1
|
Schleinhege R, Neumann I, Oeckinghaus A, Schwab A, Pethő Z. A CNA-35-based high-throughput fibrosis assay reveals ORAI1 as a regulator of collagen release from pancreatic stellate cells. Matrix Biol 2025; 135:70-86. [PMID: 39662708 DOI: 10.1016/j.matbio.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 11/22/2024] [Accepted: 12/08/2024] [Indexed: 12/13/2024]
Abstract
RATIONALE Pancreatic stellate cells (PSCs) produce a collagen-rich connective tissue in chronic pancreatitis and pancreatic ductal adenocarcinoma (PDAC). Ca2+-permeable ion channels such as ORAI1 are known to affect PSC proliferation and myofibroblastic phenotype. However, it is unknown whether these channels play a role in collagen secretion. METHODS Using the PSC cell line PS-1, we characterized their cell-derived matrices using staining, mass spectroscopy, and cell migration assays. We developed and validated a high-throughput in vitro fibrosis assay to rapidly determine collagen quantity either with Sirius Red or, in the optimized version, with the collagen-binding peptide CNA-35-tdTomato. We assessed collagen deposition upon stimulating cells with transforming growth factor β1 (TGF-β1) and/or vitamin C without or with ORAI1 modulation. Orai1 expression was assessed by immunohistochemistry in the fibrotic tumor tissue of a murine PDAC model (KPfC). RESULTS We found that TGF-β1 and vitamin C promote collagen deposition from PSCs. We used small interfering RNA (siRNA) and the inhibitor Synta-66 to demonstrate that ORAI1 regulates collagen secretion of PSCs but not NIH-3T3 fibroblasts. Physiological levels of vitamin C induce a drastic increase of the intracellular [Ca2+] in PSCs, with Synta-66 inhibiting Ca2+ influx. Lastly, we revealed Orai1 expression in cancer-associated fibroblasts (CAFs) in murine PDAC (KPfC) samples. CONCLUSION In conclusion, our study introduces a robust in vitro assay for fibrosis and identifies ORAI1 as being engaged in PSC-driven fibrosis.
Collapse
Affiliation(s)
- Rieke Schleinhege
- Institute of Physiology II, University of Münster, Robert-Koch Str. 27B, 48149, Germany
| | - Ilka Neumann
- Institute of Physiology II, University of Münster, Robert-Koch Str. 27B, 48149, Germany
| | - Andrea Oeckinghaus
- Institute of Molecular Tumor Biology, University of Münster, 48149, Germany
| | - Albrecht Schwab
- Institute of Physiology II, University of Münster, Robert-Koch Str. 27B, 48149, Germany
| | - Zoltán Pethő
- Institute of Physiology II, University of Münster, Robert-Koch Str. 27B, 48149, Germany.
| |
Collapse
|
2
|
Lim YW, Quinn R, Bharti K, Ferrer M, Zarkoob H, Song MJ. Development of immunocompetent full thickness skin tissue constructs to model skin fibrosis for high-throughput drug screening. Biofabrication 2024; 17:015033. [PMID: 39622178 PMCID: PMC11638742 DOI: 10.1088/1758-5090/ad998c] [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: 05/20/2024] [Revised: 10/30/2024] [Accepted: 12/02/2024] [Indexed: 12/14/2024]
Abstract
The lack of the immune component in most of the engineered skin models remains a challenge to study the interplay between different immune and non-immune cell types of the skin. Immunocompetent humanin vitroskin models offer potential advantages in recapitulatingin vivolike behavior which can serve to accelerate translational research and therapeutics development for skin diseases. Here we describe a three-dimensional human full-thickness skin (FTS) equivalent incorporating polarized M1 and M2 macrophages from human peripheral CD14+monocytes. This macrophage-incorporated FTS model demonstrates discernible immune responses with physiologically relevant cytokine production and macrophage plasticity under homeostatic and lipopolysaccharide stimulation conditions. M2-incorporated FTS recapitulates skin fibrosis phenotypes with transforming growth factor-β1 treatment as reflected by significant collagen deposition and myofibroblast expression, demonstrating a M2 potentiation effect. In conclusion, we successfully biofabricated an immunocompetent FTS with functional macrophages in a high-throughput (HT) amenable format. This model is the first step towards a HT-assay platform to develop new therapeutics for skin diseases.
Collapse
Affiliation(s)
- Yi Wei Lim
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, United States of America
| | - Russell Quinn
- National Eye Institute, National Institutes of Health, Bethesda, MD 20814, United States of America
| | - Kapil Bharti
- National Eye Institute, National Institutes of Health, Bethesda, MD 20814, United States of America
| | - Marc Ferrer
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, United States of America
| | - Hoda Zarkoob
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, United States of America
| | - Min Jae Song
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, United States of America
| |
Collapse
|
3
|
Sander EA, El-Hattab MY, Jacobson KR, Klingelhutz AJ, Ankrum JA, Calve S. Fibroblast-Adipocyte Lineage Cell Interactions Result in Differential Production of Extracellular Matrix Proteins. Cell Mol Bioeng 2024; 17:597-608. [PMID: 39926384 PMCID: PMC11799492 DOI: 10.1007/s12195-024-00829-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 10/08/2024] [Indexed: 02/11/2025] Open
Abstract
Introduction Scarring from traumatic injury, burns, and other complications remains a significant problem that diminishes quality of life for millions of people worldwide. A common target for the development of new therapies to promote healing and reduce scarring are myofibroblasts because of their central role in pathological scarring. Recent work indicates that adipocyte lineage cells also contribute to the wound healing process, including clinical reports that indicate that the placement of autologous adipose micrografts at the surgical site improves the appearance and pliability of existing scars. Methods To better understand how adipocyte lineage cells interact with fibroblasts to promote healing, we first utilized an in vitro model of wound healing to visualize fibroblast spheroid collagen deposition via time-lapse imaging. We then introduced pre-adipocyte and adipocyte spheroids to visualize pair-wise spheroid interactions and collagen deposition among all three cell types. Finally, we quantified differences in the extracellular matrix (ECM) proteins produced using liquid chromatography with tandem mass spectrometry (LC-MS/MS). Results We found that all three cell-types contribute to ECM deposition and that the composition of the ECM proteins, or matrisome, was significantly different depending on which cells were co-cultured together. Conclusions By better understanding the interactions among these cell types, novel adipose-tissue-based therapeutic approaches can be developed to improve wound healing and reduce scar tissue. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-024-00829-8.
Collapse
Affiliation(s)
- Edward A. Sander
- Roy J. Carver Department of Biomedical Engineering, College of Engineering, University of Iowa, 5629 Seamans Center, Iowa City, IA 52242 USA
| | - Mariam Y. El-Hattab
- Roy J. Carver Department of Biomedical Engineering, College of Engineering, University of Iowa, 5629 Seamans Center, Iowa City, IA 52242 USA
| | - Kathryn R. Jacobson
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, 1111 Engineering Dr, Boulder, CO USA
| | - Aloysius J. Klingelhutz
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA USA
| | - James A. Ankrum
- Roy J. Carver Department of Biomedical Engineering, College of Engineering, University of Iowa, 5629 Seamans Center, Iowa City, IA 52242 USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA USA
| | - Sarah Calve
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, 1111 Engineering Dr, Boulder, CO USA
| |
Collapse
|
4
|
Schaart JM, Kea-Te Lindert M, Roverts R, Nijhuis WH, Sommerdijk N, Akiva A. Cell-induced collagen alignment in a 3D in vitro culture during extracellular matrix production. J Struct Biol 2024; 216:108096. [PMID: 38697586 DOI: 10.1016/j.jsb.2024.108096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 04/24/2024] [Accepted: 04/27/2024] [Indexed: 05/05/2024]
Abstract
The bone extracellular matrix consists of a highly organized collagen matrix that is mineralized with carbonated hydroxyapatite. Even though the structure and composition of bone have been studied extensively, the mechanisms underlying collagen matrix organization remain elusive. In this study, we used a 3D cell culture system in which osteogenic cells deposit and orient the collagen matrix that is subsequently mineralized. Using live fluorescence imaging combined with volume electron microscopy, we visualize the organization of the cells and collagen in the cell culture. We show that the osteogenically induced cells are organizing the collagen matrix during development. Based on the observation of tunnel-like structures surrounded by aligned collagen in the center of the culture, we propose that osteoblasts organize the deposited collagen during migration through the culture. Overall, we show that cell-matrix interactions are involved in collagen alignment during early-stage osteogenic differentiation and that the matrix is organized by the osteoblasts in the absence of osteoclast activity.
Collapse
Affiliation(s)
- Judith M Schaart
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein Zuid 28, 6525GA Nijmegen, The Netherlands
| | - Mariska Kea-Te Lindert
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein Zuid 28, 6525GA Nijmegen, The Netherlands; Electron Microscopy Center, Radboudumc Technology Center Microscopy, Radboud University Medical Center, Geert Grooteplein Noord 29, 6525EZ Nijmegen, The Netherlands
| | - Rona Roverts
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein Zuid 28, 6525GA Nijmegen, The Netherlands; Electron Microscopy Center, Radboudumc Technology Center Microscopy, Radboud University Medical Center, Geert Grooteplein Noord 29, 6525EZ Nijmegen, The Netherlands
| | - Wouter H Nijhuis
- Department of Orthopaedic Surgery, University Medical Centre Utrecht, Wilhelmina Children's Hospital, 3508GA Utrecht, The Netherlands
| | - Nico Sommerdijk
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein Zuid 28, 6525GA Nijmegen, The Netherlands; Electron Microscopy Center, Radboudumc Technology Center Microscopy, Radboud University Medical Center, Geert Grooteplein Noord 29, 6525EZ Nijmegen, The Netherlands.
| | - Anat Akiva
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein Zuid 28, 6525GA Nijmegen, The Netherlands; Electron Microscopy Center, Radboudumc Technology Center Microscopy, Radboud University Medical Center, Geert Grooteplein Noord 29, 6525EZ Nijmegen, The Netherlands.
| |
Collapse
|
5
|
Perez KA, Deppe DW, Filas A, Singh SA, Aikawa E. Multimodal Analytical Tools to Enhance Mechanistic Understanding of Aortic Valve Calcification. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:539-550. [PMID: 37517686 PMCID: PMC10988764 DOI: 10.1016/j.ajpath.2023.06.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 06/14/2023] [Accepted: 06/29/2023] [Indexed: 08/01/2023]
Abstract
This review focuses on technologies at the core of calcific aortic valve disease (CAVD) and drug target research advancement, including transcriptomics, proteomics, and molecular imaging. We examine how bulk RNA sequencing and single-cell RNA sequencing have engendered organismal genomes and transcriptomes, promoting the analysis of tissue gene expression profiles and cell subpopulations, respectively. We bring into focus how the field is also largely influenced by increasingly accessible proteome profiling techniques. In unison, global transcriptional and protein expression analyses allow for increased understanding of cellular behavior and pathogenic pathways under pathologic stimuli including stress, inflammation, low-density lipoprotein accumulation, increased calcium and phosphate levels, and vascular injury. We also look at how direct investigation of protein signatures paves the way for identification of targetable pathways for pharmacologic intervention. Here, we note that imaging techniques, once a clinical diagnostic tool for late-stage CAVD, have since been refined to address a clinical need to identify microcalcifications using positron emission tomography/computed tomography and even detect in vivo cellular events indicative of early stage CAVD and map the expression of identified proteins in animal models. Together, these techniques generate a holistic approach to CAVD investigation, with the potential to identify additional novel regulatory pathways.
Collapse
Affiliation(s)
- Katelyn A Perez
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Daniel W Deppe
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Aidan Filas
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Sasha A Singh
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
6
|
Werner MP, Kučikas V, Voß K, Abel D, Jockenhoevel S, van Zandvoort MAMJ, Schmitz-Rode T. Multiphoton Imaging of Maturation in Tissue Engineering. Tissue Eng Part C Methods 2024; 30:38-48. [PMID: 38115629 DOI: 10.1089/ten.tec.2023.0141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2023] Open
Abstract
Donor cell-specific tissue-engineered (TE) implants are a promising therapy for personalized treatment of cardiovascular diseases, but current development protocols lack a stable longitudinal assessment of tissue development at subcellular resolution. As a first step toward such an assessment approach, in this study we establish a generalized labeling and imaging protocol to obtain quantified maturation parameters of TE constructs in three dimensions (3D) without the need of histological slicing, thus leaving the tissue intact. Focusing on intracellular matrix (ICM) and extracellular matrix (ECM) networks, multiphoton laser scanning microscopy (MPLSM) was used to investigate TE patches of different conditioning durations of up to 21 days. We show here that with a straightforward labeling procedure of whole-mount samples (so without slicing into thin histological sections), followed by an easy-to-use multiphoton imaging process, we obtained high-quality images of the tissue in 3D at various time points during development. The stacks of images could then be further analyzed to visualize and quantify the volume of cell coverage as well as the volume fraction and network of structural proteins. We showed that collagen and alpha-smooth muscle actin (α-SMA) volume fractions increased as normalized to full tissue volume and proportional to the cell count, with a converging trend to the final density of (4.0% ± 0.6%) and (7.6% ± 0.7%), respectively. The image analysis of ICM and ECM revealed a developing and widely branched interconnected matrix. We are currently working on the second step, that is, to integrate MPLSM endoscopy into a dynamic bioreactor system to monitor the maturation of intact TE constructs over time, thus without the need to take them out.
