1
|
Gill A, Kinghorn K, Bautch VL, Mac Gabhann F. Mechanistic computational modeling of sFLT1 secretion dynamics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.12.637983. [PMID: 40027776 PMCID: PMC11870409 DOI: 10.1101/2025.02.12.637983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Constitutively secreted by endothelial cells, soluble FLT1 (sFLT1 or sVEGFR1) binds and sequesters extracellular vascular endothelial growth factors (VEGF), thereby reducing VEGF binding to VEGF receptor tyrosine kinases and their downstream signaling. In doing so, sFLT1 plays an important role in vascular development and in the patterning of new blood vessels in angiogenesis. Here, we develop multiple mechanistic models of sFLT1 secretion and identify a minimal mechanistic model that recapitulates key qualitative and quantitative features of temporal experimental datasets of sFLT1 secretion from multiple studies. We show that the experimental data on sFLT1 secretion is best represented by a delay differential equation (DDE) system including a maturation term, reflecting the time required between synthesis and secretion. Using optimization to identify appropriate values for the key mechanistic parameters in the model, we show that two model parameters (extracellular degradation rate constant and maturation time) are very strongly constrained by the experimental data, and that the remaining parameters are related by two strongly constrained constants. Thus, only one degree of freedom remains, and measurements of the intracellular levels of sFLT1 would fix the remaining parameters. Comparison between simulation predictions and additional experimental data of the outcomes of chemical inhibitors and genetic perturbations suggest that intermediate values of the secretion rate constant best match the simulation with experiments, which would completely constrain the model. However, some of the inhibitors tested produce results that cannot be reproduced by the model simulations, suggesting that additional mechanisms not included here are required to explain those inhibitors. Overall, the model reproduces most available experimental data and suggests targets for further quantitative investigation of the sFLT1 system.
Collapse
Affiliation(s)
- Amy Gill
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Karina Kinghorn
- Department of Biology, University of North Carolina, Chapel Hill, NC, USA
- Curriculum in Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA
| | - Victoria L Bautch
- Department of Biology, University of North Carolina, Chapel Hill, NC, USA
- Curriculum in Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Feilim Mac Gabhann
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
2
|
Mutsuddy A, Huggins JR, Amrit A, Erdem C, Calhoun JC, Birtwistle MR. Mechanistic modeling of cell viability assays with in silico lineage tracing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.23.609433. [PMID: 39253474 PMCID: PMC11383287 DOI: 10.1101/2024.08.23.609433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Data from cell viability assays, which measure cumulative division and death events in a population and reflect substantial cellular heterogeneity, are widely available. However, interpreting such data with mechanistic computational models is hindered because direct model/data comparison is often muddled. We developed an algorithm that tracks simulated division and death events in mechanistically detailed single-cell lineages to enable such a model/data comparison and suggest causes of cell-cell drug response variability. Using our previously developed model of mammalian single-cell proliferation and death signaling, we simulated drug dose response experiments for four targeted anti-cancer drugs (alpelisib, neratinib, trametinib and palbociclib) and compared them to experimental data. Simulations are consistent with data for strong growth inhibition by trametinib (MEK inhibitor) and overall lack of efficacy for alpelisib (PI-3K inhibitor), but are inconsistent with data for palbociclib (CDK4/6 inhibitor) and neratinib (EGFR inhibitor). Model/data inconsistencies suggest (i) the importance of CDK4/6 for driving the cell cycle may be overestimated, and (ii) that the cellular balance between basal (tonic) and ligand-induced signaling is a critical determinant of receptor inhibitor response. Simulations show subpopulations of rapidly and slowly dividing cells in both control and drug-treated conditions. Variations in mother cells prior to drug treatment all impinging on ERK pathway activity are associated with the rapidly dividing phenotype and trametinib resistance. This work lays a foundation for the application of mechanistic modeling to large-scale cell viability assay datasets and better understanding determinants of cellular heterogeneity in drug response.
Collapse
Affiliation(s)
- Arnab Mutsuddy
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, SC, USA
| | - Jonah R. Huggins
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, SC, USA
| | - Aurore Amrit
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, SC, USA
- Faculté de Pharmacie, Université Paris Cité, Paris, France
| | - Cemal Erdem
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, SC, USA
- Department of Medical Biosciences, Umeå University, Umeå, Sweden
| | - Jon C. Calhoun
- Holcombe Department of Electrical and Computer Engineering, Clemson University, Clemson, SC, USA
| | - Marc R. Birtwistle
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, SC, USA
- Department of Bioengineering, Clemson University, Clemson, SC, USA
| |
Collapse
|
3
|
Oliveira RHDM, Annex BH, Popel AS. Endothelial cells signaling and patterning under hypoxia: a mechanistic integrative computational model including the Notch-Dll4 pathway. Front Physiol 2024; 15:1351753. [PMID: 38455844 PMCID: PMC10917925 DOI: 10.3389/fphys.2024.1351753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 02/12/2024] [Indexed: 03/09/2024] Open
Abstract
Introduction: Several signaling pathways are activated during hypoxia to promote angiogenesis, leading to endothelial cell patterning, interaction, and downstream signaling. Understanding the mechanistic signaling differences between endothelial cells under normoxia and hypoxia and their response to different stimuli can guide therapies to modulate angiogenesis. We present a novel mechanistic model of interacting endothelial cells, including the main pathways involved in angiogenesis. Methods: We calibrate and fit the model parameters based on well-established modeling techniques that include structural and practical parameter identifiability, uncertainty quantification, and global sensitivity. Results: Our results indicate that the main pathways involved in patterning tip and stalk endothelial cells under hypoxia differ, and the time under hypoxia interferes with how different stimuli affect patterning. Additionally, our simulations indicate that Notch signaling might regulate vascular permeability and establish different Nitric Oxide release patterns for tip/stalk cells. Following simulations with various stimuli, our model suggests that factors such as time under hypoxia and oxygen availability must be considered for EC pattern control. Discussion: This project provides insights into the signaling and patterning of endothelial cells under various oxygen levels and stimulation by VEGFA and is our first integrative approach toward achieving EC control as a method for improving angiogenesis. Overall, our model provides a computational framework that can be built on to test angiogenesis-related therapies by modulation of different pathways, such as the Notch pathway.
Collapse
Affiliation(s)
| | - Brian H. Annex
- Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Aleksander S. Popel
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
4
|
Li S, Wu H, Wang F, Kong L, Yu Y, Zuo R, Zhao H, Xu J, Kang Q. Enhanced Bone Regeneration through Regulation of Mechanoresponsive FAK-ERK1/2 Signaling by ZINC40099027 during Distraction Osteogenesis. Int J Med Sci 2024; 21:137-150. [PMID: 38164350 PMCID: PMC10750334 DOI: 10.7150/ijms.88298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 10/21/2023] [Indexed: 01/03/2024] Open
Abstract
Background: Focal adhesion kinase (FAK) is activated by mechanical stimulation and plays a vital role in distraction osteogenesis (DO), a well-established but lengthy procedure for repairing large bone defects. Both angiogenesis and osteogenesis contribute to bone regeneration during DO. However, the effects of ZINC40099027 (ZN27), a potent FAK activator, on angiogenesis, osteogenesis, and bone regeneration in DO remain unknown. Methods: The angiogenic potential of human umbilical vein endothelial cells (HUVECs) was evaluated using transwell migration and tube formation assays. The osteogenic activity of bone marrow mesenchymal stem cells (BMSCs) was assessed using alkaline phosphatase (ALP) and alizarin red s (ARS) staining. Additionally, quantitative real-time polymerase chain reaction (qRT-PCR), western blot, and immunofluorescence staining were used to assay angiogenic markers, osteogenic markers, and FAK-extracellular signal-regulated kinase 1/2 (ERK1/2) signaling. In vivo, a rat tibia DO model was established to verify the effects of ZN27 on neovascularization and bone regeneration using radiological and histological analyses. Results: ZN27 promoted the migration and angiogenesis of HUVECs. Additionally, ZN27 facilitated the osteogenic differentiation of BMSCs, as revealed by increased ALP activity, calcium deposition, and expression of osteogenesis-specific markers. The ERK1/2-specific inhibitor PD98059 significantly hindered the effects of ZN27, suggesting the participation of FAK-ERK1/2 signaling in ZN27-enhanced angiogenesis and osteogenesis. As indicated by improved radiological and histological features, ZN27 induced active angiogenesis within the distraction area and accelerated bone regeneration in a rat DO model. Conclusion: Our results show that ZN27 targets FAK-ERK1/2 signaling to stimulate both angiogenesis and osteogenesis, and ZN27 accelerates bone regeneration in DO, suggesting the therapeutic potential of ZN27 for repairing large bone defects in the mechanobiological environment during DO.
Collapse
Affiliation(s)
- Shanyu Li
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Hongxiao Wu
- Department of Orthopedics, Dongying People's Hospital, Dongying, Shandong, PR China
| | - Feng Wang
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Lingchi Kong
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Yifan Yu
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Rongtai Zuo
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Haoyu Zhao
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Jia Xu
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Qinglin Kang
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| |
Collapse
|
5
|
Maduabuchi WO, Tansi FL, Heller R, Hilger I. Hyperthermia Influences the Secretion Signature of Tumor Cells and Affects Endothelial Cell Sprouting. Biomedicines 2023; 11:2256. [PMID: 37626752 PMCID: PMC10452125 DOI: 10.3390/biomedicines11082256] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/03/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Tumors are a highly heterogeneous mass of tissue showing distinct therapy responses. In particular, the therapeutic outcome of tumor hyperthermia treatments has been inconsistent, presumably due to tumor versus endothelial cell cross-talks related to the treatment temperature and the tumor tissue environment. Here, we investigated the impact of the average or strong hyperthermic treatment (43 °C or 47 °C for 1 h) of the human pancreatic adenocarcinoma cell line (PANC-1 and BxPC-3) on endothelial cells (HUVECs) under post-treatment normoxic or hypoxic conditions. Immediately after the hyperthermia treatment, the distinct repression of secreted pro-angiogenic factors (e.g., VEGF, PDGF-AA, PDGF-BB, M-CSF), intracellular HIF-1α and the enhanced phosphorylation of ERK1/2 in tumor cells were detectable (particularly for strong hyperthermia, 2D cell monolayers). Notably, there was a significant increase in endothelial sprouting when 3D self-organized pancreatic cancer cells were treated with strong hyperthermia and the post-treatment conditions were hypoxic. Interestingly, for the used treatment temperatures, the intracellular HIF-1α accumulation in tumor cells seems to play a role in MAPK/ERK activation and mediator secretion (e.g., VEGF, PDGF-AA, Angiopoietin-2), as shown by inhibition experiments. Taken together, the hyperthermia of pancreatic adenocarcinoma cells in vitro impacts endothelial cells under defined environmental conditions (cell-to-cell contact, oxygen status, treatment temperature), whereby HIF-1α and VEGF secretion play a role in a complex context. Our observations could be exploited for the hyperthermic treatment of pancreatic cancer in the future.
