1
|
Ebner-Peking P, Krisch L, Wolf M, Hochmann S, Hoog A, Vári B, Muigg K, Poupardin R, Scharler C, Schmidhuber S, Russe E, Stachelscheid H, Schneeberger A, Schallmoser K, Strunk D. Self-assembly of differentiated progenitor cells facilitates spheroid human skin organoid formation and planar skin regeneration. Theranostics 2021; 11:8430-8447. [PMID: 34373751 PMCID: PMC8344006 DOI: 10.7150/thno.59661] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 07/02/2021] [Indexed: 01/01/2023] Open
Abstract
Self-assembly of solid organs from single cells would greatly expand applicability of regenerative medicine. Stem/progenitor cells can self-organize into micro-sized organ units, termed organoids, partially modelling tissue function and regeneration. Here we demonstrated 3D self-assembly of adult and induced pluripotent stem cell (iPSC)-derived fibroblasts, keratinocytes and endothelial progenitors into both, planar human skin in vivo and a novel type of spheroid-shaped skin organoids in vitro, under the aegis of human platelet lysate. Methods: Primary endothelial colony forming cells (ECFCs), skin fibroblasts (FBs) and keratinocytes (KCs) were isolated from human tissues and polyclonally propagated under 2D xeno-free conditions. Human tissue-derived iPSCs were differentiated into endothelial cells (hiPSC-ECs), fibroblasts (hiPSC-FBs) and keratinocytes (hiPSC-KCs) according to efficiency-optimized protocols. Cell identity and purity were confirmed by flow cytometry and clonogenicity indicated their stem/progenitor potential. Triple cell type floating spheroids formation was promoted by human platelet-derived growth factors containing culture conditions, using nanoparticle cell labelling for monitoring the organization process. Planar human skin regeneration was assessed in full-thickness wounds of immune-deficient mice upon transplantation of hiPSC-derived single cell suspensions. Results: Organoids displayed a distinct architecture with surface-anchored keratinocytes surrounding a stromal core, and specific signaling patterns in response to inflammatory stimuli. FGF-7 mRNA transfection was required to accelerate keratinocyte long-term fitness. Stratified human skin also self-assembled within two weeks after either adult- or iPSC-derived skin cell-suspension liquid-transplantation, healing deep wounds of mice. Transplant vascularization significantly accelerated in the presence of co-transplanted endothelial progenitors. Mechanistically, extracellular vesicles mediated the multifactorial platelet-derived trophic effects. No tumorigenesis occurred upon xenografting. Conclusion: This illustrates the superordinate progenitor self-organization principle and permits novel rapid 3D skin-related pharmaceutical high-content testing opportunities with floating spheroid skin organoids. Multi-cell transplant self-organization facilitates development of iPSC-based organ regeneration strategies using cell suspension transplantation supported by human platelet factors.
Collapse
Affiliation(s)
- Patricia Ebner-Peking
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), University Clinic, Paracelsus Medical University, Salzburg, Austria
| | - Linda Krisch
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), University Clinic, Paracelsus Medical University, Salzburg, Austria
- Department of Transfusion Medicine, University Clinic, Paracelsus Medical University, Salzburg, Austria
| | - Martin Wolf
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), University Clinic, Paracelsus Medical University, Salzburg, Austria
| | - Sarah Hochmann
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), University Clinic, Paracelsus Medical University, Salzburg, Austria
| | - Anna Hoog
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), University Clinic, Paracelsus Medical University, Salzburg, Austria
| | - Balázs Vári
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), University Clinic, Paracelsus Medical University, Salzburg, Austria
| | - Katharina Muigg
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), University Clinic, Paracelsus Medical University, Salzburg, Austria
| | - Rodolphe Poupardin
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), University Clinic, Paracelsus Medical University, Salzburg, Austria
| | - Cornelia Scharler
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), University Clinic, Paracelsus Medical University, Salzburg, Austria
| | | | - Elisabeth Russe
- Department of Plastic, Aesthetic and Reconstructive Surgery, Hospital Barmherzige Brueder, Salzburg, Austria
| | | | | | - Katharina Schallmoser
- Department of Transfusion Medicine, University Clinic, Paracelsus Medical University, Salzburg, Austria
| | - Dirk Strunk
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), University Clinic, Paracelsus Medical University, Salzburg, Austria
