1
|
Liao CY, Hundscheid JH, Crawford J, ten Dijke P, Coornaert B, Danen EH. Novel high throughput 3D ECM remodeling assay identifies MEK as key driver of fibrotic fibroblast activity. Mater Today Bio 2025; 32:101800. [PMID: 40343164 PMCID: PMC12059351 DOI: 10.1016/j.mtbio.2025.101800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 01/21/2025] [Accepted: 04/24/2025] [Indexed: 05/11/2025] Open
Abstract
In fibrotic tissues, activated fibroblasts remodel the collagen-rich extracellular matrix (ECM). Intervening with this process represents a candidate therapeutic strategy to attenuate disease progression. Models that generate quantitative data on 3D fibroblast-mediated ECM remodeling with the reproducibility and throughput needed for drug testing are lacking. Here, we develop a model that fits this purpose and produces combined quantitative information on drug efficacy and cytotoxicity. We use microinjection robotics to design patterns of fibrillar collagen-embedded fibroblast clusters and apply automated microscopy and image analysis to quantify ECM remodeling between-, and cell viability within clusters of TGFβ-activated primary human skin or lung fibroblasts. We apply this assay to compound screening and reveal actionable targets to suppress fibrotic ECM remodeling. Strikingly, we find that after an initial phase of fibroblast activation by TGFβ, canonical TGFβ signaling is dispensable and, instead, non-canonical activation of MEK-ERK signaling drives ECM remodeling. Moreover, we reveal that higher concentrations of two TGFβ receptor inhibitors while blocking canonical TGFβ signaling, in fact stimulate this MEK-mediated profibrotic ECM remodeling activity.
Collapse
Affiliation(s)
- Chen-Yi Liao
- Leiden Academic Center for Drug Research, Leiden University, Leiden, the Netherlands
| | | | | | - Peter ten Dijke
- Oncode Institute and Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Erik H.J. Danen
- Leiden Academic Center for Drug Research, Leiden University, Leiden, the Netherlands
| |
Collapse
|
2
|
Kent O, Casey ER, Brown M, Bell S, Ehrman MC, Flagler MJ, Määttä A, Benham AM, Hawkins TJ. New imaging tools reveal live cellular collagen secretion, fibril dynamics and network organisation. Sci Rep 2025; 15:13764. [PMID: 40258849 PMCID: PMC12012225 DOI: 10.1038/s41598-025-96280-4] [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: 08/15/2024] [Accepted: 03/27/2025] [Indexed: 04/23/2025] Open
Abstract
Although light microscopy has been used to examine the early trafficking of collagen within the cell, much of our understanding of the detailed organisation of cell deposited collagen is from static electron microscopy studies. To understand the dynamics of live cell collagen deposition and fibril organisation, we generated a bright photostable mNGCol1α2 fusion protein and employed a range of microscopy techniques to follow its intracellular transport and elucidate extracellular fibril formation. Our findings reveal the dynamics of fibril growth and the dynamic nature of collagen network interactions at the cellular level. Notably we observed molecular events that build network organisation, including fibril bundling, bifurcation, directionality along existing fibrils, and looping/intertwining behaviours. Strikingly, mNGCol1α2 fluorescence intensity maxima can mark a fibril before another growing collagen fibril intersects at this location. Real-time, high-resolution imaging of collagen has enabled fibrillogenesis and organisational dynamics to be visualised together in an actively secreting cellular system. We also show that the N-terminal protease site is not an absolute requirement for collagen fibril incorporation. This approach paves the way for assessing the dynamic organisation and assembly of collagen into the extracellular matrix in skin models and other tissues during health, ageing and disease.
Collapse
Affiliation(s)
- Olivia Kent
- Department of Biosciences, Durham University, South Road, Durham, DH1 3LE, UK
| | - Eleanor R Casey
- Department of Biosciences, Durham University, South Road, Durham, DH1 3LE, UK
| | - Max Brown
- The Procter & Gamble Company, Newcastle Innovation Centre, Newcastle-Upon-Tyne, NE12 9TS, UK
| | - Steven Bell
- Department of Biosciences, Durham University, South Road, Durham, DH1 3LE, UK
| | - Matthew C Ehrman
- Procter & Gamble International Operations SA SG Branch, 70 Biopolis Street, Singapore, 138547, Singapore
| | | | - Arto Määttä
- Department of Biosciences, Durham University, South Road, Durham, DH1 3LE, UK
| | - Adam M Benham
- Department of Biosciences, Durham University, South Road, Durham, DH1 3LE, UK
| | - Timothy J Hawkins
- Department of Biosciences, Durham University, South Road, Durham, DH1 3LE, UK.
| |
Collapse
|
3
|
Patten J, Halligan P, Bashiri G, Kegel M, Bonadio JD, Wang K. EDA Fibronectin Microarchitecture and YAP Translocation during Wound Closure. ACS Biomater Sci Eng 2025; 11:2249-2262. [PMID: 40029610 DOI: 10.1021/acsbiomaterials.4c02019] [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: 03/05/2025]
Abstract
Fibronectin (Fn) is an extracellular matrix glycoprotein with mechanosensitive structure-function. Extra domain A (EDA) Fn, a Fn isoform, is not present in adult tissue but is required for tissue repair. Curiously, EDA Fn is linked to both regenerative and fibrotic tissue repair. Given that Fn mechanoregulates cell behavior, EDA Fn organization during wound closure might play a role in mediating these differing responses. One mechanism by which cells sense and respond to their microenvironment is by activating a transcriptional coactivator, yes-associated protein (YAP). Interestingly, YAP activity is not only required for wound closure but similarly linked to both regenerative and fibrotic repair. Therefore, this study aims to evaluate how, during normal and fibrotic wound closure, EDA Fn organization might modulate YAP translocation by culturing human dermal fibroblasts on polydimethylsiloxane substrates mimicking normal (soft: 18 kPa) and fibrotic (stiff: 146 kPa) wounded skin. On stiffer substrates mimicking fibrotic wounds, fibroblasts assembled an aligned EDA Fn matrix comprising thinner fibers, suggesting increased microenvironmental tension. To evaluate if cell binding to the EDA domain of Fn was essential to overall matrix organization, fibroblasts were treated with Irigenin, which inhibits binding to the EDA domain within Fn. Blocking adhesion to EDA led to randomly organized EDA Fn matrices with thicker fibers, suggesting reduced microenvironmental tension even during fibrotic wound closure. To evaluate whether YAP signaling plays a role in EDA Fn organization, fibroblasts were treated with CA3, which suppresses YAP activity in a dose-dependent manner. Treatment with CA3 also led to randomly organized EDA Fn matrices with thicker fibers, suggesting a potential connected mechanism of reducing tension during fibrotic wound closure. Next, YAP activity was assessed to evaluate the impact of EDA Fn organization. Interestingly, fibroblasts migrating on softer substrates mimicking normal wounds increased YAP activity, but on stiffer substrates, they decreased YAP activity. When fibroblasts on stiffer substrates were treated with Irigenin or CA3, fibroblasts increased YAP activity. These results suggest that there may be disrupted signaling between EDA Fn organization and YAP translocation during fibrotic wound closure that could be restored when reestablishing normal EDA Fn matrix organization to instead drive regenerative wound repair.
Collapse
Affiliation(s)
- Jennifer Patten
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Patrick Halligan
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Ghazal Bashiri
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Michael Kegel
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Jacob D Bonadio
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Karin Wang
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, United States
| |
Collapse
|
4
|
Sun Y, Hamlin AJ, Schwarzbauer JE. Fibronectin matrix assembly at a glance. J Cell Sci 2025; 138:jcs263834. [PMID: 40130407 PMCID: PMC12050093 DOI: 10.1242/jcs.263834] [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] [Indexed: 03/26/2025] Open
Abstract
The organization and mechanics of extracellular matrix (ECM) protein polymers determine tissue structure and function. Secreted ECM components are assembled into polymers via a cell-mediated process. The specific mechanisms that cells use for assembly are crucial for generating tissue-appropriate matrices. Fibronectin (FN) is a ubiquitous and abundant ECM protein that is assembled into a fibrillar matrix by a receptor-mediated process, and the FN matrix provides a foundation for incorporation of many other proteins into the ECM. In this Cell Science at a Glance article and the accompanying poster, we describe the domain organization of FN and the events that initiate and propagate a stable insoluble network of FN fibrils. We also discuss intracellular pathways that regulate FN assembly and the impact of changes in assembly on disease progression.
Collapse
Affiliation(s)
- Yu Sun
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Aaron J. Hamlin
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | | |
Collapse
|
5
|
Kuebart T, Oezel L, Gürsoy B, Maus U, Windolf J, Bittersohl B, Grotheer V. Periostin Splice Variant Expression in Human Osteoblasts from Osteoporotic Patients and Its Effects on Interleukin-6 and Osteoprotegerin. Int J Mol Sci 2025; 26:932. [PMID: 39940700 PMCID: PMC11816753 DOI: 10.3390/ijms26030932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 01/10/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
Osteoporosis is an inflammatory disease characterised by low bone mass and quality, resulting in weaker bone strength and fragility fractures. Periostin is a matricellular protein expressed in the periosteum of bone by osteoblasts. It regulates cell recruitment and differentiation in response to fracture and contributes to extracellular matrix (ECM) formation. The aim of the following study was to determine the splice variants of Periostin expressed in human osteoblasts and Periostin's function in the pathophysiology of osteoporosis. Osteoblasts isolated from femoral heads from 29 patients with or without osteoporosis were utilised. Periostin splice variants were compared by quantitative real-time polymerase chain reaction (qPCR). Furthermore, the effect of Periostin inhibition on osteoblast differentiation was investigated using alizarin red S staining. Lastly, the interaction of IL-6 and Periostin and their effect on osteoprotegerin (OPG) secretion were analysed with the implantation of enzyme-linked immunosorbent assays (ELISAs). It could be demonstrated that human osteoblasts preferentially express Periostin isoform 4, even if splice variant expression was not altered in osteoporosis conditions, indicating that Periostin's functions in bone are primarily attributable to this isoform. The inhibition of Periostin resulted in significantly reduced osteoblast differentiation. However, Periostin was secreted in significantly higher amounts in osteoblasts from patients with osteoporosis. Additionally, Periostin significantly reduces OPG secretion and, thereby, rather promotes bone resorption. Furthermore, it could be determined that Periostin and IL-6 induce each other, and both significantly decrease OPG secretion. A positive feedback loop exacerbates the dysregulation found in human osteoblasts from patients with osteoporosis, thereby contributing to bone loss.
