plastic, Reconstructive, and Aesthetic Surgery, KUL–University Hospitals, Leuven, Belgium
| | - Jan J. Vranckx
- Laboratory of Plastic Surgery and Tissue Engineering Research, Department of Plastic, Reconstructive, and Aesthetic Surgery, KUL–University Hospitals, Leuven, Belgium
| | - Aernout Luttun
- Center for Molecular and Vascular Biology, Katholieke Universiteit Leuven, Leuven, Belgium
| |
Collapse
|
34
|
Battula VL, Evans KW, Hollier BG, Shi Y, Marini FC, Ayyanan A, Wang RY, Brisken C, Guerra R, Andreeff M, Mani SA. Epithelial-mesenchymal transition-derived cells exhibit multilineage differentiation potential similar to mesenchymal stem cells. Stem Cells 2011; 28:1435-45. [PMID: 20572012 DOI: 10.1002/stem.467] [Citation(s) in RCA: 212] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The epithelial-to-mesenchymal transition (EMT) is an embryonic process that becomes latent in most normal adult tissues. Recently, we have shown that induction of EMT endows breast epithelial cells with stem cell traits. In this report, we have further characterized the EMT-derived cells and shown that these cells are similar to mesenchymal stem cells (MSCs) with the capacity to differentiate into multiple tissue lineages. For this purpose, we induced EMT by ectopic expression of Twist, Snail, or transforming growth factor-beta in immortalized human mammary epithelial cells. We found that the EMT-derived cells and MSCs share many properties including the antigenic profile typical of MSCs, that is, CD44(+), CD24(-), and CD45(-). Conversely, MSCs express EMT-associated genes, such as Twist, Snail, and mesenchyme forkhead 1 (FOXC2). Interestingly, CD140b (platelet-derived growth factor receptor-beta), a marker for naive MSCs, is exclusively expressed in EMT-derived cells and not in their epithelial counterparts. Moreover, functional analyses revealed that EMT-derived cells but not the control cells can differentiate into alizarin red S-positive mature osteoblasts, oil red O-positive adipocytes and alcian blue-positive chondrocytes similar to MSCs. We also observed that EMT-derived cells but not the control cells invade and migrate towards MDA-MB-231 breast cancer cells similar to MSCs. In vivo wound homing assays in nude mice revealed that the EMT-derived cells home to wound sites similar to MSCs. In conclusion, we have demonstrated that the EMT-derived cells are similar to MSCs in gene expression, multilineage differentiation, and ability to migrate towards tumor cells and wound sites.
Collapse
Affiliation(s)
- Venkata Lokesh Battula
- Section of Molecular Hematology and Therapy, Department of Stem Cell Transplantation, The University of Texas-M.D. Anderson Cancer Center, Houston, Texas, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Alexeev V, Uitto J, Igoucheva O. Gene expression signatures of mouse bone marrow-derived mesenchymal stem cells in the cutaneous environment and therapeutic implications for blistering skin disorder. Cytotherapy 2010; 13:30-45. [PMID: 20854215 DOI: 10.3109/14653249.2010.518609] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND AIMS Multiple studies have demonstrated that mesenchymal stromal cells (MSC) can be utilized therapeutically for various congenital and acquired disorders. The involvement of MSC in the maintenance of skin homeostasis and their curative application for the treatment of skin wounds have also been documented. However, it is not known whether MSC can commit to cutaneous lineages, produce structural proteins essential for the skin integrity or be used for hereditary skin disorders. METHODS To address these questions, we conducted a comparative expression analysis between MSC and potentially adjacent cutaneous cells, fibroblasts and keratinocytes, with specific emphasis on extracellular matrix encoding and related genes. RESULTS Our data demonstrated that MSC share many features with cutaneous fibroblasts. We also observed that under direct influence of cutaneous fibroblasts in vitro and fibroblast-derived matrix in vivo, MSC acquired a fibroblastic phenotype, suggesting that specific cell-cell interactions play a key regulatory role in the differentiation of MSC. Additionally, the observed fibroblastic transition of MSC was underlined by a significant up-regulation of several cutaneous-specific genes encoding lumican, decorin, type VII collagen, laminin and other structural proteins. As many of the identified genes have considerable therapeutic value for dermatologic afflictions, particularly type VII collagen, we evaluated further the therapeutic potential of congenic MSC in the skin of Col7a1-null mice recapitulating human recessive dystrophic epidermolysis bullosa (RDEB). Remarkably, MSC-derived type VII collagen was sufficient for restoration of the damaged dermal-epidermal junction and partial reversal of the RDEB phenotype. CONCLUSIONS Collectively, our results suggest that MSC may offer promising therapeutics for the treatment of RDEB and potentially other genodermatoses.
