101
|
Suryani L, Foo JKR, Cardilla A, Dong Y, Muthukumaran P, Hassanbhai A, Wen F, Simon DT, Iandolo D, Yu N, Ng KW, Teoh SH. Effects of Pulsed Electromagnetic Field Intensity on Mesenchymal Stem Cells. Bioelectricity 2021. [DOI: 10.1089/bioe.2021.0002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Luvita Suryani
- School of Chemical and Biomedical Engineering, College of Engineering, Nanyang Technological University, Singapore, Singapore
| | - Jyong Kiat Reuben Foo
- School of Chemical and Biomedical Engineering, College of Engineering, Nanyang Technological University, Singapore, Singapore
| | - Angelysia Cardilla
- School of Chemical and Biomedical Engineering, College of Engineering, Nanyang Technological University, Singapore, Singapore
| | - Yibing Dong
- School of Materials Science and Engineering, Nanyang Technological University, Singapore, Singapore
| | - Padmalosini Muthukumaran
- School of Chemical and Biomedical Engineering, College of Engineering, Nanyang Technological University, Singapore, Singapore
| | - Ammar Hassanbhai
- School of Chemical and Biomedical Engineering, College of Engineering, Nanyang Technological University, Singapore, Singapore
| | - Feng Wen
- School of Chemical and Biomedical Engineering, College of Engineering, Nanyang Technological University, Singapore, Singapore
| | - Daniel T. Simon
- Laboratory of Organic Electronics, Department of Science and Technology, Linköping University, Norrköping, Sweden
| | - Donata Iandolo
- Laboratory of Organic Electronics, Department of Science and Technology, Linköping University, Norrköping, Sweden
- UMR5510 MATEIS, CNRS, INSA-Lyon, University of Lyon, Lyon, France
- Mines Saint-Etienne, INSERM, U1059 SAINBIOSE, Saint-Étienne, France
| | - Na Yu
- National Dental Centre Singapore, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Kee Woei Ng
- School of Materials Science and Engineering, Nanyang Technological University, Singapore, Singapore
- Environmental Chemistry & Materials Centre, Nanyang Environment and Water Research Institute (NEWRI), Nanyang Technological University, Singapore, Singapore
- Center for Nanotechnology and Nanotoxicology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, USA
| | - Swee-Hin Teoh
- School of Chemical and Biomedical Engineering, College of Engineering, Nanyang Technological University, Singapore, Singapore
- Lee Kong Chian School of Medicine, Singapore, Singapore
| |
Collapse
|
102
|
Bluhmki T, Traub S, Müller AK, Bitzer S, Schruf E, Bammert MT, Leist M, Gantner F, Garnett JP, Heilker R. Functional human iPSC-derived alveolar-like cells cultured in a miniaturized 96‑Transwell air-liquid interface model. Sci Rep 2021; 11:17028. [PMID: 34426605 PMCID: PMC8382767 DOI: 10.1038/s41598-021-96565-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 08/11/2021] [Indexed: 02/06/2023] Open
Abstract
In order to circumvent the limited access and donor variability of human primary alveolar cells, directed differentiation of human pluripotent stem cells (hiPSCs) into alveolar-like cells, provides a promising tool for respiratory disease modeling and drug discovery assays. In this work, a unique, miniaturized 96-Transwell microplate system is described where hiPSC-derived alveolar-like cells were cultured at an air-liquid interface (ALI). To this end, hiPSCs were differentiated into lung epithelial progenitor cells (LPCs) and subsequently matured into a functional alveolar type 2 (AT2)-like epithelium with monolayer-like morphology. AT2-like cells cultured at the physiological ALI conditions displayed characteristics of AT2 cells with classical alveolar surfactant protein expressions and lamellar-body like structures. The integrity of the epithelial barriers between the AT2-like cells was confirmed by applying a custom-made device for 96-parallelized transepithelial electric resistance (TEER) measurements. In order to generate an IPF disease-like phenotype in vitro, the functional AT2-like cells were stimulated with cytokines and growth factors present in the alveolar tissue of IPF patients. The cytokines stimulated the secretion of pro-fibrotic biomarker proteins both on the mRNA (messenger ribonucleic acid) and protein level. Thus, the hiPSC-derived and cellular model system enables the recapitulation of certain IPF hallmarks, while paving the route towards a miniaturized medium throughput approach of pharmaceutical drug discovery.
Collapse
Affiliation(s)
- Teresa Bluhmki
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397, Biberach an der Riss, Germany.
| | - Stefanie Traub
- Trenzyme GmbH, Byk-Gulden-Str. 2, 78467, Constance, Germany
| | | | - Sarah Bitzer
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397, Biberach an der Riss, Germany
| | - Eva Schruf
- Department of Immunology & Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397, Biberach an der Riss, Germany
| | - Marie-Therese Bammert
- Department of Immunology & Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397, Biberach an der Riss, Germany
| | - Marcel Leist
- In-vitro Toxicology and Biomedicine, University of Konstanz, 78457, Constance, Germany
| | - Florian Gantner
- Department of Translational Medicine and Clinical Pharmacology, C. H. Boehringer Sohn AG & Co. KG, 88397, Biberach an der Riss, Germany
| | - James P Garnett
- Department of Immunology & Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397, Biberach an der Riss, Germany
| | - Ralf Heilker
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397, Biberach an der Riss, Germany
| |
Collapse
|
103
|
Savorani C, Malinverno M, Seccia R, Maderna C, Giannotta M, Terreran L, Mastrapasqua E, Campaner S, Dejana E, Giampietro C. A dual role of YAP in driving TGFβ-mediated endothelial-to-mesenchymal transition. J Cell Sci 2021; 134:271139. [PMID: 34338295 PMCID: PMC8353525 DOI: 10.1242/jcs.251371] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 06/23/2021] [Indexed: 12/12/2022] Open
Abstract
Endothelial-to-mesenchymal transition (EndMT) is the biological process through which endothelial cells transdifferentiate into mesenchymal cells. During embryo development, EndMT regulates endocardial cushion formation via TGFβ/BMP signaling. In adults, EndMT is mainly activated during pathological conditions. Hence, it is necessary to characterize molecular regulators cooperating with TGFβ signaling in driving EndMT, to identify potential novel therapeutic targets to treat these pathologies. Here, we studied YAP, a transcriptional co-regulator involved in several biological processes, including epithelial-to-mesenchymal transition (EMT). As EndMT is the endothelial-specific form of EMT, and YAP (herein referring to YAP1) and TGFβ signaling cross-talk in other contexts, we hypothesized that YAP contributes to EndMT by modulating TGFβ signaling. We demonstrate that YAP is required to trigger TGFβ-induced EndMT response, specifically contributing to SMAD3-driven EndMT early gene transcription. We provide novel evidence that YAP acts as SMAD3 transcriptional co-factor and prevents GSK3β-mediated SMAD3 phosphorylation, thus protecting SMAD3 from degradation. YAP is therefore emerging as a possible candidate target to inhibit pathological TGFβ-induced EndMT at early stages. Summary: A new crucial role for YAP as a co-activator of early pathological TGFβ-mediated endothelial-to-mesenchymal transition program and characterization of the underlying molecular mechanism.
Collapse
Affiliation(s)
- Cecilia Savorani
- Institute of Molecular Oncology (IFOM), The Fondazione Italiana per la Ricerca sul Cancro (FIRC) Institute of Molecular Oncology, Milan 20139, Italy
| | - Matteo Malinverno
- Institute of Molecular Oncology (IFOM), The Fondazione Italiana per la Ricerca sul Cancro (FIRC) Institute of Molecular Oncology, Milan 20139, Italy
| | - Roberta Seccia
- Institute of Molecular Oncology (IFOM), The Fondazione Italiana per la Ricerca sul Cancro (FIRC) Institute of Molecular Oncology, Milan 20139, Italy
| | - Claudio Maderna
- Institute of Molecular Oncology (IFOM), The Fondazione Italiana per la Ricerca sul Cancro (FIRC) Institute of Molecular Oncology, Milan 20139, Italy
| | - Monica Giannotta
- Institute of Molecular Oncology (IFOM), The Fondazione Italiana per la Ricerca sul Cancro (FIRC) Institute of Molecular Oncology, Milan 20139, Italy
| | - Linda Terreran
- Institute of Molecular Oncology (IFOM), The Fondazione Italiana per la Ricerca sul Cancro (FIRC) Institute of Molecular Oncology, Milan 20139, Italy
| | - Eleonora Mastrapasqua
- Institute of Molecular Oncology (IFOM), The Fondazione Italiana per la Ricerca sul Cancro (FIRC) Institute of Molecular Oncology, Milan 20139, Italy
| | - Stefano Campaner
- Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia (IIT), Milan 20139, Italy
| | - Elisabetta Dejana
- Institute of Molecular Oncology (IFOM), The Fondazione Italiana per la Ricerca sul Cancro (FIRC) Institute of Molecular Oncology, Milan 20139, Italy.,Department of Immunology, Genetics and Pathology, Vascular Biology, Uppsala University, Uppsala 751 85, Sweden
| | - Costanza Giampietro
- Institute of Molecular Oncology (IFOM), The Fondazione Italiana per la Ricerca sul Cancro (FIRC) Institute of Molecular Oncology, Milan 20139, Italy.,Swiss Federal Laboratories for Materials Science and Technology (EMPA), Dübendorf 8600, Switzerland.,Department of Mechanical and Process Engineering, ETH Zurich, Zurich 8092, Switzerland
| |
Collapse
|
104
|
Akarapipad P, Kaarj K, Liang Y, Yoon JY. Environmental Toxicology Assays Using Organ-on-Chip. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2021; 14:155-183. [PMID: 33974806 DOI: 10.1146/annurev-anchem-091620-091335] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Adverse effects of environmental toxicants to human health have traditionally been assayed using in vitro assays. Organ-on-chip (OOC) is a new platform that can bridge the gaps between in vitro assays (or 3D cell culture) and animal tests. Microenvironments, physical and biochemical stimuli, and adequate sensing and biosensing systems can be integrated into OOC devices to better recapitulate the in vivo tissue and organ behavior and metabolism. While OOCs have extensively been studied for drug toxicity screening, their implementation in environmental toxicology assays is minimal and has limitations. In this review, recent attempts of environmental toxicology assays using OOCs, including multiple-organs-on-chip, are summarized and compared with OOC-based drug toxicity screening. Requirements for further improvements are identified and potential solutions are suggested.
Collapse
Affiliation(s)
- Patarajarin Akarapipad
- Department of Biomedical Engineering, University of Arizona, Tucson, Arizona 85721, USA;
| | - Kattika Kaarj
- Department of Biosystems Engineering, University of Arizona, Tucson, Arizona 85721, USA
| | - Yan Liang
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona 85721, USA
| | - Jeong-Yeol Yoon
- Department of Biomedical Engineering, University of Arizona, Tucson, Arizona 85721, USA;
- Department of Biosystems Engineering, University of Arizona, Tucson, Arizona 85721, USA
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona 85721, USA
| |
Collapse
|
105
|
Label-free two-photon imaging of mitochondrial activity in murine macrophages stimulated with bacterial and viral ligands. Sci Rep 2021; 11:14081. [PMID: 34234166 PMCID: PMC8263786 DOI: 10.1038/s41598-021-93043-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 06/16/2021] [Indexed: 11/12/2022] Open
Abstract
Mitochondria are the metabolic hub of the cell, playing a central role in regulating immune responses. Dysfunction of mitochondrial reprogramming can occur during bacterial and viral infections compromising hosts’ immune signaling. Comparative evaluation of these alterations in response to bacterial and viral ligands can provide insights into a cell’s ability to mount pathogen-specific responses. In this study, we used two-photon excitation fluorescence (TPEF) imaging to quantify reduced nicotinamide adenine dinucleotide phosphate (NAD(P)H) and flavin adenine dinucleotide (FAD) levels in the cell and to calculate the optical redox ratio (ORR), an indicator of mitochondrial dysfunction. Analyses were performed on RAW264.7 cells and murine bone marrow derived macrophages (BMM) stimulated with bacterial (LPS) and viral (Poly(I:C)) ligands. Responses were cell type dependent, with primary cells having significantly higher levels of FAD and higher oxygen consumption rates suggesting BMM may be more dependent on mitochondrial metabolism. Our findings also suggest that FAD-TPEF intensity may be a better predictor of mitochondrial activity and localization since it demonstrates unique mitochondrial clustering patterns in LPS vs. Poly(I:C) stimulated macrophages. Collectively, we demonstrate that TPEF imaging is a powerful label-free approach for quantifying changes in mitochondrial function and organization in macrophages following bacterial and viral stimuli.
Collapse
|
106
|
Pelepenko LE, Saavedra F, Antunes TBM, Bombarda GF, Gomes BPFDA, Zaia AA, Marciano MA. Investigation of a modified hydraulic calcium silicate-based material - Bio-C Pulpo. Braz Oral Res 2021; 35:e077. [PMID: 34161414 DOI: 10.1590/1807-3107bor-2021.vol35.0077] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 01/04/2021] [Indexed: 12/12/2022] Open
Abstract
This study evaluated the physicochemical, biological, and antimicrobial properties of a new hydraulic calcium silicate-based modified material, and compared it with MTA Repair HP and MTA Angelus. The materials were assessed regarding color luminosity (L), color change, radiopacity, setting time, and ISO 6876:2012 linear flow. Volumetric filling and volume change were evaluated using microcomputed-tomography (µCT). Chemical characterization after 28 days in Hank's Balanced Salt Solution (HBSS) and pH analysis were also assessed. Biological characterization of cytotoxicity and microbiological assessment were also undertaken. Shapiro-Wilk, ANOVA, Levene and post hoc analyses with Bonferroni correction were performed, adopting a 5% significance level (p <0.05). Bio-C Pulpo exhibited the highest L values after 90 days. All tested materials demonstrated color change during the analyses, and had radiopacity above 5 mm Al. MTA Repair HP set faster than Bio-C Pulpo, whereas the latter had the highest linear flow. MTA Repair HP had the highest volumetric filling in µCT analysis. Bio-C Pulpo showed the highest alkalinity during all tested periods, and the highest volumetric loss (above 9%), in comparison with MTA Repair HP and MTA Angelus. Bio-C Pulpo did not form calcium hydroxide after hydration. MTA Repair HP demonstrated the highest cytocompatibility, and Bio-C Pulpo, the highest cytotoxicity. No inhibition halos were observed for any material, and similar higher turbidity values were seen after direct contact. Composition additives used in Bio-C Pulpo modified its properties, and both the absence of calcium hydroxide deposition after hydration, and the related cytotoxicity of this material are of particular concern.
Collapse
Affiliation(s)
- Lauter Eston Pelepenko
- Universidade Estadual de Campinas - Unicamp, Piracicaba Dental School, Department of Restorative Dentistry, Piracicaba, SP, Brazil
| | - Flávia Saavedra
- Universidade Estadual de Campinas - Unicamp, Piracicaba Dental School, Department of Restorative Dentistry, Piracicaba, SP, Brazil
| | - Thiago Bessa Marconato Antunes
- Universidade Estadual de Campinas - Unicamp, Piracicaba Dental School, Department of Restorative Dentistry, Piracicaba, SP, Brazil
| | - Gabriela Fernanda Bombarda
- Universidade Estadual de Campinas - Unicamp, Piracicaba Dental School, Department of Restorative Dentistry, Piracicaba, SP, Brazil
| | | | - Alexandre Augusto Zaia
- Universidade Estadual de Campinas - Unicamp, Piracicaba Dental School, Department of Restorative Dentistry, Piracicaba, SP, Brazil
| | - Marina Angélica Marciano
- Universidade Estadual de Campinas - Unicamp, Piracicaba Dental School, Department of Restorative Dentistry, Piracicaba, SP, Brazil
| |
Collapse
|
107
|
Zheng F, Xiao Y, Liu H, Fan Y, Dao M. Patient-Specific Organoid and Organ-on-a-Chip: 3D Cell-Culture Meets 3D Printing and Numerical Simulation. Adv Biol (Weinh) 2021; 5:e2000024. [PMID: 33856745 PMCID: PMC8243895 DOI: 10.1002/adbi.202000024] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 02/13/2021] [Indexed: 12/11/2022]
Abstract
The last few decades have witnessed diversified in vitro models to recapitulate the architecture and function of living organs or tissues and contribute immensely to advances in life science. Two novel 3D cell culture models: 1) Organoid, promoted mainly by the developments of stem cell biology and 2) Organ-on-a-chip, enhanced primarily due to microfluidic technology, have emerged as two promising approaches to advance the understanding of basic biological principles and clinical treatments. This review describes the comparable distinct differences between these two models and provides more insights into their complementarity and integration to recognize their merits and limitations for applicable fields. The convergence of the two approaches to produce multi-organoid-on-a-chip or human organoid-on-a-chip is emerging as a new approach for building 3D models with higher physiological relevance. Furthermore, rapid advancements in 3D printing and numerical simulations, which facilitate the design, manufacture, and results-translation of 3D cell culture models, can also serve as novel tools to promote the development and propagation of organoid and organ-on-a-chip systems. Current technological challenges and limitations, as well as expert recommendations and future solutions to address the promising combinations by incorporating organoids, organ-on-a-chip, 3D printing, and numerical simulation, are also summarized.
