1
|
Iwoń Z, Krogulec E, Kierlańczyk A, Wojasiński M, Jastrzębska E. Hypoxia and re-oxygenation effects on human cardiomyocytes cultured on polycaprolactone and polyurethane nanofibrous mats. J Biol Eng 2024; 18:37. [PMID: 38844979 PMCID: PMC11157810 DOI: 10.1186/s13036-024-00432-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 05/24/2024] [Indexed: 06/09/2024] Open
Abstract
Heart diseases are caused mainly by chronic oxygen insufficiency (hypoxia), leading to damage and apoptosis of cardiomyocytes. Research into the regeneration of a damaged human heart is limited due to the lack of cellular models that mimic damaged cardiac tissue. Based on the literature, nanofibrous mats affect the cardiomyocyte morphology and stimulate the growth and differentiation of cells cultured on them; therefore, nanofibrous materials can support the production of in vitro models that faithfully mimic the 3D structure of human cardiac tissue. Nanofibrous mats were used as scaffolds for adult primary human cardiomyocytes (HCM) and immature human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs). This work focuses on understanding the effects of hypoxia and re-oxygenation on human cardiac cells cultured on polymer nanofibrous mats made of poly(ε-caprolactone) (PCL) and polyurethane (PU). The expression of selected genes and proteins in cardiomyocytes during hypoxia and re-oxygenation were evaluated. In addition, the type of cell death was analyzed. To the best of our knowledge, there are no studies on the effects of hypoxia on cardiomyocyte cells cultured on nanofibrous mats. The present study aimed to use nanofiber mats as scaffolds that structurally could mimic cardiac extracellular matrix. Understanding the impact of 3D structural properties in vitro cardiac models on different human cardiomyocytes is crucial for advancing cardiac tissue engineering and regenerative medicine. Observing how 3D scaffolds affect cardiomyocyte function under hypoxic conditions is necessary to understand the functioning of the entire human heart.
Collapse
Affiliation(s)
- Zuzanna Iwoń
- Chair of Medical Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland
| | - Ewelina Krogulec
- Laboratory of Cell Signaling and Metabolic Disorders, Nencki Institute of Experimental Biology PAS, Warsaw, Poland
| | - Aleksandra Kierlańczyk
- Chair of Medical Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland
| | - Michał Wojasiński
- Department of Biotechnology and Bioprocess Engineering, Faculty of Chemical and Process Engineering, Warsaw University of Technology, Warsaw, Poland
| | - Elżbieta Jastrzębska
- Chair of Medical Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland.
- Centre for Advanced Materials and Technologies, CEZAMAT Warsaw University of Technology, Warsaw, Poland.
| |
Collapse
|
2
|
Kang H, Strong AL, Sun Y, Guo L, Juan C, Bancroft AC, Choi JH, Pagani CA, Fernandes AA, Woodard M, Lee J, Ramesh S, James AW, Hudson D, Dalby KN, Xu L, Tower RJ, Levi B. The HIF-1α/PLOD2 axis integrates extracellular matrix organization and cell metabolism leading to aberrant musculoskeletal repair. Bone Res 2024; 12:17. [PMID: 38472175 PMCID: PMC10933265 DOI: 10.1038/s41413-024-00320-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 01/04/2024] [Accepted: 02/01/2024] [Indexed: 03/14/2024] Open
Abstract
While hypoxic signaling has been shown to play a role in many cellular processes, its role in metabolism-linked extracellular matrix (ECM) organization and downstream processes of cell fate after musculoskeletal injury remains to be determined. Heterotopic ossification (HO) is a debilitating condition where abnormal bone formation occurs within extra-skeletal tissues. Hypoxia and hypoxia-inducible factor 1α (HIF-1α) activation have been shown to promote HO. However, the underlying molecular mechanisms by which the HIF-1α pathway in mesenchymal progenitor cells (MPCs) contributes to pathologic bone formation remain to be elucidated. Here, we used a proven mouse injury-induced HO model to investigate the role of HIF-1α on aberrant cell fate. Using single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics analyses of the HO site, we found that collagen ECM organization is the most highly up-regulated biological process in MPCs. Zeugopod mesenchymal cell-specific deletion of Hif1α (Hoxa11-CreERT2; Hif1afl/fl) significantly mitigated HO in vivo. ScRNA-seq analysis of these Hoxa11-CreERT2; Hif1afl/fl mice identified the PLOD2/LOX pathway for collagen cross-linking as downstream of the HIF-1α regulation of HO. Importantly, our scRNA-seq data and mechanistic studies further uncovered that glucose metabolism in MPCs is most highly impacted by HIF-1α deletion. From a translational aspect, a pan-LOX inhibitor significantly decreased HO. A newly screened compound revealed that the inhibition of PLOD2 activity in MPCs significantly decreased osteogenic differentiation and glycolytic metabolism. This suggests that the HIF-1α/PLOD2/LOX axis linked to metabolism regulates HO-forming MPC fate. These results suggest that the HIF-1α/PLOD2/LOX pathway represents a promising strategy to mitigate HO formation.
Collapse
Affiliation(s)
- Heeseog Kang
- Center for Organogenesis, Regeneration and Trauma, Department of Surgery, University of Texas Southwestern, Dallas, TX, 75390, USA
| | - Amy L Strong
- Section of Plastic Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Yuxiao Sun
- Center for Organogenesis, Regeneration and Trauma, Department of Surgery, University of Texas Southwestern, Dallas, TX, 75390, USA
| | - Lei Guo
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern, Dallas, TX, 75390, USA
| | - Conan Juan
- Center for Organogenesis, Regeneration and Trauma, Department of Surgery, University of Texas Southwestern, Dallas, TX, 75390, USA
| | - Alec C Bancroft
- Center for Organogenesis, Regeneration and Trauma, Department of Surgery, University of Texas Southwestern, Dallas, TX, 75390, USA
| | - Ji Hae Choi
- Center for Organogenesis, Regeneration and Trauma, Department of Surgery, University of Texas Southwestern, Dallas, TX, 75390, USA
| | - Chase A Pagani
- Center for Organogenesis, Regeneration and Trauma, Department of Surgery, University of Texas Southwestern, Dallas, TX, 75390, USA
| | - Aysel A Fernandes
- Department of Orthopedics and Sports Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Michael Woodard
- Center for Organogenesis, Regeneration and Trauma, Department of Surgery, University of Texas Southwestern, Dallas, TX, 75390, USA
| | - Juhoon Lee
- Division of Chemical Biology and Medicinal Chemistry, College of Pharmacy, University of Texas at Austin, Austin, TX, 78712, USA
| | - Sowmya Ramesh
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Aaron W James
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - David Hudson
- Department of Orthopedics and Sports Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Kevin N Dalby
- Division of Chemical Biology and Medicinal Chemistry, College of Pharmacy, University of Texas at Austin, Austin, TX, 78712, USA
| | - Lin Xu
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern, Dallas, TX, 75390, USA
| | - Robert J Tower
- Center for Organogenesis, Regeneration and Trauma, Department of Surgery, University of Texas Southwestern, Dallas, TX, 75390, USA
| | - Benjamin Levi
- Center for Organogenesis, Regeneration and Trauma, Department of Surgery, University of Texas Southwestern, Dallas, TX, 75390, USA.
| |
Collapse
|
3
|
Quantitative Phase Imaging Detecting the Hypoxia-Induced Patterns in Healthy and Neoplastic Human Colonic Epithelial Cells. Cells 2022; 11:cells11223599. [PMID: 36429026 PMCID: PMC9688862 DOI: 10.3390/cells11223599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/08/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
Hypoxia is a frequent phenomenon during carcinogenesis and may lead to functional and structural changes in proliferating cancer cells. Colorectal cancer (CRC) is one of the most common neoplasms in which hypoxia is associated with progression. The aim of this study was to assess the optical parameters and microanatomy of CRC and the normal intestinal epithelium cells using the digital holotomography (DHT) method. The examination was conducted on cancer (HT-29, LoVo) and normal colonic cells (CCD-18Co) cultured in normoxic and hypoxic environments. The assessment included optical parameters such as the refractive index (RI) and dry mass as well as the morphological features. Hypoxia decreased the RI in all cells as well as in their cytoplasm, nucleus, and nucleoli. The opposite tendency was noted for spheroid-vesicular structures, where the RI was higher for the hypoxic state. The total volume of hypoxic CCD-18Co and LoVo cells was decreased, while an increase in this parameter was observed for HT-29 cells. Hypoxia increased the radius and cell volume, including the dry mass of the vesicular content. The changes in the optics and morphology of hypoxic cells may suggest the possibility of using DHT in the detection of circulating tumor cells (CTCs).
Collapse
|
4
|
Van Meijel RLJ, Wang P, Bouwman F, Blaak EE, Mariman ECM, Goossens GH. The Effects of Mild Intermittent Hypoxia Exposure on the Abdominal Subcutaneous Adipose Tissue Proteome in Overweight and Obese Men: A First-in-Human Randomized, Single-Blind, and Cross-Over Study. Front Physiol 2022; 12:791588. [PMID: 35058800 PMCID: PMC8764283 DOI: 10.3389/fphys.2021.791588] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 11/30/2021] [Indexed: 11/13/2022] Open
Abstract
Adipose tissue (AT) oxygen tension (pO2) has been implicated in AT dysfunction and metabolic perturbations in both rodents and humans. Compelling evidence suggests that hypoxia exposure alters metabolism, at least partly through effects on AT. However, it remains to be elucidated whether mild intermittent hypoxia (MIH) exposure impacts the AT proteome. We performed a randomized, single-blind, and cross-over study to investigate the effects of seven consecutive days of MIH (FiO2 15%, 3x2h/d) compared to normoxia (FiO2 21%) exposure on the AT proteome in overweight/obese men. In vivo AT insulin sensitivity was determined by the gold standard hyperinsulinemic-euglycemic clamp, and abdominal subcutaneous AT biopsies were collected under normoxic fasting conditions following both exposure regimens (day 8). AT proteins were isolated and quantified using liquid chromatography-mass spectrometry. After correction for blood contamination, 1,022 AT protein IDs were identified, of which 123 were differentially expressed following MIH (p < 0.05). We demonstrate for the first time that MIH exposure, which markedly reduces in vivo AT oxygen tension, impacts the human AT proteome. Although we cannot exclude that a single differentially expressed protein might be a false positive finding, several functional pathways were altered by MIH exposure, also after adjustment for multiple testing. Specifically, differentially expressed proteins were involved in redox systems, cell-adhesion, actin cytoskeleton organization, extracellular matrix composition, and energy metabolism. The MIH-induced change in AT TMOD3 expression was strongly related to altered in vivo AT insulin sensitivity, thus linking MIH-induced effects on the AT proteome to metabolic changes in overweight/obese humans.
