1
|
Poliwoda S, Noor N, Downs E, Schaaf A, Cantwell A, Ganti L, Kaye AD, Mosel LI, Carroll CB, Viswanath O, Urits I. Stem cells: a comprehensive review of origins and emerging clinical roles in medical practice. Orthop Rev (Pavia) 2022; 14:37498. [PMID: 36034728 PMCID: PMC9404248 DOI: 10.52965/001c.37498] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/10/2023] Open
Abstract
Stem cells are types of cells that have unique ability to self-renew and to differentiate into more than one cell lineage. They are considered building blocks of tissues and organs. Over recent decades, they have been studied and utilized for repair and regenerative medicine. One way to classify these cells is based on their differentiation capacity. Totipotent stem cells can give rise to any cell of an embryo but also to extra-embryonic tissue as well. Pluripotent stem cells are limited to any of the three embryonic germ layers; however, they cannot differentiate into extra-embryonic tissue. Multipotent stem cells can only differentiate into one germ line tissue. Oligopotent and unipotent stem cells are seen in adult organ tissues that have committed to a cell lineage. Another way to differentiate these cells is based on their origins. Stem cells can be extracted from different sources, including bone marrow, amniotic cells, adipose tissue, umbilical cord, and placental tissue. Stem cells began their role in modern regenerative medicine in the 1950's with the first bone marrow transplantation occurring in 1956. Stem cell therapies are at present indicated for a range of clinical conditions beyond traditional origins to treat genetic blood diseases and have seen substantial success. In this regard, emerging use for stem cells is their potential to treat pain states and neurodegenerative diseases such as Parkinson's and Alzheimer's disease. Stem cells offer hope in neurodegeneration to replace neurons damaged during certain disease states. This review compares stem cells arising from these different sources of origin and include clinical roles for stem cells in modern medical practice.
Collapse
Affiliation(s)
| | - Nazir Noor
- Department of Anesthesiology, Mount Sinai Medical Center
| | - Evan Downs
- LSU Health Science Center Shreveport School of Medicine, Shreveport, LA
| | - Amanda Schaaf
- University of Arizona College of Medicine-Phoenix, Phoenix, AZ
| | | | - Latha Ganti
- Department of Emergency Medicine, University of Central Florida
| | - Alan D Kaye
- Department of Anesthesiology, Louisiana State University Health Sciences Center Shreveport
| | - Luke I Mosel
- Department of Anesthesiology, Louisiana State University Health Sciences Center Shreveport
| | - Caroline B Carroll
- Department of Anesthesiology, Louisiana State University Health Sciences Center Shreveport
| | - Omar Viswanath
- Department of Anesthesiology, Louisiana State University Health Sciences Center Shreveport, Innovative Pain and Wellness, Creighton University School of Medicine
| | - Ivan Urits
- Department of Anesthesiology, Louisiana State University Health Sciences Center Shreveport
| |
Collapse
|
2
|
Pishavar E, Copus JS, Atala A, Lee SJ. Comparison Study of Stem Cell-Derived Extracellular Vesicles for Enhanced Osteogenic Differentiation. Tissue Eng Part A 2020; 27:1044-1054. [PMID: 33045930 DOI: 10.1089/ten.tea.2020.0194] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Stem cell-derived extracellular vesicles (EVs) have shown great promise in the field of regenerative medicine and tissue engineering. Recently, human bone marrow-derived mesenchymal stem cell (BMSC)-derived EVs have been considered for bone tissue engineering applications. In this study, we evaluated the osteogenic capability of placental stem cell (PSC)-derived EVs and compared them to the well-characterized BMSC-derived EVs. EVs were extracted from three designated time points (0, 7, and 21 days) after osteogenic differentiation. The results showed that the PSC-derived EVs had much higher protein and lipid concentrations than EVs derived from BMSCs. The extracted EVs were characterized by observing their morphology and size distribution before utilizing next-generation sequencing to determine their microRNA (miRNA) profiles. A total of 306 miRNAs within the EVs were identified, of which 64 were significantly expressed in PSC-derived EVs that related to osteogenic differentiation. In vitro osteogenic differentiation study indicated the late-stage (21-day extracted)-derived EVs higher osteogenic enhancing capability when compared with the early stage-derived EVs. We demonstrated that EVs derived from PSCs could be a new source of EVs for bone tissue engineering applications.
Collapse
Affiliation(s)
- Elham Pishavar
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Joshua S Copus
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
- School of Biomedical Engineering and Sciences, Wake Forest University-Virginia Tech, Winston-Salem, North Carolina, USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
- School of Biomedical Engineering and Sciences, Wake Forest University-Virginia Tech, Winston-Salem, North Carolina, USA
| | - Sang Jin Lee
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
- School of Biomedical Engineering and Sciences, Wake Forest University-Virginia Tech, Winston-Salem, North Carolina, USA
| |
Collapse
|
3
|
Salehinejad P, Moshrefi M, Eslaminejad T. An Overview on Mesenchymal Stem Cells Derived from Extraembryonic Tissues: Supplement Sources and Isolation Methods. STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2020; 13:57-65. [PMID: 32753904 PMCID: PMC7354009 DOI: 10.2147/sccaa.s248519] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 06/25/2020] [Indexed: 11/23/2022]
Abstract
Purpose The main aim of this review was to provide an updated comprehensive report regarding isolation methods of MSCs from human extra embryonic tissues, including cord blood, amniotic fluid, and different parts of the placenta and umbilical cord, with respect to the efficacy of these methods. Results Extra embryonic tissues are the most available source for harvesting of mesenchymal stem cells (MSCs). They make a large number of cells accessible using non-invasive methods of isolation and the least immune-rejection reactions. A successful culture of primary cells requires obtaining a maximum yield of functional and viable cells from the tissues. In addition, there are many reports associated with their differentiation into various kinds of cells, and there are some clinical trials regarding their utilization for patients. Conclusion Currently, cord blood-MSCs have been tested for cartilage and lung diseases. Umbilical cord-MSCs were tested for liver and neural disorders. However, these MSCs can be isolated, expanded, and cryopreserved in a cell bank for patients in need.
