51
|
Calpe B, Kovacs WJ. High-throughput screening in multicellular spheroids for target discovery in the tumor microenvironment. Expert Opin Drug Discov 2020; 15:955-967. [PMID: 32364413 DOI: 10.1080/17460441.2020.1756769] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Solid tumors are highly influenced by a complex tumor microenvironment (TME) that cannot be modeled with conventional two-dimensional (2D) cell culture. In addition, monolayer culture conditions tend to induce undesirable molecular and phenotypic cellular changes. The discrepancy between in vitro and in vivo is an important factor accounting for the high failure rate in drug development. Three-dimensional (3D) multicellular tumor spheroids (MTS) more closely resemble the in vivo situation in avascularized tumors. AREAS COVERED This review describes the use of MTS for anti-cancer drug discovery, with an emphasis on high-throughput screening (HTS) compatible assays. In particular, we focus on how these assays can be used for target discovery in the context of the TME. EXPERT OPINION Arrayed MTS in microtiter plates are HTS compatible but remain more expensive and time consuming than their 2D culture counterpart. It is therefore imperative to use assays with multiplexed readouts, in order to maximize the information that can be gained with the screen. In this context, high-content screening allowing to uncover microenvironmental dependencies is the true added value of MTS-based screening compared to 2D culture-based screening. Hit translation in animal models will, however, be key to allow a broader use of MTS-based screening in industry.
Collapse
Affiliation(s)
- Blaise Calpe
- Institute of Molecular Health Sciences, ETH Zurich , Zurich, Switzerland.,Department of Biology, Debiopharm , Lausanne, Switzerland
| | - Werner J Kovacs
- Institute of Molecular Health Sciences, ETH Zurich , Zurich, Switzerland
| |
Collapse
|
52
|
Zhang S, Kim B, Zhu X, Gui X, Wang Y, Lan Z, Prabhu P, Fond K, Wang A, Guo F. Glial type specific regulation of CNS angiogenesis by HIFα-activated different signaling pathways. Nat Commun 2020; 11:2027. [PMID: 32332719 PMCID: PMC7181614 DOI: 10.1038/s41467-020-15656-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 03/12/2020] [Indexed: 01/13/2023] Open
Abstract
The mechanisms by which oligodendroglia modulate CNS angiogenesis remain elusive. Previous in vitro data suggest that oligodendroglia regulate CNS endothelial cell proliferation and blood vessel formation through hypoxia inducible factor alpha (HIFα)-activated Wnt (but not VEGF) signaling. Using in vivo genetic models, we show that HIFα in oligodendroglia is necessary and sufficient for angiogenesis independent of CNS regions. At the molecular level, HIFα stabilization in oligodendroglia does not perturb Wnt signaling but rather activates VEGF. At the functional level, genetically blocking oligodendroglia-derived VEGF but not Wnt significantly decreases oligodendroglial HIFα-regulated CNS angiogenesis. Blocking astroglia-derived Wnt signaling reduces astroglial HIFα-regulated CNS angiogenesis. Together, our in vivo data demonstrate that oligodendroglial HIFα regulates CNS angiogenesis through Wnt-independent and VEGF-dependent signaling. These findings suggest an alternative mechanistic understanding of CNS angiogenesis by postnatal glial cells and unveil a glial cell type-dependent HIFα-Wnt axis in regulating CNS vessel formation. In the central nervous system, the maturation of glial cells is temporally and functionally coupled with that of the vascular network during postnatal development. Here the authors show that oligodendroglial HIFα regulates CNS angiogenesis through Wnt-independent and VEGF-dependent signaling, while astroglial HIFα participates through Wnt-dependent signaling.
Collapse
Affiliation(s)
- Sheng Zhang
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children/UC Davis School of Medicine, Sacramento, CA, 95817, USA.,Department of Neurology, School of Medicine, UC Davis, Sacramento, CA, 95817, USA
| | - Bokyung Kim
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children/UC Davis School of Medicine, Sacramento, CA, 95817, USA.,Department of Neurology, School of Medicine, UC Davis, Sacramento, CA, 95817, USA
| | - Xiaoqing Zhu
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children/UC Davis School of Medicine, Sacramento, CA, 95817, USA.,Qingdao University, Qingdao, China
| | - Xuehong Gui
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children/UC Davis School of Medicine, Sacramento, CA, 95817, USA
| | - Yan Wang
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children/UC Davis School of Medicine, Sacramento, CA, 95817, USA.,Department of Neurology, School of Medicine, UC Davis, Sacramento, CA, 95817, USA
| | - Zhaohui Lan
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children/UC Davis School of Medicine, Sacramento, CA, 95817, USA.,Department of Neurology, School of Medicine, UC Davis, Sacramento, CA, 95817, USA
| | - Preeti Prabhu
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children/UC Davis School of Medicine, Sacramento, CA, 95817, USA
| | - Kenneth Fond
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children/UC Davis School of Medicine, Sacramento, CA, 95817, USA
| | - Aijun Wang
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children/UC Davis School of Medicine, Sacramento, CA, 95817, USA.,Department of Surgery, School of Medicine, UC Davis, Sacramento, CA, 95817, USA
| | - Fuzheng Guo
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children/UC Davis School of Medicine, Sacramento, CA, 95817, USA. .,Department of Neurology, School of Medicine, UC Davis, Sacramento, CA, 95817, USA.
| |
Collapse
|
53
|
Adolfsson E, Helenius G, Friberg Ö, Samano N, Frøbert O, Johansson K. Bone marrow- and adipose tissue-derived mesenchymal stem cells from donors with coronary artery disease; growth, yield, gene expression and the effect of oxygen concentration. Scandinavian Journal of Clinical and Laboratory Investigation 2020; 80:318-326. [PMID: 32189529 DOI: 10.1080/00365513.2020.1741023] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Mesenchymal stem cells (MSCs) for cardiovascular cell therapy are procured from different sources including bone marrow and adipose tissue. Differently located MSCs differ in growth potential, differentiation ability and gene expression when cultured in vitro, and studies show different healing abilities for different MSC subgroups. In this study, bone marrow derived MSCs (BMSCs) and adipose tissue derived MSCs (ADSCs) from six human donors with coronary artery disease were compared for growth potential and expression of target genes (Angpt1, LIF, HGF, TGF-β1 and VEGF-A) in response to exposure to 1% and 5% O2, for up to 48 h. We found greater growth of ADSCs compared to BMSCs. ADSCs expressed higher levels of Angpt1, LIF and TGF-β1 and equal levels of VEGF-A and HGF as BMSCs. In BMSCs, exposure to low oxygen resulted in upregulation of TGF-β1, whereas other target genes were unaffected. Upregulation was only present at 1% O2. In ADSCs, LIF was upregulated in both oxygen concentrations, whereas Angpt1 was upregulated only at 1% O2. Different response to reduced oxygen culture conditions is of relevance when expanding cells in vitro prior to administration. These findings indicate ADSCs as better suited for cardiovascular cell therapy compared to BMSCs.
Collapse
Affiliation(s)
- Emma Adolfsson
- Department of Laboratory Medicine, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Gisela Helenius
- Department of Laboratory Medicine, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Örjan Friberg
- Department of Cardiothoracic Surgery, Faculty of Health, Örebro University, Örebro, Sweden
| | - Ninos Samano
- Department of Cardiothoracic Surgery, Faculty of Health, Örebro University, Örebro, Sweden
| | - Ole Frøbert
- Department of Cardiology, Faculty of Health, Örebro University, Örebro, Sweden
| | - Karin Johansson
- Department of Laboratory Medicine, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| |
Collapse
|
54
|
Three-dimensional culture models mimic colon cancer heterogeneity induced by different microenvironments. Sci Rep 2020; 10:3156. [PMID: 32081957 PMCID: PMC7035265 DOI: 10.1038/s41598-020-60145-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 02/07/2020] [Indexed: 11/09/2022] Open
Abstract
Colorectal cancer demonstrates intra-tumour heterogeneity formed by a hierarchical structure comprised of cancer stem cells (CSCs) and their differentiated progenies. The mechanism by which CSCs are maintained and differentiated needs to be further elucidated, and there is evidence that the tumour microenvironment governs cancer stemness. Using PLR123, a colon cancer cell line with CSC properties, we determined the culture conditions necessary to establish a pair of three-dimensional (3D) culture models grown in Matrigel, designated stemCO and diffCO. The conditions were determined by comparing the phenotypes in the models with PLR123 mouse xenografts colonising lung and liver. StemCO resembled LGR5-positive undifferentiated tumours in the lung, and diffCO had lumen structures composed of polarised cells that were similar to the ductal structures found in differentiated tumours in the liver. In a case using the models for biomedical research, treatment with JAG-1 peptide or a γ-secretase inhibitor modified the Notch signaling and induced changes indicating that the signal participates in lumen formation in the models. Our results demonstrate that culture conditions affect the stemness of 3D culture models generated from CSCs and show that comparing models with different phenotypes is useful for studying how the tumour environment regulates cancer.
Collapse
|
55
|
Development and Evaluation of Cell Culture Devices with the Gas-permeable Membrane. BIOTECHNOL BIOPROC E 2020. [DOI: 10.1007/s12257-019-0149-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
56
|
Adipose-derived stromal cell immunosuppression of T cells is enhanced under "physiological" hypoxia. Tissue Cell 2019; 63:101320. [PMID: 32223948 DOI: 10.1016/j.tice.2019.101320] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 11/12/2019] [Accepted: 12/02/2019] [Indexed: 01/10/2023]
Abstract
Multipotent mesenchymal stromal cells (MSCs) are characterized by immunomodulatory properties along with the high proliferative and paracrine activity, as well as multilineage potency. The effects of MSCs on the T cell adaptive immunity are of a special interest. Low O2 level (1-7 %) is known to be typical for the putative site of the MSC - T cell interactions. A comparative evaluation of the effects of adipose tissue derived MSC (ASCs) on the mitogen-stimulated T cells at the ambient (20 %) and tissue-related (5 %) O2 levels demonstrated reduced T cell activation by the HLA-DR expression, decreased pro-inflammatory and increased anti-inflammatory cytokine production in co-culture, inhibited T cell proliferation, with the effects increased at hypoxia. T cell interactions with ASCs resulted in the up-regulation of PDCD1, Foxp3, and TGFβ1 known to play an important role in the immune response suppression, and in the down-regulation of genes involved in the inflammatory reaction (IL2, IFNG). These changes were significantly increased under hypoxia. At the same time, neither ASCs nor the reduced O2 level had negative effects on the viability of T cells.
Collapse
|
57
|
Zhang K, Cui X, Zhang B, Song X, Liu Q, Yang S. Multipotent stem cells with neural crest stem cells characteristics exist in bovine adipose tissue. Biochem Biophys Res Commun 2019; 522:819-825. [PMID: 31791582 DOI: 10.1016/j.bbrc.2019.11.176] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 11/26/2019] [Indexed: 01/12/2023]
Abstract
Neural crest stem cells (NCSCs) often referred to as the fourth germ layer, comprise a migratory, stem and progenitor cell population and are synonymous with vertebrate evolution and development. The cells follow specific paths to migrate to different locations of the body where they generate a diverse array of cell types and tissues. There are NCSCs which are maintained in an undifferentiated state throughout the life in the animal tissues. Based on some cells migratory property, we successfully developed a separation strategy to isolate and identify a population of adipose-derived stem cells with neural crest stem cell features in adult bovine adipose tissues within minimally-invasive surgical procedures. The cells have a high degree of multi-potency and self-renewal capabilities, can be cultured and maintained in feeder-free adhesion conditions as monolayer cells, and also be able to grow in the suspension condition in the form of neurosphere. For the purpose of simple description, we name this type cell as bovine adipose-derived neural crest stem cell (baNCSC). Taken together our study describes a readily accessible source of multipotent baNCSC for autologous tissue engineer and cell-based therapeutic researches.