Collapse
Affiliation(s)
- Till Kuebart
- Department of Orthopedics and Trauma Surgery, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University, 40225 Duesseldorf, Germany; (T.K.)
| | - Lisa Oezel
- Department of Orthopedics and Trauma Surgery, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University, 40225 Duesseldorf, Germany; (T.K.)
| | - Beyza Gürsoy
- Department of Orthopedics and Trauma Surgery, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University, 40225 Duesseldorf, Germany; (T.K.)
| | - Uwe Maus
- Department of Orthopedics and Trauma Surgery, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University, 40225 Duesseldorf, Germany; (T.K.)
| | - Joachim Windolf
- Department of Orthopedics and Trauma Surgery, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University, 40225 Duesseldorf, Germany; (T.K.)
| | - Bernd Bittersohl
- Department of Orthopedics, Medical School and University Medical Center Ostwestalen-Lippe (OWL), Klinikum Bielefeld-Mitte, Bielefeld University, 33615 Bielefeld, Germany (V.G.)
| | - Vera Grotheer
- Department of Orthopedics, Medical School and University Medical Center Ostwestalen-Lippe (OWL), Klinikum Bielefeld-Mitte, Bielefeld University, 33615 Bielefeld, Germany (V.G.)
| |
Collapse
|
6
|
Magdaleno C, Tschumperlin DJ, Rajasekaran N, Varadaraj A. SOCS domain targets ECM assembly in lung fibroblasts and experimental lung fibrosis. Sci Rep 2024; 14:31855. [PMID: 39738247 PMCID: PMC11686354 DOI: 10.1038/s41598-024-83187-9] [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: 03/04/2024] [Accepted: 12/12/2024] [Indexed: 01/01/2025] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal disease defined by a progressive decline in lung function due to scarring and accumulation of extracellular matrix (ECM) proteins. The SOCS (Suppressor Of Cytokine Signaling) domain is a 40 amino acid conserved domain known to form a functional ubiquitin ligase complex targeting the Von Hippel Lindau (VHL) protein for proteasomal degradation. Here we show that the SOCS conserved domain operates as a molecular tool, to disrupt collagen and fibronectin fibrils in the ECM associated with fibrotic lung myofibroblasts. Our results demonstrate that fibroblasts differentiated using TGFβ, followed by transduction with the SOCS domain, exhibit significantly reduced levels of the contractile myofibroblast-marker, α-SMA. Furthermore, in support of its role to retard differentiation, we find that lung fibroblasts expressing the SOCS domain present with significantly reduced levels of α-SMA and fibrillar fibronectin after differentiation with TGFβ. We show that adenoviral delivery of the SOCS domain in the fibrotic phase of experimental lung fibrosis in mice, significantly reduces collagen accumulation in disease lungs. These data underscore a novel function for the SOCS domain and its potential in ameliorating pathologic matrix deposition in lung fibroblasts and experimental lung fibrosis.
Collapse
Affiliation(s)
- Carina Magdaleno
- Department of Chemistry and Biochemistry, Northern Arizona University, Flagstaff, AZ, USA
- Cancer Center, University of Arizona, Tucson, AZ, USA
| | - Daniel J Tschumperlin
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Narendiran Rajasekaran
- Department of Chemistry and Biochemistry, Northern Arizona University, Flagstaff, AZ, USA.
| | - Archana Varadaraj
- Department of Chemistry and Biochemistry, Northern Arizona University, Flagstaff, AZ, USA.
| |
Collapse
|
7
|
Lunde IG, Rypdal KB, Van Linthout S, Diez J, González A. Myocardial fibrosis from the perspective of the extracellular matrix: Mechanisms to clinical impact. Matrix Biol 2024; 134:1-22. [PMID: 39214156 DOI: 10.1016/j.matbio.2024.08.008] [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: 05/07/2024] [Revised: 08/08/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Fibrosis is defined by the excessive accumulation of extracellular matrix (ECM) and constitutes a central pathophysiological process that underlies tissue dysfunction, across organs, in multiple chronic diseases and during aging. Myocardial fibrosis is a key contributor to dysfunction and failure in numerous diseases of the heart and is a strong predictor of poor clinical outcome and mortality. The excess structural and matricellular ECM proteins deposited by cardiac fibroblasts, is found between cardiomyocytes (interstitial fibrosis), in focal areas where cardiomyocytes have died (replacement fibrosis), and around vessels (perivascular fibrosis). Although myocardial fibrosis has important clinical prognostic value, access to cardiac tissue biopsies for histological evaluation is limited. Despite challenges with sensitivity and specificity, cardiac magnetic resonance imaging (CMR) is the most applicable diagnostic tool in the clinic, and the scientific community is currently actively searching for blood biomarkers reflecting myocardial fibrosis, to complement the imaging techniques. The lack of mechanistic insights into specific pro- and anti-fibrotic molecular pathways has hampered the development of effective treatments to prevent or reverse myocardial fibrosis. Development and implementation of anti-fibrotic therapies is expected to improve patient outcomes and is an urgent medical need. Here, we discuss the importance of the ECM in the heart, the central role of fibrosis in heart disease, and mechanistic pathways likely to impact clinical practice with regards to diagnostics of myocardial fibrosis, risk stratification of patients, and anti-fibrotic therapy.
Collapse
Affiliation(s)
- Ida G Lunde
- Oslo Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Oslo, Norway; KG Jebsen Center for Cardiac Biomarkers, Campus Ahus, University of Oslo, Oslo, Norway.
| | - Karoline B Rypdal
- Oslo Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Oslo, Norway; KG Jebsen Center for Cardiac Biomarkers, Campus Ahus, University of Oslo, Oslo, Norway
| | - Sophie Van Linthout
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Javier Diez
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra, Department of Cardiology, Clínica Universidad de Navarra and IdiSNA Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - Arantxa González
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra, Department of Cardiology, Clínica Universidad de Navarra and IdiSNA Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain
| |
Collapse
|
8
|
Musiime M, Erusappan PM, Cukierman E, Chang J, Molven A, Hansen U, Zeltz C, Gullberg D. Fibroblast integrin α11β1 is a collagen assembly receptor in mechanoregulated fibrillar adhesions. Matrix Biol 2024; 134:144-161. [PMID: 39406317 DOI: 10.1016/j.matbio.2024.10.006] [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: 04/19/2024] [Revised: 10/12/2024] [Accepted: 10/12/2024] [Indexed: 11/25/2024]
Abstract
Solid epithelial cancers with significant desmoplasia are characterized by an excessive deposition of collagen-based matrix, which often supports tumor progression. However, the mechanism of how collagen receptors mediate collagen fibrillogenesis still remains mostly unclear. We show that the collagen-binding integrin α11β1 can co-localize with tensin-1 and deposited collagen I in human pancreatic ductal adenocarcinoma (PDAC) stroma. In addition to the canonical fibrillar adhesion integrin α5β1 expressed by human PDAC cancer-associated fibroblasts (CAFs), tensin-1-positive fibrillar adhesions contained α11β1 but lacked α1β1 and α2β1. CAFs lacking α5β1 expression displayed mechanoregulated and tensin-1 dependent α11β1 fibrillar adhesions, suggesting independent roles of the two integrins with regards to fibrillar adhesions-based de novo fibrillogenesis. Further, we demonstrate that cell surface-associated collagen I assembly necessitated α11β1, but not α5β1 expression. In summary, α11β1 integrin is a novel component of fibrillar adhesions, which is strategically positioned to mediate de novo collagen fibrillogenesis at the cell surface under pro-fibrotic conditions.
Collapse
Affiliation(s)
- Moses Musiime
- University of Bergen, Department of Biomedicine and Centre for Cancer Biomarkers, Jonas Lies vei 91, 5009 Bergen, Norway
| | - Pugazendhi Murugan Erusappan
- University of Bergen, Department of Biomedicine and Centre for Cancer Biomarkers, Jonas Lies vei 91, 5009 Bergen, Norway; Institute for Experimental Medical Research, Oslo university Hospital and university of Oslo, Kirkeveien 166, 0450, Oslo, Norway
| | - Edna Cukierman
- Cancer Signaling & Microenvironment Program, M&C Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Temple Health, Philadelphia, PA, 19111, USA
| | - Joan Chang
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, UK
| | - Anders Molven
- Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, NO-5020 Bergen, Norway; Department of Pathology and Section for Cancer Genomics, Haukeland University Hospital, NO-5020 Bergen, Norway
| | - Uwe Hansen
- Institute for Musculoskeletal Medicine, University Hospital of Münster, Münster, Germany
| | - Cédric Zeltz
- University of Bergen, Department of Biomedicine and Centre for Cancer Biomarkers, Jonas Lies vei 91, 5009 Bergen, Norway.
| | - Donald Gullberg
- University of Bergen, Department of Biomedicine and Centre for Cancer Biomarkers, Jonas Lies vei 91, 5009 Bergen, Norway.
| |
Collapse
|
9
|
Zeyer KA, Bornert O, Nelea V, Bao X, Leytens A, Sharoyan S, Sengle G, Antonyan A, Bruckner-Tuderman L, Dengjel J, Reinhardt DP, Nyström A. Dipeptidyl Peptidase-4-Mediated Fibronectin Processing Evokes a Profibrotic Extracellular Matrix. J Invest Dermatol 2024; 144:2477-2487.e13. [PMID: 38570029 DOI: 10.1016/j.jid.2024.03.020] [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: 11/09/2023] [Revised: 02/21/2024] [Accepted: 03/13/2024] [Indexed: 04/05/2024]
Abstract
Fibronectin serves as a platform to guide and facilitate deposition of collagen and fibrillin microfibrils. During development of fibrotic diseases, altered fibronectin deposition in the extracellular matrix (ECM) is generally an early event. After this, dysregulated organization of fibrillins and fibrillar collagens occurs. Because fibronectin is an essential orchestrator of healthy ECM, perturbation of its ECM-organizational capacity may be involved in development of fibrosis. To investigate this, we employed recessive dystrophic epidermolysis bullosa as a disease model with progressive, severe dermal fibrosis. Fibroblasts from donors with recessive dystrophic epidermolysis bullosa in 2-dimensional and 3-dimensional cultures displayed dysregulated fibronectin deposition. Our analyses revealed that increase of profibrotic dipeptidyl peptidase-4-positive fibroblasts coincides with altered fibronectin deposition. Dipeptidyl peptidase-4 inhibitors normalized deposition of fibronectin and subsequently of fibrillin microfibrils and collagen I. Intriguingly, proteomics and inhibitor and mutagenesis studies disclosed that dipeptidyl peptidase-4 modulates ECM deposition through the proteolysis of the fibronectin N-terminus. Our study provides mechanistic insights into the observed profibrotic activities of dipeptidyl peptidase-4 and extends the understanding of fibronectin-guided ECM assembly in health and disease.
