1
|
Abbasi-Malati Z, Khanicheragh P, Narmi MT, Mardi N, Khosrowshahi ND, Hiradfar A, Rezabakhsh A, Sadeghsoltani F, Rashidi S, Chegeni SA, Roozbahani G, Rahbarghazi R. Tumoroids, a valid preclinical screening platform for monitoring cancer angiogenesis. Stem Cell Res Ther 2024; 15:267. [PMID: 39183337 PMCID: PMC11346257 DOI: 10.1186/s13287-024-03880-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 08/06/2024] [Indexed: 08/27/2024] Open
Abstract
In recent years, biologists and clinicians have witnessed prominent advances in in vitro 3D culture techniques related to biomimetic human/animal tissue analogs. Numerous data have confirmed that unicellular and multicellular (tumoroids) tumor spheroids with dense native cells in certain matrices are sensitive and valid analytical tools for drug screening, cancer cell dynamic growth, behavior, etc. in laboratory settings. Angiogenesis/vascularization is a very critical biological phenomenon to support oxygen and nutrients to tumor cells within the deep layer of solid masses. It has been shown that endothelial cell (EC)-incorporated or -free spheroid/tumoroid systems provide a relatively reliable biological platform for monitoring the formation of nascent blood vessels in micron/micrometer scales. Besides, the paracrine angiogenic activity of cells within the spheroid/tumoroid systems can be monitored after being treated with different therapeutic approaches. Here, we aimed to collect recent advances and findings related to the monitoring of cancer angiogenesis using unicellular and multicellular tumor spheroids. Vascularized spheroids/tumoroids can help us in the elucidation of mechanisms related to cancer formation, development, and metastasis by monitoring the main influencing factors.
Collapse
Affiliation(s)
- Zahra Abbasi-Malati
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parisa Khanicheragh
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Narges Mardi
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nafiseh Didar Khosrowshahi
- Stem Cell and Tissue Engineering Research Laboratory, Sahand University of Technology, Tabriz, 51335-1996, Iran
| | - Amirataollah Hiradfar
- Pediatric Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aysa Rezabakhsh
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Somayyeh Rashidi
- Department of Medical Biotechnology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | | | - Golbarg Roozbahani
- Department of Plant, Cell and Molecular Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
2
|
Vahldieck C, Löning S, Hamacher C, Fels B, Rudzewski B, Nickel L, Weil J, Nording H, Baron L, Kleingarn M, Karsten CM, Kusche-Vihrog K. Dysregulated complement activation during acute myocardial infarction leads to endothelial glycocalyx degradation and endothelial dysfunction via the C5a:C5a-Receptor1 axis. Front Immunol 2024; 15:1426526. [PMID: 39055717 PMCID: PMC11269135 DOI: 10.3389/fimmu.2024.1426526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
Introduction Complement-mediated damage to the myocardium during acute myocardial infarction (AMI), particularly the late components of the terminal pathway (C5-convertase and C5b-9), have previously been characterized. Unfortunately, only few studies have reported a direct association between dysregulated complement activation and endothelial function. Hence, little attention has been paid to the role of the anaphylatoxin C5a. The endothelial glycocalyx (eGC) together with the cellular actin cortex provide a vasoprotective barrier against chronic vascular inflammation. Changes in their nanomechanical properties (stiffness and height) are recognized as hallmarks of endothelial dysfunction as they correlate with the bioavailability of vasoactive substances, such as nitric oxide (NO). Here, we determined how the C5a:C5aR1 axis affects the eGC and endothelial function in AMI. Methods Samples of fifty-five patients with ST-elevation myocardial infarction (STEMI) vs. healthy controls were analyzed in this study. eGC components and C5a levels were determined via ELISA; NO levels were quantified chemiluminescence-based. Endothelial cells were stimulated with C5a or patient sera (with/without C5a-receptor1 antagonist "PMX53") and the nanomechanical properties of eGC quantified using the atomic force microscopy (AFM)-based nanoindentation technique. To measure actin cytoskeletal tension regulator activation (RhoA and Rac1) G-LISA assays were applied. Vascular inflammation was examined by quantifying monocyte-endothelium interaction via AFM-based single-cell-force spectroscopy. Results Serum concentrations of eGC components and C5a were significantly increased during STEMI. Serum and solely C5a stimulation decreased eGC height and stiffness, indicating shedding of the eGC. C5a enhanced RhoA activation, resulting in increased cortical stiffness with subsequent reduction in NO concentrations. Monocyte adhesion to the endothelium was enhanced after both C5a and stimulation with STEMI serum. eGC degradation- and RhoA-induced cortical stiffening with subsequent endothelial dysfunction were attenuated after administering PMX53. Conclusion This study demonstrates that dysregulated C5a activation during AMI results in eGC damage with subsequent endothelial dysfunction and reduced NO bioavailability, indicating progressively developing vascular inflammation. This could be prevented by antagonizing C5aR1, highlighting the role of the C5a:C5a-Receptor1 axis in vascular inflammation development and endothelial dysfunction in AMI, offering new therapeutic approaches for future investigations.