| |
Collapse
|
2
|
Andrade AC, Wolf M, Binder HM, Gomes FG, Manstein F, Ebner-Peking P, Poupardin R, Zweigerdt R, Schallmoser K, Strunk D. Hypoxic Conditions Promote the Angiogenic Potential of Human Induced Pluripotent Stem Cell-Derived Extracellular Vesicles. Int J Mol Sci 2021; 22:ijms22083890. [PMID: 33918735 PMCID: PMC8070165 DOI: 10.3390/ijms22083890] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 12/11/2022] Open
Abstract
Stem cells secrete paracrine factors including extracellular vesicles (EVs) which can mediate cellular communication and support the regeneration of injured tissues. Reduced oxygen (hypoxia) as a key regulator in development and regeneration may influence cellular communication via EVs. We asked whether hypoxic conditioning during human induced pluripotent stem cell (iPSC) culture effects their EV quantity, quality or EV-based angiogenic potential. We produced iPSC-EVs from large-scale culture-conditioned media at 1%, 5% and 18% air oxygen using tangential flow filtration (TFF), with or without subsequent concentration by ultracentrifugation (TUCF). EVs were quantified by tunable resistive pulse sensing (TRPS), characterized according to MISEV2018 guidelines, and analyzed for angiogenic potential. We observed superior EV recovery by TFF compared to TUCF. We confirmed hypoxia efficacy by HIF-1α stabilization and pimonidazole hypoxyprobe. EV quantity did not differ significantly at different oxygen conditions. Significantly elevated angiogenic potential was observed for iPSC-EVs derived from 1% oxygen culture by TFF or TUCF as compared to EVs obtained at higher oxygen or the corresponding EV-depleted soluble factor fractions. Data thus demonstrate that cell-culture oxygen conditions and mode of EV preparation affect iPSC-EV function. We conclude that selecting appropriate protocols will further improve production of particularly potent iPSC-EV-based therapeutics.
Collapse
Affiliation(s)
- André Cronemberger Andrade
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (A.C.A.); (M.W.); (H.-M.B.); (P.E.-P.); (R.P.)
| | - Martin Wolf
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (A.C.A.); (M.W.); (H.-M.B.); (P.E.-P.); (R.P.)
| | - Heide-Marie Binder
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (A.C.A.); (M.W.); (H.-M.B.); (P.E.-P.); (R.P.)
| | - Fausto Gueths Gomes
- Department of Transfusion Medicine and SCI-TReCS, Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (F.G.G.); (K.S.)
| | - Felix Manstein
- Department of Cardiac, Thoracic, Transplantation and Vascular Surgery, Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, 30625 Hannover, Germany; (F.M.); (R.Z.)
| | - Patricia Ebner-Peking
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (A.C.A.); (M.W.); (H.-M.B.); (P.E.-P.); (R.P.)
| | - Rodolphe Poupardin
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (A.C.A.); (M.W.); (H.-M.B.); (P.E.-P.); (R.P.)
| | - Robert Zweigerdt
- Department of Cardiac, Thoracic, Transplantation and Vascular Surgery, Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, 30625 Hannover, Germany; (F.M.); (R.Z.)
| | - Katharina Schallmoser
- Department of Transfusion Medicine and SCI-TReCS, Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (F.G.G.); (K.S.)
| | - Dirk Strunk
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (A.C.A.); (M.W.); (H.-M.B.); (P.E.-P.); (R.P.)
- Correspondence:
| |
Collapse
|
3
|
Rohban R, Prietl B, Pieber TR. Crosstalk between Stem and Progenitor Cellular Mediators with Special Emphasis on Vasculogenesis. Transfus Med Hemother 2017. [PMID: 28626368 DOI: 10.1159/000477677] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The cellular components and molecular processes of signaling during vasculogenesis have been investigated for decades. Considerable efforts have been made to unravel regulatory mechanisms of vasculogenesis through crosstalk between vasculogenic playmakers located in the vascular niche, namely hematopoietic stem cells, endothelial progenitor cells, and mesenchymal stem and progenitor cells. Recent studies have increased the knowledge about signaling events within vascular microenvironment that leads to vasculogenesis. Findings from these recent studies indicate the impact of cellular crosstalk through signaling pathways such as vascular endothelial growth factor signaling, wingless and Notch signaling in vasculogenesis and vascular development. In this review, we highlight the signaling signature between stem and progenitor cellular mediators during vasculogenesis. We further focus on hematopoietic stem cell-endothelial progenitor cell crosstalk during vasculogenesis and discuss their potential implications and benefits for therapeutic interventions and regenerative therapy.