Collapse
Affiliation(s)
- Karina A Zeyer
- Department of Dermatology, Medical Faculty, Medical Center - University of Freiburg, Freiburg, Germany
| | - Olivier Bornert
- Department of Dermatology, Medical Faculty, Medical Center - University of Freiburg, Freiburg, Germany
| | - Valentin Nelea
- Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada; Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Canada
| | - Xinyi Bao
- Department of Dermatology, Medical Faculty, Medical Center - University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Alexandre Leytens
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Svetlana Sharoyan
- H. Buniatian Institute of Biochemistry of Armenian NAS, Yerevan, Republic of Armenia
| | - Gerhard Sengle
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), Medical Faculty, University of Cologne, Cologne, Germany; Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Cologne Center for Musculoskeletal Biomechanics (CCMB), Cologne, Germany
| | - Alvard Antonyan
- H. Buniatian Institute of Biochemistry of Armenian NAS, Yerevan, Republic of Armenia
| | - Leena Bruckner-Tuderman
- Department of Dermatology, Medical Faculty, Medical Center - University of Freiburg, Freiburg, Germany
| | - Jörn Dengjel
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Dieter P Reinhardt
- Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada; Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Canada
| | - Alexander Nyström
- Department of Dermatology, Medical Faculty, Medical Center - University of Freiburg, Freiburg, Germany.
| |
Collapse
|
10
|
Dinesh NEH, Baratang N, Rosseau J, Mohapatra R, Li L, Mahalingam R, Tiedemann K, Campeau PM, Reinhardt DP. Fibronectin isoforms promote postnatal skeletal development. Matrix Biol 2024; 133:86-102. [PMID: 39159790 DOI: 10.1016/j.matbio.2024.08.002] [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: 04/11/2024] [Revised: 08/02/2024] [Accepted: 08/12/2024] [Indexed: 08/21/2024]
Abstract
Fibronectin (FN) is a ubiquitous extracellular matrix glycoprotein essential for the development of various tissues. Mutations in FN cause a unique form of spondylometaphyseal dysplasia, emphasizing its importance in cartilage and bone development. However, the relevance and functional role of FN during skeletal development has remained elusive. To address these aspects, we have generated conditional knockout mouse models targeting the cellular FN isoform in cartilage (cFNKO), the plasma FN isoform in hepatocytes (pFNKO), and both isoforms together in a double knockout (FNdKO). We used these mice to determine the relevance of the two principal FN isoforms in skeletal development from postnatal day one to the adult stage at two months. We identified a distinct topological FN deposition pattern in the mouse limb during different gestational and postnatal skeletal development phases, with prominent levels at the resting and hypertrophic chondrocyte zones and in the trabecular bone. Cartilage-specific cFN emerged as the predominant isoform in the growth plate, whereas circulating pFN remained excluded from the growth plate and confined to the primary and secondary ossification centers. Deleting either isoform independently (cFNKO or pFNKO) yielded only relatively subtle changes in the analyzed skeletal parameters. However, the double knockout of cFN in the growth plate and pFN in the circulation of the FNdKO mice significantly reduced postnatal body weight, body length, and bone length. Micro-CT analysis of the adult bone microarchitecture in FNdKO mice exposed substantial reductions in trabecular bone parameters and bone mineral density. The mice also showed elevated bone marrow adiposity. Analysis of chondrogenesis in FNdKO mice demonstrated changes in the resting, proliferating and hypertrophic growth plate zones, consistent alterations in chondrogenic markers such as collagen type II and X, decreased apoptosis of hypertrophic chondrocytes, and downregulation of bone formation markers. Transforming growth factor-β1 and downstream phospho-AKT levels were significantly lower in the FNdKO than in the control mice, revealing a crucial FN-mediated regulatory pathway in chondrogenesis and bone formation. In conclusion, the data demonstrate that FN is essential for chondrogenesis and bone development. Even though cFN and pFN act in different regions of the bone, both FN isoforms are required for the regulation of chondrogenesis, cartilage maturation, trabecular bone formation, and overall skeletal growth.
Collapse
Affiliation(s)
- Neha E H Dinesh
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
| | | | | | - Ronit Mohapatra
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
| | - Ling Li
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
| | - Ramshaa Mahalingam
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada
| | | | | | - Dieter P Reinhardt
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada; Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada.
| |
Collapse
|
11
|
Rusbjerg-Weberskov CE, Scavenius C, Enghild JJ, Nielsen NS. Periostin Is a Disulfide-Bonded Homodimer and Forms a Complex with Fibronectin in the Human Skin. Biochemistry 2024; 63:2658-2669. [PMID: 39352075 DOI: 10.1021/acs.biochem.4c00240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2024]
Abstract
The protein periostin is a matricellular protein that is expressed in connective tissue. It is composed of five globular domains arranged in an elongated structure with an extensive disordered C-terminal tail. Periostin contains 11 cysteine residues, of which one is unpaired and the rest form five intramolecular disulfide bonds. Periostin plays an important role during wound healing and is also involved in driving the inflammatory state in atopic diseases. This study provides a comprehensive biochemical characterization of periostin in human skin and in dermal and pulmonary fibroblasts in vitro. Through the application of Western blotting, co-immunoprecipitation, and LC-MS/MS, we show for the first time that periostin is a disulfide-bonded homodimer and engages in a novel disulfide-bonded complex with fibronectin both in vivo and in vitro. This inherent characteristic of periostin holds the potential to redefine our approach to exploring and understanding its functional role in future research endeavors.
Collapse
Affiliation(s)
| | - Carsten Scavenius
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | - Jan J Enghild
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | - Nadia Sukusu Nielsen
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| |
Collapse
|
12
|
Patten J, Halligan P, Bashiri G, Kegel M, Bonadio JD, Wang K. EDA Fibronectin Microarchitecture and YAP Translocation During Wound Closure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.23.614581. [PMID: 39386582 PMCID: PMC11463502 DOI: 10.1101/2024.09.23.614581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Fibronectin (Fn) is an extracellular matrix glycoprotein with mechanosensitive structure-function. EDA Fn, a Fn isoform, is not present in adult tissue but is required for tissue repair. Curiously, EDA Fn is linked to both regenerative and fibrotic tissue repair. Given that Fn mechanoregulates cell behavior, Fn EDA organization during wound closure might play a role in mediating these differing responses. One mechanism by which cells sense and respond to their microenvironment is by activating a transcriptional co-activator, Yes-associated protein (YAP). Interestingly, YAP activity is not only required for wound closure, but similarly linked to both regenerative and fibrotic repair. Therefore, this study aims to evaluate how, during normal and fibrotic wound closure, EDA Fn organization might modulate YAP translocation by culturing human dermal fibroblasts on polydimethylsiloxane (PDMS) substrates mimicking normal (soft: 18 kPa) and fibrotic (stiff: 146 kPa) wounded skin. On stiffer substrates mimicking fibrotic wounds, fibroblasts assembled an aligned EDA Fn matrix comprising thinner fibers, suggesting increased microenvironmental tension. To evaluate if cell binding to the EDA domain of Fn was essential to overall matrix organization, fibroblasts were treated with Irigenin, which inhibits binding to the EDA domain within Fn. Blocking adhesion to EDA led to randomly organized EDA Fn matrices with thicker fibers, suggesting reduced microenvironmental tension even during fibrotic wound closure. To evaluate if YAP signaling plays a role in EDA Fn organization, fibroblasts were treated with CA3, which suppresses YAP activity in a dose-dependent manner. Treatment with CA3 also led to randomly organized EDA Fn matrices with thicker fibers, suggesting a potential connected mechanism of reducing tension during fibrotic wound closure. Next, YAP activity was assessed to evaluate the impact of EDA Fn organization. Interestingly, fibroblasts migrating on softer substrates mimicking normal wounds increased YAP activity but on stiffer substrates, decreased YAP activity. When fibroblasts on stiffer substrates were treated with Irigenin or CA3, fibroblasts increased YAP activity. These results suggest there may be disrupted signaling between EDA Fn organization and YAP translocation during fibrotic wound closure that could be restored when reestablishing normal EDA Fn matrix organization to instead drive regenerative wound repair.
Collapse
Affiliation(s)
- Jennifer Patten
- Department of Bioengineering, Temple University, Pennsylvania
| | | | - Ghazal Bashiri
- Department of Bioengineering, Temple University, Pennsylvania
| | - Michael Kegel
- Department of Bioengineering, Temple University, Pennsylvania
| | - Jacob D Bonadio
- Department of Bioengineering, Temple University, Pennsylvania
| | - Karin Wang
- Department of Bioengineering, Temple University, Pennsylvania
| |
Collapse
|
13
|
Seyed-Razavi Y, Lee SR, Fan J, Shen W, Cornish EE, Gillies MC. JR5558 mice are a reliable model to investigate subretinal fibrosis. Sci Rep 2024; 14:18752. [PMID: 39138242 PMCID: PMC11322289 DOI: 10.1038/s41598-024-66068-z] [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: 09/29/2023] [Accepted: 06/26/2024] [Indexed: 08/15/2024] Open
Abstract
Subretinal fibrosis is a major untreatable cause of poor outcomes in neovascular age-related macular degeneration. Mouse models of subretinal fibrosis all possess a degree of invasiveness and tissue damage not typical of fibrosis progression. This project characterises JR5558 mice as a model to study subretinal fibrosis. Fundus and optical coherence tomography (OCT) imaging was used to non-invasively track lesions. Lesion number and area were quantified with ImageJ. Retinal sections, wholemounts and Western blots were used to characterise alterations. Subretinal lesions expand between 4 and 8 weeks and become established in size and location around 12 weeks. Subretinal lesions were confirmed to be fibrotic, including various cell populations involved in fibrosis development. Müller cell processes extended from superficial retina into subretinal lesions at 8 weeks. Western blotting revealed increases in fibronectin (4 wk and 8 wk, p < 0.001), CTGF (20 wks, p < 0.001), MMP2 (12 wks and 20 wks p < 0.05), αSMA (12 wks and 20 wks p < 0.05) and GFAP (8 wk and 12 wk, p ≤ 0.01), consistent with our immunofluorescence results. Intravitreal injection of Aflibercept reduced subretinal lesion growth. Our study provides evidence JR5558 mice have subretinal fibrotic lesions that grow between 4 and 8 weeks and confirms this line to be a good model to study subretinal fibrosis development and assess treatment options.
