103 results on '"Susanne Wolbank"'
Search Results
2. Human-based New Approach Methodologies in Developmental Toxicity Testing: A Step Ahead from the State-of-the-art with a Feto-placental Organ-on-a-chip Platform
- Author
-
Michaela Luconi, Miguel Angel Sogorb, Udo R. Markert, Emilio Benfenati, Tobias May, Susanne Wolbank, Alessandra Roncaglioni, Astrid Schmidt, Marco Straccia, and Sabrina Tait
- Subjects
life_sciences_other - Abstract
Developmental toxicity testing urgently requires the implementation of human relevant new approach methodologies (NAMs) that better recapitulate the peculiar nature of human physiology during pregnancy, especially the placenta and the maternal/fetal interface, which represent a key stage for the human lifelong health. Fit-for-purpose NAMs for the placental-fetal interface are desirable to improve the biological knowledge of environmental exposure at molecular level and to reduce the high cost, time and ethical impact of animal studies. This article reviews the state of the art on the available in vitro (placental, fetal and amniotic cell-based systems) and in silico NAMs of human relevance for developmental toxicity testing purposes, as well as of the available Adverse Outcome Pathways related to developmental toxicity. The OECD TG 414 for the identification and assessment of deleterious effects of prenatal exposure to chemicals on developing organisms will be discussed to delineate the regulatory context and to better debate what is missing and needed in the context of the developmental origins of health and disease hypothesis to significantly improve this sector. Starting from this analysis, the development of a novel human feto-placental organ-on-chip platform will be introduced as an innovative alternative tool for developmental toxicity testing, considering possible implementation and validation strategies to overcome the limitation of the current animal studies and NAMs available in regulatory toxicology and in the biomedical field.
- Published
- 2022
- Full Text
- View/download PDF
3. Methods and criteria for validating the multimodal functions of perinatal derivatives when used in oncological and antimicrobial applications
- Author
-
Antonietta R. Silini, Taja Železnik Ramuta, Ana Salomé Pires, Asmita Banerjee, Marie Dubus, Florelle Gindraux, Halima Kerdjoudj, Justinas Maciulatis, Adelheid Weidinger, Susanne Wolbank, Günther Eissner, Bernd Giebel, Michela Pozzobon, Ornella Parolini, and Mateja Erdani Kreft
- Subjects
biological assays ,mechanisms of action ,potency assays ,perinatal derivatives ,Histology ,functional assays ,Biomedical Engineering ,cancer ,Settore BIO/13 - BIOLOGIA APPLICATA ,pharmacologic actions ,Bioengineering ,infections ,Biotechnology - Abstract
Perinatal derivatives or PnDs refer to tissues, cells and secretomes from perinatal, or birth-associated tissues. In the past 2 decades PnDs have been highly investigated for their multimodal mechanisms of action that have been exploited in various disease settings, including in different cancers and infections. Indeed, there is growing evidence that PnDs possess anticancer and antimicrobial activities, but an urgent issue that needs to be addressed is the reproducible evaluation of efficacy, both in vitro and in vivo. Herein we present the most commonly used functional assays for the assessment of antitumor and antimicrobial properties of PnDs, and we discuss their advantages and disadvantages in assessing the functionality. This review is part of a quadrinomial series on functional assays for the validation of PnDs spanning biological functions such as immunomodulation, anticancer and antimicrobial, wound healing, and regeneration.
- Published
- 2022
- Full Text
- View/download PDF
4. Establishment of In Vitro Models by Stress-Induced Premature Senescence for Characterizing the Stromal Vascular Niche in Human Adipose Tissue
- Author
-
Marlene Wahlmueller, Marie-Sophie Narzt, Karin Missfeldt, Verena Arminger, Anna Krasensky, Ingo Lämmermann, Barbara Schaedl, Mario Mairhofer, Susanne Suessner, Susanne Wolbank, and Eleni Priglinger
- Subjects
Space and Planetary Science ,Paleontology ,General Biochemistry, Genetics and Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,senescence ,stress-induced premature senescence (SIPS) ,doxorubicin ,adipose tissue ,adipose-derived stromal/stem cells (ASCs) ,stromal vascular fraction (SVF) ,microtissue - Abstract
Acting as the largest energy reservoir in the body, adipose tissue is involved in longevity and progression of age-related metabolic dysfunction. Here, cellular senescence plays a central role in the generation of a pro-inflammatory environment and in the evolution of chronic diseases. Within the complexity of a tissue, identification and targeting of senescent cells is hampered by their heterogeneity. In this study, we generated stress-induced premature senescence 2D and 3D in vitro models for the stromal vascular niche of human adipose tissue. We established treatment conditions for senescence induction using Doxorubicin (Dox), starting from adipose-derived stromal/stem cells (ASCs), which we adapted to freshly isolated microtissue-stromal vascular fraction (MT-SVF), where cells are embedded within their native extracellular matrix. Senescence hallmarks for the established in vitro models were verified on different cellular levels, including morphology, cell cycle arrest, senescence-associated β-galactosidase activity (SA-βgal) and gene expression. Two subsequent exposures with 200 nM Dox for six days were suitable to induce senescence in our in vitro models. We demonstrated induction of senescence in the 2D in vitro models through SA-βgal activity, at the mRNA level (LMNB1, CDK1, p21) and additionally by G2/M phase cell cycle arrest in ASCs. Significant differences in Lamin B1 and p21 protein expression confirmed senescence in our MT-SVF 3D model. MT-SVF 3D cultures were composed of multiple cell types, including CD31, CD34 and CD68 positive cells, while cell death remained unaltered upon senescence induction. As heterogeneity and complexity of adipose tissue senescence is given by multiple cell types, our established senescence models that represent the perivascular niche embedded within its native extracellular matrix are highly relevant for future clinical studies.
- Published
- 2022
- Full Text
- View/download PDF
5. SVF-derived extracellular vesicles carry characteristic miRNAs in lipedema
- Author
-
Heinz Redl, Susanne Wolbank, Martin Barsch, Jaroslaw Jacak, Matthias Sandhofer, Moritz Weigl, Daniela Auer, Eleni Priglinger, Johannes Grillari, Mario Gimona, Karin Strohmeier, Carolin Lindner, and Matthias Hackl
- Subjects
Adult ,Male ,Pathology ,medicine.medical_specialty ,Adolescent ,Adipose tissue ,lcsh:Medicine ,Diseases ,Biology ,Extracellular vesicles ,Article ,Extracellular Vesicles ,microRNA ,Conditioned medium ,medicine ,Extracellular ,Humans ,lcsh:Science ,Multidisciplinary ,Lipedema ,Significant difference ,lcsh:R ,Stromal vascular fraction ,Middle Aged ,MicroRNAs ,Mechanisms of disease ,Adipose Tissue ,Healthy individuals ,miRNAs ,Female ,lcsh:Q ,Biomarkers - Abstract
Lipedema is a chronic, progressive disease of adipose tissue with lack of consistent diagnostic criteria. The aim of this study was a thorough comparative characterization of extracellular microRNAs (miRNAs) from the stromal vascular fraction (SVF) of healthy and lipedema adipose tissue. For this, we analyzed 187 extracellular miRNAs in concentrated conditioned medium (cCM) and specifically in small extracellular vesicles (sEVs) enriched thereof by size exclusion chromatography. No significant difference in median particle size and concentration was observed between sEV fractions in healthy and lipedema. We found the majority of miRNAs located predominantly in cCM compared to sEV enriched fraction. Surprisingly, hierarchical clustering of the most variant miRNAs showed that only sEVmiRNA profiles – but not cCMmiRNAs – were impacted by lipedema. Seven sEVmiRNAs (miR–16-5p, miR-29a-3p, miR-24-3p, miR-454-p, miR–144-5p, miR-130a-3p, let-7c-5p) were differently regulated in lipedema and healthy individuals, whereas only one cCMmiRNA (miR-188-5p) was significantly downregulated in lipedema. Comparing SVF from healthy and lipedema patients, we identified sEVs as the lipedema relevant miRNA fraction. This study contributes to identify the potential role of SVF secreted miRNAs in lipedema.
- Published
- 2020
- Full Text
- View/download PDF
6. Multi-Level Analysis of Adipose Tissue Reveals the Relevance of Perivascular Subpopulations and an Increased Endothelial Permeability in Early-Stage Lipedema
- Author
-
Karin Strohmeier, Martina Hofmann, Jaroslaw Jacak, Marie-Sophie Narzt, Marlene Wahlmueller, Mario Mairhofer, Barbara Schaedl, Wolfgang Holnthoner, Martin Barsch, Matthias Sandhofer, Susanne Wolbank, and Eleni Priglinger
- Subjects
lipedema ,endothelial permeability ,endothelial cells ,pericytes ,stromal vascular fraction (SVF) ,adipose-derived stromal/stem cells (ASC) ,adipose tissue ,aromatase ,ZNF423 ,machine learning ,Medicine (miscellaneous) ,General Biochemistry, Genetics and Molecular Biology - Abstract
Lipedema is a chronic, progressive disease of adipose tissue with unknown etiology. Based on the relevance of the stromal vascular fraction (SVF) cell population in lipedema, we performed a thorough characterization of subcutaneous adipose tissue, SVF isolated thereof and the sorted populations of endothelial cells (EC), pericytes and cultured adipose-derived stromal/stem cells (ASC) of early-stage lipedema patients. We employed histological and gene expression analysis and investigated the endothelial barrier by immunofluorescence and analysis of endothelial permeability in vitro. Although there were no significant differences in histological stainings, we found altered gene expression of factors relevant for local estrogen metabolism (aromatase), preadipocyte commitment (ZNF423) and immune cell infiltration (CD11c) in lipedema on tissue level as well as in distinct cellular subpopulations. Machine learning analysis of immunofluorescence images of CD31 and ZO-1 revealed a morphological difference in the cellular junctions of EC cultures derived from healthy and lipedema individuals. Furthermore, the secretome of lipedema-derived SVF cells was sufficient to significantly increase leakiness of healthy human primary EC, which was also reflected by decreased mRNA expression of VE-cadherin. Here we showed for the first time, that the secretome of SVF cells creates an environment that triggers endothelial barrier dysfunction in early-stage lipedema. Moreover, since alterations in gene expression were detected on the cellular and/or tissue level, the choice of sample material is of high importance in elucidating this complex disease.
- Published
- 2022
7. General consensus on multimodal functions and validation analysis of perinatal derivatives for regenerative medicine applications
- Author
-
Michela Pozzobon, Stefania D’Agostino, Maria G. Roubelakis, Anna Cargnoni, Roberto Gramignoli, Susanne Wolbank, Florelle Gindraux, Sveva Bollini, Halima Kerdjoudj, Mathilde Fenelon, Roberta Di Pietro, Mariangela Basile, Veronika Borutinskaitė, Roberta Piva, Andreina Schoeberlein, Guenther Eissner, Bernd Giebel, and Peter Ponsaerts
- Subjects
amniotic membrane and fluid stem cells ,Histology ,Biomedical Engineering ,extracellular vesicles ,perinatal derivatives ,regenerative medicine ,tissue regeneration ,Bioengineering ,610 Medicine & health ,610 Medizin und Gesundheit ,Biology ,Engineering sciences. Technology ,Biotechnology - Abstract
Perinatal tissues, such as placenta and umbilical cord contain a variety of somatic stem cell types, spanning from the largely used hematopoietic stem and progenitor cells to the most recently described broadly multipotent epithelial and stromal cells. As perinatal derivatives (PnD), several of these cell types and related products provide an interesting regenerative potential for a variety of diseases. Within COST SPRINT Action, we continue our review series, revising and summarizing the modalities of action and proposed medical approaches using PnD products: cells, secretome, extracellular vesicles, and decellularized tissues. Focusing on the brain, bone, skeletal muscle, heart, intestinal, liver, and lung pathologies, we discuss the importance of potency testing in validating PnD therapeutics, and critically evaluate the concept of PnD application in the field of tissue regeneration. Hereby we aim to shed light on the actual therapeutic properties of PnD, with an open eye for future clinical application. This review is part of a quadrinomial series on functional/potency assays for validation of PnD, spanning biological functions, such as immunomodulation, anti-microbial/anti-cancer, anti-inflammation, wound healing, angiogenesis, and regeneration.
- Published
- 2022
- Full Text
- View/download PDF
8. Label‐free characterization of an extracellular vesicle‐based therapeutic
- Author
-
Eva Grasmann, Jaroslaw Jacak, Juergen Strasser, Florian Weber, Claudia Arzt, Mario Gimona, Susanne Wolbank, Dmitry Sivun, Eleni Priglinger, Daniela Auer, Johannes Preiner, Boris Buchroithner, and Johannes Grillari
- Subjects
Histology ,Materials science ,QH573-671 ,Atomic force microscopy ,quartz crystal microbalance (QCM) ,Mesenchymal Stem Cells ,Cell Biology ,Extracellular vesicle ,Quartz crystal microbalance ,Microscopy, Atomic Force ,Extracellular vesicles ,Characterization (materials science) ,label‐free sensors ,Extracellular Vesicles ,Characterization methods ,Humans ,atomic force microscopy (AFM) ,extracellular vesicles (EVs) ,Cytology ,Cytometry ,Research Articles ,Biomedical engineering ,Label free ,Research Article - Abstract
Interest in mesenchymal stem cell derived extracellular vesicles (MSC‐EVs) as therapeutic agents has dramatically increased over the last decade. Current approaches to the characterization and quality control of EV‐based therapeutics include particle tracking techniques, Western blotting, and advanced cytometry, but standardized methods are lacking. In this study, we established and verified quartz crystal microbalance (QCM) as highly sensitive label‐free immunosensing technique for characterizing clinically approved umbilical cord MSC‐EVs enriched by tangential flow filtration and ultracentrifugation. Using QCM in conjunction with common characterization methods, we were able to specifically detect EVs via EV (CD9, CD63, CD81) and MSC (CD44, CD49e, CD73) markers. Furthermore, analysis of QCM dissipation versus frequency allowed us to quantitatively determine the ratio of marker‐specific EVs versus non‐vesicular particles (NVPs) – a parameter that cannot be obtained by any other technique so far. Additionally, we characterized the topography and elasticity of these EVs by atomic force microscopy (AFM), enabling us to distinguish between EVs and NVPs in our EV preparations. This measurement modality makes it possible to identify EV sub‐fractions, discriminate between EVs and NVPs, and to characterize EV surface proteins, all with minimal sample preparation and using label‐free measurement devices with low barriers of entry for labs looking to widen their spectrum of characterization techniques. Our combination of QCM with impedance measurement (QCM‐I) and AFM measurements provides a robust multi‐marker approach to the characterization of clinically approved EV therapeutics and opens the door to improved quality control.