Collapse
Affiliation(s)
- Rokhsareh Rohban
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, Graz, Austria.,Center for Medical Research (ZMF), Medical University of Graz, Graz, Austria
| | - Barbara Prietl
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, Graz, Austria.,Competence Center for Biomarker Research in Medicine, CBmed, Graz, Austria
| | - Thomas R Pieber
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, Graz, Austria.,Competence Center for Biomarker Research in Medicine, CBmed, Graz, Austria.,HEALTH-Institute for Biomedicine and Health Sciences, Joanneum Research Forschungsgesellschaft m.b.H, Graz, Austria
| |
Collapse
|
4
|
Abstract
In the mid-1990s, my research group began to devise a method to establish endothelial cell cultures from human peripheral blood, with an ultimate goal of examining interindividual heterogeneity of endothelial biology. The initial work, published in the JCI in 2000, described the method enabling successful attainment of blood outgrowth endothelial cells (BOEC). Truly endothelial, BOEC are progeny of a transplantable cell that originates in bone marrow, a putative endothelial progenitor. Our subsequent experimental work focused upon practical applications of BOEC: their use for gene therapy, tissue engineering, assessment of mutant gene effect, and discovery of heterogeneity in endothelial biology.
Collapse
|
5
|
Souidi N, Stolk M, Rudeck J, Strunk D, Schallmoser K, Volk HD, Seifert M. Stromal Cells Act as Guardians for Endothelial Progenitors by Reducing Their Immunogenicity After Co-Transplantation. Stem Cells 2017; 35:1233-1245. [PMID: 28100035 DOI: 10.1002/stem.2573] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 12/07/2016] [Accepted: 12/18/2016] [Indexed: 01/15/2023]
Abstract
Regeneration of injured tissues requires effective therapeutic strategies supporting vasculogenesis. The lack of instantly available autologous cell sources and immunogenicity of allogeneic endothelial (progenitor) cells limits clinical progress. Based on the immunosuppressive potency of mesenchymal stem/progenitor cells (MSCs), we investigated whether crosstalk between endothelial colony-forming progenitor cells (ECFCs) and MSCs during vasculogenesis could lower allogeneic T cell responses against ECFCs allowing long-term engraftment in vivo. Immunodeficient mice received subcutaneous grafts containing human ECFCs alone, or pairs of human ECFCs/MSCs from the same umbilical cord (UC) to study vasculogenesis in the presence of human leukocyte antigen (HLA)-mismatched human peripheral blood mononuclear cells (PBMCs). In vitro, cell surface marker changes due to interferon gamma (IFNγ) stimulation during ECFC/MSC coculture were determined and further effects on allostimulated T cell proliferation and cytotoxic lysis were measured. IFNγ-induced HLA-DR expression on ECFCs and MSCs, but both cell types had significantly less HLA-DR in cocultures. ECFC-induced T cell proliferation was abolished after MSC coculture as a result of HLA-DR downregulation and indolamin-2,3-dioxygenase activation. Additionally, allospecific CD8+ T cell-mediated lysis of ECFCs was reduced in cocultures. ECFC/MSC coapplication in immunodeficient mice not only promoted the generation of improved blood vessel architecture after 6 weeks, but also reduced intragraft immune cell infiltration and endothelial HLA-DR expression following PBMC reconstitution. Crosstalk between UC-derived ECFCs and MSCs after combined transplantation can lower the risk of ECFC rejection, thus enabling their coapplication for therapeutic vasculogenesis. Stem Cells 2017;35:1233-1245.