Collapse
Affiliation(s)
- Rens L J Van Meijel
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Ping Wang
- Department of Clinical Genetics, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Freek Bouwman
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Ellen E Blaak
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Edwin C M Mariman
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Gijs H Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| |
Collapse
|
5
|
Mehta V, Vilikkathala Sudhakaran S, Rath SN. Facile Route for 3D Printing of Transparent PETg-Based Hybrid Biomicrofluidic Devices Promoting Cell Adhesion. ACS Biomater Sci Eng 2021; 7:3947-3963. [PMID: 34282888 DOI: 10.1021/acsbiomaterials.1c00633] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
3D printing has emerged as a promising fabrication technique for microfluidic devices, overcoming some of the challenges associated with conventional soft lithography. Filament-based polymer extrusion (popularly known as fused deposition modeling (FDM)) is one of the most accessible 3D printing techniques available, offering a wide range of low-cost thermoplastic polymer materials for microfluidic device fabrication. However, low optical transparency is one of the significant limitations of extrusion-based microfluidic devices, rendering them unsuitable for cell culture-related biological applications. Moreover, previously reported extrusion-based devices were largely dependent on fluorescent dyes for cell imaging because of their poor transparency. First, we aim to improve the optical transparency of FDM-based microfluidic devices to enable bright-field microscopy of cells. This is achieved using (1) transparent polymer filament materials such as poly(ethylene terephthalate) glycol (PETg), (2) optimized 3D printing process parameters, and (3) a hybrid approach by integrating 3D printed microfluidic devices with cast poly(dimethylsiloxane) (PDMS) blocks. We begin by optimizing four essential 3D printing process parameters (layer height, printing speed, cooling fan speed, and extrusion flow), affecting the overall transparency of 3D printed devices. Optimized parameters produce exceptional optical transparency close to 80% in 3D printed PETg devices. Next, we demonstrate the potential of FDM-based 3D printing to fabricate transparent micromixing devices with complex planar and nonplanar channel networks. Most importantly, cells cultured on native 3D printed PETg surfaces show excellent cell attachment, spreading, and proliferation during 3 days of culture without extracellular matrix coating or surface treatment. Next, we introduce L929 cells inside hybrid PETg-PDMS biomicrofluidic devices as a proof of concept. We demonstrate that 3D printed hybrid biomicrofluidic devices promote cell adhesion, allow bright-field microscopy, and maintain high cell viability for 3 days. Finally, we demonstrate the applicability of the proposed fabrication approach for developing 3D printed microfluidic devices from other FDM-compatible transparent polymers such as polylactic acid (PLA) and poly(methyl methacrylate) (PMMA).
Collapse
Affiliation(s)
- Viraj Mehta
- Regenerative Medicine and Stem Cell Laboratory (RMS), Department of Biomedical Engineering, Indian Institute of Technology Hyderabad (IITH), Kandi, Sangareddy 502285, Telangana, India
| | - Sukanya Vilikkathala Sudhakaran
- Regenerative Medicine and Stem Cell Laboratory (RMS), Department of Biomedical Engineering, Indian Institute of Technology Hyderabad (IITH), Kandi, Sangareddy 502285, Telangana, India
| | - Subha Narayan Rath
- Regenerative Medicine and Stem Cell Laboratory (RMS), Department of Biomedical Engineering, Indian Institute of Technology Hyderabad (IITH), Kandi, Sangareddy 502285, Telangana, India
| |
Collapse
|
6
|
Dynamic flow priming programs allow tuning up the cell layers properties for engineered vascular graft. Sci Rep 2021; 11:14666. [PMID: 34282200 PMCID: PMC8290030 DOI: 10.1038/s41598-021-94023-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 06/29/2021] [Indexed: 12/15/2022] Open
Abstract
Tissue engineered vascular grafts (TEVG) are potentially clear from ethical and epidemiological concerns sources for reconstructive surgery for small diameter blood vessels replacement. Here, we proposed a novel method to create three-layered TEVG on biocompatible glass fiber scaffolds starting from flat sheet state into tubular shape and to train the resulting tissue by our developed bioreactor system. Constructed tubular tissues were matured and trained under 3 types of individual flow programs, and their mechanical and biological properties were analyzed. Training in the bioreactor significantly increased the tissue burst pressure resistance (up to 18 kPa) comparing to untrained tissue. Fluorescent imaging and histological examination of trained vascular tissue revealed that each cell layer has its own individual response to training flow rates. Histological analysis suggested reverse relationship between tissue thickness and shear stress, and the thickness variation profiles were individual between all three types of cell layers. Concluding: a three-layered tissue structure similar to physiological can be assembled by seeding different cell types in succession; the following training of the formed tissue with increasing flow in a bioreactor is effective for promoting cell survival, improving pressure resistance, and cell layer formation of desired properties.
Collapse
|
7
|
DeFrates KG, Franco D, Heber-Katz E, Messersmith PB. Unlocking mammalian regeneration through hypoxia inducible factor one alpha signaling. Biomaterials 2021; 269:120646. [PMID: 33493769 PMCID: PMC8279430 DOI: 10.1016/j.biomaterials.2020.120646] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 12/19/2020] [Accepted: 12/29/2020] [Indexed: 02/08/2023]
Abstract
Historically, the field of regenerative medicine has aimed to heal damaged tissue through the use of biomaterials scaffolds or delivery of foreign progenitor cells. Despite 30 years of research, however, translation and commercialization of these techniques has been limited. To enable mammalian regeneration, a more practical approach may instead be to develop therapies that evoke endogenous processes reminiscent of those seen in innate regenerators. Recently, investigations into tadpole tail regrowth, zebrafish limb restoration, and the super-healing Murphy Roths Large (MRL) mouse strain, have identified ancient oxygen-sensing pathways as a possible target to achieve this goal. Specifically, upregulation of the transcription factor, hypoxia-inducible factor one alpha (HIF-1α) has been shown to modulate cell metabolism and plasticity, as well as inflammation and tissue remodeling, possibly priming injuries for regeneration. Since HIF-1α signaling is conserved across species, environmental or pharmacological manipulation of oxygen-dependent pathways may elicit a regenerative response in non-healing mammals. In this review, we will explore the emerging role of HIF-1α in mammalian healing and regeneration, as well as attempts to modulate protein stability through hyperbaric oxygen treatment, intermittent hypoxia therapy, and pharmacological targeting. We believe that these therapies could breathe new life into the field of regenerative medicine.
Collapse
Affiliation(s)
- Kelsey G DeFrates
- Department of Bioengineering and Materials Science and Engineering, University of California, Berkeley, CA, USA.
| | - Daniela Franco
- Department of Bioengineering and Materials Science and Engineering, University of California, Berkeley, CA, USA.
| | - Ellen Heber-Katz
- Laboratory of Regenerative Medicine, Lankenau Institute for Medical Research, Wynnewood, PA, USA.
| | - Phillip B Messersmith
- Department of Bioengineering and Materials Science and Engineering, University of California, Berkeley, CA, USA; Materials Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.
| |
Collapse
|
8
|
Wang XT, Cheng K, Zhu L. Hypoxia Accelerate β-Actin Expression through Transcriptional Activation of ACTB by Nuclear Respiratory Factor-1. Mol Biol 2020. [DOI: 10.1134/s0026893321020011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
9
|
The effect of hypoxia on the proteomic signature of pig adipose-derived stromal/stem cells (pASCs). Sci Rep 2020; 10:20035. [PMID: 33208768 PMCID: PMC7676232 DOI: 10.1038/s41598-020-76796-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 10/16/2020] [Indexed: 01/16/2023] Open
Abstract
Human adipose-derived stem cells (ASCs) have potential to improve wound healing; however, their equivalents from domestic animals have received less attention as an alternative cell-based therapy for animals or even humans. Hypoxia is essential for maintaining stem cell functionality in tissue-specific niches. However, a cellular response to low oxygen levels has not been demonstrated in pig ASCs. Hence, the goal of our study was to characterize ASCs isolated from the subcutaneous fat of domestic pigs (pASCs) and examine the effect of hypoxia on their proteome and functional characteristics that might reproduce pASCs wound healing ability. Analysis of immunophenotypic and functional markers demonstrated that pASCs exhibited characteristics of mesenchymal stem cells. Proteomic analysis revealed 70 differentially abundant proteins between pASCs cultured under hypoxia (1% O2) or normoxia (21% O2). Among them, 42 proteins were enriched in the cells exposed to low oxygen, whereas 28 proteins showed decrease expression following hypoxia. Differentially expressed proteins were predominantly involved in cell metabolism, regulation of focal and intracellular communication, and attributed to wound healing. Functional examination of hypoxic pASCs demonstrated acquisition of contractile abilities in vitro. Overall, our results demonstrate that hypoxia pre-conditioning impacts the pASC proteome signature and contractile function in vitro and hence, they might be considered for further cell-based therapy study on wound healing.
Collapse
|
10
|
Fareed SA, Mostafa HES. Could aspartame exacerbate caffeine effects on renal maturation in rat's offspring? A biochemical and histological study. Birth Defects Res 2020; 113:90-107. [PMID: 33128303 DOI: 10.1002/bdr2.1836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 10/03/2020] [Accepted: 10/13/2020] [Indexed: 11/06/2022]
Abstract
BACKGROUND Caffeine and aspartame (ASP) are mostly used as a diet regimen to reduce overweight. The risk increase if used during critical life periods that may affect the development of fetal organs. OBJECTIVE To evaluate the individual and combined effects of maternal exposure to caffeine and ASP during gestation and lactation on the kidneys' development of rats' offspring. METHODS Pregnant rats were divided randomly into four groups; Group I (control group). Group II (ASP group): ASP was given at a dose of 40 mg of /kg/day. Group III (Caffeine group): caffeine was given at a dose of 80 mg/kg/day. Group IV (ASP & caffeine group); where previous doses of ASP and caffeine were given at the same time. All the treatments were given by oral gavage from the first day of pregnancy until postnatal day 30. Kidneys of rats' offspring were dissected and tested for detection of oxidative stress markers and for histopathological & immunohistochemical examination. RESULTS This study showed a high significant increase in oxidative load (malondialdehyde) in renal tissues in group IV associated with decreased activities of total glutathione and antioxidant enzymes (superoxide dismutase and glutathione peroxidase). Histological and morphometric examination results showed delayed maturation of renal tissues in Group II and III, but more deleterious effects were observed in group IV with a lot of pathological changes in renal tissues. CONCLUSION The extensive use of caffeine and ASP should be controlled to avoid the risk of their toxicity.
Collapse
|
11
|
Down syndrome iPSC model: endothelial perspective on tumor development. Oncotarget 2020; 11:3387-3404. [PMID: 32934781 PMCID: PMC7486695 DOI: 10.18632/oncotarget.27712] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 08/01/2020] [Indexed: 12/12/2022] Open
Abstract
Trisomy 21 (T21), known as Down syndrome (DS), is a widely studied chromosomal abnormality. Previous studies have shown that DS individuals have a unique cancer profile. While exhibiting low solid tumor prevalence, DS patients are at risk for hematologic cancers, such as acute megakaryocytic leukemia and acute lymphoblastic leukemia. We speculated that endothelial cells are active players in this clinical background. To this end, we hypothesized that impaired DS endothelial development and functionality, impacted by genome-wide T21 alterations, potentially results in a suboptimal endothelial microenvironment with the capability to prevent solid tumor growth. To test this hypothesis, we assessed molecular and phenotypic differences of endothelial cells differentiated from Down syndrome and euploid iPS cells. Microarray, RNA-Seq, and bioinformatic analyses revealed that most significantly expressed genes belong to angiogenic, cytoskeletal rearrangement, extracellular matrix remodeling, and inflammatory pathways. Interestingly, the majority of these genes are not located on Chromosome 21. To substantiate these findings, we carried out functional assays. The obtained phenotypic results correlated with the molecular data and showed that Down syndrome endothelial cells exhibit decreased proliferation, reduced migration, and a weak TNF-α inflammatory response. Based on this data, we provide a set of genes potentially associated with Down syndrome’s elevated leukemic incidence and its unfavorable solid tumor microenvironment—highlighting the potential use of these genes as therapeutic targets in translational cancer research.