Collapse
Affiliation(s)
- Vitali Alexeev
- Department of Dermatology and Cutaneous Biology, Jefferson Medical College, Thomas Jefferson University, 233 South 10th Street, Philadelphia, PA 19107, USA
| | | | | |
Collapse
|
36
|
Nichols JE, Niles J, Walls S, Cortiella J. In vitro human bone marrow analog: clinical potential. Regen Med 2010; 5:289-98. [PMID: 20210588 DOI: 10.2217/rme.10.7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Bone marrow is the primary site of hematopoiesis in adult humans. Bone marrow can be cultured in vitro but few simple culture systems fully support hematopoiesis beyond a few months. Human bone marrow analogs are long-term in vitro cultures of marrow stromal and hematopoietic stem cells that can be used to produce cells and products normally harvested from human donors. Bone marrow analog systems should exhibit confluence of the stromal cell populations, persistence of hematopoietic progenitor cells, presence of active regions of hematopoiesis and capacity to produce mature cell types for extended periods of time. Although we are still years away from realizing clinical application of products formed by artificial bone marrow analogs, the process of transitioning this research tool from bench to bedside should be fairly straightforward. The most obvious application of artificial marrow would be for production of autologous hematopoietic CD34(+) stem cells as a stem cell therapy for individuals experiencing bone marrow failure due to disease or injury. Another logical application is for 'blood farming', a process for large-scale in vitro production of red blood cells, white blood cells or platelets, for transfusion or treatment. Other possibilities include production of nonhematopoietic stem cells such as osteogenic stromal cells, osteoblasts and rare pluripotent stem cells. Bone marrow analogs also have great potential as ex vivo human test systems and could play a critical role in drug discovery, drug development and toxicity testing in the future.
Collapse
Affiliation(s)
- Joan E Nichols
- Laboratory of Regenerative & Nano-Medicine, Department of Internal Medicine, University of Texas Medical Branch at Galveston, Galveston, TX 77555-0435, USA.
| | | | | | | |
Collapse
|
37
|
Zhao X, Wu N, Huang L. Endothelial progenitor cells and spleen: new insights in regeneration medicine. Cytotherapy 2010; 12:7-16. [PMID: 19878079 DOI: 10.3109/14653240903300674] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
As a promising concept for regeneration medicine, endothelial progenitor cell (EPC) therapy represents a novel strategy for a variety of diseases. Increasing evidence suggests that the spleen, a traditionally dispensable organ, acts as a major reservoir during EPC trafficking and plays an important role regarding the modulation of circulating EPC kinetics. Moreover, infusion of splenic EPC can restore endothelial function and promote neovascularization, indicating an available resource for EPC transplantation. Thus a discussion of the role of the spleen with respect to EPC may provide novel information for management of EPC therapy.