- Published
- 2021
9. Stromal vascular fraction cells as biologic coating of mesh for hernia repair
- Author
-
Susanne Wolbank, Eleni Priglinger, René H. Fortelny, Alexander H. Petter-Puchner, B. Schädl, Bárbara Pérez-Köhler, Olivier Guillaume, Heinz Redl, N. Saxenhuber, Patrick Heimel, and Claudia Keibl
- Subjects
Male ,medicine.medical_specialty ,Stromal vascular fraction ,Angiogenesis ,Adipose tissue ,Regenerative medicine ,Fibrin ,Rats, Nude ,03 medical and health sciences ,0302 clinical medicine ,Vascularity ,Tissue engineering ,In vivo ,medicine ,Animals ,Humans ,Herniorrhaphy ,030304 developmental biology ,Mesh ,Biological Products ,0303 health sciences ,biology ,business.industry ,Abdominal hernia ,Surgical Mesh ,Rats ,Surgery ,Disease Models, Animal ,030220 oncology & carcinogenesis ,biology.protein ,Original Article ,medicine.symptom ,business ,Biomedical engineering - Abstract
Background The interest in non-manipulated cells originating from adipose tissue has raised tremendously in the field of tissue engineering and regenerative medicine. The resulting stromal vascular fraction (SVF) cells have been successfully used in numerous clinical applications. The aim of this experimental work is, first to combine a macroporous synthetic mesh with SVF isolated using a mechanical disruption process, and to assess the effect of those cells on the early healing phase of hernia. Methods Human SVF cells combined with fibrin were used to coat commercial titanized polypropylene meshes. In vitro, viability and growth of the SVF cells were assessed using live/dead staining and scanning electron microscopy. The influence of SVF cells on abdominal wall hernia healing was conducted on immunodeficient rats, with a focus on short-term vascularization and fibrogenesis. Results Macroporous meshes were easily coated with SVF using a fibrin gel as temporary carrier. The in vitro experiments showed that the whole process including the isolation of human SVF cells and their coating on PP meshes did not impact on the SVF cells’ viability and on their capacity to attach and to proliferate. In vivo, the SVF cells were well tolerated by the animals, and coating mesh with SVF resulted in a decrease degree of vascularity compared to control group at day 21. Conclusions The utilization of SVF-coated mesh influences the level of angiogenesis during the early onset of tissue healing. Further long-term animal experiments are needed to confirm that this effect correlates with a more robust mesh integration compared to non-SVF-coated mesh.
- Published
- 2020
- Full Text
- View/download PDF
10. Human-Based New Approach Methodologies in Developmental Toxicity Testing: A Step Ahead from the State of the Art with a Feto–Placental Organ-on-Chip Platform
- Author
-
Michaela Luconi, Miguel A. Sogorb, Udo R. Markert, Emilio Benfenati, Tobias May, Susanne Wolbank, Alessandra Roncaglioni, Astrid Schmidt, Marco Straccia, and Sabrina Tait
- Subjects
Health, Toxicology and Mutagenesis ,Public Health, Environmental and Occupational Health - Abstract
Developmental toxicity testing urgently requires the implementation of human-relevant new approach methodologies (NAMs) that better recapitulate the peculiar nature of human physiology during pregnancy, especially the placenta and the maternal/fetal interface, which represent a key stage for human lifelong health. Fit-for-purpose NAMs for the placental–fetal interface are desirable to improve the biological knowledge of environmental exposure at the molecular level and to reduce the high cost, time and ethical impact of animal studies. This article reviews the state of the art on the available in vitro (placental, fetal and amniotic cell-based systems) and in silico NAMs of human relevance for developmental toxicity testing purposes; in addition, we considered available Adverse Outcome Pathways related to developmental toxicity. The OECD TG 414 for the identification and assessment of deleterious effects of prenatal exposure to chemicals on developing organisms will be discussed to delineate the regulatory context and to better debate what is missing and needed in the context of the Developmental Origins of Health and Disease hypothesis to significantly improve this sector. Starting from this analysis, the development of a novel human feto–placental organ-on-chip platform will be introduced as an innovative future alternative tool for developmental toxicity testing, considering possible implementation and validation strategies to overcome the limitation of the current animal studies and NAMs available in regulatory toxicology and in the biomedical field.
- Published
- 2022
- Full Text
- View/download PDF
11. Sub-Regional Differences of the Human Amniotic Membrane and Their Potential Impact on Tissue Regeneration Application
- Author
-
Adelheid Weidinger, Laura Poženel, Susanne Wolbank, and Asmita Banerjee
- Subjects
0301 basic medicine ,human amniotic membrane epithelial cells ,Scaffold ,Histology ,lcsh:Biotechnology ,Biomedical Engineering ,Bioengineering ,Review ,Germ layer ,human amniotic membrane mesenchymal stromal cells ,scaffold ,Biology ,sub-regions ,bioactive factors ,Extracellular matrix ,03 medical and health sciences ,0302 clinical medicine ,lcsh:TP248.13-248.65 ,medicine ,human amnion ,placental ,Amnion ,Bioengineering and Biotechnology ,food and beverages ,Chondrogenesis ,Cell biology ,030104 developmental biology ,Membrane ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,reflected ,Stem cell ,Reprogramming ,Biotechnology - Abstract
For more than 100 years, the human amniotic membrane (hAM) has been used in multiple tissue regeneration applications. The hAM consists of cells with stem cell characteristics and a rich layer of extracellular matrix. Undoubtedly, the hAM with viable cells has remarkable properties such as the differentiation potential into all three germ layers, immuno-modulatory, and anti-fibrotic properties. At first sight, the hAM seems to be one structural entity. However, by integrating its anatomical location, the hAM can be divided into placental, reflected, and umbilical amniotic membrane. Recent studies show that cells of these amniotic sub-regions differ considerably in their properties such as morphology, structure, and content/release of certain bioactive factors. The aim of this review is to summarize these findings and discuss the relevance of these different properties for tissue regeneration. In summary, reflected amnion seems to be more immuno-modulatory and could have a higher reprogramming efficiency, whereas placental amnion seems to be pro-inflammatory, pro-angiogenic, with higher proliferation and differentiation capacity (e.g., chondrogenic and osteogenic), and could be more suitable for certain graft constructions. Therefore, we suggest that the respective hAM sub-region should be selected in consideration of its desired outcome. This will help to optimize and fine-tune the clinical application of the hAM.
- Published
- 2021
- Full Text
- View/download PDF
12. Use of Amniotic Membrane and Its Derived Products for Bone Regeneration: A Systematic Review
- Author
-
Marion Etchebarne, Jean-Christophe Fricain, Halima Kerdjoudj, Roberta Di Pietro, Susanne Wolbank, Florelle Gindraux, Mathilde Fenelon, Bioingénierie tissulaire (BIOTIS), Université de Bordeaux (UB)-Institut National de la Santé et de la Recherche Médicale (INSERM), Service de chirurgie maxillo-faciale [CHU Bordeaux], CHU Bordeaux [Bordeaux], Biomatériaux et inflammation en site osseux - EA 4691 (BIOS), Université de Reims Champagne-Ardenne (URCA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-SFR CAP Santé (Champagne-Ardenne Picardie Santé), Université de Reims Champagne-Ardenne (URCA)-Université de Picardie Jules Verne (UPJV)-Université de Reims Champagne-Ardenne (URCA)-Université de Picardie Jules Verne (UPJV), University 'G. d'Annunzio' of Chieti-Pescara [Chieti], Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in the Research Center of AUVA, Austrian Cluster for Tissue Regeneration, Service de Chirurgie Orthopédique Traumatologique et Plastique [Besançon], Centre Hospitalier Régional Universitaire de Besançon (CHRU Besançon)-Université de Franche-Comté (UFC), Université Bourgogne Franche-Comté [COMUE] (UBFC)-Université Bourgogne Franche-Comté [COMUE] (UBFC), Nanomédecine, imagerie, thérapeutique - UFC (EA 4662) (NIT / NANOMEDECINE), and Université de Franche-Comté (UFC)
- Subjects
0301 basic medicine ,Scaffold ,Histology ,[SDV]Life Sciences [q-bio] ,Biomedical Engineering ,regenerative medicine ,Bioengineering ,Bone healing ,Bioinformatics ,Regenerative medicine ,bone ,natural scaffold ,03 medical and health sciences ,0302 clinical medicine ,Tissue engineering ,Medicine ,Bone regeneration ,bone tissue engineering ,amniotic membrane ,business.industry ,Mesenchymal stem cell ,Bioengineering and Biotechnology ,030206 dentistry ,amniotic epithelial cells ,3. Good health ,030104 developmental biology ,Amniotic epithelial cells ,amniotic mesenchymal stromal cells ,Systematic Review ,Stem cell ,business ,TP248.13-248.65 ,Biotechnology - Abstract
International audience; Thanks to their biological properties, amniotic membrane (AM), and its derivatives are considered as an attractive reservoir of stem cells and biological scaffolds for bone regenerative medicine. The objective of this systematic review was to assess the benefit of using AM and amniotic membrane-derived products for bone regeneration. An electronic search of the MEDLINE-Pubmed database and the Scopus database was carried out and the selection of articles was performed following PRISMA guidelines. This systematic review included 42 articles taking into consideration the studies in which AM, amniotic-derived epithelial cells (AECs), and amniotic mesenchymal stromal cells (AMSCs) show promising results for bone regeneration in animal models. Moreover, this review also presents some commercialized products derived from AM and discusses their application modalities. Finally, AM therapeutic benefit is highlighted in the reported clinical studies. This study is the first one to systematically review the therapeutic benefits of AM and amniotic membrane-derived products for bone defect healing. The AM is a promising alternative to the commercially available membranes used for guided bone regeneration. Additionally, AECs and AMSCs associated with an appropriate scaffold may also be ideal candidates for tissue engineering strategies applied to bone healing. Here, we summarized these findings and highlighted the relevance of these different products for bone regeneration.
- Published
- 2021
- Full Text
- View/download PDF
13. Repopulation of decellularised articular cartilage by laser-based matrix engraving
- Author
-
M. Nalbach, Susanne Wolbank, Sylvia Nürnberger, Andreas H. Teuschl, Christoph Schneider, Claudia Keibl, Patrick Heimel, Johannes Grillari, Philipp J. Thurner, Barbara Schädl, Xavier Monforte, and Heinz Redl
- Subjects
0301 basic medicine ,Cartilage, Articular ,Scaffold ,Stromal cell ,lcsh:Medicine ,Biocompatible Materials ,Matrix (biology) ,General Biochemistry, Genetics and Molecular Biology ,Chondrocyte ,03 medical and health sciences ,0302 clinical medicine ,Laser engraving ,medicine ,Cell Adhesion ,Animals ,Humans ,Cell adhesion ,Mechanical Phenomena ,lcsh:R5-920 ,Tissue Engineering ,Tissue Scaffolds ,Chemistry ,Guided Tissue Regeneration ,Cartilage ,lcsh:R ,Mesenchymal stem cell ,Mechanical testing ,Cell Differentiation ,Mesenchymal Stem Cells ,General Medicine ,X-Ray Microtomography ,Decellularisation ,Chondrogenesis ,Immunohistochemistry ,Extracellular Matrix ,030104 developmental biology ,medicine.anatomical_structure ,Ectopic animal model ,Cartilage regeneration ,030220 oncology & carcinogenesis ,Lasers, Gas ,Cattle ,lcsh:Medicine (General) ,Biomarkers ,Biomedical engineering ,Research Paper ,Repopulation - Abstract
Background In spite of advances in the treatment of cartilage defects using cell and scaffold-based therapeutic strategies, the long-term outcome is still not satisfying since clinical scores decline years after treatment. Scaffold materials currently used in clinical settings have shown limitations in providing suitable biomechanical properties and an authentic and protective environment for regenerative cells. To tackle this problem, we developed a scaffold material based on decellularised human articular cartilage. Methods Human articular cartilage matrix was engraved using a CO2 laser and treated for decellularisation and glycosaminoglycan removal. Characterisation of the resulting scaffold was performed via mechanical testing, DNA and GAG quantification and in vitro cultivation with adipose-derived stromal cells (ASC). Cell vitality, adhesion and chondrogenic differentiation were assessed. An ectopic, unloaded mouse model was used for the assessment of the in vivo performance of the scaffold in combination with ASC and human as well as bovine chondrocytes. The novel scaffold was compared to a commercial collagen type I/III scaffold. Findings Crossed line engravings of the matrix allowed for a most regular and ubiquitous distribution of cells and chemical as well as enzymatic matrix treatment was performed to increase cell adhesion. The biomechanical characteristics of this novel scaffold that we term CartiScaff were found to be superior to those of commercially available materials. Neo-tissue was integrated excellently into the scaffold matrix and new collagen fibres were guided by the laser incisions towards a vertical alignment, a typical feature of native cartilage important for nutrition and biomechanics. In an ectopic, unloaded in vivo model, chondrocytes and mesenchymal stromal cells differentiated within the incisions despite the lack of growth factors and load, indicating a strong chondrogenic microenvironment within the scaffold incisions. Cells, most noticeably bone marrow-derived cells, were able to repopulate the empty chondrocyte lacunae inside the scaffold matrix. Interpretation Due to the better load-bearing, its chondrogenic effect and the ability to guide matrix-deposition, CartiScaff is a promising biomaterial to accelerate rehabilitation and to improve long term clinical success of cartilage defect treatment. Funding Austrian Research Promotion Agency FFG (“CartiScaff” #842455), Lorenz Böhler Fonds (16/13), City of Vienna Competence Team Project Signaltissue (MA23, #18-08), Graphical abstract Image, graphical abstract
- Published
- 2020
14. Adipose-tissue-derived therapeutic cells in their natural environment as an autologous cell therapy strategy: the microtissue-stromal vascular fraction
- Author
-
Sylvia Nürnberger, Paul Slezak, Carolin Lindner, Susanne Wolbank, J. Maier, Susanne Suessner, Christoph Wurzer, Wolfgang Holnthoner, Karin Strohmeier, Heinz Redl, and Eleni Priglinger
- Subjects
Adult ,Cell type ,Stromal cell ,adipose-derived stromal/stem cells ,lcsh:Diseases of the musculoskeletal system ,Cell Survival ,Stromal vascular fraction ,extracellular matrix ,0206 medical engineering ,Cell- and Tissue-Based Therapy ,lcsh:Surgery ,Neovascularization, Physiologic ,Adipose tissue ,02 engineering and technology ,Transplantation, Autologous ,Extracellular matrix ,angiogenesis ,Humans ,Cell Lineage ,Progenitor cell ,Cell Shape ,endothelial progenitor cells ,Chemistry ,Mesenchymal stem cell ,Cell Differentiation ,lcsh:RD1-811 ,020601 biomedical engineering ,Cell biology ,Adipose Tissue ,Stromal Cells ,Stem cell ,lcsh:RC925-935 ,Biomarkers - Abstract
The prerequisite for a successful clinical use of autologous adipose-tissue-derived cells is the highest possible regenerative potential of the applied cell population, the stromal vascular fraction (SVF). Current isolation methods depend on high enzyme concentration, lysis buffer, long incubation steps and mechanical stress, resulting in single cell dissociation. The aim of the study was to limit cell manipulation and obtain a derivative comprising therapeutic cells (microtissue-SVF) without dissociation from their natural extracellular matrix, by employing a gentle good manufacturing practice (GMP)-grade isolation. The microtissue-SVF yielded larger numbers of viable cells as compared to the improved standard-SVF, both with low enzyme concentration and minimal dead cell content. It comprised stromal tissue compounds (collagen, glycosaminoglycans, fibroblasts), capillaries and vessel structures (CD31+, smooth muscle actin+). A broad range of cell types was identified by surface-marker characterisation, including mesenchymal, haematopoietic, pericytic, blood and lymphatic vascular and epithelial cells. Subpopulations such as supra-adventitial adipose-derived stromal/stem cells and endothelial progenitor cells were significantly more abundant in the microtissue-SVF, corroborated by significantly higher potency for angiogenic tube-like structure formation in vitro. The microtissue-SVF showed the characteristic phenotype and tri-lineage mesenchymal differentiation potential in vitro and an immunomodulatory and pro-angiogenic secretome. In vivo implantation of the microtissue-SVF combined with fat demonstrated successful graft integration in nude mice. The present study demonstrated a fast and gentle isolation by minor manipulation of liposuction material, achieving a therapeutically relevant cell population with high vascularisation potential and immunomodulatory properties still embedded in a fraction of its original matrix.