Collapse
|
12
|
Galat Y, Perepitchka M, Elcheva I, Iannaccone S, Iannaccone PM, Galat V. iPSC-derived progenitor stromal cells provide new insights into aberrant musculoskeletal development and resistance to cancer in down syndrome. Sci Rep 2020; 10:13252. [PMID: 32764607 PMCID: PMC7414019 DOI: 10.1038/s41598-020-69418-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 07/06/2020] [Indexed: 12/13/2022] Open
Abstract
Down syndrome (DS) is a congenital disorder caused by trisomy 21 (T21). It is associated with cognitive impairment, muscle hypotonia, heart defects, and other clinical anomalies. At the same time, individuals with Down syndrome have lower prevalence of solid tumor formation. To gain new insights into aberrant DS development during early stages of mesoderm formation and its possible connection to lower solid tumor prevalence, we developed the first model of two types of DS iPSC-derived stromal cells. Utilizing bioinformatic and functional analyses, we identified over 100 genes with coordinated expression among mesodermal and endothelial cell types. The most significantly down-regulated processes in DS mesodermal progenitors were associated with decreased stromal progenitor performance related to connective tissue organization as well as muscle development and functionality. The differentially expressed genes included cytoskeleton-related genes (actin and myosin), ECM genes (Collagens, Galectin-1, Fibronectin, Heparan Sulfate, LOX, FAK1), cell cycle genes (USP16, S1P complexes), and DNA damage repair genes. For DS endothelial cells, our analysis revealed most down-regulated genes associated with cellular response to external stimuli, cell migration, and immune response (inflammation-based). Together with functional assays, these results suggest an impairment in mesodermal development capacity during early stages, which likely translates into connective tissue impairment in DS patients. We further determined that, despite differences in functional processes and characteristics, a significant number of differentially regulated genes involved in tumorigenesis were expressed in a highly coordinated manner across endothelial and mesodermal cells. These findings strongly suggest that microRNAs (miR-24-4, miR-21), cytoskeleton remodeling, response to stimuli, and inflammation can impact resistance to tumorigenesis in DS patients. Furthermore, we also show that endothelial cell functionality is impaired, and when combined with angiogenic inhibition, it can provide another mechanism for decreased solid tumor development. We propose that the same processes, which specify the basis of connective tissue impairment observed in DS patients, potentially impart a resistance to cancer by hindering tumor progression and metastasis. We further establish that cancer-related genes on Chromosome 21 are up-regulated, while genome-wide cancer-related genes are down-regulated. These results suggest that trisomy 21 induces a modified regulation and compensation of many biochemical pathways across the genome. Such downstream interactions may contribute toward promoting tumor resistant mechanisms.
Collapse
Affiliation(s)
- Yekaterina Galat
- Developmental Biology Program, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital, Chicago, IL, USA.
- Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| | - Mariana Perepitchka
- Developmental Biology Program, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital, Chicago, IL, USA.
| | - Irina Elcheva
- Developmental Biology Program, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital, Chicago, IL, USA
- Pediatrics, Division of Hematology and Oncology, Penn State Hershey College of Medicine, Hershey, PA, USA
| | - Stephen Iannaccone
- Developmental Biology Program, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital, Chicago, IL, USA
| | - Philip M Iannaccone
- Developmental Biology Program, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital, Chicago, IL, USA
- Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Vasiliy Galat
- Developmental Biology Program, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital, Chicago, IL, USA.
- Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
- ARTEC Biotech Inc, Chicago, IL, USA.
| |
Collapse
|
13
|
Zonderland J, Gomes DB, Pallada Y, Moldero IL, Camarero‐Espinosa S, Moroni L. Mechanosensitive regulation of stanniocalcin-1 by zyxin and actin-myosin in human mesenchymal stromal cells. Stem Cells 2020; 38:948-959. [PMID: 32379914 PMCID: PMC7497098 DOI: 10.1002/stem.3198] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 03/29/2020] [Accepted: 04/16/2020] [Indexed: 12/16/2022]
Abstract
Stanniocalcin-1 (STC1) secreted by mesenchymal stromal cells (MSCs) has anti-inflammatory functions, reduces apoptosis, and aids in angiogenesis, both in vitro and in vivo. However, little is known about the molecular mechanisms of its regulation. Here, we show that STC1 secretion is increased only under specific cell-stress conditions. We find that this is due to a change in actin stress fibers and actin-myosin tension. Abolishment of stress fibers by blebbistatin and knockdown of the focal adhesion protein zyxin leads to an increase in STC1 secretion. To also study this connection in 3D, where few focal adhesions and actin stress fibers are present, STC1 expression was analyzed in 3D alginate hydrogels and 3D electrospun scaffolds. Indeed, STC1 secretion was increased in these low cellular tension 3D environments. Together, our data show that STC1 does not directly respond to cell stress, but that it is regulated through mechanotransduction. This research takes a step forward in the fundamental understanding of STC1 regulation and can have implications for cell-based regenerative medicine, where cell survival, anti-inflammatory factors, and angiogenesis are critical.
Collapse
Affiliation(s)
- Jip Zonderland
- Complex Tissue Regeneration Department, MERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastrichtThe Netherlands
| | - David B. Gomes
- Complex Tissue Regeneration Department, MERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastrichtThe Netherlands
| | - Yves Pallada
- Complex Tissue Regeneration Department, MERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastrichtThe Netherlands
| | - Ivan L. Moldero
- Complex Tissue Regeneration Department, MERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastrichtThe Netherlands
| | - Sandra Camarero‐Espinosa
- Complex Tissue Regeneration Department, MERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastrichtThe Netherlands
| | - Lorenzo Moroni
- Complex Tissue Regeneration Department, MERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastrichtThe Netherlands
| |
Collapse
|
14
|
Nilnumkhum A, Kanlaya R, Yoodee S, Thongboonkerd V. Caffeine inhibits hypoxia-induced renal fibroblast activation by antioxidant mechanism. Cell Adh Migr 2020; 13:260-272. [PMID: 31271106 PMCID: PMC6650197 DOI: 10.1080/19336918.2019.1638691] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Caffeine has been demonstrated to possess anti-fibrotic activity against liver fibrosis. However, its role in renal fibrosis remained unclear. This study investigated the effects of caffeine on renal fibroblast activation induced by hypoxia (one of the inducers for renal fibrosis). BHK-21 fibroblasts were cultured under normoxia or hypoxia with or without caffeine treatment. Hypoxia increased levels of fibronectin, α-smooth muscle actin, actin stress fibers, intracellular reactive oxygen species (ROS), and oxidized proteins. However, caffeine successfully preserved all these activated fibroblast markers to their basal levels. Cellular catalase activity was dropped under hypoxic condition but could be reactivated by caffeine. Hif1a gene and stress-responsive Nrf2 signaling molecule were elevated/activated by hypoxia, but only Nrf2 could be partially recovered by caffeine. These data suggest that caffeine exhibits anti-fibrotic effect against hypoxia-induced renal fibroblast activation through its antioxidant property to eliminate intracellular ROS, at least in part, via downstream catalase and Nrf2 mechanisms.
Collapse
Affiliation(s)
- Angkhana Nilnumkhum
- a Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital , Mahidol University , Bangkok , Thailand
| | - Rattiyaporn Kanlaya
- a Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital , Mahidol University , Bangkok , Thailand
| | - Sunisa Yoodee
- a Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital , Mahidol University , Bangkok , Thailand
| | - Visith Thongboonkerd
- a Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital , Mahidol University , Bangkok , Thailand
| |
Collapse
|
15
|
Zhu NN, Lu MJ, Chen YQ, Jin XJ, Zhou X, Wei HW, Liu XQ, Duan LS, Yin L, Guo JR. Autologous blood transfusion stimulates wound healing in diabetic mice through activation of the HIF-1α pathway by improving the blood preservation solution. FASEB J 2020; 34:6038-6054. [PMID: 32202355 DOI: 10.1096/fj.201900324rrr] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 12/05/2019] [Accepted: 12/31/2019] [Indexed: 11/11/2022]
Abstract
Transfusion of autologous blood is a timesaving, convenient, safe, and effective therapy from a clinical perspective, and often employed for the treatment of diabetic patients. Stabilization of HIF-1α has been widely reported to be a critical factor in the improvement of wound healing in diabetes. Therefore, our study reveals the roles of improved autologous blood in wound healing in diabetes, through autologous blood transfusion in a mouse model. Initially, BALB/c mice were subjected to streptozotocin for diabetic mouse model establishment. Diabetic mice were transfused with improved or standard autologous blood in perfusion culture system. Roles of improved autologous blood in mediating HIF-1α pathway were determined by measuring expression of VEGF, EGF, HIF-1α, and HSP-90. In order to assess the detailed regulatory mechanism of improved autologous blood in perspective of wound healing, cell proliferation, migration and cell cycle, fibroblasts isolated from diabetic mice were transfected with HIF-1α siRNA. Mice transfused with improved autologous blood exhibited increased levels of CD31 and α-SMA in skin tissues, and reduced TNF-α, IL-1β, and IL-6 levels, indicating that improved autologous blood promoted wound healing ability and reduced the release of inflammatory factors. Diabetic mice transfused with improved autologous blood presented activated HIF-1α pathway. The survival rate, proliferation, and migration of fibroblasts were elevated via activation of the HIF-1α pathway. Taken together, improved blood preservation solution could enhance the oxygen carrying capacity of red blood cells and wound healing in mice with diabetes, which is achieved through regulation of HIF-1α pathway.