Collapse
Affiliation(s)
- Parvin Salehinejad
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mojgan Moshrefi
- Medical Nanotechnology and Tissue Engineering Research Center, Yazd Reproductive Science Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Research and Clinical Center for Infertility, Yazd Reproductive Science Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Touba Eslaminejad
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
4
|
Nonoperative Applications of Placental Tissue Matrix in Orthopaedic Sports Injuries: A Review of Literature. Clin J Sport Med 2020; 30:383-389. [PMID: 30365472 DOI: 10.1097/jsm.0000000000000684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Recently, various amniotic tissue and placental-based tissue matrix (PTM) products have become increasingly available as a nonoperative treatment for tendinopathies and orthopaedic sports injuries. The aim of this review was to evaluate: (1) safety and efficacy of nonoperative use of PTM products, in acute and chronic tendon injuries and (2) the commercially available tissue options to better understand their differences. DATA SOURCES A comprehensive literature search was performed. Inclusion criteria were studies reporting on: (1) nonoperative uses of PTM therapy in sports injuries; and (2) clinical outcomes; in (3) human subjects. We excluded: (1) animal studies; (2) basic science studies; (3) non-English language literature; (4) review articles; and (5) duplicate studies. In addition, to determine the various product formulations, their tissue contents, and indications for use, we searched publicly available website content, marketing literature, and Food and Drug Administration (FDA) registration documents. MAIN RESULTS Current evidence investigated various PTM products for the treatment of various tendon injuries with demonstrated efficacy mainly in the short term with follow-up ranging between 6 weeks and 3 months. In addition, across all studies, no specific adverse events were reported. Substantial differences exist among the currently available products due to variations in their tissue source, formulations, processing methods, method of sterilization, preservation, and storage, indications for use, and FDA regulation. CONCLUSIONS Placental- and amniotic membrane-derived tissues seem to be safe for the nonoperative treatment of tendinopathies. However, several factors may affect the efficacy and safety profile of these products, and the orthopaedic surgeons should be aware of the differences.
Collapse
|
5
|
McIntyre JA, Jones IA, Han B, Vangsness CT. Intra-articular Mesenchymal Stem Cell Therapy for the Human Joint: A Systematic Review. Am J Sports Med 2018; 46:3550-3563. [PMID: 29099618 DOI: 10.1177/0363546517735844] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Stem cell therapy is emerging as a potential treatment of osteoarthritis (OA) and chondral defects (CDs). However, there is a great deal of heterogeneity in the literature. The indications for stem cell use, the ideal tissue source, and the preferred outcome measures for stem cell-based treatments have yet to be determined. PURPOSE To provide clinicians with a comprehensive overview of the entire body of the current human literature investigating the safety and efficacy of intra-articular mesenchymal stem cell (MSC) therapy in all joints. METHODS To provide a comprehensive overview of the current literature, all clinical studies investigating the safety and efficacy of intra-articular MSC therapy were included. PubMed, MEDLINE, and Cochrane Library databases were searched for published human clinical trials involving the use of MSCs for the treatment of OA and CDs in all joints. A total of 3867 publications were screened. RESULTS Twenty-eight studies met the criteria to be included in this review. Fourteen studies treating osteoarthritis and 14 studies treating focal chondral defects were included. MSCs originating from bone marrow (13), adipose tissue (12), synovial tissue (2), or peripheral blood (2) were administered to 584 distinct individuals. MSCs were administered into the knee (523 knees), foot/ankle (61), and hip (5). The mean follow-up time was 24.4 months after MSC therapy. All studies reported improvement from baseline in at least 1 clinical outcome measure, and no study reported major adverse events attributable to MSC therapy. DISCUSSION The studies included in this review suggest that intra-articular MSC therapy is safe. While clinical and, in some cases, radiological improvements were reported for both OA and CD trials, the overall quality of the literature was poor, and heterogeneity and lack of reproducibility limit firm conclusions regarding the efficacy of these treatments. CONCLUSION This review provides strong evidence that autologous intra-articular MSC therapy is safe, with generally positive clinical outcomes.
Collapse
Affiliation(s)
- James A McIntyre
- School of Medicine and Health Sciences, George Washington University, Washington, DC, USA
| | - Ian A Jones
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Bo Han
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - C Thomas Vangsness
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
6
|
Naseer N, Bashir S, Latief N, Latif F, Khan SN, Riazuddin S. Human amniotic membrane as differentiating matrix for in vitro chondrogenesis. Regen Med 2018; 13:821-832. [PMID: 30299207 DOI: 10.2217/rme-2018-0017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Aim: The aim of the present study is to use human amniotic membrane (HAM) for in vitro chondrogenesis of placenta-derived mesenchymal stem cells (MSCs) and umbilical cord-derived MSCs. Materials & methods: MSCs from the placenta and umbilical cord were isolated, characterized by immunophenotyping and after analyzing their rate of proliferation, cytotoxicity and viability, chondrogenesis was performed on plastic adherent surface and on HAM. Results: Successfully isolated and characterized placenta-derived MSCs and umbilical cord-derived MSCs revealed positive expression of MSCs markers CD90, CD73, CD105 and CD49d, while they were negative for CD45. Both types of cells in the presence of chondrogenic induction medium on plastic adherent surface and HAM showed aggregates of proteoglycan and strong expression of COL2A1 (collagen 2) and ACAN1 (aggrecan). Conclusion: HAM supported proliferation as well as chondrogenesis of MSCs and provide novelty of HAM utilization as an efficient natural delivery matrix for stem cell transplantation.