Collapse
Affiliation(s)
- Fuyin Zheng
- Key Laboratory for Biomechanics and Mechanobiology, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- School of Biological Sciences, Nanyang Technological University, Singapore, 639798, Singapore
| | - Yuminghao Xiao
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Hui Liu
- Key Laboratory for Biomechanics and Mechanobiology, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Ming Dao
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- School of Biological Sciences, Nanyang Technological University, Singapore, 639798, Singapore
| |
Collapse
|
108
|
Song Y, Wu Y, Xu L, Jiang T, Tang C, Yin C. Caveolae-Mediated Endocytosis Drives Robust siRNA Delivery of Polymeric Nanoparticles to Macrophages. ACS NANO 2021; 15:8267-8282. [PMID: 33915044 DOI: 10.1021/acsnano.0c08596] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Cytosolic delivery of small interfering RNA (siRNA) remains challenging, and a profound understanding of the cellular uptake and intracellular processing of siRNA delivery systems could greatly improve the development of siRNA-based therapeutics. Here, we show that caveolae-mediated endocytosis (CvME) accounts for the robust siRNA delivery of mannose-modified trimethyl chitosan-cysteine/tripolyphosphate nanoparticles (MTC/TPP NPs) to macrophages by circumventing lysosomes. We show that the Golgi complex and ER are key organelles required for the efficient delivery of siRNA to macrophages in which the siRNA accumulation positively correlates with its silencing efficiency (r = 0.94). We also identify syntaxin6 and Niemann-Pick type C1 (NPC1) as indispensable regulators for MTC/TPP NPs-delivered siRNA into macrophages both in vitro and in vivo. Syntaxin6 and NPC1 knockout substantially decrease the cellular uptake and gene silencing of the siRNA delivered in MTC/TPP NPs in macrophages, which result in poor therapeutic outcomes for mice bearing acute hepatic injury. Our results suggest that highly efficient siRNA delivery can be achieved via CvME, which would give ideas for designing optimal delivery vectors to facilitate the clinical translation of siRNA drugs.
Collapse
Affiliation(s)
- Yudong Song
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200438, P.R. China
| | - Yanhua Wu
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200438, P.R. China
| | - Lu Xu
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200438, P.R. China
| | - Ting Jiang
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200438, P.R. China
| | - Cui Tang
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200438, P.R. China
| | - Chunhua Yin
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200438, P.R. China
| |
Collapse
|
109
|
Tott S, Grosicki M, Glowacz J, Mohaissen T, Wojnar-Lason K, Chlopicki S, Baranska M. Raman imaging-based phenotyping of murine primary endothelial cells to identify disease-associated biochemical alterations. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166180. [PMID: 34048923 DOI: 10.1016/j.bbadis.2021.166180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/28/2021] [Accepted: 05/17/2021] [Indexed: 12/15/2022]
Abstract
Raman spectroscopy is successfully becoming an analytical tool used to characterize alterations in the biochemical composition of cells. In this work, we identify the features of Raman spectra of murine primary endothelial cells (EC) isolated from lungs, heart, liver, brain, kidney and aorta of normal mice, as well as from heart, lung and liver in a murine model of heart failure (HF) in Tgαq*44 mice. Primary cells were measured in suspension immediately after their isolation. Raman images showed that isolated primary EC were elliptical or circular, and did not show organ-specific spectral features for any of the studied organ, i.e. lungs, heart, liver, brain, kidney and aorta. Principal Component Analysis pairwise analysis of primary endothelial cells from FVB mice and Tgαq*44 mice revealed an increased protein content in EC isolated from the heart and increased lipid content in EC isolated from the lung in Tgαq*44 mice. No significant differences were found in the EC isolated from the liver using the same chemometric procedure. To our knowledge, this is the first report in which Raman spectroscopy has been used to characterize the biochemical phenotype of primary murine EC with developing HF. This pilot study shows that Raman-based analysis of freshly isolated primary EC did not revealed organ-specific features, however disease-associated changes were found in the coronary and pulmonary EC in the early stage of heart failure in Tgαq*44 mice.
Collapse
Affiliation(s)
- Szymon Tott
- Jagiellonian University, Faculty of Chemistry, Gronostajowa 2, 30-387 Kraków, Poland; Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego 14, 30-348 Krakow, Poland
| | - Marek Grosicki
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego 14, 30-348 Krakow, Poland
| | - Jacek Glowacz
- Jagiellonian University, Faculty of Chemistry, Gronostajowa 2, 30-387 Kraków, Poland
| | - Tasnim Mohaissen
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego 14, 30-348 Krakow, Poland; Jagiellonian University Medical College, Faculty of Pharmacy, Medyczna 9, 30-688 Krakow, Poland
| | - Kamila Wojnar-Lason
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego 14, 30-348 Krakow, Poland; Jagiellonian University Medical College, Department of Pharmacology, Grzegorzecka 16, 31-531 Krakow, Poland
| | - Stefan Chlopicki
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego 14, 30-348 Krakow, Poland; Jagiellonian University Medical College, Department of Pharmacology, Grzegorzecka 16, 31-531 Krakow, Poland
| | - Malgorzata Baranska
- Jagiellonian University, Faculty of Chemistry, Gronostajowa 2, 30-387 Kraków, Poland; Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego 14, 30-348 Krakow, Poland.
| |
Collapse
|
110
|
Zhou Z, Zhu J, Jiang M, Sang L, Hao K, He H. The Combination of Cell Cultured Technology and In Silico Model to Inform the Drug Development. Pharmaceutics 2021; 13:pharmaceutics13050704. [PMID: 34065907 PMCID: PMC8151315 DOI: 10.3390/pharmaceutics13050704] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/29/2021] [Accepted: 05/05/2021] [Indexed: 12/12/2022] Open
Abstract
Human-derived in vitro models can provide high-throughput efficacy and toxicity data without a species gap in drug development. Challenges are still encountered regarding the full utilisation of massive data in clinical settings. The lack of translated methods hinders the reliable prediction of clinical outcomes. Therefore, in this study, in silico models were proposed to tackle these obstacles from in vitro to in vivo translation, and the current major cell culture methods were introduced, such as human-induced pluripotent stem cells (hiPSCs), 3D cells, organoids, and microphysiological systems (MPS). Furthermore, the role and applications of several in silico models were summarised, including the physiologically based pharmacokinetic model (PBPK), pharmacokinetic/pharmacodynamic model (PK/PD), quantitative systems pharmacology model (QSP), and virtual clinical trials. These credible translation cases will provide templates for subsequent in vitro to in vivo translation. We believe that synergising high-quality in vitro data with existing models can better guide drug development and clinical use.
Collapse
Affiliation(s)
- Zhengying Zhou
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China; (Z.Z.); (M.J.)
| | - Jinwei Zhu
- State Key Laboratory of Natural Medicines, Jiangsu Province Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China; (J.Z.); (L.S.)
| | - Muhan Jiang
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China; (Z.Z.); (M.J.)
| | - Lan Sang
- State Key Laboratory of Natural Medicines, Jiangsu Province Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China; (J.Z.); (L.S.)
| | - Kun Hao
- State Key Laboratory of Natural Medicines, Jiangsu Province Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China; (J.Z.); (L.S.)
- Correspondence: (K.H.); (H.H.)
| | - Hua He
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China; (Z.Z.); (M.J.)
- Correspondence: (K.H.); (H.H.)
| |
Collapse
|
111
|
Zhu I, Song W, Ovcharenko I, Landsman D. A model of active transcription hubs that unifies the roles of active promoters and enhancers. Nucleic Acids Res 2021; 49:4493-4505. [PMID: 33872375 PMCID: PMC8096258 DOI: 10.1093/nar/gkab235] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 01/27/2021] [Accepted: 03/22/2021] [Indexed: 12/31/2022] Open
Abstract
An essential questions of gene regulation is how large number of enhancers and promoters organize into gene regulatory loops. Using transcription-factor binding enrichment as an indicator of enhancer strength, we identified a portion of H3K27ac peaks as potentially strong enhancers and found a universal pattern of promoter and enhancer distribution: At actively transcribed regions of length of ∼200-300 kb, the numbers of active promoters and enhancers are inversely related. Enhancer clusters are associated with isolated active promoters, regardless of the gene's cell-type specificity. As the number of nearby active promoters increases, the number of enhancers decreases. At regions where multiple active genes are closely located, there are few distant enhancers. With Hi-C analysis, we demonstrate that the interactions among the regulatory elements (active promoters and enhancers) occur predominantly in clusters and multiway among linearly close elements and the distance between adjacent elements shows a preference of ∼30 kb. We propose a simple rule of spatial organization of active promoters and enhancers: Gene transcriptions and regulations mainly occur at local active transcription hubs contributed dynamically by multiple elements from linearly close enhancers and/or active promoters. The hub model can be represented with a flower-shaped structure and implies an enhancer-like role of active promoters.
Collapse
Affiliation(s)
- Iris Zhu
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wei Song
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ivan Ovcharenko
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20892, USA
| | - David Landsman
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
112
|
Almeida-Paes R, Figueiredo-Carvalho MHG, da Silva LBR, Gerfen G, de S Araújo GR, Frases S, Zancopé-Oliveira RM, Nosanchuk JD. Candida glabrata produces a melanin-like pigment that protects against stress conditions encountered during parasitism. Future Microbiol 2021; 16:509-520. [PMID: 33960816 PMCID: PMC11835011 DOI: 10.2217/fmb-2020-0228] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 03/26/2021] [Indexed: 11/21/2022] Open
Abstract
Aim: Melanin has been linked to pathogenesis in several fungi. They often produce melanin-like pigments in the presence of L-dihydroxyphenylalanine (L-DOPA), but this is poorly studied in Candida glabrata. Methods & materials:C. glabrata was grown in minimal medium with or without L-DOPA supplementation and submitted to a chemical treatment with denaturant and hot acid. Results:C. glabrata turned black when grown in the presence of L-DOPA, whereas cells grown without L-DOPA supplementation remained white. Biophysical properties demonstrated that the pigment was melanin. Melanized C. glabrata cells were effectively protected from azoles and amphotericin B, incubation at 42°C and macrophage killing. Conclusion: In the presence of L-DOPA, C. glabrata produces melanin, increases antifungal resistance and enhances host survival.
Collapse
Affiliation(s)
- Rodrigo Almeida-Paes
- Laboratório de Micologia,Instituto Nacional de Infectologia Evandro Chagas,Fundação Oswaldo Cruz,Rio de Janeiro,RJ,Brazil
- Departments of Medicine (Division of Infectious Diseases) & Microbiology & Immunology,Albert Einstein College of Medicine,Bronx,NY,USA
| | - Maria HG Figueiredo-Carvalho
- Laboratório de Micologia,Instituto Nacional de Infectologia Evandro Chagas,Fundação Oswaldo Cruz,Rio de Janeiro,RJ,Brazil
| | - Leandro BR da Silva
- Departments of Medicine (Division of Infectious Diseases) & Microbiology & Immunology,Albert Einstein College of Medicine,Bronx,NY,USA
- Instituto de Ciências Biomédicas,Departamento de Microbiologia,Universidade de São Paulo,São Paulo,SP,Brazil
| | - Gary Gerfen
- Department of Biophysics,Albert Einstein College of Medicine,Bronx,NY,USA
| | - Glauber R de S Araújo
- Laboratório de Biofísica de Fungos,Instituto de Biofísica Carlos Chagas Filho,Universidade Federal do Rio de Janeiro,Rio de Janeiro,RJ,Brazil
| | - Susana Frases
- Laboratório de Biofísica de Fungos,Instituto de Biofísica Carlos Chagas Filho,Universidade Federal do Rio de Janeiro,Rio de Janeiro,RJ,Brazil
| | - Rosely M Zancopé-Oliveira
- Laboratório de Micologia,Instituto Nacional de Infectologia Evandro Chagas,Fundação Oswaldo Cruz,Rio de Janeiro,RJ,Brazil
| | - Joshua D Nosanchuk
- Departments of Medicine (Division of Infectious Diseases) & Microbiology & Immunology,Albert Einstein College of Medicine,Bronx,NY,USA
| |
Collapse
|
113
|
Ustun M, Rahmani Dabbagh S, Ilci IS, Bagci-Onder T, Tasoglu S. Glioma-on-a-Chip Models. MICROMACHINES 2021; 12:490. [PMID: 33926127 PMCID: PMC8145995 DOI: 10.3390/mi12050490] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 12/16/2022]
Abstract
Glioma, as an aggressive type of cancer, accounts for virtually 80% of malignant brain tumors. Despite advances in therapeutic approaches, the long-term survival of glioma patients is poor (it is usually fatal within 12-14 months). Glioma-on-chip platforms, with continuous perfusion, mimic in vivo metabolic functions of cancer cells for analytical purposes. This offers an unprecedented opportunity for understanding the underlying reasons that arise glioma, determining the most effective radiotherapy approach, testing different drug combinations, and screening conceivable side effects of drugs on other organs. Glioma-on-chip technologies can ultimately enhance the efficacy of treatments, promote the survival rate of patients, and pave a path for personalized medicine. In this perspective paper, we briefly review the latest developments of glioma-on-chip technologies, such as therapy applications, drug screening, and cell behavior studies, and discuss the current challenges as well as future research directions in this field.
Collapse
Affiliation(s)
- Merve Ustun
- Graduate School of Sciences and Engineering, Koc University, Sariyer, 34450 Istanbul, Turkey;
| | - Sajjad Rahmani Dabbagh
- Department of Mechanical Engineering, Koç University, Sariyer, 34450 Istanbul, Turkey;
- Koç University Arçelik Research Center for Creative Industries (KUAR), Koç University, Sariyer, 34450 Istanbul, Turkey
| | - Irem Sultan Ilci
- Department of Bioengineering, Yildiz Technical University, 34220 Istanbul, Turkey;
| | - Tugba Bagci-Onder
- Brain Cancer Research and Therapy Lab, Koç University School of Medicine, 34450 Istanbul, Turkey;
- Koç University Research Center for Translational Medicine, Koç University, Sariyer, 34450 Istanbul, Turkey
| | - Savas Tasoglu
- Department of Mechanical Engineering, Koç University, Sariyer, 34450 Istanbul, Turkey;
- Koç University Arçelik Research Center for Creative Industries (KUAR), Koç University, Sariyer, 34450 Istanbul, Turkey
- Koç University Research Center for Translational Medicine, Koç University, Sariyer, 34450 Istanbul, Turkey
- Center for Life Sciences and Technologies, Bogazici University, Bebek, 34342 Istanbul, Turkey
- Boğaziçi Institute of Biomedical Engineering, Boğaziçi University, Çengelköy, 34684 Istanbul, Turkey
| |
Collapse
|
114
|
de Sousa HR, de Frazão S, de Oliveira Júnior GP, Albuquerque P, Nicola AM. Cryptococcal Virulence in Humans: Learning From Translational Studies With Clinical Isolates. Front Cell Infect Microbiol 2021; 11:657502. [PMID: 33968804 PMCID: PMC8097041 DOI: 10.3389/fcimb.2021.657502] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/31/2021] [Indexed: 01/17/2023] Open
Abstract
Cryptococcosis, an invasive mycosis caused by Cryptococcus spp, kills between 20% and 70% of the patients who develop it. There are no vaccines for prevention, and treatment is based on a limited number of antifungals. Studying fungal virulence and how the host responds to infection could lead to new therapies, improving outcomes for patients. The biggest challenge, however, is that experimental cryptococcosis models do not completely recapitulate human disease, while human experiments are limited due to ethical reasons. To overcome this challenge, one of the approaches used by researchers and clinicians is to: 1) collect cryptococcal clinical isolates and associated patient data; 2) study the set of isolates in the laboratory (virulence and host-pathogen interaction variables, molecular markers); 3) correlate the laboratory and patient data to understand the roles fungal attributes play in the human disease. Here we review studies that have shed light on the cryptococcosis pathophysiology using these approaches, with a special focus on human disease. Isolates that more effectively evade macrophage responses, that secrete more laccase, melanize faster and have larger capsules in the cerebrospinal fluid are associated with poorer patient outcomes. Additionally, molecular studies have also shown that cryptococcal clades vary in virulence, with clinical impact. Limitations of those studies include the use of a small number of isolates or retrospectively collected clinical data. The fact that they resulted in very important information is a reflection of the impact this strategy has in understanding cryptococcosis and calls for international collaboration that could boost our knowledge.