Collapse
Affiliation(s)
- Maximilian P Werner
- Department of Biohybrid & Medical Textiles (BioTex), Institute of Applied Medical Engineering (AME), Helmholtz Institute, RWTH Aachen University, Aachen, Germany
- Aachen-Maastricht-Institute for Biobased Materials (AMIBM), Maastricht University, Geleen, The Netherlands
| | - Vytautas Kučikas
- Institute of Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany
| | - Kirsten Voß
- Institute of Automatic Control (IRT), RWTH Aachen University, Aachen, Germany
| | - Dirk Abel
- Institute of Automatic Control (IRT), RWTH Aachen University, Aachen, Germany
| | - Stefan Jockenhoevel
- Department of Biohybrid & Medical Textiles (BioTex), Institute of Applied Medical Engineering (AME), Helmholtz Institute, RWTH Aachen University, Aachen, Germany
- Aachen-Maastricht-Institute for Biobased Materials (AMIBM), Maastricht University, Geleen, The Netherlands
| | - Marc A M J van Zandvoort
- Institute of Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany
- Department of Genetics and Cell Biology, Cardiovascular Research Institute Maastricht (CARIM), School for Oncology and Developmental Biology (GROW), Maastricht, The Netherlands
| | - Thomas Schmitz-Rode
- Department of Biohybrid & Medical Textiles (BioTex), Institute of Applied Medical Engineering (AME), Helmholtz Institute, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
7
|
Flajnik MF, Stanfield R, Pokidysheva EN, Boudko SP, Wilson I, Ohta Y. An Ancient MHC-Linked Gene Encodes a Nonrearranging Shark Antibody, UrIg, Convergent with IgG. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1042-1051. [PMID: 37540118 PMCID: PMC10530332 DOI: 10.4049/jimmunol.2300361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 07/12/2023] [Indexed: 08/05/2023]
Abstract
Gnathostome adaptive immunity is defined by the Ag receptors, Igs and TCRs, and the MHC. Cartilaginous fish are the oldest vertebrates with these adaptive hallmarks. We and others have unearthed nonrearranging Ag receptor-like genes in several vertebrates, some of which are encoded in the MHC or in MHC paralogous regions. One of these genes, named UrIg, was detected in the class III region of the shark MHC that encodes a protein with typical V and C domains such as those found in conventional Igs and TCRs. As no transmembrane region was detected in gene models or cDNAs, the protein does not appear to act as a receptor. Unlike some other shark Ig genes, the UrIg V region shows no evidence of RAG-mediated rearrangement, and thus it is likely related to other V genes that predated the invasion of the RAG transposon. The UrIg gene is present in all elasmobranchs and evolves conservatively, unlike Igs and TCRs. Also, unlike Ig/TCR, the gene is not expressed in secondary lymphoid tissues, but mainly in the liver. Recombinant forms of the molecule form disulfide-linked homodimers, which is the form also detected in many shark tissues by Western blotting. mAbs specific for UrIg identify the protein in the extracellular matrix of several shark tissues by immunohistochemistry. We propose that UrIg is related to the V gene invaded by the RAG transposon, consistent with the speculation of emergence of Ig/TCR within the MHC or proto-MHC.
Collapse
Affiliation(s)
- Martin F Flajnik
- Department of Microbiology and Immunology, University of Maryland, Baltimore, MD
| | - Robyn Stanfield
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA
| | - Elena N Pokidysheva
- Division of Nephrology and Hypertension, Department of Medicine, Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, TN
| | - Sergei P Boudko
- Division of Nephrology and Hypertension, Department of Medicine, Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, TN
- Department of Biochemistry, Vanderbilt University, Nashville, TN
| | - Ian Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA
| | - Yuko Ohta
- Department of Microbiology and Immunology, University of Maryland, Baltimore, MD
| |
Collapse
|
8
|
Zhao Z, Deng J, Fan D. Green biomanufacturing in recombinant collagen biosynthesis: trends and selection in various expression systems. Biomater Sci 2023; 11:5439-5461. [PMID: 37401335 DOI: 10.1039/d3bm00724c] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2023]
Abstract
Collagen, classically derived from animal tissue, is an all-important protein material widely used in biomedical materials, cosmetics, fodder, food, etc. The production of recombinant collagen through different biological expression systems using bioengineering techniques has attracted significant interest in consideration of increasing market demand and the process complexity of extraction. Green biomanufacturing of recombinant collagen has become one of the focus topics. While the bioproduction of recombinant collagens (type I, II, III, etc.) has been commercialized in recent years, the biosynthesis of recombinant collagen is extremely challenging due to protein immunogenicity, yield, degradation, and other issues. The rapid development of synthetic biology allows us to perform a heterologous expression of proteins in diverse expression systems, thus optimizing the production and bioactivities of recombinant collagen. This review describes the research progress in the bioproduction of recombinant collagen over the past two decades, focusing on different expression systems (prokaryotic organisms, yeasts, plants, insects, mammalian and human cells, etc.). We also discuss the challenges and future trends in developing market-competitive recombinant collagens.
Collapse
Affiliation(s)
- Zilong Zhao
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, Xi'an 710069, Shaanxi, China.
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710069, Shaanxi, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, Shaanxi, China
| | - Jianjun Deng
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, Xi'an 710069, Shaanxi, China.
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710069, Shaanxi, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, Shaanxi, China
| | - Daidi Fan
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, Xi'an 710069, Shaanxi, China.
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710069, Shaanxi, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, Shaanxi, China
| |
Collapse
|
9
|
Luu RJ, Hoefler BC, Gard AL, Ritenour CR, Rogers MT, Kim ES, Coppeta JR, Cain BP, Isenberg BC, Azizgolshani H, Fajardo-Ramirez OR, García-Cardeña G, Lech MP, Tomlinson L, Charest JL, Williams C. Fibroblast activation in response to TGFβ1 is modulated by co-culture with endothelial cells in a vascular organ-on-chip platform. Front Mol Biosci 2023; 10:1160851. [PMID: 37577751 PMCID: PMC10421749 DOI: 10.3389/fmolb.2023.1160851] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 06/06/2023] [Indexed: 08/15/2023] Open
Abstract
Background: Tissue fibrosis is a major healthcare burden that affects various organs in the body for which no effective treatments exist. An underlying, emerging theme across organs and tissue types at early stages of fibrosis is the activation of pericytes and/or fibroblasts in the perivascular space. In hepatic tissue, it is well known that liver sinusoidal endothelial cells (EC) help maintain the quiescence of stellate cells, but whether this phenomenon holds true for other endothelial and perivascular cell types is not well studied. Methods: The goal of this work was to develop an organ-on-chip microvascular model to study the effect of EC co-culture on the activation of perivascular cells perturbed by the pro-fibrotic factor TGFβ1. A high-throughput microfluidic platform, PREDICT96, that was capable of imparting physiologically relevant fluid shear stress on the cultured endothelium was utilized. Results: We first studied the activation response of several perivascular cell types and selected a cell source, human dermal fibroblasts, that exhibited medium-level activation in response to TGFβ1. We also demonstrated that the PREDICT96 high flow pump triggered changes in select shear-responsive factors in human EC. We then found that the activation response of fibroblasts was significantly blunted in co-culture with EC compared to fibroblast mono-cultures. Subsequent studies with conditioned media demonstrated that EC-secreted factors play at least a partial role in suppressing the activation response. A Luminex panel and single cell RNA-sequencing study provided additional insight into potential EC-derived factors that could influence fibroblast activation. Conclusion: Overall, our findings showed that EC can reduce myofibroblast activation of perivascular cells in response to TGFβ1. Further exploration of EC-derived factors as potential therapeutic targets in fibrosis is warranted.
Collapse
Affiliation(s)
- Rebeccah J. Luu
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| | - B. Christopher Hoefler
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| | - Ashley L. Gard
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| | | | - Miles T. Rogers
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| | - Ernest S. Kim
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| | - Jonathan R. Coppeta
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| | - Brian P. Cain
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| | - Brett C. Isenberg
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| | - Hesham Azizgolshani
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| | - Oscar R. Fajardo-Ramirez
- Laboratory for Systems Mechanobiology, Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Guillermo García-Cardeña
- Laboratory for Systems Mechanobiology, Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | | | | | - Joseph L. Charest
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| | - Corin Williams
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| |
Collapse
|
10
|
Stashko C, Hayward MK, Northey JJ, Pearson N, Ironside AJ, Lakins JN, Oria R, Goyette MA, Mayo L, Russnes HG, Hwang ES, Kutys ML, Polyak K, Weaver VM. A convolutional neural network STIFMap reveals associations between stromal stiffness and EMT in breast cancer. Nat Commun 2023; 14:3561. [PMID: 37322009 PMCID: PMC10272194 DOI: 10.1038/s41467-023-39085-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 05/26/2023] [Indexed: 06/17/2023] Open
Abstract
Intratumor heterogeneity associates with poor patient outcome. Stromal stiffening also accompanies cancer. Whether cancers demonstrate stiffness heterogeneity, and if this is linked to tumor cell heterogeneity remains unclear. We developed a method to measure the stiffness heterogeneity in human breast tumors that quantifies the stromal stiffness each cell experiences and permits visual registration with biomarkers of tumor progression. We present Spatially Transformed Inferential Force Map (STIFMap) which exploits computer vision to precisely automate atomic force microscopy (AFM) indentation combined with a trained convolutional neural network to predict stromal elasticity with micron-resolution using collagen morphological features and ground truth AFM data. We registered high-elasticity regions within human breast tumors colocalizing with markers of mechanical activation and an epithelial-to-mesenchymal transition (EMT). The findings highlight the utility of STIFMap to assess mechanical heterogeneity of human tumors across length scales from single cells to whole tissues and implicates stromal stiffness in tumor cell heterogeneity.
Collapse
Affiliation(s)
- Connor Stashko
- Department of Surgery, University of California, San Francisco, CA, USA
- Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA, USA
| | - Mary-Kate Hayward
- Department of Surgery, University of California, San Francisco, CA, USA
- Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA, USA
| | - Jason J Northey
- Department of Surgery, University of California, San Francisco, CA, USA
- Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA, USA
| | | | - Alastair J Ironside
- Department of Pathology, Western General Hospital, NHS Lothian, Edinburgh, UK
| | - Johnathon N Lakins
- Department of Surgery, University of California, San Francisco, CA, USA
- Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA, USA
| | - Roger Oria
- Department of Surgery, University of California, San Francisco, CA, USA
- Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA, USA
| | - Marie-Anne Goyette
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Lakyn Mayo
- Department of Cell and Tissue Biology, School of Dentistry, University of California, San Francisco, San Francisco, CA, USA
| | - Hege G Russnes
- Department of Pathology and Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - E Shelley Hwang
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Matthew L Kutys
- Department of Cell and Tissue Biology, School of Dentistry, University of California, San Francisco, San Francisco, CA, USA
- UCSF Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Kornelia Polyak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Valerie M Weaver
- Department of Surgery, University of California, San Francisco, CA, USA.
- Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA, USA.
- UCSF Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA.
- Department of Radiation Oncology, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
11
|
Brennecke BR, Yang US, Liu S, Ilerisoy FS, Ilerisoy BN, Joglekar A, Kim LB, Peachee SJ, Richtsmeier SL, Stephens SB, Sander EA, Strack S, Moninger TO, Ankrum JA, Imai Y. Utilization of commercial collagens for preparing well-differentiated human beta cells for confocal microscopy. Front Endocrinol (Lausanne) 2023; 14:1187216. [PMID: 37305047 PMCID: PMC10248405 DOI: 10.3389/fendo.2023.1187216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 05/09/2023] [Indexed: 06/13/2023] Open
Abstract
Introduction With technical advances, confocal and super-resolution microscopy have become powerful tools to dissect cellular pathophysiology. Cell attachment to glass surfaces compatible with advanced imaging is critical prerequisite but remains a considerable challenge for human beta cells. Recently, Phelps et al. reported that human beta cells plated on type IV collagen (Col IV) and cultured in neuronal medium preserve beta cell characteristics. Methods We examined human islet cells plated on two commercial sources of Col IV (C6745 and C5533) and type V collagen (Col V) for differences in cell morphology by confocal microscopy and secretory function by glucose-stimulated insulin secretion (GSIS). Collagens were authenticated by mass spectrometry and fluorescent collagen-binding adhesion protein CNA35. Results All three preparations allowed attachment of beta cells with high nuclear localization of NKX6.1, indicating a well-differentiated status. All collagen preparations supported robust GSIS. However, the morphology of islet cells differed between the 3 preparations. C5533 showed preferable features as an imaging platform with the greatest cell spread and limited stacking of cells followed by Col V and C6745. A significant difference in attachment behavior of C6745 was attributed to the low collagen contents of this preparation indicating importance of authentication of coating material. Human islet cells plated on C5533 showed dynamic changes in mitochondria and lipid droplets (LDs) in response to an uncoupling agent 2-[2-[4-(trifluoromethoxy)phenyl]hydrazinylidene]-propanedinitrile (FCCP) or high glucose + oleic acid. Discussion An authenticated preparation of Col IV provides a simple platform to apply advanced imaging for studies of human islet cell function and morphology.
Collapse
Affiliation(s)
- Brianna R. Brennecke
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, United States
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, United States
| | - USeong Yang
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, United States
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, United States
| | - Siming Liu
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, United States
| | - Fatma S. Ilerisoy
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, United States
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, United States
| | - Beyza N. Ilerisoy
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, United States
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, United States
| | - Aditya Joglekar
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, United States
| | - Lucy B. Kim
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, United States
| | - Spencer J. Peachee
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, United States
| | - Syreine L. Richtsmeier
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, United States
| | - Samuel B. Stephens
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, United States
| | - Edward A. Sander
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, United States
| | - Stefan Strack
- Department of Neuroscience and Pharmacology, Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, United States
| | - Thomas O. Moninger
- Central Microscopy Research Facility, Roy G. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - James A. Ankrum
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, United States
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, United States
| | - Yumi Imai
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, United States
- Medical Service, Endocrinology Section, Iowa City Veterans Affairs Medical Center, Iowa City, IA, United States
| |
Collapse
|
12
|
Ge B, Wei M, Bao B, Pan Z, Elango J, Wu W. The Role of Integrin Receptor's α and β Subunits of Mouse Mesenchymal Stem Cells on the Interaction of Marine-Derived Blacktip Reef Shark ( Carcharhinus melanopterus) Skin Collagen. Int J Mol Sci 2023; 24:ijms24119110. [PMID: 37298062 DOI: 10.3390/ijms24119110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/25/2023] [Accepted: 05/10/2023] [Indexed: 06/12/2023] Open
Abstract
Marine collagen (MC) has recently attracted more attention in tissue engineering as a biomaterial substitute due to its significant role in cellular signaling mechanisms, especially in mesenchymal stem cells (MSCs). However, the actual signaling mechanism of MC in MSC growth, which is highly influenced by their molecular pattern, is poorly understood. Hence, we investigated the integrin receptors (α1β1, α2β1, α10β1, and α11β1) binding mechanism and proliferation of MCs (blacktip reef shark collagen (BSC) and blue shark collagen (SC)) compared to bovine collagen (BC) on MSCs behavior through functionalized collagen molecule probing for the first time. The results showed that BSC and SC had higher proliferation rates and accelerated scratch wound healing by increasing migratory rates of MSCs. Cell adhesion and spreading results demonstrated that MC had a better capacity to anchor MSCs and maintain cell morphology than controls. Living cell observations showed that BSC was gradually assembled by cells into the ECM network within 24 h. Interestingly, qRT-PCR and ELISA revealed that the proliferative effect of MC was triggered by interacting with specific integrin receptors such as α2β1, α10β1, and α11β1 of MSCs. Accordingly, BSC accelerated MSCs' growth, adhesion, shape, and spreading by interacting with specific integrin subunits (α2 and β1) and thereby triggering further signaling cascade mechanisms.