Collapse
Affiliation(s)
- Yashar Seyed-Razavi
- Save Sight Institute, Discipline of Ophthalmology, Sydney Medical School, The University of Sydney, Sydney, NSW, 2000, Australia.
- Centre for Vision Research, Westmead Institute for Medical Research, Faculty of Medicine and Health, Sydney University, Sydney, Westmead, NSW, 2145, Australia.
| | - So-Ra Lee
- Save Sight Institute, Discipline of Ophthalmology, Sydney Medical School, The University of Sydney, Sydney, NSW, 2000, Australia
| | - Jiawen Fan
- Save Sight Institute, Discipline of Ophthalmology, Sydney Medical School, The University of Sydney, Sydney, NSW, 2000, Australia
| | - Weiyong Shen
- Save Sight Institute, Discipline of Ophthalmology, Sydney Medical School, The University of Sydney, Sydney, NSW, 2000, Australia
| | - Elisa E Cornish
- Save Sight Institute, Discipline of Ophthalmology, Sydney Medical School, The University of Sydney, Sydney, NSW, 2000, Australia
| | - Mark C Gillies
- Save Sight Institute, Discipline of Ophthalmology, Sydney Medical School, The University of Sydney, Sydney, NSW, 2000, Australia.
| |
Collapse
|
14
|
Bonadio JD, Bashiri G, Halligan P, Kegel M, Ahmed F, Wang K. Delivery technologies for therapeutic targeting of fibronectin in autoimmunity and fibrosis applications. Adv Drug Deliv Rev 2024; 209:115303. [PMID: 38588958 PMCID: PMC11111362 DOI: 10.1016/j.addr.2024.115303] [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: 10/02/2023] [Revised: 02/29/2024] [Accepted: 04/03/2024] [Indexed: 04/10/2024]
Abstract
Fibronectin (FN) is a critical component of the extracellular matrix (ECM) contributing to various physiological processes, including tissue repair and immune response regulation. FN regulates various cellular functions such as adhesion, proliferation, migration, differentiation, and cytokine release. Alterations in FN expression, deposition, and molecular structure can profoundly impact its interaction with other ECM proteins, growth factors, cells, and associated signaling pathways, thus influencing the progress of diseases such as fibrosis and autoimmune disorders. Therefore, developing therapeutics that directly target FN or its interaction with cells and other ECM components can be an intriguing approach to address autoimmune and fibrosis pathogenesis.
Collapse
Affiliation(s)
- Jacob D Bonadio
- Department of Bioengineering, Temple University, Philadelphia, PA, United States
| | - Ghazal Bashiri
- Department of Bioengineering, Temple University, Philadelphia, PA, United States
| | - Patrick Halligan
- Department of Bioengineering, Temple University, Philadelphia, PA, United States
| | - Michael Kegel
- Department of Bioengineering, Temple University, Philadelphia, PA, United States
| | - Fatima Ahmed
- Department of Bioengineering, Temple University, Philadelphia, PA, United States
| | - Karin Wang
- Department of Bioengineering, Temple University, Philadelphia, PA, United States.
| |
Collapse
|
15
|
Sakai T, Choo YY, Mitsuhashi S, Ikebe R, Jeffers A, Idell S, Tucker TA, Ikebe M. Myocardin regulates fibronectin expression and secretion from human pleural mesothelial cells. Am J Physiol Lung Cell Mol Physiol 2024; 326:L419-L430. [PMID: 38349126 PMCID: PMC11281794 DOI: 10.1152/ajplung.00271.2023] [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: 08/25/2023] [Revised: 01/18/2024] [Accepted: 01/26/2024] [Indexed: 03/20/2024] Open
Abstract
During the progression of pleural fibrosis, pleural mesothelial cells (PMCs) undergo a phenotype switching process known as mesothelial-mesenchymal transition (MesoMT). During MesoMT, transformed PMCs become myofibroblasts that produce increased extracellular matrix (ECM) proteins, including collagen and fibronectin (FN1) that is critical to develop fibrosis. Here, we studied the mechanism that regulates FN1 expression in myofibroblasts derived from human pleural mesothelial cells (HPMCs). We found that myocardin (Myocd), a transcriptional coactivator of serum response factor (SRF) and a master regulator of smooth muscle and cardiac muscle differentiation, strongly controls FN1 gene expression. Myocd gene silencing markedly inhibited FN1 expression. FN1 promoter analysis revealed that deletion of the Smad3-binding element diminished FN1 promoter activity, whereas deletion of the putative SRF-binding element increased FN1 promoter activity. Smad3 gene silencing decreased FN1 expression, whereas SRF gene silencing increased FN1 expression. Moreover, SRF competes with Smad3 for binding to Myocd. These results indicate that Myocd activates FN1 expression through Smad3, whereas SRF inhibits FN1 expression in HPMCs. In HPMCs, TGF-β induced Smad3 nuclear localization, and the proximity ligation signal between Myocd and Smad3 was markedly increased after TGF-β stimulation at nucleus, suggesting that TGF-β facilitates nuclear translocation of Smad3 and interaction between Smad3 and Myocd. Moreover, Myocd and Smad3 were coimmunoprecipitated and isolated Myocd and Smad3 proteins directly bound each other. Chromatin immunoprecipitation assays revealed that Myocd interacts with the FN1 promoter at the Smad3-binding consensus sequence. The results indicate that Myocd regulates FN1 gene activation through interaction and activation of the Smad3 transcription factor.NEW & NOTEWORTHY During phenotype switching from mesothelial to mesenchymal, pleural mesothelial cells (PMCs) produce extracellular matrix (ECM) proteins, including collagen and fibronectin (FN1), critical components in the development of fibrosis. Here, we found that myocardin, a transcriptional coactivator of serum response factor (SRF), strongly activates FN1 expression through Smad3, whereas SRF inhibits FN1 expression. This study provides insights about the regulation of FN1 that could lead to the development of novel interventional approaches to prevent pleural fibrosis.
Collapse
Affiliation(s)
- Tsuyoshi Sakai
- Department of Cellular and Molecular Biology, University of Texas at Tyler Health Science Center, Tyler, Texas, United States
| | - Young-Yeon Choo
- Department of Cellular and Molecular Biology, University of Texas at Tyler Health Science Center, Tyler, Texas, United States
| | - Shinya Mitsuhashi
- Department of Cellular and Molecular Biology, University of Texas at Tyler Health Science Center, Tyler, Texas, United States
| | - Reiko Ikebe
- Department of Cellular and Molecular Biology, University of Texas at Tyler Health Science Center, Tyler, Texas, United States
| | - Ann Jeffers
- Department of Cellular and Molecular Biology, University of Texas at Tyler Health Science Center, Tyler, Texas, United States
| | - Steven Idell
- Department of Cellular and Molecular Biology, University of Texas at Tyler Health Science Center, Tyler, Texas, United States
| | - Torry A Tucker
- Department of Cellular and Molecular Biology, University of Texas at Tyler Health Science Center, Tyler, Texas, United States
| | - Mitsuo Ikebe
- Department of Cellular and Molecular Biology, University of Texas at Tyler Health Science Center, Tyler, Texas, United States
| |
Collapse
|
16
|
Ioannidis V, Pandey R, Bauer HF, Schön M, Bockmann J, Boeckers TM, Lutz AK. Disrupted extracellular matrix and cell cycle genes in autism-associated Shank3 deficiency are targeted by lithium. Mol Psychiatry 2024; 29:704-717. [PMID: 38123724 PMCID: PMC11153165 DOI: 10.1038/s41380-023-02362-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 11/20/2023] [Accepted: 12/01/2023] [Indexed: 12/23/2023]
Abstract
The Shank3 gene encodes the major postsynaptic scaffolding protein SHANK3. Its mutation causes a syndromic form of autism spectrum disorder (ASD): Phelan-McDermid Syndrome (PMDS). It is characterized by global developmental delay, intellectual disorders (ID), ASD behavior, affective symptoms, as well as extra-cerebral symptoms. Although Shank3 deficiency causes a variety of molecular alterations, they do not suffice to explain all clinical aspects of this heterogenic syndrome. Since global gene expression alterations in Shank3 deficiency remain inadequately studied, we explored the transcriptome in vitro in primary hippocampal cells from Shank3∆11(-/-) mice, under control and lithium (Li) treatment conditions, and confirmed the findings in vivo. The Shank3∆11(-/-) genotype affected the overall transcriptome. Remarkably, extracellular matrix (ECM) and cell cycle transcriptional programs were disrupted. Accordingly, in the hippocampi of adolescent Shank3∆11(-/-) mice we found proteins of the collagen family and core cell cycle proteins downregulated. In vitro Li treatment of Shank3∆11(-/-) cells had a rescue-like effect on the ECM and cell cycle gene sets. Reversed ECM gene sets were part of a network, regulated by common transcription factors (TF) such as cAMP responsive element binding protein 1 (CREB1) and β-Catenin (CTNNB1), which are known downstream effectors of synaptic activity and targets of Li. These TFs were less abundant and/or hypo-phosphorylated in hippocampi of Shank3∆11(-/-) mice and could be rescued with Li in vitro and in vivo. Our investigations suggest the ECM compartment and cell cycle genes as new players in the pathophysiology of Shank3 deficiency, and imply involvement of transcriptional regulators, which can be modulated by Li. This work supports Li as potential drug in the management of PMDS symptoms, where a Phase III study is ongoing.