Collapse
Affiliation(s)
- Herdson Renney de Sousa
- Microbiology, Immunology and Biotechnology Laboratory, Faculty of Medicine, University of Brasília, Brasília, Brazil
| | - Stefânia de Frazão
- Laboratory of Molecular Biology of Pathogenic Fungi, Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Getúlio Pereira de Oliveira Júnior
- Division of Allergy and Inflammation, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Patrícia Albuquerque
- Microbiology, Immunology and Biotechnology Laboratory, Faculty of Medicine, University of Brasília, Brasília, Brazil
- Laboratory of Molecular Biology of Pathogenic Fungi, Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
- Faculty of Ceilândia, University of Brasília, Brasília, Brazil
| | - André Moraes Nicola
- Microbiology, Immunology and Biotechnology Laboratory, Faculty of Medicine, University of Brasília, Brasília, Brazil
- Graduate Program in Genomic Sciences and Biotechnology, Catholic University of Brasília, Brasília, Brazil
| |
Collapse
|
115
|
Orr-Burks N, Murray J, Todd KV, Bakre A, Tripp RA. G-Protein-Coupled Receptor and Ion Channel Genes Used by Influenza Virus for Replication. J Virol 2021; 95:e02410-20. [PMID: 33536179 PMCID: PMC8104092 DOI: 10.1128/jvi.02410-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 01/24/2021] [Indexed: 02/07/2023] Open
Abstract
Influenza virus causes epidemics and sporadic pandemics resulting in morbidity, mortality, and economic losses. Influenza viruses require host genes to replicate. RNA interference (RNAi) screens can identify host genes coopted by influenza virus for replication. Targeting these proinfluenza genes can provide therapeutic strategies to reduce virus replication. Nineteen proinfluenza G-protein-coupled receptor (GPCR) and 13 proinfluenza ion channel genes were identified in human lung (A549) cells by use of small interfering RNAs (siRNAs). These proinfluenza genes were authenticated by testing influenza virus A/WSN/33-, A/CA/04/09-, and B/Yamagata/16/1988-infected A549 cells, resulting in the validation of 16 proinfluenza GPCR and 5 proinfluenza ion channel genes. These findings showed that several GPCR and ion channel genes are needed for the production of infectious influenza virus. These data provide potential targets for the development of host-directed therapeutic strategies to impede the influenza virus productive cycle so as to limit infection.IMPORTANCE Influenza epidemics result in morbidity and mortality each year. Vaccines are the most effective preventive measure but require annual reformulation, since a mismatch of vaccine strains can result in vaccine failure. Antiviral measures are desirable particularly when vaccines fail. In this study, we used RNAi screening to identify several GPCR and ion channel genes needed for influenza virus replication. Understanding the host genes usurped by influenza virus during viral replication can help identify host genes that can be targeted for drug repurposing or for the development of antiviral drugs. The targeting of host genes is refractory to drug resistance generated by viral mutations, as well as providing a platform for the development of broad-spectrum antiviral drugs.
Collapse
Affiliation(s)
- Nichole Orr-Burks
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Jackelyn Murray
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Kyle V Todd
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Abhijeet Bakre
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Ralph A Tripp
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
116
|
Antoine-Lorquin A, Arensburger P, Arnaoty A, Asgari S, Batailler M, Beauclair L, Belleannée C, Buisine N, Coustham V, Guyetant S, Helou L, Lecomte T, Pitard B, Stévant I, Bigot Y. Two repeated motifs enriched within some enhancers and origins of replication are bound by SETMAR isoforms in human colon cells. Genomics 2021; 113:1589-1604. [PMID: 33812898 DOI: 10.1016/j.ygeno.2021.03.032] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 03/25/2021] [Accepted: 03/30/2021] [Indexed: 11/15/2022]
Abstract
Setmar is a gene specific to simian genomes. The function(s) of its isoforms are poorly understood and their existence in healthy tissues remains to be validated. Here we profiled SETMAR expression and its genome-wide binding landscape in colon tissue. We found isoforms V3 and V6 in healthy and tumour colon tissues as well as incell lines. In two colorectal cell lines SETMAR binds to several thousand Hsmar1 and MADE1 terminal ends, transposons mostly located in non-genic regions of active chromatin including in enhancers. It also binds to a 12-bp motifs similar to an inner motif in Hsmar1 and MADE1 terminal ends. This motif is interspersed throughout the genome and is enriched in GC-rich regions as well as in CpG islands that contain constitutive replication origins. It is also found in enhancers other than those associated with Hsmar1 and MADE1. The role of SETMAR in the expression of genes, DNA replication and in DNA repair are discussed.
Collapse
Affiliation(s)
| | - Peter Arensburger
- Biological Sciences Department, California State Polytechnic University, Pomona, CA 91768, - United States
| | - Ahmed Arnaoty
- EA GICC, 7501, CHRU de Tours, 37044 TOURS, Cedex 09, France
| | - Sassan Asgari
- School of Biological Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Martine Batailler
- PRC, UMR INRA 0085, CNRS 7247, Centre INRA Val de Loire, 37380 Nouzilly, France
| | - Linda Beauclair
- PRC, UMR INRA 0085, CNRS 7247, Centre INRA Val de Loire, 37380 Nouzilly, France
| | | | - Nicolas Buisine
- UMR CNRS 7221, Muséum National d'Histoire Naturelle, 75005 Paris, France
| | | | - Serge Guyetant
- Tumorothèque du CHRU de Tours, 37044 Tours, Cedex, France
| | - Laura Helou
- PRC, UMR INRA 0085, CNRS 7247, Centre INRA Val de Loire, 37380 Nouzilly, France
| | | | - Bruno Pitard
- Université de Nantes, CNRS ERL6001, Inserm 1232, CRCINA, F-44000 Nantes, France
| | - Isabelle Stévant
- Institut de Génomique Fonctionnelle de Lyon, Univ Lyon, CNRS UMR 5242, Ecole Normale Supérieure de Lyon, Université Claude Bernard Lyon, 1, 46 allée d'Italie, 69364 Lyon, France
| | - Yves Bigot
- PRC, UMR INRA 0085, CNRS 7247, Centre INRA Val de Loire, 37380 Nouzilly, France.
| |
Collapse
|
117
|
Fan CW, Lu R, Fang C, Zhang XL, Lv ZY, Li Y, Zhang H, Zhou ZG, Mo XM, Sun XF. Expression profile, molecular functions, and prognostic significance of miRNAs in primary colorectal cancer stem cells. Aging (Albany NY) 2021; 13:12067-12085. [PMID: 33793420 PMCID: PMC8109135 DOI: 10.18632/aging.202914] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 03/13/2021] [Indexed: 02/05/2023]
Abstract
MicroRNAs (miRNAs) are known to drive the pathogenesis of colorectal cancer (CRC) via the regulation of cancer stem cells (CSCs). We studied the miRNA expression profile of primary CSCs isolated from patients with CRC (pCRCSCs). Compared to pCRCSC-derived differentiated cells, 98 differentially expressed miRNAs were identified in pCRCSCs. Target genes encoding pCRCSC-related miRNAs were identified using a combination of miRNA target databases and miRNA-mRNA regulatory networks from the same patient. The pCRCSC-related miRNA target genes were associated with pathways contributing to malignant phenotypes, including I-kappa B kinase/NF-kappa B signaling, signal transduction by p53 class mediator, Ras signaling, and cGMP-PKG signaling. The pCRCSC-related miRNA expression signature was independently associated with poor overall survival in both the training and validation cohorts. We have thus identified several pCRCSC-related miRNAs with oncogenic potential that could serve as prognostic biomarkers for CRC.
Collapse
Affiliation(s)
- Chuan-Wen Fan
- Institute of Digestive Surgery, Sichuan University, and Department of Gastrointestinal Surgery, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China.,Department of Gastrointestinal Surgery and Breast and Thyroid Surgery, Minimally Invasive Surgery, West China Fourth Hospital, Sichuan University, Chengdu, China.,Department of Oncology and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.,Laboratory of Stem Cell Biology, West China Hospital, Sichuan University, Chengdu, China
| | - Ran Lu
- Laboratory of Stem Cell Biology, West China Hospital, Sichuan University, Chengdu, China
| | - Chao Fang
- Institute of Digestive Surgery, Sichuan University, and Department of Gastrointestinal Surgery, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Xue-Li Zhang
- School of Medicine, Institute of Medical Sciences, Örebro University, Örebro, Sweden
| | - Zhao-Ying Lv
- Institute of Digestive Surgery, Sichuan University, and Department of Gastrointestinal Surgery, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Yuan Li
- Institute of Digestive Surgery, Sichuan University, and Department of Gastrointestinal Surgery, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Hong Zhang
- School of Medicine, Institute of Medical Sciences, Örebro University, Örebro, Sweden
| | - Zong-Guang Zhou
- Institute of Digestive Surgery, Sichuan University, and Department of Gastrointestinal Surgery, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Xian-Ming Mo
- Laboratory of Stem Cell Biology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiao-Feng Sun
- Department of Oncology and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
118
|
Cell Surface Profiling of Retinal Müller Glial Cells Reveals Association to Immune Pathways after LPS Stimulation. Cells 2021; 10:cells10030711. [PMID: 33806940 PMCID: PMC8004686 DOI: 10.3390/cells10030711] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/17/2021] [Accepted: 03/19/2021] [Indexed: 12/19/2022] Open
Abstract
Retinal Müller glial cells (RMG) are involved in virtually every retinal disease; however, the role of these glial cells in neuroinflammation is still poorly understood. Since cell surface proteins play a decisive role in immune system signaling pathways, this study aimed at characterizing the changes of the cell surface proteome of RMG after incubation with prototype immune system stimulant lipopolysaccharide (LPS). While mass spectrometric analysis of the human Müller glia cell line MIO-M1 revealed 507 cell surface proteins in total, with 18 proteins significantly more abundant after stimulation (ratio ≥ 2), the surfaceome of primary RMG comprised 1425 proteins, among them 79 proteins with significantly higher abundance in the stimulated state. Pathway analysis revealed notable association with immune system pathways such as “antigen presentation”, “immunoregulatory interactions between a lymphoid and a non-lymphoid cell” and “cell migration”. We could demonstrate a higher abundance of proteins that are usually ascribed to antigen-presenting cells (APCs) and function to interact with T-cells, suggesting that activated RMG might act as atypical APCs in the course of ocular neuroinflammation. Our data provide a detailed description of the unstimulated and stimulated RMG surfaceome and offer fundamental insights regarding the capacity of RMG to actively participate in neuroinflammation in the retina.
Collapse
|
119
|
Prajapati M, Pettiglio MA, Conboy HL, Mercadante CJ, Hojyo S, Fukada T, Bartnikas TB. Characterization of in vitro models of SLC30A10 deficiency. Biometals 2021; 34:573-588. [PMID: 33713241 DOI: 10.1007/s10534-021-00296-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 02/26/2021] [Indexed: 11/25/2022]
Abstract
Manganese (Mn), an essential metal, can be toxic at elevated levels. In 2012, the first inherited cause of Mn excess was reported in patients with mutations in SLC30A10, a Mn efflux transporter. To explore the function of SLC30A10 in vitro, the current study used CRISPR/Cas9 gene editing to develop a stable SLC30A10 mutant Hep3B hepatoma cell line and collagenase perfusion in live mice to isolate primary hepatocytes deficient in Slc30a10. We also compared phenotypes of primary vs. non-primary cell lines to determine if they both serve as reliable in vitro models for the known physiological roles of SLC30A10. Mutant SLC30A10 Hep3B cells had increased Mn levels and decreased viability when exposed to excess Mn. Transport studies indicated a reduction of 54Mn import and export in mutant cells. While impaired 54Mn export was hypothesized given the essential role for SLC30A10 in cellular Mn export, impaired 54Mn import was unexpected. Whole genome sequencing did not identify any additional mutations in known Mn transporters in the mutant Hep3B mutant cell line. We then evaluated 54Mn transport in primary hepatocytes cultures isolated from genetically altered mice with varying liver Mn levels. Based on results from these experiments, we suggest that the effects of SLC30A10 deficiency on Mn homeostasis can be interrogated in vitro but only in specific types of cell lines.
Collapse
Affiliation(s)
- Milankumar Prajapati
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI, 02912, USA.
| | - Michael A Pettiglio
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI, 02912, USA.,Vor Biopharma, Cambridge, MA, USA
| | - Heather L Conboy
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI, 02912, USA
| | - Courtney J Mercadante
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI, 02912, USA.,Sanofi-Bioverativ, Waltham, MA, USA
| | - Shintaro Hojyo
- Deutsches Rheuma-Forschungszentrum Berlin, 10117, Berlin, Germany.,Division of Molecular Psychoimmunology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Hokkaido, 060-0815, Japan
| | - Toshiyuki Fukada
- Molecular and Cellular Physiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, 770-8514, Japan
| | - Thomas B Bartnikas
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI, 02912, USA
| |
Collapse
|
120
|
Schmidt CX, Tsang AH, Oster H. Generation of Mouse Primary Hypothalamic Neuronal Cultures for Circadian Bioluminescence Assays. Bio Protoc 2021; 11:e3944. [PMID: 33796618 DOI: 10.21769/bioprotoc.3944] [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: 10/30/2020] [Revised: 01/11/2021] [Accepted: 01/31/2021] [Indexed: 11/02/2022] Open
Abstract
An endogenous circadian clock system enables organisms to adapt to time-of-day dependent environmental changes. In consequence, most physiological processes exhibit daily rhythms of, e.g., energy metabolism, immune function, sleep, or hormone production. Hypothalamic circadian clocks have been identified to play a particular role in coordinating many of these processes. Primary neuronal cultures are widely used as a physiologically relevant model to study molecular events within neurons. However, as circadian rhythms include dynamic molecular changes over longer timescales that vary between individual cells, longitudinal measurement methods are essential to investigate the regulation of circadian clocks of hypothalamic neurons. Here we provide a protocol for generating primary hypothalamic neuronal cultures expressing a circadian luciferase reporter. Such reporter cells can be used to longitudinally monitor cellular circadian rhythms at high temporal resolution by performing bioluminescence measurements.