Collapse
Affiliation(s)
- Wisdom O. Maduabuchi
- Department of Experimental Radiology, Institute of Diagnostic and Interventional Radiology, Jena University Hospital—Friedrich Schiller University Jena, Am Klinikum 1, D-07747 Jena, Germany; (W.O.M.); (F.L.T.)
| | - Felista L. Tansi
- Department of Experimental Radiology, Institute of Diagnostic and Interventional Radiology, Jena University Hospital—Friedrich Schiller University Jena, Am Klinikum 1, D-07747 Jena, Germany; (W.O.M.); (F.L.T.)
| | - Regine Heller
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), Hans-Knöll-Str. 2, D-07745 Jena, Germany;
| | - Ingrid Hilger
- Department of Experimental Radiology, Institute of Diagnostic and Interventional Radiology, Jena University Hospital—Friedrich Schiller University Jena, Am Klinikum 1, D-07747 Jena, Germany; (W.O.M.); (F.L.T.)
| |
Collapse
|
6
|
Oliveira RHM, Annex BH, Popel AS. Endothelial cells signaling and patterning under hypoxia: a mechanistic integrative computational model including the Notch-Dll4 pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.03.539270. [PMID: 37205581 PMCID: PMC10187169 DOI: 10.1101/2023.05.03.539270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Several signaling pathways are activated during hypoxia to promote angiogenesis, leading to endothelial cell patterning, interaction, and downstream signaling. Understanding the mechanistic signaling differences between normoxia and hypoxia can guide therapies to modulate angiogenesis. We present a novel mechanistic model of interacting endothelial cells, including the main pathways involved in angiogenesis. We calibrate and fit the model parameters based on well-established modeling techniques. Our results indicate that the main pathways involved in the patterning of tip and stalk endothelial cells under hypoxia differ, and the time under hypoxia affects how a reaction affects patterning. Interestingly, the interaction of receptors with Neuropilin1 is also relevant for cell patterning. Our simulations under different oxygen concentrations indicate time- and oxygen-availability-dependent responses for the two cells. Following simulations with various stimuli, our model suggests that factors such as period under hypoxia and oxygen availability must be considered for pattern control. This project provides insights into the signaling and patterning of endothelial cells under hypoxia, contributing to studies in the field.
Collapse
Affiliation(s)
- Rebeca Hannah M Oliveira
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, 21205, USA
| | - Brian H Annex
- Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Aleksander S Popel
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, 21205, USA
| |
Collapse
|
7
|
Barrasa-Ramos S, Dessalles CA, Hautefeuille M, Barakat AI. Mechanical regulation of the early stages of angiogenesis. J R Soc Interface 2022; 19:20220360. [PMID: 36475392 PMCID: PMC9727679 DOI: 10.1098/rsif.2022.0360] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Favouring or thwarting the development of a vascular network is essential in fields as diverse as oncology, cardiovascular disease or tissue engineering. As a result, understanding and controlling angiogenesis has become a major scientific challenge. Mechanical factors play a fundamental role in angiogenesis and can potentially be exploited for optimizing the architecture of the resulting vascular network. Largely focusing on in vitro systems but also supported by some in vivo evidence, the aim of this Highlight Review is dual. First, we describe the current knowledge with particular focus on the effects of fluid and solid mechanical stimuli on the early stages of the angiogenic process, most notably the destabilization of existing vessels and the initiation and elongation of new vessels. Second, we explore inherent difficulties in the field and propose future perspectives on the use of in vitro and physics-based modelling to overcome these difficulties.
Collapse
Affiliation(s)
- Sara Barrasa-Ramos
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Claire A. Dessalles
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Mathieu Hautefeuille
- Laboratoire de Biologie du Développement (UMR7622), Institut de Biologie Paris Seine, Sorbonne Université, Paris, France,Facultad de Ciencias, Universidad Nacional Autónoma de México, CDMX, Mexico
| | - Abdul I. Barakat
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| |
Collapse
|
8
|
Stepien TL, Secomb TW. Spreading mechanics and differentiation of astrocytes during retinal development. J Theor Biol 2022; 549:111208. [DOI: 10.1016/j.jtbi.2022.111208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 05/27/2022] [Accepted: 06/21/2022] [Indexed: 11/30/2022]
|
9
|
The transcription factor complex LMO2/TAL1 regulates branching and endothelial cell migration in sprouting angiogenesis. Sci Rep 2022; 12:7226. [PMID: 35508511 PMCID: PMC9068620 DOI: 10.1038/s41598-022-11297-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 04/08/2022] [Indexed: 11/09/2022] Open
Abstract
The transcription factor complex, consisting of LMO2, TAL1 or LYL1, and GATA2, plays an important role in capillary sprouting by regulating VEGFR2, DLL4, and angiopoietin 2 in tip cells. Overexpression of the basic helix-loop-helix transcription factor LYL1 in transgenic mice results in shortened tails. This phenotype is associated with vessel hyperbranching and a relative paucity of straight vessels due to DLL4 downregulation in tip cells by forming aberrant complex consisting of LMO2 and LYL1. Knockdown of LMO2 or TAL1 inhibits capillary sprouting in spheroid-based angiogenesis assays, which is associated with decreased angiopoietin 2 secretion. In the same assay using mixed TAL1- and LYL1-expressing endothelial cells, TAL1 was found to be primarily located in tip cells, while LYL1-expressing cells tended to occupy the stalk position in sprouts by upregulating VEGFR1 than TAL1. Thus, the interaction between LMO2 and TAL1 in tip cells plays a key role in angiogenic switch of sprouting angiogenesis.
Collapse
|
10
|
Electrophysiological Recordings from Embryonic Mouse Motoneurons Cultured on Electrospun Poly-Lactic Acid (PLA) and Polypyrrole-Coated PLA Scaffolds. IRANIAN BIOMEDICAL JOURNAL 2022; 26:183-92. [PMID: 35373542 PMCID: PMC9440684 DOI: 10.52547/ibj.26.3.183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
11
|
Zhang Y, Wang H, Oliveira RHM, Zhao C, Popel AS. Systems biology of angiogenesis signaling: Computational models and omics. WIREs Mech Dis 2021; 14:e1550. [PMID: 34970866 PMCID: PMC9243197 DOI: 10.1002/wsbm.1550] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 01/10/2023]
Abstract
Angiogenesis is a highly regulated multiscale process that involves a plethora of cells, their cellular signal transduction, activation, proliferation, differentiation, as well as their intercellular communication. The coordinated execution and integration of such complex signaling programs is critical for physiological angiogenesis to take place in normal growth, development, exercise, and wound healing, while its dysregulation is critically linked to many major human diseases such as cancer, cardiovascular diseases, and ocular disorders; it is also crucial in regenerative medicine. Although huge efforts have been devoted to drug development for these diseases by investigation of angiogenesis‐targeted therapies, only a few therapeutics and targets have proved effective in humans due to the innate multiscale complexity and nonlinearity in the process of angiogenic signaling. As a promising approach that can help better address this challenge, systems biology modeling allows the integration of knowledge across studies and scales and provides a powerful means to mechanistically elucidate and connect the individual molecular and cellular signaling components that function in concert to regulate angiogenesis. In this review, we summarize and discuss how systems biology modeling studies, at the pathway‐, cell‐, tissue‐, and whole body‐levels, have advanced our understanding of signaling in angiogenesis and thereby delivered new translational insights for human diseases. This article is categorized under:Cardiovascular Diseases > Computational Models Cancer > Computational Models
Collapse
Affiliation(s)
- Yu Zhang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hanwen Wang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Rebeca Hannah M Oliveira
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Chen Zhao
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Aleksander S Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
12
|
Campion O, Thevenard Devy J, Billottet C, Schneider C, Etique N, Dupuy JW, Raymond AA, Boulagnon Rombi C, Meunier M, Djermoune EH, Lelièvre E, Wahart A, Bour C, Hachet C, Cairo S, Bikfalvi A, Dedieu S, Devy J. LRP-1 Matricellular Receptor Involvement in Triple Negative Breast Cancer Tumor Angiogenesis. Biomedicines 2021; 9:biomedicines9101430. [PMID: 34680548 PMCID: PMC8533426 DOI: 10.3390/biomedicines9101430] [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: 08/09/2021] [Revised: 09/24/2021] [Accepted: 09/29/2021] [Indexed: 01/15/2023] Open
Abstract
Background: LRP-1 is a multifunctional scavenger receptor belonging to the LDLR family. Due to its capacity to control pericellular levels of various growth factors and proteases, LRP-1 plays a crucial role in membrane proteome dynamics, which appears decisive for tumor progression. Methods: LRP-1 involvement in a TNBC model was assessed using an RNA interference strategy in MDA-MB-231 cells. In vivo, tumorigenic and angiogenic effects of LRP-1-repressed cells were evaluated using an orthotopic xenograft model and two angiogenic assays (Matrigel® plugs, CAM). DCE-MRI, FMT, and IHC were used to complete a tumor longitudinal follow-up and obtain morphological and functional vascular information. In vitro, HUVECs’ angiogenic potential was evaluated using a tumor secretome, subjected to a proteomic analysis to highlight LRP-1-dependant signaling pathways. Results: LRP-1 repression in MDA-MB-231 tumors led to a 60% growth delay because of, inter alia, morphological and functional vascular differences, confirmed by angiogenic models. In vitro, the LRP-1-repressed cells secretome restrained HUVECs’ angiogenic capabilities. A proteomics analysis revealed that LRP-1 supports tumor growth and angiogenesis by regulating TGF-β signaling and plasminogen/plasmin system. Conclusions: LRP-1, by its wide spectrum of interactions, emerges as an important matricellular player in the control of cancer-signaling events such as angiogenesis, by supporting tumor vascular morphology and functionality.
Collapse
Affiliation(s)
- Océane Campion
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, 51687 Reims, France; (O.C.); (J.T.D.); (C.S.); (N.E.); (M.M.); (E.L.); (A.W.); (C.B.); (C.H.); (S.D.)
- Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, 51687 Reims, France;
| | - Jessica Thevenard Devy
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, 51687 Reims, France; (O.C.); (J.T.D.); (C.S.); (N.E.); (M.M.); (E.L.); (A.W.); (C.B.); (C.H.); (S.D.)
- Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, 51687 Reims, France;
| | - Clotilde Billottet
- INSERM, LAMC, U1029, Université de Bordeaux, 33600 Pessac, France; (C.B.); (A.B.)
| | - Christophe Schneider
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, 51687 Reims, France; (O.C.); (J.T.D.); (C.S.); (N.E.); (M.M.); (E.L.); (A.W.); (C.B.); (C.H.); (S.D.)
- Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, 51687 Reims, France;
| | - Nicolas Etique
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, 51687 Reims, France; (O.C.); (J.T.D.); (C.S.); (N.E.); (M.M.); (E.L.); (A.W.); (C.B.); (C.H.); (S.D.)
- Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, 51687 Reims, France;
| | | | | | - Camille Boulagnon Rombi
- Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, 51687 Reims, France;
- Laboratoire d’Anatomie Pathologie, CHU Reims, 51100 Reims, France
| | - Marie Meunier
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, 51687 Reims, France; (O.C.); (J.T.D.); (C.S.); (N.E.); (M.M.); (E.L.); (A.W.); (C.B.); (C.H.); (S.D.)
- Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, 51687 Reims, France;
| | | | - Elodie Lelièvre
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, 51687 Reims, France; (O.C.); (J.T.D.); (C.S.); (N.E.); (M.M.); (E.L.); (A.W.); (C.B.); (C.H.); (S.D.)
- Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, 51687 Reims, France;
| | - Amandine Wahart
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, 51687 Reims, France; (O.C.); (J.T.D.); (C.S.); (N.E.); (M.M.); (E.L.); (A.W.); (C.B.); (C.H.); (S.D.)
- Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, 51687 Reims, France;
| | - Camille Bour
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, 51687 Reims, France; (O.C.); (J.T.D.); (C.S.); (N.E.); (M.M.); (E.L.); (A.W.); (C.B.); (C.H.); (S.D.)
- Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, 51687 Reims, France;
| | - Cathy Hachet
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, 51687 Reims, France; (O.C.); (J.T.D.); (C.S.); (N.E.); (M.M.); (E.L.); (A.W.); (C.B.); (C.H.); (S.D.)
- Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, 51687 Reims, France;
| | | | - Andréas Bikfalvi
- INSERM, LAMC, U1029, Université de Bordeaux, 33600 Pessac, France; (C.B.); (A.B.)
| | - Stéphane Dedieu
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, 51687 Reims, France; (O.C.); (J.T.D.); (C.S.); (N.E.); (M.M.); (E.L.); (A.W.); (C.B.); (C.H.); (S.D.)
- Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, 51687 Reims, France;
| | - Jérôme Devy
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, 51687 Reims, France; (O.C.); (J.T.D.); (C.S.); (N.E.); (M.M.); (E.L.); (A.W.); (C.B.); (C.H.); (S.D.)
- Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, 51687 Reims, France;
- Correspondence:
| |
Collapse
|
13
|
Fan X, Bilir EK, Kingston OA, Oldershaw RA, Kearns VR, Willoughby CE, Sheridan CM. Replacement of the Trabecular Meshwork Cells-A Way Ahead in IOP Control? Biomolecules 2021; 11:biom11091371. [PMID: 34572584 PMCID: PMC8464777 DOI: 10.3390/biom11091371] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/09/2021] [Accepted: 09/13/2021] [Indexed: 12/11/2022] Open
Abstract
Glaucoma is one of the leading causes of vision loss worldwide, characterised with irreversible optic nerve damage and progressive vision loss. Primary open-angle glaucoma (POAG) is a subset of glaucoma, characterised by normal anterior chamber angle and raised intraocular pressure (IOP). Reducing IOP is the main modifiable factor in the treatment of POAG, and the trabecular meshwork (TM) is the primary site of aqueous humour outflow (AH) and the resistance to outflow. The structure and the composition of the TM are key to its function in regulating AH outflow. Dysfunction and loss of the TM cells found in the natural ageing process and more so in POAG can cause abnormal extracellular matrix (ECM) accumulation, increased TM stiffness, and increased IOP. Therefore, repair or regeneration of TM's structure and function is considered as a potential treatment for POAG. Cell transplantation is an attractive option to repopulate the TM cells in POAG, but to develop a cell replacement approach, various challenges are still to be addressed. The choice of cell replacement covers autologous or allogenic approaches, which led to investigations into TM progenitor cells, induced pluripotent stem cells (iPSCs), and mesenchymal stem cells (MSCs) as potential stem cell source candidates. However, the potential plasticity and the lack of definitive cell markers for the progenitor and the TM cell population compound the biological challenge. Morphological and differential gene expression of TM cells located within different regions of the TM may give rise to different cell replacement or regenerative approaches. As such, this review describes the different approaches taken to date investigating different cell sources and their differing cell isolation and differentiation methodologies. In addition, we highlighted how these approaches were evaluated in different animal and ex vivo model systems and the potential of these methods in future POAG treatment.
Collapse
Affiliation(s)
- Xiaochen Fan
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L69 3BX, UK; (X.F.); (E.K.B.); (O.A.K.); (V.R.K.)
| | - Emine K. Bilir
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L69 3BX, UK; (X.F.); (E.K.B.); (O.A.K.); (V.R.K.)
| | - Olivia A. Kingston
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L69 3BX, UK; (X.F.); (E.K.B.); (O.A.K.); (V.R.K.)
| | - Rachel A. Oldershaw
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L69 3BX, UK;
| | - Victoria R. Kearns
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L69 3BX, UK; (X.F.); (E.K.B.); (O.A.K.); (V.R.K.)
| | - Colin E. Willoughby
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L69 3BX, UK; (X.F.); (E.K.B.); (O.A.K.); (V.R.K.)
- Genomic Medicine, Biomedical Sciences Research Institute, Ulster University, Coleraine BT52 1SA, UK
- Correspondence: (C.E.W.); (C.M.S.); Tel.: +44-(28)-701-2338 (C.E.W.); +44-(151)-794-9031 (C.M.S.)
| | - Carl M. Sheridan
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L69 3BX, UK; (X.F.); (E.K.B.); (O.A.K.); (V.R.K.)
- Correspondence: (C.E.W.); (C.M.S.); Tel.: +44-(28)-701-2338 (C.E.W.); +44-(151)-794-9031 (C.M.S.)
| |
Collapse
|
14
|
Tsuji-Tamura K, Morino-Koga S, Suzuki S, Ogawa M. The canonical smooth muscle cell marker TAGLN is present in endothelial cells and is involved in angiogenesis. J Cell Sci 2021; 134:jcs254920. [PMID: 34338296 DOI: 10.1242/jcs.254920] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 06/30/2021] [Indexed: 12/24/2022] Open
Abstract
Elongation of vascular endothelial cells (ECs) is an important process in angiogenesis; however, the molecular mechanisms remain unknown. The actin-crosslinking protein TAGLN (transgelin, also known as SM22 or SM22α) is abundantly expressed in smooth muscle cells (SMCs) and is widely used as a canonical marker for this cell type. In the course of studies using mouse embryonic stem cells (ESCs) carrying an Tagln promoter-driven fluorescence marker, we noticed activation of the Tagln promoter during EC elongation. Tagln promoter activation co-occurred with EC elongation in response to vascular endothelial growth factor A (VEGF-A). Inhibition of phosphoinositide 3-kinase (PI3K)-Akt signaling and mTORC1 also induced EC elongation and Tagln promoter activation. Human umbilical vein endothelial cells (HUVECs) elongated, activated the TAGLN promoter and increased TAGLN transcripts in an angiogenesis model. Genetic disruption of TAGLN augmented angiogenic behaviors of HUVECs, as did the disruption of TAGLN2 and TAGLN3 genes. Tagln expression was found in ECs in mouse embryos. Our results identify TAGLN as a putative regulator of angiogenesis whose expression is activated in elongating ECs. This finding provides insight into the cytoskeletal regulation of EC elongation and an improved understanding of the molecular mechanisms underlying the regulation of angiogenesis.
Collapse
Affiliation(s)
- Kiyomi Tsuji-Tamura
- Department of Cell Differentiation, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Kumamoto 860-0811, Japan
| | - Saori Morino-Koga
- Department of Cell Differentiation, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Kumamoto 860-0811, Japan
| | - Shingo Suzuki
- Support Section for Education and Research, Faculty of Dental Medicine, Hokkaido University, Kita 13, Nishi 7, Kita-ku, Sapporo 060-8586, Japan
| | - Minetaro Ogawa
- Department of Cell Differentiation, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Kumamoto 860-0811, Japan
| |
Collapse
|
15
|
Bischoff-Kont I, Brabenec L, Ingelfinger R, Nausch B, Fürst R. BNO 1095, a Standardized Dry Extract from the Fruits of Vitex agnus-castus, Impairs Angiogenesis-related Endothelial Cell Functions In Vitro. PLANTA MEDICA 2021; 87:611-619. [PMID: 33530113 PMCID: PMC8277439 DOI: 10.1055/a-1351-1038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
BNO 1095, a standardized dry extract from the fruits of Vitex agnus-castus, represents an approved herbal medicinal product for the treatment of premenstrual syndrome. Angiogenesis, the formation of new blood vessels from pre-existing capillaries, plays a major role in physiological situations, such as wound healing or tissue growth in female reproductive organs, but it is also of great importance in pathophysiological conditions such as chronic inflammatory diseases or cancer. Angiogenesis is a highly regulated multi-step process consisting of distinct key events that can be influenced pharmacologically. Few studies suggested anti-angiogenic actions of V. agnus-castus fruit extracts in in vivo and ex vivo models. Here, we provide for the first time profound in vitro data on BNO 1095-derived anti-angiogenic effects focusing on distinct angiogenesis-related endothelial cell functions that are inevitable for the process of new blood vessel formation. We found that V. agnus-castus extract significantly attenuated undirected and chemotactic migration of primary human endothelial cells. Moreover, the extract efficiently inhibited endothelial cell proliferation and reduced the formation of tube-like structures on Matrigel. Of note, the treatment of endothelial cell spheroids almost blocked endothelial sprouting in a 3D collagen gel. Our data present new and detailed insights into the anti-angiogenic actions of BNO 1095 and, therefore, suggest a novel scope of potential therapeutic applications of the extract for which these anti-angiogenic properties are required.
Collapse
Affiliation(s)
- Iris Bischoff-Kont
- Institute of Pharmaceutical Biology, Goethe University Frankfurt/Main, Germany
- Correspondence Dr. Iris Bischoff-Kont Institute of Pharmaceutical BiologyGoethe University FrankfurtMax-von-Laue-Str. 960438 Frankfurt am MainGermany+ 49 69 79 82 96 45+ 49 69 79 82 96 62
| | - Laura Brabenec
- Institute of Pharmaceutical Biology, Goethe University Frankfurt/Main, Germany
| | - Rebecca Ingelfinger
- Institute of Pharmaceutical Biology, Goethe University Frankfurt/Main, Germany
- LOEWE Center Translational Biodiversity Genomics (TBG), Frankfurt/Main, Germany
| | | | - Robert Fürst
- Institute of Pharmaceutical Biology, Goethe University Frankfurt/Main, Germany
- LOEWE Center Translational Biodiversity Genomics (TBG), Frankfurt/Main, Germany
| |
Collapse
|
16
|
Ye Li, Xu J, Mi J, He X, Pan Q, Zheng L, Zu H, Chen Z, Dai B, Li X, Pang Q, Zou L, Zhou L, Huang L, Tong W, Li G, Qin L. Biodegradable magnesium combined with distraction osteogenesis synergistically stimulates bone tissue regeneration via CGRP-FAK-VEGF signaling axis. Biomaterials 2021; 275:120984. [PMID: 34186235 DOI: 10.1016/j.biomaterials.2021.120984] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 06/04/2021] [Accepted: 06/18/2021] [Indexed: 01/05/2023]
Abstract
Critical size bone defects are frequently caused by accidental trauma, oncologic surgery, and infection. Distraction osteogenesis (DO) is a useful technique to promote the repair of critical size bone defects. However, DO is usually a lengthy treatment, therefore accompanied with increased risks of complications such as infections and delayed union. Here, we demonstrated that magnesium (Mg) nail implantation into the marrow cavity degraded gradually accompanied with about 4-fold increase of new bone formation and over 5-fold of new vessel formation as compared with DO alone group in the 5 mm femoral segmental defect rat model at 2 weeks after distraction. Mg nail upregulated the expression of calcitonin gene-related peptide (CGRP) in the new bone as compared with the DO alone group. We further revealed that blockade of the sensory nerve by overdose capsaicin blunted Mg nail enhanced critical size bone defect repair during the DO process. CGRP concentration-dependently promoted endothelial cell migration and tube formation. Meanwhile, CGRP promoted the phosphorylation of focal adhesion kinase (FAK) at Y397 site and elevated the expression of vascular endothelial growth factor A (VEGFA). Moreover, inhibitor/antagonist of CGRP receptor, FAK, and VEGF receptor blocked the Mg nail stimulated vessel and bone formation. We revealed, for the first time, a CGRP-FAK-VEGF signaling axis linking sensory nerve and endothelial cells, which may be the main mechanism underlying Mg-enhanced critical size bone defect repair when combined with DO, suggesting a great potential of Mg implants in reducing DO treatment time for clinical applications.