- Published
- 2019
15. Photobiomodulation of freshly isolated human adipose tissue-derived stromal vascular fraction cells by pulsed light-emitting diodes for direct clinical application
- Author
-
Eleni Priglinger, Susanne Wolbank, Carolin Lindner, Sidrah Chaudary, Stefan Rieger, Heinz Redl, Peter Dungel, Christoph Wurzer, and J. Maier
- Subjects
0301 basic medicine ,Stromal cell ,Cell Survival ,Cell ,Population ,Biomedical Engineering ,Neovascularization, Physiologic ,Medicine (miscellaneous) ,Adipose tissue ,Cell Separation ,Biomaterials ,030207 dermatology & venereal diseases ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,medicine ,Humans ,Low-Level Light Therapy ,education ,Fibroblast ,education.field_of_study ,Chemistry ,Cell Differentiation ,Middle Aged ,Stromal vascular fraction ,Molecular biology ,Vascular endothelial growth factor ,030104 developmental biology ,medicine.anatomical_structure ,Adipose Tissue ,Female ,Stromal Cells ,Adult stem cell - Abstract
A highly interesting source for adult stem cells is adipose tissue, from which the stromal vascular fraction (SVF)-a heterogeneous cell population including the adipose-derived stromal/stem cells-can be obtained. To enhance the regenerative potential of freshly isolated SVF cells, low-level light therapy (LLLT) was used. The effects of pulsed blue (475 nm), green (516 nm), and red (635 nm) light from light-emitting diodes applied on freshly isolated SVF were analysed regarding cell phenotype, cell number, viability, adenosine triphosphate content, cytotoxicity, and proliferation but also osteogenic, adipogenic, and proangiogenic differentiation potential. The colony-forming unit fibroblast assay revealed a significantly increased colony size after LLLT with red light compared with untreated cells, whereas the frequency of colony-forming cells was not affected. LLLT with green and red light resulted in a stronger capacity to form vascular tubes by SVF when cultured within 3D fibrin matrices compared with untreated cells, which was corroborated by increased number and length of the single tubes and a significantly higher concentration of vascular endothelial growth factor. Our study showed beneficial effects after LLLT on the vascularization potential and proliferation capacity of SVF cells. Therefore, LLLT using pulsed light-emitting diode light might represent a new approach for activation of freshly isolated SVF cells for direct clinical application.
- Published
- 2018
- Full Text
- View/download PDF
16. Regenerative Zellen in der ästhetischen und rekonstruktiven Medizin
- Author
-
Susanne Wolbank, Matthias Sandhofer, Eleni Priglinger, and Heinz Redl
- Subjects
Gynecology ,medicine.medical_specialty ,business.industry ,medicine ,General Medicine ,business - Abstract
Regenerative Zellen werden zur Gewebekonstruktion und -regeneration genutzt. Dabei hat sich Fettgewebe als Quelle aufgrund guter Verfugbarkeit und leichter Gewinnung in der kosmetischen und plastischen Chirurgie durchgesetzt. Jedoch gibt es bei der Aufbereitung des zu transplantierenden Zellgemischs einiges zu beachten.
- Published
- 2019
- Full Text
- View/download PDF
17. Hydrogel composition and laser micropatterning to regulate sciatic nerve regeneration
- Author
-
Hildner Florian, Andreas H. Teuschl, Dvir Yelin, Talia Cohen, Susanne Wolbank, Eli Peled, Robert Schmidhammer, Dror Seliktar, Yulia Berkovitch, and Heinz Redl
- Subjects
Male ,0301 basic medicine ,food.ingredient ,Biomedical Engineering ,Nerve guidance conduit ,Medicine (miscellaneous) ,02 engineering and technology ,Gelatin ,Fibrin ,Biomaterials ,03 medical and health sciences ,food ,medicine ,Animals ,biology ,Guided Tissue Regeneration ,Chemistry ,Lasers ,Regeneration (biology) ,Hydrogels ,Sciatic nerve injury ,021001 nanoscience & nanotechnology ,medicine.disease ,Sciatic Nerve ,Nerve Regeneration ,Rats ,030104 developmental biology ,Rats, Inbred Lew ,Self-healing hydrogels ,biology.protein ,Sciatic nerve ,0210 nano-technology ,Biomedical engineering ,Micropatterning - Abstract
Treatment of peripheral nerve injuries has evolved over the past several decades to include the use of sophisticated new materials endowed with trophic and topographical cues that are essential for in vivo nerve fibre regeneration. In this research, we explored the use of an advanced design strategy for peripheral nerve repair, using biological and semi-synthetic hydrogels that enable controlled environmental stimuli to regenerate neurons and glial cells in a rat sciatic nerve resection model. The provisional nerve growth conduits were composed of either natural fibrin or adducts of synthetic polyethylene glycol and fibrinogen or gelatin. A photo-patterning technique was further applied to these 3D hydrogel biomaterials, in the form of laser-ablated microchannels, to provide contact guidance for unidirectional growth following sciatic nerve injury. We tested the regeneration capacity of subcritical nerve gap injuries in rats treated with photo-patterned materials and compared these with injuries treated with unpatterned hydrogels, either stiff or compliant. Among the factors tested were shear modulus, biological composition, and micropatterning of the materials. The microchannel guidance patterns, combined with appropriately matched degradation and stiffness properties of the material, proved most essential for the uniform tissue propagation during the nerve regeneration process.
- Published
- 2018
- Full Text
- View/download PDF
18. Transplantation of human amnion prevents recurring adhesions and ameliorates fibrosis in a rat model of sciatic nerve scarring
- Author
-
Rupert L. Mayer, Susanne Wolbank, Carina Penzenstadler, James Ferguson, Angela Lemke, Heinz Redl, Monika Bradl, Kelly Gross, and Christopher Gerner
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Pathology ,Biomedical Engineering ,Tissue Adhesions ,Biochemistry ,Regenerative medicine ,Rats, Sprague-Dawley ,Biomaterials ,Cicatrix ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Fibrosis ,Cell Adhesion ,medicine ,Animals ,Humans ,Amnion ,Molecular Biology ,Fetus ,business.industry ,Regeneration (biology) ,Recovery of Function ,General Medicine ,medicine.disease ,Sciatic Nerve ,Nerve Regeneration ,Surgery ,Transplantation ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Amniotic epithelial cells ,NIH 3T3 Cells ,Female ,Sciatic nerve ,business ,030217 neurology & neurosurgery ,Biotechnology - Abstract
Peripheral nerve fibrosis and painful adhesions are common, recurring pathological sequelae following injury. In this study, vital human amnion (hAM), an increasingly interesting biomaterial for regenerative medicine, was investigated as a novel therapy. hAM was first analyzed in vitro regarding its anti-adhesive characteristics. Then, the reflected region of hAM which was identified as more suitable, was transplanted into female Sprague Dawley rats with recurring sciatic nerve scarring (n = 24) and compared with untreated controls (n = 30) at one, four and twelve weeks. Immune response and fibrosis were investigated by (immuno)histochemical analysis. Nerve structure was examined and function determined using electrophysiology and gait analysis. Here we identified strongly reduced adhesions in the hAM-treated rats, displaying a significant difference at four weeks post transplantation compared to untreated controls (p = .0052). This correlated with the in vitro cell attachment test on hAM explants, which demonstrated a distinctly limited ability of fibroblasts to adhere to amniotic epithelial cells. Upon hAM transplantation, significantly less intraneural fibrosis was identified at the later time points. Moreover, hAM-treated rats exhibited a significantly higher sciatic functional index (SFI) after four weeks compared to controls (p .05), which indicated a potentially pro-regenerative effect of hAM. As a possible explanation, an impact of hAM on the endogenous immune response, including T cell and macrophage subsets, was indicated. We conclude that hAM is strongly effective against recurring nerve scarring and induces an anti-fibrotic and pro-regenerative effect, making it highly promising for treating adhesion-related disorders.Abnormal fibrotic bonding of tissues, frequently involving peripheral nerves, affects millions of people worldwide. These so-called adhesions usually cause severe pain and drastically reduce quality of life. To date, no adequate treatment exists and none is routinely used in the clinical practice. In this study, vital human amnion, the innermost of the fetal membranes, was transplanted in a rat model of peripheral nerve scarring and recurring adhesions as novel therapeutic approach. Amniotic cells have already demonstrated to feature stem-cell like properties and produce pro-regenerative factors, which makes the amnion an increasingly promising biomaterial for regenerative medicine. We identified that its transplantation was very effective against peripheral nerve scarring and distinctly reduced recurring adhesions. Moreover, we identified a pro-regenerative effect. This study showed that the amnion is a highly promising novel therapeutic approach for adhesion-related disorders.
- Published
- 2018
- Full Text
- View/download PDF
19. Cellular and Site-Specific Mitochondrial Characterization of Vital Human Amniotic Membrane
- Author
-
Heinz Redl, Simone Hennerbichler, Asmita Banerjee, Ralf Steinborn, Philipp Steindorf, Susanne Wolbank, Andrey V. Kozlov, Andrea Lindenmair, and Adelheid Weidinger
- Subjects
0301 basic medicine ,Biomedical Engineering ,lcsh:Medicine ,Mitochondrion ,03 medical and health sciences ,Adenosine Triphosphate ,human amniotic membrane ,Humans ,Amnion ,reactive oxygen species ,chemistry.chemical_classification ,Transplantation ,Reactive oxygen species ,lcsh:R ,Cell Differentiation ,Mesenchymal Stem Cells ,human amniotic mesenchymal stromal cells ,Articles ,Cell Biology ,Cell biology ,mitochondria ,human amniotic epithelial cells ,030104 developmental biology ,Membrane ,chemistry ,Stromal Cells - Abstract
Over a century ago, clinicians started to use the human amniotic membrane for coverage of wounds and burn injuries. To date, literally thousands of different clinical applications exist for this biomaterial almost exclusively in a decellularized or denuded form. Recent reconsiderations for the use of vital human amniotic membrane for clinical applications would take advantage of the versatile cells of embryonic origin including the entirety of their cell organelles. Recently, more and more evidence was found, showing mitochondria to be involved in most fundamental cellular processes, such as differentiation and cell death. In this study, we focused on specific properties of mitochondria of vital human amniotic membrane and characterized bioenergetical parameters of 2 subregions of the human amniotic membrane, the placental and reflected amnion. We found significantly different levels of adenosine triphosphate (ATP) and extracellular reactive oxygen species, concentrations of succinate dehydrogenase, and lactate upon inhibition of ATP synthase in placental and reflected amnion. We also found significantly different rates of mitochondrial respiration in isolated human amniotic epithelial cells and human amniotic mesenchymal stromal cells, according to the subregions. Differences in metabolic activities were inversely related to mitochondrial DNA copy numbers in isolated cells of placental and reflected amnion. Based on significant differences of several key parameters of energy metabolism in 2 subregions of vital amnion, we propose that these metabolic differences of vital placental and reflected amnion could have critical impact on therapeutic applications. Inclusion of region-specific metabolic properties could optimize and fine-tune the clinical application of the human amniotic membrane and improve the outcome significantly.
- Published
- 2018
- Full Text
- View/download PDF
20. Extracorporeal shock wave therapy in situ — novel approach to obtain an activated fat graft
- Author
-
Susanne Wolbank, J. Maier, Anja Peterbauer, Carolin Steffenhagen, Matthias Sandhofer, Wolfgang Holnthoner, Sylvia Nuernberger, Heinz Redl, Eleni Priglinger, and Christoph Wurzer
- Subjects
Extracorporeal Shockwave Therapy ,0301 basic medicine ,Cell Survival ,Angiogenesis ,Facial rejuvenation ,Cell ,Biomedical Engineering ,Medicine (miscellaneous) ,Adipose tissue ,Biomaterials ,Andrology ,03 medical and health sciences ,Adenosine Triphosphate ,0302 clinical medicine ,medicine ,Humans ,Cell Proliferation ,Adipogenesis ,Chemistry ,Mesenchymal stem cell ,Cell Differentiation ,Stromal vascular fraction ,030104 developmental biology ,medicine.anatomical_structure ,Secretory protein ,Adipose Tissue ,030220 oncology & carcinogenesis ,Immunology ,Female ,Stromal Cells ,Biomarkers - Abstract
One of the mainstays of facial rejuvenation strategies is volume restoration, which can be achieved by autologous fat grafting. In our novel approach, we treated the adipose tissue harvest site with extracorporeal shock wave therapy (ESWT) in order to improve the quality of the regenerative cells in situ. The latter was demonstrated by characterizing the cells of the stromal vascular fraction (SVF) in the harvested liposuction material regarding cell yield, adenosine triphosphate (ATP) content, proliferative capacity, surface marker profile, differentiation potential and secretory protein profile. Although the SVF cell yield was only slightly enhanced, viability and ATP concentration of freshly isolated cells as well as proliferation doublings after 3 weeks in culture were significantly increased in the ESWT compared with the untreated group. Likewise, cells expressing mesenchymal and endothelial/pericytic markers were significantly elevated concomitant with an improved differentiation capacity towards the adipogenic lineage and enhancement in specific angiogenic proteins. Hence, in situ ESWT might be applied in the future to promote cell fitness, adipogenesis and angiogenesis within the fat graft for successful facial rejuvenation strategies with potential long-term graft survival.