Collapse
Affiliation(s)
- Baolin Ge
- Department of Marine Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Mingjun Wei
- Department of Marine Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Bin Bao
- Department of Marine Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Zhilin Pan
- Department of Marine Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Jeevithan Elango
- Department of Marine Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
- Department of Biomaterials Engineering, Faculty of Health Sciences, UCAM-Universidad Católica San Antonio de Murcia, Guadalupe, 30107 Murcia, Spain
| | - Wenhui Wu
- Department of Marine Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| |
Collapse
|
13
|
Kim J. Characterization of Biocompatibility of Functional Bioinks for 3D Bioprinting. Bioengineering (Basel) 2023; 10:bioengineering10040457. [PMID: 37106644 PMCID: PMC10135811 DOI: 10.3390/bioengineering10040457] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/02/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Three-dimensional (3D) bioprinting with suitable bioinks has become a critical tool for fabricating 3D biomimetic complex structures mimicking physiological functions. While enormous efforts have been devoted to developing functional bioinks for 3D bioprinting, widely accepted bioinks have not yet been developed because they have to fulfill stringent requirements such as biocompatibility and printability simultaneously. To further advance our knowledge of the biocompatibility of bioinks, this review presents the evolving concept of the biocompatibility of bioinks and standardization efforts for biocompatibility characterization. This work also briefly reviews recent methodological advances in image analyses to characterize the biocompatibility of bioinks with regard to cell viability and cell-material interactions within 3D constructs. Finally, this review highlights a number of updated contemporary characterization technologies and future perspectives to further advance our understanding of the biocompatibility of functional bioinks for successful 3D bioprinting.
Collapse
Affiliation(s)
- Jinku Kim
- Department of Biological and Chemical Engineering, Hongik University, Sejong 30016, Republic of Korea
| |
Collapse
|
14
|
de Sousa Lobo Ferreira Querido R, Ji X, Lakha R, Goodyear RJ, Richardson GP, Vizcarra CL, Olson ES. Visualizing Collagen Fibrils in the Cochlea's Tectorial and Basilar Membranes Using a Fluorescently Labeled Collagen-Binding Protein Fragment. J Assoc Res Otolaryngol 2023; 24:147-157. [PMID: 36725777 PMCID: PMC10121988 DOI: 10.1007/s10162-023-00889-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 01/16/2023] [Indexed: 02/03/2023] Open
Abstract
PURPOSE A probe that binds to unfixed collagen fibrils was used to image the shapes and fibrous properties of the TM and BM. The probe (CNA35) is derived from the bacterial adhesion protein CNA. We present confocal images of hydrated gerbil TM, BM, and other cochlear structures stained with fluorescently labeled CNA35. A primary purpose of this article is to describe the use of the CNA35 collagen probe in the cochlea. METHODS Recombinant poly-histidine-tagged CNA35 was expressed in Escherichia coli, purified by cobalt-affinity chromatography, fluorescence labeled, and further purified by gel filtration chromatography. Cochleae from freshly harvested gerbil bullae were irrigated with and then incubated in CNA35 for periods ranging from 2 h - overnight. The cochleae were fixed, decalcified, and dissected. Isolated cochlear turns were imaged by confocal microscopy. RESULTS The CNA35 probe stained the BM and TM, and volumetric imaging revealed the shape of these structures and the collagen fibrils within them. The limbal zone of the TM stained intensely. In samples from the cochlear base, intense staining was detected on the side of the TM that faces hair cells. In the BM pectinate zone, staining was intense at the upper and lower boundaries. The BM arcuate zone was characterized by a prominent longitudinal collagenous structure. The spiral ligament, limbus and lamina stained for collagen, and within the spiral limbus the habenula perforata were outlined with intense staining. CONCLUSION The CNA35 probe provides a unique and useful view of collagenous structures in the cochlea.
Collapse
Affiliation(s)
| | - Xiang Ji
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Department of Chemistry, Barnard College, New York, NY, USA
| | - Rabina Lakha
- Department of Chemistry, Barnard College, New York, NY, USA
| | - Richard J Goodyear
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, UK
| | - Guy P Richardson
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, UK
| | | | - Elizabeth S Olson
- Department of Otolaryngology, Head and Neck Surgery, Columbia University, New York, NY, USA.
- Department of Biomedical Engineering, Columbia University, New York, NY, USA.
| |
Collapse
|
15
|
Timin G, Milinkovitch MC. High-resolution confocal and light-sheet imaging of collagen 3D network architecture in very large samples. iScience 2023; 26:106452. [PMID: 37020961 PMCID: PMC10067766 DOI: 10.1016/j.isci.2023.106452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 02/06/2023] [Accepted: 03/15/2023] [Indexed: 04/03/2023] Open
Abstract
Although notoriously difficult, imaging collagen network architecture, a key element affecting tissue mechanical properties, is of paramount importance in developmental and cancer biology. Here, we introduce a simple and robust method of whole-mount collagen staining with the 'Fast Green' dye that provides unmatched visualization of collagen 3D network architecture, via confocal or light-sheet microscopy, compatible with solvent-based tissue clearing and immunostaining.
Collapse
|
16
|
Hsu KS, Dunleavey JM, Szot C, Yang L, Hilton MB, Morris K, Seaman S, Feng Y, Lutz EM, Koogle R, Tomassoni-Ardori F, Saha S, Zhang XM, Zudaire E, Bajgain P, Rose J, Zhu Z, Dimitrov DS, Cuttitta F, Emenaker NJ, Tessarollo L, St. Croix B. Cancer cell survival depends on collagen uptake into tumor-associated stroma. Nat Commun 2022; 13:7078. [PMID: 36400786 PMCID: PMC9674701 DOI: 10.1038/s41467-022-34643-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 11/01/2022] [Indexed: 11/19/2022] Open
Abstract
Collagen I, the most abundant protein in humans, is ubiquitous in solid tumors where it provides a rich source of exploitable metabolic fuel for cancer cells. While tumor cells were unable to exploit collagen directly, here we show they can usurp metabolic byproducts of collagen-consuming tumor-associated stroma. Using genetically engineered mouse models, we discovered that solid tumor growth depends upon collagen binding and uptake mediated by the TEM8/ANTXR1 cell surface protein in tumor-associated stroma. Tumor-associated stromal cells processed collagen into glutamine, which was then released and internalized by cancer cells. Under chronic nutrient starvation, a condition driven by the high metabolic demand of tumors, cancer cells exploited glutamine to survive, an effect that could be reversed by blocking collagen uptake with TEM8 neutralizing antibodies. These studies reveal that cancer cells exploit collagen-consuming stromal cells for survival, exposing an important vulnerability across solid tumors with implications for developing improved anticancer therapy.
Collapse
Affiliation(s)
- Kuo-Sheng Hsu
- grid.48336.3a0000 0004 1936 8075Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, MD 21702 USA
| | - James M. Dunleavey
- grid.48336.3a0000 0004 1936 8075Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, MD 21702 USA
| | - Christopher Szot
- grid.48336.3a0000 0004 1936 8075Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, MD 21702 USA
| | - Liping Yang
- grid.48336.3a0000 0004 1936 8075Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, MD 21702 USA
| | - Mary Beth Hilton
- grid.48336.3a0000 0004 1936 8075Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, MD 21702 USA ,grid.418021.e0000 0004 0535 8394Basic Research Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research (FNLCR), Frederick, MD 21702 USA
| | - Karen Morris
- grid.48336.3a0000 0004 1936 8075Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, MD 21702 USA ,grid.418021.e0000 0004 0535 8394Basic Research Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research (FNLCR), Frederick, MD 21702 USA
| | - Steven Seaman
- grid.48336.3a0000 0004 1936 8075Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, MD 21702 USA
| | - Yang Feng
- grid.48336.3a0000 0004 1936 8075Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, MD 21702 USA
| | - Emily M. Lutz
- grid.48336.3a0000 0004 1936 8075Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, MD 21702 USA
| | - Robert Koogle
- grid.418021.e0000 0004 0535 8394MCGP, NCI, Frederick, MD 21702 USA
| | | | - Saurabh Saha
- BioMed Valley Discoveries, Inc, Kansas City, MO 64111 USA ,Present Address: Centessa Pharmaceuticals, Cambridge, MA 02139 USA
| | - Xiaoyan M. Zhang
- BioMed Valley Discoveries, Inc, Kansas City, MO 64111 USA ,Present Address: Ikena Oncology, Cambridge, MA 02210 USA
| | - Enrique Zudaire
- grid.48336.3a0000 0004 1936 8075Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, MD 21702 USA ,Present Address: Janssen Pharmaceutical Companies, J&J, R&D, Welsh Road McKean Road, Spring House, PA 19477 USA
| | - Pradip Bajgain
- grid.48336.3a0000 0004 1936 8075Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, MD 21702 USA
| | - Joshua Rose
- grid.48336.3a0000 0004 1936 8075Biomolecular Structure Section, Center for Structural Biology, NCI, NIH, Frederick, MD 21702 USA
| | - Zhongyu Zhu
- grid.48336.3a0000 0004 1936 8075Protein Interactions Section, Cancer and Inflammation Program, NCI, NIH, Frederick, MD 21702 USA ,grid.420872.bPresent Address: Lentigen Technology, Inc. 1201 Clopper Road, Gaithersburg, MD 20878 USA
| | - Dimiter S. Dimitrov
- grid.48336.3a0000 0004 1936 8075Protein Interactions Section, Cancer and Inflammation Program, NCI, NIH, Frederick, MD 21702 USA ,grid.21925.3d0000 0004 1936 9000Present Address: Center for Antibody Therapeutics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261 USA
| | - Frank Cuttitta
- grid.48336.3a0000 0004 1936 8075Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, MD 21702 USA
| | - Nancy J. Emenaker
- grid.48336.3a0000 0004 1936 8075Division of Cancer Prevention, NCI, NIH, Bethesda, MD 20892 USA
| | - Lino Tessarollo
- grid.48336.3a0000 0004 1936 8075Neural Development Section, MCGP, NCI, NIH, Frederick, MD 21702 USA
| | - Brad St. Croix
- grid.48336.3a0000 0004 1936 8075Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, MD 21702 USA
| |
Collapse
|
17
|
Hintzen KW, Simons C, Schaffrath K, Roessler G, Johnen S, Jakob F, Walter P, Schwaneberg U, Lohmann T. BioAdhere: tailor-made bioadhesives for epiretinal visual prostheses. Biomater Sci 2022; 10:3282-3295. [PMID: 35583519 DOI: 10.1039/d1bm01946e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Introduction: Visual prostheses, i.e. epiretinal stimulating arrays, are a promising therapy in treating retinal dystrophies and degenerations. In the wake of a new generation of devices, an innovative method for epiretinal fixation of stimulator arrays is required. We present the development of tailor-made bioadhesive peptides (peptesives) for fixating epiretinal stimulating arrays omitting the use of traumatic retinal tacks. Materials and methods: Binding motifs on the stimulating array (poly[chloro-p-xylylene] (Parylene C)) and in the extracellular matrix of the retinal surface (collagens I and IV, laminin, fibronectin) were identified. The anchor peptides cecropin A (CecA), KH1, KH2 (author's initials) and osteopontin (OPN) were genetically fused to reporter proteins to assess their binding behavior to coated microtiter plates via fluorescence-based assays. Domain Z (DZ) of staphylococcal protein A was used as a separator to generate a bioadhesive peptide. Following ISO 10993 "biological evaluation of medical materials", direct and non-direct cytotoxicity testing (L-929 and R28 retinal progenitor cells) was performed. Lastly, the fixating capabilities of the peptesives were tested in proof-of-principle experiments. Results: The generation of the bioadhesive peptide required evaluation of the N- and C-anchoring of investigated APs. The YmPh-CecA construct showed the highest activity on Parylene C in comparison with the wildtype phytase without the anchor peptide. eGFP-OPN was binding to all four investigated ECM proteins (collagen I, laminin > collagen IV, fibronectin). The strongest binding to collagen I was observed for eGFP-KH1, while the strongest binding to fibronectin was observed for eGFP-KH2. The selectivity of binding was checked by incubating eGFP-CecA and eGFP-OPN on ECM proteins and on Parylene C, respectively. Direct and non-direct cytotoxicity testing of the peptide cecropin-A-DZ-OPN using L-929 and R28 cells showed good biocompatibility properties. Proof-of-concept experiments in post-mortem rabbit eyes suggested an increased adhesion of CecA-DZ-OPN-coated stimulating arrays. Conclusion: This is the first study to prove the applicability and biocompatibility of peptesives for the fixation of macroscopic objects.
Collapse
Affiliation(s)
- Kai-Wolfgang Hintzen
- Institute of Biotechnology, RWTH Aachen University, Aachen, Germany.,DWI - Leibniz-Institute for Interactive Materials, Aachen, Germany
| | - Christian Simons
- DWI - Leibniz-Institute for Interactive Materials, Aachen, Germany
| | - Kim Schaffrath
- Department of Ophthalmology, RWTH Aachen University, Aachen, Germany.
| | - Gernot Roessler
- Department of Ophthalmology, RWTH Aachen University, Aachen, Germany.
| | - Sandra Johnen
- Department of Ophthalmology, RWTH Aachen University, Aachen, Germany.
| | - Felix Jakob
- Institute of Biotechnology, RWTH Aachen University, Aachen, Germany.,DWI - Leibniz-Institute for Interactive Materials, Aachen, Germany
| | - Peter Walter
- Department of Ophthalmology, RWTH Aachen University, Aachen, Germany.
| | - Ulrich Schwaneberg
- Institute of Biotechnology, RWTH Aachen University, Aachen, Germany.,DWI - Leibniz-Institute for Interactive Materials, Aachen, Germany
| | - Tibor Lohmann
- Department of Ophthalmology, RWTH Aachen University, Aachen, Germany.
| |
Collapse
|
18
|
Mesenchymal Stem-Cell Remodeling of Adsorbed Type-I Collagen-The Effect of Collagen Oxidation. Int J Mol Sci 2022; 23:ijms23063058. [PMID: 35328478 PMCID: PMC8953637 DOI: 10.3390/ijms23063058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/05/2022] [Accepted: 03/09/2022] [Indexed: 12/10/2022] Open
Abstract
This study describes the effect of collagen type I (Col I) oxidation on its physiological remodeling by adipose tissue-derived mesenchymal stem cells (ADMSCs), both mechanical and proteolytic, as an in vitro model for the acute oxidative stress that may occur in vivo upon distinct environmental changes. Morphologically, remodeling was interpreted as the mechanical rearrangement of adsorbed FITC-labelled Col I into a fibril-like pattern. This process was strongly abrogated in cells cultured on oxidized Col I albeit without visible changes in cell morphology. Proteolytic activity was quantified utilizing fluorescence de-quenching (FRET effect). The presence of ADMSCs caused a significant increase in native FITC-Col I fluorescence, which was almost absent in the oxidized samples. Parallel studies in a cell-free system confirmed the enzymatic de-quenching of native FITC-Col I by Clostridial collagenase with statistically significant inhibition occurring in the oxidized samples. Structural changes to the oxidized Col I were further studied by differential scanning calorimetry. In the oxidized samples, an additional endotherm with sustained enthalpy (∆H) was observed at 33.6 °C along with Col I’s typical one at 40.5 °C. Collectively, these data support that the remodeling of Col I by ADMSCs is altered upon oxidation due to intrinsic changes to the protein’s structure, which represents a novel mechanism for the control of stem cell behavior.