Collapse
Affiliation(s)
- Cosima X Schmidt
- Institute of Neurobiology, Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany
| | - Anthony H Tsang
- Institute of Neurobiology, Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany
| | - Henrik Oster
- Institute of Neurobiology, Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany
| |
Collapse
|
121
|
Rajendran RL, Paudel S, Gangadaran P, Oh JM, Oh EJ, Hong CM, Lee S, Chung HY, Lee J, Ahn BC. Extracellular Vesicles Act as Nano-Transporters of Tyrosine Kinase Inhibitors to Revert Iodine Avidity in Thyroid Cancer. Pharmaceutics 2021; 13:248. [PMID: 33578882 PMCID: PMC7916551 DOI: 10.3390/pharmaceutics13020248] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/04/2021] [Accepted: 02/05/2021] [Indexed: 12/21/2022] Open
Abstract
A new approach for using extracellular vesicles (EVs) to deliver tyrosine kinase inhibitors (TKIs) to enhance iodine avidity in radioactive iodine-refractory thyroid cancer is needed. We isolated and characterized primary human adipose-derived stem cells (ADSCs) and isolated their EVs. The EVs were characterized by transmission electron microscopy, nanoparticle tracking analysis, and western blotting. A new TKI was loaded into the EVs by incubation (37 °C; 10 min) or sonication (18 cycles; 4 s per cycle) with 2 s intervals and a 2 min ice bath every six cycles. TKI loading was confirmed and measured by mass spectrometry. EV uptake into radioactive iodine-refractory thyroid cancer cells (SW1736 cells) was confirmed by microscopy. We treated the SW1736 cells with vehicle, TKI, or TKI-loaded EVs (sonication TKI-loaded EVs [EVsTKI(S)]) and examined the expression of iodide-metabolizing proteins and radioiodine uptake in the SW1736 cells. ADSCs cells showed >99% of typical stem cell markers, such as CD90 and CD105. The EVs displayed a round morphology, had an average size of 211.4 ± 3.83 nm, and were positive for CD81 and Alix and negative for cytochrome c. The mass spectrometry results indicate that the sonication method loaded ~4 times more of the TKI than did the incubation method. The EVsTKI(S) were used for further experiments. Higher expression levels of iodide-metabolizing mRNA and proteins in the EVsTKI(S)-treated SW1736 cells than in TKI-treated SW1736 cells were confirmed. EVsTKI(S) treatment enhanced 125I uptake in the recipient SW1736 cells compared with free-TKI treatment. This is the first study that demonstrated successful delivery of a TKI to radioactive iodine-refractory thyroid cancer cells using EVs as the delivery vehicle. This approach can revert radioiodine-resistant thyroid cancer cells back to radioiodine-sensitive thyroid cancer cells.
Collapse
Affiliation(s)
- Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (R.L.R.); (P.G.); (J.M.O.); (J.L.)
| | - Sanjita Paudel
- BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (S.P.); (S.L.)
| | - Prakash Gangadaran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (R.L.R.); (P.G.); (J.M.O.); (J.L.)
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, School of Medicine, Kyungpook National University, Daegu 41944, Korea;
| | - Ji Min Oh
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (R.L.R.); (P.G.); (J.M.O.); (J.L.)
| | - Eun Jung Oh
- Department of Plastic and Reconstructive Surgery, CMRI, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea;
| | - Chae Moon Hong
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu 41404, Korea;
| | - Sangkyu Lee
- BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (S.P.); (S.L.)
| | - Ho Yun Chung
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, School of Medicine, Kyungpook National University, Daegu 41944, Korea;
- Department of Plastic and Reconstructive Surgery, CMRI, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea;
| | - Jaetae Lee
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (R.L.R.); (P.G.); (J.M.O.); (J.L.)
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu 41404, Korea;
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (R.L.R.); (P.G.); (J.M.O.); (J.L.)
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, School of Medicine, Kyungpook National University, Daegu 41944, Korea;
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu 41404, Korea;
| |
Collapse
|
122
|
Cheng X, Uchida T, Xia Y, Umarova R, Liu CJ, Chen PJ, Gaggar A, Suri V, Mücke MM, Vermehren J, Zeuzem S, Teraoka Y, Osawa M, Aikata H, Tsuji K, Mori N, Hige S, Karino Y, Imamura M, Chayama K, Liang TJ. Diminished hepatic IFN response following HCV clearance triggers HBV reactivation in coinfection. J Clin Invest 2021; 130:3205-3220. [PMID: 32163375 DOI: 10.1172/jci135616] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 03/05/2020] [Indexed: 12/12/2022] Open
Abstract
In patients with HBV and HCV coinfection, HBV reactivation leading to severe hepatitis has been reported with the use of direct-acting antivirals (DAAs) to treat HCV infection. Here we studied the molecular mechanisms behind this viral interaction. In coinfected cell culture and humanized mice, HBV replication was suppressed by HCV coinfection. In vitro, HBV suppression was attenuated when interferon (IFN) signaling was blocked. In vivo, HBV viremia, after initial suppression by HCV superinfection, rebounded following HCV clearance by DAA treatment that was accompanied by a reduced hepatic IFN response. Using blood samples of coinfected patients, IFN-stimulated gene products including C-X-C motif chemokine 10 (CXCL10), C-C motif chemokine ligand 5 (CCL5), and alanine aminotransferase (ALT) were identified to have predictive value for HBV reactivation after HCV clearance. Taken together, our data suggest that HBV reactivation is a result of diminished hepatic IFN response following HCV clearance and identify serologic markers that can predict HBV reactivation in DAA-treated HBV-HCV-coinfected persons.
Collapse
Affiliation(s)
- Xiaoming Cheng
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Takuro Uchida
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA.,Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yuchen Xia
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Regina Umarova
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Chun-Jen Liu
- Graduate Institute of Clinical Medicine, Hepatitis Research Center and Department of Internal Medicine, National Taiwan University College of Medicine and Hospital, Taipei, Taiwan
| | - Pei-Jer Chen
- Graduate Institute of Clinical Medicine, Hepatitis Research Center and Department of Internal Medicine, National Taiwan University College of Medicine and Hospital, Taipei, Taiwan
| | - Anuj Gaggar
- Gilead Sciences, Foster City, California, USA
| | | | - Marcus M Mücke
- Department of Internal Medicine 1, University Hospital Frankfurt, Frankfurt, Germany
| | - Johannes Vermehren
- Department of Internal Medicine 1, University Hospital Frankfurt, Frankfurt, Germany
| | - Stefan Zeuzem
- Department of Internal Medicine 1, University Hospital Frankfurt, Frankfurt, Germany
| | - Yuji Teraoka
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Mitsutaka Osawa
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hiroshi Aikata
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Keiji Tsuji
- Department of Gastroenterology, Hiroshima Red Cross Hospital and Atomic-bomb Survivors Hospital, Hiroshima, Japan
| | - Nami Mori
- Department of Gastroenterology, Hiroshima Red Cross Hospital and Atomic-bomb Survivors Hospital, Hiroshima, Japan
| | - Shuhei Hige
- Department of Hepatology, Sapporo Kosei General Hospital, Sapporo, Japan
| | - Yoshiyasu Karino
- Department of Hepatology, Sapporo Kosei General Hospital, Sapporo, Japan
| | - Michio Imamura
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kazuaki Chayama
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - T Jake Liang
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| |
Collapse
|
123
|
Chaousis S, Leusch FDL, Nouwens A, Melvin SD, van de Merwe JP. Changes in global protein expression in sea turtle cells exposed to common contaminants indicates new biomarkers of chemical exposure. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 751:141680. [PMID: 32890801 DOI: 10.1016/j.scitotenv.2020.141680] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/10/2020] [Accepted: 08/11/2020] [Indexed: 05/14/2023]
Abstract
Non-targeted protein expression at the cellular level can provide insights into mechanistic effects of contaminants in wildlife, and hence new and potentially more accurate biomarkers of exposure and effect. However, this technique has been relatively unexplored in the realm of in vitro biomarker discovery in threatened wildlife, despite the vulnerability of this group of animals to adverse sublethal effects of contaminant exposure. Here we examined the usefulness of non-targeted protein expression for biomarker discovery in green sea turtles (Chelonia mydas) by investigating differences in the response of primary cells from five different tissue types that were exposed to three contaminants known to accumulate in this species. Cells derived from C. mydas skin, liver, kidney, ovary and small intestine were exposed to 100 μg/L of either polychlorinated biphenyl 153 (PCB153), perfluorononanoic acid (PFNA) or phenanthrene for 24 h. The global protein expression was then quantitatively evaluated using sequential window acquisition of all theoretical mass spectra (SWATH-MS). Comparison of the global protein profiles revealed that, while a majority of proteins were mutually expressed in controls of all tissue types (~90%), the response to exposure in terms of protein expression strength was significantly different between tissue types. Furthermore, a comparison to known markers of chemical exposure in sea turtles from the literature indicated that in vitro response can reflect known in vivo responses. In particular, markers such as heat shock protein (HSP) 60, glutathione S-transferases (GSTs) and superoxide dismutases (SODs), cytochrome P450 and catalase were dysregulated in response to exposure. Furthermore, potential new markers of exposure were discovered such as annexin, an important protein in cell signalling processes. While this methodology proved promising further studies are required to confirm the accuracy of in vitro protein expression as a tool for biomarker discovery in wildlife.
Collapse
Affiliation(s)
- Stephanie Chaousis
- Griffith School of Science and Environment, Building 51, Griffith University, Gold Coast Campus, QLD 4222, Australia; The Australian Rivers Institute, Building 51, Griffith University, Gold Coast Campus, QLD 4222, Australia.
| | - Frederic D L Leusch
- Griffith School of Science and Environment, Building 51, Griffith University, Gold Coast Campus, QLD 4222, Australia; The Australian Rivers Institute, Building 51, Griffith University, Gold Coast Campus, QLD 4222, Australia
| | - Amanda Nouwens
- School of Chemistry and Molecular Biology, Building 76, The University of Queensland, QLD 4067, Australia
| | - Steven D Melvin
- Griffith School of Science and Environment, Building 51, Griffith University, Gold Coast Campus, QLD 4222, Australia; The Australian Rivers Institute, Building 51, Griffith University, Gold Coast Campus, QLD 4222, Australia
| | - Jason P van de Merwe
- Griffith School of Science and Environment, Building 51, Griffith University, Gold Coast Campus, QLD 4222, Australia; The Australian Rivers Institute, Building 51, Griffith University, Gold Coast Campus, QLD 4222, Australia
| |
Collapse
|
124
|
Endothelial Cells as Tools to Model Tissue Microenvironment in Hypoxia-Dependent Pathologies. Int J Mol Sci 2021; 22:ijms22020520. [PMID: 33430201 PMCID: PMC7825710 DOI: 10.3390/ijms22020520] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/27/2020] [Accepted: 01/05/2021] [Indexed: 12/11/2022] Open
Abstract
Endothelial cells (ECs) lining the blood vessels are important players in many biological phenomena but are crucial in hypoxia-dependent diseases where their deregulation contributes to pathology. On the other hand, processes mediated by ECs, such as angiogenesis, vessel permeability, interactions with cells and factors circulating in the blood, maintain homeostasis of the organism. Understanding the diversity and heterogeneity of ECs in different tissues and during various biological processes is crucial in biomedical research to properly develop our knowledge on many diseases, including cancer. Here, we review the most important aspects related to ECs’ heterogeneity and list the available in vitro tools to study different angiogenesis-related pathologies. We focus on the relationship between functions of ECs and their organo-specificity but also point to how the microenvironment, mainly hypoxia, shapes their activity. We believe that taking into account the specific features of ECs that are relevant to the object of the study (organ or disease state), especially in a simplified in vitro setting, is important to truly depict the biology of endothelium and its consequences. This is possible in many instances with the use of proper in vitro tools as alternative methods to animal testing.
Collapse
|
125
|
Grabowska K, Harwood E, Ciborowski P. HIV and Proteomics: What We Have Learned from High Throughput Studies. Proteomics Clin Appl 2021; 15:e2000040. [PMID: 32978881 PMCID: PMC7900993 DOI: 10.1002/prca.202000040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 09/04/2020] [Indexed: 12/17/2022]
Abstract
The accelerated development of technology over the last three decades has driven biological sciences to high-throughput profiling experiments, now broadly referred to as systems biology. The unprecedented improvement of analytical instrumentation has opened new avenues for more complex experimental designs and expands the knowledge in genomics, proteomics, and other omics fields. Despite the collective efforts of hundreds of researchers, gleaning all the expected information from omics experiments is still quite far. This paper summarizes what has been learned from high-throughput proteomics studies thus far, and what is believed should be done to reveal even more valuable information from such studies. It is drawn from the background in using proteomics to study human immunodeficiency virus 1 infection of macrophages and/or T cells, but it is believed that some conclusions will be more broadly applicable.
Collapse
Affiliation(s)
- Kinga Grabowska
- Laboratory of Virus Molecular BiologyIntercollegiate Faculty of BiotechnologyUniversity of GdanskGdansk80‐307Poland
- Department of Pharmacology and Experimental NeuroscienceCollege of MedicineUniversity of Nebraska Medical CenterOmahaNE68198‐5800USA
| | - Emma Harwood
- Department of Pharmacology and Experimental NeuroscienceCollege of MedicineUniversity of Nebraska Medical CenterOmahaNE68198‐5800USA
| | - Pawel Ciborowski
- Department of Pharmacology and Experimental NeuroscienceCollege of MedicineUniversity of Nebraska Medical CenterOmahaNE68198‐5800USA
| |
Collapse
|
126
|
Miya C, Cueno ME, Suzuki R, Maruoka S, Gon Y, Kaneko T, Yonehara Y, Imai K. Porphyromonas gingivalis gingipains potentially affect MUC5AC gene expression and protein levels in respiratory epithelial cells. FEBS Open Bio 2020; 11:446-455. [PMID: 33332733 PMCID: PMC7876492 DOI: 10.1002/2211-5463.13066] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/02/2020] [Accepted: 12/15/2020] [Indexed: 01/08/2023] Open
Abstract
Porphyromonas gingivalis (Pg) is a periodontopathic pathogen that may affect MUC5AC‐related mucus hypersecretion along airway epithelial cells. Here, we attempted to establish whether Pg virulence factors (lipopolysaccharide, FimA fimbriae, gingipains) affect MUC5AC in immortalized and primary bronchial cells. We report that MUC5AC gene expression and protein levels are affected by Pg culture supernatant, but not by lipopolysaccharide or FimA fimbriae. Cells treated with either Pg single (Kgp or Rgp) or double (Kgp/Rgp) mutants had altered levels of MUC5AC gene expression and protein levels, and MUC5AC staining of double mutant‐treated mouse lung cells showed that MUC5AC protein levels were unaffected. Taken together, we propose that Pg gingipains may be the primary virulence factor that influences both MUC5AC gene expression and protein levels.
Collapse
Affiliation(s)
- Chihiro Miya
- Department of Oral and Maxillofacial Surgery II, Nihon University School of Dentistry, Tokyo, Japan.,Department of Microbiology, Nihon University School of Dentistry, Tokyo, Japan
| | - Marni E Cueno
- Department of Microbiology, Nihon University School of Dentistry, Tokyo, Japan
| | - Ryuta Suzuki
- Department of Oral and Maxillofacial Surgery II, Nihon University School of Dentistry, Tokyo, Japan.,Department of Microbiology, Nihon University School of Dentistry, Tokyo, Japan
| | - Shuichiro Maruoka
- Department of Internal Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Yasuhiro Gon
- Department of Internal Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Tadayoshi Kaneko
- Department of Oral and Maxillofacial Surgery II, Nihon University School of Dentistry, Tokyo, Japan
| | - Yoshiyuki Yonehara
- Department of Oral and Maxillofacial Surgery II, Nihon University School of Dentistry, Tokyo, Japan
| | - Kenichi Imai
- Department of Microbiology, Nihon University School of Dentistry, Tokyo, Japan
| |
Collapse
|
127
|
Pei R, Feng J, Zhang Y, Sun H, Li L, Yang X, He J, Xiao S, Xiong J, Lin Y, Wen K, Zhou H, Chen J, Rong Z, Chen X. Host metabolism dysregulation and cell tropism identification in human airway and alveolar organoids upon SARS-CoV-2 infection. Protein Cell 2020; 12:717-733. [PMID: 33314005 PMCID: PMC7732737 DOI: 10.1007/s13238-020-00811-w] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 11/13/2020] [Indexed: 12/12/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic is caused by infection with the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which is spread primary via respiratory droplets and infects the lungs. Currently widely used cell lines and animals are unable to accurately mimic human physiological conditions because of the abnormal status of cell lines (transformed or cancer cells) and species differences between animals and humans. Organoids are stem cell-derived self-organized three-dimensional culture in vitro and model the physiological conditions of natural organs. Here we showed that SARS-CoV-2 infected and extensively replicated in human embryonic stem cells (hESCs)-derived lung organoids, including airway and alveolar organoids which covered the complete infection and spread route for SARS-CoV-2 within lungs. The infected cells were ciliated, club, and alveolar type 2 (AT2) cells, which were sequentially located from the proximal to the distal airway and terminal alveoli, respectively. Additionally, RNA-seq revealed early cell response to virus infection including an unexpected downregulation of the metabolic processes, especially lipid metabolism, in addition to the well-known upregulation of immune response. Further, Remdesivir and a human neutralizing antibody potently inhibited SARS-CoV-2 replication in lung organoids. Therefore, human lung organoids can serve as a pathophysiological model to investigate the underlying mechanism of SARS-CoV-2 infection and to discover and test therapeutic drugs for COVID-19.