Collapse
Affiliation(s)
- Carl Vahldieck
- Department of Anesthesiology and Intensive Care Medicine, University Medical Centre Schleswig-Holstein Campus Luebeck, Luebeck, Germany
- Institute of Physiology, University of Luebeck, Luebeck, Germany
- DZHK (German Research Centre for Cardiovascular Research), Partner Site Hamburg/Luebeck/Kiel, Luebeck, Germany
| | - Samuel Löning
- Institute of Physiology, University of Luebeck, Luebeck, Germany
| | | | - Benedikt Fels
- Institute of Physiology, University of Luebeck, Luebeck, Germany
- DZHK (German Research Centre for Cardiovascular Research), Partner Site Hamburg/Luebeck/Kiel, Luebeck, Germany
| | | | - Laura Nickel
- Medizinische Klinik II, Sana Kliniken Luebeck, Luebeck, Germany
| | - Joachim Weil
- Medizinische Klinik II, Sana Kliniken Luebeck, Luebeck, Germany
| | - Henry Nording
- DZHK (German Research Centre for Cardiovascular Research), Partner Site Hamburg/Luebeck/Kiel, Luebeck, Germany
- Cardioimmunology Group, Medical Clinic II, University Heart Center Luebeck, Luebeck, Germany
| | - Lasse Baron
- Cardioimmunology Group, Medical Clinic II, University Heart Center Luebeck, Luebeck, Germany
| | - Marie Kleingarn
- Institute for Systemic Inflammation Research (ISEF), University of Luebeck, Luebeck, Germany
| | | | - Kristina Kusche-Vihrog
- Institute of Physiology, University of Luebeck, Luebeck, Germany
- DZHK (German Research Centre for Cardiovascular Research), Partner Site Hamburg/Luebeck/Kiel, Luebeck, Germany
| |
Collapse
|
3
|
Margolis EA, Choi LS, Friend NE, Putnam AJ. Engineering primitive multiscale chimeric vasculature by combining human microvessels with explanted murine vessels. Sci Rep 2024; 14:4036. [PMID: 38369633 PMCID: PMC10874928 DOI: 10.1038/s41598-024-54880-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 02/17/2024] [Indexed: 02/20/2024] Open
Abstract
Strategies to separately manufacture arterial-scale tissue engineered vascular grafts and microvascular networks have been well-established, but efforts to bridge these two length scales to create hierarchical vasculature capable of supporting parenchymal cell functions or restoring perfusion to ischemic tissues have been limited. This work aimed to create multiscale vascular constructs by assessing the capability of macroscopic vessels isolated from mice to form functional connections to engineered capillary networks ex vivo. Vessels of venous and arterial origins from both thoracic and femoral locations were isolated from mice, and then evaluated for their abilities to sprout endothelial cells (EC) capable of inosculating with surrounding human cell-derived microvasculature within bulk fibrin hydrogels. Comparing aortae, vena cavae, and femoral vessel bundles, we identified the thoracic aorta as the rodent macrovessel that yielded the greatest degree of sprouting and interconnection to surrounding capillaries. The presence of cells undergoing vascular morphogenesis in the surrounding hydrogel attenuated EC sprouting from the macrovessel compared to sprouting into acellular hydrogels, but ultimately sprouted mouse EC interacted with human cell-derived capillary networks in the bulk, yielding chimeric vessels. We then integrated micromolded mesovessels into the constructs to engineer a primitive 3-scale vascular hierarchy comprising capillaries, mesovessels, and macrovessels. Overall, this study yielded a primitive hierarchical vasculature suitable as proof-of-concept for regenerative medicine applications and as an experimental model to better understand the spontaneous formation of host-graft vessel anastomoses.
Collapse
Affiliation(s)
- Emily A Margolis
- Department of Biomedical Engineering, University of Michigan, 2204 Lurie Biomedical Eng. Bldg., 1101 Beal Ave., Ann Arbor, MI, 48109, USA
| | - Lucia S Choi
- Department of Biomedical Engineering, University of Michigan, 2204 Lurie Biomedical Eng. Bldg., 1101 Beal Ave., Ann Arbor, MI, 48109, USA
| | - Nicole E Friend
- Department of Biomedical Engineering, University of Michigan, 2204 Lurie Biomedical Eng. Bldg., 1101 Beal Ave., Ann Arbor, MI, 48109, USA
| | - Andrew J Putnam
- Department of Biomedical Engineering, University of Michigan, 2204 Lurie Biomedical Eng. Bldg., 1101 Beal Ave., Ann Arbor, MI, 48109, USA.
| |
Collapse
|
4
|
Iqbal F, Johnston A, Wyse B, Rabani R, Mander P, Hoseini B, Wu J, Li RK, Gauthier-Fisher A, Szaraz P, Librach C. Combination human umbilical cord perivascular and endothelial colony forming cell therapy for ischemic cardiac injury. NPJ Regen Med 2023; 8:45. [PMID: 37626067 PMCID: PMC10457300 DOI: 10.1038/s41536-023-00321-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
Cell-based therapeutics are promising interventions to repair ischemic cardiac tissue. However, no single cell type has yet been found to be both specialized and versatile enough to heal the heart. The synergistic effects of two regenerative cell types including endothelial colony forming cells (ECFC) and first-trimester human umbilical cord perivascular cells (FTM HUCPVC) with endothelial cell and pericyte properties respectively, on angiogenic and regenerative properties were tested in a rat model of myocardial infarction (MI), in vitro tube formation and Matrigel plug assay. The combination of FTM HUCPVCs and ECFCs synergistically reduced fibrosis and cardiomyocyte apoptosis, while promoting favorable cardiac remodeling and contractility. These effects were in part mediated by ANGPT2, PDGF-β, and VEGF-C. PDGF-β signaling-dependent synergistic effects on angiogenesis were also observed in vitro and in vivo. FTM HUCPVCs and ECFCs represent a cell combination therapy for promoting and sustaining vascularization following ischemic cardiac injury.
Collapse
Affiliation(s)
- Farwah Iqbal
- Create Fertility Centre, Toronto, ON, Canada
- Virginia Tech Carillion School of Medicine, Roanoke, VA, USA
| | | | | | | | | | | | - Jun Wu
- Toronto General Research Institute (TGRI), University Health Network (UHN), Toronto, ON, Canada
| | - Ren-Ke Li
- Toronto General Research Institute (TGRI), University Health Network (UHN), Toronto, ON, Canada
| | | | | | - Clifford Librach
- Create Fertility Centre, Toronto, ON, Canada.
- Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON, Canada.
- Institute of Medical Sciences, Department of Physiology, University of Toronto, Toronto, ON, Canada.
- Department of Obstetrics and Gynecology, Women's College Hospital, Toronto, ON, Canada.
| |
Collapse
|
5
|
Shuster-Hyman H, Siddiqui F, Gallagher D, Gauthier-Fisher A, Librach CL. Time course and mechanistic analysis of human umbilical cord perivascular cell mitigation of lipopolysaccharide-induced systemic and neurological inflammation. Cytotherapy 2023; 25:125-137. [PMID: 36473795 DOI: 10.1016/j.jcyt.2022.10.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 10/11/2022] [Accepted: 10/20/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND AIMS Because of their potent immunomodulatory and anti-inflammatory properties, mesenchymal stromal cells are a major focus in the field of stem cell therapy. However, the precise mechanisms underlying this are not entirely understood. Human umbilical cord perivascular cells (HUCPVCs) are a promising cell therapy candidate. This study was designed to evaluate the time course and mechanisms by which HUCPVCs mitigate lipopolysaccharide (LPS)-induced systemic and neurological inflammation in immunocompetent mice. To explore the underlying mechanisms, the authors investigated the biodistribution and fate of HUCPVCs. METHODS Male C57BL/6 mice were randomly allocated to four groups: control, LPS, HUCPVCs or LPS + HUCPVCs. Quantitative polymerase chain reaction, enzyme-linked immunosorbent assay and cytokine arrays were used to assess changes in pro-inflammatory mediators systemically and in the brain. Depressive-like behavioral changes were evaluated via a forced swim test. Quantum dot (qDot) labeling and immunohistochemistry were used to assess the biodistribution and fate of HUCPVCs and interactions with recipient innate immune cells. RESULTS A single intravenously delivered dose of HUCPVCs significantly reduced the systemic inflammation induced by LPS within the first 24 h after administration. HUCPVC treatment abrogated the upregulated expression of pro-inflammatory genes in the hippocampus and cortex and attenuated depressive-like behavior induced by LPS treatment. Biodistribution analysis revealed that HUCPVC-derived qDots rapidly accumulated in the lungs and demonstrated limited in vivo persistence. Furthermore, qDot signals were associated with major recipient innate immune cells and promoted a shift in macrophages toward a regulatory phenotype in the lungs. CONCLUSIONS Overall, this study demonstrates that HUCPVCs can successfully reduce systemic and neurological inflammation induced by LPS within the first 24 h after administration. Biodistribution and fate analyses suggest a critical role for the innate immune system in the HUCPVC-based immunomodulation mechanism.
Collapse
Affiliation(s)
- Hannah Shuster-Hyman
- CReATe Fertility Center, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | - Clifford L Librach
- CReATe Fertility Center, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Department of Obstetrics and Gynecology, University of Toronto, Toronto, Ontario, Canada; Department of Physiology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
6
|
Chinnadurai R. Advanced Technologies for Potency Assay Measurement. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1420:81-95. [PMID: 37258785 DOI: 10.1007/978-3-031-30040-0_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Crucial for their application, cell products need to be well-characterized in the cell manufacturing facilities and conform to regulatory approval criteria before infusion into the patients. Mesenchymal Stromal Cells (MSCs) are the leading cell therapy candidate in clinical trials worldwide. Early phase clinical trials have demonstrated that MSCs display an excellent safety profile and are well tolerated. However, MSCs have also exhibited contradictory efficacy in later-phase clinical trials with reasons for this discrepancy including poorly understood mechanism of MSC therapeutic action. With likelihood that a number of attributes are involved in MSC derived clinical benefit, an assay that measures a single quality of may not adequately reflect potency, thus a combination of bioassays and analytical methods, collectively called "assay matrix" are favoured for defining the potency of MSC more adequately. This chapter highlights advanced technologies and targets that can achieve quantitative measurement for a range of MSC attributes, including immunological, genomic, secretome, phosphorylation, morphological, biomaterial, angiogenic and metabolic assays.
Collapse
Affiliation(s)
- Raghavan Chinnadurai
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA, USA.
| |
Collapse
|
7
|
Torggler R, Margreiter E, Marksteiner R, Thurner M. Potency Assay Development: A Keystone for Clinical Use. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1420:13-28. [PMID: 37258781 DOI: 10.1007/978-3-031-30040-0_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Potency can be described as the quantitative measure of biological activity, that is, the ability of an Advanced Therapy Medicinal Product (ATMP) to elicit the intended effect necessary for clinical efficacy. Potency testing is part of the quality control strategy necessary for batch release and is required for market approval application of an ATMP. Thus, it is crucial to develop a reliable and accurate potency assay. As a prerequisite for potency assay development, it is essential to define the mode of action of the product and thereby also the relevant biological activity that should be measured. The establishment of a potency assay should be initiated already during early product development followed by its progressive implementation into an ATMP's manufacturing, quality control and release process. Potency testing is indispensable for clinical use with a wide range of applications. A potency assay is a valuable tool to determine the product's stability, detect the impact of changes in the manufacturing process on the product, demonstrate quality and manufacturing consistency from batch to batch, estimate clinical efficacy and define the effective dose. This chapter describes the requirements and challenges to be considered for potency assay development and the importance of a well-established potency assay for clinical use.