Collapse
|
19
|
Passive biaxial mechanical behavior of newborn mouse aorta with and without elastin. J Mech Behav Biomed Mater 2022; 126:105021. [PMID: 34864571 PMCID: PMC9808670 DOI: 10.1016/j.jmbbm.2021.105021] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 11/16/2021] [Accepted: 11/28/2021] [Indexed: 02/03/2023]
Abstract
Aortic wall material properties are needed for computational models and for comparisons across developmental and disease states. There has been abundant work in comparing aortic material properties across disease states, but limited work across developmental states. We performed passive biaxial mechanical testing on newborn mouse aorta with (Eln+/+) and without (Eln-/-) elastin. Elastin provides elasticity to the aortic wall and is necessary for survival beyond birth in the mouse. Mechanically functional elastin is challenging to create in vitro and so Eln-/- aorta can be a comparison for tissue engineered arteries with limited elastin amounts. We found that a traditional arterial strain energy function provided reasonable fits to newborn mouse aorta and generally predicted lower material constants in Eln-/- compared to Eln+/+ aorta. At physiologic pressures, the circumferential stresses and moduli trended lower in Eln-/- compared to Eln+/+ aorta. Increased blood pressure in Eln-/- mice helps to alleviate the differences in stresses and moduli. Increased blood pressure also serves to partially offload stresses in the isotropic compared to the anisotropic component of the wall. The baseline material parameters can be used in computational models of growth and remodeling to improve understanding of developmental mechanobiology and tissue engineering strategies.
Collapse
|
20
|
Sibole SC, Moo EK, Federico S, Herzog W. The Protective Function of Directed Asymmetry in the Pericellular Matrix Enveloping Chondrocytes. Ann Biomed Eng 2022; 50:39-55. [PMID: 34993700 DOI: 10.1007/s10439-021-02900-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 12/01/2021] [Indexed: 01/10/2023]
Abstract
The specialized pericellular matrix (PCM) surrounding chondrocytes within articular cartilage is critical to the tissue's health and longevity. Growing evidence suggests that PCM alterations are ubiquitous across all trajectories of osteoarthritis, a crippling and prevalent joint disease. The PCM geometry is of particular interest as it influences the cellular mechanical environment. Observations of asymmetrical PCM thickness have been reported, but a quantified characterization is lacking. To this end, a novel microscopy protocol was developed and applied to acquire images of the PCM surrounding live cells. Morphometric analysis indicated a statistical bias towards thicker PCM on the inferior cellular surface. The mechanical effects of this bias were investigated with multiscale modelling, which revealed potentially damaging, high tensile strains in the direction perpendicular to the membrane and localized on the inferior surface. These strains varied substantially between PCM asymmetry cases. Simulations with a thicker inferior PCM, representative of the observed geometry, resulted in strain magnitudes approximately half of those calculated for a symmetric geometry, and a third of those with a thin inferior PCM. This strain attenuation suggests that synthesis of a thicker inferior PCM may be a protective adaptation. PCM asymmetry may thus be important in cartilage development, pathology, and engineering.
Collapse
Affiliation(s)
- Scott C Sibole
- Human Performance Laboratory, University of Calgary, Calgary, Canada.
| | - Eng Kuan Moo
- Human Performance Laboratory, University of Calgary, Calgary, Canada.,Department of Applied Physics, University of Eastern Finland, Kuopio, Finland
| | - Salvatore Federico
- Human Performance Laboratory, University of Calgary, Calgary, Canada.,Department of Mechanical and Manufacturing Engineering, University of Calgary, Calgary, Canada
| | - Walter Herzog
- Human Performance Laboratory, University of Calgary, Calgary, Canada.,Department of Mechanical and Manufacturing Engineering, University of Calgary, Calgary, Canada
| |
Collapse
|
21
|
Bosworth LA, Lanaro M, O'Loughlin DA, D'Sa RA, Woodruff MA, Williams RL. Melt electro-written scaffolds with box-architecture support orthogonally oriented collagen. Biofabrication 2021; 14. [PMID: 34883476 DOI: 10.1088/1758-5090/ac41a1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 12/09/2021] [Indexed: 11/12/2022]
Abstract
Melt electro-writing (MEW) is a state-of-the-art technique that supports fabrication of 3D, precisely controlled and reproducible fiber structures. A standard MEW scaffold design is a box-structure, where a repeat layer of 90° boxes is produced from a single fiber. In 3D form (i.e. multiple layers), this structure has the potential to mimic orthogonal arrangements of collagen, as observed in the corneal stroma. In this study, we determined the response of human primary corneal stromal cells and their deposited fibrillar collagen (detected using a CNA35 probe) following six weeksin vitroculture on these box-structures made from poly(ϵ-caprolactone) (PCL). Comparison was also made to glass substrates (topography-free) and electrospun PCL fibers (aligned topography). Cell orientation and collagen deposition were non-uniform on glass substrates. Electrospun scaffolds supported an excellent parallel arrangement of cells and deposited collagen to the underlying architecture of aligned fibers, but there was no evidence of bidirectional collagen. In contrast, MEW scaffolds encouraged the formation of a dense, interconnected cellular network and deposited fibrillar collagen layers with a distinct orthogonal-arrangement. Collagen fibrils were particularly dominant through the middle layers of the MEW scaffolds' total thickness and closer examination revealed these fibrils to be concentrated within the pores' central regions. With the demand for donor corneas far exceeding the supply-leaving many with visual impairment-the application of MEW as a potential technique to recreate the corneal stroma with spontaneous, bidirectional collagen organization warrants further study.
Collapse
Affiliation(s)
- Lucy A Bosworth
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L7 8TX, United Kingdom
| | - Matthew Lanaro
- Science and Engineering Faculty, Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - Danielle A O'Loughlin
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L7 8TX, United Kingdom
| | - Raechelle A D'Sa
- Department of Mechanical, Materials and Aerospace Engineering, Faculty of Science and Engineering, University of Liverpool, Liverpool L69 3GH, United Kingdom
| | - Maria A Woodruff
- Science and Engineering Faculty, Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - Rachel L Williams
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L7 8TX, United Kingdom
| |
Collapse
|
22
|
Siadat SM, Silverman AA, Susilo ME, Paten JA, DiMarzio CA, Ruberti JW. Development of Fluorescently Labeled, Functional Type I Collagen Molecules. Macromol Biosci 2021; 22:e2100144. [PMID: 34856056 DOI: 10.1002/mabi.202100144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 10/22/2021] [Indexed: 11/11/2022]
Abstract
While de novo collagen fibril formation is well-studied, there are few investigations into the growth and remodeling of extant fibrils, where molecular collagen incorporation into and erosion from the fibril surface must delicately balance during fibril growth and remodeling. Observing molecule/fibril interactions is difficult, requiring the tracking of molecular dynamics while, at the same time, minimizing the effect of the observation on fibril structure and assembly. To address the observation-interference problem, exogenous collagen molecules are tagged with small fluorophores and the fibrillogenesis kinetics of labeled collagen molecules as well as the structure and network morphology of assembled fibrils are examined. While excessive labeling significantly disturbs fibrillogenesis kinetics and network morphology of assembled fibrils, adding less than ≈1.2 labels per collagen molecule preserves these characteristics. Applications of the functional, labeled collagen probe are demonstrated in both cellular and acellular systems. The functional, labeled collagen associates strongly with native fibrils and when added to an in vitro model of corneal stromal development at low concentration, the labeled collagen is incorporated into a fine extracellular matrix (ECM) network associated with the cells within 24 h.
Collapse
Affiliation(s)
| | | | - Monica E Susilo
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA
| | - Jeffrey A Paten
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02134, USA
| | - Charles A DiMarzio
- Department of Electrical and Computer Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Jeffrey W Ruberti
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA
| |
Collapse
|
23
|
Klontzas ME, Protonotarios A. High-Resolution Imaging for the Analysis and Reconstruction of 3D Microenvironments for Regenerative Medicine: An Application-Focused Review. Bioengineering (Basel) 2021; 8:182. [PMID: 34821748 PMCID: PMC8614770 DOI: 10.3390/bioengineering8110182] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/07/2021] [Accepted: 11/08/2021] [Indexed: 11/29/2022] Open
Abstract
The rapid evolution of regenerative medicine and its associated scientific fields, such as tissue engineering, has provided great promise for multiple applications where replacement and regeneration of damaged or lost tissue is required. In order to evaluate and optimise the tissue engineering techniques, visualisation of the material of interest is crucial. This includes monitoring of the cellular behaviour, extracellular matrix composition, scaffold structure, and other crucial elements of biomaterials. Non-invasive visualisation of artificial tissues is important at all stages of development and clinical translation. A variety of preclinical and clinical imaging methods-including confocal multiphoton microscopy, optical coherence tomography, magnetic resonance imaging (MRI), and computed tomography (CT)-have been used for the evaluation of artificial tissues. This review attempts to present the imaging methods available to assess the composition and quality of 3D microenvironments, as well as their integration with human tissues once implanted in the human body. The review provides tissue-specific application examples to demonstrate the applicability of such methods on cardiovascular, musculoskeletal, and neural tissue engineering.
Collapse
Affiliation(s)
- Michail E. Klontzas
- Department of Medical Imaging, University Hospital of Heraklion, 71110, Heraklion, Crete, Greece
- Computational Biomedicine Laboratory, Institute of Computer Science, Foundation for Research and Technology (FORTH), 70013 Heraklion, Crete, Greece
- Department of Radiology, School of Medicine, Voutes Campus, University of Crete, 71003 Heraklion, Crete, Greece
| | | |
Collapse
|
24
|
Katsuno-Kambe H, Teo JL, Ju RJ, Hudson J, Stehbens SJ, Yap AS. Collagen polarization promotes epithelial elongation by stimulating locoregional cell proliferation. eLife 2021; 10:e67915. [PMID: 34661524 PMCID: PMC8550756 DOI: 10.7554/elife.67915] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 10/13/2021] [Indexed: 12/21/2022] Open
Abstract
Epithelial networks are commonly generated by processes where multicellular aggregates elongate and branch. Here, we focus on understanding cellular mechanisms for elongation using an organotypic culture system as a model of mammary epithelial anlage. Isotropic cell aggregates broke symmetry and slowly elongated when transplanted into collagen 1 gels. The elongating regions of aggregates displayed enhanced cell proliferation that was necessary for elongation to occur. Strikingly, this locoregional increase in cell proliferation occurred where collagen 1 fibrils reorganized into bundles that were polarized with the elongating aggregates. Applying external stretch as a cell-independent way to reorganize the extracellular matrix, we found that collagen polarization stimulated regional cell proliferation to precipitate symmetry breaking and elongation. This required β1-integrin and ERK signaling. We propose that collagen polarization supports epithelial anlagen elongation by stimulating locoregional cell proliferation. This could provide a long-lasting structural memory of the initial axis that is generated when anlage break symmetry.
Collapse
Affiliation(s)
- Hiroko Katsuno-Kambe
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of QueenslandBrisbaneAustralia
| | - Jessica L Teo
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of QueenslandBrisbaneAustralia
| | - Robert J Ju
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of QueenslandBrisbaneAustralia
| | - James Hudson
- QIMR Berghofer Medical Research InstituteBrisbaneAustralia
| | - Samantha J Stehbens
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of QueenslandBrisbaneAustralia
| | - Alpha S Yap
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of QueenslandBrisbaneAustralia
| |
Collapse
|
25
|
Meerman M, Driessen R, van Engeland NCA, Bergsma I, Steenhuijsen JLG, Kozono D, Aikawa E, Hjortnaes J, Bouten CVC. Radiation Induces Valvular Interstitial Cell Calcific Response in an in vitro Model of Calcific Aortic Valve Disease. Front Cardiovasc Med 2021; 8:687885. [PMID: 34527708 PMCID: PMC8435633 DOI: 10.3389/fcvm.2021.687885] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 08/13/2021] [Indexed: 12/14/2022] Open
Abstract
Background: Mediastinal ionizing radiotherapy is associated with an increased risk of valvular disease, which demonstrates pathological hallmarks similar to calcific aortic valve disease (CAVD). Despite advances in radiotherapy techniques, the prevalence of comorbidities such as radiation-associated valvular disease is still increasing due to improved survival of patients receiving radiotherapy. However, the mechanisms of radiation-associated valvular disease are largely unknown. CAVD is considered to be an actively regulated disease process, mainly controlled by valvular interstitial cells (VICs). We hypothesize that radiation exposure catalyzes the calcific response of VICs and, therefore, contributes to the development of radiation-associated valvular disease. Methods and Results: To delineate the relationship between radiation and VIC behavior (morphology, calcification, and matrix turnover), two different in vitro models were established: (1) VICs were cultured two-dimensional (2D) on coverslips in control medium (CM) or osteogenic medium (OM) and irradiated with 0, 2, 4, 8, or 16 Gray (Gy); and (2) three-dimensional (3D) hydrogel system was designed, loaded with VICs and exposed to 0, 4, or 16 Gy of radiation. In both models, a dose-dependent decrease in cell viability and proliferation was observed in CM and OM. Radiation exposure caused myofibroblast-like morphological changes and differentiation of VICs, as characterized by decreased αSMA expression. Calcification, as defined by increased alkaline phosphatase activity, was mostly present in the 2D irradiated VICs exposed to 4 Gy, while after exposure to higher doses VICs acquired a unique giant fibroblast-like cell morphology. Finally, matrix turnover was significantly affected by radiation exposure in the 3D irradiated VICs, as shown by decreased collagen staining and increased MMP-2 and MMP-9 activity. Conclusions: The presented work demonstrates that radiation exposure enhances the calcific response in VICs, a hallmark of CAVD. In addition, high radiation exposure induces differentiation of VICs into a terminally differentiated giant-cell fibroblast. Further studies are essential to elucidate the underlying mechanisms of these radiation-induced valvular changes.