Collapse
Affiliation(s)
- Nadia Naseer
- Centre of Excellence in Molecular Biology, 87 West Canal Bank Road, Thokar Niazbaig Lahore, Punjab, 53700 Pakistan
| | - Saliha Bashir
- Centre of Excellence in Molecular Biology, 87 West Canal Bank Road, Thokar Niazbaig Lahore, Punjab, 53700 Pakistan
| | - Noreen Latief
- Centre of Excellence in Molecular Biology, 87 West Canal Bank Road, Thokar Niazbaig Lahore, Punjab, 53700 Pakistan
| | - Farzana Latif
- Ameer-ud-din Medical College, Post Graduate Medical Institute (PGMI), Lahore General Hospital, 6-Abdur Rehman Chughtai Road (Birdwood Road), Jail Road, Shadman, Lahore,54000 Pakistan
| | - Shaheen N Khan
- Centre of Excellence in Molecular Biology, 87 West Canal Bank Road, Thokar Niazbaig Lahore, Punjab, 53700 Pakistan
| | - Sheikh Riazuddin
- Centre of Excellence in Molecular Biology, 87 West Canal Bank Road, Thokar Niazbaig Lahore, Punjab, 53700 Pakistan
- Allama Iqbal Medical College, University of Health Sciences, Lahore, Jinnah hospital Moulana Shabir Ahmed Usmani Road, Faisal Town Lahore 54550 Pakistan
- Shaheed Zulfiqar Ali Bhutto Medical University (SZABMU), PIMS, G-8/3, Islamabad, 44000 Pakistan
| |
Collapse
|
7
|
McIntyre JA, Jones IA, Danilkovich A, Vangsness CT. The Placenta: Applications in Orthopaedic Sports Medicine. Am J Sports Med 2018; 46:234-247. [PMID: 28375638 DOI: 10.1177/0363546517697682] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Placenta has a long history of use for treating burns and wounds. It is a rich source of collagen and other extracellular matrix proteins, tissue reparative growth factors, and stem cells, including mesenchymal stem cells (MSCs). Recent data show its therapeutic potential for orthopaedic sports medicine indications. PURPOSE To provide orthopaedic surgeons with an anatomic description of the placenta, to characterize its cellular composition, and to review the literature reporting the use of placenta-derived cells and placental tissue allografts for orthopaedic sports medicine indications in animal models and in humans. STUDY DESIGN Systematic review. METHODS Using a total of 63 keyword combinations, the PubMed and MEDLINE databases were searched for published articles describing the use of placental cells and/or tissue for orthopaedic sports medicine indications. Information was collected on placental tissue type, indications, animal model, study design, treatment regimen, safety, and efficacy outcomes. Results were categorized by indication and subcategorized by animal model. RESULTS Outcomes for 29 animal studies and 6 human studies reporting the use of placenta-derived therapeutics were generally positive; however, the placental tissue source, clinical indication, and administration route were highly variable across these studies. Fourteen animal studies described the use of placental tissue for tendon injuries, 13 studies for osteoarthritis or articular cartilage injuries, 3 for ligament injuries, and 1 for synovitis. Both placenta-derived culture-expanded cells (epithelial cells or MSCs) and placental tissue allografts were used in animal studies. In all human studies, commercial placental allografts were used. Five of 6 human studies examined the treatment of foot and ankle pathological conditions, and 1 studied the treatment of knee osteoarthritis. CONCLUSION A review of the small number of reported studies revealed a high degree of variability in placental cell types, placental tissue preparation, routes of administration, and treatment regimens, which prohibits making any definitive conclusions. Currently, the clinical use of placenta is limited to only commercial placental tissue allografts, as there are no placenta-derived biological drugs approved for the treatment of orthopaedic sports medicine conditions in the United States. However, this review shows that the application of placental cells or tissue allografts appears to be safe and has potential to improve outcomes for orthopaedic sports medicine indications.
Collapse
Affiliation(s)
- James Alexander McIntyre
- School of Medicine & Health Sciences, George Washington University, Washington, District of Columbia, USA
| | - Ian A Jones
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | | | | |
Collapse
|
8
|
Jung JH, Kang KW, Kim J, Hong SC, Park Y, Kim BS. CXCR2 Inhibition in Human Pluripotent Stem Cells Induces Predominant Differentiation to Mesoderm and Endoderm Through Repression of mTOR, β-Catenin, and hTERT Activities. Stem Cells Dev 2017; 25:1006-19. [PMID: 27188501 PMCID: PMC4931345 DOI: 10.1089/scd.2015.0395] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
On the basis of our previous report verifying that chemokine (C-X-C motif) receptor 2 (CXCR2) ligands in human placenta-derived cell conditioned medium (hPCCM) support human pluripotent stem cell (hPSC) propagation without exogenous basic fibroblast growth factor (bFGF), this study was designed to identify the effect of CXCR2 manipulation on the fate of hPSCs and the underlying mechanism, which had not been previously determined. We observed that CXCR2 inhibition in hPSCs induces predominant differentiation to mesoderm and endoderm with concomitant loss of hPSC characteristics and accompanying decreased expression of mammalian target of rapamycin (mTOR), β-catenin, and human telomerase reverse transcriptase (hTERT). These phenomena are recapitulated in hPSCs propagated in conventional culture conditions, including bFGF as well as those in hPCCM without exogenous bFGF, suggesting that the action of CXCR2 on hPSCs might not be associated with a bFGF-related mechanism. In addition, the specific CXCR2 ligand growth-related oncogene α (GROα) markedly increased the expression of ectodermal markers in differentiation-committed embryoid bodies derived from hPSCs. This finding suggests that CXCR2 inhibition in hPSCs prohibits the propagation of hPSCs and leads to predominant differentiation to mesoderm and endoderm owing to the blockage of ectodermal differentiation. Taken together, our results indicate that CXCR2 preferentially supports the maintenance of hPSC characteristics as well as facilitates ectodermal differentiation after the commitment to differentiation, and the mechanism might be associated with mTOR, β-catenin, and hTERT activities.
Collapse
Affiliation(s)
- Ji-Hye Jung
- 1 Institute of Stem Cell Research, Korea University , Seoul, Korea.,2 Department of Biomedical Science, Graduate School of Medicine, Korea University , Seoul, Korea
| | - Ka-Won Kang
- 1 Institute of Stem Cell Research, Korea University , Seoul, Korea.,3 Department of Hematology/Oncology, Korea University Anam Medical Center , Seoul, Korea
| | - Jihea Kim
- 1 Institute of Stem Cell Research, Korea University , Seoul, Korea
| | - Soon-Chul Hong
- 4 Department of Obstetrics/Gynecology, Korea University Anam Medical Center , Seoul, Korea
| | - Yong Park
- 1 Institute of Stem Cell Research, Korea University , Seoul, Korea.,3 Department of Hematology/Oncology, Korea University Anam Medical Center , Seoul, Korea
| | - Byung Soo Kim
- 1 Institute of Stem Cell Research, Korea University , Seoul, Korea.,2 Department of Biomedical Science, Graduate School of Medicine, Korea University , Seoul, Korea.,3 Department of Hematology/Oncology, Korea University Anam Medical Center , Seoul, Korea
| |
Collapse
|
9
|
Clevenger TN, Luna G, Fisher SK, Clegg DO. Strategies for bioengineered scaffolds that support adipose stem cells in regenerative therapies. Regen Med 2016; 11:589-99. [PMID: 27484203 DOI: 10.2217/rme-2016-0064] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Regenerative medicine possesses the potential to ameliorate damage to tissue that results from a vast range of conditions, including traumatic injury, tumor resection and inherited tissue defects. Adult stem cells, while more limited in their potential than pluripotent stem cells, are still capable of differentiating into numerous lineages and provide feasible allogeneic and autologous treatment options for many conditions. Adipose stem cells are one of the most abundant types of stem cell in the adult human. Here, we review recent advances in the development of synthetic scaffolding systems used in concert with adipose stem cells and assess their potential use for clinical applications.