- Published
- 2017
- Full Text
- View/download PDF
21. A novel experimental rat model of peripheral nerve scarring that reliably mimics post-surgical complications and recurring adhesions
- Author
-
Heinz Redl, Monika Bradl, James Ferguson, Angela Lemke, Dominika Lidinsky, Susanne Wolbank, Carina Penzenstadler, Rudolf Hopf, and Thomas Hausner
- Subjects
Pathology ,T-Lymphocytes ,Medicine (miscellaneous) ,Action Potentials ,lcsh:Medicine ,Tissue Adhesions ,Rats, Sprague-Dawley ,0302 clinical medicine ,Postoperative Complications ,Immunology and Microbiology (miscellaneous) ,Fibrosis ,Recurrence ,Gait ,Neurolysis ,Nerve fibrosis ,Nerve scarring ,Chronic pain ,Anatomy ,Peripheral nerve adhesions ,Sciatic Nerve ,Peripheral ,030220 oncology & carcinogenesis ,Female ,Sciatic nerve ,medicine.symptom ,Research Article ,lcsh:RB1-214 ,medicine.medical_specialty ,Neuroscience (miscellaneous) ,Inflammation ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Cicatrix ,medicine ,lcsh:Pathology ,Animals ,Pathological ,business.industry ,Macrophages ,lcsh:R ,Nerve injury ,Nerve inflammation ,medicine.disease ,Perineural adhesions ,Disease Models, Animal ,Glutaral ,Nerve Degeneration ,business ,030217 neurology & neurosurgery - Abstract
Inflammation, fibrosis and perineural adhesions with the surrounding tissue are common pathological processes following nerve injury and surgical interventions on peripheral nerves in human patients. These features can reoccur following external neurolysis, currently the most common surgical treatment for peripheral nerve scarring, thus leading to renewed nerve function impairment and chronic pain. To enable a successful evaluation of new therapeutic approaches, it is crucial to use a reproducible animal model that mimics the main clinical symptoms occurring in human patients. However, a clinically relevant model combining both histological and functional alterations has not been published to date. We therefore developed a reliable rat model that exhibits the essential pathological processes of peripheral nerve scarring. In our study, we present a novel method for the induction of nerve scarring by applying glutaraldehyde-containing glue that is known to cause nerve injury in humans. After a 3-week contact period with the sciatic nerve in female Sprague Dawley rats, we could demonstrate severe intra- and perineural scarring that resulted in grade 3 adhesions and major impairments in the electrophysiological peak amplitude compared with sham control (P=0.0478). Immunohistochemical analysis of the nerve structure revealed vigorous nerve inflammation and recruitment of T cells and macrophages. Also, distinct nerve degeneration was determined by immunostaining. These pathological alterations were further reflected in significant functional deficiencies, as determined by the analysis of relevant gait parameters as well as the quantification of the sciatic functional index starting at week 1 post-operation (P, Summary: An easily reproducible and reliable rat model for peripheral nerve scarring that allows for the effective testing of new therapeutic strategies.
- Published
- 2017
22. The adipose tissue–derived stromal vascular fraction cells from lipedema patients: Are they different?
- Author
-
Matthias Sandhofer, Carolin Steffenhagen, Victoria Hofer, Anja Peterbauer, Heinz Redl, J. Maier, Christoph Wurzer, Susanne Wolbank, Eleni Priglinger, and Sylvia Nuernberger
- Subjects
Adult ,0301 basic medicine ,Cancer Research ,Pathology ,medicine.medical_specialty ,Stromal cell ,Immunology ,Population ,Adipose tissue ,CD146 Antigen ,Biology ,030207 dermatology & venereal diseases ,03 medical and health sciences ,Adenosine Triphosphate ,0302 clinical medicine ,medicine ,Humans ,Immunology and Allergy ,CD90 ,education ,Cells, Cultured ,Genetics (clinical) ,Transplantation ,education.field_of_study ,Adipogenesis ,Lipedema ,Stem Cells ,Mesenchymal stem cell ,Cell Differentiation ,Cell Biology ,Middle Aged ,Stromal vascular fraction ,030104 developmental biology ,Adipose Tissue ,Oncology ,Case-Control Studies ,Thy-1 Antigens ,CD146 ,Female ,Stromal Cells ,Stem cell - Abstract
Background aims Lipedema is a hormone-related disease of women characterized by enlargement of the extremities caused by subcutaneous deposition of adipose tissue. In healthy patients application of autologous adipose tissue–derived cells has shown great potential in several clinical studies for engrafting of soft tissue reconstruction in recent decades. The majority of these studies have used the stromal vascular fraction (SVF), a heterogeneous cell population containing adipose-derived stromal/stem cells (ASC), among others. Because cell identity and regenerative properties might be affected by the health condition of patients, we characterized the SVF cells of 30 lipedema patients in comparison to 22 healthy patients. Methods SVF cells were analyzed regarding cell yield, viability, adenosine triphosphate content, colony forming units and proliferative capacity, as well as surface marker profile and differentiation potential in vitro. Results Our results demonstrated a significantly enhanced SVF cell yield isolated from lipedema compared with healthy patients. In contrast, the adipogenic differentiation potential of SVF cells isolated from lipedema patients was significantly reduced compared with healthy patients. Interestingly, expression of the mesenchymal marker CD90 and the endothelial/pericytic marker CD146 was significantly enhanced when isolated from lipedema patients. Discussion The enhanced number of CD90+ and CD146+ cells could explain the increased cell yield because the other tested surface marker were not reduced in lipedema patients. Because the cellular mechanism and composition in lipedema is largely unknown, our findings might contribute to a better understanding of its etiology.
- Published
- 2017
- Full Text
- View/download PDF
23. Cell Engineering and Regeneration
- Author
-
Jeffrey M. Gimble, Susanne Wolbank, Darja Marolt, Richard O.C. Oreffo, and Heinz Redl
- Subjects
Cell engineering ,business.industry ,Regeneration (biology) ,Medicine ,business ,Cell biology - Published
- 2020
- Full Text
- View/download PDF
24. Perinatal derivatives: where do we stand? A roadmap of the human placenta and consensus for tissue and cell nomenclature
- Author
-
Antonietta Rosa Silini, Roberta Di Pietro, Ingrid Lang-Olip, Francesco Alviano, Asmita Banerjee, Mariangela Basile, Veronika Borutinskaite, Günther Eissner, Alexandra Gellhaus, Bernd Giebel, Yong-Can Huang, Aleksandar Janev, Mateja Erdani Kreft, Nadja Kupper, Ana Clara Abadía-Molina, Enrique G. Olivares, Assunta Pandolfi, Andrea Papait, Michela Pozzobon, Carmen Ruiz-Ruiz, Olga Soritau, Sergiu Susman, Dariusz Szukiewicz, Adelheid Weidinger, Susanne Wolbank, Berthold Huppertz, Ornella Parolini, Silini A.R., Di Pietro R., Lang-Olip I., Alviano F., Banerjee A., Basile M., Borutinskaite V., Eissner G., Gellhaus A., Giebel B., Huang Y.-C., Janev A., Kreft M.E., Kupper N., Abadia-Molina A.C., Olivares E.G., Pandolfi A., Papait A., Pozzobon M., Ruiz-Ruiz C., Soritau O., Susman S., Szukiewicz D., Weidinger A., Wolbank S., Huppertz B., and Parolini O.
- Subjects
0301 basic medicine ,Histology ,placenta ,lcsh:Biotechnology ,Cell ,Biomedical Engineering ,Medizin ,Consensus criteria ,Bioengineering ,Review ,cells ,consensus nomenclature ,derivatives ,fetal annexes ,perinatal ,tissues ,Biology ,Bioinformatics ,Cell morphology ,03 medical and health sciences ,0302 clinical medicine ,lcsh:TP248.13-248.65 ,Placenta ,medicine ,derivative ,Settore BIO/13 - BIOLOGIA APPLICATA ,fetal annexe ,Bioengineering and Biotechnology ,Human placenta ,cell ,3. Good health ,Clinical trial ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Identification (biology) ,Biotechnology - Abstract
Progress in the understanding of the biology of perinatal tissues has contributed to the breakthrough revelation of the therapeutic effects of perinatal derivatives (PnD), namely birth-associated tissues, cells, and secreted factors. The significant knowledge acquired in the past two decades, along with the increasing interest in perinatal derivatives, fuels an urgent need for the precise identification of PnD and the establishment of updated consensus criteria policies for their characterization. The aim of this review is not to go into detail on preclinical or clinical trials, but rather we address specific issues that are relevant for the definition/characterization of perinatal cells, starting from an understanding of the development of the human placenta, its structure, and the different cell populations that can be isolated from the different perinatal tissues. We describe where the cells are located within the placenta and their cell morphology and phenotype. We also propose nomenclature for the cell populations and derivatives discussed herein. This review is a joint effort from the COST SPRINT Action (CA17116), which broadly aims at approaching consensus for different aspects of PnD research, such as providing inputs for future standards for the processing and in vitro characterization and clinical application of PnD., Austrian Science Fund (FWF) DOC 31-B26, Medical University Graz, Universita Cattolica del Sacro Cuore, PRIN 2017 program of Italian Ministry of Research and University (MIUR) 2017RSAFK7, Ministry of Health, Italy GR-2018-12366992, Slovenian Research Agency - Slovenia P3-0108, MRIC UL IP-0510, Plan Estatal de Investigacion Cientifica y Tecnica y de Innovacion, ISCIII Subdireccion General de Evaluacion y Fomento de la Investigacion, Ministerio de Economia y Competitividad, Spain PI16/01642, European Union (EU), European Community (EC), German Research Foundation (DFG) GE-2223/2-1
- Published
- 2020
25. A novel fluorescent hydroxyapatite based on iron quantum cluster template to enhance osteogenic differentiation
- Author
-
Alireza Naderi Sohi, Veronika Hruschka, Susanne Wolbank, Darja Marolt Presen, Hossein Naderi-Manesh, Zahra Vaezi, Somayeh Khanmohammadi, Naimeh Hashemi, Heinz Redl, and Saeed Masoumi
- Subjects
Bone sialoprotein ,Stromal cell ,Materials science ,Cell Survival ,Iron ,Osteocalcin ,Cell Culture Techniques ,Bioengineering ,Biocompatible Materials ,Core Binding Factor Alpha 1 Subunit ,02 engineering and technology ,010402 general chemistry ,01 natural sciences ,Biomaterials ,stomatognathic system ,Osteogenesis ,Quantum Dots ,Humans ,Cells, Cultured ,Fluorescent Dyes ,biology ,Mesenchymal stem cell ,Biomaterial ,Cell Differentiation ,Mesenchymal Stem Cells ,021001 nanoscience & nanotechnology ,Alkaline Phosphatase ,Fluorescence ,Microspheres ,0104 chemical sciences ,RUNX2 ,Durapatite ,Mechanics of Materials ,biology.protein ,Biophysics ,Alkaline phosphatase ,Collagen ,0210 nano-technology - Abstract
Template-mediated self-assembly synthesis has produced a diverse range of biomimetic materials with unique physicochemical properties. Here, we fabricated novel fluorescent three-dimensional (3-D) hydroxyapatite (HAP) nanorod-assembled microspheres using iron quantum cluster (FeQC) as a hybrid template, containing three organic components: hemoglobin chains, piperidine, and iron clusters. The material characterization indicated that the synthesized HAP possessed a uniform rod-like morphology, ordered 3-D architecture, high crystallinity, self-activated fluorescence, and remarkable photostability. Our study proposed that this FeQC template is a promising regulating agent to fabricate fluorescent self-assembled HAP microspheres with a controlled morphology. The effect of HAP on stem cell fate and their osteogenic differentiation was investigated by culturing human bone marrow-derived mesenchymal stromal/stem cells (BMSCs) with HAP microspheres. Significant increases in collagen matrix production and gene expression of osteogenic markers, including osteocalcin (OCN), Runt-related transcription factor 2 (Runx2), bone sialoprotein (BSP) and alkaline phosphatase (ALP), were observed compared to the controls after 21 days of culture. Taken together, our data suggest that synthetic HAP nanorod-assembled microspheres represent a promising new biomaterial which exhibits enhanced fluorescent properties and osteoinductive effects on human BMSCs.
- Published
- 2019
26. SVF-derived extracellular vesicles carry characteristic miRNAs in lipedema
- Author
-
Moritz Weigl, Susanne Wolbank, Martin Barsch, Jaroslaw Jacak, Heinz Redl, Eleni Priglinger, Carolin Lindner, Karin Strohmeier, Matthias Hackl, Johannes Grillari, and Matthias Sandhofer
- Subjects
Pathology ,medicine.medical_specialty ,microRNA ,Significant difference ,Extracellular ,medicine ,Conditioned medium ,Adipose tissue ,Stromal vascular fraction ,Biology ,Extracellular vesicles - Abstract
Lipedema is a chronic, progressive disease of adipose tissue with lack of consistent diagnostic criteria. The aim of this study was a thorough comparative characterization of extracellular microRNAs from the stromal vascular fraction (SVF) of healthy and lipedema adipose tissue. For this, we analyzed 187 extracellular microRNAs in concentrated conditioned media (cCM) and specifically in small extracellular vesicles (sEVs) enriched thereof by size exclusion chromatography. No significant difference in median particle size and concentration was observed between sEV fractions in healthy and lipedema. We found the majority of miRNAs located predominantly in cCM compared to sEV enriched fraction. Surprisingly, hierarchical clustering of the most variant miRNAs showed that only sEV miRNA profiles – but not cCM miRNAs – were impacted by lipedema. Seven sEV miRNAs (miR–16-5p, miR-29a-3p, miR-24-3p, miR-454-p, miR–144-5p, miR-130a-3p, let-7c-5p) were differently regulated in lipedema and healthy, whereas only one cCM miRNA (miR-188-5p) was significantly downregulated in lipedema. Comparing SVF from healthy and lipedema patients, we identified sEVs as the lipedema relevant miRNA fraction. This study contributes to identify the potential role of SVF secreted miRNAs in lipedema.