Collapse
Affiliation(s)
- Na-Na Zhu
- Department of Anesthesiology, Gongli Hospital, the Second Military Medical University, Shanghai, P. R. China
| | - Ming-Jun Lu
- Department of Anesthesiology, Gongli Hospital, the Second Military Medical University, Shanghai, P. R. China
| | - Yong-Quan Chen
- Department of Anesthesiology, Yijishan Hospital, the Wannan Medical College, Wuhu, P. R. China
| | - Xiao-Ju Jin
- Department of Anesthesiology, Yijishan Hospital, the Wannan Medical College, Wuhu, P. R. China
| | - Xun Zhou
- Department of Anesthesiology, Gongli Hospital, the Second Military Medical University, Shanghai, P. R. China.,Ningxia Medical University, Gongli Hospital of Shanghai Pudong New Area Training Base, Shanghai, P. R. China
| | - Han-Wei Wei
- Department of Anesthesiology, Yijishan Hospital, the Wannan Medical College, Wuhu, P. R. China
| | - Xiao-Qian Liu
- Department of Anesthesiology, Gongli Hospital, the Second Military Medical University, Shanghai, P. R. China
| | - Li-Shuang Duan
- Department of Anesthesiology, Gongli Hospital, the Second Military Medical University, Shanghai, P. R. China
| | - Lei Yin
- Department of Anesthesiology, Gongli Hospital, the Second Military Medical University, Shanghai, P. R. China
| | - Jian-Rong Guo
- Department of Anesthesiology, Gongli Hospital, the Second Military Medical University, Shanghai, P. R. China.,Ningxia Medical University, Gongli Hospital of Shanghai Pudong New Area Training Base, Shanghai, P. R. China
| |
Collapse
|
16
|
Kosol W, Kumar S, Marrero-BerrÍos I, Berthiaume F. Medium conditioned by human mesenchymal stromal cells reverses low serum and hypoxia-induced inhibition of wound closure. Biochem Biophys Res Commun 2020; 522:335-341. [PMID: 31761327 PMCID: PMC10660584 DOI: 10.1016/j.bbrc.2019.11.071] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 11/12/2019] [Indexed: 01/08/2023]
Abstract
Chronic wounds, such as pressure ulcers, are a common complication of impaired peripheral circulation, such as in advanced diabetes. Factors secreted by mesenchymal stromal cells (MSCs) have been shown to enhance wound healing in vitro and in vivo. However, there is little understanding of the impact of the chronic wound environment, namely the limited supply of nutrients and oxygen, on the ability of wound cells to respond to MSCs. In this study, we first established the effects of hypoxia (1% O2) and low serum (1% serum) concentration on the proliferation and migration of keratinocytes. We found that hypoxia and low serum significantly slowed down these processes. Next, we found that supplementation with human MSC-concentrated conditioned media (hMSC-CM) enhanced both cell migration and proliferation in the presence of hypoxia and low serum. Furthermore, low serum and hypoxia decreased cell spreading and F-actin expression, which was reversed in the presence of hMSC-CM. Several wound healing mediators were identified in hMSC-CM, including IL-5, IL-6, IL-8, IL-9, IP-10, MCP-1, FGF-2, and VEGF. This study suggests that the concentrated secretome of human MSCs can reverse the inhibitory effect of hypoxia and low serum on keratinocyte proliferation and migration. This phenomenon may contribute to the beneficial effects of hMSC-CM on wound healing in vivo.
Collapse
Affiliation(s)
- Wilai Kosol
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ, 08854, USA
| | - Suneel Kumar
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ, 08854, USA
| | - Ileana Marrero-BerrÍos
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ, 08854, USA
| | - Francois Berthiaume
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ, 08854, USA.
| |
Collapse
|
17
|
Gutierrez-Pajares JL, Ben Hassen C, Oger C, Galano JM, Durand T, Frank PG. Oxidized Products of α-Linolenic Acid Negatively Regulate Cellular Survival and Motility of Breast Cancer Cells. Biomolecules 2019; 10:biom10010050. [PMID: 31905626 PMCID: PMC7023043 DOI: 10.3390/biom10010050] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 12/24/2019] [Accepted: 12/25/2019] [Indexed: 12/09/2022] Open
Abstract
Despite recent advances in our understanding of the biological processes leading to the development and progression of cancer, there is still a need for new and effective agents to treat this disease. Phytoprostanes (PhytoPs) and phytofurans (PhytoFs) are non-enzymatically oxidized products of α-linolenic acid that are present in seeds and vegetable oils. They have been shown to possess anti-inflammatory and apoptosis-promoting activities in macrophages and leukemia cells, respectively. In this work, seven PhytoPs (PP1–PP7) and one PhytoFs (PF1) were evaluated for their cytotoxic, chemosensitization, and anti-migratory activities using the MCF-7 and MDA-MB-231 breast cancer cell lines. Among the tested compounds, only three PhytoPs had a significant effect on cell viability compared to the control group: Ent-9-L1-PhytoP (PP6) decreased cell viability in both cell lines, while 16-F1t-PhytoP (PP1) and 9-L1-PhytoP (PP5) decreased viability of MCF-7 and MDA-MB-231 cells, respectively. When combined with a sub-cytotoxic dose of doxorubicin, these three PhytoPs displayed significantly enhanced cytotoxic effects on MCF-7 cells while the chemotherapeutic drug alone had no effect. In cellular motility assays, Ent-9-(RS)-12-epi-ST-Δ10-13-PhytoF could significantly inhibit cellular migration of MDA-MB-231 cells. In addition, Ent-9-(RS)-12-epi-ST-Δ10-13-PhytoF also enhanced cellular adhesion of MDA-MB-231 cells.
Collapse
Affiliation(s)
| | - Celine Ben Hassen
- INSERM, Faculté de Médecine, Université de Tours, UMR1069 Tours, France
| | - Camille Oger
- Institut des Biomolécules Max Mousseron (IBMM), Université de Montpellier, CNRS, ENSCM Faculté de Pharmacie, UMR5247 Montpellier, France
| | - Jean-Marie Galano
- Institut des Biomolécules Max Mousseron (IBMM), Université de Montpellier, CNRS, ENSCM Faculté de Pharmacie, UMR5247 Montpellier, France
| | - Thierry Durand
- Institut des Biomolécules Max Mousseron (IBMM), Université de Montpellier, CNRS, ENSCM Faculté de Pharmacie, UMR5247 Montpellier, France
| | - Philippe G Frank
- INSERM, Faculté de Médecine, Université de Tours, UMR1069 Tours, France
| |
Collapse
|
18
|
Effects of an Acetic Acid and Acetone Mixture on the Characteristics and Scaffold–Cell Interaction of Electrospun Polycaprolactone Membranes. APPLIED SCIENCES-BASEL 2019. [DOI: 10.3390/app9204350] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Green electrospinning has attracted great interest since non-toxic solvents were shown to be applicable in the fabrication of fibrous materials while ensuring health safety and environmental protection. Less harmful reagents such as acetone (AC) and acetic acid (AA) have been employed in this field in recent years. However, research in this area is still rare, yielding only preliminary results. In this study, two different types of solvents (pure AC and an AA/AC mixture) were used to fabricate electrospun polycaprolactone (PCL) membranes. Sample morphology, wettability, tensile strength, and chemical composition were compared between two types of membranes. Cell–scaffold interaction was also examined by cell adhesion and proliferation assays. The results demonstrate that the two types of solvents had significant effects on membrane morphology, physical strength, and cell adherence behaviors, which should be considered for different application purposes.
Collapse
|
19
|
Gli signaling pathway modulates fibroblast activation and facilitates scar formation in pulmonary fibrosis. Biochem Biophys Res Commun 2019; 514:684-690. [DOI: 10.1016/j.bbrc.2019.05.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 05/02/2019] [Indexed: 12/20/2022]
|
20
|
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
|
21
|
"Prostate telocytes change their phenotype in response to castration or testosterone replacement". Sci Rep 2019; 9:3761. [PMID: 30842587 PMCID: PMC6403354 DOI: 10.1038/s41598-019-40465-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 02/07/2019] [Indexed: 12/24/2022] Open
Abstract
Telocytes are CD34-positive cells with a fusiform cell body and long, thin cytoplasmic projections called telopodes. These cells were detected in the stroma of various organs, including the prostate. The prostate is a complex gland capable of undergoing involution due to low testosterone levels; and this condition can be reversed with testosterone replacement. Telocyte function in the mature prostate remains to be dermined, and it is not known whether telocytes can take place in tissue remodeling during prostate involution and regrowth. The present study employed structural, ultrastructural and immunohistochemical methods to investigate the telocyte's phenotypes in the ventral prostate (VP) from control (CT), castrated (CS) and testosterone replacement (TR) groups of adult male Wistar rats. Telocytes were found in the subepithelial, perimuscular and interstitical regions around glandular acini. Telocytes from CT animals have condensed chromatin and long and thin telopodes. In CS group, telocytes appeared quiescent and exhibited layers of folded up telopodes. After TR, telocytes presented loose chromatin, abundant rough endoplasmic reticulum and enlarged telopodes, closely associated with bundles of collagen fibrils. We called these cells "telocytes with a synthetic phenotype". As testosterone levels and glandular morphology returned toward to the CT group parameters, after 10 days of TR, these telocytes progressively switched to the normal phenotype. Our results demonstrate that telocytes exhibit phenotypic plasticity upon androgen manipulation and interact with fibroblast and smooth muscle cells to maintain glandular architecture in control animals and during tissue remodeling after hormonal manipulation.
Collapse
|
22
|
Leinhos L, Peters J, Krull S, Helbig L, Vogler M, Levay M, van Belle GJ, Ridley AJ, Lutz S, Katschinski DM, Zieseniss A. Hypoxia suppresses myofibroblast differentiation by changing RhoA activity. J Cell Sci 2019; 132:jcs223230. [PMID: 30659117 DOI: 10.1242/jcs.223230] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 01/09/2019] [Indexed: 12/15/2022] Open
Abstract
Fibroblasts show a high range of phenotypic plasticity, including transdifferentiation into myofibroblasts. Myofibroblasts are responsible for generation of the contraction forces that are important for wound healing and scar formation. Overactive myofibroblasts, by contrast, are involved in abnormal scarring. Cell stretching and extracellular signals such as transforming growth factor β can induce the myofibroblastic program, whereas microenvironmental conditions such as reduced tissue oxygenation have an inhibitory effect. We investigated the effects of hypoxia on myofibroblastic properties and linked this to RhoA activity. Hypoxia reversed the myofibroblastic phenotype of primary fibroblasts. This was accompanied by decreased αSMA (ACTA2) expression, alterations in cell contractility, actin reorganization and RhoA activity. We identified a hypoxia-inducible induction of ARHGAP29, which is critically involved in myocardin-related transcription factor-A (MRTF-A) signaling, the differentiation state of myofibroblasts and modulates RhoA activity. This novel link between hypoxia and MRTF-A signaling is likely to be important for ischemia-induced tissue remodeling and the fibrotic response.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Lisa Leinhos
- Institute of Cardiovascular Physiology, University Medical Center, Georg-August University Göttingen, 37073 Göttingen, Germany
| | - Johannes Peters
- Institute of Cardiovascular Physiology, University Medical Center, Georg-August University Göttingen, 37073 Göttingen, Germany
| | - Sabine Krull
- Institute of Cardiovascular Physiology, University Medical Center, Georg-August University Göttingen, 37073 Göttingen, Germany
| | - Lena Helbig
- Institute of Cardiovascular Physiology, University Medical Center, Georg-August University Göttingen, 37073 Göttingen, Germany
| | - Melanie Vogler
- Institute of Cardiovascular Physiology, University Medical Center, Georg-August University Göttingen, 37073 Göttingen, Germany
| | - Magdolna Levay
- Experimental Pharmacology, European Center of Angioscience, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Gijsbert J van Belle
- Institute of Cardiovascular Physiology, University Medical Center, Georg-August University Göttingen, 37073 Göttingen, Germany
| | - Anne J Ridley
- Randall Centre of Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| | - Susanne Lutz
- Institute of Pharmacology and Toxicology, University Medical Center, Georg-August University Göttingen, 37075 Göttingen, Germany
| | - Dörthe M Katschinski
- Institute of Cardiovascular Physiology, University Medical Center, Georg-August University Göttingen, 37073 Göttingen, Germany
| | - Anke Zieseniss
- Institute of Cardiovascular Physiology, University Medical Center, Georg-August University Göttingen, 37073 Göttingen, Germany
| |
Collapse
|
23
|
Qiu S, Jia Y, Sun Y, Han P, Xu J, Wen G, Chai Y. Von Hippel-Lindau (VHL) Protein Antagonist VH298 Improves Wound Healing in Streptozotocin-Induced Hyperglycaemic Rats by Activating Hypoxia-Inducible Factor- (HIF-) 1 Signalling. J Diabetes Res 2019; 2019:1897174. [PMID: 30911550 PMCID: PMC6398031 DOI: 10.1155/2019/1897174] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 11/01/2018] [Accepted: 11/29/2018] [Indexed: 01/04/2023] Open
Abstract
AIMS The purpose of the present research is to investigate the effects of the VHL protein antagonist, VH298, on functional activities of fibroblasts and vascular endothelial cells and the effects on the wound healing process in a streptozotocin-induced hyperglycaemic rat model. METHODS HIF-1α and hydroxy-HIF-1α protein levels in VH298-treated rat fibroblasts (rFb) were measured by immunoblotting, rFb proliferation was detected by the CCK-8 assay, and mRNA levels of related genes were measured by quantitative RT-PCR. In vitro wound healing was simulated by the scratch test; angiogenesis was measured by the human umbilical vein endothelial cell (hUVEC) tube formation assay. VH298 or PBS was locally injected into wounds in rat models with streptozotocin- (STZ-) induced hyperglycaemia, the wound tissues were harvested, and haematoxylin-eosin (HE) and Masson trichrome staining and immunohistochemical processes were conducted. RESULTS HIF-1α and hydroxy-HIF-1α levels increased in VH298-treated rFb, in a time- and dose-dependent manner. Thirty micromolar VH298 could significantly increase cell proliferation, angiogenesis, and gene expression of type I collagen-α1 (Col1-α1), vascular endothelial growth factor A (VEGF-A), and insulin-like growth factor 1 (IGF-1). The VH298-treated wound had a better healing pattern, activation of HIF-1 signalling, and vascularization. CONCLUSIONS Taken together, VH298 activated the HIF-1 signalling pathway by stabilizing both HIF-1α and hydroxy-HIF-1α. VH298 enhanced rFb functions, promoted hUVEC angiogenesis, and accelerated wound healing in the rat model mimicking diabetes mellitus.