Collapse
Affiliation(s)
- Kai Zhang
- College of Animal Sciences and Veterinary Medicines, Shanxi Agricultural University, 030801, Taigu, Shanxi Province, PR China; Feed and Veterinary Medicine Research Institute, Shanxi Academy of Agriculture Sciences, 030036, Taiyuan, Shanxi Province, PR China
| | - Xiaozhen Cui
- Feed and Veterinary Medicine Research Institute, Shanxi Academy of Agriculture Sciences, 030036, Taiyuan, Shanxi Province, PR China
| | - Bochi Zhang
- Feed and Veterinary Medicine Research Institute, Shanxi Academy of Agriculture Sciences, 030036, Taiyuan, Shanxi Province, PR China
| | - Xianyi Song
- Feed and Veterinary Medicine Research Institute, Shanxi Academy of Agriculture Sciences, 030036, Taiyuan, Shanxi Province, PR China
| | - Qiang Liu
- College of Animal Sciences and Veterinary Medicines, Shanxi Agricultural University, 030801, Taigu, Shanxi Province, PR China.
| | - Shiyu Yang
- Department of Clinical Neurosciences, UCL Institute of Neurology, Rowland Hill Street, London, NW3 2PF, UK.
| |
Collapse
|
58
|
Isolation and Culture of Human Stem Cells from Apical Papilla under Low Oxygen Concentration Highlight Original Properties. Cells 2019; 8:cells8121485. [PMID: 31766521 PMCID: PMC6952825 DOI: 10.3390/cells8121485] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/07/2019] [Accepted: 11/14/2019] [Indexed: 12/12/2022] Open
Abstract
Stem cells isolated from the apical papilla of wisdom teeth (SCAPs) are an attractive model for tissue repair due to their availability, high proliferation rate and potential to differentiate in vitro towards mesodermal and neurogenic lineages. Adult stem cells, such as SCAPs, develop in stem cell niches in which the oxygen concentration [O2] is low (3–8% compared with 21% of ambient air). In this work, we evaluate the impact of low [O2] on the physiology of SCAPs isolated and processed in parallel at 21% or 3% O2 without any hyperoxic shock in ambient air during the experiment performed at 3% O2. We demonstrate that SCAPs display a higher proliferation capacity at 3% O2 than in ambient air with elevated expression levels of two cell surface antigens: the alpha-6 integrin subunit (CD49f) and the embryonic stem cell marker (SSEA4). We show that the mesodermal differentiation potential of SCAPs is conserved at early passage in both [O2], but is partly lost at late passage and low [O2], conditions in which SCAPs proliferate efficiently without any sign of apoptosis. Unexpectedly, we show that autophagic flux is active in SCAPs irrespective of [O2] and that this process remains high in cells even after prolonged exposure to 3% O2.
Collapse
|
59
|
Godet I, Shin YJ, Ju JA, Ye IC, Wang G, Gilkes DM. Fate-mapping post-hypoxic tumor cells reveals a ROS-resistant phenotype that promotes metastasis. Nat Commun 2019; 10:4862. [PMID: 31649238 PMCID: PMC6813355 DOI: 10.1038/s41467-019-12412-1] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 09/06/2019] [Indexed: 12/30/2022] Open
Abstract
Hypoxia is known to be detrimental in cancer and contributes to its development. In this work, we present an approach to fate-map hypoxic cells in vivo in order to determine their cellular response to physiological O2 gradients as well as to quantify their contribution to metastatic spread. We demonstrate the ability of the system to fate-map hypoxic cells in 2D, and in 3D spheroids and organoids. We identify distinct gene expression patterns in cells that experienced intratumoral hypoxia in vivo compared to cells exposed to hypoxia in vitro. The intratumoral hypoxia gene-signature is a better prognostic indicator for distant metastasis-free survival. Post-hypoxic tumor cells have an ROS-resistant phenotype that provides a survival advantage in the bloodstream and promotes their ability to establish overt metastasis. Post-hypoxic cells retain an increase in the expression of a subset of hypoxia-inducible genes at the metastatic site, suggesting the possibility of a 'hypoxic memory.'
Collapse
Affiliation(s)
- Inês Godet
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Yu Jung Shin
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Julia A Ju
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, 21218, USA
| | - I Chae Ye
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Guannan Wang
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Daniele M Gilkes
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA.
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, 21218, USA.
- Cellular and Molecular Medicine Program, The Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA.
| |
Collapse
|
60
|
Wang J, Xue X, Fan K, Liu Q, Zhang S, Peng M, Zhou J, Cao Z. Moderate hypoxia modulates ABCG2 to promote the proliferation of mouse spermatogonial stem cells by maintaining mild ROS levels. Theriogenology 2019; 145:149-157. [PMID: 31733931 DOI: 10.1016/j.theriogenology.2019.10.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 09/25/2019] [Accepted: 10/09/2019] [Indexed: 12/25/2022]
Abstract
The aim of this study was to investigate the effects of different oxygen (O2) concentrations on the growth of mouse spermatogonial stem cells (SSCs) and the possible mechanisms of cell proliferation in vitro. The SSCs from testicular cells were cultured in various O2 concentrations (1%, 2.5%, 5%, and 20% O2) for 7 days. Colonies of SSCs were identified morphologically and by immunofluorescence. The number of mouse SSC colonies and the area covered by them were measured. Cell cycle progression of the SSCs was analyzed to identify the state of cell proliferation. The effects of O2 concentrations on the levels of intracellular reactive oxygen species (ROS) and expression of ATP binding cassette subfamily G member 2 (ABCG2) were also analyzed in the SSCs. Following culturing for 7 days, the SSCs were treated with Ko143 (a specific inhibitor of ABCG2) for 1 h, and the ROS level and expression of bcl-2, bax, and p53 were analyzed. The results showed that mouse SSCs formed compact colonies and had unclear borders in different O2 concentrations for 7 days, and there were no major morphologic differences between the O2 treatment groups. The expression of the SSC marker, GFR α1 was studied in each O2 treatment group. The number and area of SSC colonies, and the number of GFR α1 positive cells were the highest in the 2.5% O2 treatment group. Compared with other O2 concentrations, the number of cells in G0 cycle was significantly higher, while the level of intracellular ROS was lower at 1% O2. Moreover, the intracellular ROS levels gradually increased with increasing O2 concentration from 1% to 20%. The expression of ABCG2 in the SSCs cultured at 2.5% O2 was higher than in the other O2 groups. Inhibition of ABCG2 increased intracellular ROS generation, and the expression of the pro-apoptotic genes bax and p53, and decreased the expression of the anti-apoptotic gene bcl-2. In conclusion, moderate to low O2 tension increases ABCG2 expression to maintain mild ROS levels, triggers the expression of the anti-apoptotic genes, suppresses the proapoptotic gene pathway, and further promotes the proliferation of mouse SSCs in vitro.
Collapse
Affiliation(s)
- Juhua Wang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China; Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding in Anhui Provincial, Hefei, China; Key Laboratory of Veterinary Pathobiology and Disease Control in Anhui Provincial, Hefei, China.
| | - Xiuheng Xue
- College of Tea & Food Science and Technology, Anhui Agricultural University, Hefei, China.
| | - Kai Fan
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Qi Liu
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Suzi Zhang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Mengling Peng
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China; Key Laboratory of Veterinary Pathobiology and Disease Control in Anhui Provincial, Hefei, China
| | - Jie Zhou
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China; Key Laboratory of Veterinary Pathobiology and Disease Control in Anhui Provincial, Hefei, China
| | - Zubing Cao
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China; Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding in Anhui Provincial, Hefei, China
| |
Collapse
|
61
|
Hadida M, Marchat D. Strategy for achieving standardized bone models. Biotechnol Bioeng 2019; 117:251-271. [PMID: 31531968 PMCID: PMC6915912 DOI: 10.1002/bit.27171] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 09/05/2019] [Accepted: 09/06/2019] [Indexed: 12/24/2022]
Abstract
Reliably producing functional in vitro organ models, such as organ-on-chip systems, has the potential to considerably advance biology research, drug development time, and resource efficiency. However, despite the ongoing major progress in the field, three-dimensional bone tissue models remain elusive. In this review, we specifically investigate the control of perfusion flow effects as the missing link between isolated culture systems and scientifically exploitable bone models and propose a roadmap toward this goal.
Collapse
Affiliation(s)
- Mikhael Hadida
- Mines Saint-Etienne, Univ Lyon, Univ Jean Monnet, INSERM, U 1059 Sainbiose, Centre CIS, Saint-Etienne, France
| | - David Marchat
- Mines Saint-Etienne, Univ Lyon, Univ Jean Monnet, INSERM, U 1059 Sainbiose, Centre CIS, Saint-Etienne, France
| |
Collapse
|
62
|
Yu JSL, Palano G, Lim C, Moggio A, Drowley L, Plowright AT, Bohlooly‐Y M, Rosen BS, Hansson EM, Wang Q, Yusa K. CRISPR-Knockout Screen Identifies Dmap1 as a Regulator of Chemically Induced Reprogramming and Differentiation of Cardiac Progenitors. Stem Cells 2019; 37:958-972. [PMID: 30932271 PMCID: PMC6767549 DOI: 10.1002/stem.3012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 02/11/2019] [Accepted: 03/02/2019] [Indexed: 12/12/2022]
Abstract
Direct in vivo reprogramming of cardiac fibroblasts into myocytes is an attractive therapeutic intervention in resolving myogenic deterioration. Current transgene-dependent approaches can restore cardiac function, but dependence on retroviral delivery and persistent retention of transgenic sequences are significant therapeutic hurdles. Chemical reprogramming has been established as a legitimate method to generate functional cell types, including those of the cardiac lineage. Here, we have extended this approach to generate progenitor cells that can differentiate into endothelial cells and cardiomyocytes using a single inhibitor protocol. Depletion of terminally differentiated cells and enrichment for proliferative cells result in a second expandable progenitor population that can robustly give rise to myofibroblasts and smooth muscle. Deployment of a genome-wide knockout screen with clustered regularly interspaced short palindromic repeats-guide RNA library to identify novel mediators that regulate the reprogramming revealed the involvement of DNA methyltransferase 1-associated protein 1 (Dmap1). Loss of Dmap1 reduced promoter methylation, increased the expression of Nkx2-5, and enhanced the retention of self-renewal, although further differentiation is inhibited because of the sustained expression of Cdh1. Our results hence establish Dmap1 as a modulator of cardiac reprogramming and myocytic induction. Stem Cells 2019;37:958-972.
Collapse
MESH Headings
- Animals
- Benzamides/pharmacology
- CRISPR-Cas Systems
- Cadherins/genetics
- Cadherins/metabolism
- Cell Differentiation/drug effects
- Cell Proliferation/drug effects
- Cellular Reprogramming/drug effects
- Cellular Reprogramming/genetics
- Dioxoles/pharmacology
- Fibroblasts/cytology
- Fibroblasts/drug effects
- Fibroblasts/metabolism
- Gene Editing/methods
- Homeobox Protein Nkx-2.5/genetics
- Homeobox Protein Nkx-2.5/metabolism
- Humans
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth/cytology
- Muscle, Smooth/metabolism
- Myocardium/cytology
- Myocardium/metabolism
- Myocytes, Cardiac/cytology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Primary Cell Culture
- Pyrazoles/pharmacology
- Pyridines/pharmacology
- RNA, Guide, CRISPR-Cas Systems/genetics
- RNA, Guide, CRISPR-Cas Systems/metabolism
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Stem Cells/cytology
- Stem Cells/drug effects
- Stem Cells/metabolism
Collapse
Affiliation(s)
- Jason S. L. Yu
- Stem Cell Genetics, Wellcome Sanger InstituteHinxton, CambridgeUnited Kingdom
- Department of Cell BiologyThe Francis Crick InstituteLondonUnited Kingdom
| | - Giorgia Palano
- KI/AZ Integrated CardioMetabolic Center (ICMC), Department of MedicineKarolinska InstitutetHuddingeSweden
| | - Cindy Lim
- Bioscience Heart Failure, Cardiovascular, Renal and Metabolism, IMED Biotech UnitAstraZenecaGothenburgSweden
| | - Aldo Moggio
- KI/AZ Integrated CardioMetabolic Center (ICMC), Department of MedicineKarolinska InstitutetHuddingeSweden
| | - Lauren Drowley
- Bioscience Heart Failure, Cardiovascular, Renal and Metabolism, IMED Biotech UnitAstraZenecaGothenburgSweden
| | - Alleyn T. Plowright
- Medicinal Chemistry, Cardiovascular, Renal and Metabolism, IMED Biotech UnitAstraZenecaGothenburgSweden
| | | | - Barry S. Rosen
- Discovery Sciences, IMED Biotech UnitAstraZenecaGothenburgSweden
| | - Emil M. Hansson
- KI/AZ Integrated CardioMetabolic Center (ICMC), Department of MedicineKarolinska InstitutetHuddingeSweden
| | - Qing‐Dong Wang
- Bioscience Heart Failure, Cardiovascular, Renal and Metabolism, IMED Biotech UnitAstraZenecaGothenburgSweden
| | - Kosuke Yusa
- Stem Cell Genetics, Wellcome Sanger InstituteHinxton, CambridgeUnited Kingdom
- Stem Cell GeneticsInstitute for Frontier Life and Medical Sciences, Kyoto UniversityKyotoJapan
| |
Collapse
|
63
|
Musah-Eroje A, Watson S. Adaptive Changes of Glioblastoma Cells Following Exposure to Hypoxic (1% Oxygen) Tumour Microenvironment. Int J Mol Sci 2019; 20:ijms20092091. [PMID: 31035344 PMCID: PMC6539006 DOI: 10.3390/ijms20092091] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/21/2019] [Accepted: 04/24/2019] [Indexed: 12/11/2022] Open
Abstract
Glioblastoma multiforme is the most aggressive and malignant primary brain tumour, with a median survival rate of between 15 to 17 months. Heterogeneous regions occur in glioblastoma as a result of oxygen gradients which ranges from 0.1% to 10% in vivo. Emerging evidence suggests that tumour hypoxia leads to increased aggressiveness and chemo/radio resistance. Yet, few in vitro studies have been performed in hypoxia. Using three glioblastoma cell-lines (U87, U251, and SNB19), the adaptation of glioblastoma cells in a 1% (hypoxia) and 20% (normoxia) oxygen microenvironment on proliferation, metabolism, migration, neurosphere formation, CD133 and VEGF expression was investigated. Compared to cells maintained in normoxia (20% oxygen), glioblastoma cells adapted to 1% oxygen tension by reducing proliferation and enhancing metabolism. Both migratory tendency and neurosphere formation ability were greatly limited. In addition, hypoxic-mediated gene upregulation (CD133 and VEGF) was reversed when cells were removed from the hypoxic environment. Collectively, our results reveal that hypoxia plays a pivotal role in changing the behaviour of glioblastoma cells. We have also shown that genetic modulation can be reversed, supporting the concept of reversibility. Thus, understanding the degree of oxygen gradient in glioblastoma will be crucial in personalising treatment for glioblastoma patients.