Collapse
Affiliation(s)
- Xiaohui Zhao
- Cardiovascular Department, XinQiao Hospital, China
| | | | | |
Collapse
|
38
|
Nakatsuka R, Nozaki T, Uemura Y, Matsuoka Y, Sasaki Y, Shinohara M, Ohura K, Sonoda Y. 5-Aza-2'-deoxycytidine treatment induces skeletal myogenic differentiation of mouse dental pulp stem cells. Arch Oral Biol 2010; 55:350-7. [PMID: 20362276 DOI: 10.1016/j.archoralbio.2010.03.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2009] [Revised: 03/03/2010] [Accepted: 03/06/2010] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Tissue stem cells in dental pulp are assumed to possess differentiation potentials similar to mesenchymal stem cells (MSCs). The aim of this in vitro study is to examine the differentiation potentials of mouse dental pulp stem cells (DPSCs) and develop the appropriate differentiation assay systems for skeletal myogenic differentiation of these cells. METHODS Dental pulps were extracted from mandible sections of C57/BL6 mice, and adherent dental pulp cells were isolated in culture. These cells were cultured in osteogenic or adipogenic induction medium to induce osteogenic and adipogenic differentiation. On the other hand, the skeletal myogenic differentiation potential of these cells was investigated using different conditions, such as serum-free medium, Myod1 overexpression, or 5-Aza-2'-deoxycytidine (5-Aza) treatment for DNA demethylation. Muscle-specific transcriptional factor expression was evaluated by RT-PCR, and myotube formation and myosin heavy chain expression were evaluated by phase-contrast microscopy and immunofluorescence staining, respectively. RESULTS The adherent dental pulp cells exhibited a proliferative capacity and they showed osteogenic and adipogenic differentiation as seen in previous studies. Although the expression of Myod1 mRNA and myotube formation was not detected in serum-free conditions, the forced expression of Myod1 up-regulated the expression of Myogenin and Pax7 mRNA. However, myotube formation was not confirmed. Interestingly, myosin heavy chain expression and myotube formation were observed following 5-Aza treatment of these cells. CONCLUSIONS These results demonstrated that mouse DPSCs possess MSC-like differentiation potential. DNA demethylation induced by 5-Aza treatment resulted in the skeletal muscle differentiation in mouse DPSCs, suggesting that DNA demethylation might trigger this differential induction of mouse DPSCs.
Collapse
Affiliation(s)
- Ryusuke Nakatsuka
- Department of Stem Cell Biology and Regenerative Medicine, Graduate School of Medical Science, Kansai Medical University, 10-15 Fumizono-cho, Moriguchi, Osaka 570-8506, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
SUMMARY Chronic wounds remain a major challenge in modern medicine and represent a significant burden, affecting not only physical and mental health, but also productivity, health care expenditure, and long-term morbidity. Even under optimal conditions, the healing process leads to fibrosis or scar. One promising solution, cell therapy, involves the transplantation of progenitor/stem cells to patients through local or systemic delivery, and offers a novel approach to many chronic diseases, including nonhealing wounds. Mesenchymal stem cells are multipotent, adult progenitor cells of great interest because of their unique immunologic properties and regenerative potential. A variety of preclinical and clinical studies have shown that mesenchymal stem cells may have a useful role in wound-healing and tissue-engineering strategies and both aesthetic and reconstructive surgery. Recent advances in stem cell immunobiology can offer insight into the multiple mechanisms through which mesenchymal stem cells could affect underlying pathophysiologic processes associated with nonhealing mesenchymal stem cells. Critical evaluation of the current literature is necessary for understanding how mesenchymal stem cells could potentially revolutionize our approach to skin and soft-tissue defects and designing clinical trials to address their role in wound repair and regeneration.