Collapse
Affiliation(s)
| | | | | | - Marco Thurner
- Innovacell AG, Innsbruck, Austria
- Finnegan, Henderson, Farabow, Garrett & Dunner LLP, Munich, Germany
| |
Collapse
|
8
|
Herre C, Nshdejan A, Klopfleisch R, Corte GM, Bahramsoltani M. Expression of vimentin, TPI and MAT2A in human dermal microvascular endothelial cells during angiogenesis in vitro. PLoS One 2022; 17:e0266774. [PMID: 35482724 PMCID: PMC9049311 DOI: 10.1371/journal.pone.0266774] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 03/27/2022] [Indexed: 11/19/2022] Open
Abstract
Introduction
In vitro assays of angiogenesis face immense problems considering their reproducibility based on the inhomogeneous characters of endothelial cells (ECs). It is necessary to detect influencing factors, which affect the angiogenic potency of ECs.
Objective
This study aimed to analyse expression profiles of vimentin (VIM), triosephosphate isomerase (TPI) and adenosylmethionine synthetase isoform type–2 (MAT2A) during the whole angiogenic cascade in vitro. Furthermore, the impact of knocking down vimentin (VIM) on angiogenesis in vitro was evaluated, while monitoring TPI and MAT2A expression.
Methods
A long–term cultivation and angiogenic stimulation of human dermal microvascular ECs was performed. Cells were characterized via VEGFR–1 and VEGFR–2 expression and a shRNA–mediated knockdown of VIM was performed. The process of angiogenesis in vitro was quantified via morphological staging and mRNA–and protein–levels of all proteins were analysed.
Results
While native cells ran through the angiogenic cascade chronologically, knockdown cells only entered beginning stages of angiogenesis and died eventually. Cell cultures showing a higher VEGFR–1 expression survived exclusively and displayed an upregulation of MAT2A and TPI expression. Native cells highly expressed VIM in early stages, MAT2A mainly in the beginning and TPI during the course of angiogenesis in vitro.
Conclusion
VIM knockdown led to a deceleration of angiogenesis in vitro and knockdown cells displayed expressional changes in TPI and MAT2A. Cell populations with a higher number of stalk cells emerged as being more stable against manipulations and native expression profiles provided an indication of VIM and MAT2A being relevant predominantly in beginning stages and TPI during the whole angiogenic cascade in vitro.
Collapse
Affiliation(s)
- Christina Herre
- Department of Veterinary Medicine, Institute of Veterinary Anatomy, Freie Universität Berlin, Berlin, Germany
- * E-mail:
| | - Arpenik Nshdejan
- Department of Veterinary Medicine, Institute of Veterinary Anatomy, Freie Universität Berlin, Berlin, Germany
| | - Robert Klopfleisch
- Department of Veterinary Medicine, Institute of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Giuliano Mario Corte
- Department of Veterinary Medicine, Institute of Veterinary Anatomy, Freie Universität Berlin, Berlin, Germany
| | - Mahtab Bahramsoltani
- Department of Veterinary Medicine, Institute of Veterinary Anatomy, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
9
|
Abstract
The ex vivo aortic ring assay is one of the most widely used protocols to study sprouting angiogenesis. It is a highly adaptable method that can be utilized to investigate the effects of different growth factors, small-molecule drugs, and genetic modifications on vascular sprouting in a physiologically relevant setting. In this chapter we describe a simple and optimized protocol for investigating vascular sprouting in the mouse aortic ring model. The protocol describes the harvesting and embedding of the aortic rings in a collagen matrix, treatment of the rings with agents of interest, and the visualization and quantification of the vascular sprouts.
Collapse
Affiliation(s)
- Vedanta Mehta
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Wellcome Centre for Human Genetics, Oxford, UK
| | - Marwa Mahmoud
- Faculty of Health, Education, Medicine and Social Care , Anglia Ruskin University, Chelmsford, UK.
| |
Collapse
|
10
|
Szklanny AA, Machour M, Redenski I, Chochola V, Goldfracht I, Kaplan B, Epshtein M, Simaan Yameen H, Merdler U, Feinberg A, Seliktar D, Korin N, Jaroš J, Levenberg S. 3D Bioprinting of Engineered Tissue Flaps with Hierarchical Vessel Networks (VesselNet) for Direct Host-To-Implant Perfusion. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2102661. [PMID: 34510579 PMCID: PMC11468543 DOI: 10.1002/adma.202102661] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/28/2021] [Indexed: 05/09/2023]
Abstract
Engineering hierarchical vasculatures is critical for creating implantable functional thick tissues. Current approaches focus on fabricating mesoscale vessels for implantation or hierarchical microvascular in vitro models, but a combined approach is yet to be achieved to create engineered tissue flaps. Here, millimetric vessel-like scaffolds and 3D bioprinted vascularized tissues interconnect, creating fully engineered hierarchical vascular constructs for implantation. Endothelial and support cells spontaneously form microvascular networks in bioprinted tissues using a human collagen bioink. Sacrificial molds are used to create polymeric vessel-like scaffolds and endothelial cells seeded in their lumen form native-like endothelia. Assembling endothelialized scaffolds within vascularizing hydrogels incites the bioprinted vasculature and endothelium to cooperatively create vessels, enabling tissue perfusion through the scaffold lumen. Using a cuffing microsurgery approach, the engineered tissue is directly anastomosed with a rat femoral artery, promoting a rich host vasculature within the implanted tissue. After two weeks in vivo, contrast microcomputer tomography imaging and lectin perfusion of explanted engineered tissues verify the host ingrowth vasculature's functionality. Furthermore, the hierarchical vessel network (VesselNet) supports in vitro functionality of cardiomyocytes. Finally, the proposed approach is expanded to mimic complex structures with native-like millimetric vessels. This work presents a novel strategy aiming to create fully-engineered patient-specific thick tissue flaps.