Collapse
Affiliation(s)
- Valentin Ioannidis
- Institute for Anatomy and Cell Biology, Ulm University, 89081, Ulm, Germany
| | - Rakshita Pandey
- Institute for Anatomy and Cell Biology, Ulm University, 89081, Ulm, Germany
- International Graduate School in Molecular Medicine Ulm, Ulm University, Ulm, Germany
| | - Helen Friedericke Bauer
- Institute for Anatomy and Cell Biology, Ulm University, 89081, Ulm, Germany
- International Graduate School in Molecular Medicine Ulm, Ulm University, Ulm, Germany
| | - Michael Schön
- Institute for Anatomy and Cell Biology, Ulm University, 89081, Ulm, Germany
| | - Jürgen Bockmann
- Institute for Anatomy and Cell Biology, Ulm University, 89081, Ulm, Germany
| | - Tobias M Boeckers
- Institute for Anatomy and Cell Biology, Ulm University, 89081, Ulm, Germany
- German Center for Neurodegenerative Diseases (DZNE), Ulm site, 89081, Ulm, Germany
| | - Anne-Kathrin Lutz
- Institute for Anatomy and Cell Biology, Ulm University, 89081, Ulm, Germany.
| |
Collapse
|
17
|
Magdaleno C, Tschumperlin DJ, Rajasekaran N, Varadaraj A. SOCS domain targets ECM assembly in lung fibroblasts and experimental lung fibrosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.14.580347. [PMID: 38469152 PMCID: PMC10926664 DOI: 10.1101/2024.02.14.580347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal disease defined by a progressive decline in lung function due to scarring and accumulation of extracellular matrix (ECM) proteins. The SOCS (Suppressor Of Cytokine Signaling) domain is a 40 amino acid conserved domain known to form a functional ubiquitin ligase complex targeting the Von Hippel Lindau (VHL) protein for proteasomal degradation. Here we show that the SOCS conserved domain operates as a molecular tool, to disrupt collagen and fibronectin fibrils in the ECM associated with fibrotic lung myofibroblasts. Our results demonstrate that fibroblasts differentiated using TGFß, followed by transduction with the SOCS domain, exhibit significantly reduced levels of the contractile myofibroblast-marker, α-SMA. Furthermore, in support of its role to retard differentiation, we find that lung fibroblasts expressing the SOCS domain present with significantly reduced levels of α-SMA and fibrillar fibronectin after differentiation with TGFß. We show that adenoviral delivery of the SOCS domain in the fibrotic phase of experimental lung fibrosis in mice, significantly reduces collagen accumulation in disease lungs. These data underscore a novel function for the SOCS domain and its potential in ameliorating pathologic matrix deposition in lung fibroblasts and experimental lung fibrosis.
Collapse
Affiliation(s)
- Carina Magdaleno
- Department of Chemistry and Biochemistry, Northern Arizona University, Flagstaff, Arizona, USA
| | - Daniel J. Tschumperlin
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
| | - Narendiran Rajasekaran
- Correspondence to: Archana Varadaraj, PO Box 5698, Science and Health Building, Rm430, Flagstaff, AZ, 86011, USA. Tel: (928) 523-6394, Fax: (928) 523-8111, ; Narendiran Rajasekaran, PO Box 5698, Science and Health Building, Rm430, Flagstaff, AZ, 86011, USA. Tel: (928) 523-6394, Fax: (928) 523-8111,
| | - Archana Varadaraj
- Department of Chemistry and Biochemistry, Northern Arizona University, Flagstaff, Arizona, USA
| |
Collapse
|
18
|
Yu W, Ye H, Li Y, Bao X, Ni Y, Chen X, Sun Y, Chen A, Zhou W, Li J. Pneumocystis carinii infection drives upregulation of Fn1 expression that causes pulmonary fibrosis with an inflammatory response. Rev Iberoam Micol 2024; 41:17-26. [PMID: 39332977 DOI: 10.1016/j.riam.2024.04.002] [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: 05/04/2023] [Revised: 03/15/2024] [Accepted: 04/09/2024] [Indexed: 09/29/2024] Open
Abstract
BACKGROUND Pneumocystis carinii is an opportunistic fungal pathogen that may cause pneumonia and lead to pulmonary fibrosis. AIMS This study attempted to investigate the role of P. carinii infection-related genes in regulating lung fibrosis in mice. METHODS A screening of P. carinii infection-related differential mRNAs was performed using the GEO database, followed by protein-protein interaction (PPI) network construction using the STRING website in order to obtain P. carinii infection-related key genes. The development of a mouse model with gene aberrant expression was achieved by utilizing mice carrying the Cre-LoxP recombinase system. Dexamethasone was employed to induce tracheal infection in order to develop a model of pulmonary fibrosis, and the magnitude of lung injury was assessed by performing hematoxylin-eosin (H&E) staining and Masson staining. Lung coefficient and hydroxyproline level were assessed on sections of lung tissue as well. Finally, the magnitude of lung fibrosis and inflammation in mice was determined based on immunofluorescence and on the expression of genes associated with lung fibrosis and inflammation. RESULTS Fn1 was found by PPI with the highest connectivity in the PPI network associated with immunity and inflammation. Besides, Fn1 was significantly highly expressed in P. carinii-infected mice samples. The P carinii pneumonia (PCP)+Fn1fl/fl group had significantly higher lung coefficients, hydroxyproline levels and TNF-α, IL-6, IL-1β, IL-8 and NLRP3 expression levels, and significantly lower IL-10 expression levels. The results found in PCP+SPC-Cre:Fn1fl/fl group were the opposite. The results of the pulmonary fibrosis level study showed that the PCP+Fn1fl/fl group had the most intense H&E and Masson staining, and significantly higher expression levels of Col1A2, Col3A1 and α-SMA, which were lower in the PCP+SPC-Cre:Fn1fl/fl group. CONCLUSIONS P. carinii infection may promote the upregulation of Fn1, which causes pulmonary fibrosis with an inflammatory response.
Collapse
Affiliation(s)
- Wenwen Yu
- Department of Respiratory and Critical Care Medicine of Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang China
| | - Hua Ye
- Department of Respiratory and Critical Care Medicine of Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang China
| | - Yunlei Li
- Department of Respiratory and Critical Care Medicine of Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang China
| | - Xiaoqiong Bao
- Department of Respiratory and Critical Care Medicine of Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang China
| | - Yangyang Ni
- Department of Respiratory and Critical Care Medicine of Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang China
| | - Xiangxiang Chen
- Department of Respiratory and Critical Care Medicine of Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang China
| | - Yangjie Sun
- Department of Respiratory and Critical Care Medicine of Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang China
| | - Ali Chen
- Department of Respiratory and Critical Care Medicine of Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang China
| | - Weilong Zhou
- Department of Respiratory and Critical Care Medicine of Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang China
| | - Jifa Li
- Department of Respiratory and Critical Care Medicine of Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang China.
| |
Collapse
|
19
|
Faralli JA, Filla MS, Peters DM. Role of integrins in the development of fibrosis in the trabecular meshwork. FRONTIERS IN OPHTHALMOLOGY 2023; 3:1274797. [PMID: 38983065 PMCID: PMC11182094 DOI: 10.3389/fopht.2023.1274797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 10/09/2023] [Indexed: 07/11/2024]
Abstract
Primary open angle glaucoma (POAG) is a progressive and chronic disease exhibiting many of the features of fibrosis. The extracellular matrix (ECM) in the trabecular meshwork (TM) undergoes extensive remodeling and enhanced rigidity, resembling fibrotic changes. In addition, there are changes associated with myofibroblast activation and cell contractility that further drives tissue fibrosis and stiffening. This review discusses what is known about the integrins in the TM and their involvement in fibrotic processes.
Collapse
Affiliation(s)
- Jennifer A. Faralli
- Department of Pathology & Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Mark S. Filla
- Department of Pathology & Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Donna M. Peters
- Department of Pathology & Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
- Department of Ophthalmology & Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| |
Collapse
|
20
|
Luangmonkong T, Parichatikanond W, Olinga P. Targeting collagen homeostasis for the treatment of liver fibrosis: Opportunities and challenges. Biochem Pharmacol 2023; 215:115740. [PMID: 37567319 DOI: 10.1016/j.bcp.2023.115740] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 07/24/2023] [Accepted: 08/08/2023] [Indexed: 08/13/2023]
Abstract
Liver fibrosis is an excessive production, aberrant deposition, and deficit degradation of extracellular matrix (ECM). Patients with unresolved fibrosis ultimately undergo end-stage liver diseases. To date, the effective and safe strategy to cease fibrosis progression remains an unmet clinical need. Since collagens are the most abundant ECM protein which play an essential role in fibrogenesis, the suitable regulation of collagen homeostasis could be an effective strategy for the treatment of liver fibrosis. Therefore, this review provides a brief overview on the dysregulation of ECM homeostasis, focusing on collagens, in the pathogenesis of liver fibrosis. Most importantly, promising therapeutic mechanisms related to biosynthesis, deposition and extracellular interactions, and degradation of collagens, together with preclinical and clinical antifibrotic evidence of drugs affecting each target are orderly criticized. In addition, challenges for targeting collagen homeostasis in the treatment of liver fibrosis are discussed.
Collapse
Affiliation(s)
- Theerut Luangmonkong
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Thailand; Centre of Biopharmaceutical Science for Healthy Ageing (BSHA), Faculty of Pharmacy, Mahidol University, Bangkok, Thailand.
| | - Warisara Parichatikanond
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Thailand; Centre of Biopharmaceutical Science for Healthy Ageing (BSHA), Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
| | - Peter Olinga
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, The Netherlands
| |
Collapse
|
21
|
Guinard I, Nguyen T, Brassard-Jollive N, Weber J, Ruch L, Reininger L, Brouard N, Eckly A, Collin D, Lanza F, Léon C. Matrix stiffness controls megakaryocyte adhesion, fibronectin fibrillogenesis, and proplatelet formation through Itgβ3. Blood Adv 2023; 7:4003-4018. [PMID: 37171626 PMCID: PMC10410137 DOI: 10.1182/bloodadvances.2022008680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 04/13/2023] [Accepted: 04/17/2023] [Indexed: 05/13/2023] Open
Abstract
Megakaryocytes (MKs) are the precursor cells of platelets, located in the bone marrow (BM). Once mature, they extend elongated projections named proplatelets through sinusoid vessels, emerging from the marrow stroma into the circulating blood. Not all signals from the microenvironment that regulate proplatelet formation are understood, particularly those from the BM biomechanics. We sought to investigate how MKs perceive and adapt to modifications of the stiffness of their environment. Although the BM is one of the softest tissue of the body, its rigidification results from excess fibronectin (FN), and other matrix protein deposition occur upon myelofibrosis. Here, we have shown that mouse MKs are able to detect the stiffness of a FN-coated substrate and adapt their morphology accordingly. Using a polydimethylsiloxane substrate with stiffness varying from physiological to pathological marrow, we found that a stiff matrix favors spreading, intracellular contractility, and FN fibrils assembly at the expense of proplatelet formation. Itgb3, but not Itgb1, is required for stiffness sensing, whereas both integrins are involved in fibrils assembly. In contrast, soft substrates promote proplatelet formation in an Itgb3-dependent manner, consistent with the ex vivo decrease in proplatelet formation and the in vivo decrease in platelet number in Itgb3-deficient mice. Our findings demonstrate the importance of environmental stiffness for MK functions with potential pathophysiological implications during pathologies that deregulate FN deposition and modulate stiffness in the marrow.