Collapse
Affiliation(s)
- Ahmed Musah-Eroje
- Division of Cancer and Stem Cells, Cancer Biology, University of Nottingham, Nottingham NG7 2UH, UK.
- School of Life Sciences, University of Bedfordshire, Luton LU1 3JU, UK.
| | - Sue Watson
- Division of Cancer and Stem Cells, Cancer Biology, University of Nottingham, Nottingham NG7 2UH, UK.
| |
Collapse
|
64
|
Gaudichon J, Jakobczyk H, Debaize L, Cousin E, Galibert MD, Troadec MB, Gandemer V. Mechanisms of extramedullary relapse in acute lymphoblastic leukemia: Reconciling biological concepts and clinical issues. Blood Rev 2019; 36:40-56. [PMID: 31010660 DOI: 10.1016/j.blre.2019.04.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 04/03/2019] [Accepted: 04/15/2019] [Indexed: 12/17/2022]
Abstract
Long-term survival rates in childhood acute lymphoblastic leukemia (ALL) are currently above 85% due to huge improvements in treatment. However, 15-20% of children still experience relapses. Relapses can either occur in the bone marrow or at extramedullary sites, such as gonads or the central nervous system (CNS), formerly referred to as ALL-blast sanctuaries. The reason why ALL cells migrate to and stay in these sites is still unclear. In this review, we have attempted to assemble the evidence concerning the microenvironmental factors that could explain why ALL cells reside in such sites. We present criteria that make extramedullary leukemia niches and solid tumor metastatic niches comparable. Indeed, considering extramedullary leukemias as metastases could be a useful approach for proposing more effective treatments. In this context, we conclude with several examples of potential niche-based therapies which could be successfully added to current treatments of ALL.
Collapse
Affiliation(s)
- Jérémie Gaudichon
- CNRS, IGDR (Institut de Génétique et Développement de Rennes), Univ Rennes, UMR 6290, Rennes F-35000, France; Pediatric Hematology and Oncology Department, University Hospital, Caen, France.
| | - Hélène Jakobczyk
- CNRS, IGDR (Institut de Génétique et Développement de Rennes), Univ Rennes, UMR 6290, Rennes F-35000, France
| | - Lydie Debaize
- CNRS, IGDR (Institut de Génétique et Développement de Rennes), Univ Rennes, UMR 6290, Rennes F-35000, France
| | - Elie Cousin
- CNRS, IGDR (Institut de Génétique et Développement de Rennes), Univ Rennes, UMR 6290, Rennes F-35000, France; Pediatric Hematology Department, University Hospital, Rennes, France
| | - Marie-Dominique Galibert
- CNRS, IGDR (Institut de Génétique et Développement de Rennes), Univ Rennes, UMR 6290, Rennes F-35000, France.
| | - Marie-Bérengère Troadec
- CNRS, IGDR (Institut de Génétique et Développement de Rennes), Univ Rennes, UMR 6290, Rennes F-35000, France
| | - Virginie Gandemer
- CNRS, IGDR (Institut de Génétique et Développement de Rennes), Univ Rennes, UMR 6290, Rennes F-35000, France; Pediatric Hematology Department, University Hospital, Rennes, France.
| |
Collapse
|
65
|
Molecular characterization of manganese superoxide dismutase (MnSOD) from sterlet Acipenser ruthenus and its responses to Aeromonas hydrophila challenge and hypoxia stress. Comp Biochem Physiol A Mol Integr Physiol 2019; 234:68-76. [PMID: 30999108 DOI: 10.1016/j.cbpa.2019.04.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 02/26/2019] [Accepted: 04/11/2019] [Indexed: 01/14/2023]
Abstract
A novel gene encoding the mitochondrial manganese superoxide dismutase from sterlet Acipenser ruthenus (Ar-MnSOD) was cloned. The full-length cDNA of MnSOD was of 1040 bp with a 672 bp open reading frame encoding 224 amino acids and the deduced amino acid sequence was located in mitochondria. Sequence comparison analysis showed that Ar-MnSOD was highly similar to MnSODs of invertebrates and vertebrates, especially those of freshwater Cyprinidae fishes and mammals. Phylogenetic analysis revealed that Ar-MnSOD was distant from MnSODs of other fishes and belonged to the family of mitochondrial MnSODs (mMnSOD). Consistently, Ar-MnSOD was located in mitochondria. The 3D structure of Ar-MnSOD was predicted and the overall structure was similar to that of MnSODs of humans and the bay scallop Argopecten irradians. In addition, mRNA of Ar-MnSOD was detected to extensively express in all tissues, with the highest level in brain and liver. Spleen and head kidney inoculation of Aeromonas hydrophila led to a significant up-regulation of Ar-MnSOD transcript levels. Also, hypoxia induced a transient increase in transcription of Ar-MnSOD in the gills, but not in the heart and brain, suggesting metabolic depression in these vital organs. The results also implied the anti-hypoxia properties of Ar-MnSOD in the related tissues and proved that Ar-MnSOD was involved in the stress response and (anti) oxidative processes triggered by hypoxia. The results indicated that Ar-MnSOD is induced upon A. hydrophila infection and hypoxia, consistent with its role in host immune and stress-induced anti-oxidative responses.
Collapse
|
66
|
Tissue "Hypoxia" and the Maintenance of Leukemia Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1143:129-145. [PMID: 31338818 DOI: 10.1007/978-981-13-7342-8_6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The relationship of the homing of normal hematopoietic stem cells (HSC) in the bone marrow to specific environmental conditions, referred to as the stem cell niche (SCN), has been intensively studied over the last three decades. These conditions include the action of a number of molecular and cellular players, as well as critical levels of nutrients, oxygen and glucose in particular, involved in energy production. These factors are likely to act also in leukemias, due to the strict analogy between the hierarchical structure of normal hematopoietic cell populations and that of leukemia cell populations. This led to propose that leukemic growth is fostered by cells endowed with stem cell properties, the leukemia stem cells (LSC), a concept readily extended to comprise the cancer stem cells (CSC) of solid tumors. Two alternative routes have been proposed for CSC generation, that is, the oncogenic staminalization (acquisition of self-renewal) of a normal progenitor cell (the "CSC in normal progenitor cell" model) and the oncogenic transformation of a normal (self-renewing) stem cell (the "CSC in normal stem cell" model). The latter mechanism, in the hematological context, makes LSC derive from HSC, suggesting that LSC share SCN homing with HSC. This chapter is focused on the availability of oxygen and glucose in the regulation of LSC maintenance within the SCN. In this respect, the most critical aspect in view of the outcome of therapy is the long-term maintenance of the LSC subset capable to sustain minimal residual disease and the related risk of relapse of disease.
Collapse
|
67
|
Chalmin F, Bruchard M, Vegran F, Ghiringhelli F. Regulation of T cell antitumor immune response by tumor induced metabolic stress. Cell Stress 2018; 3:9-18. [PMID: 31225495 PMCID: PMC6551678 DOI: 10.15698/cst2019.01.171] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Adaptive T cell immune response is essential for tumor growth control. The efficacy of immune checkpoint inhibitors is regulated by intratumoral immune response. The tumor microenvironment has a major role in adaptive immune response tuning. Tumor cells generate a particular metabolic environment in comparison to other tissues. Tumors are characterized by glycolysis, hypoxia, acidosis, amino acid depletion and fatty acid metabolism modification. Such metabolic changes promote tumor growth, impair immune response and lead to resistance to therapies. This review will detail how these modifications strongly affect CD8 and CD4 T cell functions and impact immunotherapy efficacy.
Collapse
Affiliation(s)
- Fanny Chalmin
- Cancer Biology Research Platform, Centre Georges-François Leclerc, Dijon, France.,Université de Bourgogne-Franche Comté.,GIMI Genetic and Immunology Medical Institute, Dijon, France.,INSERM UMR1231, Dijon, France
| | - Mélanie Bruchard
- Cancer Biology Research Platform, Centre Georges-François Leclerc, Dijon, France.,Université de Bourgogne-Franche Comté.,GIMI Genetic and Immunology Medical Institute, Dijon, France.,INSERM UMR1231, Dijon, France
| | - Frederique Vegran
- Cancer Biology Research Platform, Centre Georges-François Leclerc, Dijon, France.,Université de Bourgogne-Franche Comté.,GIMI Genetic and Immunology Medical Institute, Dijon, France.,INSERM UMR1231, Dijon, France
| | - Francois Ghiringhelli
- Cancer Biology Research Platform, Centre Georges-François Leclerc, Dijon, France.,Université de Bourgogne-Franche Comté.,GIMI Genetic and Immunology Medical Institute, Dijon, France.,INSERM UMR1231, Dijon, France
| |
Collapse
|
68
|
Fernandes C, Suares D, Yergeri MC. Tumor Microenvironment Targeted Nanotherapy. Front Pharmacol 2018; 9:1230. [PMID: 30429787 PMCID: PMC6220447 DOI: 10.3389/fphar.2018.01230] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 10/08/2018] [Indexed: 12/12/2022] Open
Abstract
Recent developments in nanotechnology have brought new approaches to cancer diagnosis and therapy. While enhanced permeability and retention effect promotes nano-chemotherapeutics extravasation, the abnormal tumor vasculature, high interstitial pressure and dense stroma structure limit homogeneous intratumoral distribution of nano-chemotherapeutics and compromise their imaging and therapeutic effect. Moreover, heterogeneous distribution of nano-chemotherapeutics in non-tumor-stroma cells damages the non-tumor cells, and interferes with tumor-stroma crosstalk. This can lead not only to inhibition of tumor progression, but can also paradoxically induce acquired resistance and facilitate tumor cell proliferation and metastasis. Overall, the tumor microenvironment plays a vital role in regulating nano-chemotherapeutics distribution and their biological effects. In this review, the barriers in tumor microenvironment, its consequential effects on nano-chemotherapeutics, considerations to improve nano-chemotherapeutics delivery and combinatory strategies to overcome acquired resistance induced by tumor microenvironment have been summarized. The various strategies viz., nanotechnology based approach as well as ligand-mediated, redox-responsive, and enzyme-mediated based combinatorial nanoapproaches have been discussed in this review.
Collapse
Affiliation(s)
| | | | - Mayur C Yergeri
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM's Narsee Monjee Institute of Management Studies - NMIMS, Mumbai, India
| |
Collapse
|
69
|
Banerjee A, Lindenmair A, Steinborn R, Dumitrescu SD, Hennerbichler S, Kozlov AV, Redl H, Wolbank S, Weidinger A. Oxygen Tension Strongly Influences Metabolic Parameters and the Release of Interleukin-6 of Human Amniotic Mesenchymal Stromal Cells In Vitro. Stem Cells Int 2018; 2018:9502451. [PMID: 30510589 PMCID: PMC6230389 DOI: 10.1155/2018/9502451] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 05/18/2018] [Accepted: 09/06/2018] [Indexed: 12/11/2022] Open
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.
Collapse
Affiliation(s)
- Asmita Banerjee
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Donaueschingenstraße 13, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Andrea Lindenmair
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Garnisonstraße 21, 4020 Linz, Austria
| | - Ralf Steinborn
- Genomics Core Facility, VetCore, University of Veterinary Medicine, Veterinaerplatz 1, 1210 Vienna, Austria
| | - Sergiu Dan Dumitrescu
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Donaueschingenstraße 13, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Simone Hennerbichler
- Red Cross Blood Transfusion Service for Upper Austria, Krankenhausstraße 7, 4017 Linz, Austria
| | - Andrey V. Kozlov
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Donaueschingenstraße 13, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Heinz Redl
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Donaueschingenstraße 13, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Susanne Wolbank
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Donaueschingenstraße 13, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Adelheid Weidinger
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Donaueschingenstraße 13, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
70
|
Al-Ani A, Toms D, Kondro D, Thundathil J, Yu Y, Ungrin M. Oxygenation in cell culture: Critical parameters for reproducibility are routinely not reported. PLoS One 2018; 13:e0204269. [PMID: 30325922 PMCID: PMC6191109 DOI: 10.1371/journal.pone.0204269] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 09/04/2018] [Indexed: 01/01/2023] Open
Abstract
Mammalian cell culture is foundational to biomedical research, and the reproducibility of research findings across the sciences is drawing increasing attention. While many components contribute to reproducibility, the reporting of factors that impact oxygen delivery in the general biomedical literature has the potential for both significant impact, and immediate improvement. The relationship between the oxygen consumption rate of cells and the diffusive delivery of oxygen through the overlying medium layer means parameters such as medium depth and cell type can cause significant differences in oxygenation for cultures nominally maintained under the same conditions. While oxygenation levels are widely understood to significantly impact the phenotype of cultured cells in the abstract, in practise the importance of the above parameters does not appear to be well recognized in the non-specialist research community. On analyzing two hundred articles from high-impact journals we find a large majority missing at least one key piece of information necessary to ensure consistency in replication. We propose that explicitly reporting these values should be a requirement for publication.