Collapse
Affiliation(s)
- Tracy N Clevenger
- Center for Stem Cell Biology & Engineering, University of California, Santa Barbara, CA, USA.,Neuroscience Research Institute, University of California, Santa Barbara, CA, USA.,Department of Molecular, Cellular & Developmental Biology, University of California, Santa Barbara, CA, USA.,Institute for Collaborative Biotechnologies, University of California, Santa Barbara, CA, USA
| | - Gabriel Luna
- Neuroscience Research Institute, University of California, Santa Barbara, CA, USA.,Center for Bio-Image Informatics, University of California, Santa Barbara, CA, USA
| | - Steven K Fisher
- Neuroscience Research Institute, University of California, Santa Barbara, CA, USA.,Center for Bio-Image Informatics, University of California, Santa Barbara, CA, USA
| | - Dennis O Clegg
- Center for Stem Cell Biology & Engineering, University of California, Santa Barbara, CA, USA.,Neuroscience Research Institute, University of California, Santa Barbara, CA, USA.,Department of Molecular, Cellular & Developmental Biology, University of California, Santa Barbara, CA, USA.,Institute for Collaborative Biotechnologies, University of California, Santa Barbara, CA, USA
| |
Collapse
|
10
|
Platelet-Rich Plasma Obtained with Different Anticoagulants and Their Effect on Platelet Numbers and Mesenchymal Stromal Cells Behavior In Vitro. Stem Cells Int 2016; 2016:7414036. [PMID: 27340410 PMCID: PMC4909912 DOI: 10.1155/2016/7414036] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 04/09/2016] [Accepted: 04/27/2016] [Indexed: 02/06/2023] Open
Abstract
There are promising results in the use of platelet-rich plasma (PRP) for musculoskeletal tissue repair. However, the variability in the methodology for its obtaining may cause different and opposing findings in the literature. Particularly, the choice of the anticoagulant is the first definition to be made. In this work, blood was collected with sodium citrate (SC), ethylenediaminetetraacetic acid (EDTA), or anticoagulant citrate dextrose (ACD) solution A, as anticoagulants, prior to PRP obtaining. Hematological analysis and growth factors release quantification were performed, and the effects on mesenchymal stromal cell (MSC) culture, such as cytotoxicity and cell proliferation (evaluated by MTT method) and gene expression, were evaluated. The use of EDTA resulted in higher platelet yield in whole blood; however, it induced an increase in the mean platelet volume (MPV) following the blood centrifugation steps for PRP obtaining. The use of SC and ACD resulted in higher induction of MSC proliferation. On the other hand, PRP obtained in SC presented the higher platelet recovery after the blood first centrifugation step and a minimal change in MSC gene expression. Therefore, we suggest the use of SC as the anticoagulant for PRP obtaining.
Collapse
|
11
|
Current View on Osteogenic Differentiation Potential of Mesenchymal Stromal Cells Derived from Placental Tissues. Stem Cell Rev Rep 2016; 11:570-85. [PMID: 25381565 PMCID: PMC4493719 DOI: 10.1007/s12015-014-9569-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Mesenchymal stromal cells (MSC) isolated from human term placental tissues possess unique characteristics, including their peculiar immunomodulatory properties and their multilineage differentiation potential. The osteogenic differentiation capacity of placental MSC has been widely disputed, and continues to be an issue of debate. This review will briefly discuss the different MSC populations which can be obtained from different regions of human term placenta, along with their unique properties, focusing specifically on their osteogenic differentiation potential. We will present the strategies used to enhance osteogenic differentiation potential in vitro, such as through the selection of subpopulations more prone to differentiate, the modification of the components of osteo-inductive medium, and even mechanical stimulation. Accordingly, the applications of three-dimensional environments in vitro and in vivo, such as non-synthetic, polymer-based, and ceramic scaffolds, will also be discussed, along with results obtained from pre-clinical studies of placental MSC for the regeneration of bone defects and treatment of bone-related diseases.
Collapse
|
12
|
Montemurro T, Viganò M, Ragni E, Barilani M, Parazzi V, Boldrin V, Lavazza C, Montelatici E, Banfi F, Lauri E, Giovanelli S, Baccarin M, Guerneri S, Giordano R, Lazzari L. Angiogenic and anti-inflammatory properties of mesenchymal stem cells from cord blood: soluble factors and extracellular vesicles for cell regeneration. Eur J Cell Biol 2016; 95:228-38. [PMID: 27139721 DOI: 10.1016/j.ejcb.2016.04.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 02/25/2016] [Accepted: 04/06/2016] [Indexed: 12/18/2022] Open
Abstract
In a recent work, our group showed the existence of two distinct mesenchymal stem cell (MSC) subsets within human umbilical cord blood. One less proliferative and short-living (SL-CBMSC), the other with higher growth rate and long-living (LL-CBMSC), and therefore better suited for regenerative medicine applications. We examined whether LL-CBMSC possess peculiar paracrine properties able to affect angiogenesis or inflammatory processes. It was shown for the first time that pro-angiogenic, proliferation-stimulating and tissue repairing factors were released at high level not only as soluble cytokines, but also as mRNA precursors embedded in membrane vesicles. The combination of this primary (proteic factors interacting with surface receptors) and delayed (mRNA transferred and translated via vesicle fusion and cargo release) interaction in endothelial target cells resulted in strong blood vessel induction with the development of capillary-like structures. In addition, LL-CBMSC dynamically modulated their release of pro-angiogenic and anti-inflammatory factors in an in vitro model of damage. In conclusion, LL-CBMSC synthesize and secrete multiple factors that may be attuned in response to the status of the target cell, a crucial requisite when paracrine mechanisms are needed at onset of tissue regeneration.
Collapse
Affiliation(s)
- Tiziana Montemurro
- Unit of Cell Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Mariele Viganò
- Unit of Cell Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Enrico Ragni
- Unit of Cell Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Mario Barilani
- Unit of Cell Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Valentina Parazzi
- Unit of Cell Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Valentina Boldrin
- Unit of Cell Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Cristiana Lavazza
- Unit of Cell Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Elisa Montelatici
- Unit of Cell Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Federica Banfi
- Unit of Cell Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Eleonora Lauri
- Anatomia Patologica, Ospedale Sacco, Università degli Studi di Milano, Milano, Italy
| | - Silvia Giovanelli
- Milano Cord Blood Bank, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Marco Baccarin
- Laboratorio Citogenetica e Genetica molecolare, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Silvana Guerneri
- Laboratorio Citogenetica e Genetica molecolare, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Rosaria Giordano
- Unit of Cell Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Lorenza Lazzari
- Unit of Cell Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy.