Collapse
Affiliation(s)
- Shuo Qiu
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yachao Jia
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yunchu Sun
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Pei Han
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Jia Xu
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Gen Wen
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yimin Chai
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
24
|
Celikkan FT, Mungan C, Sucu M, Ulus AT, Cinar O, Ili EG, Can A. Optimizing the transport and storage conditions of current Good Manufacturing Practice -grade human umbilical cord mesenchymal stromal cells for transplantation (HUC-HEART Trial). Cytotherapy 2018; 21:64-75. [PMID: 30455106 DOI: 10.1016/j.jcyt.2018.10.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 10/01/2018] [Accepted: 10/13/2018] [Indexed: 01/12/2023]
Abstract
BACKGROUND The HUC-HEART Trial is a clinical study of intramyocardial delivery of current Good Manufacturing Practice (cGMP)-grade human umbilical cord multipotent stromal cells (HUC-MSCs) in ischemic cardiomyopathy where 2 × 107 cells are administered to peri-infarcted myocardium. Prior to the onset of the trial, we aimed to optimize the transport/storage conditions for obtaining the highest cell viability and proliferation rate of cells to be transplanted. METHODS Cells were tested after being transported in phosphate-buffered saline (PBS) or Ringer's lactate-based (RL) transport media supplemented with human serum albumin (HSA) and/or hydroxyethyl starch (HES) at two temperatures (2-10°C or 22-24°C). RESULTS The effects of transport conditions on cell viability following 6 h were found highest (93.4 ± 1.5) in RL-based media at 2-10°C. Karyotypes were found normal upon transportation in any of the formulations and temperatures. However, the highest proliferation rate was noted (3.1-fold increase) in RL (1% HSA) media at 2-10°C over 6 days in culture. From that point, RL (1% HSA) media at 2-10°C was used for further experiments. The maximum cell storage time was detected around 24 h at 2-10°C. Extended storage periods resulted in a decrease in cell viability but not in MSC marker expression. An increase in actin quantity was detected in hypoxia (5% O2) groups in early culture days; no difference was noted between hypoxic versus normoxic (21% O2) conditions in later days. DISCUSSION The overall results suggest that non-commercial, simple media formulations with extended storage intervals at 2-10°C temperatures are capable of retaining the characteristics of clinical-grade HUC-MSCs. The above findings led us to use RL (1% HSA) media at 2-10°C for transport and storage in the HUC-HEART Trial; 23 patients received HUC-MSCs by August 2018; no adverse effects were noted related to cell processing and transplantation.
Collapse
Affiliation(s)
- Ferda Topal Celikkan
- Department of Histology and Embryology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Ceren Mungan
- Ankara University Biotechnology Institute, Ankara, Turkey
| | - Merve Sucu
- Ankara University Biotechnology Institute, Ankara, Turkey
| | - A Tulga Ulus
- Division of Cardiovascular Surgery, Acibadem Hospital, Ankara, Turkey
| | - Ozgur Cinar
- Department of Histology and Embryology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Ezgi Gokpinar Ili
- Department of Medical Genetics, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Alp Can
- Department of Histology and Embryology, Faculty of Medicine, Ankara University, Ankara, Turkey.
| |
Collapse
|
25
|
Chhabra V, Anand AS, Baidya AK, Malik SM, Kohli E, Reddy MPK. Hypobaric hypoxia induced renal damage is mediated by altering redox pathway. PLoS One 2018; 13:e0195701. [PMID: 30005088 PMCID: PMC6044529 DOI: 10.1371/journal.pone.0195701] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 03/28/2018] [Indexed: 12/29/2022] Open
Abstract
Systemic hypobaric hypoxia is reported to cause renal damage; nevertheless the exact pathophysiological mechanisms are not completely understood. Therefore, the present study aims to explore renal pathophysiology by using proteomics approach under hypobaric hypoxia. Six to eight week old male Sprague Dawley rats were exposed to hypobaric hypoxia equivalent to altitude of 7628 metres (pO2-282mmhg) at 28°C and 55% humidity in decompression chamber for different time intervals; 1, 3, and7 days. Various physiological, proteomic and bioinformatic studies were carried out to examine the effect of chronic hypobaric hypoxia on kidney. Our data demonstrated mild to moderate degenerative tubular changes, altered renal function, injury biomarkers and systolic blood pressure with increase in duration of hypobaric hypoxia exposure. Renal proteomic analysis showed 38 differential expressed spots, out of which 25 spots were down regulated and 13 were up regulated in 7 dayhypobarichypoxic exposure group of rats as compared to normoxia control. Identified proteins were involved in specific molecular changes pertinent to endogenous redox pathways, cellular integrity and energy metabolism. The study provides an empirical evidence of renal homeostasis under hypobaric hypoxia by investigating both physiological and proteomics changes. The identification of explicit key proteins provides a valuable clue about redox signalling mediated renal damage under hypobaric hypoxia.
Collapse
Affiliation(s)
- Varun Chhabra
- Cardio-Respiratory Division, Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Development Organisation (DRDO), Ministry of Defence, Timarpur, Delhi, India
| | - Avnika Singh Anand
- Neurobiology Division, Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Development Organisation (DRDO), Ministry of Defence, Timarpur, Delhi, India
| | - Amit Kumar Baidya
- Cardio-Respiratory Division, Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Development Organisation (DRDO), Ministry of Defence, Timarpur, Delhi, India
| | - Shajer Manzoor Malik
- Cardio-Respiratory Division, Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Development Organisation (DRDO), Ministry of Defence, Timarpur, Delhi, India
| | - Ekta Kohli
- Neurobiology Division, Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Development Organisation (DRDO), Ministry of Defence, Timarpur, Delhi, India
| | - Maramreddy Prasanna Kumar Reddy
- Cardio-Respiratory Division, Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Development Organisation (DRDO), Ministry of Defence, Timarpur, Delhi, India
- * E-mail: ,
| |
Collapse
|
26
|
Moen I, Ugland H, Strömberg N, Sjöström E, Karlson A, Ringstad L, Bysell H, Amiry‐Moghaddam M, Haglerød C. Development of a novel in situ gelling skin dressing: Delivering high levels of dissolved oxygen at pH 5.5. Health Sci Rep 2018; 1:e57. [PMID: 30623087 PMCID: PMC6266454 DOI: 10.1002/hsr2.57] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 04/05/2018] [Accepted: 05/16/2018] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND AND AIMS Wound healing requires appropriate oxygen and pH levels. Oxygen therapy and pH-modulating treatments have shown positive effects on wound healing. Thus, a dressing, which combines high levels of dissolved oxygen (DO) with the pH of intact skin, may improve wound healing. Our aims were to (1) formulate an in situ gelling dressing with high levels of DO and with the pH level of intact skin, (2) evaluate physical and chemical properties of the dressing, and (3) elucidate basic effects of elevated levels of DO on human skin cells in vitro. METHODS A dressing was formulated with 15 to 16 wt% poloxamer 407, acetate buffer, and oxygenated water. Stability of pH and DO, rheology, and shelf life were analysed. Furthermore, in vitro studies of the effect of increased levels of DO were performed. RESULTS An in situ gelling wound dressing, with a DO concentration ranging between 25 and 35 mg/L and a pH of 5.5, was formulated. The DO concentration was stable above 22 mg/L for at least 30 hours when applied on a surface at 35°C and covered for directed diffusion into the intended wound area. At storage, the dressing had stable pH for 3 months and stable DO concentration over 30 mg/L for 7 weeks. Increasing DO significantly enhanced intracellular ATP in human skin cells, without changing reactive oxygen species production, proliferation rate, or viability. CONCLUSION The developed dressing may facilitate wound healing by delivering controlled and stable oxygen levels, providing adjustable pH for optimized healing, and increasing intracellular ATP availability.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Mahmood Amiry‐Moghaddam
- Oxy Solutions ASOsloNorway
- Laboratory of Molecular Neuroscience, Department of Molecular Medicine, Institute of Basic Medical SciencesUniversity of OsloNorway
| | | |
Collapse
|
27
|
Gaur P, Saini S, Vats P, Kumar B. Regulation, signalling and functions of hormonal peptides in pulmonary vascular remodelling during hypoxia. Endocrine 2018; 59:466-480. [PMID: 29383676 DOI: 10.1007/s12020-018-1529-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 01/10/2018] [Indexed: 01/06/2023]
Abstract
Hypoxic state affects organism primarily by decreasing the amount of oxygen reaching the cells and tissues. To adjust with changing environment organism undergoes mechanisms which are necessary for acclimatization to hypoxic stress. Pulmonary vascular remodelling is one such mechanism controlled by hormonal peptides present in blood circulation for acclimatization. Activation of peptides regulates constriction and relaxation of blood vessels of pulmonary and systemic circulation. Thus, understanding of vascular tone maintenance and hypoxic pulmonary vasoconstriction like pathophysiological condition during hypoxia is of prime importance. Endothelin-1 (ET-1), atrial natriuretic peptide (ANP), and renin angiotensin system (RAS) function, their receptor functioning and signalling during hypoxia in different body parts point them as disease markers. In vivo and in vitro studies have helped understanding the mechanism of hormonal peptides for better acclimatization to hypoxic stress and interventions for better management of vascular remodelling in different models like cell, rat, and human is discussed in this review.