Collapse
Affiliation(s)
- Abdullah Al-Ani
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Derek Toms
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Douglas Kondro
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Jarin Thundathil
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Yang Yu
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Mark Ungrin
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB, Canada
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
- * E-mail:
| |
Collapse
|
71
|
Bahsoun S, Coopman K, Forsyth NR, Akam EC. The Role of Dissolved Oxygen Levels on Human Mesenchymal Stem Cell Culture Success, Regulatory Compliance, and Therapeutic Potential. Stem Cells Dev 2018; 27:1303-1321. [DOI: 10.1089/scd.2017.0291] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Soukaina Bahsoun
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, United Kingdom
| | - Karen Coopman
- Centre for Biological Engineering, Loughborough University, Loughborough, United Kingdom
| | - Nicholas R. Forsyth
- Guy Hilton Research Centre, Institute for Science and Technology in Medicine, Keele University, Keele, United Kingdom
| | - Elizabeth C. Akam
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, United Kingdom
| |
Collapse
|
72
|
Rivera KR, Pozdin VA, Young AT, Erb PD, Wisniewski NA, Magness ST, Daniele M. Integrated phosphorescence-based photonic biosensor (iPOB) for monitoring oxygen levels in 3D cell culture systems. Biosens Bioelectron 2018; 123:131-140. [PMID: 30060990 DOI: 10.1016/j.bios.2018.07.035] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 07/13/2018] [Accepted: 07/16/2018] [Indexed: 12/16/2022]
Abstract
Physiological processes, such as respiration, circulation, digestion, and many pathologies alter oxygen concentration in the blood and tissue. When designing culture systems to recapitulate the in vivo oxygen environment, it is important to integrate systems for monitoring and controlling oxygen concentration. Herein, we report the design and engineering of a system to remotely monitor and control oxygen concentration inside a device for 3D cell culture. We integrate a photonic oxygen biosensor into the 3D tissue scaffold and regulate oxygen concentration via the control of purging gas flow. The integrated phosphorescence-based oxygen biosensor employs the quenching of palladium-benzoporphyrin by molecular oxygen to transduce the local oxygen concentration in the 3D tissue scaffold. The system is validated by testing the effects of normoxic and hypoxic culture conditions on healthy and tumorigenic breast epithelial cells, MCF-10A cells and BT474 cells, respectively. Under hypoxic conditions, both cell types exhibited upregulation of downstream target genes for the hypoxia marker gene, hypoxia-inducible factor 1α (HIF1A). Lastly, by monitoring the real-time fluctuation of oxygen concentration, we illustrated the formation of hypoxic culture conditions due to limited diffusion of oxygen through 3D tissue scaffolds.
Collapse
Affiliation(s)
- Kristina R Rivera
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, 911 Oval Dr., Raleigh, NC 27695, USA
| | - Vladimir A Pozdin
- Department of Electrical & Computer Engineering, North Carolina State University, 890 Oval Dr., Raleigh, NC 27695, USA
| | - Ashlyn T Young
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, 911 Oval Dr., Raleigh, NC 27695, USA
| | - Patrick D Erb
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, 911 Oval Dr., Raleigh, NC 27695, USA
| | | | - Scott T Magness
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, 911 Oval Dr., Raleigh, NC 27695, USA; Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Michael Daniele
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, 911 Oval Dr., Raleigh, NC 27695, USA; Department of Electrical & Computer Engineering, North Carolina State University, 890 Oval Dr., Raleigh, NC 27695, USA.
| |
Collapse
|
73
|
Gdynia G, Robak T, Kopitz J, Heller A, Grekova S, Duglova K, Laukemper G, Heinzel-Gutenbrunner M, Gutenbrunner C, Roth W, Ho AD, Schirmacher P, Schmitt M, Dreger P, Sellner L. Distinct Activities of Glycolytic Enzymes Identify Chronic Lymphocytic Leukemia Patients with a more Aggressive Course and Resistance to Chemo-Immunotherapy. EBioMedicine 2018; 32:125-133. [PMID: 29884457 PMCID: PMC6021262 DOI: 10.1016/j.ebiom.2018.05.030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 05/24/2018] [Accepted: 05/24/2018] [Indexed: 12/12/2022] Open
Abstract
A higher capacity to grow under hypoxic conditions can lead to a more aggressive behavior of tumor cells. Determining tumor activity under hypoxia may identify chronic lymphocytic leukemia (CLL) with aggressive clinical course and predict response to chemo-immunotherapy (CIT). A metabolic score was generated by determining pyruvate kinase and lactate dehydrogenase, key enzymes of glycolysis, ex vivo in primary CLL samples under normoxic and hypoxic conditions. This score was further correlated with clinical endpoints and response to CIT in 96 CLL patients. 45 patients were classified as metabolic high risk (HR), 51 as low risk (LR). Treatment-free survival (TFS) was significantly shorter in HR patients (median 394 vs 723 days, p = .021). 15 HR patients and 14 LR patients received CIT after sample acquisition. HR patients had a significantly shorter progression-free survival after treatment compared to LR patients (median 216 days vs not reached, p = .008). Multivariate analysis evaluating age, IGHV, TP53 deletion or mutation and 11q22–23 deletion besides the capacity of tumor cells to grow under severe hypoxic conditions identified the metabolic profile as the strongest independent risk factor for shorter TFS (hazard ratio 2.37, p = .011). The metabolic risk can provide prognostic and predictive information complementary to genetic biomarkers and identify patients who might benefit from alternative treatment approaches. The activity of distinct glycolytic enzymes can identify CLL patients with resistance to chemo-immunotherapy The activity of distinct glycolytic enzymes can identify CLL patients who may benefit from specific pathway inhibitors We provide a tool for the evaluation of specific glycolytic enzymes in primary CLL cells for clinical diagnostics
Collapse
Affiliation(s)
- Georg Gdynia
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Tadeusz Robak
- Medical University of Lodz, Copernicus Memorial Hospital, Lodz, Poland
| | - Jürgen Kopitz
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Anette Heller
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Svetlana Grekova
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Katarina Duglova
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Gloria Laukemper
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | | | | | - Wilfried Roth
- Institute of Pathology, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Anthony D Ho
- Department of Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Peter Schirmacher
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Michael Schmitt
- Department of Medicine V, University Hospital Heidelberg, Heidelberg, Germany; National Center for Tumor Diseases (NCT), German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Peter Dreger
- Department of Medicine V, University Hospital Heidelberg, Heidelberg, Germany; National Center for Tumor Diseases (NCT), German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Leopold Sellner
- Department of Medicine V, University Hospital Heidelberg, Heidelberg, Germany; National Center for Tumor Diseases (NCT), German Cancer Consortium (DKTK), Heidelberg, Germany.
| |
Collapse
|
74
|
Barmaki S, Jokinen V, Obermaier D, Blokhina D, Korhonen M, Ras RHA, Vuola J, Franssila S, Kankuri E. A microfluidic oxygen sink to create a targeted cellular hypoxic microenvironment under ambient atmospheric conditions. Acta Biomater 2018; 73:167-179. [PMID: 29649636 DOI: 10.1016/j.actbio.2018.04.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 03/28/2018] [Accepted: 04/03/2018] [Indexed: 12/12/2022]
Abstract
Physiological oxygen levels within the tissue microenvironment are usually lower than 14%, in stem cell niches these levels can be as low as 0-1%. In cell cultures, such low oxygen levels are usually mimicked by altering the global culture environment either by O2 removal (vacuum or oxygen absorption) or by N2 supplementation for O2 replacement. To generate a targeted cellular hypoxic microenvironment under ambient atmospheric conditions, we characterised the ability of the dissolved oxygen-depleting sodium sulfite to generate an in-liquid oxygen sink. We utilised a microfluidic design to place the cultured cells in the vertical oxygen gradient and to physically separate the cells from the liquid. We demonstrate generation of a chemical in-liquid oxygen sink that modifies the surrounding O2 concentrations. O2 level control in the sink-generated hypoxia gradient is achievable by varying the thickness of the polydimethylsiloxane membrane. We show that intracellular hypoxia and hypoxia response element-dependent signalling is instigated in cells exposed to the microfluidic in-liquid O2 sink-generated hypoxia gradient. Moreover, we show that microfluidic flow controls site-specific microenvironmental kinetics of the chemical O2 sink reaction, which enables generation of intermittent hypoxia/re-oxygenation cycles. The microfluidic O2 sink chip targets hypoxia to the cell culture microenvironment exposed to the microfluidic channel architecture solely by depleting O2 while other sites in the same culture well remain unaffected. Thus, responses of both hypoxic and bystander cells can be characterised. Moreover, control of microfluidic flow enables generation of intermittent hypoxia or hypoxia/re-oxygenation cycles. STATEMENT OF SIGNIFICANCE Specific manipulation of oxygen concentrations in cultured cells' microenvironment is important when mimicking low-oxygen tissue conditions and pathologies such as tissue infarction or cancer. We utilised a sodium sulfite-based in-liquid chemical reaction to consume dissolved oxygen. When this liquid was pumped into a microfluidic channel, lowered oxygen levels could be measured outside the channel through a polydimethylsiloxane PDMS membrane allowing only for gaseous exchange. We then utilised this setup to deplete oxygen from the microenvironment of cultured cells, and showed that cells responded to hypoxia on molecular level. Our setup can be used for specifically removing oxygen from the cell culture microenvironment for experimental purposes and for generating a low oxygen environment that better mimics the cells' original tissue environments.
Collapse
Affiliation(s)
- Samineh Barmaki
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Ville Jokinen
- Aalto University, School of Chemical Engineering, Department of Chemistry and Materials Science, Espoo, Finland
| | | | - Daria Blokhina
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Matti Korhonen
- Advanced Cell Therapy Centre, Finnish Red Cross Blood Service, Helsinki, Finland
| | - Robin H A Ras
- Aalto University, School of Chemical Engineering, Department of Chemistry and Materials Science, Espoo, Finland; Aalto University, School of Science, Department of Applied Physics, Espoo, Finland
| | - Jyrki Vuola
- Helsinki Burn Centre, Jorvi Hospital, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Sami Franssila
- Aalto University, School of Chemical Engineering, Department of Chemistry and Materials Science, Espoo, Finland
| | - Esko Kankuri
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
75
|
Chen C, Tang Q, Zhang Y, Yu M, Jing W, Tian W. Physioxia: a more effective approach for culturing human adipose-derived stem cells for cell transplantation. Stem Cell Res Ther 2018; 9:148. [PMID: 29793517 PMCID: PMC5968705 DOI: 10.1186/s13287-018-0891-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 04/18/2018] [Accepted: 05/01/2018] [Indexed: 02/05/2023] Open
Abstract
Background Although typically cultured at an atmospheric oxygen concentration (20–21%), adipose-derived stem cells (ASCs) reside under considerable low oxygen tension (physioxia) in vivo. In the present study, we explored whether and how physioxia could be a more effective strategy for culturing ASCs for transplantation. Methods After isolation, human ASCs were cultured under physioxia (2% O2) and hyperoxia (20% O2) until assayed. WST-8, Transwell, tube formation, β-galactosidase staining, and annexin V-FITC/PI assays were used to evaluate cell proliferation, migration, angiogenesis, senescence, and apoptosis, respectively. Survivability was determined by an ischemia model in vitro and nude mouse model in vivo, and the underlying metabolic alterations were investigated by fluorescence staining, flow cytometry, and real-time polymerase chain reaction. Results Compared with those in the hyperoxia group, cells in the physioxia group exhibited increased proliferation, migration, and angiogenesis, and decreased senescence and apoptosis. The increased survival rate of ASCs cultured in physioxia was found both in ischemia model in vitro and in vivo. The underlying metabolic reprogramming was also monitored and showed decreased mitochondrial mass, alkalized intracellular pH, and increased glucose uptake and glycogen synthesis. Conclusions These results suggest that physioxia is a more effective environment in which to culture ASCs for transplantation owing to the maintenance of native bioactivities without injury by hyperoxia.