Collapse
|
40
|
Turner NJ, Johnson SA, Badylak SF. A histomorphologic study of the normal healing response following digit amputation in C57bl/6 and MRL/MpJ mice. ACTA ACUST UNITED AC 2010; 73:103-11. [DOI: 10.1679/aohc.73.103] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Neill J. Turner
- McGowan Institute for Regenerative Medicine, University of Pittsburgh
| | - Scott A. Johnson
- McGowan Institute for Regenerative Medicine, University of Pittsburgh
| | - Stephen F. Badylak
- McGowan Institute for Regenerative Medicine, University of Pittsburgh
- Department of Surgery, University of Pittsburgh
| |
Collapse
|
41
|
Zhuge Y, Liu ZJ, Habib B, Velazquez OC. Diabetic foot ulcers: effects of hyperoxia and SDF-1α on endothelial progenitor cells. Expert Rev Endocrinol Metab 2010; 5:113-125. [PMID: 30934386 DOI: 10.1586/eem.09.61] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Diabetes mellitus is a common disease afflicting many people. In addition to coronary artery disease, diabetic retinopathy and renal failure, diabetic patients face abnormal wound healing and have increased lower extremity ulcers and amputations. In diabetes, wound healing is altered due to both macrovascular and microvascular processes. While the former can be addressed with surgical intervention, the latter is more difficult to correct. Neovascularization within the granulation tissue via angiogenesis and vasculogenesis is critical for wound healing. Endothelial progenitor cells (EPCs) have been implicated in vasculogenesis. Mobilization of EPCs from the bone marrow is impaired in diabetes and homing of EPCs to the wound is also abnormal. Recent studies show that hyperoxia and administration of exogenous stromal-derived factor-1α increases circulatory and wound levels of EPCs and improves wound healing in diabetic mice. These findings have great potential for translation into human counterparts as the treatment for this prevalent disease matures.
Collapse
Affiliation(s)
- Ying Zhuge
- a University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Zhao-Jun Liu
- b University of Miami Sylvester Comprehensive Cancer Center, Miami, FL, USA and Division of Vascular and Endovascular Surgery, DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Room 3016, Holtz Center - JMH East Tower, 1611 NW 12th Avenue, Miami, FL 33136, USA
| | - Bianca Habib
- a University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Omaida C Velazquez
- c University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL, USA and Chief, Division of Vascular and Endovascular Surgery, DeWitt Daughtry Family Department of Surgery, University of Miami, Miller School of Medicine, Room 3016, Holtz Center - JMH East Tower, 1611 NW 12th Avenue, Miami, FL 33136, USA.
| |
Collapse
|
42
|
Battiwalla M, Hematti P. Mesenchymal stem cells in hematopoietic stem cell transplantation. Cytotherapy 2009; 11:503-15. [PMID: 19728189 DOI: 10.1080/14653240903193806] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Mesenchymal stromal/stem cells (MSC) of bone marrow (BM) origin not only provide the supportive microenvironmental niche for hematopoietic stem cells (HSC) but are capable of differentiating into various cell types of mesenchymal origin, such as bone, fat and cartilage. In vitro and in vivo data suggest that MSC have low inherent immunogenicity, modulate/suppress immunologic responses through interactions with immune cells, and home to damaged tissues to participate in regeneration processes through their diverse biologic properties. MSC derived from BM are being evaluated for a wide range of clinical applications, including disorders as diverse as myocardial infarction and newly diagnosed diabetes mellitus type 1. However, their use in HSC transplantation, either for enhancement of hematopoietic engraftment or for treatment/prevention of graft-versus-host disease, is far ahead of other indications. Ease of isolation and ex vivo expansion of MSC, combined with their intriguing immunomodulatory properties and their impressive record of safety in a wide variety of clinical trials, make these cells promising candidates for further investigation.