Collapse
Affiliation(s)
- Priya Gaur
- Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi, India
| | - Supriya Saini
- Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi, India
| | - Praveen Vats
- Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi, India.
| | - Bhuvnesh Kumar
- Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi, India
| |
Collapse
|
28
|
Swaminathan A, Kasiviswanathan D, Balaguru UM, Kolluru GK, SuryaKumar G, Chatterjee S. Hypoxia perturbs endothelium by re-organizing cellular actin architecture: Nitric oxide offers limited protection. Tissue Cell 2018; 50:114-124. [PMID: 29429511 DOI: 10.1016/j.tice.2017.12.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 12/09/2017] [Accepted: 12/18/2017] [Indexed: 11/26/2022]
Abstract
Exposure to hypoxia causes structural changes in the endothelial cell (EC) monolayer that alter its permeability. There was a report earlier of impairment of nitric oxide (NO) production in endothelium. The intervention of NO in the altered cellular arrangements of actin cytoskeleton in endothelium for rectification of paracellular gaps in endothelium under hypoxia was observed. The present study demonstrates hypoxia inducing paracellular gaps in hypoxia-exposed blood capillaries in chick embryo extravascular model. Phalloidin staining confirmed significant polymerization of actin and unique cellular localization of the F-actin bands under hypoxia treatments. Addition of spermine NONOate (SPNO), a NO donor, or reoxygenation to endothelial monolayer attenuated hypoxia-mediated effects on endothelial permeability with partial recovery of endothelial integrity through actin remodeling. The present study indicates link of hypoxia-induced actin-associated cytoskeletal rearrangements and paracellular gaps in the endothelium with a low NO availability in the hypoxia milieu. The author concludes that NO confers protection against hypoxia-mediated cytoskeletal remodeling and endothelial leakiness.
Collapse
Affiliation(s)
- Akila Swaminathan
- Vascular Biology Lab, AU-KBC Research Centre, MIT Campus of Anna University, Chennai, India
| | | | - Uma Maheswari Balaguru
- Vascular Biology Lab, AU-KBC Research Centre, MIT Campus of Anna University, Chennai, India
| | | | - Geetha SuryaKumar
- Defence Institute of Physiology and Allied Sciences, DIPAS, Delhi, India.
| | - Suvro Chatterjee
- Vascular Biology Lab, AU-KBC Research Centre, MIT Campus of Anna University, Chennai, India; Department of Biotechnology, Anna University, Chennai, India.
| |
Collapse
|
29
|
Lasocka I, Szulc-Dąbrowska L, Skibniewski M, Skibniewska E, Strupinski W, Pasternak I, Kmieć H, Kowalczyk P. Biocompatibility of pristine graphene monolayer: Scaffold for fibroblasts. Toxicol In Vitro 2018; 48:276-285. [PMID: 29409908 DOI: 10.1016/j.tiv.2018.01.028] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 12/29/2017] [Accepted: 01/30/2018] [Indexed: 12/29/2022]
Abstract
The aim of the present study was to evaluate the cytotoxicity of pristine graphene monolayer and its utility as a scaffold for murine fibroblast L929 cell line. Cell viability, morphology, cytoskeleton architecture (microfilaments and microtubules), cell adhesion and migration into the scratch-wound area were determined using pristine graphene-coated microscopic slides. We found that fibroblasts cultured on pristine graphene monolayer exhibited changes in cell attachment, motility and cytoskeleton organization. Graphene was found to have no cytotoxicity on L929 fibroblasts and increased cell adhesion and proliferation within 24 h of culture. The area of cells growing on graphene was comparable to the area of fibroblasts cultured on glass. Migration of cells on the surface of graphene substrate appeared to be more regular in comparison to uncoated glass surface, however in both control (glass) and experimental (graphene) groups the scratch wound was closed after 48 h of culture. Taken together, our results indicate that pristine graphene monolayer is non-toxic for murine subcutaneous connective tissue fibroblasts and could be beneficial for recovery of damaged tissues after injury. These studies could be helpful in evaluating biocompatibility of graphene, which still remains ambiguous.
Collapse
Affiliation(s)
- Iwona Lasocka
- Department of Biology of Animal Environment, Faculty of Animal Science, Warsaw University of Life Sciences, Ciszewskiego street 8, 02-786 Warsaw, Poland
| | - Lidia Szulc-Dąbrowska
- Department of Preclinical Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences, Ciszewskiego street 8, 02-786 Warsaw, Poland
| | - Michał Skibniewski
- Department of Morphological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences, Nowoursynowska street 159, 02-776 Warsaw, Poland.
| | - Ewa Skibniewska
- Department of Biology of Animal Environment, Faculty of Animal Science, Warsaw University of Life Sciences, Ciszewskiego street 8, 02-786 Warsaw, Poland
| | - Włodzimierz Strupinski
- Institute of Electronic Materials Technology, Wólczyńska street 133, 01-919 Warsaw, Poland; Faculty of Physics, Warsaw University of Technology, Koszykowa street 75, 00-662 Warsaw, Poland
| | - Iwona Pasternak
- Institute of Electronic Materials Technology, Wólczyńska street 133, 01-919 Warsaw, Poland; Faculty of Physics, Warsaw University of Technology, Koszykowa street 75, 00-662 Warsaw, Poland
| | - Hubert Kmieć
- Department of Biology of Animal Environment, Faculty of Animal Science, Warsaw University of Life Sciences, Ciszewskiego street 8, 02-786 Warsaw, Poland
| | - Paweł Kowalczyk
- Department of Animal Nutrition, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka street 3, 05-110 Jabłonna, Poland
| |
Collapse
|
30
|
Hypoxia regulates RhoA and Wnt/β-catenin signaling in a context-dependent way to control re-differentiation of chondrocytes. Sci Rep 2017; 7:9032. [PMID: 28831110 PMCID: PMC5567364 DOI: 10.1038/s41598-017-09505-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 07/27/2017] [Indexed: 01/16/2023] Open
Abstract
Cartilage tissue is avascular and hypoxic which regulates chondrocyte phenotype via stabilization of HIFs. Here, we investigated the role of hypoxia and HIFs in regulation of Rho and canonical Wnt signaling in chondrocytes. Our data demonstrates that hypoxia controls the expression of RhoA in chondrocytes in a context-dependent manner on the culturing conditions. Within a 3D microenvironment, hypoxia suppresses RhoA on which hypoxia-driven expression of chondrogenic markers depends. Conversely, hypoxia leads to upregulation of RhoA in chondrocytes on 2D with a failure in re-expression of chondrogenic markers. Similarly to RhoA, hypoxic regulation of Wnt/β-catenin signaling depends on the microenvironment. Hypoxia downregulates β-catenin within 3D hydrogels whereas it causes a potent increase on 2D. Hypoxia-induced suppression of canonical Wnt signaling in 3D contributes to the promotion of chondrogenic phenotype as induction of Wnt signaling abrogates the hypoxic re-differentiation of chondrocytes. Inhibiting Wnt/β-catenin signaling via stabilization of Axin2 leads to a synergistic enhancement of hypoxia-induced expression of chondrogenic markers. The effects of hypoxia on Rho and Wnt/β-catenin signaling are HIF-dependent as stabilizing HIFs under normoxia revealed similar effects on chondrocytes. The study reveals important insights on hypoxic signaling of chondrocytes and how hypoxia regulates cellular mechanisms depending on the cellular microenvironment.
Collapse
|
31
|
Khakshour S, Labrecque MP, Esmaeilsabzali H, Lee FJS, Cox ME, Park EJ, Beischlag TV. Retinoblastoma protein (Rb) links hypoxia to altered mechanical properties in cancer cells as measured by an optical tweezer. Sci Rep 2017; 7:7833. [PMID: 28798482 PMCID: PMC5552853 DOI: 10.1038/s41598-017-07947-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 07/06/2017] [Indexed: 12/18/2022] Open
Abstract
Hypoxia modulates actin organization via multiple pathways. Analyzing the effect of hypoxia on the biophysical properties of cancer cells is beneficial for studying modulatory signalling pathways by quantifying cytoskeleton rearrangements. We have characterized the biophysical properties of human LNCaP prostate cancer cells that occur in response to loss of the retinoblastoma protein (Rb) under hypoxic stress using an oscillating optical tweezer. Hypoxia and Rb-loss increased cell stiffness in a fashion that was dependent on activation of the extracellular signal-regulated kinase (ERK) and the protein kinase B (AKT)- mammalian target of rapamycin (MTOR) pathways. Pharmacological inhibition of MEK1/2, AKT or MTOR impeded hypoxia-inducible changes in the actin cytoskeleton and inhibited cell migration in Rb-deficient cells conditioned with hypoxia. These results suggest that loss of Rb in transformed hypoxic cancer cells affects MEK1/2-ERK/AKT-MTOR signalling and promotes motility. Thus, the mechanical characterization of cancer cells using an optical tweezer provides an additional technique for cancer diagnosis/prognosis and evaluating therapeutic performance.
Collapse
Affiliation(s)
- S Khakshour
- School of Mechatronic Systems Engineering, Faculty of Applied Sciences, Simon Fraser University, Surrey, BC, Canada
| | - M P Labrecque
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, Canada
| | - H Esmaeilsabzali
- School of Mechatronic Systems Engineering, Faculty of Applied Sciences, Simon Fraser University, Surrey, BC, Canada
| | - F J S Lee
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, Canada
| | - M E Cox
- Department of Urologic Sciences, The Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - E J Park
- School of Mechatronic Systems Engineering, Faculty of Applied Sciences, Simon Fraser University, Surrey, BC, Canada. .,Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, Canada.
| | - T V Beischlag
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, Canada.
| |
Collapse
|
32
|
Mathew SA, Chandravanshi B, Bhonde R. Hypoxia primed placental mesenchymal stem cells for wound healing. Life Sci 2017. [PMID: 28625360 DOI: 10.1016/j.lfs.2017.06.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
AIMS To investigate how Placental Mesenchymal Stem Cells (P-MSCs) would adapt themselves and survive under hypoxic conditions which are prevalent in most injury sites. MAIN METHODS P-MSCs were isolated from term placenta and characterised under normoxia and hypoxia (2-2.5% O2). Cells were examined for morphology and surface marker variations by flow cytometry analysis. Glucose stimulated insulin secretion was assayed by Insulin ELISA Kit. Gene expression levels were estimated using Real Time PCR for hypoxia inducible factor1 alpha, Insulin (INS), Glucose transporters (GLUT-1, GLUT-2 and GLUT-3), Adhesion Proteins- Integrins, Fibronectin1 (FN1), E-Cadherin (CDH1), and N-Cadherin (CDH2) and angiogenesis marker VEGFA. Immunofluorescence assay was done to confirm the presence of C-Peptide, GLUT 2, E-Cadherin and ITGB3. Adhesion was confirmed assessed on fibronectin binding. KEY FINDINGS We show that insulin secretion is not hampered under hypoxia. We found an upregulation of glucose transporters under hypoxia indicating enhanced glucose uptake needed to cater to metabolic demands of proliferating cells. Up regulation of adhesion molecules was seen under hypoxia indicative of a favoured environment for retention of cells at the injury site. We also found increased level of angiogenesis of P-MSCs under hypoxia. SIGNIFICANCE Our present study thus demonstrates for the first time that P-MSCs modulate themselves under hypoxic conditions by secreting insulin, up regulating glucose transporters and adhesion molecules and eventually exhibiting an increased angiogenic potential. We thus infer that priming P-MSCs under hypoxia, could make them more suitable for wound healing applications.