Collapse
Affiliation(s)
- Naima Souidi
- Institute of Medical Immunology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | - Meaghan Stolk
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | - Juliane Rudeck
- Institute of Medical Immunology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | - Dirk Strunk
- Institute of Experimental and Clinical Cell Therapy, Paracelsus Medical University, Salzburg, Austria.,Spinal Cord & Tissue Regeneration Center, Paracelsus Medical University, Salzburg, Austria
| | - Katharina Schallmoser
- Spinal Cord & Tissue Regeneration Center, Paracelsus Medical University, Salzburg, Austria.,Department for Blood Group Serology and Transfusion Medicine, Paracelsus Medical University, Salzburg, Austria
| | - Hans-Dieter Volk
- Institute of Medical Immunology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | - Martina Seifert
- Institute of Medical Immunology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| |
Collapse
|
6
|
Mesenchymal Stem and Progenitor Cells in Regeneration: Tissue Specificity and Regenerative Potential. Stem Cells Int 2017; 2017:5173732. [PMID: 28286525 PMCID: PMC5327785 DOI: 10.1155/2017/5173732] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 12/07/2016] [Indexed: 12/15/2022] Open
Abstract
It has always been an ambitious goal in medicine to repair or replace morbid tissues for regaining the organ functionality. This challenge has recently gained momentum through considerable progress in understanding the biological concept of the regenerative potential of stem cells. Routine therapeutic procedures are about to shift towards the use of biological and molecular armamentarium. The potential use of embryonic stem cells and invention of induced pluripotent stem cells raised hope for clinical regenerative purposes; however, the use of these interventions for regenerative therapy showed its dark side, as many health concerns and ethical issues arose in terms of using these cells in clinical applications. In this regard, adult stem cells climbed up to the top list of regenerative tools and mesenchymal stem cells (MSC) showed promise for regenerative cell therapy with a rather limited level of risk. MSC have been successfully isolated from various human tissues and they have been shown to offer the possibility to establish novel therapeutic interventions for a variety of hard-to-noncurable diseases. There have been many elegant studies investigating the impact of MSC in regenerative medicine. This review provides compact information on the role of stem cells, in particular, MSC in regeneration.
Collapse
|
7
|
Laner-Plamberger S, Lener T, Schmid D, Streif DA, Salzer T, Öller M, Hauser-Kronberger C, Fischer T, Jacobs VR, Schallmoser K, Gimona M, Rohde E. Mechanical fibrinogen-depletion supports heparin-free mesenchymal stem cell propagation in human platelet lysate. J Transl Med 2015; 13:354. [PMID: 26554451 PMCID: PMC4641400 DOI: 10.1186/s12967-015-0717-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 10/29/2015] [Indexed: 01/10/2023] Open
Abstract
Background Pooled human platelet lysate (pHPL) is an efficient alternative to xenogenic supplements for ex vivo expansion of mesenchymal stem cells (MSCs) in clinical studies. Currently, porcine heparin is used in pHPL-supplemented medium to prevent clotting due to plasmatic coagulation factors. We therefore searched for an efficient and reproducible medium preparation method that avoids clot formation while omitting animal-derived heparin. Methods We established a protocol to deplete fibrinogen by clotting of pHPL in medium, subsequent mechanical hydrogel disruption and removal of the fibrin pellet. After primary culture, bone-marrow and umbilical cord derived MSCs were tested for surface markers by flow cytometry and for trilineage differentiation capacity. Proliferation and clonogenicity were analyzed for three passages. Results The proposed clotting procedure reduced fibrinogen more than 1000-fold, while a volume recovery of 99.5 % was obtained. All MSC types were propagated in standard and fibrinogen-depleted medium. Flow cytometric phenotype profiles and adipogenic, osteogenic and chondrogenic differentiation potential in vitro were independent of MSC-source or medium type. Enhanced proliferation of MSCs was observed in the absence of fibrinogen but presence of heparin compared to standard medium. Interestingly, this proliferative response to heparin was not detected after an initial contact with fibrinogen during the isolation procedure. Conclusions Here, we present an efficient, reproducible and economical method in compliance to good manufacturing practice for the preparation of MSC media avoiding xenogenic components and suitable for clinical studies. Electronic supplementary material The online version of this article (doi:10.1186/s12967-015-0717-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sandra Laner-Plamberger
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria. .,Department of Blood Group Serology and Transfusion Medicine, Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Lindhofstrasse 20-22, 5020, Salzburg, Austria.