Collapse
Affiliation(s)
- Minoo Battiwalla
- Department of Medicine, Roswell Park Cancer Institute, New York, New York, USA
| | | |
Collapse
|
43
|
Ejaz S, Ashraf M, Nawaz M, Lim CW. Total particulate matter and wound healing: an in vivo study with histological insights. BIOMEDICAL AND ENVIRONMENTAL SCIENCES : BES 2009; 22:278-287. [PMID: 19950522 DOI: 10.1016/s0895-3988(09)60057-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
OBJECTIVES Wound healing in the skin is a multifarious orchestration of cellular processes and cigarette smoking may be a cause for delayed wound healing. The aim of this study was to investigate the plausible association between exposures of cigarette total particulate matter (TPM) and wound healing. METHODS An in vivo wound healing model of mice was established for determination of assorted events of wound healing, dermal matrix regeneration, re-epithelialization, and neovascularization. A total of 72 adult mice, separated in eight groups, were exposed to TPM for 12 days. RESULTS A highly considerable diminution in wound closure (P < 0.001) was pragmatic among all TPM-treated mice from day 6 to day 8 post-wounding. Histological investigations unveiled a noteworthy impede in the outcome of re-epithelialization, dermal matrix regeneration and maturation of collagen bundles among all TPM-exposed wounds. Delayed commencement of neovascularization was pragmatic among all TPM-treated mice, on day 12 post wounding. Abbot curve, angular spectrum, and other different parameters of 3D surface behavior of wounds revealed a very highly significant reduction (P < 0.001) in angiogenesis on days 6 and 8 post-wounding, which points that application of TPM instigates extensive delay in trigging the progression of angiogenesis, resulting in delayed onset of wound healing. CONCLUSION Our annotations validate the damaging effects of TPM on wound healing and excessive use of TPM may lead to the production of chronic wounds and oral ulcers.
Collapse
Affiliation(s)
- Sohail Ejaz
- Department of Clinical Neurosciences, Neurology Unit, Addenbrookes Hospital, University of Cambridge, UK.
| | | | | | | |
Collapse
|
44
|
Ejaz S, Iqbal A, Rahman SA, Bari F, Ashraf M, Nawaz M, Lim CW, Kim B. Toxicological evaluation of the effects of 2-stroke auto-rickshaw smoke solutions on wound healing. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2009; 27:373-383. [PMID: 21783967 DOI: 10.1016/j.etap.2008.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2008] [Revised: 12/10/2008] [Accepted: 12/12/2008] [Indexed: 05/31/2023]
Abstract
Vehicle exhaust from traffic is a widespread air pollutant. The use of 3-wheel auto-rickshaws powered by a 2-stroke engine is widespread in south Asia; exhaust from these vehicles may cause different types of toxicities resulting in different pathologies. The aim of this study was to explore the association between exposure to 2-stroke auto-rickshaw smoke solution (2SARSS) and wound healing. The in vivo model of wound healing was customized to evaluate different stages of wound healing: dermal matrix regeneration, re-epithelialization, and neovascularization. A total of 72 adult mice were divided into 8 groups and exposed to 2SARSS for 12 days. A highly significant reduction (p<0.001) in wound closure was observed among all 2SARSS-treated groups at day 8 post-wounding. Histological examination revealed a significant delay in the outcome of re-epithelialization, dermal matrix regeneration, and maturation of collagen bundles among all 2SARSS-exposed wounds. Delayed activation of neovascularization was seen in the 2SARSS-treated groups at day 12 post-wounding. The Abbot curve, angular spectrum, and several other 3D surface parameters of reverse wound topographies revealed a highly significant reduction (p<0.001) in angiogenesis. These results demonstrate that application of 2SARSS causes a substantial delay in the progression of angiogenesis, resulting in delayed onset of wound healing. These observations validate the damaging effects of 2SARSS on wound healing. Thus, people who are directly or indirectly exposed to this toxic exhaust are expected to have delayed wound healing, which could result in chronic wounds.
Collapse
Affiliation(s)
- Sohail Ejaz
- Department of Clinical Neurosciences, Neurology Unit, Addenbrookes Hospital, University of Cambridge, Cambridge, UK; Angiogenesis and Toxicology Research Laboratory, Department of Pharmacology and Toxicology, University of Veterinary and Animal Sciences, Lahore, Pakistan; Biosafety Research Institute and College of Veterinary Medicine, Chonbuk National University, Jeonju, 561-756, South Korea
| | | | | | | | | | | | | | | |
Collapse
|