Collapse
Affiliation(s)
- Suja Ann Mathew
- School of Regenerative Medicine, Manipal University, MAHE, GKVK Post, Bellary Road, Allalasandra, Near Royal Orchid, Yelahanka, Bangalore 560 065, India
| | - Bhawna Chandravanshi
- School of Regenerative Medicine, Manipal University, MAHE, GKVK Post, Bellary Road, Allalasandra, Near Royal Orchid, Yelahanka, Bangalore 560 065, India
| | - Ramesh Bhonde
- Dr D Y Patil University, Sant Tukaram Nagar, Maharashtra, Pune 411018, India.
| |
Collapse
|
33
|
Chaudhary A, Bag S, Banerjee P, Chatterjee J. Honey Extracted Polyphenolics Reduce Experimental Hypoxia in Human Keratinocytes Culture. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:3460-3473. [PMID: 28406019 DOI: 10.1021/acs.jafc.7b00366] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Hypoxic assault affects fundamental cellular processes and generates oxidative stress on healthy cells/molecules. Honey extracted polyphenolics (HEP) as a natural antioxidant reduced hypoxic cytotoxicity in this study. Different honey samples were physicochemically characterized to identify preferred (jamun) honey [pH 3.55 ± 0.04, conductivity (μs/cm) = 6.66 ± 0.14, water content % (w/w) = 14.70 ± 0.35, total solid content % (w/w) = 85.30 ± 0.35, phenol content (mg GAE/100 g) = 403.55 ± 0.35, flavonoid content (mg QE/100 g) = 276.76 ± 4.10, radical scavenging activity (% 500 μL) = 147.75 ± 3.13, catalase activity (absorbance at 620 nm) = 0.226 ± 0.01]. HEP was tested in different doses on hypoxic and normoxic cells (HaCaT) using viability and antioxidant assays. Cardinal molecular expressions such as cadherin-catenin-cytoskeleton complex (namely, E-cadherin, β-catenin, and F-actin), hypoxia marker (Hif 1 α), proliferation marker (Ki67), and epithelial master regulator (p63) were studied by immuno-cytochemisty (ICC) and qRT-PCR. The 0.063 mg/mL HEP demonstrated better vitality and functionality of HaCaT cells as per viability assay (*, P < 0.01) even under hypoxia. ICC and qRT-PCR observations indicated restoration of cellular survival and homeostasis under 0.063 mg/mL HEP after hypoxic assault. Furthermore, major spectral changes for nucleic acid and membrane phospholipid reorganizations by Fourier transform infrared spectroscopy illustrated a positive impact of 0.063 mg/mL HEP on hypoxic cells considering proliferation and cellular integrity. It was concluded that a specific dose of jamun HEP reduces hypoxic cytotoxicity.
Collapse
Affiliation(s)
- Amrita Chaudhary
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur , Kharagpur 721302, West Bengal, India
| | - Swarnendu Bag
- Department of Biotechnology, National Institute of Technology Sikkim , Barfung Block Ravangla Sub-Division South Sikkim, Ravangla 737139, Sikkim, India
| | - Provas Banerjee
- Banerjees' Biomedical Research Foundation , Sainthia, Birbhum 731234, West Bengal, India
| | - Jyotirmoy Chatterjee
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur , Kharagpur 721302, West Bengal, India
| |
Collapse
|
34
|
MBD3 mediates epigenetic regulation on EPAS1 promoter in cancer. Tumour Biol 2016; 37:13455-13467. [PMID: 27465550 DOI: 10.1007/s13277-016-5237-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 07/15/2016] [Indexed: 10/21/2022] Open
Abstract
Hypoxia-inducible factor 2α (HIF2α) plays critical roles in cancer progression. Although the mechanisms of HIF2α translation and degradation have been well studied, the mechanism for HIF2α regulation at transcriptional level is still not fully understood. Here, we present evidence that DNA methylation in promoter contributes to transcription of EPAS1 coding HIF2α. Methylated CpG binding protein 3 (MBD3) contributes to the intricate regulatory mechanism. We showed that MBD3 bound to the EPAS1 promoter in breast cancer cells and amplified EPAS1 transcription through demethylating CpG located around transcriptional start site in MDA-MB-468 cells. This enabled MDA-MB-468 cells to activate HIF2α-mediated angiogenesis. However, in 7860 cells, the demethylation function of MBD3 on EPAS1 was not observed because of the poor methylated-CpG promoter. Nevertheless, depletion of MBD3 induced by shRNA decreased EPAS1 transcription and therefore decreased HIF2α-mediated cellular response in both MDA-MB-468 and 7860 cancer cells. These results indicated that the endogenous MBD3 was involved in regulating the transcription and therefore the transcriptional activities of HIF2α, suggesting that MBD3 may be a potential therapeutic target of tumor.
Collapse
|
35
|
Hoogewijs D, Vogler M, Zwenger E, Krull S, Zieseniss A. Oxygen-dependent regulation of aquaporin-3 expression. HYPOXIA 2016; 4:91-97. [PMID: 27800511 PMCID: PMC5085303 DOI: 10.2147/hp.s97681] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The purpose of this study was to investigate whether aquaporin-3 (AQP3) expression is altered in hypoxia and whether hypoxia-inducible transcription factor (HIF)-1 regulates the hypoxic expression. AQP3 mRNA expression was studied in L929 fibrosarcoma cells and in several tissues derived from mice that were subjected to hypoxia. Computational analysis of the AQP3 promoter revealed conserved HIF binding sites within close proximity to the translational start site, and chromatin immunoprecipitation assays confirmed binding of HIF-1α to the endogenous hypoxia response elements. Furthermore, hypoxia resulted in increased expression of AQP3 mRNA in L929 fibrosarcoma cells. Consistently, shRNA-mediated knockdown of HIF-1α greatly reduced the hypoxic induction of AQP3. In addition, mRNA analysis of organs from mice exposed to inspiratory hypoxia demonstrated pronounced hypoxia-inducible expression of AQP3 in the kidney. Overall, our findings suggest that AQP3 expression can be regulated at the transcriptional level and that AQP3 represents a novel HIF-1 target gene.
Collapse
Affiliation(s)
- David Hoogewijs
- Institute of Physiology, University of Duisburg-Essen, Essen, Germany; Institute of Physiology, University of Zürich, Zürich, Switzerland
| | - Melanie Vogler
- Institute of Cardiovascular Physiology, University Medical Center Göttingen, University of Göttingen, Göttingen, Germany
| | - Eveline Zwenger
- Institute of Cardiovascular Physiology, University Medical Center Göttingen, University of Göttingen, Göttingen, Germany
| | - Sabine Krull
- Institute of Cardiovascular Physiology, University Medical Center Göttingen, University of Göttingen, Göttingen, Germany
| | - Anke Zieseniss
- Institute of Cardiovascular Physiology, University Medical Center Göttingen, University of Göttingen, Göttingen, Germany
| |
Collapse
|
36
|
Rho-Associated Kinase Inhibitors Promote Microglial Uptake Via the ERK Signaling Pathway. Neurosci Bull 2016; 32:83-91. [PMID: 26779919 DOI: 10.1007/s12264-016-0013-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 11/26/2015] [Indexed: 12/17/2022] Open
Abstract
Microglia are immunocompetent cells in the central nervous system that take up tissue debris and pathogens. Rho-associated kinase (ROCK) has been identified as an important regulator of uptake, proliferation, secretion, and differentiation in a number of cell types. Although ROCK plays critical roles in the microglial secretion of inflammatory factors, migration, and morphology, its effects on microglial uptake activity have not been well characterized. In the present study, we found that treatment of BV2 microglia and primary microglia with the ROCK inhibitors Y27632 and fasudil increased uptake activity and was associated with morphological changes. Furthermore, western blots showed that this increase in uptake activity was mediated through the extracellular-signal-regulated kinase (ERK) signaling cascade, indicating the importance of ROCK in regulating microglial uptake activity.
Collapse
|
37
|
Glass JJ, Phillips PA, Gunning PW, Stehn JR. Hypoxia alters the recruitment of tropomyosins into the actin stress fibres of neuroblastoma cells. BMC Cancer 2015; 15:712. [PMID: 26475688 PMCID: PMC4608101 DOI: 10.1186/s12885-015-1741-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 10/09/2015] [Indexed: 01/27/2023] Open
Abstract
Background Neuroblastoma is the most common extracranial solid tumor of childhood. The heterogeneous microenvironment of solid tumors contains hypoxic regions associated with poor prognosis and chemoresistance. Hypoxia implicates the actin cytoskeleton through its essential roles in motility, invasion and proliferation. However, hypoxia-induced changes in the actin cytoskeleton have only recently been observed in human cells. Tropomyosins are key regulators of the actin cytoskeleton and we hypothesized that tropomyosins may mediate hypoxic phenotypes. Methods Neuroblastoma (SH-EP) cells were incubated ± hypoxia (1 % O2, 5 % CO2) for up to 144 h, before examining the cytoskeleton by confocal microscopy and Western blotting. Results Hypoxic cells were characterized by a more organized actin cytoskeleton and a reduced ability to degrade gelatin substrates. Hypoxia significantly increased mean actin filament bundle width (72 h) and actin filament length (72–96 h). This correlated with increased hypoxic expression and filamentous organization of stabilizing tropomyosins Tm1 and Tm2. However, isoform specific changes in tropomyosin expression were more evident at 96 h. Conclusions This study demonstrates hypoxia-induced changes in the recruitment of high molecular weight tropomyosins into the actin stress fibres of a human cancer. While hypoxia induced clear changes in actin organization compared with parallel normoxic cultures of neuroblastoma, the precise role of tropomyosins in this hypoxic actin reorganization remains to be determined. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1741-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Joshua J Glass
- Oncology Research Unit, School of Medical Sciences, UNSW Australia, Room 229, Wallace Wurth Building, Sydney, NSW, 2052, Australia. .,Current address: ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, 3010, Australia.
| | - Phoebe A Phillips
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, UNSW Australia, Sydney, NSW, 2052, Australia.
| | - Peter W Gunning
- Oncology Research Unit, School of Medical Sciences, UNSW Australia, Room 229, Wallace Wurth Building, Sydney, NSW, 2052, Australia.
| | - Justine R Stehn
- Oncology Research Unit, School of Medical Sciences, UNSW Australia, Room 229, Wallace Wurth Building, Sydney, NSW, 2052, Australia.