Collapse
Affiliation(s)
- Rongjuan Pei
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Jianqi Feng
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yecheng Zhang
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Hao Sun
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Lian Li
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xuejie Yang
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- Joint School of Life Sciences, Guangzhou Medical University and Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 511436, China
| | - Jiangping He
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- The Centre of Cell Lineage and Atlas (CCLA), Bioland Laboratory (Guangzhou Regenerative Medicine and Health-Guangdong Laboratory), Guangzhou, 510530, China
| | - Shuqi Xiao
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Jin Xiong
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Ying Lin
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Kun Wen
- Microbiome Medicine Center, Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Hongwei Zhou
- Microbiome Medicine Center, Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jiekai Chen
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- The Centre of Cell Lineage and Atlas (CCLA), Bioland Laboratory (Guangzhou Regenerative Medicine and Health-Guangdong Laboratory), Guangzhou, 510530, China
- Joint School of Life Sciences, Guangzhou Medical University and Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 511436, China
| | - Zhili Rong
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, China.
- Dermatology Hospital, Southern Medical University, Guangzhou, 510091, China.
| | - Xinwen Chen
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
| |
Collapse
|
128
|
Addario G, Djudjaj S, Farè S, Boor P, Moroni L, Mota C. Microfluidic bioprinting towards a renal in vitro model. ACTA ACUST UNITED AC 2020. [DOI: 10.1016/j.bprint.2020.e00108] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
129
|
Demers I, Donkers J, Kremer B, Speel EJ. Ex Vivo Culture Models to Indicate Therapy Response in Head and Neck Squamous Cell Carcinoma. Cells 2020; 9:E2527. [PMID: 33238461 PMCID: PMC7700693 DOI: 10.3390/cells9112527] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/17/2020] [Accepted: 11/20/2020] [Indexed: 12/24/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is characterized by a poor 5 year survival and varying response rates to both standard-of-care and new treatments. Despite advances in medicine and treatment methods, mortality rates have hardly decreased in recent decades. Reliable patient-derived tumor models offer the chance to predict therapy response in a personalized setting, thereby improving treatment efficacy by identifying the most appropriate treatment regimen for each patient. Furthermore, ex vivo tumor models enable testing of novel therapies before introduction in clinical practice. A literature search was performed to identify relevant literature describing three-dimensional ex vivo culture models of HNSCC to examine sensitivity to chemotherapy, radiotherapy, immunotherapy and targeted therapy. We provide a comprehensive overview of the currently used three-dimensional ex vivo culture models for HNSCC with their advantages and limitations, including culture success percentage and comparison to the original tumor. Furthermore, we evaluate the potential of these models to predict patient therapy response.
Collapse
Affiliation(s)
- Imke Demers
- Department of Pathology, GROW-school for Oncology and Development Biology, Maastricht University Medical Centre, PO Box 5800, 6202 AZ Maastricht, The Netherlands;
| | - Johan Donkers
- Department of Otorhinolaryngology, Head and Neck Surgery, GROW-School for Oncology and Development Biology, Maastricht University Medical Centre, PO Box 5800, 6202 AZ Maastricht, The Netherlands; (J.D.); (B.K.)
| | - Bernd Kremer
- Department of Otorhinolaryngology, Head and Neck Surgery, GROW-School for Oncology and Development Biology, Maastricht University Medical Centre, PO Box 5800, 6202 AZ Maastricht, The Netherlands; (J.D.); (B.K.)
| | - Ernst Jan Speel
- Department of Pathology, GROW-school for Oncology and Development Biology, Maastricht University Medical Centre, PO Box 5800, 6202 AZ Maastricht, The Netherlands;
| |
Collapse
|
130
|
Jain A, Behera M, Ravi V, Mishra S, Sundaresan NR, Chatterjee K. Recapitulating pathophysiology of skeletal muscle diseases in vitro using primary mouse myoblasts on a nanofibrous platform. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 32:102341. [PMID: 33227539 DOI: 10.1016/j.nano.2020.102341] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 11/05/2020] [Accepted: 11/09/2020] [Indexed: 01/10/2023]
Abstract
Tissue engineering approaches are used to mimic the microenvironment of the skeletal muscle in vitro. However, the validation of a bioengineered muscle as a model to study diseases is inadequate. Here, we present polycaprolactone nanofibers as a robust platform that mimics cellular organization and recapitulates critical functions of the myotubes observed in vivo. We isolated myoblasts from mice following a simplified protocol and cultured them on aligned nanofibers. Myotubes grown on aligned nanofibers maintained alignment for 14 days and exhibited a time-dependent increase in levels of p-AKT upon insulin stimulation. Treatment with matrix-assisted integrin inhibitor led to reduction in p-AKT levels, underscoring the critical role of environment on the biological processes. We demonstrate the suitability of myotubes grown on nanofibrous platform to study corticosteroid-induced muscle degeneration. This study, thus, demonstrates that aligned nanofibers retain myotubes in culture for longer duration and recapitulate the functions of skeletal muscle under pathophysiological conditions.
Collapse
Affiliation(s)
- Aditi Jain
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India
| | - Manisha Behera
- Department of Materials Engineering, Indian Institute of Science, Bangalore, India
| | - Venkatraman Ravi
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | - Sneha Mishra
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | - Nagalingam R Sundaresan
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India; Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India.
| | - Kaushik Chatterjee
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India; Department of Materials Engineering, Indian Institute of Science, Bangalore, India.
| |
Collapse
|
131
|
Landázuri AC, Gualle A, Castañeda V, Morales E, Caicedo A, Orejuela-Escobar LM. Moringa oleifera Lam. leaf powder antioxidant activity and cytotoxicity in human primary fibroblasts. Nat Prod Res 2020; 35:6194-6199. [PMID: 33118387 DOI: 10.1080/14786419.2020.1837804] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Moringa oleifera Lam. (MO) leaf powder has been well studied, however, understanding how extraction methods of antioxidant compounds affect human primary fibroblasts still needs to be determined. The antioxidant capacity was analyzed through a copper reduction capacity method and primary human skin fibroblasts were evaluated for cytotoxicity using MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay. Antioxidant activity under the influence of methanolic solvents (Trolox equivalents of 160.18 nmol/µL) was 17 times higher than under aqueous solvents. Interestingly, the aqueous extract showed less toxicity in comparison to the methanolic, as cells resulted more susceptible to concentrations ranging from 0.05 to 5 mg/L. Although, MO methanol solvent showed a higher antioxidant capacity in comparison to the aqueous solvent, it presented greater cytotoxicity. Thus, it is concluded that the aqueous extract could be suitable for downstream processing and applications.
Collapse
Affiliation(s)
- Andrea C Landázuri
- Chemical Engineering Department, Engineering, Applied Sciences & Simulation Group (GICAS), Colegio de Ciencias e Ingenierías, Universidad San Francisco de Quito USFQ, Quito, Ecuador.,Instituto de Investigaciones Biológicas y Ambientales BIÓSFERA, Universidad San Francisco de Quito USFQ, Quito, Ecuador.,Instituto de Investigaciones en Biomedicina, Universidad San Francisco de Quito USFQ, Quito, Ecuador
| | - Arleth Gualle
- Chemical Engineering Department, Engineering, Applied Sciences & Simulation Group (GICAS), Colegio de Ciencias e Ingenierías, Universidad San Francisco de Quito USFQ, Quito, Ecuador
| | - Verónica Castañeda
- Instituto de Investigaciones en Biomedicina, Universidad San Francisco de Quito USFQ, Quito, Ecuador.,Ingeniería en Procesos Biotecnológicos, Colegio de Ciencias Biológicas y Ambientales COCIBA, Universidad San Francisco de Quito USFQ, Quito, Ecuador
| | - Emilia Morales
- Instituto de Investigaciones en Biomedicina, Universidad San Francisco de Quito USFQ, Quito, Ecuador.,Ingeniería en Procesos Biotecnológicos, Colegio de Ciencias Biológicas y Ambientales COCIBA, Universidad San Francisco de Quito USFQ, Quito, Ecuador
| | - Andrés Caicedo
- Chemical Engineering Department, Engineering, Applied Sciences & Simulation Group (GICAS), Colegio de Ciencias e Ingenierías, Universidad San Francisco de Quito USFQ, Quito, Ecuador.,Instituto de Investigaciones en Biomedicina, Universidad San Francisco de Quito USFQ, Quito, Ecuador.,Escuela de Medicina, Colegio de Ciencias de la Salud COCSA, Universidad San Francisco de Quito USFQ, Quito, Ecuador.,Sistemas Médicos SIME, Universidad San Francisco de Quito USFQ, Quito, Ecuador.,CEDIA-USFQ Research Initiative, Corporación Ecuatoriana para el Desarrollo de la Investigación y Académica CEDIA & Universidad San Francisco de Quito USFQ, Quito, Ecuador
| | - Lourdes M Orejuela-Escobar
- Chemical Engineering Department, Engineering, Applied Sciences & Simulation Group (GICAS), Colegio de Ciencias e Ingenierías, Universidad San Francisco de Quito USFQ, Quito, Ecuador.,Instituto de Investigaciones Biológicas y Ambientales BIÓSFERA, Universidad San Francisco de Quito USFQ, Quito, Ecuador.,Instituto de Investigaciones en Biomedicina, Universidad San Francisco de Quito USFQ, Quito, Ecuador
| |
Collapse
|
132
|
Khalil AS, Jaenisch R, Mooney DJ. Engineered tissues and strategies to overcome challenges in drug development. Adv Drug Deliv Rev 2020; 158:116-139. [PMID: 32987094 PMCID: PMC7518978 DOI: 10.1016/j.addr.2020.09.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/29/2020] [Accepted: 09/23/2020] [Indexed: 12/16/2022]
Abstract
Current preclinical studies in drug development utilize high-throughput in vitro screens to identify drug leads, followed by both in vitro and in vivo models to predict lead candidates' pharmacokinetic and pharmacodynamic properties. The goal of these studies is to reduce the number of lead drug candidates down to the most likely to succeed in later human clinical trials. However, only 1 in 10 drug candidates that emerge from preclinical studies will succeed and become an approved therapeutic. Lack of efficacy or undetected toxicity represents roughly 75% of the causes for these failures, despite these parameters being the primary exclusion criteria in preclinical studies. Recently, advances in both biology and engineering have created new tools for constructing new preclinical models. These models can complement those used in current preclinical studies by helping to create more realistic representations of human tissues in vitro and in vivo. In this review, we describe current preclinical models to identify their value and limitations and then discuss select areas of research where improvements in preclinical models are particularly needed to advance drug development. Following this, we discuss design considerations for constructing preclinical models and then highlight recent advances in these efforts. Taken together, we aim to review the advances as of 2020 surrounding the prospect of biological and engineering tools for adding enhanced biological relevance to preclinical studies to aid in the challenges of failed drug candidates and the burden this poses on the drug development enterprise and thus healthcare.
Collapse
Affiliation(s)
- Andrew S Khalil
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02115, USA; Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Rudolf Jaenisch
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.
| | - David J Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02115, USA.
| |
Collapse
|
133
|
Fotopoulou E, Lykogianni M, Papadimitriou E, Mavrikou S, Machera K, Kintzios S, Thomaidou D, Aliferis ΚΑ. Mining the effect of the neonicotinoids imidacloprid and clothianidin on the chemical homeostasis and energy equilibrium of primary mouse neural stem/progenitor cells using metabolomics. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2020; 168:104617. [PMID: 32711778 DOI: 10.1016/j.pestbp.2020.104617] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/19/2020] [Accepted: 05/20/2020] [Indexed: 06/11/2023]
Abstract
The projection of plant protection products' (PPPs) toxicity to non-target organisms at early stages of their development is challenging and demanding. Recent developments in bioanalytics, however, have facilitated the study of fluctuations in the metabolism of biological systems in response to treatments with bioactives and the discovery of corresponding toxicity biomarkers. Neonicotinoids are improved insecticides that target nicotinic acetylocholine receptors (nAChR) in insects which are similar to mammals. Nonetheless, they have sparked controversy due to effects on non-target organisms. Within this context, mammalian cell cultures represent ideal systems for the development of robust models for the dissection of PPPs' toxicity. Thus, we have investigated the toxicity of imidacloprid, clothianidin, and their mixture on primary mouse (Mus musculus) neural stem/progenitor (NSPCs) and mouse neuroblastoma-derived Neuro-2a (N2a) cells, and the undergoing metabolic changes applying metabolomics. Results revealed that NSPCs, which in vitro resemble those that reside in the postnatal and adult central nervous system, are five to seven-fold more sensitive than N2a to the applied insecticides. The energy equilibrium of NSPCs was substantially altered, as it is indicated by fluctuations of metabolites involved in energy production (e.g. glucose, lactate), Krebs cycle intermediates, and fatty acids, which are important components of cell membranes. Such evidence plausibly suggests a switch of cells' energy-producing mechanism to the direct metabolism of glucose to lactate in response to insecticides. The developed pipeline could be further exploited in the discovery of unintended effects of PPPs at early steps of development and for regulatory purposes.
Collapse
Affiliation(s)
- E Fotopoulou
- Laboratory of Pesticide Science, Agricultural University of Athens, Iera Odos 75, 11855 Athens, Greece
| | - M Lykogianni
- Laboratory of Pesticide Science, Agricultural University of Athens, Iera Odos 75, 11855 Athens, Greece; Laboratory of Biological Control of Pesticides, Benaki Phytopathological Institute, St. Delta 8, 14561 Kifissia, Greece
| | - E Papadimitriou
- Neural Stem Cells and Neuroimaging Group, Neurobiology, Hellenic Pasteur Institute, Vasilissis Sofias 127, 11521 Athens, Greece
| | - S Mavrikou
- Laboratory of Cell Technology, Agricultural University of Athens, Iera Odos 75, 11855 Athens, Greece
| | - K Machera
- Laboratory of Toxicological Control of Pesticides, Benaki Phytopathological Institute, St. Delta 8, 14561 Kifissia, Greece
| | - S Kintzios
- Laboratory of Cell Technology, Agricultural University of Athens, Iera Odos 75, 11855 Athens, Greece
| | - D Thomaidou
- Neural Stem Cells and Neuroimaging Group, Neurobiology, Hellenic Pasteur Institute, Vasilissis Sofias 127, 11521 Athens, Greece.
| | - Κ Α Aliferis
- Laboratory of Pesticide Science, Agricultural University of Athens, Iera Odos 75, 11855 Athens, Greece; Department of Plant Science, McGill University, 21111 Lakeshore Road, Sainte-Anne-de-Bellevue, Quebec H9X 3V9C, Canada.
| |
Collapse
|
134
|
Ghodsi Z, Kalbassi MR, Farzaneh P, Mobarez AM, Beemelmanns C, Amiri Moghaddam J. Immunomodulatory function of antimicrobial peptide EC-Hepcidin1 modulates the induction of inflammatory gene expression in primary cells of Caspian Trout (Salmo trutta caspius Kessler, 1877). FISH & SHELLFISH IMMUNOLOGY 2020; 104:55-61. [PMID: 32473358 DOI: 10.1016/j.fsi.2020.05.067] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 05/08/2020] [Accepted: 05/25/2020] [Indexed: 06/11/2023]
Abstract
Hepcidins, a group of antimicrobial peptides (AMPs), play a key role in the innate immune system of fishes and act against different pathogens. In this study, antimicrobial and immune-inflammatory activity of a synthetic EC-hepcidin1, previously identified from orange-spotted grouper, were evaluated. EC-hepcidin1 showed weak activity against the zoonotic fish pathogen Streptococcus iniae (MIC 100 μg mL-1 and MBC 150 μg mL-1). To study the effect of AMPs in general, and EC-hepcidin1 in particular, a primary cell culture (SC) from the fin tissue of the Caspian Trout (Salmo trutta caspius) was established. The neutral Red method on SC cells revealed that EC-hepcidin1 has no or very low cytotoxic properties. Treatment of cells with either EC-hepcidin1 (150 μg mL-1) or fish pathogen Streptococcus iniae (MOI = 10) and a mixture of both resulted in the up-regulation of gene expression of MHC-UBA, IL-6, and TNFα indicating the modulatory function on inflammatory processes. These findings indicate that EC-hepcidin1 might act as a candidate for modulation of the innate immune system in S. iniae-based infection.