Collapse
Affiliation(s)
- Ines Guinard
- UMR_S1255, INSERM, Etablissement Français du Sang-Grand Est, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Thao Nguyen
- UMR_S1255, INSERM, Etablissement Français du Sang-Grand Est, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Noémie Brassard-Jollive
- UMR_S1255, INSERM, Etablissement Français du Sang-Grand Est, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Josiane Weber
- UMR_S1255, INSERM, Etablissement Français du Sang-Grand Est, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Laurie Ruch
- UMR_S1255, INSERM, Etablissement Français du Sang-Grand Est, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Laura Reininger
- UMR_S1255, INSERM, Etablissement Français du Sang-Grand Est, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Nathalie Brouard
- UMR_S1255, INSERM, Etablissement Français du Sang-Grand Est, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Anita Eckly
- UMR_S1255, INSERM, Etablissement Français du Sang-Grand Est, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | | | - François Lanza
- UMR_S1255, INSERM, Etablissement Français du Sang-Grand Est, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Catherine Léon
- UMR_S1255, INSERM, Etablissement Français du Sang-Grand Est, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| |
Collapse
|
22
|
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
|
23
|
Jääskeläinen I, Petäistö T, Mirzarazi Dahagi E, Mahmoodi M, Pihlajaniemi T, Kaartinen MT, Heljasvaara R. Collagens Regulating Adipose Tissue Formation and Functions. Biomedicines 2023; 11:biomedicines11051412. [PMID: 37239083 DOI: 10.3390/biomedicines11051412] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/28/2023] [Accepted: 05/03/2023] [Indexed: 05/28/2023] Open
Abstract
The globally increasing prevalence of obesity is associated with the development of metabolic diseases such as type 2 diabetes, dyslipidemia, and fatty liver. Excess adipose tissue (AT) often leads to its malfunction and to a systemic metabolic dysfunction because, in addition to storing lipids, AT is an active endocrine system. Adipocytes are embedded in a unique extracellular matrix (ECM), which provides structural support to the cells as well as participating in the regulation of their functions, such as proliferation and differentiation. Adipocytes have a thin pericellular layer of a specialized ECM, referred to as the basement membrane (BM), which is an important functional unit that lies between cells and tissue stroma. Collagens form a major group of proteins in the ECM, and some of them, especially the BM-associated collagens, support AT functions and participate in the regulation of adipocyte differentiation. In pathological conditions such as obesity, AT often proceeds to fibrosis, characterized by the accumulation of large collagen bundles, which disturbs the natural functions of the AT. In this review, we summarize the current knowledge on the vertebrate collagens that are important for AT development and function and include basic information on some other important ECM components, principally fibronectin, of the AT. We also briefly discuss the function of AT collagens in certain metabolic diseases in which they have been shown to play central roles.
Collapse
Affiliation(s)
- Iida Jääskeläinen
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90014 Oulu, Finland
| | - Tiina Petäistö
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90014 Oulu, Finland
| | - Elahe Mirzarazi Dahagi
- Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC H3A 0C7, Canada
| | - Mahdokht Mahmoodi
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montréal, QC H3A 0C7, Canada
| | - Taina Pihlajaniemi
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90014 Oulu, Finland
| | - Mari T Kaartinen
- Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC H3A 0C7, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montréal, QC H3A 0C7, Canada
- Division of Experimental Medicine, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC H3A 0C7, Canada
| | - Ritva Heljasvaara
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90014 Oulu, Finland
| |
Collapse
|
24
|
Benn MC, Pot SA, Moeller J, Yamashita T, Fonta CM, Orend G, Kollmannsberger P, Vogel V. How the mechanobiology orchestrates the iterative and reciprocal ECM-cell cross-talk that drives microtissue growth. SCIENCE ADVANCES 2023; 9:eadd9275. [PMID: 36989370 PMCID: PMC10058249 DOI: 10.1126/sciadv.add9275] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 02/24/2023] [Indexed: 06/19/2023]
Abstract
Controlled tissue growth is essential for multicellular life and requires tight spatiotemporal control over cell proliferation and differentiation until reaching homeostasis. As cells synthesize and remodel extracellular matrix, tissue growth processes can only be understood if the reciprocal feedback between cells and their environment is revealed. Using de novo-grown microtissues, we identified crucial actors of the mechanoregulated events, which iteratively orchestrate a sharp transition from tissue growth to maturation, requiring a myofibroblast-to-fibroblast transition. Cellular decision-making occurs when fibronectin fiber tension switches from highly stretched to relaxed, and it requires the transiently up-regulated appearance of tenascin-C and tissue transglutaminase, matrix metalloprotease activity, as well as a switch from α5β1 to α2β1 integrin engagement and epidermal growth factor receptor signaling. As myofibroblasts are associated with wound healing and inflammatory or fibrotic diseases, crucial knowledge needed to advance regenerative strategies or to counter fibrosis and cancer progression has been gained.
Collapse
Affiliation(s)
- Mario C. Benn
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, Zurich 8093, Switzerland
| | - Simon A. Pot
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, Zurich 8093, Switzerland
| | - Jens Moeller
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, Zurich 8093, Switzerland
| | - Tadahiro Yamashita
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, Zurich 8093, Switzerland
| | - Charlotte M. Fonta
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, Zurich 8093, Switzerland
| | - Gertraud Orend
- The Tumor Microenvironment Laboratory, INSERM U1109, Hôpital Civil, Institut d'Hématologie et d'Immunologie, 1 Place de l'Hôpital, Strasbourg 67091, France
- Université Strasbourg, Strasbourg 67000, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg 67000, France
| | - Philip Kollmannsberger
- Biomedical Physics, Heinrich-Heine-University Düsseldorf, Universitätsstrasse 1, Düsseldorf 40225, Germany
| | - Viola Vogel
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, Zurich 8093, Switzerland
| |
Collapse
|
25
|
Bashiri G, Padilla MS, Swingle KL, Shepherd SJ, Mitchell MJ, Wang K. Nanoparticle protein corona: from structure and function to therapeutic targeting. LAB ON A CHIP 2023; 23:1432-1466. [PMID: 36655824 PMCID: PMC10013352 DOI: 10.1039/d2lc00799a] [Citation(s) in RCA: 83] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 11/29/2022] [Indexed: 05/31/2023]
Abstract
Nanoparticle (NP)-based therapeutics have ushered in a new era in translational medicine. However, despite the clinical success of NP technology, it is not well-understood how NPs fundamentally change in biological environments. When introduced into physiological fluids, NPs are coated by proteins, forming a protein corona (PC). The PC has the potential to endow NPs with a new identity and alter their bioactivity, stability, and destination. Additionally, the conformation of proteins is sensitive to their physical and chemical surroundings. Therefore, biological factors and protein-NP-interactions can induce changes in the conformation and orientation of proteins in vivo. Since the function of a protein is closely connected to its folded structure, slight differences in the surrounding environment as well as the surface characteristics of the NP materials may cause proteins to lose or gain a function. As a result, this can alter the downstream functionality of the NPs. This review introduces the main biological factors affecting the conformation of proteins associated with the PC. Then, four types of NPs with extensive utility in biomedical applications are described in greater detail, focusing on the conformation and orientation of adsorbed proteins. This is followed by a discussion on the instances in which the conformation of adsorbed proteins can be leveraged for therapeutic purposes, such as controlling protein conformation in assembled matrices in tissue, as well as controlling the PC conformation for modulating immune responses. The review concludes with a perspective on the remaining challenges and unexplored areas at the interface of PC and NP research.
Collapse
Affiliation(s)
- Ghazal Bashiri
- Department of Bioengineering, Temple University, Philadelphia, PA 19122, USA.
| | - Marshall S Padilla
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kelsey L Swingle
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sarah J Shepherd
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Karin Wang
- Department of Bioengineering, Temple University, Philadelphia, PA 19122, USA.
| |
Collapse
|
26
|
Aytemiz DG, Kambe Y, Hirata M, Nishi H, Kameda T. Effects of RGD-fused silk fibroin in a solution format on fibroblast proliferation and collagen production. Biomed Mater Eng 2023; 34:183-193. [PMID: 35871317 DOI: 10.3233/bme-221430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Collagen production in fibroblasts is important for skin tissue repair. Cell-adhesive Arg-Gly-Asp (RGD) peptides immobilized on scaffolds stimulate fibroblast collagen production, but RGD peptides in solution exhibit opposite effects. Transgenic silkworm technology enables the design of fusion positions for RGD peptides in silk fibroin molecules. The effect of RGD-fused silk fibroin in solution on fibroblast cell activity remains unclear. OBJECTIVE To clarify the effects of RGD peptides fused to silk fibroin heavy (H)-chain or light (L)-chain on fibroblast proliferation and collagen production when RGD-fused silk fibroin proteins were added to the culture medium. METHODS Silk fibers with RGD-fused H-chains (H-RGD) or L-chains (L-RGD) were degummed, dissolved, and dialyzed to prepare H-RGD or L-RGD aqueous solutions, respectively. These solutions were added to the fibroblast medium, and their proliferation and collagen production were quantified. RESULTS Both L- and H-RGD stimulated fibroblast proliferation at a similar level, even in a solution format, but L-RGD promoted fibroblast collagen production significantly, indicating the synergistic effect of the native H-chain and RGD-fused L-chain. CONCLUSION RGD-fused silk fibroin in solution stimulated fibroblast proliferation and collagen production, depending on the fusion position of the peptides.
Collapse
Affiliation(s)
- Derya G Aytemiz
- Silk Materials Research Group, Division of Silk-Producing Insect Biotechnology, Institute of Agrobiological Sciences, National Agriculture and Food Research Organization (NARO), Ibaraki, Japan
| | - Yusuke Kambe
- Silk Materials Research Group, Division of Silk-Producing Insect Biotechnology, Institute of Agrobiological Sciences, National Agriculture and Food Research Organization (NARO), Ibaraki, Japan
| | | | | | - Tsunenori Kameda
- Silk Materials Research Group, Division of Silk-Producing Insect Biotechnology, Institute of Agrobiological Sciences, National Agriculture and Food Research Organization (NARO), Ibaraki, Japan
| |
Collapse
|
27
|
Nashchekina Y, Nikonov P, Prasolov N, Sulatsky M, Chabina A, Nashchekin A. The Structural Interactions of Molecular and Fibrillar Collagen Type I with Fibronectin and Its Role in the Regulation of Mesenchymal Stem Cell Morphology and Functional Activity. Int J Mol Sci 2022; 23:ijms232012577. [PMID: 36293432 PMCID: PMC9604100 DOI: 10.3390/ijms232012577] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/14/2022] [Accepted: 10/17/2022] [Indexed: 11/16/2022] Open
Abstract
The observed differences in the structure of native tissue and tissue formed in vitro cause the loss of functional activity of cells cultured in vitro. The lack of fundamental knowledge about the protein mechanism interactions limits the ability to effectively create in vitro native tissue. Collagen is able to spontaneously assemble into fibrils in vitro, but in vivo, other proteins, for example fibronectin, have a noticeable effect on this process. The molecular or fibrillar structure of collagen plays an equally important role. Therefore, we studied the interaction of the molecular and fibrillar structure of collagen with fibronectin. Atomic force and transmission electron microscopy showed that the presence of fibronectin does not affect the native structure and diameter of collagen fibrils. Confocal microscopy demonstrated that the collagen structure affects the cell morphology. Cells are better spread on molecular collagen compared with cells cultured on fibrillar collagen. Fibronectin promotes the formation of a large number of focal contacts, while in combination with collagen of both forms, its effect is leveled. Thus, understanding the mechanisms of the relationship between the protein structure and composition will effectively manage the creation in vitro of a new tissue with native properties.