Collapse
Affiliation(s)
- Manon Meerman
- Department of Cardiothoracic Surgery, Heart and Lung Division, Leiden University Medical Center, Leiden, Netherlands
| | - Rob Driessen
- Department of Biomedical Engineering, Soft Tissue Engineering and Mechanobiology (STEM), Eindhoven University of Technology, Eindhoven, Netherlands.,Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, Netherlands
| | - Nicole C A van Engeland
- Department of Biomedical Engineering, Soft Tissue Engineering and Mechanobiology (STEM), Eindhoven University of Technology, Eindhoven, Netherlands.,Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, Netherlands.,Åbo Akademi University, Faculty of Science and Engineering, Molecular Biosciences, Turku, Finland
| | - Irith Bergsma
- Department of Biomedical Engineering, Soft Tissue Engineering and Mechanobiology (STEM), Eindhoven University of Technology, Eindhoven, Netherlands.,Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, Netherlands
| | | | - David Kozono
- Department of Radiation Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, United States
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Jesper Hjortnaes
- Department of Cardiothoracic Surgery, Heart and Lung Division, Leiden University Medical Center, Leiden, Netherlands
| | - Carlijn V C Bouten
- Department of Biomedical Engineering, Soft Tissue Engineering and Mechanobiology (STEM), Eindhoven University of Technology, Eindhoven, Netherlands.,Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, Netherlands
| |
Collapse
|
26
|
Abdolahpur Monikh F, Chupani L, Karkossa I, Gardian Z, Arenas-Lago D, von Bergen M, Schubert K, Piackova V, Zuskova E, Jiskoot W, Vijver MG, Peijnenburg WJGM. An environmental ecocorona influences the formation and evolution of the biological corona on the surface of single-walled carbon nanotubes. NANOIMPACT 2021; 22:100315. [PMID: 35559972 DOI: 10.1016/j.impact.2021.100315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/27/2021] [Accepted: 03/27/2021] [Indexed: 06/15/2023]
Abstract
Nanomaterials (NMs) taken up from the environment carry a complex ecocorona consisting of dissolved organic matter. An ecocorona is assumed to influence the interactions between NMs and endogenous biomolecules and consequently affects the formation of a biological corona (biocorona) and the biological fate of the NMs. This study shows that biomolecules in fish plasma attach immediately (within <5 min) to the surface of SWCNTs and the evolution of the biocorona is a size dependent phenomenon. Quantitative proteomics data revealed that the nanotube size also influences the plasma protein composition on the surface of SWCNTs. The presence of a pre-attached ecocorona on the surface of SWCNTs eliminated the influence of nanotube size on the formation and evolution of the biocorona. Over time, endogenous biomolecules from the plasma partially replaced the pre-attached ecocorona as measured using a fluorescently labelled ecocorona. The presence of an ecocorona offers a unique surface composition to each nanotube. This suggests that understanding the biological fate of NMs taken up from the environment by organisms to support the environmental risk assessment of NMs is a challenging task because each NM may have a unique surface composition in the body of an organism.
Collapse
Affiliation(s)
- Fazel Abdolahpur Monikh
- Institute of Environmental Sciences (CML), Leiden University, P.O. Box 9518, 2300, RA, Leiden, Netherlands; Department of Environmental & Biological Sciences, University of Eastern Finland, P.O. Box 111, FI-80101 Joensuu, Finland.
| | - Latifeh Chupani
- University of South Bohemia in Ceske Budejovice, Faculty of Fisheries and Protection of Waters, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Zátiší 728/II, 389 25 Vodňany, Czech Republic
| | - Isabel Karkossa
- Department of Molecular Systems Biology, UFZ, Helmholtz-Centre for Environmental Research, Permoserstraße 15, 04318 Leipzig, Germany
| | - Zdenko Gardian
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, University of South Bohemia, Faculty of Science, Branišovská 31, 370 05 České Budějovice, Czech Republic
| | - Daniel Arenas-Lago
- Department of Plant Biology and soil Science, University of Vigo, As Lagoas, Ourense, Spain
| | - Martin von Bergen
- Department of Molecular Systems Biology, UFZ, Helmholtz-Centre for Environmental Research, Permoserstraße 15, 04318 Leipzig, Germany; Institute of Biochemistry, Leipzig University, Permoserstraße 15, 04318 Leipzig, Germany
| | - Kristin Schubert
- Department of Molecular Systems Biology, UFZ, Helmholtz-Centre for Environmental Research, Permoserstraße 15, 04318 Leipzig, Germany
| | - Veronika Piackova
- University of South Bohemia in Ceske Budejovice, Faculty of Fisheries and Protection of Waters, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Zátiší 728/II, 389 25 Vodňany, Czech Republic
| | - Eliska Zuskova
- University of South Bohemia in Ceske Budejovice, Faculty of Fisheries and Protection of Waters, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Zátiší 728/II, 389 25 Vodňany, Czech Republic
| | - Wim Jiskoot
- Division of BioTherapeutics, Leiden University, Einsteinweg 55, 2333, CC, Leiden, the Netherlands
| | - Martina G Vijver
- Institute of Environmental Sciences (CML), Leiden University, P.O. Box 9518, 2300, RA, Leiden, Netherlands
| | - Willie J G M Peijnenburg
- Institute of Environmental Sciences (CML), Leiden University, P.O. Box 9518, 2300, RA, Leiden, Netherlands; National Institute of Public Health and the Environment (RIVM), Center for Safety of Substances and Products, Bilthoven, the Netherlands
| |
Collapse
|
27
|
Srivastava A, Tomar B, Prajapati S, Gaikwad AB, Mulay SR. Advanced non-invasive diagnostic techniques for visualization and estimation of kidney fibrosis. Drug Discov Today 2021; 26:2053-2063. [PMID: 33617976 DOI: 10.1016/j.drudis.2021.02.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/22/2020] [Accepted: 02/12/2021] [Indexed: 12/17/2022]
Abstract
Kidney fibrosis is marked by excessive extracellular matrix deposition during disease progression. Unfortunately, existing kidney function parameters do not predict the extent of kidney fibrosis. Moreover, the traditional histology methods for the assessment of kidney fibrosis require liquid and imaging biomarkers as well as needle-based biopsies, which are invasive and often associated with kidney injury. The repetitive analyses required to monitor the disease progression are therefore difficult. Hence, there is an unmet medical need for non-invasive and informative diagnostic approaches to monitor kidney fibrosis during the progression of chronic kidney disease. Here, we summarize the modern advances in diagnostic imaging techniques that have shown promise for non-invasive estimation of kidney fibrosis in pre-clinical and clinical studies.
Collapse
Affiliation(s)
- Anjali Srivastava
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Bhawna Tomar
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Smita Prajapati
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Anil Bhanudas Gaikwad
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, 333031, India
| | - Shrikant R Mulay
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031, India.
| |
Collapse
|
28
|
Siadat SM, Zamboulis DE, Thorpe CT, Ruberti JW, Connizzo BK. Tendon Extracellular Matrix Assembly, Maintenance and Dysregulation Throughout Life. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1348:45-103. [PMID: 34807415 DOI: 10.1007/978-3-030-80614-9_3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
In his Lissner Award medal lecture in 2000, Stephen Cowin asked the question: "How is a tissue built?" It is not a new question, but it remains as relevant today as it did when it was asked 20 years ago. In fact, research on the organization and development of tissue structure has been a primary focus of tendon and ligament research for over two centuries. The tendon extracellular matrix (ECM) is critical to overall tissue function; it gives the tissue its unique mechanical properties, exhibiting complex non-linear responses, viscoelasticity and flow mechanisms, excellent energy storage and fatigue resistance. This matrix also creates a unique microenvironment for resident cells, allowing cells to maintain their phenotype and translate mechanical and chemical signals into biological responses. Importantly, this architecture is constantly remodeled by local cell populations in response to changing biochemical (systemic and local disease or injury) and mechanical (exercise, disuse, and overuse) stimuli. Here, we review the current understanding of matrix remodeling throughout life, focusing on formation and assembly during the postnatal period, maintenance and homeostasis during adulthood, and changes to homeostasis in natural aging. We also discuss advances in model systems and novel tools for studying collagen and non-collagenous matrix remodeling throughout life, and finally conclude by identifying key questions that have yet to be answered.
Collapse
Affiliation(s)
| | - Danae E Zamboulis
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
| | - Chavaunne T Thorpe
- Comparative Biomedical Sciences, The Royal Veterinary College, University of London, London, UK
| | - Jeffrey W Ruberti
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Brianne K Connizzo
- Department of Biomedical Engineering, Boston University, Boston, MA, USA.
| |
Collapse
|
29
|
Aikawa E, Blaser MC. 2020 Jeffrey M. Hoeg Award Lecture: Calcifying Extracellular Vesicles as Building Blocks of Microcalcifications in Cardiovascular Disorders. Arterioscler Thromb Vasc Biol 2021; 41:117-127. [PMID: 33115271 PMCID: PMC7832175 DOI: 10.1161/atvbaha.120.314704] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 10/11/2020] [Indexed: 12/12/2022]
Abstract
Cardiovascular calcification is an insidious form of ectopic tissue mineralization that presents as a frequent comorbidity of atherosclerosis, aortic valve stenosis, diabetes, renal failure, and chronic inflammation. Calcification of the vasculature and heart valves contributes to mortality in these diseases. An inability to clinically image or detect early microcalcification coupled with an utter lack of pharmaceutical therapies capable of inhibiting or regressing entrenched and detectable macrocalcification has led to a prominent and deadly gap in care for a growing portion of our rapidly aging population. Recognition of this mounting concern has arisen over the past decade and led to a series of revolutionary works that has begun to pull back the curtain on the pathogenesis, mechanistic basis, and causative drivers of cardiovascular calcification. Central to this progress is the discovery that calcifying extracellular vesicles act as active precursors of cardiovascular microcalcification in diverse vascular beds. More recently, the omics revolution has resulted in the collection and quantification of vast amounts of molecular-level data. As the field has become poised to leverage these resources for drug discovery, new means of deriving relevant biological insights from these rich and complex datasets have come into focus through the careful application of systems biology and network medicine approaches. As we look onward toward the next decade, we envision a growing need to standardize approaches to study this complex and multifaceted clinical problem and expect that a push to translate mechanistic findings into therapeutics will begin to finally provide relief for those impacted by this disease.
Collapse
Affiliation(s)
- Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Center for Excellence in Vascular Biology, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Mark C. Blaser
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
30
|
Abstract
Chronic kidney disease is inevitably associated with the proliferation of fibroblasts following injury of the tubular system and collagen deposition in the interstitial tissue. To date, renal fibrosis has been hard to detect without histologic tissue examination. In this issue of Kidney International, Baues et al. introduce a novel technology for noninvasive detection of renal fibrosis using a multimodal optical approach with the fluorescent-labelled collagen-binding agent CNA35-Cy7.
Collapse
Affiliation(s)
- Julian Stumpf
- Division of Nephrology, Department of Internal Medicine III, Carl Gustav Carus University Hospital, Technical University Dresden, Dresden, Germany
| | - Christian Hugo
- Division of Nephrology, Department of Internal Medicine III, Carl Gustav Carus University Hospital, Technical University Dresden, Dresden, Germany.
| |
Collapse
|
31
|
Lee H, Kim SJ, Shin H, Kim YP. Collagen-Immobilized Extracellular FRET Reporter for Visualizing Protease Activity Secreted by Living Cells. ACS Sens 2020; 5:655-664. [PMID: 32036648 DOI: 10.1021/acssensors.9b01456] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Despite the diverse roles of cell-secreted proteases in the extracellular matrix (ECM), classical methods to analyze protease activity have not been explored at the cell culture site. Here, we report a stable, matrix-sticky, and protease-sensitive extracellular reporter that comprises a collagen-binding protein and a Förster resonance energy transfer (FRET) coupler of an enhanced green fluorescent protein and a small dye molecule. The extracellular FRET reporter via split intein-mediated protein trans-splicing is able to adhere to collagen matrices, leading to fluorescence changes by matrix metalloproteinase-2 (MMP2) activity during living cell culture without impeding cell viability. When a proMMP2 mutant (Y581A) with altered protease secretion and activity was transfected into cancer cells, the reporter revealed a dramatic reduction in MMP2 activity in both two- and three-dimensional culture systems, compared with cells transfected with wild-type proMMP2. Our reporter is immediately amenable to monitor protease activity in diverse ECM-resident cells as well as to study protease-related extracellular signaling and tissue remodeling.
Collapse
Affiliation(s)
- Hawon Lee
- Department of Life Science, BK21 Plus Bio-Defense Research Team, Hanyang University, Seoul 04763, Republic of Korea
| | - Se-jeong Kim
- Department of Bioengineering, BK21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, Seoul 04763, Republic of Korea
| | - Heungsoo Shin
- Department of Bioengineering, BK21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, Seoul 04763, Republic of Korea
- Institute of Nano Science and Technology, Hanyang University, Seoul 04763, Republic of Korea
| | - Young-Pil Kim
- Department of Life Science, BK21 Plus Bio-Defense Research Team, Hanyang University, Seoul 04763, Republic of Korea
- Institute of Nano Science and Technology, Hanyang University, Seoul 04763, Republic of Korea
- Research Institute for Natural Sciences and Research Institute for Convergence of Basic Sciences, Hanyang University, Seoul 04763, Republic of Korea
| |
Collapse
|
32
|
Emmert MY, Schmitt BA, Loerakker S, Sanders B, Spriestersbach H, Fioretta ES, Bruder L, Brakmann K, Motta SE, Lintas V, Dijkman PE, Frese L, Berger F, Baaijens FPT, Hoerstrup SP. Computational modeling guides tissue-engineered heart valve design for long-term in vivo performance in a translational sheep model. Sci Transl Med 2019; 10:10/440/eaan4587. [PMID: 29743347 DOI: 10.1126/scitranslmed.aan4587] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 09/20/2017] [Accepted: 04/09/2018] [Indexed: 12/14/2022]
Abstract
Valvular heart disease is a major cause of morbidity and mortality worldwide. Current heart valve prostheses have considerable clinical limitations due to their artificial, nonliving nature without regenerative capacity. To overcome these limitations, heart valve tissue engineering (TE) aiming to develop living, native-like heart valves with self-repair, remodeling, and regeneration capacity has been suggested as next-generation technology. A major roadblock to clinically relevant, safe, and robust TE solutions has been the high complexity and variability inherent to bioengineering approaches that rely on cell-driven tissue remodeling. For heart valve TE, this has limited long-term performance in vivo because of uncontrolled tissue remodeling phenomena, such as valve leaflet shortening, which often translates into valve failure regardless of the bioengineering methodology used to develop the implant. We tested the hypothesis that integration of a computationally inspired heart valve design into our TE methodologies could guide tissue remodeling toward long-term functionality in tissue-engineered heart valves (TEHVs). In a clinically and regulatory relevant sheep model, TEHVs implanted as pulmonary valve replacements using minimally invasive techniques were monitored for 1 year via multimodal in vivo imaging and comprehensive tissue remodeling assessments. TEHVs exhibited good preserved long-term in vivo performance and remodeling comparable to native heart valves, as predicted by and consistent with computational modeling. TEHV failure could be predicted for nonphysiological pressure loading. Beyond previous studies, this work suggests the relevance of an integrated in silico, in vitro, and in vivo bioengineering approach as a basis for the safe and efficient clinical translation of TEHVs.