Collapse
Affiliation(s)
- Ye Li
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong SAR, China; Center for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Science, China
| | - Jiankun Xu
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jie Mi
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xuan He
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qi Pan
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Lizhen Zheng
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong SAR, China; Center for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Science, China
| | - Haiyue Zu
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ziyi Chen
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Bingyang Dai
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xu Li
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qianqian Pang
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Li Zou
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Liangbin Zhou
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Le Huang
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Wenxue Tong
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Gang Li
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Ling Qin
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong SAR, China; CHUK Hong Kong - Shenzhen Innovation and Technology Institute (Futian), China.
| |
Collapse
|
17
|
Link PA, Heise RL, Weinberg SH. Cellular mitosis predicts vessel stability in a mechanochemical model of sprouting angiogenesis. Biomech Model Mechanobiol 2021; 20:1195-1208. [PMID: 33715101 DOI: 10.1007/s10237-021-01442-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 02/22/2021] [Indexed: 11/28/2022]
Abstract
Angiogenesis, the formation of new vessels, occurs in both developmental and pathological contexts. Prior research has investigated vessel formation to identify cellular phenotypes and dynamics associated with angiogenic disease. One major family of proteins involved in angiogenesis are the Rho GTPases, which govern function related to cellular elongation, migration, and proliferation. Using a mechanochemical model coupling Rho GTPase activity and cellular and intercellular mechanics, we investigate the role of cellular mitosis on sprouting angiogenesis. Mitosis-GTPase synchronization was not a strong predictor of GTPase and thus vessel signaling instability, whereas the location of mitotic events was predicted to alter GTPase cycling instabilities. Our model predicts that middle stalk cells undergoing mitosis introduce irregular dynamics in GTPase cycling and may provide a source of aberrant angiogenesis. We also find that cellular and junctional tension exhibit spatial heterogeneity through the vessel, and that tension feedback, specifically in stalk cells, tends to increase the maximum forces generated in the vessel.
Collapse
Affiliation(s)
- Patrick A Link
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Rebecca L Heise
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Seth H Weinberg
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
18
|
Comparative analysis of continuum angiogenesis models. J Math Biol 2021; 82:21. [PMID: 33619643 PMCID: PMC7900093 DOI: 10.1007/s00285-021-01570-w] [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: 07/20/2020] [Revised: 12/07/2020] [Accepted: 01/17/2021] [Indexed: 11/06/2022]
Abstract
Although discrete approaches are increasingly employed to model biological phenomena, it remains unclear how complex, population-level behaviours in such frameworks arise from the rules used to represent interactions between individuals. Discrete-to-continuum approaches, which are used to derive systems of coarse-grained equations describing the mean-field dynamics of a microscopic model, can provide insight into such emergent behaviour. Coarse-grained models often contain nonlinear terms that depend on the microscopic rules of the discrete framework, however, and such nonlinearities can make a model difficult to mathematically analyse. By contrast, models developed using phenomenological approaches are typically easier to investigate but have a more obscure connection to the underlying microscopic system. To our knowledge, there has been little work done to compare solutions of phenomenological and coarse-grained models. Here we address this problem in the context of angiogenesis (the creation of new blood vessels from existing vasculature). We compare asymptotic solutions of a classical, phenomenological “snail-trail” model for angiogenesis to solutions of a nonlinear system of partial differential equations (PDEs) derived via a systematic coarse-graining procedure (Pillay et al. in Phys Rev E 95(1):012410, 2017. https://doi.org/10.1103/PhysRevE.95.012410). For distinguished parameter regimes corresponding to chemotaxis-dominated cell movement and low branching rates, both continuum models reduce at leading order to identical PDEs within the domain interior. Numerical and analytical results confirm that pointwise differences between solutions to the two continuum models are small if these conditions hold, and demonstrate how perturbation methods can be used to determine when a phenomenological model provides a good approximation to a more detailed coarse-grained system for the same biological process.
Collapse
|
19
|
Martinson WD, Byrne HM, Maini PK. Evaluating snail-trail frameworks for leader-follower behavior with agent-based modeling. Phys Rev E 2020; 102:062417. [PMID: 33466087 DOI: 10.1103/physreve.102.062417] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 11/18/2020] [Indexed: 06/12/2023]
Abstract
Branched networks constitute a ubiquitous structure in biology, arising in plants, lungs, and the circulatory system; however, the mechanisms behind their creation are not well understood. A commonly used model for network morphogenesis proposes that sprouts develop through interactions between leader (tip) cells and follower (stalk) cells. In this description, tip cells emerge from existing structures, travel up chemoattractant gradients, and form new networks by guiding the movement of stalk cells. Such dynamics have been mathematically represented by continuum "snail-trail" models in which the tip cell flux contributes to the stalk cell proliferation rate. Although snail-trail models constitute a classical depiction of leader-follower behavior, their accuracy has yet to be evaluated in a rigorous quantitative setting. Here, we extend the snail-trail modeling framework to two spatial dimensions by introducing a novel multiplicative factor to the stalk cell rate equation, which corrects for neglected network creation in directions other than that of the migrating front. Our derivation of this factor demonstrates that snail-trail models are valid descriptions of cell dynamics when chemotaxis dominates cell movement. We confirm that our snail-trail model accurately predicts the dynamics of tip and stalk cells in an existing agent-based model (ABM) for network formation [Pillay et al., Phys. Rev. E 95, 012410 (2017)10.1103/PhysRevE.95.012410]. We also derive conditions for which it is appropriate to use a reduced, one-dimensional snail-trail model to analyze ABM results. Our analysis identifies key metrics for cell migration that may be used to anticipate when simple snail-trail models will accurately describe experimentally observed cell dynamics in network formation.
Collapse
Affiliation(s)
- W Duncan Martinson
- Mathematical Institute, University of Oxford, Woodstock Road, Oxford OX2 6GG, United Kingdom
| | - Helen M Byrne
- Mathematical Institute, University of Oxford, Woodstock Road, Oxford OX2 6GG, United Kingdom
| | - Philip K Maini
- Mathematical Institute, University of Oxford, Woodstock Road, Oxford OX2 6GG, United Kingdom
| |
Collapse
|
20
|
Mamer SB, Page P, Murphy M, Wang J, Gallerne P, Ansari A, Imoukhuede PI. The Convergence of Cell-Based Surface Plasmon Resonance and Biomaterials: The Future of Quantifying Bio-molecular Interactions-A Review. Ann Biomed Eng 2020; 48:2078-2089. [PMID: 31811474 PMCID: PMC8637426 DOI: 10.1007/s10439-019-02429-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 11/29/2019] [Indexed: 12/20/2022]
Abstract
Cell biology is driven by complex networks of biomolecular interactions. Characterizing the kinetic and thermodynamic properties of these interactions is crucial to understanding their role in different physiological processes. Surface plasmon resonance (SPR)-based approaches have become a key tool in quantifying biomolecular interactions, however conventional approaches require isolating the interacting components from the cellular system. Cell-based SPR approaches have recently emerged, promising to enable precise measurements of biomolecular interactions within their normal biological context. Two major approaches have been developed, offering their own advantages and limitations. These approaches currently lack a systematic exploration of 'best practices' like those existing for traditional SPR experiments. Toward this end, we describe the two major approaches, and identify the experimental parameters that require exploration, and discuss the experimental considerations constraining the optimization of each. In particular, we discuss the requirements of future biomaterial development needed to advance the cell-based SPR technique.
Collapse
Affiliation(s)
- Spencer B Mamer
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | | | | | - Jiaojiao Wang
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Pierrick Gallerne
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Ecole Centrale de Lille, Villeneuve d'Ascq, Hauts-De-France, France
| | - Ali Ansari
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - P I Imoukhuede
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
21
|
Rubin JB, Lagas JS, Broestl L, Sponagel J, Rockwell N, Rhee G, Rosen SF, Chen S, Klein RS, Imoukhuede P, Luo J. Sex differences in cancer mechanisms. Biol Sex Differ 2020; 11:17. [PMID: 32295632 PMCID: PMC7161126 DOI: 10.1186/s13293-020-00291-x] [Citation(s) in RCA: 178] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 03/18/2020] [Indexed: 02/07/2023] Open
Abstract
We now know that cancer is many different diseases, with great variation even within a single histological subtype. With the current emphasis on developing personalized approaches to cancer treatment, it is astonishing that we have not yet systematically incorporated the biology of sex differences into our paradigms for laboratory and clinical cancer research. While some sex differences in cancer arise through the actions of circulating sex hormones, other sex differences are independent of estrogen, testosterone, or progesterone levels. Instead, these differences are the result of sexual differentiation, a process that involves genetic and epigenetic mechanisms, in addition to acute sex hormone actions. Sexual differentiation begins with fertilization and continues beyond menopause. It affects virtually every body system, resulting in marked sex differences in such areas as growth, lifespan, metabolism, and immunity, all of which can impact on cancer progression, treatment response, and survival. These organismal level differences have correlates at the cellular level, and thus, males and females can fundamentally differ in their protections and vulnerabilities to cancer, from cellular transformation through all stages of progression, spread, and response to treatment. Our goal in this review is to cover some of the robust sex differences that exist in core cancer pathways and to make the case for inclusion of sex as a biological variable in all laboratory and clinical cancer research. We finish with a discussion of lab- and clinic-based experimental design that should be used when testing whether sex matters and the appropriate statistical models to apply in data analysis for rigorous evaluations of potential sex effects. It is our goal to facilitate the evaluation of sex differences in cancer in order to improve outcomes for all patients.
Collapse
Affiliation(s)
- Joshua B Rubin
- Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA.
- Department of Neuroscience, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA.
| | - Joseph S Lagas
- Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - Lauren Broestl
- Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - Jasmin Sponagel
- Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - Nathan Rockwell
- Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - Gina Rhee
- Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - Sarah F Rosen
- Department of Medicine, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - Si Chen
- Department of Biomedical Engineering, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - Robyn S Klein
- Department of Neuroscience, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
- Department of Medicine, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - Princess Imoukhuede
- Department of Biomedical Engineering, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - Jingqin Luo
- Department of Surgery, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| |
Collapse
|
22
|
Roy V, Magne B, Vaillancourt-Audet M, Blais M, Chabaud S, Grammond E, Piquet L, Fradette J, Laverdière I, Moulin VJ, Landreville S, Germain L, Auger FA, Gros-Louis F, Bolduc S. Human Organ-Specific 3D Cancer Models Produced by the Stromal Self-Assembly Method of Tissue Engineering for the Study of Solid Tumors. BIOMED RESEARCH INTERNATIONAL 2020; 2020:6051210. [PMID: 32352002 PMCID: PMC7178531 DOI: 10.1155/2020/6051210] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 02/07/2020] [Accepted: 02/28/2020] [Indexed: 12/24/2022]
Abstract
Cancer research has considerably progressed with the improvement of in vitro study models, helping to understand the key role of the tumor microenvironment in cancer development and progression. Over the last few years, complex 3D human cell culture systems have gained much popularity over in vivo models, as they accurately mimic the tumor microenvironment and allow high-throughput drug screening. Of particular interest, in vitrohuman 3D tissue constructs, produced by the self-assembly method of tissue engineering, have been successfully used to model the tumor microenvironment and now represent a very promising approach to further develop diverse cancer models. In this review, we describe the importance of the tumor microenvironment and present the existing in vitro cancer models generated through the self-assembly method of tissue engineering. Lastly, we highlight the relevance of this approach to mimic various and complex tumors, including basal cell carcinoma, cutaneous neurofibroma, skin melanoma, bladder cancer, and uveal melanoma.