| |
Collapse
|
13
|
Qin SQ, Kusuma GD, Al-Sowayan B, Pace RA, Isenmann S, Pertile MD, Gronthos S, Abumaree MH, Brennecke SP, Kalionis B. Establishment and characterization of fetal and maternal mesenchymal stem/stromal cell lines from the human term placenta. Placenta 2016; 39:134-46. [DOI: 10.1016/j.placenta.2016.01.018] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 01/19/2016] [Accepted: 01/21/2016] [Indexed: 12/31/2022]
|
14
|
Antoniadou E, David AL. Placental stem cells. Best Pract Res Clin Obstet Gynaecol 2015; 31:13-29. [PMID: 26547389 DOI: 10.1016/j.bpobgyn.2015.08.014] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 08/31/2015] [Indexed: 12/14/2022]
Abstract
The placenta represents a reservoir of progenitor, stem cells and epithelial cells that have been shown to differentiate into various types, including adipogenic, osteogenic, myogenic, hepatogenic, cardiac, pancreatic, endothelial, pulmonary and neurogenic lineages. This review focuses on the properties of placenta-derived cells, and it evaluates their current therapeutic applications in regenerative medicine and cell transplantations. Ongoing clinical and preclinical studies are investigating the safety and efficacy of the human amniotic epithelial cells (hAECs), human amniotic mesenchymal stromal cells (hAMSCs) and chorionic mesenchymal stromal cells (hCMSCs). The establishment of biobanks for placental stem cells will enable the translation of scientific research into the clinic. The advantage of the placenta as a cellular source is that it contains different cell lineages, such as the haematopoietic lineage that originates from the chorion, allantois and yolk sac, and the mesenchymal lineage that originates from the chorion and amnion. In this review, we address advances in placental stem cell characterization, and we explore their possible uses in cell therapy.
Collapse
Affiliation(s)
- Eleni Antoniadou
- Stem Cells and Regenerative Medicine Section, Developmental Biology and Cancer Programme, UCL Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK.
| | - Anna L David
- Institute for Women's Health, University College London, 86-96 Chenies Mews, London WC1E 6HX, UK.
| |
Collapse
|
15
|
Placenta as a Source of Stem Cells for Regenerative Medicine. CURRENT PATHOBIOLOGY REPORTS 2015. [DOI: 10.1007/s40139-015-0070-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
16
|
Resca E, Zavatti M, Maraldi T, Bertoni L, Beretti F, Guida M, La Sala G, Guillot P, David A, Sebire N, De Pol A, De Coppi P. Enrichment in c-Kit improved differentiation potential of amniotic membrane progenitor/stem cells. Placenta 2015; 36:18-26. [DOI: 10.1016/j.placenta.2014.11.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 10/31/2014] [Accepted: 11/03/2014] [Indexed: 12/15/2022]
|
17
|
Garg RK, Rennert RC, Duscher D, Sorkin M, Kosaraju R, Auerbach LJ, Lennon J, Chung MT, Paik K, Nimpf J, Rajadas J, Longaker MT, Gurtner GC. Capillary force seeding of hydrogels for adipose-derived stem cell delivery in wounds. Stem Cells Transl Med 2014; 3:1079-89. [PMID: 25038246 DOI: 10.5966/sctm.2014-0007] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Effective skin regeneration therapies require a successful interface between progenitor cells and biocompatible delivery systems. We previously demonstrated the efficiency of a biomimetic pullulan-collagen hydrogel scaffold for improving bone marrow-derived mesenchymal stem cell survival within ischemic skin wounds by creating a "stem cell niche" that enhances regenerative cytokine secretion. Adipose-derived mesenchymal stem cells (ASCs) represent an even more appealing source of stem cells because of their abundance and accessibility, and in this study we explored the utility of ASCs for hydrogel-based therapies. To optimize hydrogel cell seeding, a rapid, capillary force-based approach was developed and compared with previously established cell seeding methods. ASC viability and functionality following capillary hydrogel seeding were then analyzed in vitro and in vivo. In these experiments, ASCs were seeded more efficiently by capillary force than by traditional methods and remained viable and functional in this niche for up to 14 days. Additionally, hydrogel seeding of ASCs resulted in the enhanced expression of multiple stemness and angiogenesis-related genes, including Oct4, Vegf, Mcp-1, and Sdf-1. Moving in vivo, hydrogel delivery improved ASC survival, and application of both murine and human ASC-seeded hydrogels to splinted murine wounds resulted in accelerated wound closure and increased vascularity when compared with control wounds treated with unseeded hydrogels. In conclusion, capillary seeding of ASCs within a pullulan-collagen hydrogel bioscaffold provides a convenient and simple way to deliver therapeutic cells to wound environments. Moreover, ASC-seeded constructs display a significant potential to accelerate wound healing that can be easily translated to a clinical setting.
Collapse
Affiliation(s)
- Ravi K Garg
- Hagey Laboratory, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA; Max F. Perutz Laboratories, Department of Medical Biochemistry and Molecular Biology, Medical University of Vienna, Vienna, Austria; Biomaterials and Advanced Drug Delivery Center, Stanford University, Stanford, California, USA
| | - Robert C Rennert
- Hagey Laboratory, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA; Max F. Perutz Laboratories, Department of Medical Biochemistry and Molecular Biology, Medical University of Vienna, Vienna, Austria; Biomaterials and Advanced Drug Delivery Center, Stanford University, Stanford, California, USA
| | - Dominik Duscher
- Hagey Laboratory, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA; Max F. Perutz Laboratories, Department of Medical Biochemistry and Molecular Biology, Medical University of Vienna, Vienna, Austria; Biomaterials and Advanced Drug Delivery Center, Stanford University, Stanford, California, USA
| | - Michael Sorkin
- Hagey Laboratory, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA; Max F. Perutz Laboratories, Department of Medical Biochemistry and Molecular Biology, Medical University of Vienna, Vienna, Austria; Biomaterials and Advanced Drug Delivery Center, Stanford University, Stanford, California, USA
| | - Revanth Kosaraju
- Hagey Laboratory, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA; Max F. Perutz Laboratories, Department of Medical Biochemistry and Molecular Biology, Medical University of Vienna, Vienna, Austria; Biomaterials and Advanced Drug Delivery Center, Stanford University, Stanford, California, USA
| | - Lauren J Auerbach
- Hagey Laboratory, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA; Max F. Perutz Laboratories, Department of Medical Biochemistry and Molecular Biology, Medical University of Vienna, Vienna, Austria; Biomaterials and Advanced Drug Delivery Center, Stanford University, Stanford, California, USA
| | - James Lennon
- Hagey Laboratory, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA; Max F. Perutz Laboratories, Department of Medical Biochemistry and Molecular Biology, Medical University of Vienna, Vienna, Austria; Biomaterials and Advanced Drug Delivery Center, Stanford University, Stanford, California, USA
| | - Michael T Chung