Collapse
Affiliation(s)
- Ariel A. Szklanny
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifa32000Israel
| | - Majd Machour
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifa32000Israel
| | - Idan Redenski
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifa32000Israel
| | - Václav Chochola
- Department of Histology and EmbryologyFaculty of MedicineMasaryk UniversityBrno625 00Czech Republic
| | - Idit Goldfracht
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifa32000Israel
| | - Ben Kaplan
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifa32000Israel
| | - Mark Epshtein
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifa32000Israel
| | - Haneen Simaan Yameen
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifa32000Israel
| | - Uri Merdler
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifa32000Israel
| | - Adam Feinberg
- Department of Biomedical EngineeringCarnegie Mellon UniversityPittsburghPA15213USA
| | - Dror Seliktar
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifa32000Israel
| | - Netanel Korin
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifa32000Israel
| | - Josef Jaroš
- Cell and Tissue RegenerationInternational Clinical Research CenterSt. Anne's University Hospital BrnoBrno65691Czech Republic
| | - Shulamit Levenberg
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifa32000Israel
| |
Collapse
|
11
|
Mashiach J, Zohni K, Lopez L, Filice M, Garcia M, Wyse B, Glass K, Dviri M, Baram S, Gauthier-Fisher A, Librach CL. Human umbilical cord perivascular cells prevent chemotherapeutic drug-induced male infertility in a mouse model. F&S SCIENCE 2021; 2:24-32. [PMID: 35559762 DOI: 10.1016/j.xfss.2020.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 11/13/2020] [Accepted: 12/03/2020] [Indexed: 06/15/2023]
Abstract
OBJECTIVE To study whether intratesticular (IT) administration of 2 sources of human umbilical cord perivascular cells (HUCPVC), rich and potent sources of mesenchymal stromal cells (MSC), before chemotherapy can prevent infertility in a mouse model. DESIGN Two control groups of CD1 male mice without busulfan (BUS) administration (untreated and IT media injection groups) were included. Experimental groups included IT administration of media, first trimester (FTM) HUCPVCs or term HUCPVCs (n = 5 each) injected 3 days before BUS treatment (20 mg/kg). All groups were included in a mating time course study over 6 months. SETTING Preclinical study in a fertility center research laboratory. PATIENTS Not applicable. INTERVENTION IT delivery of FTM or term HUCPVC before BUS treatment. MAIN OUTCOME MEASURES Pregnancies, litter sizes, and gross morphology of offspring were monitored. Caudal epididymal sperm concentration, motility, and progressive motility were assessed by computer-assisted sperm analysis. Spermatogenesis was also assessed histologically in testicular tissue sections. RESULTS FTM and term HUCPVC displayed an MSC-associated immunophenotype and expressed transcripts encoding paracrine factors known to regulate the testicular cell niche. IT administration of FTM and term HUCPVC before chemotherapy promoted the recovery of spermatogenesis and fertility compared with BUS-treated animals that received a media injection. Although the total number of pups sired over 6 months by males treated with FTM or term HUCPVC was reduced compared with untreated or media-injected controls, litter size and sperm parameters in fertile animals did not differ between control and cell-treated groups. CONCLUSION HUCPVC represent a promising source of MSC-based therapy to prevent gonadotoxic chemotherapeutic drug-induced infertility.
Collapse
Affiliation(s)
| | - Khaled Zohni
- CReATe Fertility Centre, Toronto, Canada; Department of Obstetrics and Gynecology, University of Toronto, Toronto, Canada; Department of Obstetrics and Gynecology, University of Manitoba, Winnipeg, Manitoba, Canada; Heartland Fertility and Gynecology clinic, Winnipeg, Manitoba, Canada
| | | | | | | | | | - Karen Glass
- CReATe Fertility Centre, Toronto, Canada; Department of Obstetrics and Gynecology, University of Toronto, Toronto, Canada
| | - Michal Dviri
- CReATe Fertility Centre, Toronto, Canada; Department of Obstetrics and Gynecology, University of Toronto, Toronto, Canada
| | - Shira Baram
- CReATe Fertility Centre, Toronto, Canada; Department of Obstetrics and Gynecology, University of Toronto, Toronto, Canada; Emek Medical Center, Afula, Israel
| | | | - Clifford L Librach
- CReATe Fertility Centre, Toronto, Canada; Department of Obstetrics and Gynecology, University of Toronto, Toronto, Canada; Department of Physiology University of Toronto, Toronto, Canada; Institute of Medical Sciences, University of Toronto, Toronto, Canada; Department of Gynecology, Women's College Hospital, Toronto, Canada
| |
Collapse
|
12
|
Kapoor A, Chen CG, Iozzo RV. Endorepellin evokes an angiostatic stress signaling cascade in endothelial cells. J Biol Chem 2020; 295:6344-6356. [PMID: 32205445 PMCID: PMC7212646 DOI: 10.1074/jbc.ra120.012525] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 03/17/2020] [Indexed: 12/25/2022] Open
Abstract
Endorepellin, the C-terminal fragment of the heparan sulfate proteoglycan perlecan, influences various signaling pathways in endothelial cells by binding to VEGFR2. In this study, we discovered that soluble endorepellin activates the canonical stress signaling pathway consisting of PERK, eIF2α, ATF4, and GADD45α. Specifically, endorepellin evoked transient activation of VEGFR2, which, in turn, phosphorylated PERK at Thr980 Subsequently, PERK phosphorylated eIF2α at Ser51, upregulating its downstream effector proteins ATF4 and GADD45α. RNAi-mediated knockdown of PERK or eIF2α abrogated the endorepellin-mediated up-regulation of GADD45α, the ultimate effector protein of this stress signaling cascade. To functionally validate these findings, we utilized an ex vivo model of angiogenesis. Exposure of the aortic rings embedded in 3D fibrillar collagen to recombinant endorepellin for 2-4 h activated PERK and induced GADD45α vis à vis vehicle-treated counterparts. Similar effects were obtained with the established cellular stress inducer tunicamycin. Notably, chronic exposure of aortic rings to endorepellin for 7-9 days markedly suppressed vessel sprouting, an angiostatic effect that was rescued by blocking PERK kinase activity. Our findings unravel a mechanism by which an extracellular matrix protein evokes stress signaling in endothelial cells, which leads to angiostasis.