| | - Thomas Lener
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria. .,Department of Blood Group Serology and Transfusion Medicine, Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Lindhofstrasse 20-22, 5020, Salzburg, Austria.
| | - Doris Schmid
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria. .,Department of Blood Group Serology and Transfusion Medicine, Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Lindhofstrasse 20-22, 5020, Salzburg, Austria.
| | - Doris A Streif
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria. .,Department of Blood Group Serology and Transfusion Medicine, Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Lindhofstrasse 20-22, 5020, Salzburg, Austria.
| | - Tina Salzer
- Department of Blood Group Serology and Transfusion Medicine, Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Lindhofstrasse 20-22, 5020, Salzburg, Austria.
| | - Michaela Öller
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria. .,Department of Blood Group Serology and Transfusion Medicine, Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Lindhofstrasse 20-22, 5020, Salzburg, Austria.
| | | | - Thorsten Fischer
- Department of Gynecology and Obstetrics, Paracelsus Medical University, Salzburg, Austria.
| | - Volker R Jacobs
- Department of Gynecology and Obstetrics, Paracelsus Medical University, Salzburg, Austria.
| | - Katharina Schallmoser
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria. .,Department of Blood Group Serology and Transfusion Medicine, Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Lindhofstrasse 20-22, 5020, Salzburg, Austria.
| | - Mario Gimona
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria. .,Department of Blood Group Serology and Transfusion Medicine, Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Lindhofstrasse 20-22, 5020, Salzburg, Austria.
| | - Eva Rohde
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria. .,Department of Blood Group Serology and Transfusion Medicine, Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Lindhofstrasse 20-22, 5020, Salzburg, Austria.
| |
Collapse
|
8
|
Burnouf T, Strunk D, Koh MBC, Schallmoser K. Human platelet lysate: Replacing fetal bovine serum as a gold standard for human cell propagation? Biomaterials 2015; 76:371-87. [PMID: 26561934 DOI: 10.1016/j.biomaterials.2015.10.065] [Citation(s) in RCA: 329] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 10/16/2015] [Accepted: 10/26/2015] [Indexed: 02/07/2023]
Abstract
The essential physiological role of platelets in wound healing and tissue repair builds the rationale for the use of human platelet derivatives in regenerative medicine. Abundant growth factors and cytokines stored in platelet granules can be naturally released by thrombin activation and clotting or artificially by freeze/thaw-mediated platelet lysis, sonication or chemical treatment. Human platelet lysate prepared by the various release strategies has been established as a suitable alternative to fetal bovine serum as culture medium supplement, enabling efficient propagation of human cells under animal serum-free conditions for a multiplicity of applications in advanced somatic cell therapy and tissue engineering. The rapidly increasing number of studies using platelet derived products for inducing human cell proliferation and differentiation has also uncovered a considerable variability of human platelet lysate preparations which limits comparability of results. The main variations discussed herein encompass aspects of donor selection, preparation of the starting material, the possibility for pooling in plasma or additive solution, the implementation of pathogen inactivation and consideration of ABO blood groups, all of which can influence applicability. This review outlines the current knowledge about human platelet lysate as a powerful additive for human cell propagation and highlights its role as a prevailing supplement for human cell culture capable to replace animal serum in a growing spectrum of applications.
Collapse
Affiliation(s)
- Thierry Burnouf
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Dirk Strunk
- Experimental & Clinical Cell Therapy Institute, Paracelsus Medical University, Salzburg, Austria; Spinal Cord Injury & Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria.
| | - Mickey B C Koh
- Blood Services Group, Health Sciences Authority, Singapore; Department for Hematology, St George's Hospital and Medical School, London, UK
| | - Katharina Schallmoser
- Spinal Cord Injury & Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria; Department for Blood Group Serology and Transfusion Medicine, Paracelsus Medical University, Salzburg, Austria.