Collapse
Affiliation(s)
- Chang Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China.,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China.,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China
| | - Qi Tang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China.,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China.,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China
| | - Yan Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China.,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China.,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China
| | - Mei Yu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China.,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China
| | - Wei Jing
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China. .,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China. .,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China.
| | - Weidong Tian
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China. .,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China. .,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China.
| |
Collapse
|
76
|
Shin HS, Lee S, Kim YM, Lim JY. Hypoxia-Activated Adipose Mesenchymal Stem Cells Prevents Irradiation-Induced Salivary Hypofunction by Enhanced Paracrine Effect Through Fibroblast Growth Factor 10. Stem Cells 2018; 36:1020-1032. [PMID: 29569790 DOI: 10.1002/stem.2818] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 02/12/2018] [Accepted: 02/17/2018] [Indexed: 12/13/2022]
Abstract
To explore the effects and mechanisms of paracrine factors secreted from human adipose mesenchymal stem cell (hAdMSCs) that are activated by hypoxia on radioprotection against irradiation-induced salivary hypofunction in subjects undergoing radiotherapy for head and neck cancers. An organotypic spheroid coculture model to mimic irradiation (IR)-induced salivary hypofunction was set up for in vitro experiments. Human parotid gland epithelial cells were organized to form three-dimensional (3D) acinus-like spheroids on growth factor reduced -Matrigel. Cellular, structural, and functional damage following IR were examined after cells were cocultured with hAdMSCs preconditioned with either normoxia (hAdMSCNMX ) or hypoxia (hAdMSCHPX ). A key paracrine factor secreted by hAdMSCsHPX was identified by high-throughput microarray-based enzyme-linked immunosorbent assay. Molecular mechanisms and signaling pathways on radioprotection were explored. Therapeutic effects of hAdMSCsHPX were evaluated after in vivo transplant into mice with IR-induced salivary hypofunction. In our 3D coculture experiment, hAdMSCsHPX significantly enhanced radioresistance of spheroidal human parotid epithelial cells, and led to greater preservation of salivary epithelial integrity and acinar secretory function relative to hAdMSCsNMX . Coculture with hAdMSCsHPX promoted FGFR expression and suppressed FGFR diminished antiapoptotic activity of hAdMSCsHPX . Among FGFR-binding secreted factors, we found that fibroblast growth factor 10 (FGF10) contributed to therapeutic effects of hAdMSCsHPX by enhancing antiapoptotic effect, which was dependent on FGFR-PI3K signaling. An in vivo transplant of hAdMSCsHPX into irradiated salivary glands of mice reversed IR-induced salivary hypofunction where hAdMSC-released FGF10 contributed to tissue remodeling. Our results suggest that hAdMSCsHPX protect salivary glands from IR-induced apoptosis and preserve acinar structure and functions by activation of FGFR-PI3K signaling via actions of hAdMSC-secreted factors, including FGF10. Stem Cells 2018;36:1020-1032.
Collapse
Affiliation(s)
- Hyun-Soo Shin
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Songyi Lee
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young-Mo Kim
- Department of Otorhinolaryngology, Inha University College of Medicine, Incheon, Republic of Korea
| | - Jae-Yol Lim
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
77
|
Irigoyen M, García-Ruiz JC, Berra E. The hypoxia signalling pathway in haematological malignancies. Oncotarget 2018; 8:36832-36844. [PMID: 28415662 PMCID: PMC5482702 DOI: 10.18632/oncotarget.15981] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 02/27/2017] [Indexed: 12/25/2022] Open
Abstract
Haematological malignancies are tumours that affect the haematopoietic and the lymphatic systems. Despite the huge efforts to eradicate these tumours, the percentage of patients suffering resistance to therapies and relapse still remains significant. The tumour environment favours drug resistance of cancer cells, and particularly of cancer stem/initiating cells. Hypoxia promotes aggressiveness, metastatic spread and relapse in most of the solid tumours. Furthermore, hypoxia is associated with worse prognosis and resistance to conventional treatments through activation of the hypoxia-inducible factors. Haematological malignancies are not considered solid tumours, and therefore, the role of hypoxia in these diseases was initially presumed to be inconsequential. However, hypoxia is a hallmark of the haematopoietic niche. Here, we will review the current understanding of the role of both hypoxia and hypoxia-inducible factors in different haematological tumours.
Collapse
Affiliation(s)
- Marta Irigoyen
- Centro de Investigación Cooperativa en Biociencias CIC bioGUNE, Derio, Spain
| | - Juan Carlos García-Ruiz
- Servicio de Hematología y Hemoterapia, BioCruces Health Research Institute, Hospital Universitario Cruces, Spain
| | - Edurne Berra
- Centro de Investigación Cooperativa en Biociencias CIC bioGUNE, Derio, Spain
| |
Collapse
|
78
|
Shan J, Ramachandran A, Thanki AM, Vukusic FBI, Barylski J, Clokie MRJ. Bacteriophages are more virulent to bacteria with human cells than they are in bacterial culture; insights from HT-29 cells. Sci Rep 2018; 8:5091. [PMID: 29572482 PMCID: PMC5865146 DOI: 10.1038/s41598-018-23418-y] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 03/13/2018] [Indexed: 02/07/2023] Open
Abstract
Bacteriophage therapeutic development will clearly benefit from understanding the fundamental dynamics of in vivo phage-bacteria interactions. Such information can inform animal and human trials, and much can be ascertained from human cell-line work. We have developed a human cell-based system using Clostridium difficile, a pernicious hospital pathogen with limited treatment options, and the phage phiCDHS1 that effectively kills this bacterium in liquid culture. The human colon tumorigenic cell line HT-29 was used because it simulates the colon environment where C. difficile infection occurs. Studies on the dynamics of phage-bacteria interactions revealed novel facets of phage biology, showing that phage can reduce C. difficile numbers more effectively in the presence of HT-29 cells than in vitro. Both planktonic and adhered Clostridial cell numbers were successfully reduced. We hypothesise and demonstrate that this observation is due to strong phage adsorption to the HT-29 cells, which likely promotes phage-bacteria interactions. The data also showed that the phage phiCDHS1 was not toxic to HT-29 cells, and phage-mediated bacterial lysis did not cause toxin release and cytotoxic effects. The use of human cell lines to understand phage-bacterial dynamics offers valuable insights into phage biology in vivo, and can provide informative data for human trials.
Collapse
Affiliation(s)
- Jinyu Shan
- Department of Infection, Immunity, and Inflammation, University of Leicester, Leicester, LE1 9HN, UK.
| | - Ananthi Ramachandran
- Department of Infection, Immunity, and Inflammation, University of Leicester, Leicester, LE1 9HN, UK
| | - Anisha M Thanki
- Department of Infection, Immunity, and Inflammation, University of Leicester, Leicester, LE1 9HN, UK
| | - Fatima B I Vukusic
- Department of Infection, Immunity, and Inflammation, University of Leicester, Leicester, LE1 9HN, UK
| | - Jakub Barylski
- Department of Molecular Virology, Faculty of Biology, Adam Mickiewicz University, 61-614, Poznan, Poland
| | - Martha R J Clokie
- Department of Infection, Immunity, and Inflammation, University of Leicester, Leicester, LE1 9HN, UK.
| |
Collapse
|
79
|
Murugesan M, Premkumar K. Hypoxia stimulates microenvironment in human embryonic stem cell through inflammatory signalling: An integrative analysis. Biochem Biophys Res Commun 2018; 498:437-444. [PMID: 29501494 DOI: 10.1016/j.bbrc.2018.02.194] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Accepted: 02/27/2018] [Indexed: 02/08/2023]
Abstract
Despite, several lines of evidence suggesting the possible role of hypoxia in stem cell development and differentiation its significance in conferring the stemness and pluripotency remains elusive. In the present study we sought to delineate the candidate genes and molecular pathways imposed during hypoxic microenvironment and its physiological relevance in tipping the balance between the niche and cellular differentiation. Integrated meta-analysis was performed between the hypoxia exposed and normal human embryonic stem cells, employing three transcriptomic cohorts (GSE35819, GSE9510 and GSE37761) retrieved from Gene expression omnibus (GEO) database. Results reveal that a total number of 12 genes were consistently differentially expressed (6up regulated and 6 down regulated) with FDR <0.05 and fold change >1.5. The Gene Ontology (GO) functions and Kyoto encyclopedia of genes and genomes (KEGG) pathway analysis was performed using DAVID. The GO analysis showed DEG significantly enriched in terms of Cellular process (GO:0009987), protein binding (GO:0005515) and cell part (GO:0044464). KEGG analysis indicated participation of genes associated with circadian rthyum regulation and PPAR signalling pathway. Further, gene-set signature (MsigDB) enrichment analysis showed positive regulation with inflammatory signals and negative association with PPAR and p53 pathway. Protein-protein network of gene modules suggests significant hub proteins viz. CTTNB1 (Degree = 18), IL8 (Degree = 15), NFKB1 (Degree = 15) and RELA (Degree = 15) in the PPI network. MCODE algorithm was used for subnetworks of the PPI network. Our integrative analysis documents the potential candidate genes which serves distinct roles influencing metabolic shift and induce inflammatory effectors contributing to hypoxic mediated stem cell niche.
Collapse
Affiliation(s)
- Manikandan Murugesan
- Cancer Genetics and Nanomedicine Laboratory, Department of Biomedical Science, School of Basic Medical Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Kumpati Premkumar
- Cancer Genetics and Nanomedicine Laboratory, Department of Biomedical Science, School of Basic Medical Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India.
| |
Collapse
|
80
|
Bono S, Lulli M, D'Agostino VG, Di Gesualdo F, Loffredo R, Cipolleschi MG, Provenzani A, Rovida E, Dello Sbarba P. Different BCR/Abl protein suppression patterns as a converging trait of chronic myeloid leukemia cell adaptation to energy restriction. Oncotarget 2018; 7:84810-84825. [PMID: 27852045 PMCID: PMC5356700 DOI: 10.18632/oncotarget.13319] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 10/28/2016] [Indexed: 02/07/2023] Open
Abstract
BCR/Abl protein drives the onset and progression of Chronic Myeloid Leukemia (CML). We previously showed that BCR/Abl protein is suppressed in low oxygen, where viable cells retain stem cell potential. This study addressed the regulation of BCR/Abl protein expression under oxygen or glucose shortage, characteristic of the in vivo environment where cells resistant to tyrosine kinase inhibitors (TKi) persist. We investigated, at transcriptional, translational and post-translational level, the mechanisms involved in BCR/Abl suppression in K562 and KCL22 CML cells. BCR/abl mRNA steady-state analysis and ChIP-qPCR on BCR promoter revealed that BCR/abl transcriptional activity is reduced in K562 cells under oxygen shortage. The SUnSET assay showed an overall reduction of protein synthesis under oxygen/glucose shortage in both cell lines. However, only low oxygen decreased polysome-associated BCR/abl mRNA significantly in KCL22 cells, suggesting a decreased BCR/Abl translation. The proteasome inhibitor MG132 or the pan-caspase inhibitor z-VAD-fmk extended BCR/Abl expression under oxygen/glucose shortage in K562 cells. Glucose shortage induced autophagy-dependent BCR/Abl protein degradation in KCL22 cells. Overall, our results showed that energy restriction induces different cell-specific BCR/Abl protein suppression patterns, which represent a converging route to TKi-resistance of CML cells. Thus, the interference with BCR/Abl expression in environment-adapted CML cells may become a useful implement to current therapy.
Collapse
Affiliation(s)
- Silvia Bono
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Università degli Studi di Firenze, Florence, Italy
| | - Matteo Lulli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Università degli Studi di Firenze, Florence, Italy
| | | | - Federico Di Gesualdo
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Università degli Studi di Firenze, Florence, Italy
| | - Rosa Loffredo
- Centre For Integrative Biology (CIBIO), Università degli Studi di Trento, Trento, Italy
| | - Maria Grazia Cipolleschi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Università degli Studi di Firenze, Florence, Italy
| | - Alessandro Provenzani
- Centre For Integrative Biology (CIBIO), Università degli Studi di Trento, Trento, Italy
| | - Elisabetta Rovida
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Università degli Studi di Firenze, Florence, Italy
| | - Persio Dello Sbarba
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Università degli Studi di Firenze, Florence, Italy
| |
Collapse
|
81
|
Refined control of cell stemness allowed animal evolution in the oxic realm. Nat Ecol Evol 2018; 2:220-228. [DOI: 10.1038/s41559-017-0410-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 11/10/2017] [Indexed: 12/26/2022]
|
82
|
Evaluation of committed and primitive cord blood progenitors after expansion on adipose stromal cells. Cell Tissue Res 2018; 372:523-533. [DOI: 10.1007/s00441-017-2766-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 11/29/2017] [Indexed: 02/07/2023]
|
83
|
Abstract
The therapeutic efficacy of tissue-engineered constructs is often compromised by inadequate inosculation and neo-vascularization. This problem is considered one of the biggest hurdles in the field and finding a solution is currently the focus of a great fraction of the research community. Many of the methodologies designed to address this issue propose the use of endothelial cells and angiogenic growth factors, or combinations of both, to accelerate neo-vascularization after transplantation. However, an adequate solution is still elusive. In this context, we describe a methodology that combines the use of the stromal vascular fraction (SVF) isolated from adipose tissue with low oxygen culture to produce pre-vascularized cell sheets as angiogenic tools for Tissue Engineering. The herein proposed approach takes advantage of the SVF angiogenic nature conferred by adipose stem cells, endothelial progenitors, endothelial and hematopoietic cells, and pericytes and further potentiates it using low oxygen, or hypoxic, culture. Freshly isolated nucleated SVF cells are cultured in hyperconfluent conditions under hypoxia (pO2 = 5 %) for up to 5 days in medium without extrinsic growth factors enabling the generation of contiguous sheets as described by the cell sheet engineering technique. Flow cytometry and immunocytochemistry allow confirming the phenotype of the different cell types composing the cell-sheets as well the organization of the CD31(+) cells in branched and highly complex tube-like structures. Overall, a simple and flexible approach to promote growth factor-free pre-vascularization of cell sheets for tissue engineering (TE) applications is described.