Collapse
Affiliation(s)
- Yuliya Nashchekina
- Institute of Cytology of the Russian Academy of Sciences, Center of Cell Technologies, Tikhoretsky pr. 4, St. Petersburg 194064, Russia
- Correspondence:
| | - Pavel Nikonov
- Institute of Cytology of the Russian Academy of Sciences, Center of Cell Technologies, Tikhoretsky pr. 4, St. Petersburg 194064, Russia
| | - Nikita Prasolov
- Laboratory «Characterization of Materials and Structures of Solid State Electronics», Ioffe Institute, Polytekhnicheskaya Str. 26, St. Petersburg 194021, Russia
| | - Maksim Sulatsky
- Institute of Cytology of the Russian Academy of Sciences, Center of Cell Technologies, Tikhoretsky pr. 4, St. Petersburg 194064, Russia
| | - Alina Chabina
- Institute of Cytology of the Russian Academy of Sciences, Center of Cell Technologies, Tikhoretsky pr. 4, St. Petersburg 194064, Russia
| | - Alexey Nashchekin
- Laboratory «Characterization of Materials and Structures of Solid State Electronics», Ioffe Institute, Polytekhnicheskaya Str. 26, St. Petersburg 194021, Russia
| |
Collapse
|
28
|
Pancreatic ductal adenocarcinoma: tumor microenvironment and problems in the development of novel therapeutic strategies. Clin Exp Med 2022:10.1007/s10238-022-00886-1. [DOI: 10.1007/s10238-022-00886-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 08/30/2022] [Indexed: 12/19/2022]
|
29
|
Kweon B, Kim DU, Oh JY, Oh H, Kim YC, Mun YJ, Bae GS, Park SJ. Arecae pericarpium water extract alleviates chronic pancreatitis by deactivating pancreatic stellate cells. Front Pharmacol 2022; 13:941955. [PMID: 36105227 PMCID: PMC9465814 DOI: 10.3389/fphar.2022.941955] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 08/02/2022] [Indexed: 01/30/2023] Open
Abstract
Chronic pancreatitis (CP) is a chronic inflammatory disease of the pancreas with irreversible morphological changes. Arecae pericarpium (ARP), known to improve gastrointestinal disorders, has not yet been reported to inhibit fibrosis in CP. Therefore, we investigated the beneficial effects of ARP on cerulein-induced CP. Cerulein (50 μg/kg) was administered intraperitoneally to mice every hour, six times a day, four times a week for a total of 3 weeks to induce CP. To ascertain the prophylactic effects of ARP, ARP water extract (50, 100, or 200 mg/kg) or saline was administered intraperitoneally 1 h before the onset of CP. To determine the therapeutic effects of ARP, ARP water extract (200 mg/kg) or saline was administered for a total of 1 week or 2 weeks, starting 2 weeks or 1 week after the onset of CP. The pancreas was collected immediately for histological analysis. Additionally, to determine the effectiveness and mechanism of ARP in alleviating pancreatic fibrosis, pancreatic stellate cells (PSCs) were isolated. ARP treatment considerably improved glandular atrophy and inflammation and repressed collagen deposition in the pancreas. Furthermore, ARP water extract inhibited extracellular matrix (ECM) constituents such as alpha-smooth muscle actin (α-SMA), collagen I, and fibronectin 1 (FN1) in pancreatic tissue and PSCs. ARP also suppressed transforming growth factor-β (TGF-β) signaling by inhibiting Smad2 phosphorylation. Our study suggests that ARP exhibits anti-fibrotic effects in cerulein-induced CP by inhibiting TGF-β/Smad signaling.
Collapse
Affiliation(s)
- Bitna Kweon
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, Iksan, Jeollabuk-do, South Korea
| | - Dong-Uk Kim
- Hanbang Cardio-Renal Syndrome Research Center, School of Korean Medicine, Wonkwang University, Iksan, Jeollabuk-do, South Korea
| | - Jin-Young Oh
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, Iksan, Jeollabuk-do, South Korea
| | - Hyuncheol Oh
- Hanbang Cardio-Renal Syndrome Research Center, School of Korean Medicine, Wonkwang University, Iksan, Jeollabuk-do, South Korea
- Institute of Pharmaceutical Research and Development, College of Pharmacy, Wonkwang University, Iksan, Jeollabuk-do, South Korea
| | - Youn-Chul Kim
- Institute of Pharmaceutical Research and Development, College of Pharmacy, Wonkwang University, Iksan, Jeollabuk-do, South Korea
| | - Yeun-Ja Mun
- Department of Anatomy, College of Korean Medicine, Wonkwang University, Iksan, Jeollabuk-do, South Korea
- Research Center of Traditional Korean Medicine, Wonkwang University, Iksan, Jeollabuk-do, South Korea
| | - Gi-Sang Bae
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, Iksan, Jeollabuk-do, South Korea
- Hanbang Cardio-Renal Syndrome Research Center, School of Korean Medicine, Wonkwang University, Iksan, Jeollabuk-do, South Korea
- Research Center of Traditional Korean Medicine, Wonkwang University, Iksan, Jeollabuk-do, South Korea
- *Correspondence: Gi-Sang Bae, ; Sung-Joo Park,
| | - Sung-Joo Park
- Hanbang Cardio-Renal Syndrome Research Center, School of Korean Medicine, Wonkwang University, Iksan, Jeollabuk-do, South Korea
- Department of Herbology, School of Korean Medicine, Wonkwang University, Iksan, Jeollabuk-do, South Korea
- *Correspondence: Gi-Sang Bae, ; Sung-Joo Park,
| |
Collapse
|
30
|
Dooling LJ, Saini K, Anlaş AA, Discher DE. Tissue mechanics coevolves with fibrillar matrisomes in healthy and fibrotic tissues. Matrix Biol 2022; 111:153-188. [PMID: 35764212 PMCID: PMC9990088 DOI: 10.1016/j.matbio.2022.06.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 06/16/2022] [Accepted: 06/23/2022] [Indexed: 12/12/2022]
Abstract
Fibrillar proteins are principal components of extracellular matrix (ECM) that confer mechanical properties to tissues. Fibrosis can result from wound repair in nearly every tissue in adults, and it associates with increased ECM density and crosslinking as well as increased tissue stiffness. Such fibrotic tissues are a major biomedical challenge, and an emerging view posits that the altered mechanical environment supports both synthetic and contractile myofibroblasts in a state of persistent activation. Here, we review the matrisome in several fibrotic diseases, as well as normal tissues, with a focus on physicochemical properties. Stiffness generally increases with the abundance of fibrillar collagens, the major constituent of ECM, with similar mathematical trends for fibrosis as well as adult tissues from soft brain to stiff bone and heart development. Changes in expression of other core matrisome and matrisome-associated proteins or proteoglycans contribute to tissue stiffening in fibrosis by organizing collagen, crosslinking ECM, and facilitating adhesion of myofibroblasts. Understanding how ECM composition and mechanics coevolve during fibrosis can lead to better models and help with antifibrotic therapies.
Collapse
Affiliation(s)
- Lawrence J Dooling
- Molecular and Cellular Biophysics Lab, University of Pennsylvania,Philadelphia, PA 19104, USA
| | - Karanvir Saini
- Molecular and Cellular Biophysics Lab, University of Pennsylvania,Philadelphia, PA 19104, USA
| | - Alişya A Anlaş
- Molecular and Cellular Biophysics Lab, University of Pennsylvania,Philadelphia, PA 19104, USA
| | - Dennis E Discher
- Molecular and Cellular Biophysics Lab, University of Pennsylvania,Philadelphia, PA 19104, USA.
| |
Collapse
|
31
|
Resnikoff HA, Miller CG, Schwarzbauer JE. Implications of fibrotic extracellular matrix in diabetic retinopathy. Exp Biol Med (Maywood) 2022; 247:1093-1102. [PMID: 35410521 PMCID: PMC9335512 DOI: 10.1177/15353702221087175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Fibrosis is an accumulation of extracellular matrix (ECM) proteins and fibers in a disordered fashion, which compromises cell and tissue functions. High glucose-induced fibrosis, a major pathophysiological change of diabetic retinopathy (DR), severely affects vision by compromising the retinal vasculature and ultimately disrupting retinal tissue organization. The retina is a highly vascularized, stratified tissue with multiple cell types organized into distinct layers. Chronically high blood glucose stimulates certain retinal cells to increase production and assembly of ECM proteins resulting in excess ECM deposition primarily in the capillary walls on the basal side of the endothelium. This subendothelial fibrosis of the capillaries is the earliest histological change in the diabetic retina and has been linked to the vascular dysfunction that underlies DR. Proteins that are not normally abundant in the capillary basement membrane (BM) matrix, such as the ECM protein fibronectin, are assembled in significant quantities, disrupting the architecture of the BM and altering its properties. Cell culture models have identified multiple mechanisms through which elevated glucose can stimulate fibronectin matrix assembly, including intracellular signaling pathways, alternative splicing, and non-enzymatic glycation of the ECM. The fibrotic subendothelial matrix alters cell adhesion and supports further accumulation of other ECM proteins leading to disruption of endothelial cell-cell junctions. We review evidence supporting the notion that these molecular changes in the ECM contribute to the pathogenesis of DR, including vascular leakage, loss of endothelial cells and pericytes, changes in blood flow, and neovascularization. We propose that the accumulation of ECM, especially fibronectin matrix, first around the vasculature and later in extravascular locations, plays a critical role in DR and vision loss. Strategies for DR prevention and treatment should consider the ECM a potential therapeutic target.