Collapse
Affiliation(s)
- Maximilian Y Emmert
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland.,University Heart Center Zurich, University Hospital Zurich, Zurich, Switzerland.,Wyss Translational Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Boris A Schmitt
- German Heart Center Berlin and Charité-Universitätsmedizin Berlin, Department of Congenital Heart Disease, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Sandra Loerakker
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands.,Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Bart Sanders
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands.,Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Hendrik Spriestersbach
- German Heart Center Berlin and Charité-Universitätsmedizin Berlin, Department of Congenital Heart Disease, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Emanuela S Fioretta
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Leon Bruder
- German Heart Center Berlin and Charité-Universitätsmedizin Berlin, Department of Congenital Heart Disease, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Kerstin Brakmann
- German Heart Center Berlin and Charité-Universitätsmedizin Berlin, Department of Congenital Heart Disease, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Sarah E Motta
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Valentina Lintas
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Petra E Dijkman
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Laura Frese
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Felix Berger
- German Heart Center Berlin and Charité-Universitätsmedizin Berlin, Department of Congenital Heart Disease, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Frank P T Baaijens
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands.,Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Simon P Hoerstrup
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland. .,Wyss Translational Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| |
Collapse
|
33
|
A collagen-binding protein enables molecular imaging of kidney fibrosis in vivo. Kidney Int 2019; 97:609-614. [PMID: 31784048 DOI: 10.1016/j.kint.2019.08.029] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 08/02/2019] [Accepted: 08/22/2019] [Indexed: 12/13/2022]
Abstract
Pathological deposition of collagen is a hallmark of kidney fibrosis. To illustrate this process we employed multimodal optical imaging to visualize and quantify collagen deposition in murine models of kidney fibrosis (ischemia-reperfusion or unilateral ureteral obstruction) using the collagen-binding adhesion protein CNA35. For in vivo imaging, we used hybrid computed tomography-fluorescence molecular tomography and CNA35 labeled with the near-infrared fluorophore Cy7. Upon intravenous injection, CNA35-Cy7 accumulation was significantly higher in fibrotic compared to non-fibrotic kidneys. This difference was not detected for a non-specific scrambled version of CNA35-Cy7. Ex vivo, on kidney sections of mice and patients with renal fibrosis, CNA35-FITC co-localized with fibrotic collagen type I and III, but not with the basement membrane collagen type IV. Following intravenous injection, CNA35-FITC bound to both interstitial and perivascular fibrotic areas. In line with this perivascular accumulation, we observed significant perivascular fibrosis in the mouse models and in biopsy sections from patients with chronic kidney disease using computer-based morphometry quantification. Thus, molecular imaging of collagen using CNA35 enabled specific non-invasive quantification of kidney fibrosis. Collagen imaging revealed significant perivascular fibrosis as a consistent component next to the more commonly assessed interstitial fibrosis. Our results lay the basis for further probe and protocol optimization towards the clinical translation of molecular imaging of kidney fibrosis.
Collapse
|
34
|
Zhou Q, Zeng Y, Xiong Q, Zhong S, Li P, Ran H, Yin Y, Reutelingsperger C, Prinze FW, Ling Z. Construction of CNA35 Collagen-Targeted Phase-Changeable Nanoagents for Low-Intensity Focused Ultrasound-Triggered Ultrasound Molecular Imaging of Myocardial Fibrosis in Rabbits. ACS APPLIED MATERIALS & INTERFACES 2019; 11:23006-23017. [PMID: 31136144 DOI: 10.1021/acsami.9b05999] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Myocardial fibrosis plays an important role in the development of heart failure and malignant arrhythmia, which potentially increases the incidence of sudden cardiac death. Therefore, early detection of myocardial fibrosis is of great significance for evaluating the prognosis of patients and formulating appropriate treatment strategies. Late gadolinium-enhanced magnetic resonance imaging is considered as the currently effective strategy for noninvasive detection of myocardial fibrosis, but it still suffers from some critical issues. In this work, multifunctional CNA35-labeled perfluoropentane nanoparticles (CNA35-PFP NPs) have been elaborately designed and constructed for molecular imaging of fibrotic myocardium based on ultrasound imaging. These as-constructed CNA35-PFP NPs are intravenously infused into rabbit circulation with an animal model of myocardial infarction. Especially, these targeted CNA35-PFP NPs with nanoscale size could efficiently pass through the endothelial cell gap and adhere to the surface of fibroblasts in the fibrotic myocardium. Importantly, followed by low-intensity focused ultrasound irradiation on the myocardium, these intriguing CNA35-PFP NPs could transform from liquid into gaseous microbubbles, which further significantly enhanced the ultrasound contrast in the fibrotic area, facilitating the detection by diagnostic ultrasound imaging. Therefore, this work provides a desirable noninvasive, economical, and real-time imaging technique for the assessment of cardiac fibrosis with diagnostic ultrasound based on the rational design of liquid-to-gas phase-changeable nanoplatforms.
Collapse
Affiliation(s)
- Qin Zhou
- Department of Cardiology , Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Yalin Zeng
- Department of Cardiology , Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Qingsong Xiong
- Department of Cardiology , Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Shigen Zhong
- Institute of Ultrasound Imaging, Chongqing Key Laboratory of Ultrasound Molecular Imaging , The Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Pan Li
- Institute of Ultrasound Imaging, Chongqing Key Laboratory of Ultrasound Molecular Imaging , The Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Haitao Ran
- Institute of Ultrasound Imaging, Chongqing Key Laboratory of Ultrasound Molecular Imaging , The Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Yuehui Yin
- Department of Cardiology , Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Chris Reutelingsperger
- Department of Physiology, Cardiovascular Research Institute Maastricht , Maastricht University , P.O. Box 616 , 6200 MD , Maastricht , The Netherlands
| | - Frits W Prinze
- Department of Biochemistry, Cardiovascular Research Institute Maastricht , University of Maastricht , P.O. Box 616 , 6200 MD , Maastricht , The Netherlands
| | - Zhiyu Ling
- Department of Cardiology , Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
- Institute of Ultrasound Imaging, Chongqing Key Laboratory of Ultrasound Molecular Imaging , The Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| |
Collapse
|
35
|
Tan K, Keegan P, Rogers M, Lu M, Gosset JR, Charest J, Bale SS. A high-throughput microfluidic microphysiological system (PREDICT-96) to recapitulate hepatocyte function in dynamic, re-circulating flow conditions. LAB ON A CHIP 2019; 19:1556-1566. [PMID: 30855604 DOI: 10.1039/c8lc01262h] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Microphysiological systems (MPSs) are dynamic cell culture systems that provide micro-environmental and external cues to support physiologically relevant, organ-specific functions. Recent progresses in MPS fabrication technologies have enabled the development of advanced models to capture microenvironments with physiological relevance, while increasing throughput and reducing material-based artefacts. In addition to conventional cell culture systems, advanced MPSs are emerging as ideal contenders for disease modeling and incorporation into drug screening. Since liver is a central organ for drug metabolism, liver-on-chip models have been developed to recapitulate hepatic microenvironment with varying complexities, while allowing long-term culture. Recently, we have developed a novel thermoplastic, oxygen-permeable MPS for primary human hepatocyte (PHH) culture. Herein, we have adapted and extended the MPS to a) a 96 microfluidic array (PREDICT-96 array) and b) integrated a novel, ultra-low volume, re-circulating pumping system (PREDICT-96 pump) - collectively known as the PREDICT-96 platform. The PREDICT-96 platform conforms to the industrial standard 96-well footprint and enables media re-circulation. First, we demonstrate the introduction of PHHs into the PREDICT-96 array using standard handling procedures for multi-well plates and allow cells to stabilize in static conditions. Next, we introduce recirculating flow into the bottom channel (using PREDICT-96 pump) to mimic mass transport in vivo. Our results indicate an increase in metabolic and secretory functions of PHHs in the PREDICT-96 platform, and their maintenance over 10 days of flow. Furthermore, long-term culture with fluid flow allows for the periodic introduction of media components (e.g., fatty acids, cytokines) and capture cellular responses to chronic stimuli. The low-volume footprint of the pump and small media volume in the MPS allow for the interrogation of hepatic responses incorporating secretion feedback to a stimulus, which is essential for disease model development and drug interrogation. We envision future development of this liver model to incorporate key primary hepatic cells, multi-cellular co-cultures and adaptation, integration with high-throughput analytical tools.
Collapse
Affiliation(s)
- Kelly Tan
- Draper, 555 Technology Square, Cambridge, MA 02138, USA.
| | | | | | | | | | | | | |
Collapse
|
36
|
Development and characterization of Lyophilized Transparized Decellularized stroma as a replacement for living cornea in deep anterior lamellar keratoplasty. Cell Tissue Bank 2019; 20:49-59. [DOI: 10.1007/s10561-018-9742-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 12/01/2018] [Indexed: 11/27/2022]
|
37
|
Sato K, Matsubara O, Hase E, Minamikawa T, Yasui T. Quantitative in situ time-series evaluation of osteoblastic collagen synthesis under cyclic strain using second-harmonic-generation microscopy. JOURNAL OF BIOMEDICAL OPTICS 2019; 24:1-8. [PMID: 30635995 PMCID: PMC6975189 DOI: 10.1117/1.jbo.24.3.031019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 12/11/2018] [Indexed: 06/09/2023]
Abstract
The aim of this study is to evaluate the osteoblastic collagen synthesis under mechanical stimulation using second-harmonic-generation (SHG) microscopy. We apply SHG microscopy to monitor the collagen fibers synthesized by osteoblast-like cells (MC3T3-E1) without the need for fixation and staining. To quantitatively evaluate the influence of mechanical stimulation on osteoblastic collagen synthesis, we compare SHG images of osteoblast-synthesized collagen fibers with and without a cyclic stretch stimulus applied using a lab-made stretching device. We acquire SHG images every 7 days for 3 weeks at different stimulus conditions (5 min/day and 3 h/day with a strain magnitude of 5% and a frequency of 0.5 Hz). Image analysis of the average SHG intensity indicates that the amount of osteoblastic collagen synthesis is significantly enhanced by the cyclic stretch compared with the nonstretched condition, while there is no significant difference between the two mechanical stimulation conditions. Furthermore, the maturity of the collagen fibers in the early stage of bone formation is not affected by the mechanical stimulation. The results can be used in bone regenerative medicine to apply feedback control of collagen synthesis by artificial stimulation.
Collapse
Affiliation(s)
- Katsuya Sato
- Tokushima University, Graduate School of Technology, Industrial and Social Sciences, Tokushima City, Tokushima, Japan
| | - Oki Matsubara
- Tokushima University, Graduate School of Technology, Industrial and Social Sciences, Tokushima City, Tokushima, Japan
| | - Eiji Hase
- Japan Synchrotron Radiation Research Institute, Research and Utilization Division, Sayo, Hyogo, Japan
| | - Takeo Minamikawa
- Tokushima University, Graduate School of Technology, Industrial and Social Sciences, Tokushima City, Tokushima, Japan
| | - Takeshi Yasui
- Tokushima University, Graduate School of Technology, Industrial and Social Sciences, Tokushima City, Tokushima, Japan
| |
Collapse
|
38
|
Staiculescu MC, Cocciolone AJ, Procknow JD, Kim J, Wagenseil JE. Comparative gene array analyses of severe elastic fiber defects in late embryonic and newborn mouse aorta. Physiol Genomics 2018; 50:988-1001. [PMID: 30312140 PMCID: PMC6293116 DOI: 10.1152/physiolgenomics.00080.2018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 10/09/2018] [Accepted: 10/09/2018] [Indexed: 01/17/2023] Open
Abstract
Elastic fibers provide reversible elasticity to the large arteries and are assembled during development when hemodynamic forces are increasing. Mutations in elastic fiber genes are associated with cardiovascular disease. Mice lacking expression of the elastic fiber genes elastin ( Eln-/-), fibulin-4 ( Efemp2-/-), or lysyl oxidase ( Lox-/-) die at birth with severe cardiovascular malformations. All three genetic knockout models have elastic fiber defects, aortic wall thickening, and arterial tortuosity. However, Eln-/- mice develop arterial stenoses, while Efemp2-/- and Lox-/- mice develop ascending aortic aneurysms. We performed comparative gene array analyses of these three genetic models for two vascular locations and developmental stages to determine differentially expressed genes and pathways that may explain the common and divergent phenotypes. We first examined arterial morphology and wall structure in newborn mice to confirm that the lack of elastin, fibulin-4, or lysyl oxidase expression provided the expected phenotypes. We then compared gene expression levels for each genetic model by three-way ANOVA for genotype, vascular location, and developmental stage. We found three genes upregulated by genotype in all three models, Col8a1, Igfbp2, and Thbs1, indicative of a common response to severe elastic fiber defects in developing mouse aorta. Genes that are differentially regulated by vascular location or developmental stage in all three models suggest mechanisms for location or stage-specific disease pathology. Comparison of signaling pathways enriched in all three models shows upregulation of integrins and matrix proteins involved in early wound healing, but not of mature matrix molecules such as elastic fiber proteins or fibrillar collagens.
Collapse
Affiliation(s)
| | - Austin J Cocciolone
- Department of Biomedical Engineering, Washington University , St. Louis, Missouri
| | - Jesse D Procknow
- Department of Mechanical Engineering and Materials Science, Washington University , St. Louis, Missouri
| | - Jungsil Kim
- Department of Mechanical Engineering and Materials Science, Washington University , St. Louis, Missouri
| | - Jessica E Wagenseil
- Department of Mechanical Engineering and Materials Science, Washington University , St. Louis, Missouri
| |
Collapse
|
39
|
Burns DP, Canavan L, Rowland J, O'Flaherty R, Brannock M, Drummond SE, O'Malley D, Edge D, O'Halloran KD. Recovery of respiratory function in mdx mice co-treated with neutralizing interleukin-6 receptor antibodies and urocortin-2. J Physiol 2018; 596:5175-5197. [PMID: 30160301 PMCID: PMC6209753 DOI: 10.1113/jp276954] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Accepted: 08/22/2018] [Indexed: 12/17/2022] Open
Abstract
KEY POINTS Impaired ventilatory capacity and diaphragm muscle weakness are prominent features of Duchenne muscular dystrophy, with strong evidence of attendant systemic and muscle inflammation. We performed a 2-week intervention in young wild-type and mdx mice, consisting of either injection of saline or co-administration of a neutralizing interleukin-6 receptor antibody (xIL-6R) and urocortin-2 (Ucn2), a corticotrophin releasing factor receptor 2 agonist. We examined breathing and diaphragm muscle form and function. Breathing and diaphragm muscle functional deficits are improved following xIL-6R and Ucn2 co-treatment in mdx mice. The functional improvements were associated with a preservation of mdx diaphragm muscle myosin heavy chain IIx fibre complement. The concentration of the pro-inflammatory cytokine interleukin-1β was reduced and the concentration of the anti-inflammatory cytokine interleukin-10 was increased in mdx diaphragm following drug co-treatment. Our novel findings may have implications for the development of pharmacotherapies for the dystrophinopathies with relevance for respiratory muscle performance and breathing. ABSTRACT The mdx mouse model of Duchenne muscular dystrophy shows evidence of hypoventilation and pronounced diaphragm dysfunction. Six-week-old male mdx (n = 32) and wild-type (WT; n = 32) mice received either saline (0.9% w/v) or a co-administration of neutralizing interleukin-6 receptor antibodies (xIL-6R; 0.2 mg kg-1 ) and corticotrophin-releasing factor receptor 2 agonist (urocortin-2; 30 μg kg-1 ) subcutaneously over 2 weeks. Breathing and diaphragm muscle contractile function (ex vivo) were examined. Diaphragm structure was assessed using histology and immunofluorescence. Muscle cytokine concentration was determined using a multiplex assay. Minute ventilation and diaphragm muscle peak force at 100 Hz were significantly depressed in mdx compared with WT. Drug treatment completely restored ventilation in mdx mice during normoxia and significantly increased mdx diaphragm force- and power-generating capacity. The number of centrally nucleated muscle fibres and the areal density of infiltrates and collagen content were significantly increased in mdx diaphragm; all indices were unaffected by drug co-treatment. The abundance of myosin heavy chain (MyHC) type IIx fibres was significantly decreased in mdx diaphragm; drug co-treatment preserved MyHC type IIx complement in mdx muscle. Drug co-treatment increased the cross-sectional area of MyHC type I and IIx fibres in mdx diaphragm. The cytokines IL-1β, IL-6, KC/GRO and TNF-α were significantly increased in mdx diaphragm compared with WT. Drug co-treatment significantly decreased IL-1β and increased IL-10 in mdx diaphragm. Drug co-treatment had no significant effect on WT diaphragm muscle structure, cytokine concentrations or function. Recovery of breathing and diaphragm force in mdx mice was impressive in our studies, with implication for human dystrophinopathies.