| |
Collapse
|
9
|
Flood B, Oficjalska K, Laukens D, Fay J, O'Grady A, Caiazza F, Heetun Z, Mills KHG, Sheahan K, Ryan EJ, Doherty GA, Kay E, Creagh EM. Altered expression of caspases-4 and -5 during inflammatory bowel disease and colorectal cancer: Diagnostic and therapeutic potential. Clin Exp Immunol 2015; 181:39-50. [PMID: 25943872 PMCID: PMC4469154 DOI: 10.1111/cei.12617] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/23/2015] [Indexed: 12/27/2022] Open
Abstract
Caspases are a group of proteolytic enzymes involved in the co-ordination of cellular processes, including cellular homeostasis, inflammation and apoptosis. Altered activity of caspases, particularly caspase-1, has been implicated in the development of intestinal diseases, such as inflammatory bowel disease (IBD) and colorectal cancer (CRC). However, the involvement of two related inflammatory caspase members, caspases-4 and -5, during intestinal homeostasis and disease has not yet been established. This study demonstrates that caspases-4 and -5 are involved in IBD-associated intestinal inflammation. Furthermore, we found a clear correlation between stromal caspase-4 and -5 expression levels, inflammation and disease activity in ulcerative colitis patients. Deregulated intestinal inflammation in IBD patients is associated with an increased risk of developing CRC. We found robust expression of caspases-4 and -5 within intestinal epithelial cells, exclusively within neoplastic tissue, of colorectal tumours. An examination of adjacent normal, inflamed and tumour tissue from patients with colitis-associated CRC confirmed that stromal expression of caspases-4 and -5 is increased in inflamed and dysplastic tissue, while epithelial expression is restricted to neoplastic tissue. In addition to identifying caspases-4 and -5 as potential targets for limiting intestinal inflammation, this study has identified epithelial-expressed caspases-4 and -5 as biomarkers with diagnostic and therapeutic potential in CRC.
Collapse
Affiliation(s)
- B Flood
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College DublinIreland
| | - K Oficjalska
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College DublinIreland
| | - D Laukens
- Department of Gastroenterology, Ghent UniversityGhent, Belgium
| | - J Fay
- Pathology Department, RCSI and Beaumont HospitalDublin
| | - A O'Grady
- Pathology Department, RCSI and Beaumont HospitalDublin
| | - F Caiazza
- Centre for Colorectal Disease, St Vincent's University Hospital and School of Medicine and Medical Sciences, University College DublinIreland
| | - Z Heetun
- Centre for Colorectal Disease, St Vincent's University Hospital and School of Medicine and Medical Sciences, University College DublinIreland
| | - K H G Mills
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College DublinIreland
| | - K Sheahan
- Centre for Colorectal Disease, St Vincent's University Hospital and School of Medicine and Medical Sciences, University College DublinIreland
| | - E J Ryan
- Centre for Colorectal Disease, St Vincent's University Hospital and School of Medicine and Medical Sciences, University College DublinIreland
| | - G A Doherty
- Centre for Colorectal Disease, St Vincent's University Hospital and School of Medicine and Medical Sciences, University College DublinIreland
| | - E Kay
- Pathology Department, RCSI and Beaumont HospitalDublin
| | - E M Creagh
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College DublinIreland
| |
Collapse
|
10
|
Salter MM, Seeto WJ, DeWitt BB, Hashimi SA, Schwartz DD, Lipke EA, Wooldridge AA. Characterization of endothelial colony-forming cells from peripheral blood samples of adult horses. Am J Vet Res 2015; 76:174-87. [DOI: 10.2460/ajvr.76.2.174] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
11
|
Mesenchymal stromal cells from the human placenta promote neovascularization in a mouse model in vivo. Placenta 2014; 35:517-9. [PMID: 24814611 DOI: 10.1016/j.placenta.2014.04.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 04/10/2014] [Accepted: 04/15/2014] [Indexed: 01/14/2023]
Abstract
Cell transplantation is a promising strategy in regenerative medicine for revascularization of ischemic tissues. Based on our observation that placental mesenchymal stromal cells (PMSC) enhance endothelial cell viability in vitro via secretion of angiogenic factors, we asked whether PMSC support vascular growth in vivo. PMSC were isolated from amnion and placental endothelial cells (PLEC) from chorion and either separately or co-transplanted subcutaneously into immune-deficient mice. Co-transplantation resulted in a higher number of perfused human vessels (CD31+/vimentin+) containing mouse glycophorin A+ erythrocytes. Results indicate positive effects of PMSC on neovascularization in vivo, making them attractive candidates to create autologous PMSC/PLEC pairs for research and transplantation.
Collapse
|