- Hagey Laboratory, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA; Max F. Perutz Laboratories, Department of Medical Biochemistry and Molecular Biology, Medical University of Vienna, Vienna, Austria; Biomaterials and Advanced Drug Delivery Center, Stanford University, Stanford, California, USA
| | - Kevin Paik
- Hagey Laboratory, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA; Max F. Perutz Laboratories, Department of Medical Biochemistry and Molecular Biology, Medical University of Vienna, Vienna, Austria; Biomaterials and Advanced Drug Delivery Center, Stanford University, Stanford, California, USA
| | - Johannes Nimpf
- Hagey Laboratory, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA; Max F. Perutz Laboratories, Department of Medical Biochemistry and Molecular Biology, Medical University of Vienna, Vienna, Austria; Biomaterials and Advanced Drug Delivery Center, Stanford University, Stanford, California, USA
| | - Jayakumar Rajadas
- Hagey Laboratory, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA; Max F. Perutz Laboratories, Department of Medical Biochemistry and Molecular Biology, Medical University of Vienna, Vienna, Austria; Biomaterials and Advanced Drug Delivery Center, Stanford University, Stanford, California, USA
| | - Michael T Longaker
- Hagey Laboratory, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA; Max F. Perutz Laboratories, Department of Medical Biochemistry and Molecular Biology, Medical University of Vienna, Vienna, Austria; Biomaterials and Advanced Drug Delivery Center, Stanford University, Stanford, California, USA
| | - Geoffrey C Gurtner
- Hagey Laboratory, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA; Max F. Perutz Laboratories, Department of Medical Biochemistry and Molecular Biology, Medical University of Vienna, Vienna, Austria; Biomaterials and Advanced Drug Delivery Center, Stanford University, Stanford, California, USA
| |
Collapse
|
18
|
Qu H, Liu X, Ni Y, Jiang Y, Feng X, Xiao J, Guo Y, Kong D, Li A, Li X, Zhuang X, Wang Z, Wang Y, Chang Y, Chen S, Kong F, Zhang X, Zhao S, Sun Y, Xu D, Wang D, Zheng C. Laminin 411 acts as a potent inducer of umbilical cord mesenchymal stem cell differentiation into insulin-producing cells. J Transl Med 2014; 12:135. [PMID: 24885418 PMCID: PMC4040110 DOI: 10.1186/1479-5876-12-135] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 05/13/2014] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Diabetes mellitus (DM) is an incurable metabolic disease constituting a major threat to human health. Insulin-producing cells (IPCs) differentiated from mesenchymal stem cells (MSCs) hold great promise in the treatment of DM. The development of an efficient IPC induction system is a crucial step for the clinical application of IPCs for DM. Laminin 411 is a key component of the basement membrane and is involved in the regulation of cell differentiation; however, little is known about a role of laminin 411 in the regulation of IPC differentiation from human MSCs. METHODS MSCs were isolated from human umbilical cord (UC-MSCs) and expanded in an in vitro culture system. UC-MSCs were then cultured in the IPC induction and differentiation medium in the presence of laminin 411. Flow cytometry, Quantitative realtime PCR, immunofluorescence staining, ELISA, Western blotting and other techniques were applied to determine IPC generation, insulin expression and related mechanisms. To evaluate potential therapeutic efficacy of IPCs induced from UC-MSCs, a type-1 diabetes (T1DM) rat model was generated using streptozotocin. Blood glucose, insulin levels, and survival of rats were monitored periodically following intravenous injection of the tested cells. RESULTS Laminin 411 markedly induced the expression of the genes Foxa2 and Sox17, markers for pancreatic precursor cells, efficiently induced IPC differentiation from MSCs, and up-regulated insulin expression at both mRNA and protein levels. Furthermore, the expression of the genes known to govern insulin expression including Pdx1 and Ngn3 was markedly induced by laminin 411, which suggests that Pdx1 and Ngn3 signaling pathways are involved in laminin 411 induced-insulin expression machinery. More importantly, administration of laminin 411-induced IPCs rapidly and significantly down-regulated fasting blood glucose levels, significantly reduced the HbA1c concentration and markedly improved the symptoms and survival of T1DM rats. CONCLUSIONS Our results demonstrate that laminin 411 acts as a potent differentiation inducer of IPCs from UC-MSCs via the Pdx1 and Ngn3 signaling pathways. Moreover, transfusion of laminin 411 induced-IPCs more efficiently improves symptoms and survival of T1DM rats. These novel finding highlights a potential clinical application of laminin 411 induced-IPCs in the treatment of T1DM, which calls for further studies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Daoqing Wang
- Departments of Hematology and Cellular Therapy, the Second Hospital of Shandong University, Jinan, Shandong, PR China.
| | | |
Collapse
|
19
|
Liu H, Murthi P, Qin S, Kusuma GD, Borg AJ, Knöfler M, Haslinger P, Manuelpillai U, Pertile MD, Abumaree M, Kalionis B. A novel combination of homeobox genes is expressed in mesenchymal chorionic stem/stromal cells in first trimester and term pregnancies. Reprod Sci 2014; 21:1382-94. [PMID: 24692208 DOI: 10.1177/1933719114526471] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Human chorionic mesenchymal stem/stromal cells (CMSCs) derived from the placenta are similar to adult tissue-derived MSCs. The aim of this study was to investigate the role of these cells in normal placental development. Transcription factors, particularly members of the homeobox gene family, play crucial roles in maintaining stem cell proliferation and lineage specification in embryonic tissues. In adult tissues and organs, stem cells proliferate at low levels in their niche until they receive cues from the microenvironment to differentiate. The homeobox genes that are expressed in the CMSC niche in placental tissues have not been identified. We used the novel strategy of laser capture microdissection to isolate the stromal component of first trimester villi and excluded the cytotrophoblast and syncytiotrophoblast layers that comprise the outer layer of the chorionic villi. Microarray analysis was then used to screen for homeobox genes in the microdissected tissue. Candidate homeobox genes were selected for further RNA analysis. Immunohistochemistry of candidate genes in first trimester placental villous stromal tissue revealed homeobox genes Meis1, myeloid ectropic viral integration site 1 homolog 2 (MEIS2), H2.0-like Drosophila (HLX), transforming growth factor β-induced factor (TGIF), and distal-less homeobox 5 (DLX5) were expressed in the vascular niche where CMSCs have been shown to reside. Expression of MEIS2, HLX, TGIF, and DLX5 was also detected in scattered stromal cells. Real-time polymerase chain reaction and immunocytochemistry verified expression of MEIS2, HLX, TGIF, and DLX5 homeobox genes in first trimester and term CMSCs. These data suggest a combination of regulatory homeobox genes is expressed in CMSCs from early placental development to term, which may be required for stem cell proliferation and differentiation.