Collapse
Affiliation(s)
- Zohreh Ghodsi
- Department of Aquaculture, Marine Sciences Faculty, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Reza Kalbassi
- Department of Aquaculture, Marine Sciences Faculty, Tarbiat Modares University, Tehran, Iran.
| | - Parvaneh Farzaneh
- Human and Animal Cell Bank, Iranian Biological Resource Center, ACECR, Tehran, Iran
| | - Ashraf Mohebati Mobarez
- Department of Bacteriology, Medical Sciences Faculty, Tarbiat Modares University, Tehran, Iran
| | - Christine Beemelmanns
- Leibniz Institute for Natural Product Research and Infection Biology e.V. Hans-Knöll-Institute (HKI), Jena, Germany
| | - Jamshid Amiri Moghaddam
- Leibniz Institute for Natural Product Research and Infection Biology e.V. Hans-Knöll-Institute (HKI), Jena, Germany.
| |
Collapse
|
135
|
Keller F, Bruch R, Schneider R, Meier-Hubberten J, Hafner M, Rudolf R. A Scaffold-Free 3-D Co-Culture Mimics the Major Features of the Reverse Warburg Effect In Vitro. Cells 2020; 9:cells9081900. [PMID: 32823793 PMCID: PMC7463893 DOI: 10.3390/cells9081900] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/31/2020] [Accepted: 08/09/2020] [Indexed: 12/12/2022] Open
Abstract
Most tumors consume large amounts of glucose. Concepts to explain the mechanisms that mediate the achievement of this metabolic need have proposed a switch of the tumor mass to aerobic glycolysis. Depending on whether primarily tumor or stroma cells undergo such a commutation, the terms ‘Warburg effect’ or ‘reverse Warburg effect’ were coined to describe the underlying biological phenomena. However, current in vitro systems relying on 2-D culture, single cell-type spheroids, or basal-membrane extract (BME/Matrigel)-containing 3-D structures do not thoroughly reflect these processes. Here, we aimed to establish a BME/Matrigel-free 3-D microarray cancer model to recapitulate the metabolic interplay between cancer and stromal cells that allows mechanistic analyses and drug testing. Human HT-29 colon cancer and CCD-1137Sk fibroblast cells were used in mono- and co-cultures as 2-D monolayers, spheroids, and in a cell-chip format. Metabolic patterns were studied with immunofluorescence and confocal microscopy. In chip-based co-cultures, HT-29 cells showed facilitated 3-D growth and increased levels of hexokinase-2, TP53-induced glycolysis and apoptosis regulator (TIGAR), lactate dehydrogenase, and: translocase of outer mitochondrial membrane 20 (TOMM20), when compared with HT-29 mono-cultures. Fibroblasts co-cultured with HT-29 cells expressed higher levels of mono-carboxylate transporter 4, hexokinase-2, microtubule-associated proteins 1A/1B light chain 3, and ubiquitin-binding protein p62 than in fibroblast mono-cultures, in both 2-D cultures and chips. Tetramethylrhodamin-methylester (TMRM) live-cell imaging of chip co-cultures revealed a higher mitochondrial potential in cancer cells than in fibroblasts. The findings demonstrate a crosstalk between cancer cells and fibroblasts that affects cellular growth and metabolism. Chip-based 3-D co-cultures of cancer cells and fibroblasts mimicked features of the reverse Warburg effect.
Collapse
Affiliation(s)
- Florian Keller
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany; (F.K.); (R.B.); (M.H.)
- Institute of Medical Technology, Medical Faculty Mannheim of Heidelberg University and Mannheim University of Applied Sciences, 68167 Mannheim, Germany
| | - Roman Bruch
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany; (F.K.); (R.B.); (M.H.)
| | - Richard Schneider
- TIP Oncology, Merck Healthcare KGaA, 64289 Darmstadt, Germany; (R.S.); (J.M.-H.)
| | | | - Mathias Hafner
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany; (F.K.); (R.B.); (M.H.)
- Institute of Medical Technology, Medical Faculty Mannheim of Heidelberg University and Mannheim University of Applied Sciences, 68167 Mannheim, Germany
| | - Rüdiger Rudolf
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany; (F.K.); (R.B.); (M.H.)
- Institute of Medical Technology, Medical Faculty Mannheim of Heidelberg University and Mannheim University of Applied Sciences, 68167 Mannheim, Germany
- Correspondence: ; Tel.: +49-621-292-6804
| |
Collapse
|
136
|
Sakib S, Uchida A, Valenzuela-Leon P, Yu Y, Valli-Pulaski H, Orwig K, Ungrin M, Dobrinski I. Formation of organotypic testicular organoids in microwell culture†. Biol Reprod 2020; 100:1648-1660. [PMID: 30927418 DOI: 10.1093/biolre/ioz053] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 03/29/2019] [Indexed: 01/15/2023] Open
Abstract
Three-dimensional (3D) organoids can serve as an in vitro platform to study cell-cell interactions, tissue development, and toxicology. Development of organoids with tissue architecture similar to testis in vivo has remained a challenge. Here, we present a microwell aggregation approach to establish multicellular 3D testicular organoids from pig, mouse, macaque, and human. The organoids consist of germ cells, Sertoli cells, Leydig cells, and peritubular myoid cells forming a distinct seminiferous epithelium and interstitial compartment separated by a basement membrane. Sertoli cells in the organoids express tight junction proteins claudin 11 and occludin. Germ cells in organoids showed an attenuated response to retinoic acid compared to germ cells in 2D culture indicating that the tissue architecture of the organoid modulates response to retinoic acid similar to in vivo. Germ cells maintaining physiological cell-cell interactions in organoids also had lower levels of autophagy indicating lower levels of cellular stress. When organoids were treated with mono(2-ethylhexyl) phthalate (MEHP), levels of germ cell autophagy increased in a dose-dependent manner, indicating the utility of the organoids for toxicity screening. Ablation of primary cilia on testicular somatic cells inhibited the formation of organoids demonstrating an application to screen for factors affecting testicular morphogenesis. Organoids can be generated from cryopreserved testis cells and preserved by vitrification. Taken together, the testicular organoid system recapitulates the 3D organization of the mammalian testis and provides an in vitro platform for studying germ cell function, testicular development, and drug toxicity in a cellular context representative of the testis in vivo.
Collapse
Affiliation(s)
- Sadman Sakib
- Department of Comparative Biology and Experimental Medicine, University of Calgary Faculty of Veterinary Medicine, Calgary, Alberta, Canada.,Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Aya Uchida
- Department of Comparative Biology and Experimental Medicine, University of Calgary Faculty of Veterinary Medicine, Calgary, Alberta, Canada
| | - Paula Valenzuela-Leon
- Department of Comparative Biology and Experimental Medicine, University of Calgary Faculty of Veterinary Medicine, Calgary, Alberta, Canada
| | - Yang Yu
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, Alberta, Canada.,Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Hanna Valli-Pulaski
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Kyle Orwig
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Mark Ungrin
- Department of Comparative Biology and Experimental Medicine, University of Calgary Faculty of Veterinary Medicine, Calgary, Alberta, Canada.,Biomedical Engineering Graduate Program, University of Calgary, Calgary, Alberta, Canada.,Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Ina Dobrinski
- Department of Comparative Biology and Experimental Medicine, University of Calgary Faculty of Veterinary Medicine, Calgary, Alberta, Canada.,Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
137
|
Kreß S, Schaller-Ammann R, Feiel J, Priedl J, Kasper C, Egger D. 3D Printing of Cell Culture Devices: Assessment and Prevention of the Cytotoxicity of Photopolymers for Stereolithography. MATERIALS (BASEL, SWITZERLAND) 2020; 13:E3011. [PMID: 32640644 PMCID: PMC7372443 DOI: 10.3390/ma13133011] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/23/2020] [Accepted: 06/25/2020] [Indexed: 12/11/2022]
Abstract
3D printing is increasingly important for the rapid prototyping of advanced and tailor-made cell culture devices. In this context, stereolithography represents a method for the rapid generation of prototypes from photocurable polymers. However, the biocompatibility of commercially available photopolymers is largely unknown. Therefore, we evaluated the cytotoxicity of six polymers, two of them certified as biocompatible according to ISO 10993-5:2009, and we evaluated, if coating with Parylene, an inert polymer widely used in medical applications, might shield cells from the cytotoxic effects of a toxic polymer. In addition, we evaluated the processability, reliability, and consistency of the details printed. Human mesenchymal stem cells (MSCs) were used for cytotoxicity testing as they are widely used and promising for numerous applications in regenerative medicine. MSCs were incubated together with printed photopolymers, and the cytotoxicity was assessed. All photopolymers significantly reduced the viability of MSCs while the officially biocompatible resins displayed minor toxic effects. Further, coating with Parylene completely protected MSCs from toxic effects. In conclusion, none of the tested polymers can be fully recommended for rapid prototyping of cell culture devices. However, coating with Parylene can shield cells from toxic effects and thus might represent a viable option until more compatible materials are available.
Collapse
Affiliation(s)
- Sebastian Kreß
- Institute of Cell and Tissue Culture Technologies, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Muthgasse 18, 1190 Vienna, Austria; (S.K.); (C.K.)
| | - Roland Schaller-Ammann
- Health—Institute for Biomedicine and Health Sciences, JOANNEUM RESEARCH Forschungsgesellschaft mbH, Neue Stiftingtalstrasse 2, 8010 Graz, Austria; (R.S.-A.); (J.F.); (J.P.)
| | - Jürgen Feiel
- Health—Institute for Biomedicine and Health Sciences, JOANNEUM RESEARCH Forschungsgesellschaft mbH, Neue Stiftingtalstrasse 2, 8010 Graz, Austria; (R.S.-A.); (J.F.); (J.P.)
| | - Joachim Priedl
- Health—Institute for Biomedicine and Health Sciences, JOANNEUM RESEARCH Forschungsgesellschaft mbH, Neue Stiftingtalstrasse 2, 8010 Graz, Austria; (R.S.-A.); (J.F.); (J.P.)
| | - Cornelia Kasper
- Institute of Cell and Tissue Culture Technologies, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Muthgasse 18, 1190 Vienna, Austria; (S.K.); (C.K.)
| | - Dominik Egger
- Institute of Cell and Tissue Culture Technologies, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Muthgasse 18, 1190 Vienna, Austria; (S.K.); (C.K.)
| |
Collapse
|
138
|
Arora S, Sharma D, Singh J. GLUT-1: An Effective Target To Deliver Brain-Derived Neurotrophic Factor Gene Across the Blood Brain Barrier. ACS Chem Neurosci 2020; 11:1620-1633. [PMID: 32352752 DOI: 10.1021/acschemneuro.0c00076] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Alzheimer's disease (AD), the most common cause of dementia, inflicts enormous suffering to patients and their family members. It is the third deadliest disease, affecting 46.8 million people worldwide. Brain-derived neurotrophic factor (BDNF) is involved in the development, maintenance, and plasticity of the central nervous system. This crucial protein is significantly reduced in AD patients leading to reduced plasticity and neuronal death. In this study, we demonstrate the targeted delivery of the BDNF gene to the brain using liposome nanoparticles. These liposomes were surface modified with glucose transporter-1 targeting ligand (mannose) and cell penetrating peptides (penetratin or rabies virus glycoprotein) to promote selective and enhanced delivery to the brain. Surface modified liposomes showed significantly higher transfection of BDNF in primary astrocytes and neurons, compared to unmodified (plain) liposomes. BDNF transfection via dual modified liposomes resulted in an increase in presynaptic marker synaptophysin protein in primary neuronal cells, which is usually found to be reduced in AD patients. Liposomes surface modified with mannose and cell penetrating peptides demonstrated ∼50% higher transport across the in vitro blood brain barrier (BBB) model and showed significantly higher transfection efficiency in primary neuronal cells compared to plain liposomes. These results were correlated with significantly higher transport of surface modified liposomes (∼7% of injected dose/gram of tissue) and BDNF transfection (∼1.7 times higher than baseline level) across BBB following single intravenous administration in C57BL/6 mice without any signs of inflammation or toxicity. Overall, this study suggests a safe and targeted strategy to increase BDNF protein in the brain, which has the potential to reverse AD pathophysiology.
Collapse
Affiliation(s)
- Sanjay Arora
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Divya Sharma
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Jagdish Singh
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, North Dakota 58105, United States
| |
Collapse
|
139
|
Potts RW, Gutierrez AP, Cortés-Araya Y, Houston RD, Bean TP. Developments in marine invertebrate primary culture reveal novel cell morphologies in the model bivalve Crassostrea gigas. PeerJ 2020; 8:e9180. [PMID: 32547861 PMCID: PMC7271890 DOI: 10.7717/peerj.9180] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 04/22/2020] [Indexed: 01/23/2023] Open
Abstract
Cell culture provides useful model systems used in a wide range of biological applications, but its utility in marine invertebrates is limited due to the lack of immortalised cell lines. Primary cell and tissue cultures are typically used but remain poorly characterised for oysters, which can cause issues with experimental consistency and reproducibility. Improvements to methods of repeatable isolation, culture, and characterisation of oyster cells and tissues are required to help address these issues. In the current study, systematic improvements have been developed to facilitate the culture of primary cells from adult Pacific oyster tissues and identify novel cell morphologies that have not been reported previously. Cultures analysed by light microscopy, qPCR, and live cell imaging demonstrated maintenance of live, metabolically active Pacific oyster cells for several weeks post-explant. Interestingly, whole hearts dissected from adult oysters were found to continue contracting rhythmically up to 8 weeks after being transferred to a tissue culture system. Mantle tissue explants were also actively moving in the culture system. These improvements in primary cell culture of bivalves may be beneficial for research in ecotoxicology, virology, immunology, and genetic resistance to disease.
Collapse
Affiliation(s)
- Robert W.A. Potts
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom,Centre for Environment Fisheries and Aquaculture Science (Cefas) Weymouth Laboratory, Dorset, United Kingdom
| | - Alejandro P. Gutierrez
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Yennifer Cortés-Araya
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Ross D. Houston
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Tim P. Bean
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
140
|
Eckhardt M, Hultquist JF, Kaake RM, Hüttenhain R, Krogan NJ. A systems approach to infectious disease. Nat Rev Genet 2020; 21:339-354. [PMID: 32060427 PMCID: PMC7839161 DOI: 10.1038/s41576-020-0212-5] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/08/2020] [Indexed: 01/01/2023]
Abstract
Ongoing social, political and ecological changes in the 21st century have placed more people at risk of life-threatening acute and chronic infections than ever before. The development of new diagnostic, prophylactic, therapeutic and curative strategies is critical to address this burden but is predicated on a detailed understanding of the immensely complex relationship between pathogens and their hosts. Traditional, reductionist approaches to investigate this dynamic often lack the scale and/or scope to faithfully model the dual and co-dependent nature of this relationship, limiting the success of translational efforts. With recent advances in large-scale, quantitative omics methods as well as in integrative analytical strategies, systems biology approaches for the study of infectious disease are quickly forming a new paradigm for how we understand and model host-pathogen relationships for translational applications. Here, we delineate a framework for a systems biology approach to infectious disease in three parts: discovery - the design, collection and analysis of omics data; representation - the iterative modelling, integration and visualization of complex data sets; and application - the interpretation and hypothesis-based inquiry towards translational outcomes.