Collapse
Affiliation(s)
- Aastha Kapoor
- Department of Pathology, Anatomy, and Cell Biology and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 19107
| | - Carolyn G Chen
- Department of Pathology, Anatomy, and Cell Biology and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 19107
| | - Renato V Iozzo
- Department of Pathology, Anatomy, and Cell Biology and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 19107
| |
Collapse
|
13
|
Szaraz P, Mander P, Gasner N, Librach M, Iqbal F, Librach C. Glucose withdrawal induces Endothelin 1 release with significant angiogenic effect from first trimester (FTM), but not term human umbilical cord perivascular cells (HUCPVC). Angiogenesis 2019; 23:131-144. [PMID: 31576475 DOI: 10.1007/s10456-019-09682-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 09/19/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Perivascular cells (PVC) and their "progeny," mesenchymal stromal cells (MSC), have high therapeutic potential for ischemic diseases. While hypoxia can increase their angiogenic properties, the other aspect of ischemic conditions-glucose shortage-is deleterious for MSC and limits their therapeutic applicability. Regenerative cells in developing vascular tissues, however, can adapt to varying glucose environment and react in a tissue-protective manner. Placental development and fetal insulin production generate different glucose fluxes in early and late extraembryonic tissues. We hypothesized that FTM HUCPVC, which are isolated from a developing vascular tissue with varying glucose availability react to low-glucose conditions in a pro-angiogenic manner in vitro. METHODS Xeno-free (Human Platelet Lysate 2.5%) expanded FTM (n = 3) and term (n = 3) HUCPVC lines were cultured in low (2 mM) and regular (4 mM) glucose conditions. After 72 h, the expression (Next Generation Sequencing) and secretion (Proteome Profiler) of angiogenic factors and the functional angiogenic effect (rat aortic ring assay and Matrigel™ plug) of the conditioned media were quantified and statistically compared between all cultures. RESULTS Low-glucose conditions had a significant post-transcriptional inductive effect on FTM HUCPVC angiogenic factor secretion, resulting in significantly higher VEGFc and Endothelin 1 release in 3 days compared to term counterparts. Conditioned media from low-glucose FTM HUCPVC cultures had a significantly higher endothelial network enhancing effect compared to all other experimental groups both in vitro aortic ring assay and in subcutan Matrigel™ plugs. Endothelin 1 depletion of the low-glucose FTM HUCPVC conditioned media significantly diminished its angiogenic effect CONCLUSIONS: FTM HUCPVC isolated from an early extraembryonic tissue show significant pro-angiogenic paracrine reaction in low-glucose conditions at least in part through the excess release of Endothelin 1. This can be a substantial advantage in cell therapy applications for ischemic injuries.
Collapse
Affiliation(s)
- Peter Szaraz
- Research Department, Create Program Inc., Suite 412, Toronto, ON, M5G 1N8, Canada.
| | - Poonam Mander
- Research Department, Create Program Inc., Suite 412, Toronto, ON, M5G 1N8, Canada
| | - Nadav Gasner
- Research Department, Create Program Inc., Suite 412, Toronto, ON, M5G 1N8, Canada
| | - Max Librach
- Research Department, Create Program Inc., Suite 412, Toronto, ON, M5G 1N8, Canada
| | - Farwah Iqbal
- Department Physiology, University of Toronto, Toronto, ON, Canada
| | - Clifford Librach
- Research Department, Create Program Inc., Suite 412, Toronto, ON, M5G 1N8, Canada.,Department Physiology, University of Toronto, Toronto, ON, Canada.,Department Obstetrics and Gynaecology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
14
|
Bazzigaluppi P, Beckett TL, Koletar MM, Hill ME, Lai A, Trivedi A, Thomason L, Dorr A, Gallagher D, Librach CL, Joo IL, McLaurin J, Stefanovic B. Combinatorial Treatment Using Umbilical Cord Perivascular Cells and Aβ Clearance Rescues Vascular Function Following Transient Hypertension in a Rat Model of Alzheimer Disease. Hypertension 2019; 74:1041-1051. [PMID: 31476904 PMCID: PMC6739147 DOI: 10.1161/hypertensionaha.119.13187] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Supplemental Digital Content is available in the text. Transient hypertension is a risk factor for Alzheimer disease (AD), but the effects of this interaction on brain vasculature are understudied. Addressing vascular pathology is a promising avenue to potentiate the efficacy of treatments for AD. We used arterial spin labeling magnetic resonance imaging to longitudinally assess brain vascular function and immunohistopathology to examine cerebrovascular remodeling and amyloid load. Hypertension was induced for 1 month by administration of l-NG-nitroarginine-methyl-ester in TgF344-AD rats at the prodromal stage. Following hypertension, nontransgenic rats showed transient cerebrovascular changes, whereas TgF344-AD animals exhibited sustained alterations in cerebrovascular function. Human umbilical cord perivascular cells in combination with scyllo-inositol, an inhibitor of Aβ oligomerization, resulted in normalization of hippocampal vascular function and remodeling, in contrast to either treatment alone. Prodromal stage hypertension exacerbates latter AD pathology, and the combination of human umbilical cord perivascular cells with amyloid clearance promotes cerebrovascular functional recovery.