Collapse
Affiliation(s)
- David P. Burns
- Department of PhysiologySchool of MedicineCollege of Medicine and HealthUniversity College CorkCorkIreland
| | - Leonie Canavan
- Department of PhysiologySchool of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, the University of DublinDublinIreland
| | - Jane Rowland
- Department of PhysiologySchool of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, the University of DublinDublinIreland
| | - Robin O'Flaherty
- Department of PhysiologySchool of MedicineCollege of Medicine and HealthUniversity College CorkCorkIreland
| | - Molly Brannock
- Department of PhysiologySchool of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, the University of DublinDublinIreland
| | - Sarah E. Drummond
- Department of PhysiologySchool of MedicineCollege of Medicine and HealthUniversity College CorkCorkIreland
| | - Dervla O'Malley
- Department of PhysiologySchool of MedicineCollege of Medicine and HealthUniversity College CorkCorkIreland
| | - Deirdre Edge
- Department of PhysiologySchool of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, the University of DublinDublinIreland
| | - Ken D. O'Halloran
- Department of PhysiologySchool of MedicineCollege of Medicine and HealthUniversity College CorkCorkIreland
| |
Collapse
|
40
|
Maacha S, Hong J, von Lersner A, Zijlstra A, Belkhiri A. AXL Mediates Esophageal Adenocarcinoma Cell Invasion through Regulation of Extracellular Acidification and Lysosome Trafficking. Neoplasia 2018; 20:1008-1022. [PMID: 30189359 PMCID: PMC6126204 DOI: 10.1016/j.neo.2018.08.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 08/06/2018] [Accepted: 08/14/2018] [Indexed: 12/14/2022]
Abstract
Esophageal adenocarcinoma (EAC) is a highly aggressive malignancy that is characterized by resistance to chemotherapy and a poor clinical outcome. The overexpression of the receptor tyrosine kinase AXL is frequently associated with unfavorable prognosis in EAC. Although it is well documented that AXL mediates cancer cell invasion as a downstream effector of epithelial-to-mesenchymal transition, the precise molecular mechanism underlying this process is not completely understood. Herein, we demonstrate for the first time that AXL mediates cell invasion through the regulation of lysosomes peripheral distribution and cathepsin B secretion in EAC cell lines. Furthermore, we show that AXL-dependent peripheral distribution of lysosomes and cell invasion are mediated by extracellular acidification, which is potentiated by AXL-induced secretion of lactate through AKT-NF-κB-dependent MCT-1 regulation. Our novel mechanistic findings support future clinical studies to evaluate the therapeutic potential of the AXL inhibitor R428 (BGB324) in highly invasive EAC.
Collapse
Affiliation(s)
- Selma Maacha
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jun Hong
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Ariana von Lersner
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN 37240, USA
| | - Andries Zijlstra
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN 37240, USA
| | - Abbes Belkhiri
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
41
|
Werner M, Kurniawan NA, Korus G, Bouten CVC, Petersen A. Mesoscale substrate curvature overrules nanoscale contact guidance to direct bone marrow stromal cell migration. J R Soc Interface 2018; 15:20180162. [PMID: 30089684 PMCID: PMC6127159 DOI: 10.1098/rsif.2018.0162] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 07/13/2018] [Indexed: 12/19/2022] Open
Abstract
The intrinsic architecture of biological tissues and of implanted biomaterials provides cells with large-scale geometrical cues. To understand how cells are able to sense and respond to complex structural environments, a deeper insight into the cellular response to multi-scale and conflicting geometrical cues is needed. In this study, we subjected human bone marrow stromal cells (hBMSCs) to mesoscale cylindrical surfaces (diameter 250-5000 µm) and nanoscale collagen fibrils (diameter 100-200 nm) that were aligned perpendicular to the cylinder axis. On flat surfaces and at low substrate curvatures (cylinder diameter d > 1000 µm), cell alignment and migration were governed by the nanoscale collagen fibrils, consistent with the contact guidance effect. With increasing surface curvature (decreasing cylinder diameter, d < 1000 µm), cells increasingly aligned and migrated along the cylinder axis, i.e. the direction of zero curvature. An increase in phosphorylated myosin light chain levels was observed with increasing substrate curvature, suggesting a link between substrate-induced cell bending and the F-actin-myosin machinery. Taken together, this work demonstrates that geometrical cues of up to 10× cell size can play a dominant role in directing hBMSC alignment and migration and that the effect of nanoscale contact guidance can even be overruled by mesoscale curvature guidance.
Collapse
Affiliation(s)
- Maike Werner
- Soft Tissue Engineering and Mechanobiology, Department of Biomedical Engineering, Eindhoven University of Technology, The Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, The Netherlands
| | - Nicholas A Kurniawan
- Soft Tissue Engineering and Mechanobiology, Department of Biomedical Engineering, Eindhoven University of Technology, The Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, The Netherlands
| | - Gabriela Korus
- Julius Wolff Institute, Charité-Universitätsmedizin Berlin, Germany
| | - Carlijn V C Bouten
- Soft Tissue Engineering and Mechanobiology, Department of Biomedical Engineering, Eindhoven University of Technology, The Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, The Netherlands
| | - Ansgar Petersen
- Julius Wolff Institute, Charité-Universitätsmedizin Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Germany
| |
Collapse
|
42
|
Espinosa MG, Taber LA, Wagenseil JE. Reduced embryonic blood flow impacts extracellular matrix deposition in the maturing aorta. Dev Dyn 2018; 247:914-923. [PMID: 29696727 DOI: 10.1002/dvdy.24635] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 04/15/2018] [Accepted: 04/18/2018] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Perturbations to embryonic hemodynamics are known to adversely affect cardiovascular development. Vitelline vein ligation (VVL) is a model of reduced placental blood flow used to induce cardiac defects in early chick embryo development. The effect of these hemodynamic interventions on maturing elastic arteries is largely unknown. We hypothesize that hemodynamic changes impact maturation of the dorsal aorta (DA). RESULTS We examined the effects of VVL on hemodynamic properties well into the maturation process and the corresponding changes in aortic dimensions, wall composition, and gene expression. In chick embryos, we found that DA blood velocity was reduced immediately postsurgery at Hamburger-Hamilton (HH) stage 18 and later at HH36, but not in the interim. Throughout this period, DA diameter adapted to maintain a constant shear stress. At HH36, we found that VVL DAs showed a substantial decrease in elastin and a modest increase in collagen protein content. In VVL DAs, up-regulation of elastic fiber-related genes followed the down-regulation of flow-dependent genes. Together, these suggest the existence of a compensatory mechanism in response to shear-induced delays in maturation. CONCLUSIONS The DA's response to hemodynamic perturbations invokes coupled mechanisms for shear regulation and matrix maturation, potentially impacting the course of vascular development. Developmental Dynamics 247:914-923, 2018. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- M Gabriela Espinosa
- Department of Biomedical Engineering, Washington University, Saint Louis, Missouri
| | - Larry A Taber
- Department of Biomedical Engineering, Washington University, Saint Louis, Missouri
| | - Jessica E Wagenseil
- Department of Mechanical Engineering and Materials Science, Washington University, Saint Louis, Missouri
| |
Collapse
|
43
|
Fleszar AJ, Walker A, Porubsky V, Flanigan W, James D, Campagnola PJ, Weisman PS, Kreeger PK. The Extracellular Matrix of Ovarian Cortical Inclusion Cysts Modulates Invasion of Fallopian Tube Epithelial Cells. APL Bioeng 2018; 2:031902. [PMID: 30556046 PMCID: PMC6294138 DOI: 10.1063/1.5022595] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 03/27/2018] [Indexed: 12/26/2022] Open
Abstract
A growing body of research supports the idea that the fallopian tube epithelium (FTE) is the precursor for most high-grade serous ovarian canacers (HGSOC) but that the ovary plays a critical role in tumor metastasis. Cortical inclusion cysts (CICs) in the ovarian cortex have been hypothesized to create a niche environment that plays a role in HGSOC progression. Through histological analysis of pathology samples from human ovaries, we determined that collagen I and III were elevated near CICs and that the collagen fibers in this dense region were oriented parallel to the cyst boundary. Using this information from human samples as design parameters, we engineered an in vitro model that recreates the size, shape, and extracellular matrix (ECM) properties of CICs. We found that FTE cells within our model underwent robust invasion that was responsive to stimulation with follicular fluid, while ovarian surface epithelial (OSE) cells, the native cells of the ovary, were not invasive. We provide experimental evidence to support a role of the extracellular matrix in modulating FTE cell invasion, as decreased collagen I concentration or the addition of collagen III to the matrix surrounding FTE cells increased FTE cell invasion. Taken together, we show that an in vitro model of CICs informed by the analysis of human tissue can act as an important tool for understanding FTE cell interactions with their environment.
Collapse
Affiliation(s)
- Andrew J. Fleszar
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| | - Alyssa Walker
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| | - Veronica Porubsky
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| | - Will Flanigan
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| | - Darian James
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| | | | - Paul S. Weisman
- Department of Pathology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53705, USA
| | | |
Collapse
|
44
|
Arnold Egloff SA, Du L, Loomans HA, Starchenko A, Su PF, Ketova T, Knoll PB, Wang J, Haddad AQ, Fadare O, Cates JM, Lotan Y, Shyr Y, Clark PE, Zijlstra A. Shed urinary ALCAM is an independent prognostic biomarker of three-year overall survival after cystectomy in patients with bladder cancer. Oncotarget 2018; 8:722-741. [PMID: 27894096 PMCID: PMC5352192 DOI: 10.18632/oncotarget.13546] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 10/19/2016] [Indexed: 01/08/2023] Open
Abstract
Proteins involved in tumor cell migration can potentially serve as markers of invasive disease. Activated Leukocyte Cell Adhesion Molecule (ALCAM) promotes adhesion, while shedding of its extracellular domain is associated with migration. We hypothesized that shed ALCAM in biofluids could be predictive of progressive disease. ALCAM expression in tumor (n = 198) and shedding in biofluids (n = 120) were measured in two separate VUMC bladder cancer cystectomy cohorts by immunofluorescence and enzyme-linked immunosorbent assay, respectively. The primary outcome measure was accuracy of predicting 3-year overall survival (OS) with shed ALCAM compared to standard clinical indicators alone, assessed by multivariable Cox regression and concordance-indices. Validation was performed by internal bootstrap, a cohort from a second institution (n = 64), and treatment of missing data with multiple-imputation. While ALCAM mRNA expression was unchanged, histological detection of ALCAM decreased with increasing stage (P = 0.004). Importantly, urine ALCAM was elevated 17.0-fold (P < 0.0001) above non-cancer controls, correlated positively with tumor stage (P = 0.018), was an independent predictor of OS after adjusting for age, tumor stage, lymph-node status, and hematuria (HR, 1.46; 95% CI, 1.03–2.06; P = 0.002), and improved prediction of OS by 3.3% (concordance-index, 78.5% vs. 75.2%). Urine ALCAM remained an independent predictor of OS after accounting for treatment with Bacillus Calmette-Guerin, carcinoma in situ, lymph-node dissection, lymphovascular invasion, urine creatinine, and adjuvant chemotherapy (HR, 1.10; 95% CI, 1.02–1.19; P = 0.011). In conclusion, shed ALCAM may be a novel prognostic biomarker in bladder cancer, although prospective validation studies are warranted. These findings demonstrate that markers reporting on cell motility can act as prognostic indicators.