Collapse
|
84
|
Buzanska L, Zychowicz M, Kinsner-Ovaskainen A. Bioengineering of the Human Neural Stem Cell Niche: A Regulatory Environment for Cell Fate and Potential Target for Neurotoxicity. Results Probl Cell Differ 2018; 66:207-230. [PMID: 30209661 DOI: 10.1007/978-3-319-93485-3_9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Human neural stem/progenitor cells of the developing and adult organisms are surrounded by the microenvironment, so-called neurogenic niche. The developmental processes of stem cells, such as survival, proliferation, differentiation, and fate decisions, are controlled by the mutual interactions between cells and the niche components. Such interactions are tissue specific and determined by the biochemical and biophysical properties of the niche constituencies and the presence of other cell types. This dynamic approach of the stem cell niche, when translated into in vitro settings, requires building up "biomimetic" microenvironments resembling natural conditions, where the stem/progenitor cell is provided with diverse extracellular signals exerted by soluble and structural cues, mimicking those found in vivo. The neural stem cell niche is characterized by a unique composition of soluble components including neurotransmitters and trophic factors as well as insoluble extracellular matrix proteins and proteoglycans. Biotechnological innovations provide tools such as a new generation of tunable biomaterials capable of releasing specific signals in a spatially and temporally controlled manner, thus creating in vitro nature-like conditions and, when combined with stem cell-derived tissue specific progenitors, producing differentiated neuronal tissue structures. In addition, substantial progress has been made on the protocols to obtain stem cell-derived cell aggregates such as neurospheres and self-assembled organoids.In this chapter, we have assessed the application of bioengineered human neural stem cell microenvironments to produce in vitro models of different levels of biological complexity for the efficient control of stem cell fate. Examples of biomaterial-supported two-dimensional and three-dimensional (2D and 3D) complex culture systems that provide artificial neural stem cell niches are discussed in the context of their application for basic research and neurotoxicity testing.
Collapse
Affiliation(s)
- Leonora Buzanska
- Stem Cell Bioengineering Unit, Mossakowski Medical Research Centre Polish Academy of Sciences, Warsaw, Poland.
| | - Marzena Zychowicz
- Stem Cell Bioengineering Unit, Mossakowski Medical Research Centre Polish Academy of Sciences, Warsaw, Poland
| | - Agnieszka Kinsner-Ovaskainen
- European Commission, Joint Research Centre, Directorate for Health Consumers and Reference Materials, Ispra, Italy
| |
Collapse
|
85
|
Buizer AT, Bulstra SK, Veldhuizen AG, Kuijer R. The balance between proliferation and transcription of angiogenic factors of mesenchymal stem cells in hypoxia. Connect Tissue Res 2018; 59:12-20. [PMID: 28165799 DOI: 10.1080/03008207.2017.1289189] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Bridging large bone defects with mesenchymal stromal cells-seeded scaffolds remains a big challenge in orthopedic surgery, due to the lack of vascularization. Within such a cell-scaffold construct, cells are exposed to ischemic conditions. When human mesenchymal stem cells (hMSCs) encounter hypoxic conditions, they show higher cell proliferation than at ambient oxygen levels. However, when hMSCs are exposed to prolonged ischemia, cell proliferation ceases completely. Exposure of hMSCs to hypoxic conditions is known to result in the transcription of angiogenic factors (AGF), which can promote the development of new blood vessels. In this study, we investigated at which oxygen level hMSC proliferation and the transcription of AGF were optimal. Human bone marrow-derived hMSCs were cultured at 0.1, 1, 2, 3, 4, 5, and 21% oxygen. Cell proliferation over 14 days was assayed using a DNA quantification method. hMSC metabolic activity over 14 days was measured using a MTT test. Quantitative RT-PCR was used to assess mRNA levels of angiogenic factors at the tested oxygen percentages. hMSCs showed the highest cell proliferation rate at 1% oxygen. The highest corrected cell metabolic rate was found at 21% oxygen, followed by 2% oxygen. HIF1α transcription did not increase under hypoxic conditions compared to 21% oxygen conditions. However, transcription of VEGF and ANG-1 was significantly higher at 2% oxygen than at 21% O2. The optimum oxygen range at which hMSCs proliferated rapidly and angiogenic factors ANG-1 and VEGF simultaneously came to expression was from 1 to 2% oxygen.
Collapse
Affiliation(s)
- Arina T Buizer
- a Department of Orthopedic Surgery , University of Groningen, University Medical Center Groningen , Groningen , The Netherlands.,b Department of Biomedical Engineering , University of Groningen, University Medical Center Groningen , Groningen , The Netherlands
| | - Sjoerd K Bulstra
- a Department of Orthopedic Surgery , University of Groningen, University Medical Center Groningen , Groningen , The Netherlands
| | - Albert G Veldhuizen
- a Department of Orthopedic Surgery , University of Groningen, University Medical Center Groningen , Groningen , The Netherlands
| | - Roel Kuijer
- b Department of Biomedical Engineering , University of Groningen, University Medical Center Groningen , Groningen , The Netherlands
| |
Collapse
|
86
|
Plotkin BJ, Davis JW, Strizzi L, Lee P, Christoffersen-Cebi J, Kacmar J, Rivero OJ, Elsayed N, Zanghi N, Ito B, Sigar IM. A method for the long-term cultivation of mammalian cells in the absence of oxygen: Characterization of cell replication, hypoxia-inducible factor expression and reactive oxygen species production. Tissue Cell 2017; 50:59-68. [PMID: 29429519 DOI: 10.1016/j.tice.2017.12.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 11/29/2017] [Accepted: 12/09/2017] [Indexed: 10/18/2022]
Abstract
The center of tumors, stem cell niches and mucosal surfaces all represent areas of the body that are reported to be anoxic. However, long-term study of anoxic cell physiology is hindered by the lack of a sustainable method permitting cell cultivation in the complete absence of oxygen. A novel methodology was developed that enabled anoxic cell cultivation (17d maximum time tested) and cell passage. In the absence of oxygen, cell morphology is significantly altered. All cells tested exhibited morphologic changes, i.e., a combination of tethered (monolayer-like) and runagate (suspension-like) morphologies. Both morphologies replicated (Vero and HeLa cells tested) and could be passaged anaerobically. In the absence of exogenous oxygen, anoxic cells produced reactive oxygen species (ROS). Anaerobic runagate HeLa and Vero cells increased ROS production from day 3 to day 10 by 2- and 3-fold, respectively. In contrast, anoxic tethered HeLa and Vero cells either showed no significant change in ROS production between days 3 and 10 or exhibited a 3-fold decrease in ROS, respectively. Detection of ROS was inversely related to detection of hypoxia-inducible factor-1α (HIF1) mRNA and HIF-1 protein expression which cycled over a 10-day period. This methodology has broad applications for the study of tumor and stem cell physiology as well as gastrointestinal cell-microbiome interactions. In addition, sustainable anaerobic cell culture may lead to the identification of novel pathways and targets for chemotherapeutic drug development.
Collapse
Affiliation(s)
- Balbina J Plotkin
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL, 60515, USA.
| | - James W Davis
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL, 60515, USA
| | - Luigi Strizzi
- Department of Pathology, Midwestern University, Downers Grove, IL 60515, USA
| | - Peter Lee
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL, 60515, USA
| | | | - Joan Kacmar
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL, 60515, USA
| | - Orlando J Rivero
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL, 60515, USA
| | - Norhan Elsayed
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL, 60515, USA
| | - Nicholas Zanghi
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL, 60515, USA
| | - Brent Ito
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL, 60515, USA
| | - Ira M Sigar
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL, 60515, USA
| |
Collapse
|
87
|
Tumor Tissue Analogs for the Assessment of Radioresistance in Cancer Stem Cells. Methods Mol Biol 2017. [PMID: 28986892 DOI: 10.1007/978-1-4939-7401-6_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Over the years, radiotherapy-related research has been based on local tumor control as an experimental endpoint, yielding a wealth of data demonstrating the importance of cancer stem cells in tumor reoccurrence after radiotherapy. Literature is replete with experimental and clinical evidence that the cancer stem cell population in a tumor affects its radiocurability. An important consideration for radiotherapy is the microenvironmental stimuli in the CSC niche that results from factors such as hypoxia, extracellular matrix (ECM) elements and their intercellular interaction with non-stem cells and other cell types that prevail in the tumor milieu. In this chapter, we have described the methodology to develop in vitro 3D tumor models that incorporate these microenvironmental characteristics and design experiments that generate endpoints for understanding radioresistance in cancer stem cells.
Collapse
|
88
|
Kulkarni A, Mateus M, Thinnes CC, McCullagh JS, Schofield CJ, Taylor GP, Bangham CRM. Glucose Metabolism and Oxygen Availability Govern Reactivation of the Latent Human Retrovirus HTLV-1. Cell Chem Biol 2017; 24:1377-1387.e3. [PMID: 28965728 PMCID: PMC5696563 DOI: 10.1016/j.chembiol.2017.08.016] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 07/24/2017] [Accepted: 08/17/2017] [Indexed: 12/30/2022]
Abstract
The human retrovirus HTLV-1 causes a hematological malignancy or neuroinflammatory disease in ∼10% of infected individuals. HTLV-1 primarily infects CD4+ T lymphocytes and persists as a provirus integrated in their genome. HTLV-1 appears transcriptionally latent in freshly isolated cells; however, the chronically active anti-HTLV-1 cytotoxic T cell response observed in infected individuals indicates frequent proviral expression in vivo. The kinetics and regulation of HTLV-1 proviral expression in vivo are poorly understood. By using hypoxia, small-molecule hypoxia mimics, and inhibitors of specific metabolic pathways, we show that physiologically relevant levels of hypoxia, as routinely encountered by circulating T cells in the lymphoid organs and bone marrow, significantly enhance HTLV-1 reactivation from latency. Furthermore, culturing naturally infected CD4+ T cells in glucose-free medium or chemical inhibition of glycolysis or the mitochondrial electron transport chain strongly suppresses HTLV-1 plus-strand transcription. We conclude that glucose metabolism and oxygen tension regulate HTLV-1 proviral latency and reactivation in vivo. Physiological (1%) hypoxia enhances HTLV-1 plus-strand transcription HTLV-1 transcription is hypoxia regulated but HIF independent Inhibition of glycolysis or the mitochondrial ETC suppresses HTLV-1 transcription Extracellular glucose concentration regulates HTLV-1 reactivation from latency
Collapse
Affiliation(s)
- Anurag Kulkarni
- Section of Virology, Department of Medicine, Imperial College, London W2 1PG, UK
| | - Manuel Mateus
- Section of Virology, Department of Medicine, Imperial College, London W2 1PG, UK
| | - Cyrille C Thinnes
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, Mansfield Road, Oxford, UK
| | - James S McCullagh
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, Mansfield Road, Oxford, UK
| | - Christopher J Schofield
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, Mansfield Road, Oxford, UK
| | - Graham P Taylor
- Section of Virology, Department of Medicine, Imperial College, London W2 1PG, UK
| | - Charles R M Bangham
- Section of Virology, Department of Medicine, Imperial College, London W2 1PG, UK.
| |
Collapse
|
89
|
Schröck C, Eydt C, Geburek F, Kaiser L, Päbst F, Burk J, Pfarrer C, Staszyk C. Bone marrow-derived multipotent mesenchymal stromal cells from horses after euthanasia. Vet Med Sci 2017; 3:239-251. [PMID: 29152317 PMCID: PMC5677777 DOI: 10.1002/vms3.74] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Allogeneic equine multipotent mesenchymal stromal cells (eMSCs) have been proposed for use in regenerative therapies in veterinary medicine. A source of allogeneic eMSCs might be the bone marrow from euthanized horses. The purpose of this study was to compare in vitro characteristics of equine bone marrow derived eMSC (eBM‐MSCs) from euthanized horses (eut‐MSCs) and from narcotized horses (nar‐MSCs). Eut‐MSCs and nar‐MSCs showed typical eMSC marker profiles (positive: CD44, CD90; negative: CD11a/CD18 and MHCII) and possessed tri‐lineage differentiation characteristics. Although CD105 and MHCI expression varied, no differences were detected between eut‐MSCs and nar‐MSCs. Proliferation characteristics did not differ between eut‐MSCs and nar‐MSCs, but age dependent decrease in proliferation and increase in MHCI expression was detected. These results suggest the possible use of eut‐MSCs for therapeutic applications and production of commercial available eBM‐MSC products.