Collapse
Affiliation(s)
- Haiying Liu
- Department of Obstetrics and Gynaecology, QiLu Hospital of Shandong University, Jinan, Shandong, P.R. China
| | - Padma Murthi
- Department of Obstetrics and Gynaecology, University of Melbourne, The Royal Women's Hospital, Parkville, Victoria, Australia Department of Perinatal Medicine, Pregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, Australia
| | - Sharon Qin
- Department of Obstetrics and Gynaecology, University of Melbourne, The Royal Women's Hospital, Parkville, Victoria, Australia Department of Perinatal Medicine, Pregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, Australia
| | - Gina D Kusuma
- Department of Obstetrics and Gynaecology, University of Melbourne, The Royal Women's Hospital, Parkville, Victoria, Australia Department of Perinatal Medicine, Pregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, Australia
| | - Anthony J Borg
- Department of Perinatal Medicine, Pregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, Australia
| | - Martin Knöfler
- Department of Obstetrics and Fetal-Maternal Medicine, Reproductive Biology Unit, Medical University of Vienna, Vienna, Austria
| | - Peter Haslinger
- Department of Obstetrics and Fetal-Maternal Medicine, Reproductive Biology Unit, Medical University of Vienna, Vienna, Austria
| | - Ursula Manuelpillai
- Centre for Genetic Diseases, Monash Institute of Medical Research, Monash University, Clayton, Victoria
| | - Mark D Pertile
- VCGS, Murdoch Children's Research Institute, Royal Childrens Hospital, Flemington Road, Parkville, Victoria, Australia
| | - Mohamed Abumaree
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences/ King Abdulla International Medical Research Center, Riyadh, Saudi Arabia
| | - Bill Kalionis
- Department of Obstetrics and Gynaecology, University of Melbourne, The Royal Women's Hospital, Parkville, Victoria, Australia Department of Perinatal Medicine, Pregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, Australia
| |
Collapse
|
20
|
Ryan JM, Pettit AR, Guillot PV, Chan JKY, Fisk NM. Unravelling the pluripotency paradox in fetal and placental mesenchymal stem cells: Oct-4 expression and the case of The Emperor's New Clothes. Stem Cell Rev Rep 2014; 9:408-21. [PMID: 22161644 DOI: 10.1007/s12015-011-9336-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem cells (MSC) from fetal-placental tissues have translational advantages over their adult counterparts, and have variably been reported to express pluripotency markers. OCT-4 expression in fetal-placental MSC has been documented in some studies, paradoxically without tumourogenicity in vivo. It is possible that OCT-4 expression is insufficient to induce true "stemness", but this issue is important for the translational safety of fetal-derived MSC. To clarify this, we undertook a systematic literature review on OCT-4 in fetal or adnexal MSC to show that most studies report OCT-4 message or protein expression, but no study provides definitive evidence of true OCT-4A expression. Discrepant findings were attributable not to different culture conditions, tissue sources, or gestational ages but instead to techniques used. In assessing OCT-4 as a pluripotency marker, we highlight the challenges in detecting the correct OCT-4 isoform (OCT-4A) associated with pluripotency. Although specific detection of OCT-4A mRNA is achievable, it appears unlikely that any antibody can reliably distinguish between OCT-4A and the pseudogene OCT-4B. Finally, using five robust techniques we demonstrate that fetal derived-MSC do not express OCT-4A (or by default OCT-4B). Reports suggesting OCT-4 expression in fetal-derived MSC warrant reassessment, paying attention to gene and protein isoforms, pseudogenes, and antibody choice as well as primer design. Critical examination of the OCT-4 literature leads us to suggest that OCT-4 expression in fetal MSC may be a case of "The Emperor's New Clothes" with early reports of (false) positive expression amplified in subsequent studies without critical attention to emerging refinements in knowledge and methodology.
Collapse
Affiliation(s)
- Jennifer M Ryan
- UQ Centre for Clinical Research, University of Queensland, Herston campus, Brisbane 4029, Australia.
| | | | | | | | | |
Collapse
|
21
|
Feng Y, Yu HM, Shang DS, Fang WG, He ZY, Chen YH. The involvement of CXCL11 in bone marrow-derived mesenchymal stem cell migration through human brain microvascular endothelial cells. Neurochem Res 2014; 39:700-6. [PMID: 24526602 DOI: 10.1007/s11064-014-1257-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 02/04/2014] [Accepted: 02/07/2014] [Indexed: 12/21/2022]
Abstract
Bone marrow-derived mesenchymal stem cells (MSCs) transplant into the brain, where they play a potential therapeutic role in neurological diseases. However, the blood-brain barrier (BBB) is a native obstacle for MSCs entry into the brain. Little is known about the mechanism behind MSCs migration across the BBB. In the present study, we modeled the interactions between human MSCs (hMSCs) and human brain microvascular endothelial cells (HBMECs) to mimic the BBB microenvironment. Real-time PCR analysis indicated that the chemokine CXCL11 is produced by hMSCs and the chemokine receptor CXCR3 is expressed on HBMECs. Further results indicate that CXCL11 secreted by hMSCs may interact with CXCR3 on HBMECs to induce the disassembly of tight junctions through the activation of ERK1/2 signaling in the endothelium, which promotes MSCs transendothelial migration. These findings are relevant for understanding the biological responses of MSCs in BBB environments and helpful for the application of MSCs in neurological diseases.
Collapse
Affiliation(s)
- Yu Feng
- Department of Neurology, The First Hospital of China Medical University, 155 Nan Jing Northern Street, Shenyang, 110001, Liaoning, China
| | | | | | | | | | | |
Collapse
|
22
|
Can we fix it? Evaluating the potential of placental stem cells for the treatment of pregnancy disorders. Placenta 2014; 35:77-84. [DOI: 10.1016/j.placenta.2013.12.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 12/18/2013] [Accepted: 12/22/2013] [Indexed: 12/14/2022]
|
23
|
Bhonde RR, Sheshadri P, Sharma S, Kumar A. Making surrogate β-cells from mesenchymal stromal cells: perspectives and future endeavors. Int J Biochem Cell Biol 2013; 46:90-102. [PMID: 24275096 DOI: 10.1016/j.biocel.2013.11.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 10/29/2013] [Accepted: 11/05/2013] [Indexed: 02/06/2023]
Abstract
Generation of surrogate β-cells is the need of the day to compensate the short supply of islets for transplantation to diabetic patients requiring daily shots of insulin. Over the years several sources of stem cells have been claimed to cater to the need of insulin producing cells. These include human embryonic stem cells, induced pluripotent stem cells, human perinatal tissues such as amnion, placenta, umbilical cord and postnatal tissues involving adipose tissue, bone marrow, blood monocytes, cord blood, dental pulp, endometrium, liver, labia minora dermis-derived fibroblasts and pancreas. Despite the availability of such heterogonous sources, there is no substantial breakthrough in selecting and implementing an ideal source for generating large number of stable insulin producing cells. Although the progress in derivation of β-cell like cells from embryonic stem cells has taken a greater leap, their application is limited due to controversy surrounding the destruction of human embryo and immune rejection. Since multipotent mesenchymal stromal cells are free of ethical and immunological complications, they could provide unprecedented opportunity as starting material to derive insulin secreting cells. The main focus of this review is to discuss the merits and demerits of MSCs obtained from human peri- and post-natal tissue sources to yield abundant glucose responsive insulin producing cells as ideal candidates for prospective stem cell therapy to treat diabetes.