- Published
- 2019
- Full Text
- View/download PDF
27. Autonomous spheroid formation by culture plate compartmentation
- Author
-
Marian Fürsatz, Sylvia Nürnberger, Peter Gerges, and Susanne Wolbank
- Subjects
Tissue Engineering ,Chemistry ,Regeneration (biology) ,Cartilage ,Biomedical Engineering ,Spheroid ,Cell Differentiation ,Bioengineering ,General Medicine ,Matrix (biology) ,Chondrogenesis ,Biochemistry ,Biomaterials ,Chondrocytes ,medicine.anatomical_structure ,Tissue engineering ,Cell culture ,medicine ,Humans ,Cell Culture Techniques, Three Dimensional ,Stem cell ,Cells, Cultured ,Biotechnology ,Biomedical engineering - Abstract
Scaffold-free 3D cell cultures (e.g. pellet cultures) are widely used in medical science, including cartilage regeneration. Their drawbacks are high time/reagent consumption and lack of early readout parameters. While optimisation was achieved by automation or simplified spheroid generation, most culture systems remain expensive or require tedious procedures. The aim of this study was to establish a system for resource efficient spheroid generation with additional early readout parameters. This was achieved by a new approach for spheroid generation via self-assembly from monolayer via compartmentation of cell culture surfaces utilising laser engraving (grid plates). The compartmentation triggered contraction and rolling up of the cell monolayer, finishing in condensation into a spheroid in human adipose-derived stem cell (ASC/TERT1) and human articular chondrocytes (hACs)-ASC/TERT1 co-cultures, when cultivated on grid plates under chondrogenic conditions. Plates with 1 and 3 mm grid size yielded stable diameters (about 140 µm and 300 µm, respectively). ASC/TERT1 spheroids fully formed within 3 weeks while co-cultures took 1–2 weeks, forming significantly faster with increasing hAC ratio (p < 0.05 and 0.01 for 1:1 and 1:4 ASC/TERT1:hAC ratio, respectively). Co-cultures showed slightly lower spheroid diameters, due to earlier spheroid formation and incomplete monolayer formation. However, this was associated with a more homogeneous matrix distribution in the co-culture. Both showed differentiation capacity comparable to standard pellet culture in (immune-)histochemistry and RT-qPCR. To assess usability for cartilage repair, spheroids were embedded into a hydrogel (fibrin), yielding cellular outgrowth and matrix deposition, which was especially pronounced in co-cultures. The herein presented novel cell culture system is not only a promising tool for autonomous spheroid generation with the potential of experimental and clinical application in tissue engineering, but also for the generation of ‘building blocks’ for subsequential biofabrication strategies such as bioprinting.
- Published
- 2021
- Full Text
- View/download PDF
28. A NoninvasiveIn VitroMonitoring System Reporting Skeletal Muscle Differentiation
- Author
-
Deniz Öztürk-Kaloglu, Heinz Redl, Susanne Wolbank, Philipp Heher, David Hercher, Alice Zimmermann, Katja Posa-Markaryan, Ara Hacobian, and Simon Sperger
- Subjects
0301 basic medicine ,Cellular differentiation ,Green Fluorescent Proteins ,Cell ,Biomedical Engineering ,Medicine (miscellaneous) ,Bioengineering ,In Vitro Techniques ,Signal-To-Noise Ratio ,Biology ,Muscle Development ,Green fluorescent protein ,Mice ,03 medical and health sciences ,0302 clinical medicine ,microRNA ,medicine ,Animals ,Luciferase ,Muscle, Skeletal ,Promoter Regions, Genetic ,Creatine Kinase ,Cells, Cultured ,Myogenesis ,Skeletal muscle ,Cell Differentiation ,Molecular biology ,In vitro ,Enhancer Elements, Genetic ,030104 developmental biology ,medicine.anatomical_structure ,Microscopy, Fluorescence ,030220 oncology & carcinogenesis - Abstract
Monitoring of cell differentiation is a crucial aspect of cell-based therapeutic strategies depending on tissue maturation. In this study, we have developed a noninvasive reporter system to trace murine skeletal muscle differentiation. Either a secreted bioluminescent reporter (Metridia luciferase) or a fluorescent reporter (green fluorescent protein [GFP]) was placed under the control of the truncated muscle creatine kinase (MCK) basal promoter enhanced by variable numbers of upstream MCK E-boxes. The engineered pE3MCK vector, coding a triple tandem of E-Boxes and the truncated MCK promoter, showed twentyfold higher levels of luciferase activation compared with a Cytomegalovirus (CMV) promoter. This newly developed reporter system allowed noninvasive monitoring of myogenic differentiation in a straining bioreactor. Additionally, binding sequences of endogenous microRNAs (miRNAs; seed sequences) that are known to be downregulated in myogenesis were ligated as complementary seed sequences into the reporter vector to reduce nonspecific signal background. The insertion of seed sequences improved the signal-to-noise ratio up to 25% compared with pE3MCK. Due to the highly specific, fast, and convenient expression analysis for cells undergoing myogenic differentiation, this reporter system provides a powerful tool for application in skeletal muscle tissue engineering.
- Published
- 2017
- Full Text
- View/download PDF
29. Effects of amniotic epithelial cell transplantation in endothelial injury
- Author
-
Gabriella Vácz, Andrea Lindenmair, Levente Kiss, Rita Benkő, Susanne Wolbank, Zsombor Lacza, Zsuzsanna Cserép, Eszter Pankotai, Dénes B. Horváthy, István Hornyák, Charlotte M. Schwarz, Endre Kovács, and Attila Cselenyák
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Cell ,Vasodilation ,030204 cardiovascular system & hematology ,Constriction ,03 medical and health sciences ,0302 clinical medicine ,medicine ,BrdU ,vascular injury ,amnion ,Original Paper ,endarterectomy ,Amnion ,business.industry ,amniotic epithelial cells ,General Medicine ,Anatomy ,Epithelium ,Transplantation ,030104 developmental biology ,medicine.anatomical_structure ,Amniotic epithelial cells ,cardiovascular system ,Immunohistochemistry ,business - Abstract
Purpose Human amniotic epithelial cells (hAECs) are promising tools for endothelial repair in vascular regenerative medicine. We hypothesized that these epithelial cells are capable of repairing the damaged endothelial layer following balloon injury of the carotid artery in adult male rats. Results Two days after injury, the transplanted hAECs were observed at the luminal side of the arterial wall. Then, 4 weeks after the injury, significant intimal thickening was observed in both untreated and cell implanted vessels. Constriction was decreased in both implanted and control animals. Immunohistochemical analysis showed a few surviving cells in the intact arterial wall, but no cells were observed at the site of injury. Interestingly, acetylcholine-induced dilation was preserved in the intact side and the sham-transplanted injured arteries, but it was a trend toward decreased vasodilation in the hAECs’ transplanted vessels. Conclusion We conclude that hAECs were able to incorporate into the arterial wall without immunosuppression, but failed to improve vascular function, highlighting that morphological implantation does not necessarily result in functional benefits and underscoring the need to understand other mechanisms of endothelial regeneration.
- Published
- 2016
- Full Text
- View/download PDF
30. Effect of Diphenyleneiodonium Chloride on Intracellular Reactive Oxygen Species Metabolism with Emphasis on NADPH Oxidase and Mitochondria in Two Therapeutically Relevant Human Cell Types
- Author
-
Sergejs Zavadskis, Susanne Wolbank, Adelheid Weidinger, Cornelia Schneider, Andrey V. Kozlov, Dominik Hanetseder, Darja Marolt Presen, and Asmita Banerjee
- Subjects
Cellular respiration ,proliferation ,lcsh:RS1-441 ,Pharmaceutical Science ,Mitochondrion ,Article ,lcsh:Pharmacy and materia medica ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,diphenyleneiodonium ,030304 developmental biology ,reactive oxygen species ,chemistry.chemical_classification ,0303 health sciences ,Reactive oxygen species ,Oxidase test ,NADPH oxidase ,biology ,Mesenchymal stem cell ,differentiation ,Cell biology ,mitochondria ,chemistry ,biology.protein ,Energy source ,NADPH-oxidase ,030217 neurology & neurosurgery ,Nicotinamide adenine dinucleotide phosphate - Abstract
Reactive oxygen species (ROS) have recently been recognized as important signal transducers, particularly regulating proliferation and differentiation of cells. Diphenyleneiodonium (DPI) is known as an inhibitor of the nicotinamide adenine dinucleotide phosphate oxidase (NOX) and is also affecting mitochondrial function. The aim of this study was to investigate the effect of DPI on ROS metabolism and mitochondrial function in human amniotic membrane mesenchymal stromal cells (hAMSCs), human bone marrow mesenchymal stromal cells (hBMSCs), hBMSCs induced into osteoblast-like cells, and osteosarcoma cell line MG-63. Our data suggested a combination of a membrane potential sensitive fluorescent dye, tetramethylrhodamine methyl ester (TMRM), and a ROS-sensitive dye, CM-H2DCFDA, combined with a pretreatment with mitochondria-targeted ROS scavenger MitoTEMPO as a good tool to examine effects of DPI. We observed critical differences in ROS metabolism between hAMSCs, hBMSCs, osteoblast-like cells, and MG-63 cells, which were linked to energy metabolism. In cell types using predominantly glycolysis as the energy source, such as hAMSCs, DPI predominantly interacted with NOX, and it was not toxic for the cells. In hBMSCs, the ROS turnover was influenced by NOX activity rather than by the mitochondria. In cells with aerobic metabolism, such as MG 63, the mitochondria became an additional target for DPI, and these cells were prone to the toxic effects of DPI. In summary, our data suggest that undifferentiated cells rather than differentiated parenchymal cells should be considered as potential targets for DPI.
- Published
- 2020
- Full Text
- View/download PDF
31. Repopulation of an auricular cartilage scaffold, AuriScaff, perforated with an enzyme combination
- Author
-
Christoph Schneider, G.V.M. van Osch, Xavier Monforte, Susanne Wolbank, Bernhard Rieder, Severin Mühleder, Heinz Redl, Andreas H. Teuschl, B. Schädl, Sylvia Nürnberger, Claudia Keibl, Wolfgang Holnthoner, Claudia Gahleitner, and Otorhinolaryngology and Head and Neck Surgery
- Subjects
Male ,Scaffold ,Compressive Strength ,0206 medical engineering ,Biomedical Engineering ,02 engineering and technology ,Matrix (biology) ,Biochemistry ,Biomaterials ,Prosthesis Implantation ,Chondrocytes ,Tissue engineering ,medicine ,Animals ,Humans ,Molecular Biology ,Collagen Type II ,Cellular Senescence ,Glycosaminoglycans ,Decellularization ,Tissue Scaffolds ,Chemistry ,Hyaline cartilage ,Regeneration (biology) ,Cartilage ,Cell Differentiation ,General Medicine ,DNA ,Middle Aged ,021001 nanoscience & nanotechnology ,Chondrogenesis ,020601 biomedical engineering ,medicine.anatomical_structure ,Cattle ,Female ,Ear Cartilage ,0210 nano-technology ,Biotechnology ,Biomedical engineering - Abstract
Biomaterials currently in use for articular cartilage regeneration do not mimic the composition or architecture of hyaline cartilage, leading to the formation of repair tissue with inferior characteristics. In this study we demonstrate the use of “AuriScaff”, an enzymatically perforated bovine auricular cartilage scaffold, as a novel biomaterial for repopulation with regenerative cells and for the formation of high-quality hyaline cartilage. AuriScaff features a traversing channel network, generated by selective depletion of elastic fibers, enabling uniform repopulation with therapeutic cells. The complex collagen type II matrix is left intact, as observed by immunohistochemistry, SEM and TEM. The compressive modulus is diminished, but three times higher than in the clinically used collagen type I/III scaffold that served as control. Seeding tests with human articular chondrocytes (hAC) alone and in co-culture with human adipose-derived stromal/stem cells (ASC) confirmed that the network enabled cell migration throughout the scaffold. It also guides collagen alignment along the channels and, due to the generally traverse channel alignment, newly deposited cartilage matrix corresponds with the orientation of collagen within articular cartilage. In an osteochondral plug model, AuriScaff filled the complete defect with compact collagen type II matrix and enabled chondrogenic differentiation inside the channels. Using adult articular chondrocytes from bovine origin (bAC), filling of even deep defects with high-quality hyaline-like cartilage was achieved after 6 weeks in vivo. With its composition and spatial organization, AuriScaff provides an optimal chondrogenic environment for therapeutic cells to treat cartilage defects and is expected to improve long-term outcome by channel-guided repopulation followed by matrix deposition and alignment. Statement of Significance After two decades of tissue engineering for cartilage regeneration, there is still no optimal strategy available to overcome problems such as inconsistent clinical outcome, early and late graft failures. Especially large defects are dependent on biomaterials and their scaffolding, guiding and protective function. Considering the currently used biomaterials, structure and mechanical properties appear to be insufficient to fulfill this task. The novel scaffold developed within this study is the first approach enabling the use of dense cartilage matrix, repopulate it via channels and provide the cells with a compact collagen type II environment. Due to its density, it also provides better mechanical properties than materials currently used in clinics. We therefore think, that the auricular cartilage scaffold (AuriScaff) has a high potential to improve future cartilage regeneration approaches.
- Published
- 2018
32. Cardiac Restoration Stemming From the Placenta Tree: Insights From Fetal and Perinatal Cell Biology
- Author
-
Sveva Bollini, Antonietta R. Silini, Asmita Banerjee, Susanne Wolbank, Carolina Balbi, and Ornella Parolini
- Subjects
0301 basic medicine ,Amniotic fluid ,Modern medicine ,Physiology ,Placenta ,medicine.medical_treatment ,Paracrine effect ,Review ,Cardiomyocyte ,Cardioprotection ,Bioinformatics ,lcsh:Physiology ,Immunomodulation ,Cardiac repair ,03 medical and health sciences ,Physiology (medical) ,medicine ,Settore BIO/13 - BIOLOGIA APPLICATA ,Progenitor cell ,Umbilical cord ,Heart transplantation ,Cardiotoxicity ,lcsh:QP1-981 ,business.industry ,Regeneration (biology) ,Amniotic fluid, Cardiac repair, Cardiomyocyte, Cardioprotection, Immunomodulation, Paracrine effect, Placenta, Umbilical cord, Physiology, Physiology (medical) ,medicine.disease ,030104 developmental biology ,Heart failure ,Stem cell ,business ,Reprogramming - Abstract
Efficient cardiac repair and ultimate regeneration still represents one of the main challenges of modern medicine. Indeed, cardiovascular disease can derive from independent conditions upsetting heart structure and performance: myocardial ischemia and infarction (MI), pharmacological cardiotoxicity, and congenital heart defects, just to name a few. All these disorders have profound consequences on cardiac tissue, inducing the onset of heart failure over time. Since the cure is currently represented by heart transplantation, which is extremely difficult due to the shortage of donors, much effort is being dedicated to developing innovative therapeutic strategies based on stem cell exploitation. Among the broad scenario of stem/progenitor cell subpopulations, fetal and perinatal sources, namely amniotic fluid and term placenta, have gained interest due to their peculiar regenerative capacity, high self-renewal capability, and ease of collection from clinical waste material. In this review, we will provide the state-of-the-art on fetal perinatal stem cells for cardiac repair and regeneration. We will discuss different pathological conditions and the main therapeutic strategies proposed, including cell transplantation, putative paracrine therapy, reprogramming, and tissue engineering approaches.