Collapse
Affiliation(s)
- Manon Eckhardt
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA
- Quantitative Biosciences Institute, University of California, San Francisco, San Francisco, CA, USA
- J. David Gladstone Institutes, San Francisco, CA, USA
| | - Judd F Hultquist
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.
- Quantitative Biosciences Institute, University of California, San Francisco, San Francisco, CA, USA.
- J. David Gladstone Institutes, San Francisco, CA, USA.
- Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| | - Robyn M Kaake
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA
- Quantitative Biosciences Institute, University of California, San Francisco, San Francisco, CA, USA
- J. David Gladstone Institutes, San Francisco, CA, USA
| | - Ruth Hüttenhain
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA
- Quantitative Biosciences Institute, University of California, San Francisco, San Francisco, CA, USA
- J. David Gladstone Institutes, San Francisco, CA, USA
| | - Nevan J Krogan
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.
- Quantitative Biosciences Institute, University of California, San Francisco, San Francisco, CA, USA.
- J. David Gladstone Institutes, San Francisco, CA, USA.
| |
Collapse
|
141
|
Chen L, Yang Y, Ueno H, Esch MB. Body-in-a-Cube: a microphysiological system for multi-tissue co-culture with near-physiological amounts of blood surrogate. MICROPHYSIOLOGICAL SYSTEMS 2020; 4:10.21037/mps-19-8. [PMID: 34131641 PMCID: PMC8201523 DOI: 10.21037/mps-19-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND Decreasing the amount of liquid inside microphysiological systems (MPS) can help uncover the presence of toxic drug metabolites. However, maintaining near-physiological volume ratios among blood surrogate and multiple organ mimics is technically challenging. Here, we developed a body cube and tested its ability to support four human tissues (kidney, GI tract, liver, and bone marrow) scaled down from in vivo functional volumes by a factor of 73,000 with 80 μL of cell culture medium (corresponding to ~1/73000th of in vivo blood volume). METHODS GI tract cells (Caco-2), liver cells (HepG2/C3A), bone marrow cells (Meg-01), and kidney cells (HK-2) were co-cultured inside the body cube with 80 μL of common, recirculating cell culture medium for 72 h. The system was challenged with acetaminophen and troglitazone, and concentrations of aspartate aminotransferase (AST), albumin, and urea were monitored over time. RESULTS Cell viability analysis showed that 95.5%±3.2% of liver cells, 89.8%±4.7% of bone marrow cells, 82.8%±8.1% of GI tract cells, and 80.1%±11.5% of kidney cells were viable in co-culture for 72 h. Both acetaminophen and troglitazone significantly lowered cell viability in the liver chamber as indicated by viability analysis and a temporary increase of AST in the cell culture medium. Both drugs also lowered urea production in the liver by up to 45%. CONCLUSIONS Cell viability data and the production of urea and albumin indicate that the co-culture of GI tract, liver, bone marrow, and kidney tissues with near-physiological volume ratios of tissues to blood surrogate is possible for up to 72 h. The body-cube was capable of reproducing liver toxicity to HepG2/C3A liver cells via acetaminophen and troglitazone. The developed design provides a viable format for acute toxicity testing with near-physiological blood surrogate to tissue volume ratios.
Collapse
Affiliation(s)
- Longyi Chen
- Biomedical Technologies Group, Physical Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, MD, USA
- Institute for Research in Electronics and Applied Physics & Maryland NanoCenter, University of Maryland, College Park, MD, USA
| | - Yang Yang
- Biomedical Technologies Group, Physical Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, MD, USA
- Institute for Research in Electronics and Applied Physics & Maryland NanoCenter, University of Maryland, College Park, MD, USA
| | - Hidetaka Ueno
- Biomedical Technologies Group, Physical Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, MD, USA
- Biosensing Research Group, Health and Medical Research Institute, National Institute of Advanced Industrial Science and Technology, Takamatsu, Japan
| | - Mandy B. Esch
- Biomedical Technologies Group, Physical Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, MD, USA
| |
Collapse
|
142
|
Novel 2D and 3D Assays to Determine the Activity of Anti-Leishmanial Drugs. Microorganisms 2020; 8:microorganisms8060831. [PMID: 32492796 PMCID: PMC7356592 DOI: 10.3390/microorganisms8060831] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/21/2020] [Accepted: 05/25/2020] [Indexed: 12/17/2022] Open
Abstract
The discovery of novel anti-leishmanial compounds remains essential as current treatments have known limitations and there are insufficient novel compounds in development. We have investigated three complex and physiologically relevant in vitro assays, including: (i) a media perfusion based cell culture model, (ii) two 3D cell culture models, and (iii) iPSC derived macrophages in place of primary macrophages or cell lines, to determine whether they offer improved approaches to anti-leishmanial drug discovery and development. Using a Leishmania major amastigote-macrophage assay the activities of standard drugs were investigated to show the effect of changing parameters in these assays. We determined that drug activity was reduced by media perfusion (EC50 values for amphotericin B shifted from 54 (51–57) nM in the static system to 70 (61–75) nM under media perfusion; EC50 values for miltefosine shifted from 12 (11–15) µM in the static system to 30 (26–34) µM under media perfusion) (mean and 95% confidence intervals), with corresponding reduced drug accumulation by macrophages. In the 3D cell culture model there was a significant difference in the EC50 values of amphotericin B but not miltefosine (EC50 values for amphotericin B were 34.9 (31.4–38.6) nM in the 2D and 52.3 (46.6–58.7) nM in 3D; EC50 values for miltefosine were 5.0 (4.9–5.2) µM in 2D and 5.9 (5.5–6.2) µM in 3D (mean and 95% confidence intervals). Finally, in experiments using iPSC derived macrophages infected with Leishmania, reported here for the first time, we observed a higher level of intracellular infection in iPSC derived macrophages compared to the other macrophage types for four different species of Leishmania studied. For L. major with an initial infection ratio of 0.5 parasites per host cell the percentage infection level of the macrophages after 72 h was 11.3% ± 1.5%, 46.0% ± 1.4%, 66.4% ± 3.5% and 75.1% ± 2.4% (average ± SD) for the four cells types, THP1 a human monocytic cell line, mouse bone marrow macrophages (MBMMs), human bone marrow macrophages (HBMMs) and iPSC derived macrophages respectively. Despite the higher infection levels, drug activity in iPSC derived macrophages was similar to that in other macrophage types, for example, amphotericin B EC50 values were 35.9 (33.4–38.5), 33.5 (31.5–36.5), 33.6 (30.5—not calculated (NC)) and 46.4 (45.8–47.2) nM in iPSC, MBMMs, HBMMs and THP1 cells respectively (mean and 95% confidence intervals). We conclude that increasing the complexity of cellular assays does impact upon anti-leishmanial drug activities but not sufficiently to replace the current model used in HTS/HCS assays in drug discovery programmes. The impact of media perfusion on drug activities and the use of iPSC macrophages do, however, deserve further investigation.
Collapse
|
143
|
SPON1 Can Reduce Amyloid Beta and Reverse Cognitive Impairment and Memory Dysfunction in Alzheimer's Disease Mouse Model. Cells 2020; 9:cells9051275. [PMID: 32455709 PMCID: PMC7290723 DOI: 10.3390/cells9051275] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/08/2020] [Accepted: 05/18/2020] [Indexed: 01/07/2023] Open
Abstract
Alzheimer’s disease (AD) is a complex, age-related neurodegenerative disease that is the most common form of dementia. However, the cure for AD has not yet been founded. The accumulation of amyloid beta (Aβ) is considered to be a hallmark of AD. Beta-site amyloid precursor protein cleaving enzyme 1 (BACE1), also known as beta secretase is the initiating enzyme in the amyloidogenic pathway. Blocking BACE1 could reduce the amount of Aβ, but this would also prohibit the other functions of BACE1 in brain physiological activity. SPONDIN1 (SPON1) is known to bind to the BACE1 binding site of the amyloid precursor protein (APP) and blocks the initiating amyloidogenesis. Here, we show the effect of SPON1 in Aβ reduction in vitro in neural cells and in an in vivo AD mouse model. We engineered mouse induced neural stem cells (iNSCs) to express Spon1. iNSCs harboring mouse Spon1 secreted SPON1 protein and reduced the quantity of Aβ when co-cultured with Aβ-secreting Neuro 2a cells. The human SPON1 gene itself also reduced Aβ in HEK 293T cells expressing the human APP transgene with AD-linked mutations through lentiviral-mediated delivery. We also demonstrated that injecting SPON1 reduced the amount of Aβ and ameliorated cognitive dysfunction and memory impairment in 5xFAD mice expressing human APP and PSEN1 transgenes with five AD-linked mutations.
Collapse
|
144
|
Choi KYG, Wu BC, Lee AHY, Baquir B, Hancock REW. Utilizing Organoid and Air-Liquid Interface Models as a Screening Method in the Development of New Host Defense Peptides. Front Cell Infect Microbiol 2020; 10:228. [PMID: 32509598 PMCID: PMC7251080 DOI: 10.3389/fcimb.2020.00228] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 04/23/2020] [Indexed: 12/24/2022] Open
Abstract
Host defense peptides (HDPs), also known as antimicrobial peptides, are naturally occurring polypeptides (~12–50 residues) composed of cationic and hydrophobic amino acids that adopt an amphipathic conformation upon folding usually after contact with membranes. HDPs have a variety of biological activities including immunomodulatory, anti-inflammatory, anti-bacterial, and anti-biofilm functions. Although HDPs have the potential to address the global threat of antibiotic resistance and to treat immune and inflammatory disorders, they have yet to achieve this promise. Indeed, there are several challenges associated with bringing peptide-based drug candidates from the lab bench to clinical practice, including identifying appropriate indications, stability, toxicity, and cost. These challenges can be addressed in part by the development of innate defense regulator (IDR) peptides and peptidomimetics, which are synthetic derivatives of HDPs with similar or better efficacy, increased stability, and reduced toxicity and cost of the original HDP. However, one of the largest gaps between basic research and clinical application is the validity and translatability of conventional model systems, such as cell lines and animal models, for screening HDPs and their derivatives as potential drug therapies. Indeed, such translation has often relied on animal models, which have only limited validity. Here we discuss the recent development of human organoids for disease modeling and drug screening, assisted by the use of omics analyses. Organoids, developed from primary cells, cell lines, or human pluripotent stem cells, are three-dimensional, self-organizing structures that closely resemble their corresponding in vivo organs with regards to immune responses, tissue organization, and physiological properties; thus, organoids represent a reliable method for studying efficacy, formulation, toxicity and to some extent drug stability and pharmacodynamics. The use of patient-derived organoids enables the study of patient-specific efficacy, toxicogenomics and drug response predictions. We outline how organoids and omics data analysis can be leveraged to aid in the clinical translation of IDR peptides.
Collapse
Affiliation(s)
- Ka-Yee Grace Choi
- Department of Microbiology and Immunology, Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Bing Catherine Wu
- Department of Microbiology and Immunology, Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Amy Huei-Yi Lee
- Department of Microbiology and Immunology, Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada.,Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Beverlie Baquir
- Department of Microbiology and Immunology, Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Robert E W Hancock
- Department of Microbiology and Immunology, Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
145
|
Isolation, culture, and downstream characterization of primary microglia and astrocytes from adult rodent brain and spinal cord. J Neurosci Methods 2020; 340:108742. [PMID: 32315669 DOI: 10.1016/j.jneumeth.2020.108742] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 04/10/2020] [Accepted: 04/14/2020] [Indexed: 01/01/2023]
Abstract
BACKGROUND Neuroimmunologists aspire to understand the interactions between neurons, microglia, and astrocytes in the CNS. To study these cells, researchers work with either immortalized cell lines or primary cells acquired from animal tissue. Primary cells reflect in vivo characteristics and functionality compared to immortalized cells; however, they are challenging to acquire and maintain. NEW METHOD Established protocols to harvest primary glia use neonatal rodents, here we provide a method for simultaneously isolating microglia and astrocytes from brain and/or spinal cord from adult rodents. We utilized a discontinuous percoll density gradient enabling easy discrimination of these cell populations without enzymatic digestion or complex sorting techniques. RESULTS We found cells isolated from the percoll interface between 70 %-50 % were microglia, as they express ionizing calcium-binding adaptor molecule 1 (Iba1) in immunocytochemistry and CD11bhi and CD45lo using flow cytometry. Isolated cells from the 50 %-30 % interface were astrocytes as they express glial fibrillary acidic protein (GFAP) in immunocytochemistry and Glutamate aspartate transporter (GLAST)-1 using flow cytometry. Cultured microglia and astrocytes showed a functional increase in IL-6 production after treatment of lipopolysaccharide (LPS). COMPARISON WITH EXISTING METHODS Our method allows for rapid isolation of both microglia and astrocytes in one protocol with relatively few resources, preserves cellular phenotype, and yields high cell numbers without magnetic or antibody sorting. CONCLUSION Here we show a novel, single protocol to isolate microglia and astrocytes from brain and spinal cord tissue, allowing for culturing and other downstream applications from the cells of animals of various ages, which will be useful for researchers investigating these two major glial cell types from the brain or spinal cord of the same rodent.
Collapse
|
146
|
Annageldiyev C, Tan SF, Thakur S, Dhanyamraju PK, Ramisetti SR, Bhadauria P, Schick J, Zeng Z, Sharma V, Dunton W, Dovat S, Desai D, Zheng H, Feith DJ, Loughran TP, Amin S, Sharma AK, Claxton D, Sharma A. The PI3K/AKT Pathway Inhibitor ISC-4 Induces Apoptosis and Inhibits Growth of Leukemia in Preclinical Models of Acute Myeloid Leukemia. Front Oncol 2020; 10:393. [PMID: 32296637 PMCID: PMC7140985 DOI: 10.3389/fonc.2020.00393] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 03/05/2020] [Indexed: 12/12/2022] Open
Abstract
Acute myeloid leukemia is a heterogeneous disease with a 5-year survival rate of 28.3%, and current treatment options constrained by dose-limiting toxicities. One of the key signaling pathways known to be frequently activated and dysregulated in AML is PI3K/AKT. Its dysregulation is associated with aggressive cell growth and drug resistance. We investigated the activity of Phenybutyl isoselenocyanate (ISC-4) in primary cells obtained from newly diagnosed AML patients, diverse AML cell lines, and normal cord blood cells. ISC-4 significantly inhibited survival and clonogenicity of primary human AML cells without affecting normal cells. We demonstrated that ISC-4-mediated p-Akt inhibition caused apoptosis in primary AML (CD34+) stem cells and enhanced efficacy of cytarabine. ISC-4 impeded leukemia progression with improved overall survival in a syngeneic C1498 mouse model with no obvious toxic effects on normal myelopoiesis. In U937 xenograft model, bone marrow cells exhibited significant reduction in human CD45+ cells in ISC-4 (~87%) or AraC (~89%) monotherapy groups compared to control. Notably, combination treatment suppressed the leukemic infiltration significantly higher than the single-drug treatments (~94%). Together, the present findings suggest that ISC-4 might be a promising agent for AML treatment.