Collapse
Affiliation(s)
- Ramesh R Bhonde
- Manipal Institute of Regenerative Medicine, GKVK Post, Alalsandra, Yelahanka, Bangalore 560065, India
| | - Preethi Sheshadri
- Manipal Institute of Regenerative Medicine, GKVK Post, Alalsandra, Yelahanka, Bangalore 560065, India
| | - Shikha Sharma
- Manipal Institute of Regenerative Medicine, GKVK Post, Alalsandra, Yelahanka, Bangalore 560065, India
| | - Anujith Kumar
- Manipal Institute of Regenerative Medicine, GKVK Post, Alalsandra, Yelahanka, Bangalore 560065, India.
| |
Collapse
|
24
|
Resca E, Zavatti M, Bertoni L, Maraldi T, De Biasi S, Pisciotta A, Nicoli A, La Sala G, Guillot P, David A, Sebire N, De Coppi P, De Pol A. Enrichment in c-Kit+ enhances mesodermal and neural differentiation of human chorionic placental cells. Placenta 2013; 34:526-35. [DOI: 10.1016/j.placenta.2013.03.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Revised: 03/25/2013] [Accepted: 03/27/2013] [Indexed: 01/15/2023]
|
25
|
Jones GN, Moschidou D, Puga-Iglesias TI, Kuleszewicz K, Vanleene M, Shefelbine SJ, Bou-Gharios G, Fisk NM, David AL, De Coppi P, Guillot PV. Ontological differences in first compared to third trimester human fetal placental chorionic stem cells. PLoS One 2012; 7:e43395. [PMID: 22962584 PMCID: PMC3433473 DOI: 10.1371/journal.pone.0043395] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Accepted: 07/19/2012] [Indexed: 12/16/2022] Open
Abstract
Human mesenchymal stromal/stem cells (MSC) isolated from fetal tissues hold promise for use in tissue engineering applications and cell-based therapies, but their collection is restricted ethically and technically. In contrast, the placenta is a potential source of readily-obtainable stem cells throughout pregnancy. In fetal tissues, early gestational stem cells are known to have advantageous characteristics over neonatal and adult stem cells. Accordingly, we investigated whether early fetal placental chorionic stem cells (e-CSC) were physiologically superior to their late gestation fetal chorionic counterparts (l-CSC). We showed that e-CSC shared a common phenotype with l-CSC, differentiating down the osteogenic, adipogenic and neurogenic pathways, and containing a subset of cells endogenously expressing NANOG, SOX2, c-MYC, and KLF4, as well as an array of genes expressed in pluripotent stem cells and primordial germ cells, including CD24, NANOG, SSEA4, SSEA3, TRA-1-60, TRA-1-81, STELLA, FRAGILIS, NANOS3, DAZL and SSEA1. However, we showed that e-CSC have characteristics of an earlier state of stemness compared to l-CSC, such as smaller size, faster kinetics, uniquely expressing OCT4A variant 1 and showing higher levels of expression of NANOG, SOX2, c-MYC and KLF4 than l-CSC. Furthermore e-CSC, but not l-CSC, formed embryoid bodies containing cells from the three germ layer lineages. Finally, we showed that e-CSC demonstrate higher tissue repair in vivo; when transplanted in the osteogenesis imperfecta mice, e-CSC, but not l-CSC increased bone quality and plasticity; and when applied to a skin wound, e-CSC, but not l-CSC, accelerated healing compared to controls. Our results provide insight into the ontogeny of the stemness phenotype during fetal development and suggest that the more primitive characteristics of early compared to late gestation fetal chorionic stem cells may be translationally advantageous.
Collapse
Affiliation(s)
- Gemma N. Jones
- Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom
| | - Dafni Moschidou
- Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom
| | | | - Katarzyna Kuleszewicz
- Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom
| | - Maximilien Vanleene
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | | | - George Bou-Gharios
- Kennedy Institute of Rheumatology, University of Oxford, London, United Kingdom
| | - Nicholas M. Fisk
- UQ Centre for Clinical Research, University of Queensland, Brisbane, Queensland, Australia
| | - Anna L. David
- Prenatal Cell and Gene Therapy Group, Institute for Women's Health, University College London, London, United Kingdom
| | - Paolo De Coppi
- Surgery Unit, UCL Institute of Child Health, London, United Kingdom
| | - Pascale V. Guillot
- Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
26
|
Koo BK, Park IY, Kim J, Kim JH, Kwon A, Kim M, Kim Y, Shin JC, Kim JH. Isolation and characterization of chorionic mesenchymal stromal cells from human full term placenta. J Korean Med Sci 2012; 27:857-63. [PMID: 22876050 PMCID: PMC3410231 DOI: 10.3346/jkms.2012.27.8.857] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Accepted: 05/17/2012] [Indexed: 01/05/2023] Open
Abstract
This study focused on the characterization of mesenchymal stromal cells (MSCs) from the chorion of human full term placenta from 15 donors. Chorionic MSCs revealed homologous fibroblast-like morphology and expressed CD73, CD29, CD105, and CD90. The hematopoietic stem cell markers including HLA DR, CD11b, CD34, CD79a, and CD45 were not expressed. The growth kinetics of their serial passage was steady at the later passages (passage 10). The multilineage capability of chorionic MSCs was demonstrated by successful adipogenic, osteogenic and chondrogenic differentiation and associated gene expression. Chorionic MSCs expressed genes associated with undifferentiated cells (NANOG, OCT4, REX1) and cardiogenic or neurogenic markers such as SOX2, FGF4, NES, MAP2, and NF. TERT was negative in all the samples. These findings suggest that chorionic MSCs undifferentiated stem cells and less likely to be transformed into cancer cells. A low HLA DR expression suggests that chorionic MSCs may serve as a great source of stem cells for transplantation because of their immune-privileged status and their immunosuppressive effect. Based on these unique properties, it is concluded that chorionic MSCs are pluripotent stem cells that are probably less differentiated than BM-MSCs, and they have considerable potential for use in cell-based therapies.
Collapse
Affiliation(s)
- Bo Kyung Koo
- Department of Laboratory Medicine, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - In Yang Park
- Department of Obstetrics and Gynecology, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Jiyeon Kim
- Department of Laboratory Medicine, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Ji-Hyun Kim
- Department of Laboratory Medicine, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Ahlm Kwon
- Department of Laboratory Medicine, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Myungshin Kim
- Department of Laboratory Medicine, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Yonggoo Kim
- Department of Laboratory Medicine, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Jong Chul Shin
- Department of Obstetrics and Gynecology, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Jong-Hoon Kim
- Laboratory of Stem Cell Biology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| |
Collapse
|