- Published
- 2018
- Full Text
- View/download PDF
33. Matrix Metalloproteinase-2 Impairs Homing of Intracoronary Delivered Mesenchymal Stem Cells in a Porcine Reperfused Myocardial Infarction: Comparison With Intramyocardial Cell Delivery
- Author
-
Susanne Wolbank, Christoph Kaun, Ljubica Mandic, Rayyan Hemetsberger, Katrin Zlabinger, Mariann Gyöngyösi, Imre Repa, Noemi Nyolczas, Rainer de Martin, Zsolt Petrasi, Florian Gruber, Denise Traxler, Dominika Lukovic, Alfred Gugerell, Aniko Posa, Johannes Winkler, Dara L. Kraitchman, Kurt Huber, Renate Hofer-Warbinek, Andrea Gyenes, Örs Petneházy, Gerald Zanoni, Imre J. Pavo, Noemi Pavo, Silvia Charwat, and Andreas Spannbauer
- Subjects
0301 basic medicine ,Histology ,Receptor expression ,lcsh:Biotechnology ,Biomedical Engineering ,Bioengineering ,030204 cardiovascular system & hematology ,intracoronary ,CXCR4 ,Andrology ,Cell therapy ,03 medical and health sciences ,ischemic injured heart tissue ,0302 clinical medicine ,cell delivery ,lcsh:TP248.13-248.65 ,Medicine ,oxidative stress ,Original Research ,mesenchymal stem cells ,Ejection fraction ,business.industry ,Mesenchymal stem cell ,Bioengineering and Biotechnology ,030104 developmental biology ,translational research ,intramyocardial ,Stem cell ,homing ,business ,Perfusion ,Biotechnology ,Homing (hematopoietic) - Abstract
Background Intracoronary (IC) injection of mesenchymal stem cells (MSCs) results in a prompt decrease of absolute myocardial blood flow (AMF) with late and incomplete recovery of myocardial tissue perfusion. Here, we investigated the effect of decreased AMF on oxidative stress marker matrix metalloproteinase-2 (MMP-2) and its influence on the fate and homing and paracrine character of MSCs after IC or intramyocardial cell delivery in a closed-chest reperfused myocardial infarction (MI) model in pigs. Methods Porcine MSCs were transiently transfected with Ad-Luc and Ad-green fluorescent protein (GFP). One week after MI, the GFP-Luc-MSCs were injected either IC (group IC, 11.00 ± 1.07 × 106) or intramyocardially (group IM, 9.88 ± 1.44 × 106). AMF was measured before, immediately after, and 24 h post GFP-Luc-MSC delivery. In vitro bioluminescence signal was used to identify tissue samples containing GFP-Luc-MSCs. Myocardial tissue MMP-2 and CXCR4 receptor expression (index of homing signal) were measured in bioluminescence positive and negative infarcted and border, and non-ischemic myocardial areas 1-day post cell transfer. At 7-day follow-up, myocardial homing (cadherin, CXCR4, and stromal derived factor-1alpha) and angiogenic [fibroblast growth factor 2 (FGF2) and VEGF] were quantified by ELISA of homogenized myocardial tissues from the bioluminescence positive and negative infarcted and border, and non-ischemic myocardium. Biodistribution of the implanted cells was quantified by using Luciferase assay and confirmed by fluorescence immunochemistry. Global left ventricular ejection fraction (LVEF) was measured at baseline and 1-month post cell therapy using magnet resonance image. Results AMF decreased immediately after IC cell delivery, while no change in tissue perfusion was found in the IM group (42.6 ± 11.7 vs. 56.9 ± 16.7 ml/min, p = 0.018). IC delivery led to a significant increase in myocardial MMP-2 64 kD expression (448 ± 88 vs. 315 ± 54 intensity × mm2, p = 0.021), and decreased expression of CXCR4 (592 ± 50 vs. 714 ± 54 pg/tissue/ml, p = 0.006), with significant exponential decay between MMP-2 and CXCR4 (r = 0.679, p
- Published
- 2018
- Full Text
- View/download PDF
34. VEGF released from a fibrin biomatrix increases VEGFR-2 expression and improves early outcome after ischaemia-reperfusion injury
- Author
-
Elizabeth R. Balmayor, Susanne Wolbank, Sabine Pfeifer, Heinz Redl, Rainer Mittermayr, Martina Moritz, and Martijn van Griensven
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Biomedical Engineering ,Ischemia ,Medicine (miscellaneous) ,Inflammation ,Hindlimb ,Fibrin ,Biomaterials ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,In vivo ,medicine ,biology ,business.industry ,Kinase insert domain receptor ,medicine.disease ,Vascular endothelial growth factor ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Myeloperoxidase ,biology.protein ,medicine.symptom ,business - Abstract
Skeletal ischaemia-reperfusion (I-R) injury may influence patient outcome after severe vascular trauma or clamping of major vessels. The aim of this study was to observe whether locally applied vascular endothelial growth factor (VEGF) in fibrin could induce the expression of VEGF-receptor-2 (VEGFR-2) and improve the outcome after I-R injury. Transgenic mice expressing VEGFR-2 promoter-controlled luciferase were used for the assessment of VEGFR-2 expression. Ischaemia was induced for 2 h by a tension-controlled tourniquet to the hind limb, followed by 24 h of reperfusion. The animals were locally injected subcutaneously with fibrin sealant containing 20 or 200 ng VEGF; control animals received no treatment or fibrin sealant application. In vivo VEGFR-2 expression was quantified upon administration of luciferin at several observation times. For oedema and inflammation quantification, wet:dry ratio measurements and a myeloperoxidase assay of the muscle tissue were performed. Laser Doppler imaging showed that ischaemia was present and that the blood flow had returned to baseline levels after 24 h of reperfusion. VEGFR-2 expression levels in the fibrin + 200 ng VEGF were significantly higher than in all other groups. Granulocyte infiltration was reduced in both treatment groups, as well as reduced oedema formation. These results showed that VEGF released from fibrin had a positive effect on early I-R outcome in a mouse model, possibly via VEGFR-2. Copyright © 2016 John Wiley & Sons, Ltd.
- Published
- 2016
- Full Text
- View/download PDF
35. Systematic Comparison of Protocols for the Preparation of Human Articular Cartilage for Use as Scaffold Material in Cartilage Tissue Engineering
- Author
-
Andreas H. Teuschl, Susanne Wolbank, Florian Hildner, Sylvia Nürnberger, Heinz Redl, Johannes Lehmann, Gerjo J.V.M. van Osch, Patrick Heimel, Xavier Monforte, Cornelia Schneider, David Miosga, Eleni Priglinger, Cell biology, Otorhinolaryngology and Head and Neck Surgery, and Orthopedics and Sports Medicine
- Subjects
Cartilage, Articular ,0301 basic medicine ,Scaffold ,Cell Survival ,Biomedical Engineering ,Medicine (miscellaneous) ,Bioengineering ,Glycosaminoglycan ,03 medical and health sciences ,Chondrocytes ,Tissue engineering ,medicine ,Humans ,Viability assay ,Cells, Cultured ,Decellularization ,Tissue Engineering ,Tissue Scaffolds ,Chemistry ,Cartilage ,Biomaterial ,Mesenchymal Stem Cells ,X-Ray Microtomography ,Extracellular Matrix ,Staining ,030104 developmental biology ,medicine.anatomical_structure ,Adipose Tissue ,Biomedical engineering - Abstract
Natural extracellular matrix-derived biomaterials from decellularized allogenic tissues are of increasing interest for tissue engineering because their structure and composition provide a complexity that is not achievable with current manufacturing techniques. The prerequisite to bring allogenic tissue from bench to bedside as a functional biomaterial is the full removal of cells while preserving most of its native characteristics such as structure and composition. The exceptionally dense structure of articular cartilage, however, poses a special challenge for decellularization, scaffold preparation, and reseeding. Therefore, we tested 24 different protocols aiming to remove cells and glycosaminoglycans (GAG) while preserving the collagen backbone and ultrastructure. The resulting matrices were analyzed for cell removal (DNA quantification, haematoxylin and eosin staining), GAG content (dimethyl methylene blue assay, Alcian blue staining and micro-computed tomography), collagen integrity (immunohistochemistry and ultrastructure), and biomechanics (compression test). Furthermore, seeding tests were conducted to evaluate cell viability and attachment to the scaffolds. Sodium dodecyl sulfate-based protocols yielded satisfactory reduction of DNA content, yet had negative effects on cell viability and attachment. Hydrochloric acid efficiently decellularized the scaffold and pepsin emerged as best option for GAG depletion. Combining these two reagents led to our final protocol, most efficient in DNA and GAG depletion while preserving the collagen architecture. The compressive modulus decreased in the absence of GAG to ∼1/3 of native cartilage, which is significantly higher than that by commercially available scaffolds tested as a reference (ranging from 1/25 to 1/100 of native cartilage). Cytocompatibility tests showed that human adipose-derived stromal cells readily adhered to the scaffold. In this study, we established a protocol combining freeze-thaw cycles, osmotic shock, and treatment with hydrochloric acid followed by a pepsin digestion step, achieving successful decellularization and GAG depletion within 1 week, resulting in a cytocompatible material with intact collagen structure. The protocol provides a basis for the generation of allogeneic scaffolds, potentially substituting manufactured scaffolds currently used in clinical articular cartilage treatment.
- Published
- 2016
36. Different metabolic activity in placental and reflected regions of the human amniotic membrane
- Author
-
Andrea Lindenmair, Heinz Redl, Ralf Steinborn, Martin Hofer, Simone Hennerbichler-Lugscheider, Johann Eibl, Asmita Banerjee, Susanne Wolbank, Andrey V. Kozlov, and Adelheid Weidinger
- Subjects
Membrane Potential, Mitochondrial ,chemistry.chemical_classification ,Reactive oxygen species ,Immunogenicity ,Cell Respiration ,Cell ,Obstetrics and Gynecology ,Amniotic stem cells ,Mitochondrion ,Biology ,Cell biology ,medicine.anatomical_structure ,Reproductive Medicine ,Tissue engineering ,chemistry ,Pregnancy ,Amniotic epithelial cells ,Immunology ,medicine ,Humans ,Female ,Amnion ,Stem cell ,Developmental Biology - Abstract
Cells of the human amniotic membrane (hAM) have stem cell characteristics with low immunogenicity and anti-inflammatory properties. While hAM is an excellent source for tissue engineering, so far, its sub-regions have not been taken into account. We show that placental and reflected hAM differ distinctly in morphology and functional activity, as the placental region has significantly higher mitochondrial activity, however significantly less reactive oxygen species. Since mitochondria may participate in processes such as cell rescue, we speculate that amniotic sub-regions may have different potential for tissue regeneration, which may be crucial for clinical applications.
- Published
- 2015
- Full Text
- View/download PDF
37. Oxygen Tension Strongly Influences Metabolic Parameters and the Release of Interleukin-6 of Human Amniotic Mesenchymal Stromal Cells
- Author
-
Heinz Redl, Susanne Wolbank, Sergiu Dumitrescu, Andrey V. Kozlov, Simone Hennerbichler, Andrea Lindenmair, Ralf Steinborn, Asmita Banerjee, and Adelheid Weidinger
- Subjects
0301 basic medicine ,chemistry.chemical_classification ,Reactive oxygen species ,lcsh:Internal medicine ,Article Subject ,Mesenchymal stem cell ,chemistry.chemical_element ,Cell Biology ,Oxidative phosphorylation ,Oxygen ,Nitric oxide ,Oxygen tension ,Andrology ,Cell therapy ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,chemistry ,In vivo ,lcsh:RC31-1245 ,Molecular Biology ,Research Article - Abstract
The human amniotic membrane (hAM) has been used for tissue regeneration for over a century.In vivo(in utero), cells of the hAM are exposed to low oxygen tension (1–4% oxygen), while the hAM is usually cultured in atmospheric, meaning high, oxygen tension (20% oxygen). We tested the influence of oxygen tensions on mitochondrial and inflammatory parameters of human amniotic mesenchymal stromal cells (hAMSCs). Freshly isolated hAMSCs were incubated for 4 days at 5% and 20% oxygen. We found 20% oxygen to strongly increase mitochondrial oxidative phosphorylation, especially in placental amniotic cells. Oxygen tension did not impact levels of reactive oxygen species (ROS); however, placental amniotic cells showed lower levels of ROS, independent of oxygen tension. In contrast, the release of nitric oxide was independent of the amniotic region but dependent on oxygen tension. Furthermore, IL-6 was significantly increased at 20% oxygen. To conclude, short-time cultivation at 20% oxygen of freshly isolated hAMSCs induced significant changes in mitochondrial function and release of IL-6. Depending on the therapeutic purpose, cultivation conditions of the cells should be chosen carefully for providing the best possible quality of cell therapy.