| |
Collapse
|
38
|
Zhu T, Park HC, Son KM, Yang HC. Effects of dimethyloxalylglycine on wound healing of palatal mucosa in a rat model. BMC Oral Health 2015; 15:60. [PMID: 25981588 PMCID: PMC4434535 DOI: 10.1186/s12903-015-0047-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 05/13/2015] [Indexed: 12/13/2022] Open
Abstract
Background Rapid wound healing of oral soft tissue may reduce the opportunity of infection and discomfort of patients. Previous studies have demonstrated that enhancement of angiogenesis is an effective way to accelerate wound repair. In this study, to enhance angiogenesis and healing of palatal wounds, dimethyloxalylglycine (DMOG) was applied to a rat palatal wound model. DMOG is known to inhibit oxygen-dependent degradation of hypoxia inducible factor-1 alpha (HIF-1α), which can lead to up-regulation of angiogenesis markers, favoring wound repair. We also evaluated the effects of DMOG on cell migration and HIF-1α expression of rat palatal (RP) cells. Furthermore, mRNA and protein expression of vascular endothelial growth factor (VEGF) were analyzed in DMOG-treated RP cells. Methods Primary cultures of rat palatal (RP) cells were obtained from Sprague–Dawley (SD) rats. Effects of DMOG on cell viability and migration of RP cells were evaluated by using a formazan and culture insert, respectively. VEGF mRNA was observed by real-time PCR, and VEGF and HIF-1α proteins were detected by Western blotting. For the animal study, excisional wounds, 3 mm in diameter, were made at the central part of the palate of SD rats. DMOG with hyaluronic acid ointment was topically applied three times during 1 week, and then wound closures were quantitated photographically and histologically. Results DMOG was cytotoxic to RP cells at concentrations higher than 2 mM and did not affect cell migration at non-cytotoxic concentrations. mRNA and protein expression of VEGF were significantly stimulated by DMOG treatment. The protein level of HIF-1α was also stabilized in RP cells by DMOG. In the animal study, groups treated with 1 mg/ml DMOG showed an increase of rat palatal wound contractures. Conclusions DMOG enhanced wound healing of rat palatal mucosa, which was likely due to the angiogenic effect of the agent.
Collapse
Affiliation(s)
- Tingting Zhu
- Department of Dental Biomaterials Science and Dental Research Institute, College of Dentistry, Seoul National University, 28 Yeonkun-dong, Chongro-ku, Seoul, 110-749, South Korea.
| | - Hee Chul Park
- Department of Dental Biomaterials Science and Dental Research Institute, College of Dentistry, Seoul National University, 28 Yeonkun-dong, Chongro-ku, Seoul, 110-749, South Korea.
| | - Kyung Mi Son
- Department of Dental Biomaterials Science and Dental Research Institute, College of Dentistry, Seoul National University, 28 Yeonkun-dong, Chongro-ku, Seoul, 110-749, South Korea.
| | - Hyeong-Cheol Yang
- Department of Dental Biomaterials Science and Dental Research Institute, College of Dentistry, Seoul National University, 28 Yeonkun-dong, Chongro-ku, Seoul, 110-749, South Korea.
| |
Collapse
|
39
|
Cellular memory of hypoxia elicits neuroblastoma metastasis and enables invasion by non-aggressive neighbouring cells. Oncogenesis 2015; 4:e138. [PMID: 25664931 PMCID: PMC4338426 DOI: 10.1038/oncsis.2014.52] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 11/03/2014] [Accepted: 11/18/2014] [Indexed: 12/20/2022] Open
Abstract
Therapies targeting cancer metastasis are challenging owing to the complexity of the metastatic process and the high number of effectors involved. Although tumour hypoxia has previously been associated with increased aggressiveness as well as resistance to radio- and chemotherapy, the understanding of a direct link between the level and duration of hypoxia and the individual steps involved in metastasis is still missing. Using live imaging in a chick embryo model, we have demonstrated that the exposure of neuroblastoma cells to 1% oxygen for 3 days was capable of (1) enabling cell migration towards blood vessels, (2) slowing down their velocity within blood vessels to facilitate extravasation and (3) promoting cell proliferation in primary and secondary sites. We have shown that cells do not have to be hypoxic anymore to exhibit these acquired capabilities as a long-term memory of prior hypoxic exposure is kept. Furthermore, non-hypoxic cells can be influenced by neighbouring hypoxic preconditioned cells and be entrained in the metastatic progression. The acquired aggressive phenotype relies on hypoxia-inducible factor (HIF)-dependent transcription of a number of genes involved in metastasis and can be impaired by HIF inhibition. Altogether, our results demonstrate the need to consider both temporal and spatial tumour heterogeneity because cells can 'remember' an earlier environment and share their acquired phenotype with their close neighbours. As a consequence, it is necessary to monitor the correct hypoxic markers to be able to predict the consequences of the cells' history on their behaviour and their potential response to therapies.
Collapse
|
40
|
Kuiper C, Dachs GU, Currie MJ, Vissers MCM. Intracellular ascorbate enhances hypoxia-inducible factor (HIF)-hydroxylase activity and preferentially suppresses the HIF-1 transcriptional response. Free Radic Biol Med 2014; 69:308-17. [PMID: 24495550 DOI: 10.1016/j.freeradbiomed.2014.01.033] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 01/22/2014] [Accepted: 01/25/2014] [Indexed: 12/17/2022]
Abstract
Hypoxia-inducible factor (HIF)-1 drives the transcription of hundreds of genes to support cell survival under conditions of microenvironmental and metabolic stress. HIF-1 is downregulated by iron-containing 2-oxoglutarate-dependent enzymes that require ascorbate as a cofactor. The HIF hydroxylases control both protein stability and the formation of an active transcription complex and, consequently, ascorbate could affect HIF-1α stabilization and/or gene expression, but the relative effect of ascorbate on these separate processes has not been well characterized. In this study we examined the effects of known intracellular ascorbate concentrations on both processes in response to various means of hydroxylase inhibition, including CoCl2, NiCl2, desferrioxamine, dimethyloxalylglycine, and hypoxia. Ascorbate inhibited HIF-1 activity most dramatically with all mechanisms of iron competition. In addition, HIF-1-dependent gene expression was effectively prevented by ascorbate and was inhibited even under conditions that allowed HIF-1α protein stabilization. This suggests that (1) ascorbate acts primarily to stabilize and reduce the iron atom in the hydroxylase active site and (2) the asparagine hydroxylase controlling HIF-1 transcriptional activity is particularly susceptible to fluctuations in intracellular ascorbate. These findings suggest that ascorbate plays a significant role in supporting HIF-hydroxylase function and that it could thereby modulate the cell survival response.
Collapse
Affiliation(s)
- Caroline Kuiper
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago at Christchurch, Christchurch 8140, New Zealand.
| | - Gabi U Dachs
- Mackenzie Cancer Research Group, Department of Pathology and Biomedical Science, University of Otago at Christchurch, Christchurch 8140, New Zealand
| | - Margaret J Currie
- Mackenzie Cancer Research Group, Department of Pathology and Biomedical Science, University of Otago at Christchurch, Christchurch 8140, New Zealand
| | - Margreet C M Vissers
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago at Christchurch, Christchurch 8140, New Zealand
| |
Collapse
|
41
|
Zieseniss A. Hypoxia and the modulation of the actin cytoskeleton - emerging interrelations. HYPOXIA 2014; 2:11-21. [PMID: 27774463 PMCID: PMC5045051 DOI: 10.2147/hp.s53575] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Recent progress in understanding the influence of hypoxia on cell function has revealed new information about the interrelationship between the actin cytoskeleton and hypoxia; nevertheless, details remain cloudy. The dynamic regulation of the actin cytoskeleton during hypoxia is complex, varies in different cells and tissues, and also depends on the mode of hypoxia. Several molecular players and pathways are emerging that contribute to the modulation of the actin cytoskeleton and that affect the large repertoire of actin-binding proteins in hypoxia. This review describes and discusses the accumulated knowledge about actin cytoskeleton dynamics in hypoxia, placing special emphasis on the Rho family of small guanosine triphosphatases (Rho GTPases). Given that RhoA, Rac and Cdc42 are very well characterized, the review is focused on these family members of Rho GTPases. Notably, in several cell types and tissues, hypoxia, presumably via Rho GTPase signaling, induces actin rearrangement and actin stress fiber assembly, which is a prevalent modulation of the actin cytoskeleton in hypoxia.
Collapse
Affiliation(s)
- Anke Zieseniss
- Institute of Cardiovascular Physiology, University Medical Center, Georg-August University, Göttingen, Germany
| |
Collapse
|
42
|
Zhang X, Yan X, Cheng L, Dai J, Wang C, Han P, Chai Y. Wound healing improvement with PHD-2 silenced fibroblasts in diabetic mice. PLoS One 2013; 8:e84548. [PMID: 24376825 PMCID: PMC3869847 DOI: 10.1371/journal.pone.0084548] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 11/15/2013] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Hypoxia-inducible factor 1α is the central regulator of the hypoxia-induced response which results in the up-regulation of angiogenic factors. Its activity is under precise regulation of prolyl-hydroxylase domain 2. We hypothesized that PHD2 silenced fibroblasts would increase the expression of angiogenic factors, which might contribute to the improvement of the diabetic wound healing. MATERIALS AND METHODS 50 dB/db mice were employed and randomly assigned into five groups with 10 mice in each: group 1 (untreated cell), group 2 (PHD2 silenced cell), group 3 (L-mimosine treated cells), group 4 (nontargeting siRNA treated cells) and group 5 (sham control). Fibroblasts were cultivated from the dermis of mice in each group and treated with PHD2 targeting siRNA, L-mimosine and non-targeting siRNA respectively. A fraction of the fibroblasts were employed to verify the silencing rate of PHD2 after 48 hours. The autologous fibroblasts (treated and untreated) labeled with adenovirus-GFP were implanted around the wound (Φ6mm), which was created on the dorsum of each mouse. The status of wounds was recorded periodically. Ten days postoperatively, 3 mice from each group were sacrificed and wound tissues were harvested. Molecular biological examinations were performed to evaluate the expressions of cytokines. 28 days postoperatively, the remaining mice were sacrificed. Histological examinations were performed to evaluate the densities of GFP+ cells and capillaries. RESULTS The expression of PHD2 reduced to 12.5%, and the expressions of HIF-1α and VEGFa increased significantly after PHD2 siRNA treatment. With the increasing expressions of HIF-1α and VEGFa, the time to wound closure in group 2 was less than 2 weeks. Increased numbers of GFP+ cells and capillaries were observed in group 2. CONCLUSION PHD2 siRNA treatment not only increased the expression of HIF1α and VEGFa, but also improved the fibroblast proliferation. These effects might contribute to the improvement of the diabetic wound healing.
Collapse
Affiliation(s)
- Xiongliang Zhang
- Department of Orthopedics, Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoyu Yan
- Department of Orthopedics, Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Liang Cheng
- Department of Orthopedics, Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jiezhi Dai
- Department of Orthopedics, Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Chunyang Wang
- Department of Orthopedics, Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Pei Han
- Department of Orthopedics, Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yimin Chai
- Department of Orthopedics, Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|