Collapse
Affiliation(s)
- Carmen Schröck
- Institute for Veterinary Anatomy, -Histology and -EmbryologyJustus-Liebig-UniversityGiessenGermany
| | - Carina Eydt
- Institute of AnatomyUniversity of Veterinary Medicine HannoverHannoverGermany
| | | | - Lena Kaiser
- Institute for Veterinary Anatomy, -Histology and -EmbryologyJustus-Liebig-UniversityGiessenGermany
| | - Felicitas Päbst
- Translational Centre for Regenerative Medicine (TRM)University of LeipzigLeipzigGermany.,Faculty of Veterinary MedicineLarge Animal Clinic for SurgeryUniversity of LeipzigLeipzigGermany
| | - Janina Burk
- Translational Centre for Regenerative Medicine (TRM)University of LeipzigLeipzigGermany.,Faculty of Veterinary MedicineLarge Animal Clinic for SurgeryUniversity of LeipzigLeipzigGermany
| | - Christiane Pfarrer
- Institute of AnatomyUniversity of Veterinary Medicine HannoverHannoverGermany
| | - Carsten Staszyk
- Institute for Veterinary Anatomy, -Histology and -EmbryologyJustus-Liebig-UniversityGiessenGermany
| |
Collapse
|
90
|
Ballester-Beltrán J, Trujillo S, Alakpa EV, Compañ V, Gavara R, Meek D, West CC, Péault B, Dalby MJ, Salmerón-Sánchez M. Confined Sandwichlike Microenvironments Tune Myogenic Differentiation. ACS Biomater Sci Eng 2017; 3:1710-1718. [PMID: 28824958 PMCID: PMC5558191 DOI: 10.1021/acsbiomaterials.7b00109] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 06/09/2017] [Indexed: 12/29/2022]
Abstract
Sandwichlike (SW) cultures are engineered as a multilayer technology to simultaneously stimulate dorsal and ventral cell receptors, seeking to mimic cell adhesion in three-dimensional (3D) environments in a reductionist manner. The effect of this environment on cell differentiation was investigated for several cell types cultured in standard growth media, which promotes proliferation on two-dimensional (2D) surfaces and avoids any preferential differentiation. First, murine C2C12 myoblasts showed specific myogenic differentiation. Human mesenchymal stem cells (hMSCs) of adipose and bone marrow origin, which can differentiate toward a wider variety of lineages, showed again myodifferentiation. Overall, this study shows myogenic differentiation in normal growth media for several cell types under SW conditions, avoiding the use of growth factors and cytokines, i.e., solely by culturing cells within the SW environment. Mechanistically, it provides further insights into the balance between integrin adhesion to the dorsal substrate and the confinement imposed by the SW system.
Collapse
Affiliation(s)
- José Ballester-Beltrán
- Division
of Biomedical Engineering, School of Engineering, University of Glasgow. Rankine Building, Oakfield Avenue, Glasgow G12 8LT, United Kingdom
| | - Sara Trujillo
- Division
of Biomedical Engineering, School of Engineering, University of Glasgow. Rankine Building, Oakfield Avenue, Glasgow G12 8LT, United Kingdom
| | - Enateri V. Alakpa
- Centre
for Cell Engineering, Institute of Molecular, Cell and Systems Biology, University of Glasgow. Joseph Black Building, University Avenue, Glasgow G12 8QQ, United Kingdom
| | - Vicente Compañ
- Escuela
Técnica Superior de Ingenieros Industriales, Departamento de
Termodinámica Aplicada, Universitat
Politècnica de València, Camino de Vera s/n, Valencia, Valencia 46022, Spain
| | - Rafael Gavara
- Instituto
de Agroquímica y Tecnología de Alimentos. Consejo Superior
de Investigaciones Científicas (IATA-CSIC), Departamento de Investigación: Conservación y Calidad
de Alimentos,Calle Agustín
Escardino 7, Paterna, Valencia 46980, Spain
| | - Dominic Meek
- Centre
for Cell Engineering, Institute of Molecular, Cell and Systems Biology, University of Glasgow. Joseph Black Building, University Avenue, Glasgow G12 8QQ, United Kingdom
| | - Christopher C. West
- Centre for
Regenerative Medicine and Centre for Cardiovascular Science, University of Edinburgh. 47 Little France Crescent, Edinburgh EH16 4TJ, United
Kingdom
| | - Bruno Péault
- Centre for
Regenerative Medicine and Centre for Cardiovascular Science, University of Edinburgh. 47 Little France Crescent, Edinburgh EH16 4TJ, United
Kingdom
| | - Matthew J. Dalby
- Centre
for Cell Engineering, Institute of Molecular, Cell and Systems Biology, University of Glasgow. Joseph Black Building, University Avenue, Glasgow G12 8QQ, United Kingdom
| | - Manuel Salmerón-Sánchez
- Division
of Biomedical Engineering, School of Engineering, University of Glasgow. Rankine Building, Oakfield Avenue, Glasgow G12 8LT, United Kingdom
| |
Collapse
|
91
|
Andreeva ER, Udartseva OO, Zhidkova OV, Buravkov SV, Ezdakova MI, Buravkova LB. IFN‐gamma priming of adipose‐derived stromal cells at “physiological” hypoxia. J Cell Physiol 2017; 233:1535-1547. [DOI: 10.1002/jcp.26046] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 06/09/2017] [Indexed: 12/31/2022]
Affiliation(s)
- Elena R. Andreeva
- Cell Physiology LaboratoryInstitute of Biomedical ProblemsRussian Academy of SciencesMoscowRussia
| | - Olga O. Udartseva
- Cell Physiology LaboratoryInstitute of Biomedical ProblemsRussian Academy of SciencesMoscowRussia
| | - Olga V. Zhidkova
- Cell Physiology LaboratoryInstitute of Biomedical ProblemsRussian Academy of SciencesMoscowRussia
| | | | - Maria I. Ezdakova
- Cell Physiology LaboratoryInstitute of Biomedical ProblemsRussian Academy of SciencesMoscowRussia
| | - Ludmila B. Buravkova
- Cell Physiology LaboratoryInstitute of Biomedical ProblemsRussian Academy of SciencesMoscowRussia
| |
Collapse
|
92
|
Liu J, Edgington-Giordano F, Dugas C, Abrams A, Katakam P, Satou R, Saifudeen Z. Regulation of Nephron Progenitor Cell Self-Renewal by Intermediary Metabolism. J Am Soc Nephrol 2017; 28:3323-3335. [PMID: 28754792 DOI: 10.1681/asn.2016111246] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Accepted: 06/09/2017] [Indexed: 12/21/2022] Open
Abstract
Nephron progenitor cells (NPCs) show an age-dependent capacity to balance self-renewal with differentiation. Older NPCs (postnatal day 0) exit the progenitor niche at a higher rate than younger (embryonic day 13.5) NPCs do. This behavior is reflected in the transcript profiles of young and old NPCs. Bioenergetic pathways have emerged as important regulators of stem cell fate. Here, we investigated the mechanisms underlying this regulation in murine NPCs. Upon isolation and culture in NPC renewal medium, younger NPCs displayed a higher glycolysis rate than older NPCs. Inhibition of glycolysis enhanced nephrogenesis in cultured embryonic kidneys, without increasing ureteric tree branching, and promoted mesenchymal-to-epithelial transition in cultured isolated metanephric mesenchyme. Cotreatment with a canonical Wnt signaling inhibitor attenuated but did not entirely block the increase in nephrogenesis observed after glycolysis inhibition. Furthermore, inhibition of the phosphatidylinositol 3-kinase/Akt self-renewal signaling pathway or stimulation of differentiation pathways in the NPC decreased glycolytic flux. Our findings suggest that glycolysis is a pivotal, cell-intrinsic determinant of NPC fate, with a high glycolytic flux supporting self-renewal and inhibition of glycolysis stimulating differentiation.
Collapse
Affiliation(s)
- Jiao Liu
- Department of Pediatrics, Section of Nephrology
| | | | | | - Anna Abrams
- Department of Pediatrics, Section of Nephrology
| | - Prasad Katakam
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana
| | | | | |
Collapse
|
93
|
Ortega JA, Sirois CL, Memi F, Glidden N, Zecevic N. Oxygen Levels Regulate the Development of Human Cortical Radial Glia Cells. Cereb Cortex 2017; 27:3736-3751. [PMID: 27600849 PMCID: PMC6075453 DOI: 10.1093/cercor/bhw194] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Revised: 04/29/2016] [Accepted: 05/24/2016] [Indexed: 12/17/2022] Open
Abstract
The oxygen (O2) concentration is a vital parameter for controlling the survival, proliferation, and differentiation of neural stem cells. A prenatal reduction of O2 levels (hypoxia) often leads to cognitive and behavioral defects, attributable to altered neural development. In this study, we analyzed the effects of O2 levels on human cortical progenitors, the radial glia cells (RGCs), during active neurogenesis, corresponding to the second trimester of gestation. Small changes in O2 levels profoundly affected RGC survival, proliferation, and differentiation. Physiological hypoxia (3% O2) promoted neurogenesis, whereas anoxia (<1% O2) and severe hypoxia (1% O2) arrested the differentiation of human RGCs, mainly by altering the generation of glutamatergic neurons. The in vitro activation of Wnt-β-catenin signaling rescued the proliferation and neuronal differentiation of RGCs subjected to anoxia. Pathologic hypoxia (≤1% O2) also exerted negative effects on gliogenesis, by decreasing the number of O4+ preoligodendrocytes and increasing the number of reactive astrocytes derived from cortical RGCs. O2-dependent alterations in glutamatergic neurogenesis and oligodendrogenesis can lead to significant changes in cortical circuitry formation. A better understanding of the cellular effects caused by changes in O2 levels during human cortical development is essential to elucidating the etiology of numerous neurodevelopmental disorders.
Collapse
Affiliation(s)
- J Alberto Ortega
- Department of Neuroscience, University of Connecticut Health, Farmington, CT 06030, USA
| | - Carissa L Sirois
- Department of Neuroscience, University of Connecticut Health, Farmington, CT 06030, USA
- Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Fani Memi
- Department of Neuroscience, University of Connecticut Health, Farmington, CT 06030, USA
| | - Nicole Glidden
- Department of Neuroscience, University of Connecticut Health, Farmington, CT 06030, USA
| | - Nada Zecevic
- Department of Neuroscience, University of Connecticut Health, Farmington, CT 06030, USA
| |
Collapse
|
94
|
Ianniciello A, Dumas PY, Drullion C, Guitart A, Villacreces A, Peytour Y, Chevaleyre J, Brunet de la Grange P, Vigon I, Desplat V, Priault M, Sbarba PD, Ivanovic Z, Mahon FX, Pasquet JM. Chronic myeloid leukemia progenitor cells require autophagy when leaving hypoxia-induced quiescence. Oncotarget 2017; 8:96984-96992. [PMID: 29228587 PMCID: PMC5722539 DOI: 10.18632/oncotarget.18904] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 06/17/2017] [Indexed: 12/15/2022] Open
Abstract
Albeit tyrosine kinase inhibitors anti-Abl used in Chronic Myeloid Leukemia (CML) block the deregulated activity of the Bcr-Abl tyrosine kinase and induce remission in 90% of patients, they do not eradicate immature hematopoietic compartments of leukemic stem cells. To elucidate if autophagy is important for stem cell survival and/or proliferation, we used culture in low oxygen concentration (0.1% O2 for 7 days) followed back by non-restricted O2 supply (normoxic culture) to mimic stem cell proliferation and commitment. Knockdown of Atg7 expression, a key player in autophagy, in K562 cell line inhibited autophagy compared to control cells. Upon 7 days at 0.1% O2 both K562 and K562 shATG7 cells stopped to proliferate and a similar amount of viable cells remained. Back to non-restricted O2 supply K562 cells proliferate whereas K562 shATG7 cells exhibited strong apoptosis. Using immunomagnetic sorted normal and CML CD34+ cells, we inhibited the autophagic process by lentiviral infection expressing shATG7 or using a Vps34 inhibitor. Both, normal and CML CD34+ cells either competent or deficient for autophagy stopped to proliferate in hypoxia. Surprisingly, while normal CD34+ cells proliferate back to non restricted O2 supply, the CML CD34+ cells deficient for autophagy failed to proliferate. All together, these results suggest that autophagy is required for CML CD34+ commitment while it is dispensable for normal CD34 cells.