- Published
- 2018
38. Secretome Conveys the Protective Effects of ASCs: Therapeutic Potential Following Hemorrhagic Shock?
- Author
-
Susanne Wolbank, Heinz Redl, Carina Penzenstadler, Soheyl Bahrami, Arian Bahrami, Mohammad Jafarmadar, Mostafa Ashmwe, A Klotz, and Asmita Banerjee
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,Resuscitation ,Cell ,Renal function ,Inflammation ,Shock, Hemorrhagic ,Critical Care and Intensive Care Medicine ,Gastroenterology ,Rats, Sprague-Dawley ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Animals ,Shock, Traumatic ,Liver injury ,business.industry ,Stem Cells ,hemic and immune systems ,030208 emergency & critical care medicine ,medicine.disease ,Rats ,Traumatic Shock ,030104 developmental biology ,medicine.anatomical_structure ,Adipose Tissue ,Shock (circulatory) ,Culture Media, Conditioned ,Emergency Medicine ,medicine.symptom ,Stem cell ,business - Abstract
OBJECTIVES We tested whether resuscitation supplemented with rat adipose-derived stem cells (ASCs) or secretome (conditioned media) of ASCs can ameliorate inflammation, cell/organ injury, and/or improve outcome after hemorrhagic traumatic shock (HTS). INTERVENTIONS Rats were subjected to HTS and a resuscitation protocol that mimics prehospital restrictive reperfusion followed by an adequate reperfusion phase. Twenty minutes into the restrictive reperfusion, animals received an intravenous bolus of 2 × 10 cells (ASC group) or the secretome produced by 2 × 10 ASCs/24 h (ASC-Secretome group). Controls received the vehicle (Vehicle group). All rats were observed for 28-day survival. MEASUREMENTS AND MAIN RESULTS HTS-induced inflammation represented by IL-6 was inhibited in the ASC (80%, P
- Published
- 2017
39. Salamanders on the bench - A biocompatibility study of salamander skin secretions in cell cultures
- Author
-
Kristin A. Bakkegard, Ursula Dicke, Janek von Byern, Susanne Wolbank, Severin Mühleder, Alfred Gugerell, Sylvia Nürnberger, Heidemarie C. Fuchs, Dietrich Mebs, Egon Heiss, Oliver Wetjen, Ingo Grunwald, and Publica
- Subjects
0106 biological sciences ,0301 basic medicine ,Pleurodeles ,Cell Culture Techniques ,Toxicology ,010603 evolutionary biology ,01 natural sciences ,Plethodon glutinosus ,Tylototriton verrucosus ,Microbiology ,Cell Line ,03 medical and health sciences ,Mice ,Species Specificity ,Ambystoma maculatum ,biology.animal ,Materials Testing ,Human Umbilical Vein Endothelial Cells ,Animals ,Humans ,Skin ,Salamandridae ,biology ,Anatomy ,Fibroblasts ,biology.organism_classification ,030104 developmental biology ,Salamander ,Plethodon shermani ,Salamandra - Abstract
Salamanders have evolved a wide variety of antipredator mechanisms and behavior patterns, including toxins and noxious or adhesive skin secretions. The high bonding strength of the natural bioadhesives makes these substances interesting for biomimetic research and applications in industrial and medical sectors. Secretions of toxic species may help to understand the direct effect of harmful substances on the cellular level. In the present study, the biocompatibility of adhesive secretions from four salamander species (Plethodon shermani, Plethodon glutinosus, Ambystoma maculatum, Ambystoma opacum) were analyzed using the MTT assay in cell culture and evaluated against toxic secretions of Pleurodeles waltl, Triturus carnifex, Pseudotriton ruber, Tylototriton verrucosus, and Salamandra salamandra. Their effect on cells was tested in direct contact (direct culture) or under the influence of the extract (indirect exposure) in accordance with the protocol of the international standard norm ISO 10993-5. Human dermal fibroblasts (NHDF), umbilical vein endothelial cells (HUVEC), and articular chondrocytes (HAC), as well as the cell lines C2C12 and L929 were used in both culture types. While the adhesive secretions from Plethodon shermani are cytocompatible and those of Ambystoma opacum are even advantageous, those of Plethodon glutinosus and Ambystoma maculatum appear to be cytotoxic to NDHF and HUVEC. Toxic secretions from Salamandra salamandra exhibited harmful effects on all cell types. Pseudotriton ruber and Triturus carnifex secretions affected certain cell types marginally; those from Pleurodeles waltl and Tylototriton verrucosus were generally well tolerated. The study shows for the first time the effect of salamander secretions on the viability of different cell types in culture. Two adhesive secretions appeared to be cell compatible and are therefore promising candidates for future investigations in the field of medical bioadhesives. Among the toxic secretions tested, only two of the five had a harmful effect on cells, indicating different cell toxicity mechanisms.
- Published
- 2017
40. Thrombin as important factor for cutaneous wound healing: Comparison of fibrin biomatrices in vitro and in a rat excisional wound healing model
- Author
-
Sabine Pfeifer, Susanne Wolbank, Andreas Goppelt, Joachim Hartinger, Sylvia Nürnberger, Johanna Kober, Rainer Mittermayr, Heinz Redl, Alexandra Meinl, Alfred Gugerell, and Waltraud Pasteiner
- Subjects
medicine.medical_specialty ,biology ,Adhesion (medicine) ,Dermatology ,Pharmacology ,medicine.disease ,Fibrin ,In vitro ,Surgery ,Vascular endothelial growth factor ,chemistry.chemical_compound ,Thrombin ,chemistry ,In vivo ,Hemostasis ,medicine ,biology.protein ,Wound healing ,medicine.drug - Abstract
Fibrin biomatrices have been used for many years for hemostasis and sealing and are a well-established surgical tool. The objective of the present study was to compare two commercially available fibrin biomatrices regarding the effect of their thrombin concentration on keratinocytes and wound healing in vitro and in vivo. Keratinocytes showed significant differences in adhesion, viability, and morphology in the presence of the fibrin matrices in vitro. A high thrombin concentration (800-1,200 IU/mL) caused deteriorated cell compatibility. By using a thrombin inhibitor, those differences could be reversed. In a rat excisional wound healing model, we observed more rapid wound closure and less wound severity in wounds treated with a fibrin matrix containing a lower concentration of thrombin (4 IU/mL). Furthermore, fewer new functional vessels and a lower level of vascular endothelial growth factor were measured in wounds after 7 days treated with the matrix with higher thrombin concentration. These in vivo results may be partially explained by the in vitro biocompatibility data. Additionally, results show that low thrombin biomatrices were degraded faster than the high thrombin material. Hence, we conclude that the composition of fibrin biomatrices influences keratinocytes and therefore has an impact on wound healing.
- Published
- 2014
- Full Text
- View/download PDF
41. Non-invasivein vivotracking of fibrin degradation by fluorescence imaging
- Author
-
Susanne Wolbank, Valentin Pichler, James Ferguson, Andreas Goppelt, Alexandra Meinl, Martijn van Griensven, and Heinz Redl
- Subjects
Fluorescence-lifetime imaging microscopy ,biology ,Chemistry ,medicine.medical_treatment ,Biomedical Engineering ,Medicine (miscellaneous) ,Protein degradation ,Fibrinogen ,Fibrin ,Biomaterials ,Thrombin ,Coagulation ,In vivo ,Fibrinolysis ,medicine ,biology.protein ,Biomedical engineering ,medicine.drug - Abstract
Fibrin-based sealants consist of natural coagulation factors involved in the final phase of blood coagulation, during which fibrinogen is enzymatically converted by thrombin to form a solid-phase fibrin clot. For applications in tissue regeneration, a controlled process of matrix degradation within a certain period of time is essential for optimal wound healing. Hence, it is desirable to follow the kinetics of fibrinolysis at the application site. Non-invasive molecular imaging systems enable real-time tracking of processes in the living animal. In this study, a non-invasive fluorescence based imaging system was applied to follow and quantify site-specific degradation of fibrin sealant. To enable non-invasive tracking of fibrin in vivo, fibrin-matrix was labelled by incorporation of a fluorophore-conjugated fibrinogen component. Protein degradation and release of fluorescence were, in a first step, correlated in vitro. In vivo, fluorophore-labelled fibrin was subcutaneously implanted in mice and followed throughout the experiment using a multispectral imaging system. For the fluorescent fibrin, degradation correlated with the release of fluorescence from the clots in vitro. In vivo it was possible to follow and quantify implanted fibrin clots throughout the experiment, demonstrating degradation kinetics of approximately 16 days in the subcutaneous compartment, which was further confirmed by histological evaluation of the application site.
- Published
- 2014
- Full Text
- View/download PDF
42. Intact human amniotic membrane differentiated towards the chondrogenic lineage
- Author
-
Andrea Lindenmair, Christian Gabriel, Christa Hackl, Heinz Redl, Johann Eibl, Alexandra Meinl, Guido Stadler, Susanne Wolbank, Sylvia Nürnberger, and Simone Hennerbichler
- Subjects
Placenta ,Biomedical Engineering ,Biomaterials ,Tissue engineering ,Pregnancy ,medicine ,Humans ,Cell Lineage ,Amnion ,Cells, Cultured ,Aggrecan ,Cartilage oligomeric matrix protein ,Transplantation ,biology ,Stem Cells ,Cartilage ,Melanoma inhibitory activity ,Cell Differentiation ,Cell Biology ,Chondrogenesis ,Molecular biology ,medicine.anatomical_structure ,Immunology ,biology.protein ,Versican ,Female - Abstract
Human amniotic membrane (hAM) represents a tissue that is well established as biomaterial in the clinics with potential for new applications in regenerative medicine. For tissue engineering (TE) strategies, cells are usually combined with inductive factors and a carrier substrate. We have previously recognized that hAM represents a natural, preformed sheet including highly potent stem cells. In the present approach for cartilage regeneration we have induced chondrogenesis in hAM in vitro. For this, hAM biopsies were cultured for up to 56 days under chondrogenic conditions. The induced hAM was characterized for remaining viability, glycosaminoglycan (GAG) accumulation using histochemical analysis, and a quantitative assay. Collagen I, II and X was immunohistochemically determined and cartilage-specific mRNA expression of (sex determining region Y-) box 9, cartilage oligomeric matrix protein (COMP), aggrecan (AGC1), versican (CSPG2), COL1A1, COL9A2, melanoma inhibitory activity (MIA), and cartilage-linking protein 1 (CRTL1) analyzed by quantitative real-time polymerase chain reaction. Human AM was successfully induced to accumulate GAG, as demonstrated by Alcianblue staining and a significant (p < 0.001) increase of GAG/viability under chondrogenic conditions peaking in a 29.9 ± 0.9-fold induction on day 56. Further, upon chondrogenic induction collagen II positive areas were identified within histological sections and cartilage-specific markers including COMP, AGC1, CSPG2, COL1A1, COL9A2, MIA, and CRTL1 were found upregulated at mRNA level. This is the first study, demonstrating that upon in vitro induction viable human amnion expresses cartilage-specific markers and accumulates GAGs within the biomatrix. This is a promising first step towards a potential use of living hAM for cartilage TE.
- Published
- 2014
- Full Text
- View/download PDF
43. Critical Impact of Human Amniotic Membrane Tension on Mitochondrial Function and Cell Viability In Vitro
- Author
-
Katy Schmidt, Susanne Wolbank, Laura Poženel, Andrea Lindenmair, Asmita Banerjee, Andrey V. Kozlov, Adelheid Weidinger, and Johannes Grillari
- Subjects
Cell Survival ,Placenta ,BCL-2 ,Caspase 3 ,mitochondrial cell death ,Pregnancy ,In vivo ,human amniotic membrane ,Humans ,Amnion ,Viability assay ,bcl-2-Associated X Protein ,Membrane Potential, Mitochondrial ,ATP synthase ,biology ,Chemistry ,Communication ,apoptosis ,General Medicine ,In vitro ,Biomechanical Phenomena ,Mitochondria ,Cell biology ,tensile strength ,Proto-Oncogene Proteins c-bcl-2 ,BAX ,Apoptosis ,Cell culture ,biology.protein ,Female ,Stress, Mechanical ,Stem cell - Abstract
Amniotic cells show exciting stem cell features, which has led to the idea of using living cells of human amniotic membranes (hAMs) in toto for clinical applications. However, under common cell culture conditions, viability of amniotic cells decreases rapidly, whereby reasons for this decrease are unknown so far. Recently, it has been suggested that loss of tissue tension in vivo leads to apoptosis. Therefore, the aim of this study was to investigate the effect of tissue distention on the viability of amniotic cells in vitro. Thereby, particular focus was put on vital mitochondria-linked parameters, such as respiration and ATP synthesis. Biopsies of hAMs were incubated for 7–21 days either non-distended or distended. We observed increased B-cell lymphoma 2-associated X protein (BAX)/B-cell lymphoma (BCL)-2 ratios in non-distended hAMs at day seven, followed by increased caspase 3 expression at day 14, and, consequently, loss of viability at day 21. In contrast, under distention, caspase 3 expression increased only slightly, and mitochondrial function and cellular viability were largely maintained. Our data suggest that a mechano-sensing pathway may control viability of hAM cells by triggering mitochondria-mediated apoptosis upon loss of tension in vitro. Further studies are required to elucidate the underlying molecular mechanisms between tissue distention and viability of hAM cells.
- Published
- 2019
- Full Text
- View/download PDF
44. Long-lasting fibrin matrices ensure stable and functional angiogenesis by highly tunable, sustained delivery of recombinant VEGF164
- Author
-
Anna Hofmann, Elena Groppa, Heinz Redl, Lorentz Kristen Marie, Martin Ehrbar, Jeffrey A. Hubbell, Mikaël M. Martino, Susanne Wolbank, Jeffrey S. Marschall, Joachim Hartinger, Veronica Sacchi, Roberto Gianni-Barrera, Remo A. Largo, Andrea Banfi, Rainer Mittermayr, University of Zurich, and Banfi, Andrea
- Subjects
Vascular Endothelial Growth Factor A ,Materials science ,Angiogenesis ,medicine.medical_treatment ,Neovascularization, Physiologic ,610 Medicine & health ,Mice, Inbred Strains ,Mice, SCID ,Gene delivery ,Pharmacology ,Fibrin ,chemistry.chemical_compound ,Mice ,In vivo ,Ischemia ,medicine ,Human Umbilical Vein Endothelial Cells ,Animals ,Humans ,Therapeutic angiogenesis ,Muscle, Skeletal ,10026 Clinic for Obstetrics ,1000 Multidisciplinary ,Multidisciplinary ,biology ,Growth factor ,Gene Transfer Techniques ,Genetic Therapy ,Biological Sciences ,Recombinant Proteins ,Hindlimb ,Vascular endothelial growth factor ,10062 Urological Clinic ,Vascular endothelial growth factor A ,chemistry ,biology.protein ,Female ,Gels ,Biomedical engineering - Abstract
Clinical trials of therapeutic angiogenesis by vascular endothelial growth factor (VEGF) gene delivery failed to show efficacy. Major challenges include the need to precisely control in vivo distribution of growth factor dose and duration of expression. Recombinant VEGF protein delivery could overcome these issues, but rapid in vivo clearance prevents the stabilization of induced angiogenesis. Here, we developed an optimized fibrin platform for controlled delivery of recombinant VEGF, to robustly induce normal, stable, and functional angiogenesis. Murine VEGF(164) was fused to a sequence derived from alpha 2-plasmin inhibitor (alpha(2)-PI1-8) that is a substrate for the coagulation factor fXIIIa, to allow its covalent cross-linking into fibrin hydrogels and release only by enzymatic cleavage. An alpha(2)-PI1-8-fused variant of the fibrinolysis inhibitor aprotinin was used to control the hydrogel degradation rate, which determines both the duration and effective dose of factor release. An optimized aprotinin-alpha(2)-PI1-8 concentration ensured ideal degradation over 4 wk. Under these conditions, fibrin-alpha(2)-PI1-8-VEGF(164) allowed exquisitely dose-dependent angiogenesis: concentrations >= 25 mu g/mL caused widespread aberrant vascular structures, but a 500-fold concentration range (0.01-5.0 mu g/mL) induced exclusively normal, mature, nonleaky, and perfused capillaries, which were stable after 3 mo. Optimized delivery of fibrin-alpha(2)-PI1-8-VEGF(164) was therapeutically effective both in ischemic hind limb and wound-healing models, significantly improving angiogenesis, tissue perfusion, and healing rate. In conclusion, this optimized platform ensured (i) controlled and highly tunable delivery of VEGF protein in ischemic tissue and (ii) stable and functional angiogenesis without introducing genetic material and with a limited and controllable duration of treatment. These findings suggest a strategy to improve safety and efficacy of therapeutic angiogenesis.