Collapse
Affiliation(s)
- Vincent Roy
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
| | - Brice Magne
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
| | - Maude Vaillancourt-Audet
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
| | - Mathieu Blais
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
| | - Stéphane Chabaud
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
| | - Emil Grammond
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
| | - Léo Piquet
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
- Centre de Recherche sur le Cancer de l'Université Laval, Québec, QC, Canada
| | - Julie Fradette
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
- Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Isabelle Laverdière
- Centre de Recherche sur le Cancer de l'Université Laval, Québec, QC, Canada
- Faculty of Pharmacy, Université Laval and CHU de Québec-Université Laval Research Center, Oncology Division, Québec, QC, Canada
| | - Véronique J. Moulin
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
- Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Solange Landreville
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
- Centre de Recherche sur le Cancer de l'Université Laval, Québec, QC, Canada
- Department of Ophthalmology, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Lucie Germain
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
- Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - François A. Auger
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
- Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - François Gros-Louis
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
- Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Stéphane Bolduc
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
- Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, Canada
| |
Collapse
|
23
|
Ros E, Encina M, González F, Contreras R, Luz-Crawford P, Khoury M, Acevedo JP. Single cell migration profiling on a microenvironmentally tunable hydrogel microstructure device that enables stem cell potency evaluation. LAB ON A CHIP 2020; 20:958-972. [PMID: 31990283 DOI: 10.1039/c9lc00988d] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Cell migration is a key function in a myriad of physiological events and disease conditions. Efficient, quick and descriptive profiling of migration behaviour in response to different treatments or conditions is highly desirable in a series of applications, ranging from fundamental studies of the migration mechanism to drug discovery and cell therapy. This investigation applied the use of methacrylamide gelatin (GelMA) to microfabricate migration lanes based on GelMA hydrogel with encapsulated migration stimuli and structural stability under culture medium conditions, providing the possibility of tailoring the microenvironment during cell-based assays. The actual device provides 3D topography, cell localization and a few step protocol, allowing the quick evaluation and quantification of individual migrated distances of a cell sample by an ImageJ plugin for automated microscopy processing. The detailed profiling of migration behaviour given by the new device has demonstrated a broader assay sensitivity compared to other migration assays and higher versatility to study cell migration in different settings of applications. In this study, parametric information extracted from the migration profiling was successfully used to develop predictive models of immunosuppressive cell function that could be applied as a potency test for mesenchymal stem cells.
Collapse
Affiliation(s)
- Enrique Ros
- Cells for Cells, Santiago, Chile and Consorcio Regenero, Chilean Consortium for Regenerative Medicine, Santiago, Chile
| | - Matías Encina
- Cells for Cells, Santiago, Chile and Consorcio Regenero, Chilean Consortium for Regenerative Medicine, Santiago, Chile
| | - Fabián González
- Cells for Cells, Santiago, Chile and Consorcio Regenero, Chilean Consortium for Regenerative Medicine, Santiago, Chile
| | - Rafael Contreras
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Patricia Luz-Crawford
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Maroun Khoury
- Cells for Cells, Santiago, Chile and Consorcio Regenero, Chilean Consortium for Regenerative Medicine, Santiago, Chile and Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de los Andes, Santiago, Chile.
| | - Juan Pablo Acevedo
- Cells for Cells, Santiago, Chile and Consorcio Regenero, Chilean Consortium for Regenerative Medicine, Santiago, Chile and Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de los Andes, Santiago, Chile.
| |
Collapse
|
24
|
Urbanczyk M, Layland SL, Schenke-Layland K. The role of extracellular matrix in biomechanics and its impact on bioengineering of cells and 3D tissues. Matrix Biol 2019; 85-86:1-14. [PMID: 31805360 DOI: 10.1016/j.matbio.2019.11.005] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 11/24/2019] [Accepted: 11/24/2019] [Indexed: 12/20/2022]
Abstract
The cells and tissues of the human body are constantly exposed to exogenous and endogenous forces that are referred to as biomechanical cues. They guide and impact cellular processes and cell fate decisions on the nano-, micro- and macro-scale, and are therefore critical for normal tissue development and maintaining tissue homeostasis. Alterations in the extracellular matrix composition of a tissue combined with abnormal mechanosensing and mechanotransduction can aberrantly activate signaling pathways that promote disease development. Such processes are therefore highly relevant for disease modelling or when aiming for the development of novel therapies. In this mini review, we describe the main biomechanical cues that impact cellular fates. We highlight their role during development, homeostasis and in disease. We also discuss current techniques and tools that allow us to study the impact of biomechanical cues on cell and tissue development under physiological conditions, and we point out directions, in which in vitro biomechanics can be of use in the future.
Collapse
Affiliation(s)
- Max Urbanczyk
- Department of Women's Health, Research Institute of Women's Health, Eberhard Karls University Tübingen, Germany
| | - Shannon L Layland
- Department of Women's Health, Research Institute of Women's Health, Eberhard Karls University Tübingen, Germany
| | - Katja Schenke-Layland
- Department of Women's Health, Research Institute of Women's Health, Eberhard Karls University Tübingen, Germany; Natural and Medical Sciences Institute (NMI) at the University of Tübingen, Reutlingen, Germany; Cluster of Excellence IFIT (EXC 2180), "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University Tübingen, Germany; Dept. of Medicine/Cardiology, University of California Los Angeles (UCLA), Los Angeles, CA, USA.
| |
Collapse
|
25
|
Goud ESK, Pandey M, Singh C, Vedamurthy GV, Singh D, Onteru SK. Effect of Dioxins in Milk on the 3D Cultured Primary Buffalo Hepatocyte Model System. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:8007-8019. [PMID: 31268702 DOI: 10.1021/acs.jafc.9b03384] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Cow and human milk have been reported to contain dioxins ranging from 0.023 to 26.46 and 0.88 to 19 pg/g of fat, respectively. However, the toxic effects of the dioxins in the milk in this range of concentrations were not explored. Therefore, considering the outbred livestock tissues as better models than inbred laboratory animals, the present study targeted to study the effect of dioxins present in the milk on three-dimensionally (3D) cultured buffalo primary hepatocyte spheroids. The spheroids were treated with a model dioxin, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), directly and also through milk fat at different concentrations (i.e, 0.02-20 pg/mL) for 24 h. Among the liver-cell-specific (ALB, HNF4α, and AFP) genes, a similar ALB and upregulated HNF4α expression at all treatments indicated the functional and transcriptionally active hepatocyte spheroids. Supportingly, no significant difference in the antiapoptotic gene expression between the treatments of milk fat and milk fat containing dioxins indicated the survivability of the spheroids during dioxin treatments. Among the selected TCDD responsive (CYP1A1, CYP1A2, AHR, CYP1B1, and TIPARP) genes, a nonsignificant increasing trend of the CYP1A1 expression was observed from 0.2 to 10 pg/mL of TCDD concentration through milk fat. This pattern was similar to the reported insensitive response of human primary hepatocytes toward dioxins than that of rat primary hepatocytes. This may indicate that the buffalo hepatocyte spheroids could be better models than rats for TCDD hepatotoxic studies. Further, TCDD in the milk in the range of 0.02-20 pg/mL concentration may not be very hepatotoxic.
Collapse
Affiliation(s)
- Emmagouni Sharath Kumar Goud
- Molecular Endocrinology, Functional Genomics and Systems Biology Lab, Animal Biochemistry Division , ICAR-National Dairy Research Institute , Karnal 132001 , India
| | - Mamta Pandey
- Molecular Endocrinology, Functional Genomics and Systems Biology Lab, Animal Biochemistry Division , ICAR-National Dairy Research Institute , Karnal 132001 , India
| | - Chhama Singh
- Molecular Endocrinology, Functional Genomics and Systems Biology Lab, Animal Biochemistry Division , ICAR-National Dairy Research Institute , Karnal 132001 , India
| | - Gowdar Veerappa Vedamurthy
- Molecular Endocrinology, Functional Genomics and Systems Biology Lab, Animal Biochemistry Division , ICAR-National Dairy Research Institute , Karnal 132001 , India
| | - Dheer Singh
- Molecular Endocrinology, Functional Genomics and Systems Biology Lab, Animal Biochemistry Division , ICAR-National Dairy Research Institute , Karnal 132001 , India
| | - Suneel Kumar Onteru
- Molecular Endocrinology, Functional Genomics and Systems Biology Lab, Animal Biochemistry Division , ICAR-National Dairy Research Institute , Karnal 132001 , India
| |
Collapse
|
26
|
Single-Cell Receptor Quantification of an In Vitro Coculture Angiogenesis Model Reveals VEGFR, NRP1, Tie2, and PDGFR Regulation and Endothelial Heterogeneity. Processes (Basel) 2019. [DOI: 10.3390/pr7060356] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Angiogenesis, the formation of new blood vessels from pre-existing ones, is essential for both normal development and numerous pathologies. Systems biology has offered a unique approach to study angiogenesis by profiling tyrosine kinase receptors (RTKs) that regulate angiogenic processes and computationally modeling RTK signaling pathways. Historically, this systems biology approach has been applied on ex vivo angiogenesis assays, however, these assays are difficult to quantify and limited in their potential of temporal analysis. In this study, we adopted a simple two-dimensional angiogenesis assay comprised of human umbilical vein endothelial cells (HUVECs) and human dermal fibroblasts (HDFs) and examined temporal dynamics of a panel of six RTKs and cell heterogeneity up to 17 days. We observed ~2700 VEGFR1 (vascular endothelial growth factor receptor 1) per cell on 24-h-old cocultured HDF plasma membranes, which do not express VEGFR when cultured alone. We observed 4000–8100 VEGFR2 per cell on cocultured HUVEC plasma membranes throughout endothelial tube formation. We showed steady increase of platelet-derived growth factor receptors (PDGFRs) on cocultured HDF plasma membranes, and more interestingly, 1900–2900 PDGFRβ per plasma membrane were found on HUVECs within the first six hours of coculturing. These quantitative findings will offer us insights into molecular regulation during angiogenesis and help assess in vitro tube formation models and their physiological relevance.