Collapse
Affiliation(s)
- Charyguly Annageldiyev
- Division of Hematology and Oncology, Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA, United States.,Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Su-Fern Tan
- Division of Hematology and Oncology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Shreya Thakur
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Pavan Kumar Dhanyamraju
- Division of Hematology and Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Srinivasa R Ramisetti
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Preeti Bhadauria
- Division of Hematology and Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Jacob Schick
- Division of Hematology and Oncology, Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Zheng Zeng
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Varun Sharma
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Wendy Dunton
- Division of Hematology and Oncology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Sinisa Dovat
- Division of Hematology and Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Dhimant Desai
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, United States.,Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Hong Zheng
- Division of Hematology and Oncology, Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA, United States.,Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - David J Feith
- Division of Hematology and Oncology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States.,Division of Hematology and Oncology, Department of Medicine, University of Virginia Cancer Center, Charlottesville, VA, United States
| | - Thomas P Loughran
- Division of Hematology and Oncology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States.,Division of Hematology and Oncology, Department of Medicine, University of Virginia Cancer Center, Charlottesville, VA, United States
| | - Shantu Amin
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, United States.,Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Arun K Sharma
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, United States.,Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - David Claxton
- Division of Hematology and Oncology, Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA, United States.,Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Arati Sharma
- Division of Hematology and Oncology, Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA, United States.,Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, United States.,Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| |
Collapse
|
147
|
Cui Y, Zhang M, Leng C, Blokzijl T, Jansen BH, Dijkstra G, Faber KN. Pirfenidone Inhibits Cell Proliferation and Collagen I Production of Primary Human Intestinal Fibroblasts. Cells 2020; 9:cells9030775. [PMID: 32235767 PMCID: PMC7140656 DOI: 10.3390/cells9030775] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 03/16/2020] [Accepted: 03/20/2020] [Indexed: 12/12/2022] Open
Abstract
Intestinal fibrosis is a common complication of inflammatory bowel disease. So far, there is no safe and effective drug for intestinal fibrosis. Pirfenidone is an anti-fibrotic compound available for the treatment of idiopathic pulmonary fibrosis. Here, we explored the anti-proliferative and anti-fibrotic properties of pirfenidone on primary human intestinal fibroblasts (p-hIFs). p-hIFs were cultured in the absence and presence of pirfenidone. Cell proliferation was measured by a real-time cell analyzer (xCELLigence) and BrdU incorporation. Cell motility was monitored by live cell imaging. Cytotoxicity and cell viability were analyzed by Sytox green, Caspase-3 and Water Soluble Tetrazolium Salt-1 (WST-1) assays. Gene expression of fibrosis markers was determined by quantitative reverse transcription PCR (RT-qPCR). The mammalian target of rapamycin (mTOR) signaling was analyzed by Western blotting and type I collagen protein expression additionally by immunofluorescence microscopy. Pirfenidone dose-dependently inhibited p-hIF proliferation and motility, without inducing cell death. Pirfenidone suppressed mRNA levels of genes that contribute to extracellular matrix production, as well as basal and TGF-β1-induced collagen I protein production, which was associated with inhibition of the rapamycin-sensitive mTOR/p70S6K pathway in p-hIFs. Thus, pirfenidone inhibits the proliferation of intestinal fibroblasts and suppresses collagen I production through the TGF-β1/mTOR/p70S6K signaling pathway, which might be a novel and safe anti-fibrotic strategy to treat intestinal fibrosis.
Collapse
Affiliation(s)
- Yingying Cui
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (Y.C.); (M.Z.); (T.B.); (B.H.J.); (G.D.)
| | - Mengfan Zhang
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (Y.C.); (M.Z.); (T.B.); (B.H.J.); (G.D.)
| | - Changsen Leng
- Department of Biomedical Sciences of Cells and Systems, section Molecular Cell Biology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands;
| | - Tjasso Blokzijl
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (Y.C.); (M.Z.); (T.B.); (B.H.J.); (G.D.)
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Bernadien H. Jansen
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (Y.C.); (M.Z.); (T.B.); (B.H.J.); (G.D.)
| | - Gerard Dijkstra
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (Y.C.); (M.Z.); (T.B.); (B.H.J.); (G.D.)
| | - Klaas Nico Faber
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (Y.C.); (M.Z.); (T.B.); (B.H.J.); (G.D.)
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
- Correspondence: ; Tel.: +31-50-3612364
| |
Collapse
|
148
|
Abstract
High-throughput in vitro models lack human-relevant complexity, which undermines their ability to accurately mimic in vivo biologic and pathologic responses. The emergence of microphysiological systems (MPS) presents an opportunity to revolutionize in vitro modeling for both basic biomedical research and applied drug discovery. The MPS platform has been an area of interdisciplinary collaboration to develop new, predictive, and reliable in vitro methods for regulatory acceptance. The current MPS models have been developed to recapitulate an organ or tissue on a smaller scale. However, the complexity of these models (ie, including all cell types present in the in vivo tissue) with appropriate structural, functional, and biochemical attributes are often not fully characterized. Here, we provide an overview of the capabilities and limitations of the microfluidic MPS model (aka organs-on-chips) within the scope of drug development. We recommend the engagement of pathologists early in the MPS design, characterization, and validation phases, because this will enable development of more robust and comprehensive MPS models that can accurately replicate normal biology and pathophysiology and hence be more predictive of human responses.
Collapse
Affiliation(s)
| | - Terry Van Vleet
- Global Preclinical Safety, AbbVie Inc, North Chicago, IL, USA
| | - Brian R Berridge
- National Toxicology Program, The National Institute of Environmental Health Sciences, Durham, NC, USA
| |
Collapse
|
149
|
Feng F, Huang C, Xiao M, Wang H, Gao Q, Chen Z, Xu X, Zhou J, Li F, Li Y, Zhang D, Chang Y, Jiang X. Establishment and characterization of patient-derived primary cell lines as preclinical models for gallbladder carcinoma. Transl Cancer Res 2020; 9:1698-1710. [PMID: 35117518 PMCID: PMC8798768 DOI: 10.21037/tcr.2020.02.04] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 01/18/2020] [Indexed: 01/18/2023]
Abstract
Background Gallbladder carcinoma (GBC) is one of the most lethal malignancies which do not have a targeted drug in the clinic. Patient-derived primary cell lines (PDCs) are useful in assessment of cancer complexity and heterogeneity, drug-sensitivity tests, and personalized-drug-selection guidance. The aim of this study is to establish GBC PDCs and characterize their biological features. Methods The characterization of PDCs was defined by morphology, growth kinetics, chromosomal analysis, short tandem repeat (STR) analysis, RNA-seq and tumorigenicity. Glycosylation of PDCs derived from GBC was first studied, and the PDC model’s performance were also tested and evaluated using seven molecular target inhibitors. Results Three novel GBC cell lines from three GBC patients were successfully established and denoted as JXQ-3D-902R4, JXQ-3D-4494R, and JXQ-3D-4786R. These cell lines demonstrated the heterogeneous characteristics of tumor morphology and phenotypes which are consistent with primary GBC, such as irregular cell shape, varied chromosomal numbers, and different STR patterns. Moreover, the growth activity and tumorigenicity ability varied among the cell lines, of which JXQ-3D-4494R exhibited the best growth rate. Furthermore, glycan profiling of whole proteins were detected and characterized. Unique N-glycans of each PDC were identified, JXQ-3D-902R4, JXQ-3D-4494R and JXQ-3D-4786R contained ten, four and seven unique glycans, respectively. The epithelial origins of three PDCs were confirmed using RNA-seq based on the highly expressed typical epithelial marker genes. Moreover, the drug-sensitivity results demonstrated that the three PDCs exhibited different responses to the seven-most commonly used targeted medicines belonging to three groups: cell-cycle inhibitors, PI3K/AKT/mTOR signaling-pathway inhibitors, and ErbB inhibitors. JXQ-3D-4494R was sensitive to most of the inhibitors, JXQ-3D-4786R was sensitive to ErbB inhibitors, and JXQ-3D-902R4 was sensitive to PI3K/AKT/mTOR inhibitors. Conclusions These results indicate that PDCs may be efficient preclinical models for further investigation of the biological behaviors and potential targeted therapies of human GBC.
Collapse
Affiliation(s)
- Feiling Feng
- Department of Biliary I, Shanghai Eastern Hepatobiliary Surgery Hospital, Navy Military Medical University, Shanghai 200438, China
| | - Chuncui Huang
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Mingjia Xiao
- Department of Biliary I, Shanghai Eastern Hepatobiliary Surgery Hospital, Navy Military Medical University, Shanghai 200438, China
| | - Huizhen Wang
- Research and Development Institute of Precision Medicine, 3D Medicines Inc., Shanghai 201114, China
| | - Qingxiang Gao
- Department of Biliary I, Shanghai Eastern Hepatobiliary Surgery Hospital, Navy Military Medical University, Shanghai 200438, China
| | - Zishuo Chen
- Research and Development Institute of Precision Medicine, 3D Medicines Inc., Shanghai 201114, China
| | - Xiaoya Xu
- Research and Development Institute of Precision Medicine, 3D Medicines Inc., Shanghai 201114, China
| | - Jun Zhou
- Research and Development Institute of Precision Medicine, 3D Medicines Inc., Shanghai 201114, China
| | - Fugen Li
- Research and Development Institute of Precision Medicine, 3D Medicines Inc., Shanghai 201114, China
| | - Yan Li
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Dadong Zhang
- Research and Development Institute of Precision Medicine, 3D Medicines Inc., Shanghai 201114, China
| | - Yanxin Chang
- Biliary Tract Surgery Department, Shanghai Eastern Hepatobiliary Surgery Hospital, Navy Military Medical University, Shanghai 200438, China
| | - Xiaoqing Jiang
- Department of Biliary I, Shanghai Eastern Hepatobiliary Surgery Hospital, Navy Military Medical University, Shanghai 200438, China
| |
Collapse
|
150
|
Kuhn TC, Knobel J, Burkert-Rettenmaier S, Li X, Meyer IS, Jungmann A, Sicklinger F, Backs J, Lasitschka F, Müller OJ, Katus HA, Krijgsveld J, Leuschner F. Secretome Analysis of Cardiomyocytes Identifies PCSK6 (Proprotein Convertase Subtilisin/Kexin Type 6) as a Novel Player in Cardiac Remodeling After Myocardial Infarction. Circulation 2020; 141:1628-1644. [PMID: 32100557 DOI: 10.1161/circulationaha.119.044914] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Acute occlusion of a coronary artery results in swift tissue necrosis. Bordering areas of the infarcted myocardium can also experience impaired blood supply and reduced oxygen delivery, leading to altered metabolic and mechanical processes. Although transcriptional changes in hypoxic cardiomyocytes are well studied, little is known about the proteins that are actively secreted from these cells. METHODS We established a novel secretome analysis of cardiomyocytes by combining stable isotope labeling and click chemistry with subsequent mass spectrometry analysis. Further functional validation experiments included ELISA measurement of human samples, murine left anterior descending coronary artery ligation, and adeno-associated virus 9-mediated in vivo overexpression in mice. RESULTS The presented approach is feasible for analysis of the secretome of primary cardiomyocytes without serum starvation. A total of 1026 proteins were identified to be secreted within 24 hours, indicating a 5-fold increase in detection compared with former approaches. Among them, a variety of proteins have not yet been explored in the context of cardiovascular pathologies. One of the secreted factors most strongly upregulated upon hypoxia was PCSK6 (proprotein convertase subtilisin/kexin type 6). Validation experiments revealed an increase of PCSK6 on mRNA and protein level in hypoxic cardiomyocytes. PCSK6 expression was elevated in hearts of mice after 3 days of ligation of the left anterior descending artery, a finding confirmed by immunohistochemistry. ELISA measurements in human serum also indicate distinct kinetics for PCSK6 in patients with acute myocardial infarction, with a peak on postinfarction day 3. Transfer of PCSK6-depleted cardiomyocyte secretome resulted in decreased expression of collagen I and III in fibroblasts compared with control treated cells, and small interfering RNA-mediated knockdown of PCSK6 in cardiomyocytes impacted transforming growth factor-β activation and SMAD3 (mothers against decapentaplegic homolog 3) translocation in fibroblasts. An adeno-associated virus 9-mediated, cardiomyocyte-specific overexpression of PCSK6 in mice resulted in increased collagen expression and cardiac fibrosis, as well as decreased left ventricular function, after myocardial infarction. CONCLUSIONS A novel mass spectrometry-based approach allows investigation of the secretome of primary cardiomyocytes. Analysis of hypoxia-induced secretion led to the identification of PCSK6 as being crucially involved in cardiac remodeling after acute myocardial infarction.
Collapse
Affiliation(s)
- Tim Christian Kuhn
- Department of Cardiology, Medical University Hospital, Heidelberg, Germany (T.C.K., J.K., S.B-R., X.L., I.S.M., A.J., F.S., H.A.K., F.L.).,DZHK (German Centre for Cardiovascular Research), Heidelberg, Germany (T.C.K., J.K., S.B-R., X.L., I.S.M., A.J., F.S., J.B., H.A.K., F.L.)
| | - Johannes Knobel
- Department of Cardiology, Medical University Hospital, Heidelberg, Germany (T.C.K., J.K., S.B-R., X.L., I.S.M., A.J., F.S., H.A.K., F.L.).,DZHK (German Centre for Cardiovascular Research), Heidelberg, Germany (T.C.K., J.K., S.B-R., X.L., I.S.M., A.J., F.S., J.B., H.A.K., F.L.)
| | - Sonja Burkert-Rettenmaier
- Department of Cardiology, Medical University Hospital, Heidelberg, Germany (T.C.K., J.K., S.B-R., X.L., I.S.M., A.J., F.S., H.A.K., F.L.).,DZHK (German Centre for Cardiovascular Research), Heidelberg, Germany (T.C.K., J.K., S.B-R., X.L., I.S.M., A.J., F.S., J.B., H.A.K., F.L.)
| | - Xue Li
- Department of Cardiology, Medical University Hospital, Heidelberg, Germany (T.C.K., J.K., S.B-R., X.L., I.S.M., A.J., F.S., H.A.K., F.L.).,DZHK (German Centre for Cardiovascular Research), Heidelberg, Germany (T.C.K., J.K., S.B-R., X.L., I.S.M., A.J., F.S., J.B., H.A.K., F.L.)
| | - Ingmar Sören Meyer
- Department of Cardiology, Medical University Hospital, Heidelberg, Germany (T.C.K., J.K., S.B-R., X.L., I.S.M., A.J., F.S., H.A.K., F.L.).,DZHK (German Centre for Cardiovascular Research), Heidelberg, Germany (T.C.K., J.K., S.B-R., X.L., I.S.M., A.J., F.S., J.B., H.A.K., F.L.)
| | - Andreas Jungmann
- Department of Cardiology, Medical University Hospital, Heidelberg, Germany (T.C.K., J.K., S.B-R., X.L., I.S.M., A.J., F.S., H.A.K., F.L.).,DZHK (German Centre for Cardiovascular Research), Heidelberg, Germany (T.C.K., J.K., S.B-R., X.L., I.S.M., A.J., F.S., J.B., H.A.K., F.L.)
| | - Florian Sicklinger
- Department of Cardiology, Medical University Hospital, Heidelberg, Germany (T.C.K., J.K., S.B-R., X.L., I.S.M., A.J., F.S., H.A.K., F.L.).,DZHK (German Centre for Cardiovascular Research), Heidelberg, Germany (T.C.K., J.K., S.B-R., X.L., I.S.M., A.J., F.S., J.B., H.A.K., F.L.)
| | - Johannes Backs
- DZHK (German Centre for Cardiovascular Research), Heidelberg, Germany (T.C.K., J.K., S.B-R., X.L., I.S.M., A.J., F.S., J.B., H.A.K., F.L.).,Department of Molecular Cardiology and Epigenetics, Heidelberg, Germany (J.B.)
| | - Felix Lasitschka
- Institute of Pathology, University of Heidelberg, Germany (Fe.L.)
| | - Oliver J Müller
- Department of Internal Medicine III, University of Kiel, Germany (O.J.M.)
| | - Hugo A Katus
- Department of Cardiology, Medical University Hospital, Heidelberg, Germany (T.C.K., J.K., S.B-R., X.L., I.S.M., A.J., F.S., H.A.K., F.L.).,DZHK (German Centre for Cardiovascular Research), Heidelberg, Germany (T.C.K., J.K., S.B-R., X.L., I.S.M., A.J., F.S., J.B., H.A.K., F.L.)
| | - Jeroen Krijgsveld
- Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany (Je.K.).,Heidelberg University, Medical Faculty, Germany (Je.K.)
| | - Florian Leuschner
- Department of Cardiology, Medical University Hospital, Heidelberg, Germany (T.C.K., J.K., S.B-R., X.L., I.S.M., A.J., F.S., H.A.K., F.L.).,DZHK (German Centre for Cardiovascular Research), Heidelberg, Germany (T.C.K., J.K., S.B-R., X.L., I.S.M., A.J., F.S., J.B., H.A.K., F.L.)
| |
Collapse
|