Collapse
Affiliation(s)
- Shanna A Arnold Egloff
- Department of Veterans Affairs, Nashville, TN, USA.,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Liping Du
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Holli A Loomans
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alina Starchenko
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Pei-Fang Su
- Department of Statistics, National Cheng Kung University, Taiwan
| | - Tatiana Ketova
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Jifeng Wang
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Urology, The Fifth People's Hospital of Shanghai, Shanghai, China
| | - Ahmed Q Haddad
- Department of Urology, The University of Louisville, Louisville, KY, USA.,Department of Urology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Oluwole Fadare
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA.,University of California San Diego, La Jolla, CA, USA
| | - Justin M Cates
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Yair Lotan
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Yu Shyr
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN, USA.,Vanderbilt Ingram-Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Peter E Clark
- Vanderbilt Ingram-Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Urology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Andries Zijlstra
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA.,Vanderbilt Ingram-Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
45
|
Staiculescu MC, Kim J, Mecham RP, Wagenseil JE. Mechanical behavior and matrisome gene expression in the aneurysm-prone thoracic aorta of newborn lysyl oxidase knockout mice. Am J Physiol Heart Circ Physiol 2017; 313:H446-H456. [PMID: 28550176 PMCID: PMC5582924 DOI: 10.1152/ajpheart.00712.2016] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 05/12/2017] [Accepted: 05/23/2017] [Indexed: 12/14/2022]
Abstract
Mutations in lysyl oxidase (LOX) are associated with thoracic aortic aneurysm and dissection (TAAD). Mice that do not express Lox (Lox-/- ) die soon after birth and have 60% and 40% reductions in elastin- and collagen-specific cross-links, respectively. LOX inactivation could also change the expression of secreted factors, the structural matrix, and matrix-associated proteins that constitute the aortic matrisome. We hypothesized that absence of Lox will change the mechanical behavior of the aortic wall because of reduced elastin and collagen cross-linking and alter the expression levels of matrisome and smooth muscle cell (SMC) genes in a vascular location-specific manner. Using fluorescence microscopy, pressure myography, and gene set enrichment analysis, we visualized the microarchitecture, quantified the mechanical behavior, and examined matrisome and SMC gene expression from ascending aortas (AAs) and descending aortas (DAs) from newborn Lox+/+ and Lox-/- mice. Even though Lox-/- AAs and DAs have fragmented elastic laminae and disorganized SMCs, the unloaded outer diameter and wall thickness were similar to Lox+/+ AAs and DAs. Lox-/- AAs and DAs have altered opening angles, circumferential stresses, and circumferential stretch ratios; however, only Lox-/- AAs have increased pressurized diameters and tangent moduli. Gene set enrichment analysis showed upregulation of the extracellular matrix (ECM) regulator gene set in Lox-/- AAs and DAs as well as differential expression of secreted factors, collagens, ECM-affiliated proteins, ECM glycoproteins, and SMC cell cycle gene sets that depend on the Lox genotype and vascular location. These results provide insights into the local chemomechanical changes induced by Lox inactivation that may be important for TAAD pathogenesis.NEW & NOTEWORTHY Absence of lysyl oxidase (Lox) causes thoracic aortic aneurysms. The aortic mechanical behavior of Lox-/- mice is consistent with reduced elastin and collagen cross-linking but demonstrates vascular location-specific differences. Lox-/- aortas show upregulation of matrix remodeling genes and location-specific differential expression of other matrix and smooth muscle cell gene sets.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Aorta, Thoracic/enzymology
- Aorta, Thoracic/pathology
- Aorta, Thoracic/physiopathology
- Aortic Aneurysm, Thoracic/enzymology
- Aortic Aneurysm, Thoracic/genetics
- Aortic Aneurysm, Thoracic/pathology
- Aortic Aneurysm, Thoracic/physiopathology
- Arterial Pressure
- Biomechanical Phenomena
- Collagen/genetics
- Collagen/metabolism
- Dilatation, Pathologic
- Disease Models, Animal
- Elastin/genetics
- Elastin/metabolism
- Extracellular Matrix Proteins/genetics
- Extracellular Matrix Proteins/metabolism
- Gene Expression Profiling
- Gene Expression Regulation
- Genetic Predisposition to Disease
- Mechanotransduction, Cellular
- Mice, Knockout
- Phenotype
- Protein-Lysine 6-Oxidase/genetics
- Protein-Lysine 6-Oxidase/metabolism
- Stress, Mechanical
- Vascular Stiffness
Collapse
Affiliation(s)
- Marius Catalin Staiculescu
- Department of Mechanical Engineering and Materials Science, Washington University, St. Louis, Missouri; and
| | - Jungsil Kim
- Department of Mechanical Engineering and Materials Science, Washington University, St. Louis, Missouri; and
| | - Robert P Mecham
- Department of Cell Biology and Physiology, Washington University, St. Louis, Missouri
| | - Jessica E Wagenseil
- Department of Mechanical Engineering and Materials Science, Washington University, St. Louis, Missouri; and
| |
Collapse
|
46
|
Kapsokalyvas D, van Hoof M, Wigren S, Chimhanda T, Kuijpers HJ, Ramaekers FCS, Stokroos RJ, van Zandvoort MAMJ. Investigating the race for the surface and skin integration in clinically retrieved abutments with two-photon microscopy. Colloids Surf B Biointerfaces 2017; 159:97-107. [PMID: 28780465 DOI: 10.1016/j.colsurfb.2017.07.072] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 07/14/2017] [Accepted: 07/26/2017] [Indexed: 12/14/2022]
Abstract
Bone conduction hearing implants can rehabilitate some types of hearing loss. A hydroxyapatite (HA)-coated skin-penetrating abutment was developed to allow for soft tissue preservation and increased skin-abutment adherence. Inflammation is thought to relate to bacterial infection of pockets around the abutment. Upon integration, the host's ability to cover the abutment surface ("race for the surface"), and thus control and prevent competitive bacteria from colonizing it, is improved. However, the attachment mechanisms behind it are not clear. In this study, we applied two-photon microscopy to visualize tissue attachment on abutments retrieved from patients. Skin integration markers were validated and applied to four HA-coated abutments. Evidence of skin integration was found, including the presence of hemidesmosomes, a basement membrane, dermal collagen and vascularization. Cases with clinical signs of severe inflammation and evident biofilm formation showed limited skin integration based on these indicators, confirming the applicability of the "race for the surface" model.
Collapse
Affiliation(s)
- D Kapsokalyvas
- Department of Molecular Cell Biology, CARIM School for Cardiovascular diseases, Maastricht University Medical Center, UNS 50, 6229 ER Maastricht, The Netherlands
| | - M van Hoof
- Department of Otorhinolaryngology and Head and Neck Surgery, Maastricht University Medical Center, PO Box 5800, 6202, AZ, Maastricht, The Netherlands.
| | - S Wigren
- Cochlear Bone Anchored Solutions, AB, Mölnlycke, Sweden
| | - T Chimhanda
- Department of Molecular Cell Biology, CARIM School for Cardiovascular diseases, Maastricht University Medical Center, UNS 50, 6229 ER Maastricht, The Netherlands
| | - H J Kuijpers
- Department of Molecular Cell Biology, CARIM School for Cardiovascular diseases, Maastricht University Medical Center, UNS 50, 6229 ER Maastricht, The Netherlands
| | - F C S Ramaekers
- Department of Molecular Cell Biology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - R J Stokroos
- Department of Otorhinolaryngology and Head and Neck Surgery, Maastricht University Medical Center, PO Box 5800, 6202, AZ, Maastricht, The Netherlands
| | - M A M J van Zandvoort
- Department of Molecular Cell Biology, CARIM School for Cardiovascular diseases, Maastricht University Medical Center, UNS 50, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
47
|
Kim J, Staiculescu MC, Cocciolone AJ, Yanagisawa H, Mecham RP, Wagenseil JE. Crosslinked elastic fibers are necessary for low energy loss in the ascending aorta. J Biomech 2017; 61:199-207. [PMID: 28778385 DOI: 10.1016/j.jbiomech.2017.07.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Revised: 06/30/2017] [Accepted: 07/16/2017] [Indexed: 11/30/2022]
Abstract
In the large arteries, it is believed that elastin provides the resistance to stretch at low pressure, while collagen provides the resistance to stretch at high pressure. It is also thought that elastin is responsible for the low energy loss observed with cyclic loading. These tenets are supported through experiments that alter component amounts through protease digestion, vessel remodeling, normal growth, or in different artery types. Genetic engineering provides the opportunity to revisit these tenets through the loss of expression of specific wall components. We used newborn mice lacking elastin (Eln-/-) or two key proteins (lysyl oxidase, Lox-/-, or fibulin-4, Fbln4-/-) that are necessary for the assembly of mechanically-functional elastic fibers to investigate the contributions of elastic fibers to large artery mechanics. We determined component content and organization and quantified the nonlinear and viscoelastic mechanical behavior of Eln-/-, Lox-/-, and Fbln4-/- ascending aorta and their respective controls. We confirmed that the lack of elastin, fibulin-4, or lysyl oxidase leads to absent or highly fragmented elastic fibers in the aortic wall and a 56-97% decrease in crosslinked elastin amounts. We found that the resistance to stretch at low pressure is decreased only in Eln-/- aorta, confirming the role of elastin in the nonlinear mechanical behavior of the aortic wall. Dissipated energy with cyclic loading and unloading is increased 53-387% in Eln-/-, Lox-/-, and Fbln4-/- aorta, indicating that not only elastin, but properly assembled and crosslinked elastic fibers, are necessary for low energy loss in the aorta.
Collapse
Affiliation(s)
- Jungsil Kim
- Dept. of Mechanical Engineering and Materials Science, Washington University, St. Louis, MO, United States
| | - Marius Catalin Staiculescu
- Dept. of Mechanical Engineering and Materials Science, Washington University, St. Louis, MO, United States
| | - Austin J Cocciolone
- Dept. of Mechanical Engineering and Materials Science, Washington University, St. Louis, MO, United States
| | - Hiromi Yanagisawa
- Life Science Center of Tsukuba Advance Research Alliance, University of Tsukuba, Japan
| | - Robert P Mecham
- Dept. of Cell Biology and Physiology, Washington University, St. Louis, MO, United States
| | - Jessica E Wagenseil
- Dept. of Mechanical Engineering and Materials Science, Washington University, St. Louis, MO, United States.
| |
Collapse
|
48
|
van Haaften EE, Bouten CVC, Kurniawan NA. Vascular Mechanobiology: Towards Control of In Situ Regeneration. Cells 2017; 6:E19. [PMID: 28671618 PMCID: PMC5617965 DOI: 10.3390/cells6030019] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 06/16/2017] [Accepted: 06/23/2017] [Indexed: 01/08/2023] Open
Abstract
The paradigm of regenerative medicine has recently shifted from in vitro to in situ tissue engineering: implanting a cell-free, biodegradable, off-the-shelf available scaffold and inducing the development of functional tissue by utilizing the regenerative potential of the body itself. This approach offers a prospect of not only alleviating the clinical demand for autologous vessels but also circumventing the current challenges with synthetic grafts. In order to move towards a hypothesis-driven engineering approach, we review three crucial aspects that need to be taken into account when regenerating vessels: (1) the structure-function relation for attaining mechanical homeostasis of vascular tissues, (2) the environmental cues governing cell function, and (3) the available experimental platforms to test instructive scaffolds for in situ tissue engineering. The understanding of cellular responses to environmental cues leads to the development of computational models to predict tissue formation and maturation, which are validated using experimental platforms recapitulating the (patho)physiological micro-environment. With the current advances, a progressive shift is anticipated towards a rational and effective approach of building instructive scaffolds for in situ vascular tissue regeneration.
Collapse
Affiliation(s)
- Eline E van Haaften
- Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands.
- Institute for Complex Molecular Systems, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands.
| | - Carlijn V C Bouten
- Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands.
- Institute for Complex Molecular Systems, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands.
| | - Nicholas A Kurniawan
- Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands.
- Institute for Complex Molecular Systems, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands.
| |
Collapse
|
49
|
Reimann C, Brangsch J, Colletini F, Walter T, Hamm B, Botnar RM, Makowski MR. Molecular imaging of the extracellular matrix in the context of atherosclerosis. Adv Drug Deliv Rev 2017; 113:49-60. [PMID: 27639968 DOI: 10.1016/j.addr.2016.09.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 09/01/2016] [Accepted: 09/07/2016] [Indexed: 12/25/2022]
Abstract
This review summarizes the current status of molecular imaging of the extracellular matrix (ECM) in the context of atherosclerosis. Apart from cellular components, the ECM of the atherosclerotic plaque plays a relevant role during the initiation of atherosclerosis and its' subsequent progression. Important structural and signaling components of the ECM include elastin, collagen and fibrin. However, the ECM not only plays a structural role in the arterial wall but also interacts with different cell types and has important biological signaling functions. Molecular imaging of the ECM has emerged as a new diagnostic tool to characterize biological aspects of atherosclerotic plaques, which cannot be characterized by current clinically established imaging techniques, such as X-ray angiography. Different types of molecular probes can be detected in vivo by imaging modalities such as magnetic resonance imaging (MRI), positron emission tomography (PET) and single photon emission computed tomography (SPECT). The modality specific signaling component of the molecular probe provides information about its spatial location and local concentration. The successful introduction of molecular imaging into clinical practice and guidelines could open new pathways for an earlier detection of disease processes and a better understanding of the disease state on a biological level. Quantitative in vivo molecular parameters could also contribute to the development and evaluation of novel cardiovascular therapeutic interventions and the assessment of response to treatment.
Collapse
Affiliation(s)
| | | | | | - Thula Walter
- Department of Radiology, Charité, Berlin, Germany
| | - Bernd Hamm
- Department of Radiology, Charité, Berlin, Germany
| | - Rene M Botnar
- King's College London, Division of Imaging Sciences, United Kingdom; Wellcome Trust and EPSRC Medical Engineering Center, United Kingdom; BHF Centre of Excellence, King's College London, London, United Kingdom; NIHR Biomedical Research Centre, King's College London, London, United Kingdom
| | - Marcus R Makowski
- Department of Radiology, Charité, Berlin, Germany; King's College London, Division of Imaging Sciences, United Kingdom.
| |
Collapse
|
50
|
Eskaros AR, Egloff SAA, Boyd KL, Richardson JE, Hyndman ME, Zijlstra A. Larger core size has superior technical and analytical accuracy in bladder tissue microarray. J Transl Med 2017; 97:335-342. [PMID: 28112755 DOI: 10.1038/labinvest.2016.151] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 11/16/2016] [Accepted: 12/05/2016] [Indexed: 12/17/2022] Open
Abstract
The construction of tissue microarrays (TMAs) with cores from a large number of paraffin-embedded tissues (donors) into a single paraffin block (recipient) is an effective method of analyzing samples from many patient specimens simultaneously. For the TMA to be successful, the cores within it must capture the correct histologic areas from the donor blocks (technical accuracy) and maintain concordance with the tissue of origin (analytical accuracy). This can be particularly challenging for tissues with small histological features such as small islands of carcinoma in situ (CIS), thin layers of normal urothelial lining of the bladder, or cancers that exhibit intratumor heterogeneity. In an effort to create a comprehensive TMA of a bladder cancer patient cohort that accurately represents the tumor heterogeneity and captures the small features of normal and CIS, we determined how core size (0.6 vs 1.0 mm) impacted the technical and analytical accuracy of the TMA. The larger 1.0 mm core exhibited better technical accuracy for all tissue types at 80.9% (normal), 94.2% (tumor), and 71.4% (CIS) compared with 58.6%, 85.9%, and 63.8% for 0.6 mm cores. Although the 1.0 mm core provided better tissue capture, increasing the number of replicates from two to three allowed with the 0.6 mm core compensated for this reduced technical accuracy. However, quantitative image analysis of proliferation using both Ki67+ immunofluorescence counts and manual mitotic counts demonstrated that the 1.0 mm core size also exhibited significantly greater analytical accuracy (P=0.004 and 0.035, respectively, r2=0.979 and 0.669, respectively). Ultimately, our findings demonstrate that capturing two or more 1.0 mm cores for TMA construction provides superior technical and analytical accuracy over the smaller 0.6 mm cores, especially for tissues harboring small histological features or substantial heterogeneity.
Collapse
Affiliation(s)
- Adel Rh Eskaros
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shanna A Arnold Egloff
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Kelli L Boyd
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Joyce E Richardson
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - M Eric Hyndman
- Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Andries Zijlstra
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|