Collapse
|
27
|
Qian Z, Sharma D, Jia W, Radke D, Kamp T, Zhao F. Engineering stem cell cardiac patch with microvascular features representative of native myocardium. Theranostics 2019; 9:2143-2157. [PMID: 31149034 PMCID: PMC6531308 DOI: 10.7150/thno.29552] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 02/14/2019] [Indexed: 12/11/2022] Open
Abstract
The natural myocardium is a highly aligned tissue with an oriented vasculature. Its characteristic cellular as well as nanoscale extracellular matrix (ECM) organization along with an oriented vascular network ensures appropriate blood supply and functional performance. Although significant efforts have been made to develop anisotropic cardiac structure, currently neither an ideal biomaterial nor an effective vascularization strategy to engineer oriented and high-density capillary-like microvessels has been achieved for clinical cardiovascular therapies. A naturally derived oriented ECM nanofibrous scaffold mimics the physiological structure and components of tissue ECM and guides neovascular network formation. The objective of this study was to create an oriented and dense microvessel network with physiological myocardial microvascular features. METHODS Highly aligned decellularized human dermal fibroblast sheets were used as ECM scaffold to regulate physiological alignment of microvascular networks by co-culturing human mesenchymal stem cells (hMSCs) and endothelial cells (ECs). The influence of topographical features on hMSC and EC interaction was investigated to understand underlying mechanisms of neovasculature formation. RESULTS Results demonstrate that the ECM topography can be translated to ECs via CD166 tracks and significantly improved hMSC-EC crosstalk and vascular network formation. The aligned ECM nanofibers enhanced structure, length, and density of microvascular networks compared to randomly organized nanofibrous ECM. Moreover, hMSC-EC co-culture promoted secretion of pro-angiogenic growth factors and matrix remodeling via metalloprotease-2 (MMP-2) activation, which resulted in highly dense vascular network formation with intercapillary distance (20 μm) similar to the native myocardium. CONCLUSION HMSC-EC co-culture on the highly aligned ECM generates physiologically oriented and dense microvascular network, which holds great potential for cardiac tissue engineering.
Collapse
Affiliation(s)
- Zichen Qian
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931, USA
| | - Dhavan Sharma
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931, USA
| | - Wenkai Jia
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931, USA
| | - Daniel Radke
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931, USA
| | - Timothy Kamp
- Stem Cell and Regenerative Medicine Center, University of Wisconsin, Madison, WI 53705, USA
| | - Feng Zhao
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931, USA
| |
Collapse
|
28
|
Li X, Chen C, Dai Y, Huang C, Han Q, Jing L, Ma Y, Xu Y, Liu Y, Zhao L, Wang J, Sun X, Yao X. Cinobufagin suppresses colorectal cancer angiogenesis by disrupting the endothelial mammalian target of rapamycin/hypoxia-inducible factor 1α axis. Cancer Sci 2019; 110:1724-1734. [PMID: 30839155 PMCID: PMC6501006 DOI: 10.1111/cas.13988] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 02/16/2019] [Accepted: 03/03/2019] [Indexed: 12/25/2022] Open
Abstract
Inducing angiogenesis is a hallmark of cancers that sustains tumor growth and metastasis. Neovascularization is a surprisingly early event during the multistage progression of cancer. Cinobufagin, an important bufadienolide originating from Chan Su, has been clinically used to treat cancer in China since the Tang dynasty. Here, we show that cinobufagin suppresses colorectal cancer (CRC) growth in vivo by downregulating angiogenesis. The hierarchized neovasculature is significantly decreased and the vascular network formation is disrupted in HUVEC by cinobufagin in a dose‐dependent way. Endothelial apoptosis is observed by inducing reactive oxygen species (ROS) accumulation and mitochondrial dysfunction which can be neutralized by N‐acetyl‐l‐cysteine (NAC). Expression of hypoxia‐inducible factor 1α (HIF‐1α) is reduced and phosphorylation of mTOR at Ser2481 and Akt at Ser473 is downregulated in HUVEC. Endothelial apoptosis is triggered by cinobufagin by stimulation of Bax and cascade activation of caspase 9 and caspase 3. Increased endothelial apoptosis rate and alterations in the HIF‐1α/mTOR pathway are recapitulated in tumor‐bearing mice in vivo. Further, the anti‐angiogenesis function of cinobufagin is consolidated based on its pro‐apoptotic effects on an EOMA‐derived hemangioendothelioma model. In conclusion, cinobufagin suppresses tumor neovascularization by disrupting the endothelial mTOR/HIF‐1α pathway to trigger ROS‐mediated vascular endothelial cell apoptosis. Cinobufagin is a promising natural anti‐angiogenetic drug that has clinical translation potential and practical application value.
Collapse
Affiliation(s)
- Xiaowu Li
- Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.,The Key Laboratory of Molecular Biology, State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.,Department of General Surgery, The First Affiliated Hospital & School of Clinical Medicine of Guangdong Pharmaceutical University, Guangzhou, China
| | - Chunhui Chen
- The Key Laboratory of Molecular Biology, State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yu Dai
- The Key Laboratory of Molecular Biology, State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Chengzhi Huang
- Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Qinrui Han
- The Key Laboratory of Molecular Biology, State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Linlin Jing
- Traditional Chinese Medicine Integrated Hospital, Southern Medical University, Guangzhou, China
| | - Ye Ma
- The Key Laboratory of Molecular Biology, State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yihua Xu
- The Key Laboratory of Molecular Biology, State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yawei Liu
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Liang Zhao
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Junjiang Wang
- Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Xuegang Sun
- The Key Laboratory of Molecular Biology, State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.,School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xueqing Yao
- Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.,The Key Laboratory of Molecular Biology, State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
29
|
Abstract
The complexity of morphogenesis poses a fundamental challenge to understanding the mechanisms governing the formation of biological patterns and structures. Over the past century, numerous processes have been identified as critically contributing to morphogenetic events, but the interplay between the various components and aspects of pattern formation have been much harder to grasp. The combination of traditional biology with mathematical and computational methods has had a profound effect on our current understanding of morphogenesis and led to significant insights and advancements in the field. In particular, the theoretical concepts of reaction–diffusion systems and positional information, proposed by Alan Turing and Lewis Wolpert, respectively, dramatically influenced our general view of morphogenesis, although typically in isolation from one another. In recent years, agent-based modeling has been emerging as a consolidation and implementation of the two theories within a single framework. Agent-based models (ABMs) are unique in their ability to integrate combinations of heterogeneous processes and investigate their respective dynamics, especially in the context of spatial phenomena. In this review, we highlight the benefits and technical challenges associated with ABMs as tools for examining morphogenetic events. These models display unparalleled flexibility for studying various morphogenetic phenomena at multiple levels and have the important advantage of informing future experimental work, including the targeted engineering of tissues and organs.
Collapse
|
30
|
Hosseini A, Rasmi Y, Rahbarghazi R, Aramwit P, Daeihassani B, Saboory E. Curcumin modulates the angiogenic potential of human endothelial cells via FAK/P-38 MAPK signaling pathway. Gene 2019; 688:7-12. [DOI: 10.1016/j.gene.2018.11.062] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/13/2018] [Accepted: 11/19/2018] [Indexed: 01/15/2023]
|
31
|
Anti-Invasion and Antiangiogenic Effects of Stellettin B through Inhibition of the Akt/Girdin Signaling Pathway and VEGF in Glioblastoma Cells. Cancers (Basel) 2019; 11:cancers11020220. [PMID: 30769863 PMCID: PMC6406657 DOI: 10.3390/cancers11020220] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 02/01/2019] [Accepted: 02/11/2019] [Indexed: 02/06/2023] Open
Abstract
Angiogenesis and invasion are highly related with tumor metastatic potential and recurrence prediction in the most aggressive brain cancer, glioblastoma multiforme (GBM). For the first time, this study reveals that marine-sponge-derived stellettin B reduces angiogenesis and invasion. We discovered that stellettin B reduces migration of glioblastoma cells by scratch wound healing assay and invasion via chamber transwell assay. Further, stellettin B downregulates Akt/Mammalian Target of Rapamycin (Akt/mTOR) and Signal transducer and activator of transcription 3 (Stat3) signaling pathways, which are essential for invasion and angiogenesis in glioblastoma. This study further demonstrates that stellettin B affects filamentous actin (F-actin) rearrangement by decreasing the cross-linkage of phosphor-Girdin (p-Girdin), which attenuates glioblastoma cell invasion. Moreover, stellettin B blocks the expression and secretion of a major proangiogenic factor, vascular endothelial growth factor (VEGF), in glioblastoma cells. Stellettin B also reduces angiogenic tubule formation in human umbilical vein endothelial cells (HUVECs). In vivo, we observed that stellettin B decreased blood vesicle formation in developmental zebrafish and suppressed angiogenesis in Matrigel plug transplant assay in mice. Decreased VEGF transcriptional expression was also found in stellettin B⁻treated zebrafish embryos. Overall, we conclude that stellettin B might be a potential antiangiogenic and anti-invasion agent for future development of therapeutic agents for cancer therapy.
Collapse
|
32
|
Salavati H, Soltani M. The impact of endothelial cells proliferation in a multiscale realistic reproduction of angiogenesis. Biochem Eng J 2019. [DOI: 10.1016/j.bej.2018.11.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
33
|
Cao J, Schnittler H. Putting VE-cadherin into JAIL for junction remodeling. J Cell Sci 2019; 132:132/1/jcs222893. [DOI: 10.1242/jcs.222893] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
ABSTRACT
Junction dynamics of endothelial cells are based on the integration of signal transduction, cytoskeletal remodeling and contraction, which are necessary for the formation and maintenance of monolayer integrity, but also enable repair and regeneration. The VE-cadherin–catenin complex forms the molecular basis of the adherence junctions and cooperates closely with actin filaments. Several groups have recently described small actin-driven protrusions at the cell junctions that are controlled by the Arp2/3 complex, contributing to cell junction regulation. We identified these protrusions as the driving force for VE-cadherin dynamics, as they directly induce new VE-cadherin-mediated adhesion sites, and have accordingly referred to these structures as junction-associated intermittent lamellipodia (JAIL). JAIL extend over only a few microns and thus provide the basis for a subcellular regulation of adhesion. The local (subcellular) VE-cadherin concentration and JAIL formation are directly interdependent, which enables autoregulation. Therefore, this mechanism can contribute a subcellularly regulated adaptation of cell contact dynamics, and is therefore of great importance for monolayer integrity and relative cell migration during wound healing and angiogenesis, as well as for inflammatory responses. In this Review, we discuss the mechanisms and functions underlying these actin-driven protrusions and consider their contribution to the dynamic regulation of endothelial cell junctions.
Collapse
Affiliation(s)
- Jiahui Cao
- Institute of Anatomy and Vascular Biology, Westfälische Wilhelms-Universität Münster, Münster Germany
| | - Hans Schnittler
- Institute of Anatomy and Vascular Biology, Westfälische Wilhelms-Universität Münster, Münster Germany
| |
Collapse
|
34
|
Maniyar R, Chakraborty S, Suriano R. Ethanol Enhances Estrogen Mediated Angiogenesis in Breast Cancer. J Cancer 2018; 9:3874-3885. [PMID: 30410590 PMCID: PMC6218769 DOI: 10.7150/jca.25581] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 08/20/2018] [Indexed: 01/15/2023] Open
Abstract
Angiogenesis, a highly regulated process, is exploited by tumors like breast cancer to ensure a constant supply of oxygen and nutrients and is key for tumor survival and progression. Estrogen and alcohol independently have been observed to contribute to angiogenesis in breast cancer but their combinatorial effects have never been evaluated. The exact mechanism by which estrogen and alcohol contribute to breast cancer angiogenesis remains to be elucidated. In this study, we defined the in vitro effects of the combination of estrogen and alcohol in breast cancer angiogenesis using the tubulogenesis and scratch wound assays. Conditioned media, generated by culturing the murine mammary cancer cell line, TG1-1, in estrogen and ethanol, enhanced tubule formation and migration as well as modulated the MAP Kinase pathway in the murine endothelial cell line, SVEC4-10. Additionally, estrogen and ethanol in combination enhanced the expression of the pro-angiogenic factors VEGF, MMP-9, and eNOS, and modulated Akt activation. These observations suggest that TG1-1 cells secrete pro-angiogenic molecules in response to the combination of estrogen and ethanol that modulate the morphological and migratory properties of endothelial cells. The data presented in this study, is the first in attempting to link the cooperative activity between estrogen and ethanol in breast cancer progression, underscoring correlations first made by epidemiological observations linking the two.