Collapse
Affiliation(s)
- Angela Ianniciello
- Cellules Souches Hématopoïétiques Normales et Leucémiques, INSERM U1035 BMGIC, Université de Bordeaux, 33076 Bordeaux Cedex, France
| | - Pierre-Yves Dumas
- Cellules Souches Hématopoïétiques Normales et Leucémiques, INSERM U1035 BMGIC, Université de Bordeaux, 33076 Bordeaux Cedex, France
| | - Claire Drullion
- Cellules Souches Hématopoïétiques Normales et Leucémiques, INSERM U1035 BMGIC, Université de Bordeaux, 33076 Bordeaux Cedex, France
| | - Amélie Guitart
- Cellules Souches Hématopoïétiques Normales et Leucémiques, INSERM U1035 BMGIC, Université de Bordeaux, 33076 Bordeaux Cedex, France
| | - Arnaud Villacreces
- Cellules Souches Hématopoïétiques Normales et Leucémiques, INSERM U1035 BMGIC, Université de Bordeaux, 33076 Bordeaux Cedex, France
| | - Yan Peytour
- Cellules Souches Hématopoïétiques Normales et Leucémiques, INSERM U1035 BMGIC, Université de Bordeaux, 33076 Bordeaux Cedex, France
| | - Jean Chevaleyre
- Cellules Souches Hématopoïétiques Normales et Leucémiques, INSERM U1035 BMGIC, Université de Bordeaux, 33076 Bordeaux Cedex, France.,Etablissement Français du Sang Aquitaine-Limousin, 33075 Bordeaux, France
| | - Philippe Brunet de la Grange
- Cellules Souches Hématopoïétiques Normales et Leucémiques, INSERM U1035 BMGIC, Université de Bordeaux, 33076 Bordeaux Cedex, France.,Etablissement Français du Sang Aquitaine-Limousin, 33075 Bordeaux, France
| | - Isabelle Vigon
- Cellules Souches Hématopoïétiques Normales et Leucémiques, INSERM U1035 BMGIC, Université de Bordeaux, 33076 Bordeaux Cedex, France
| | - Vanessa Desplat
- Cellules Souches Hématopoïétiques Normales et Leucémiques, INSERM U1035 BMGIC, Université de Bordeaux, 33076 Bordeaux Cedex, France
| | - Muriel Priault
- UMR CNRS 5095, I.B.G.C, Université de Bordeaux, 33077 Bordeaux Cedex, France
| | - Persio Dello Sbarba
- Department of Experimental and Clinical Biomedical Sciences, Università degli Studi di Firenze, 50134 Firenze, Italia
| | - Zoran Ivanovic
- Cellules Souches Hématopoïétiques Normales et Leucémiques, INSERM U1035 BMGIC, Université de Bordeaux, 33076 Bordeaux Cedex, France.,Etablissement Français du Sang Aquitaine-Limousin, 33075 Bordeaux, France
| | | | - Jean-Max Pasquet
- Cellules Souches Hématopoïétiques Normales et Leucémiques, INSERM U1035 BMGIC, Université de Bordeaux, 33076 Bordeaux Cedex, France
| |
Collapse
|
95
|
Ratushnyy A, Lobanova M, Buravkova LB. Expansion of adipose tissue-derived stromal cells at "physiologic" hypoxia attenuates replicative senescence. Cell Biochem Funct 2017; 35:232-243. [PMID: 28589682 DOI: 10.1002/cbf.3267] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 03/02/2017] [Accepted: 03/28/2017] [Indexed: 12/16/2022]
Abstract
Multipotent mesenchymal stromal cells are considered as a perspective tool in cell therapy and regenerative medicine. Unfortunately, autologous cell therapy does not always provide positive outcomes in elder donors, perhaps as a result of the alterations of stem cell compartments. The mechanisms of stem and progenitor cell senescence and the factors engaged are investigated intensively. In present paper, we elucidated the effects of tissue-related O2 on morphology, functions, and transcriptomic profile of adipose tissue-derived stromal cells (ASCs) in replicative senescence in vitro model. Replicatively senescent ASCs at ambient (20%) O2 (12-21 passages) demonstrated an increased average cell size, granularity, reactive oxygen species level, including anion superoxide, lysosomal compartment activity, and IL-6 production. Decreased ASC viability and proliferation, as well as the change of more than 10 senescence-associated gene expression were detected (IGF1, CDKN1C, ID1, CCND1, etc). Long-term ASC expansion at low O2 (5%) revoked in part the replicative senescence-associated alterations.
Collapse
Affiliation(s)
- Andrey Ratushnyy
- Lab. of Cell Physiology, Institute of Biomedical Problems of Russian Academy of Science, Moscow, Russia
| | - Margarita Lobanova
- Lab. of Cell Physiology, Institute of Biomedical Problems of Russian Academy of Science, Moscow, Russia
| | - Ludmila B Buravkova
- Lab. of Cell Physiology, Institute of Biomedical Problems of Russian Academy of Science, Moscow, Russia
| |
Collapse
|
96
|
To harness stem cells by manipulation of energetic metabolism. Transfus Clin Biol 2017; 24:468-471. [PMID: 28602675 DOI: 10.1016/j.tracli.2017.05.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 05/15/2017] [Indexed: 11/20/2022]
Abstract
The maintenance of the primitive Hematopoietic Stem Cells (HSC) in course of ex vivo expansion is a critical point to preserve the long-term reconstituting capacity of a hematopoietic graft. On the basis of the numerous experimental results, the maintenance of primitive HSC is related to their specific metabolic profile shifted towards the anaerobiosis. Hence, in addition to the exposition of the cultures to more appropriate, physiologically low O2 concentrations (usually misleadingly termed "hypoxia"), a specter of "hypoxia-mimicking" factors (cytokines, growth factors, receptor-ligands, antioxidants) can be applied to maintain this specific metabolic profile enabling an appropriate genetic and epigenetic regulation. Some factors already proved to be able to achieve this goal and "hypoxia-mimicking" ex vivo cultures were already used to produce cells for clinical trials. In this article we are discussing the approaches aimed to amplify and/or to condition the HSC, based on the manipulation of energetic metabolism properties.
Collapse
|
97
|
Ivanovic Z. Stem cell evolutionary paradigm and cell engineering. Transfus Clin Biol 2017; 24:251-255. [PMID: 28596084 DOI: 10.1016/j.tracli.2017.05.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 05/15/2017] [Indexed: 12/23/2022]
Abstract
Studying hematopoietic and mesenchymal stem cells for almost three decades revealed some similarities between the stem cell entity and the single-celled eukaryotes exhibiting the anaerobic/facultative aerobic metabolic features. A careful analysis of nowadays knowledge concerning the early eukaryotic evolution allowed us to reveal some analogies between stem cells in the metazoan tissues and the single-celled eukaryotes which existed during the first phase of eukaryotes evolution in mid-Proterozoic era. In fact, it is possible to trace the principle of the self-renewal back to the first eukaryotic common ancestor, the first undifferentiated nucleated cell possessing the primitive, mostly anaerobically-respiring mitochondria and a capacity to reproduction by a simple cell division "à l'identique". Similarly, the diversification of these single-cell eukaryotes and acquiring of complex life cycle allowed/conditioned by the increase of O2 in atmosphere (and consequently in the water environment) represents a prototype for the phenomenon of commitment/differentiation. This point of view allowed to predict the ex-vivo behavior of stem cells with respect to the O2 availability and metabolic profile which enabled to conceive the successful protocols of stem cell expansion and ex vivo conditioning based on "respecting" this relationship between the anaerobiosis and stemness. In this review, the basic elements of this paradigm and a possible application in cell engineering were discussed.
Collapse
Affiliation(s)
- Z Ivanovic
- CS21010, Établissement français du sang Aquitaine-Limousin, place Amélie-Raba-Léon, 33075 Bordeaux cedex, France; U1035 INSEM, université de Bordeaux, 33076 Bordeaux cedex, France.
| |
Collapse
|
98
|
The Leukemic Stem Cell Niche: Adaptation to "Hypoxia" versus Oncogene Addiction. Stem Cells Int 2017; 2017:4979474. [PMID: 29118813 PMCID: PMC5651121 DOI: 10.1155/2017/4979474] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 04/05/2017] [Indexed: 02/08/2023] Open
Abstract
Previous studies based on low oxygen concentrations in the incubation atmosphere revealed that metabolic factors govern the maintenance of normal hematopoietic or leukemic stem cells (HSC and LSC). The physiological oxygen concentration in tissues ranges between 0.1 and 5.0%. Stem cell niches (SCN) are placed in tissue areas at the lower end of this range (“hypoxic” SCN), to which stem cells are metabolically adapted and where they are selectively hosted. The data reported here indicated that driver oncogenic proteins of several leukemias are suppressed following cell incubation at oxygen concentration compatible with SCN physiology. This suppression is likely to represent a key positive regulator of LSC survival and maintenance (self-renewal) within the SCN. On the other hand, LSC committed to differentiation, unable to stand suppression because of addiction to oncogenic signalling, would be unfit to home in SCN. The loss of oncogene addiction in SCN-adapted LSC has a consequence of crucial practical relevance: the refractoriness to inhibitors of the biological activity of oncogenic protein due to the lack of their molecular target. Thus, LSC hosted in SCN are suited to sustain the long-term maintenance of therapy-resistant minimal residual disease.
Collapse
|
99
|
Costa M, Cerqueira MT, Santos TC, Sampaio-Marques B, Ludovico P, Marques AP, Pirraco RP, Reis RL. Cell sheet engineering using the stromal vascular fraction of adipose tissue as a vascularization strategy. Acta Biomater 2017; 55:131-143. [PMID: 28347862 DOI: 10.1016/j.actbio.2017.03.034] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 03/20/2017] [Accepted: 03/23/2017] [Indexed: 12/17/2022]
Abstract
Current vascularization strategies for Tissue Engineering constructs, in particular cell sheet-based, are limited by time-consuming and expensive endothelial cell isolation and/or by the complexity of using extrinsic growth factors. Herein, we propose an alternative strategy using angiogenic cell sheets (CS) obtained from the stromal vascular fraction (SVF) of adipose tissue that can be incorporated into more complex constructs. Cells from the SVF were cultured in normoxic and hypoxic conditions for up to 8days in the absence of extrinsic growth factors. Immunocytochemistry against CD31 and CD146 revealed spontaneous organization in capillary-like structures, more complex after hypoxic conditioning. Inhibition of HIF-1α pathway hindered capillary-like structure formation in SVF cells cultured in hypoxia, suggesting a role of HIF-1α. Moreover, hypoxic SVF cells showed a trend for increased secretion of angiogenic factors, which was reflected in increased network formation by endothelial cells cultured on matrigel using that conditioned medium. In vivo implantation of SVF CS in a mouse hind limb ischemia model revealed that hypoxia-conditioned CS led to improved restoration of blood flow. Both in vitro and in vivo data suggest that SVF CS can be used as simple and cost-efficient tools to promote functional vascularization of TE constructs. STATEMENT OF SIGNIFICANCE Neovascularization after implantation is a major obstacle for producing clinically viable cell sheet-based tissue engineered constructs. Strategies using endothelial cells and extrinsic angiogenic growth factors are expensive and time consuming and may raise concerns of tumorigenicity. In this manuscript, we describe a simplified approach using angiogenic cell sheets fabricated from the stromal vascular fraction of adipose tissue. The strong angiogenic behavior of these cell sheets, achieved without the use of external growth factors, was further stimulated by low oxygen culture. When implanted in an in vivo model of hind limb ischemia, the angiogenic cell sheets contributed to blood flux recovery. These cell sheets can therefore be used as a straightforward tool to increase the neovascularization of cell sheet-based thick constructs.
Collapse
Affiliation(s)
- Marina Costa
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal; Institute of Biophysics and Biomedical Engineering, Faculty of Sciences of the University of Lisbon, Lisbon, Portugal
| | - Mariana T Cerqueira
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Tírcia C Santos
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Belém Sampaio-Marques
- Institute of Life and Health Sciences, School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Paula Ludovico
- Institute of Life and Health Sciences, School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Alexandra P Marques
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rogério P Pirraco
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rui L Reis
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
100
|
Davies JE, Walker JT, Keating A. Concise Review: Wharton's Jelly: The Rich, but Enigmatic, Source of Mesenchymal Stromal Cells. Stem Cells Transl Med 2017; 6:1620-1630. [PMID: 28488282 PMCID: PMC5689772 DOI: 10.1002/sctm.16-0492] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 02/03/2017] [Accepted: 02/24/2017] [Indexed: 12/21/2022] Open
Abstract
The umbilical cord has become an increasingly used source of mesenchymal stromal cells for preclinical and, more recently, clinical studies. Despite the increased activity, several aspects of this cell population have been under‐appreciated. Key issues are that consensus on the anatomical structures within the cord is lacking, and potentially different populations are identified as arising from a single source. To help address these points, we propose a histologically based nomenclature for cord structures and provide an analysis of their developmental origins and composition. Methods of cell isolation from Wharton's jelly are discussed and the immunophenotypic and clonal characteristics of the cells are evaluated. The perivascular origin of the cells is also addressed. Finally, clinical trials with umbilical cord cells are briefly reviewed. Interpreting the outcomes of the many clinical studies that have been undertaken with mesenchymal stromal cells from different tissue sources has been challenging, for many reasons. It is, therefore, particularly important that as umbilical cord cells are increasingly deployed therapeutically, we strive to better understand the derivation and functional characteristics of the cells from this important tissue source. Stem Cells Translational Medicine2017;6:1620–1630
Collapse
Affiliation(s)
- John E Davies
- Institute of Biomaterials and Biomedical Engineering, Toronto, Ontario, Canada.,Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - John T Walker
- Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Armand Keating
- Institute of Biomaterials and Biomedical Engineering, Toronto, Ontario, Canada.,Cell Therapy Program, Arthritis Program, Krembil Research Institute, and Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| |
Collapse
|