- Published
- 2014
- Full Text
- View/download PDF
45. Human amniotic membrane as newly identified source of amniotic fluid pulmonary surfactant
- Author
-
Florian Prodinger, Jesús Pérez-Gil, Heinz Redl, Susanne Wolbank, Angela Lemke, José Carlos Castillo-Sánchez, Asja Ceranic, and Simone Hennerbichler-Lugscheider
- Subjects
0301 basic medicine ,Bioquímica ,Cell biology ,Amniotic fluid ,1,2-Dipalmitoylphosphatidylcholine ,Molecular biology ,Science ,Alveolar Epithelium ,Amniotic sac ,Lamellar granule ,Biology ,Article ,03 medical and health sciences ,0302 clinical medicine ,Pulmonary surfactant ,Pregnancy ,medicine ,Humans ,Amnion ,Multidisciplinary ,Lung ,Biología molecular ,medicine.diagnostic_test ,Epithelial Cells ,Mesenchymal Stem Cells ,Pulmonary Surfactants ,Amniotic Fluid ,Lipid Metabolism ,Pulmonary Surfactant-Associated Protein D ,Epithelium ,030104 developmental biology ,medicine.anatomical_structure ,Bronchoalveolar lavage ,Immunology ,Medicine ,ATP-Binding Cassette Transporters ,Female ,030217 neurology & neurosurgery - Abstract
Pulmonary surfactant (PS) reduces surface tension at the air-liquid interface in the alveolar epithelium of the lung, which is required for breathing and for the pulmonary maturity of the developing foetus. However, the origin of PS had never been thoroughly investigated, although it was assumed to be secreted from the foetal developing lung. Human amniotic membrane (hAM), particularly its epithelial cell layer, composes the amniotic sac enclosing the amniotic fluid. In this study, we therefore aimed to investigate a potential contribution of the cellular components of the hAM to pulmonary surfactant found in amniotic fluid. We identified that cells within the native membrane contain lamellar bodies and express all four surfactant proteins as well as ABCA3. Lipidomic profiling by nanoESI – MS/MS revealed the presence of the essential lipid species as found in PS. Also, the biophysical activity of conditioned cell culture supernatant obtained from hAM was tested with captive bubble surfactometry. hAM supernatant showed the ability to reduce surface tension, similar to human PS obtained from bronchoalveolar lavage. This means that hAM produces the essential PS-associated components and can therefore contribute as second potential source of PS in amniotic fluid aside from the foetal lung.
- Published
- 2017
46. Secreted microvesicular miR-31 inhibits osteogenic differentiation of mesenchymal stem cells
- Author
-
Klaus Fortschegger, Heinz Redl, Andrea B. Maier, Peter Pietschmann, Paul Messner, Pidder Jansen-Dürr, Heinrich Resch, Susanne Wolbank, Matthias Wieser, Karl H. Schneider, Elisabeth Schraml, Rudi G. J. Westendorp, Sylvia Weilner, Johannes Grillari, Regina Grillari-Voglauer, Lucia Micutkova, Klemens Wassermann, Neuromechanics, and Research Institute MOVE
- Subjects
0301 basic medicine ,Cellular differentiation ,Stem cell theory of aging ,Clinical uses of mesenchymal stem cells ,osteogenic differentiation ,Biology ,03 medical and health sciences ,0302 clinical medicine ,SDG 3 - Good Health and Well-being ,Cell-Derived Microparticles ,Osteogenesis ,Human Umbilical Vein Endothelial Cells ,Humans ,RNA, Messenger ,Cellular Senescence ,Stem cell transplantation for articular cartilage repair ,mesenchymal stem cells ,Tetraspanin 30 ,Mesenchymal stem cell ,aging ,Endothelial Cells ,Cell Differentiation ,MicroRNA ,Original Articles ,Cell Biology ,Frizzled Receptors ,Microvesicles ,Cell biology ,MicroRNAs ,030104 developmental biology ,Adipose Tissue ,Gene Knockdown Techniques ,030220 oncology & carcinogenesis ,Immunology ,senescence-associated secretory phenotype ,Original Article ,senescence‐associated secretory phenotype ,Stem cell ,microvesicles ,Adult stem cell - Abstract
Summary Damage to cells and tissues is one of the driving forces of aging and age‐related diseases. Various repair systems are in place to counteract this functional decline. In particular, the property of adult stem cells to self‐renew and differentiate is essential for tissue homeostasis and regeneration. However, their functionality declines with age (Rando, 2006). One organ that is notably affected by the reduced differentiation capacity of stem cells with age is the skeleton. Here, we found that circulating microvesicles impact on the osteogenic differentiation capacity of mesenchymal stem cells in a donor‐age‐dependent way. While searching for factors mediating the inhibitory effect of elderly derived microvesicles on osteogenesis, we identified miR‐31 as a crucial component. We demonstrated that miR‐31 is present at elevated levels in the plasma of elderly and of osteoporosis patients. As a potential source of its secretion, we identified senescent endothelial cells, which are known to increase during aging in vivo (Erusalimsky, 2009). Endothelial miR‐31 is secreted within senescent cell‐derived microvesicles and taken up by mesenchymal stem cells where it inhibits osteogenic differentiation by knocking down its target Frizzled‐3. Therefore, we suggest that microvesicular miR‐31 in the plasma of elderly might play a role in the pathogenesis of age‐related impaired bone formation and that miR‐31 might be a valuable plasma‐based biomarker for aging and for a systemic environment that does not favor cell‐based therapies whenever osteogenesis is a limiting factor.
- Published
- 2016
- Full Text
- View/download PDF
47. Meeting report of the first conference of the International Placenta Stem Cell Society (IPLASS)
- Author
-
Cv V. Borlongan, Sa A. Scherjon, Francesco Alviano, C. Götherström, Susanne Wolbank, Ag G. Betz, Al L. Mellor, Ml L. Weiss, Ornella Parolini, Dw W. Bianchi, Ursula Manuelpillai, Kj J. Wood, Peter Ponsaerts, Racheli Ofir, Parolini O., Alviano F., Betz A.G., Bianchi D.W., Gotherstrom C., Manuelpillai U., Mellor A.L., Ofir R., Ponsaerts P., Scherjon S.A., Weiss M.L., Wolbank S., Wood K.J., and Borlongan C.V.
- Subjects
Placenta ,Library science ,Fetomaternal tolerance ,Article ,Term placenta ,Immunomodulation ,Female ,Fetal Stem Cells ,Fetus ,Humans ,Pregnancy ,Cell transplantation ,Adult stem cell ,Settore BIO/13 - BIOLOGIA APPLICATA ,Medicine ,Scientific activity ,Inflammation ,business.industry ,Lung fibrosis ,Champion ,Obstetrics and Gynecology ,Placental cell ,Transplantation ,Reproductive Medicine ,embryonic structures ,Immunology ,Human medicine ,Stem cell ,business ,Developmental Biology - Abstract
The International Placenta Stem Cell Society (IPLASS) was founded in June 2010. Its goal is to serve as a network for advancing research and clinical applications of stem/progenitor cells isolated from human term placental tissues, including the amnio-chorionic fetal membranes and Wharton’s jelly. The commitment of the Society to champion placenta as a stem cell source was realized with the inaugural meeting of IPLASS held in Brescia, Italy, in October 2010. Officially designated as an EMBO-endorsed scientific activity, international experts in the field gathered for a 3-day meeting, which commenced with “Meet with the experts” sessions, IPLASS member and board meetings, and welcome remarks by Dr. Ornella Parolini, President of IPLASS. The evening’s highlight was a keynote plenary lecture by Dr. Diana Bianchi. The subsequent scientific program consisted of morning and afternoon oral and poster presentations, followed by social events. Both provided many opportunities for intellectual exchange among the 120 multi-national participants. This allowed a methodical and deliberate evaluation of the status of placental cells in research in regenerative and reparative medicine. The meeting concluded with Dr. Parolini summarizing the meeting’s highlights. This further prepared the fertile ground on which to build the promising potential of placental cell research. The second IPLASS meeting will take place in September 2012 in Vienna, Austria. This meeting report summarizes the thought-provoking lectures delivered at the first meeting of IPLASS.
- Published
- 2016
48. Improvement of adipose tissue-derived cells by low-energy extracorporeal shock wave therapy
- Author
-
J. Maier, Christoph Wurzer, Carolin Steffenhagen, Sylvia Nuernberger, Susanne Wolbank, Wolfgang Holnthoner, Christina M.A.P. Schuh, Heinz Redl, Susanne Suessner, Dominik Rünzler, Christiane Fuchs, and Eleni Priglinger
- Subjects
0301 basic medicine ,Extracorporeal Shockwave Therapy ,Cancer Research ,Pathology ,medicine.medical_specialty ,Stromal cell ,Adipose tissue macrophages ,medicine.medical_treatment ,Immunology ,Adipose tissue ,Immunophenotyping ,03 medical and health sciences ,Adenosine Triphosphate ,Immunology and Allergy ,Medicine ,Humans ,Progenitor cell ,Insulin-Like Growth Factor I ,Genetics (clinical) ,Cells, Cultured ,Placenta Growth Factor ,Transplantation ,business.industry ,Growth factor ,Stem Cells ,Mesenchymal stem cell ,Cell Differentiation ,Cell Biology ,Stromal vascular fraction ,030104 developmental biology ,Oncology ,Adipose Tissue ,Stem cell ,Stromal Cells ,business ,Biomarkers - Abstract
Background Cell-based therapies with autologous adipose tissue–derived cells have shown great potential in several clinical studies in the last decades. The majority of these studies have been using the stromal vascular fraction (SVF), a heterogeneous mixture of fibroblasts, lymphocytes, monocytes/macrophages, endothelial cells, endothelial progenitor cells, pericytes and adipose-derived stromal/stem cells (ASC) among others. Although possible clinical applications of autologous adipose tissue–derived cells are manifold, they are limited by insufficient uniformity in cell identity and regenerative potency. Methods In our experimental set-up, low-energy extracorporeal shock wave therapy (ESWT) was performed on freshly obtained human adipose tissue and isolated adipose tissue SVF cells aiming to equalize and enhance stem cell properties and functionality. Results After ESWT on adipose tissue we could achieve higher cellular adenosine triphosphate (ATP) levels compared with ESWT on the isolated SVF as well as the control. ESWT on adipose tissue resulted in a significantly higher expression of single mesenchymal and vascular marker compared with untreated control. Analysis of SVF protein secretome revealed a significant enhancement in insulin-like growth factor (IGF)-1 and placental growth factor (PLGF) after ESWT on adipose tissue. Discussion Summarizing we could show that ESWT on adipose tissue enhanced the cellular ATP content and modified the expression of single mesenchymal and vascular marker, and thus potentially provides a more regenerative cell population. Because the effectiveness of autologous cell therapy is dependent on the therapeutic potency of the patient's cells, this technology might raise the number of patients eligible for autologous cell transplantation.
- Published
- 2016
49. A luciferase-based quick potency assay to predict chondrogenic differentiation
- Author
-
Carolin Steffenhagen, Eleni Oberbauer, Christian Gabriel, Georg A. Feichtinger, Martin Danzer, Ara Hacobian, Florian Hildner, Heinz Redl, and Susanne Wolbank
- Subjects
0301 basic medicine ,Cellular differentiation ,Biomedical Engineering ,Medicine (miscellaneous) ,Bioengineering ,Biology ,Polymerase Chain Reaction ,03 medical and health sciences ,Chondrocytes ,0302 clinical medicine ,Humans ,Bioassay ,Potency ,Luciferase ,Luciferases ,Collagen Type II ,Cells, Cultured ,Reporter gene ,Tissue Engineering ,Cell Differentiation ,SOX9 Transcription Factor ,Chondrogenesis ,Molecular biology ,Cell biology ,Transplantation ,030104 developmental biology ,Adipose Tissue ,030220 oncology & carcinogenesis ,Biological Assay ,Stem cell - Abstract
Chondrogenic differentiation of adipose-derived stem cells (ASC) is challenging but highly promising for cartilage repair. Large donor variability of chondrogenic differentiation potential raises the risk for transplantation of cells with reduced efficacy and a low chondrogenic potential. Therefore, quick potency assays are required to control the potency of the isolated cells before cell transplantation. Current in vitro methods to analyze the differentiation capacity are time-consuming, and thus, a novel enhancer and tissue-specific promoter combination was used for the detection of chondrogenic differentiation of ASC in a novel quick potency bioassay. Human primary ASC were cotransfected with the Metridia luciferase-based collagen type II reporter gene pCMVE_ACDCII-MetLuc together with a Renilla control plasmid and analyzed for their chondrogenic potential. On day 3 after chondrogenic induction, the luciferase activity was induced in all tested donors under three-dimensional culture conditions and, in a second approach, also under two-dimensional (2D) culture conditions. With our newly developed quick potency bioassay, we can determine chondrogenic potential already after 3 days of chondrogenic induction and under 2D culture conditions. This will enhance the efficiency of testing cell functionality, which should allow in the future to predict the suitability of cells derived from individual patients for cell therapies in a very short time and at low costs.
- Published
- 2016
50. Mesenchymal Stem or Stromal Cells from Amnion and Umbilical Cord Tissue and Their Potential for Clinical Applications
- Author
-
Heinz Redl, Andrea Lindenmair, Susanne Wolbank, Gregor Kollwig, Tim Hatlapatka, Simone Hennerbichler, Christian Gabriel, and Cornelia Kasper
- Subjects
Pathology ,medicine.medical_specialty ,Stromal cell ,Amnion ,business.industry ,Mesenchymal stem cell ,Clinical uses of mesenchymal stem cells ,General Medicine ,Review ,clinical applications ,Umbilical cord ,Cell biology ,MSC ,medicine.anatomical_structure ,Tissue engineering ,lcsh:Biology (General) ,mesenchymal stromal cell ,umbilical cord ,Medicine ,Bone marrow ,business ,lcsh:QH301-705.5 ,mesenchymal stem cell ,Stem cell transplantation for articular cartilage repair ,amnion - Abstract
Mesenchymal stem or stromal cells (MSC) have proven to offer great promise for cell-based therapies and tissue engineering applications, as these cells are capable of extensive self-renewal and display a multilineage differentiation potential. Furthermore, MSC were shown to exhibit immunomodulatory properties and display supportive functions through parakrine effects. Besides bone marrow (BM), still today the most common source of MSC, these cells were found to be present in a variety of postnatal and extraembryonic tissues and organs as well as in a large variety of fetal tissues. Over the last decade, the human umbilical cord and human amnion have been found to be a rich and valuable source of MSC that is bio-equivalent to BM-MSC. Since these tissues are discarded after birth, the cells are easily accessible without ethical concerns.
- Published
- 2012
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.