Collapse
Affiliation(s)
- Rachana Maniyar
- Department of Microbiology and Immunology, New York Medical College, Valhalla, New York, United States of America
| | - Sanjukta Chakraborty
- Department of Microbiology and Immunology, New York Medical College, Valhalla, New York, United States of America
| | - Robert Suriano
- Department of Microbiology and Immunology, New York Medical College, Valhalla, New York, United States of America
- Division of Natural Sciences, College of Mount Saint Vincent, Bronx. New York, United States of America
| |
Collapse
|
35
|
Norton KA, Jin K, Popel AS. Modeling triple-negative breast cancer heterogeneity: Effects of stromal macrophages, fibroblasts and tumor vasculature. J Theor Biol 2018; 452:56-68. [PMID: 29750999 DOI: 10.1016/j.jtbi.2018.05.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 04/13/2018] [Accepted: 05/03/2018] [Indexed: 12/20/2022]
Abstract
A hallmark of breast tumors is its spatial heterogeneity that includes its distribution of cancer stem cells and progenitor cells, but also heterogeneity in the tumor microenvironment. In this study we focus on the contributions of stromal cells, specifically macrophages, fibroblasts, and endothelial cells on tumor progression. We develop a computational model of triple-negative breast cancer based on our previous work and expand it to include macrophage infiltration, fibroblasts, and angiogenesis. In vitro studies have shown that the secretomes of tumor-educated macrophages and fibroblasts increase both the migration and proliferation rates of triple-negative breast cancer cells. In vivo studies also demonstrated that blocking signaling of selected secreted factors inhibits tumor growth and metastasis in mouse xenograft models. We investigate the influences of increased migration and proliferation rates on tumor growth, the effect of the presence on fibroblasts or macrophages on growth and morphology, and the contributions of macrophage infiltration on tumor growth. We find that while the presence of macrophages increases overall tumor growth, the increase in macrophage infiltration does not substantially increase tumor growth and can even stifle tumor growth at excessive rates.
Collapse
Affiliation(s)
| | - Kideok Jin
- Department of Biomedical Engineering; Department of Pharmaceutical Science, Albany College of Pharmacy and Health Science, USA
| | - Aleksander S Popel
- Department of Biomedical Engineering; Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University, USA
| |
Collapse
|
36
|
Agent-based modeling of the interaction between CD8+ T cells and Beta cells in type 1 diabetes. PLoS One 2018; 13:e0190349. [PMID: 29320541 PMCID: PMC5761894 DOI: 10.1371/journal.pone.0190349] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 12/13/2017] [Indexed: 12/16/2022] Open
Abstract
We propose an agent-based model for the simulation of the autoimmune response in T1D. The model incorporates cell behavior from various rules derived from the current literature and is implemented on a high-performance computing system, which enables the simulation of a significant portion of the islets in the mouse pancreas. Simulation results indicate that the model is able to capture the trends that emerge during the progression of the autoimmunity. The multi-scale nature of the model enables definition of rules or equations that govern cellular or sub-cellular level phenomena and observation of the outcomes at the tissue scale. It is expected that such a model would facilitate in vivo clinical studies through rapid testing of hypotheses and planning of future experiments by providing insight into disease progression at different scales, some of which may not be obtained easily in clinical studies. Furthermore, the modular structure of the model simplifies tasks such as the addition of new cell types, and the definition or modification of different behaviors of the environment and the cells with ease.
Collapse
|
37
|
Discovery of High-Affinity PDGF-VEGFR Interactions: Redefining RTK Dynamics. Sci Rep 2017; 7:16439. [PMID: 29180757 PMCID: PMC5704011 DOI: 10.1038/s41598-017-16610-z] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 11/14/2017] [Indexed: 01/15/2023] Open
Abstract
Nearly all studies of angiogenesis have focused on uni-family ligand-receptor binding, e.g., VEGFs bind to VEGF receptors, PDGFs bind to PDGF receptors, etc. The discovery of VEGF-PDGFRs binding challenges this paradigm and calls for investigation of other ligand-receptor binding possibilities. We utilized surface plasmon resonance to identify and measure PDGF-to-VEGFR binding rates, establishing cut-offs for binding and non-binding interactions. We quantified the kinetics of the recent VEGF-A:PDGFRβ interaction for the first time with KD = 340 pM. We discovered new PDGF:VEGFR2 interactions with PDGF-AA:R2 KD = 530 nM, PDGF-AB:R2 KD = 110 pM, PDGF-BB:R2 KD = 40 nM, and PDGF-CC:R2 KD = 70 pM. We computationally predict that cross-family PDGF binding could contribute up to 96% of VEGFR2 ligation in healthy conditions and in cancer. Together the identification, quantification, and simulation of these novel cross-family interactions posits new mechanisms for understanding anti-angiogenic drug resistance and presents an expanded role of growth factor signaling with significance in health and disease.
Collapse
|
38
|
Hanging Drop, A Best Three-Dimensional (3D) Culture Method for Primary Buffalo and Sheep Hepatocytes. Sci Rep 2017; 7:1203. [PMID: 28446763 PMCID: PMC5430879 DOI: 10.1038/s41598-017-01355-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 03/27/2017] [Indexed: 02/07/2023] Open
Abstract
Livestock, having close resemblance to humans, could be a better source of primary hepatocytes than rodents. Herein, we successfully developed three-dimensional (3D) culturing system for primary sheep and buffalo hepatocytes. The 3D-structures of sheep hepatocytes were formed on the fifth-day and maintained until the tenth-day on polyHEMA-coated plates and in hanging drops with William’s E media (HDW). Between the cultured and fresh cells, we observed a similar expression of GAPDH, HNF4α, ALB, CYP1A1, CK8 and CK18. Interestingly, a statistically significant increase was noted in the TAT, CPS, AFP, AAT, GSP and PCNA expression. In buffalo hepatocytes culture, 3D-like structures were formed on the third-day and maintained until the sixth-day on polyHEMA and HDW. The expression of HNF4α, GSP, CPS, AFP, AAT, PCNA and CK18 was similar between cultured and fresh cells. Further, a statistically significant increase in the TAT and CK8 expression, and a decrease in the GAPDH, CYP1A1 and ALB expression were noted. Among the culture systems, HDW maintained the liver transcript markers more or less similar to the fresh hepatocytes of the sheep and buffalo for ten and six days, respectively. Taken together, hanging drop is an efficient method for 3D culturing of primary sheep and buffalo hepatocytes.
Collapse
|
39
|
Huang W, Itayama M, Arai F, Furukawa KS, Ushida T, Kawahara T. An angiogenesis platform using a cubic artificial eggshell with patterned blood vessels on chicken chorioallantoic membrane. PLoS One 2017; 12:e0175595. [PMID: 28414752 PMCID: PMC5393577 DOI: 10.1371/journal.pone.0175595] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 03/28/2017] [Indexed: 12/14/2022] Open
Abstract
The chorioallantoic membrane (CAM) containing tiny blood vessels is an alternative to large animals for studies involving angiogenesis and tissue engineering. However, there is no technique to design the direction of growing blood vessels on the CAM at the microscale level for tissue engineering experiments. Here, a methodology is provided to direct blood vessel formation on the surface of a three-dimensional egg yolk using a cubic artificial eggshell with six functionalized membranes. A structure on the lateral side of the eggshell containing a straight channel and an interlinked chamber was designed, and the direction and formation area of blood vessels with blood flow was artfully defined by channels with widths of 70-2000 μm, without sharply reducing embryo viability. The relationship between the size of interlinked chamber and the induction of blood vessels was investigated to establish a theory of design. Role of negative and positive pressure in the induction of CAM with blood vessels was investigated, and air pressure change in the culture chamber was measured to demonstrate the mechanism for blood vessel induction. Histological evaluation showed that components of CAM including chorionic membrane and blood vessels were induced into the channels. Based on our design theory, blood vessels were induced into arrayed channels, and channel-specific injection and screening were realized, which demonstrated proposed applications. The platform with position- and space-controlled blood vessels is therefore a powerful tool for biomedical research, which may afford exciting applications in studies involved in local stimulation of blood vessel networks and those necessary to establish a living system with blood flow from a beating heart.
Collapse
Affiliation(s)
- Wenjing Huang
- Department of Biological Functions Engineering, Kyushu Institute of Technology, Wakamatsu-ku, Kitakyushu, Japan
| | - Makoto Itayama
- Department of Biological Functions Engineering, Kyushu Institute of Technology, Wakamatsu-ku, Kitakyushu, Japan
| | - Fumihito Arai
- Department of Micro-Nano Systems Engineering, Nagoya University, Chikusa-ku, Nagoya, Japan
| | - Katsuko S. Furukawa
- Department of Bioengineering, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Takashi Ushida
- Department of Bioengineering, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- The Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Tomohiro Kawahara
- Department of Biological Functions Engineering, Kyushu Institute of Technology, Wakamatsu-ku, Kitakyushu, Japan
| |
Collapse
|
40
|
Effects of endothelial cell proliferation and migration rates in a computational model of sprouting angiogenesis. Sci Rep 2016; 6:36992. [PMID: 27841344 PMCID: PMC5107954 DOI: 10.1038/srep36992] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 10/24/2016] [Indexed: 01/15/2023] Open
Abstract
Angiogenesis, the recruitment of new blood vessels, is a critical process for the growth, expansion, and metastatic dissemination of developing tumors. Three types of cells make up the new vasculature: tip cells, which migrate in response to gradients of vascular endothelial growth factor (VEGF), stalk cells, which proliferate and extend the vessels, and phalanx cells, which are quiescent and support the sprout. In this study we examine the contribution of tip cell migration rate and stalk cell proliferation rate on the formation of new vasculature. We calculate several vascular metrics, such as the number of vascular bifurcations per unit volume, vascular segment length per unit volume, and vascular tortuosity. These measurements predict that proliferation rate has a greater effect on the spread and extent of vascular growth compared to migration rate. Together, these findings provide strong implications for designing anti-angiogenic therapies that may differentially target endothelial cell proliferation and migration. Computational models can be used to predict optimal anti-angiogenic therapies in combination with other therapeutics to improve outcome.