Collapse
Affiliation(s)
- Paolo Bazzigaluppi
- From the Sunnybrook Research Institute, Toronto, Canada (P.B., T.L.B., M.M.K., M.E.H., A.L., A.T., L.T., A.D., C.L.L., I.L.J., J.M., B.S.)
| | - Tina L Beckett
- From the Sunnybrook Research Institute, Toronto, Canada (P.B., T.L.B., M.M.K., M.E.H., A.L., A.T., L.T., A.D., C.L.L., I.L.J., J.M., B.S.)
| | - Margaret M Koletar
- From the Sunnybrook Research Institute, Toronto, Canada (P.B., T.L.B., M.M.K., M.E.H., A.L., A.T., L.T., A.D., C.L.L., I.L.J., J.M., B.S.)
| | - Mary E Hill
- From the Sunnybrook Research Institute, Toronto, Canada (P.B., T.L.B., M.M.K., M.E.H., A.L., A.T., L.T., A.D., C.L.L., I.L.J., J.M., B.S.)
| | - Aaron Lai
- From the Sunnybrook Research Institute, Toronto, Canada (P.B., T.L.B., M.M.K., M.E.H., A.L., A.T., L.T., A.D., C.L.L., I.L.J., J.M., B.S.)
| | - Arunachala Trivedi
- From the Sunnybrook Research Institute, Toronto, Canada (P.B., T.L.B., M.M.K., M.E.H., A.L., A.T., L.T., A.D., C.L.L., I.L.J., J.M., B.S.)
| | - Lynsie Thomason
- From the Sunnybrook Research Institute, Toronto, Canada (P.B., T.L.B., M.M.K., M.E.H., A.L., A.T., L.T., A.D., C.L.L., I.L.J., J.M., B.S.)
| | - Adrienne Dorr
- From the Sunnybrook Research Institute, Toronto, Canada (P.B., T.L.B., M.M.K., M.E.H., A.L., A.T., L.T., A.D., C.L.L., I.L.J., J.M., B.S.)
| | | | - Clifford L Librach
- From the Sunnybrook Research Institute, Toronto, Canada (P.B., T.L.B., M.M.K., M.E.H., A.L., A.T., L.T., A.D., C.L.L., I.L.J., J.M., B.S.).,Division of Obstetrics and Gynaecology, Laboratory Medicine and Pathobiology (C.L.), University of Toronto, Canada.,CReATe Research Program, Toronto, Canada (D.G., C.L.L.)
| | - Illsung L Joo
- From the Sunnybrook Research Institute, Toronto, Canada (P.B., T.L.B., M.M.K., M.E.H., A.L., A.T., L.T., A.D., C.L.L., I.L.J., J.M., B.S.)
| | - JoAnne McLaurin
- From the Sunnybrook Research Institute, Toronto, Canada (P.B., T.L.B., M.M.K., M.E.H., A.L., A.T., L.T., A.D., C.L.L., I.L.J., J.M., B.S.)
| | - Bojana Stefanovic
- From the Sunnybrook Research Institute, Toronto, Canada (P.B., T.L.B., M.M.K., M.E.H., A.L., A.T., L.T., A.D., C.L.L., I.L.J., J.M., B.S.).,Department of Medical Biophysics (B.S.), University of Toronto, Canada
| |
Collapse
|
15
|
Vawda R, Badner A, Hong J, Mikhail M, Lakhani A, Dragas R, Xhima K, Barretto T, Librach CL, Fehlings MG. Early Intravenous Infusion of Mesenchymal Stromal Cells Exerts a Tissue Source Age-Dependent Beneficial Effect on Neurovascular Integrity and Neurobehavioral Recovery After Traumatic Cervical Spinal Cord Injury. Stem Cells Transl Med 2019; 8:639-649. [PMID: 30912623 PMCID: PMC6591557 DOI: 10.1002/sctm.18-0192] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 02/04/2019] [Indexed: 12/16/2022] Open
Abstract
Localized vascular disruption after traumatic spinal cord injury (SCI) triggers a cascade of secondary events, including inflammation, gliosis, and scarring, that can further impact recovery. In addition to immunomodulatory and neurotrophic properties, mesenchymal stromal cells (MSCs) possess pericytic characteristics. These features make MSCs an ideal candidate for acute cell therapy targeting vascular disruption, which could reduce the severity of secondary injury, enhance tissue preservation and repair, and ultimately promote functional recovery. A moderately severe cervical clip compression/contusion injury was induced at C7‐T1 in adult female rats, followed by an intravenous tail vein infusion 1 hour post‐SCI of (a) term‐birth human umbilical cord perivascular cells (HUCPVCs); (b) first‐trimester human umbilical cord perivascular cells (FTM HUCPVCs); (c) adult bone marrow mesenchymal stem cells; or (d) vehicle control. Weekly behavioral testing was performed. Rats were sacrificed at 24 hours or 10 weeks post‐SCI and immunohistochemistry and ultrasound imaging were performed. Both term and FTM HUCPVC‐infused rats displayed improved (p < .05) grip strength compared with vehicle controls. However, only FTM HUCPVC‐infusion led to significant weight gain. All cell infusion treatments resulted in reduced glial scarring (p < .05). Cell infusion also led to increased axonal, myelin, and vascular densities (p < .05). Although post‐traumatic cavity volume was reduced with cell infusion, this did not reach significance. Taken together, we demonstrate selective long‐term functional recovery alongside histological improvements with HUCPVC infusion in a clinically relevant model of cervical SCI. Our findings highlight the potential of these cells for acute therapeutic intervention after SCI.