Collapse
|
41
|
Palm MM, Dallinga MG, van Dijk E, Klaassen I, Schlingemann RO, Merks RMH. Computational Screening of Tip and Stalk Cell Behavior Proposes a Role for Apelin Signaling in Sprout Progression. PLoS One 2016; 11:e0159478. [PMID: 27828952 PMCID: PMC5102492 DOI: 10.1371/journal.pone.0159478] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 05/24/2016] [Indexed: 12/30/2022] Open
Abstract
Angiogenesis involves the formation of new blood vessels by sprouting or splitting of existing blood vessels. During sprouting, a highly motile type of endothelial cell, called the tip cell, migrates from the blood vessels followed by stalk cells, an endothelial cell type that forms the body of the sprout. To get more insight into how tip cells contribute to angiogenesis, we extended an existing computational model of vascular network formation based on the cellular Potts model with tip and stalk differentiation, without making a priori assumptions about the differences between tip cells and stalk cells. To predict potential differences, we looked for parameter values that make tip cells (a) move to the sprout tip, and (b) change the morphology of the angiogenic networks. The screening predicted that if tip cells respond less effectively to an endothelial chemoattractant than stalk cells, they move to the tips of the sprouts, which impacts the morphology of the networks. A comparison of this model prediction with genes expressed differentially in tip and stalk cells revealed that the endothelial chemoattractant Apelin and its receptor APJ may match the model prediction. To test the model prediction we inhibited Apelin signaling in our model and in an in vitro model of angiogenic sprouting, and found that in both cases inhibition of Apelin or of its receptor APJ reduces sprouting. Based on the prediction of the computational model, we propose that the differential expression of Apelin and APJ yields a "self-generated" gradient mechanisms that accelerates the extension of the sprout.
Collapse
Affiliation(s)
- Margriet M. Palm
- Life Sciences Group, Centrum Wiskunde & Informatica, Amsterdam, the Netherlands
| | | | - Erik van Dijk
- Life Sciences Group, Centrum Wiskunde & Informatica, Amsterdam, the Netherlands
| | - Ingeborg Klaassen
- Ocular Angiogenesis Group, Academic Medical Center, Amsterdam, the Netherlands
| | | | - Roeland M. H. Merks
- Life Sciences Group, Centrum Wiskunde & Informatica, Amsterdam, the Netherlands
- Mathematical Institute, Leiden University, Leiden, the Netherlands
| |
Collapse
|
42
|
Bloch N, Harel D. The tumor as an organ: comprehensive spatial and temporal modeling of the tumor and its microenvironment. BMC Bioinformatics 2016; 17:317. [PMID: 27553370 PMCID: PMC4995621 DOI: 10.1186/s12859-016-1168-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 08/11/2016] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Research related to cancer is vast, and continues in earnest in many directions. Due to the complexity of cancer, a better understanding of tumor growth dynamics can be gleaned from a dynamic computational model. We present a comprehensive, fully executable, spatial and temporal 3D computational model of the development of a cancerous tumor together with its environment. RESULTS The model was created using Statecharts, which were then connected to an interactive animation front-end that we developed especially for this work, making it possible to visualize on the fly the on-going events of the system's execution, as well as the effect of various input parameters. We were thus able to gain a better understanding of, e.g., how different amounts or thresholds of oxygen and VEGF (vascular endothelial growth factor) affect the progression of the tumor. We found that the tumor has a critical turning point, where it either dies or recovers. If minimum conditions are met at that time, it eventually develops into a full, active, growing tumor, regardless of the actual amount; otherwise it dies. CONCLUSIONS This brings us to the conclusion that the tumor is in fact a very robust system: changing initial values of VEGF and oxygen can increase the time it takes to become fully developed, but will not necessarily completely eliminate it.
Collapse
Affiliation(s)
- Naamah Bloch
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, 234 Herzl st, 7610001, Rehovot, Israel.
| | - David Harel
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, 234 Herzl st, 7610001, Rehovot, Israel
| |
Collapse
|
43
|
Del Amo C, Borau C, Gutiérrez R, Asín J, García-Aznar JM. Quantification of angiogenic sprouting under different growth factors in a microfluidic platform. J Biomech 2016; 49:1340-1346. [DOI: 10.1016/j.jbiomech.2015.10.026] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 10/13/2015] [Accepted: 10/18/2015] [Indexed: 01/15/2023]
|
44
|
Du Y, Herath SCB, Wang QG, Wang DA, Asada HH, Chen PCY. Three-Dimensional Characterization of Mechanical Interactions between Endothelial Cells and Extracellular Matrix during Angiogenic Sprouting. Sci Rep 2016; 6:21362. [PMID: 26903154 PMCID: PMC4763258 DOI: 10.1038/srep21362] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 01/19/2016] [Indexed: 01/15/2023] Open
Abstract
We studied the three-dimensional cell-extracellular matrix interactions of endothelial cells that form multicellular structures called sprouts. We analyzed the data collected in-situ from angiogenic sprouting experiments and identified the differentiated interaction behavior exhibited by the tip and stalk cells. Moreover, our analysis of the tip cell lamellipodia revealed the diversity in their interaction behavior under certain conditions (e.g., when the heading of a sprout is switched approximately between the long-axis direction of two different lamellipodia). This study marks the first time that new characteristics of such interactions have been identified with shape changes in the sprouts and the associated rearrangements of collagen fibers. Clear illustrations of such changes are depicted in three-dimensional views.
Collapse
Affiliation(s)
- Yue Du
- Department of Mechanical Engineering, National University of Singapore, Singapore.,BioSystems and Micromechanics Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology Program, Singapore
| | - Sahan C B Herath
- Department of Mechanical Engineering, National University of Singapore, Singapore.,BioSystems and Micromechanics Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology Program, Singapore
| | - Qing-guo Wang
- Department of Electrical and Electronic Engineering Science, University of Johannesburg, South Africa
| | - Dong-an Wang
- Division of Bioengineering, Nanyang Technological University, Singapore
| | - H Harry Asada
- BioSystems and Micromechanics Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology Program, Singapore.,Department of Mechanical Engineering, Massachusetts Institute of Technology, USA
| | - Peter C Y Chen
- Department of Mechanical Engineering, National University of Singapore, Singapore.,BioSystems and Micromechanics Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology Program, Singapore
| |
Collapse
|
45
|
Shamloo A, Mohammadaliha N, Heilshorn SC, Bauer AL. A Comparative Study of Collagen Matrix Density Effect on Endothelial Sprout Formation Using Experimental and Computational Approaches. Ann Biomed Eng 2015; 44:929-41. [DOI: 10.1007/s10439-015-1416-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Accepted: 08/04/2015] [Indexed: 12/15/2022]
|
46
|
Chen S, Guo X, Imarenezor O, Imoukhuede PI. Quantification of VEGFRs, NRP1, and PDGFRs on Endothelial Cells and Fibroblasts Reveals Serum, Intra-Family Ligand, and Cross-Family Ligand Regulation. Cell Mol Bioeng 2015. [DOI: 10.1007/s12195-015-0411-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
47
|
Bloch N, Weiss G, Szekely S, Harel D. An Interactive Tool for Animating Biology, and Its Use in Spatial and Temporal Modeling of a Cancerous Tumor and Its Microenvironment. PLoS One 2015; 10:e0133484. [PMID: 26191814 PMCID: PMC4508114 DOI: 10.1371/journal.pone.0133484] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 06/27/2015] [Indexed: 01/15/2023] Open
Abstract
The ability to visualize the ongoing events of a computational model of biology is critical, both in order to see the dynamics of the biological system in action and to enable interaction with the model from which one can observe the resulting behavior. To this end, we have built a new interactive animation tool, SimuLife, for visualizing reactive models of cellular biology. SimuLife is web-based, and is freely accessible at http://simulife.weizmann.ac.il/. We have used SimuLife to animate a model that describes the development of a cancerous tumor, based on the individual components of the system and its environment. This has helped in understanding the dynamics of the tumor and its surrounding blood vessels, and in verifying the behavior, fine-tuning the model accordingly, and learning in which way different factors affect the tumor.
Collapse
Affiliation(s)
- Naamah Bloch
- Dept. of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel
- * E-mail:
| | - Guy Weiss
- Dept. of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel
| | - Smadar Szekely
- Dept. of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel
| | - David Harel
- Dept. of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
48
|
Harkness T, McNulty JD, Prestil R, Seymour SK, Klann T, Murrell M, Ashton RS, Saha K. High-content imaging with micropatterned multiwell plates reveals influence of cell geometry and cytoskeleton on chromatin dynamics. Biotechnol J 2015; 10:1555-67. [PMID: 26097126 DOI: 10.1002/biot.201400756] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 04/24/2015] [Accepted: 06/11/2015] [Indexed: 01/14/2023]
Abstract
Understanding the mechanisms underpinning cellular responses to microenvironmental cues requires tight control not only of the complex milieu of soluble signaling factors, extracellular matrix (ECM) connections and cell-cell contacts within cell culture, but also of the biophysics of human cells. Advances in biomaterial fabrication technologies have recently facilitated detailed examination of cellular biophysics and revealed that constraints on cell geometry arising from the cellular microenvironment influence a wide variety of human cell behaviors. Here, we create an in vitro platform capable of precise and independent control of biochemical and biophysical microenvironmental cues by adapting microcontact printing technology into the format of standard six- to 96-well plates to create MicroContact Printed Well Plates (μCP Well Plates). Automated high-content imaging of human cells seeded on μCP Well Plates revealed tight, highly consistent control of single-cell geometry, cytoskeletal organization, and nuclear elongation. Detailed subcellular imaging of the actin cytoskeleton and chromatin within live human fibroblasts on μCP Well Plates was then used to describe a new relationship between cellular geometry and chromatin dynamics. In summary, the μCP Well Plate platform is an enabling high-content screening technology for human cell biology and cellular engineering efforts that seek to identify key biochemical and biophysical cues in the cellular microenvironment.
Collapse
Affiliation(s)
- Ty Harkness
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA.,Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Jason D McNulty
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA.,Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Ryan Prestil
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA.,Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Stephanie K Seymour
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA.,Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Tyler Klann
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA.,Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael Murrell
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA.,Department of Biomedical Engineering, Yale University, CT, USA.,Systems Biology Institute, Yale University, CT, USA
| | - Randolph S Ashton
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA.,Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Krishanu Saha
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA. .,Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
49
|
Bioactive fish collagen/polycaprolactone composite nanofibrous scaffolds fabricated by electrospinning for 3D cell culture. J Biotechnol 2015; 205:47-58. [DOI: 10.1016/j.jbiotec.2015.01.017] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 01/12/2015] [Accepted: 01/16/2015] [Indexed: 01/15/2023]
|
50
|
Norton KA, Popel AS. An agent-based model of cancer stem cell initiated avascular tumour growth and metastasis: the effect of seeding frequency and location. J R Soc Interface 2015; 11:20140640. [PMID: 25185580 DOI: 10.1098/rsif.2014.0640] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
It is very important to understand the onset and growth pattern of breast primary tumours as well as their metastatic dissemination. In most cases, it is the metastatic disease that ultimately kills the patient. There is increasing evidence that cancer stem cells are closely linked to the progression of the metastatic tumour. Here, we investigate stem cell seeding to an avascular tumour site using an agent-based stochastic model of breast cancer metastatic seeding. The model includes several important cellular features such as stem cell symmetric and asymmetric division, migration, cellular quiescence, senescence, apoptosis and cell division cycles. It also includes external features such as stem cell seeding frequency and location. Using this model, we find that cell seeding rate and location are important features for tumour growth. We also define conditions in which the tumour growth exhibits decremented and exponential growth patterns. Overall, we find that seeding, senescence and division limit affect not only the number of stem cells, but also their spatial and temporal distribution.
Collapse
Affiliation(s)
- Kerri-Ann Norton
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Aleksander S Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21205, USA
| |
Collapse
|