Collapse
Affiliation(s)
- Reaz Vawda
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Anna Badner
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - James Hong
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Mirriam Mikhail
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Alam Lakhani
- CReATe Fertility Centre, Toronto, Ontario, Canada
| | - Rachel Dragas
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Kristiana Xhima
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | | | - Michael G Fehlings
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.,Division of Neurosurgery and Spinal Program, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
16
|
Bieback K, Kuçi S, Schäfer R. Production and quality testing of multipotent mesenchymal stromal cell therapeutics for clinical use. Transfusion 2019; 59:2164-2173. [DOI: 10.1111/trf.15252] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 02/10/2019] [Accepted: 02/10/2019] [Indexed: 12/16/2022]
Affiliation(s)
- Karen Bieback
- Institute of Transfusion Medicine and Immunology, Medical Faculty MannheimGerman Red Cross Blood Donor Service Baden‐Württemberg‐Hessen gGmbH, Heidelberg University Mannheim Germany
- FlowCore Mannheim, Medical Faculty MannheimHeidelberg University Germany
| | - Selim Kuçi
- Department for Children and Adolescents, Division for Stem Cell Transplantation and ImmunologyUniversity Hospital Frankfurt Frankfurt am Main Germany
| | - Richard Schäfer
- Institute for Transfusion Medicine and Immunohaematology, German Red Cross Blood Donor Service Baden‐Württemberg‐Hessen gGmbHGoethe University Hospital Frankfurt am Main Germany
| |
Collapse
|
17
|
Gauthier-Fisher A, Szaraz P, Librach CL. Pericytes in the Umbilical Cord. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1122:211-233. [DOI: 10.1007/978-3-030-11093-2_12] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
18
|
Amărandi RM, Becheru DF, Vlăsceanu GM, Ioniță M, Burns JS. Advantages of Graphene Biosensors for Human Stem Cell Therapy Potency Assays. BIOMED RESEARCH INTERNATIONAL 2018; 2018:1676851. [PMID: 30003089 PMCID: PMC5996421 DOI: 10.1155/2018/1676851] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 03/22/2018] [Indexed: 12/11/2022]
Abstract
Regenerative medicine is challenged by the need to conform to rigorous guidelines for establishing safe and effective development and translation of stem cell-based therapies. Counteracting widespread concerns regarding unproven cell therapies, stringent cell-based assays seek not only to avoid harm but also to enhance quality and efficacy. Potency indicates that the cells are functionally fit for purpose before they are administered to the patient. It is a paramount quantitative critical quality attribute serving as a decisive release criterion. Given a broad range of stem cell types and therapeutic contexts the potency assay often comprises one of the most demanding hurdles for release of a cell therapy medicinal product. With need for improved biomarker assessment and expedited measurement, recent advances in graphene-based biosensors suggest that they are poised to be valuable platforms for accelerating potency assay development. Among several potential advantages, they offer versatility for sensitive measurement of a broad range of potential biomarker types, cell biocompatibility for direct measurement, and small sample sufficiency, plus ease of use and point-of-care applicability.
Collapse
Affiliation(s)
- Roxana-Maria Amărandi
- Faculty of Medical Engineering, University Politehnica of Bucharest, Gh. Polizu 1-7, 011061 Bucharest, Romania
| | - Diana F. Becheru
- Faculty of Medical Engineering, University Politehnica of Bucharest, Gh. Polizu 1-7, 011061 Bucharest, Romania
| | - George M. Vlăsceanu
- Faculty of Medical Engineering, University Politehnica of Bucharest, Gh. Polizu 1-7, 011061 Bucharest, Romania
| | - Mariana Ioniță
- Faculty of Medical Engineering, University Politehnica of Bucharest, Gh. Polizu 1-7, 011061 Bucharest, Romania
- Advanced Polymer Materials Group, University Politehnica of Bucharest, Gh. Polizu 1-7, 011061 Bucharest, Romania
| | - Jorge S. Burns
- Faculty of Medical Engineering, University Politehnica of Bucharest, Gh. Polizu 1-7, 011061 Bucharest, Romania
- Department of Medical and Surgical Sciences of Children and Adults, University Hospital of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
19
|
The WNT7B protein promotes the migration and differentiation of human dental pulp cells partly through WNT/beta-catenin and c-Jun N-terminal kinase signalling pathways. Arch Oral Biol 2017; 87:54-61. [PMID: 29268145 DOI: 10.1016/j.archoralbio.2017.12.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 12/13/2017] [Accepted: 12/14/2017] [Indexed: 02/05/2023]
Abstract
OBJECTIVE The aim of this study is to investigate the role of the WNT7B protein in the migration and differentiation of human dental pulp cells (HDPCs). DESIGN The effect of recombinant human WNT7B (rhWNT7B) on the proliferation and migration of HDPCs was evaluated by 5-ethynyl-2'-deoxyuridine (EdU), immunofluorescence staining of Ki67, flow cytometry and scratch assay; the differentiation of HDPCs was measured by alkaline phosphatase (ALP) staining, alizarin red staining, ALP activity, qPCR and western blot. The activation of the WNT/beta-catenin (WNT/β-catenin) and c-Jun N-terminal kinase (JNK) pathways was analysed by western blot, immunocytochemistry and dual luciferase assays. XAV939 and SP600125,the inhibitors of the WNT/β-catenin and JNK pathways, were further applied to verify the mechanism. RESULTS rhWNT7B repressed the proliferation but did not affect the apoptosis of HDPCs. In the presence of rhWNT7B, ALP and alizarin red staining were increased substantially in the HDPCs with osteogenic induction; the gene expression of Runx2 and Col1 in HDPCs was quite elevated compared with that induced in osteogenic medium without WNT7B measured by qPCR; The ALP activity was also increased with rhWNT7B stimulation in HDPCs after 7-day odontogenic culture; Western blot revealed that the expression of dentin sialophosphoprotein (DSPP) of HDPCs was up-regulated significantly with the addition of WNT7B as well. Further study showed that rhWNT7B activated the WNT/β-catenin and JNK signalling pathways in the differentiation of HDPCs. XAV939 and SP600125 can partly offset the effect of the WNT7B-induced differentiation of HDPCs. CONCLUSION WNT7B promoted the differentiation of HDPCs partly through the WNT/β-catenin and JNK signalling pathways.
Collapse
|