Collapse
Affiliation(s)
- Henry A Resnikoff
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544-1014, USA
| | - Charles G Miller
- Department of Ophthalmology, Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jean E Schwarzbauer
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544-1014, USA,Jean E Schwarzbauer.
| |
Collapse
|
32
|
Setiawati A, Jeong S, Brillian AI, Lee SH, Shim JG, Jung KH, Shin K. Fabrication of a Tailored, Hybrid Extracellular Matrix Composite. Macromol Biosci 2022; 22:e2200106. [PMID: 35765216 DOI: 10.1002/mabi.202200106] [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: 03/11/2022] [Revised: 05/24/2022] [Indexed: 11/10/2022]
Abstract
The extracellular matrix (ECM) is a network of connective fibers that supports cells living in their surroundings. Native ECM, generated by the secretory products of each tissue's resident cells, has a unique architecture with different protein composition depending on the tissue. Therefore, it is very difficult to artificially design in vivo architecture in tissue engineering. In this study, we fabricated a hybrid ECM scaffold from the basic structure of fibroblast-derived cellular ECMs by adding major ECM components of fibronectin (FN) and collagen (COL I) externally. It was confirmed that while maintaining the basic structure of the native ECM, major protein components can be regulated. Then, decellularization was performed to prepare hybrid ECM scaffolds with various protein compositions and we demonstrated that a liver-mimicking fibronectin (FN)-rich hybrid ECM promoted successful settling of H4IIE rat hepatoma cells. We believe that our method holds promise for the fabrication of scaffolds that provide a tailored cellular microenvironment for specific organs and serve as novel pathways for the replacement or regeneration of specific organ tissues. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Agustina Setiawati
- Department of Chemistry and Institute of Biological Interfaces, 35-Baekbeom-ro, Sogang University, Mapo-gu, Seoul, 04107, Republic of Korea.,Department of Life Science, 35-Baekbeom-ro, Sogang University, Mapo-gu, Seoul, 04107, Republic of Korea.,Faculty of Pharmacy, Paingan, Maguwoharjo, Depok, Sanata Dharma University, Sleman, Yogyakarta, 55284, Indonesia
| | - Sungwoo Jeong
- Department of Chemistry and Institute of Biological Interfaces, 35-Baekbeom-ro, Sogang University, Mapo-gu, Seoul, 04107, Republic of Korea
| | - Albertus Ivan Brillian
- Department of Chemistry and Institute of Biological Interfaces, 35-Baekbeom-ro, Sogang University, Mapo-gu, Seoul, 04107, Republic of Korea
| | - Sang Ho Lee
- Department of Chemistry and Institute of Biological Interfaces, 35-Baekbeom-ro, Sogang University, Mapo-gu, Seoul, 04107, Republic of Korea
| | - Jin-Gon Shim
- Department of Life Science, 35-Baekbeom-ro, Sogang University, Mapo-gu, Seoul, 04107, Republic of Korea
| | - Kwang-Hwan Jung
- Department of Life Science, 35-Baekbeom-ro, Sogang University, Mapo-gu, Seoul, 04107, Republic of Korea
| | - Kwanwoo Shin
- Department of Chemistry and Institute of Biological Interfaces, 35-Baekbeom-ro, Sogang University, Mapo-gu, Seoul, 04107, Republic of Korea
| |
Collapse
|
33
|
Dinesh NEH, Campeau PM, Reinhardt DP. Fibronectin isoforms in skeletal development and associated disorders. Am J Physiol Cell Physiol 2022; 323:C536-C549. [PMID: 35759430 DOI: 10.1152/ajpcell.00226.2022] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The extracellular matrix is an intricate and essential network of proteins and non-proteinaceous components that provide a conducive microenvironment for cells to regulate cell function, differentiation, and survival. Fibronectin is one key component in the extracellular matrix that participates in determining cell fate and function crucial for normal vertebrate development. Fibronectin undergoes time dependent expression patterns during stem cell differentiation, providing a unique stem cell niche. Mutations in fibronectin have been recently identified to cause a rare form of skeletal dysplasia with scoliosis and abnormal growth plates. Even though fibronectin has been extensively analyzed in developmental processes, the functional role and importance of this protein and its various isoforms in skeletal development remains less understood. This review attempts to provide a concise and critical overview of the role of fibronectin isoforms in cartilage and bone physiology and associated pathologies. This will facilitate a better understanding of the possible mechanisms through which fibronectin exerts its regulatory role on cellular differentiation during skeletal development. The review discusses the consequences of mutations in fibronectin leading to corner fracture type spondylometaphyseal dysplasia and presents a new outlook towards matrix-mediated molecular pathways in relation to therapeutic and clinical relevance.
Collapse
Affiliation(s)
- Neha E H Dinesh
- Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
| | | | - Dieter P Reinhardt
- Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada.,Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Canada
| |
Collapse
|
34
|
Sakai T, Choo YY, Sato O, Ikebe R, Jeffers A, Idell S, Tucker T, Ikebe M. Myo5b Transports Fibronectin-Containing Vesicles and Facilitates FN1 Secretion from Human Pleural Mesothelial Cells. Int J Mol Sci 2022; 23:ijms23094823. [PMID: 35563212 PMCID: PMC9101030 DOI: 10.3390/ijms23094823] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/20/2022] [Accepted: 04/23/2022] [Indexed: 12/13/2022] Open
Abstract
Pleural mesothelial cells (PMCs) play a central role in the progression of pleural fibrosis. As pleural injury progresses to fibrosis, PMCs transition to mesenchymal myofibroblast via mesothelial mesenchymal transition (MesoMT), and produce extracellular matrix (ECM) proteins including collagen and fibronectin (FN1). FN1 plays an important role in ECM maturation and facilitates ECM-myofibroblast interaction, thus facilitating fibrosis. However, the mechanism of FN1 secretion is poorly understood. We report here that myosin 5b (Myo5b) plays a critical role in the transportation and secretion of FN1 from human pleural mesothelial cells (HPMCs). TGF-β significantly increased the expression and secretion of FN1 from HPMCs and facilitates the close association of Myo5B with FN1 and Rab11b. Moreover, Myo5b directly binds to GTP bound Rab11b (Rab11b-GTP) but not GDP bound Rab11b. Myo5b or Rab11b knockdown via siRNA significantly attenuated the secretion of FN1 without changing FN1 expression. TGF-β also induced Rab11b-GTP formation, and Rab11b-GTP but not Rab11b-GDP significantly activated the actin-activated ATPase activity of Myo5B. Live cell imaging revealed that Myo5b- and FN1-containing vesicles continuously moved together in a single direction. These results support that Myo5b and Rab11b play an important role in FN1 transportation and secretion from HPMCs, and consequently may contribute to the development of pleural fibrosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Mitsuo Ikebe
- Correspondence: ; Tel.: +1-(903)-877-7785; Fax: +1-(903)-877-5438
| |
Collapse
|
35
|
Gardiner JC, Cukierman E. Meaningful connections: Interrogating the role of physical fibroblast cell-cell communication in cancer. Adv Cancer Res 2022; 154:141-168. [PMID: 35459467 PMCID: PMC9483832 DOI: 10.1016/bs.acr.2022.01.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
As part of the connective tissue, activated fibroblasts play an important role in development and disease pathogenesis, while quiescent resident fibroblasts are responsible for sustaining tissue homeostasis. Fibroblastic activation is particularly evident in the tumor microenvironment where fibroblasts transition into tumor-supporting cancer-associated fibroblasts (CAFs), with some CAFs maintaining tumor-suppressive functions. While the tumor-supporting features of CAFs and their fibroblast-like precursors predominantly function through paracrine chemical communication (e.g., secretion of cytokine, chemokine, and more), the direct cell-cell communication that occurs between fibroblasts and other cells, and the effect that the remodeled CAF-generated interstitial extracellular matrix has in these types of cellular communications, remain poorly understood. Here, we explore the reported roles fibroblastic cell-cell communication play within the cancer stroma context and highlight insights we can gain from other disciplines.
Collapse
Affiliation(s)
- Jaye C Gardiner
- Cancer Signaling and Epigenetics Program, Marvin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Temple Health, Philadelphia, PA, United States
| | - Edna Cukierman
- Cancer Signaling and Epigenetics Program, Marvin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Temple Health, Philadelphia, PA, United States.
| |
Collapse
|
36
|
Hill KE, Lovett BM, Schwarzbauer JE. Heparan sulfate is necessary for the early formation of nascent fibronectin and collagen I fibrils at matrix assembly sites. J Biol Chem 2022; 298:101479. [PMID: 34890641 PMCID: PMC8801470 DOI: 10.1016/j.jbc.2021.101479] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 11/30/2022] Open
Abstract
Fibronectin (FN), an essential component of the extracellular matrix (ECM), is assembled via a cell-mediated process in which integrin receptors bind secreted FN and mediate its polymerization into fibrils that extend between cells, ultimately forming an insoluble matrix. Our previous work using mutant Chinese hamster ovary (CHO) cells identified the glycosaminoglycan heparan sulfate (HS) and its binding to FN as essential for the formation of insoluble FN fibrils. In this study, we investigated the contributions of HS at an early stage of the assembly process using knockdown of exostosin-1 (EXT1), one of the glycosyltransferases required for HS chain synthesis. NIH 3T3 fibroblasts with decreased EXT1 expression exhibited a significant reduction in both FN and type I collagen in the insoluble matrix. We show that FN fibril formation is initiated at matrix assembly sites, and while these sites were formed by cells with EXT1 knockdown, their growth was stunted compared with wild-type cells. The most severe defect observed was in the polymerization of nascent FN fibrils, which was reduced 2.5-fold upon EXT1 knockdown. This defect was rescued by the addition of exogenous soluble heparin chains long enough to simultaneously bind multiple FN molecules. The activity of soluble heparin in this process indicates that nascent fibril formation depends on HS more so than on the protein component of a specific HS proteoglycan. Together, our results suggest that heparin or HS is necessary for concentrating and localizing FN molecules at sites of early fibril assembly.
Collapse
Affiliation(s)
- Katherine E Hill
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Benjamin M Lovett
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Jean E Schwarzbauer
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA.
| |
Collapse
|
37
|
Keller CR, Hu Y, Ruud KF, VanDeen AE, Martinez SR, Kahn BT, Zhang Z, Chen RK, Li W. Human Breast Extracellular Matrix Microstructures and Protein Hydrogel 3D Cultures of Mammary Epithelial Cells. Cancers (Basel) 2021; 13:cancers13225857. [PMID: 34831010 PMCID: PMC8616054 DOI: 10.3390/cancers13225857] [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: 10/16/2021] [Revised: 11/10/2021] [Accepted: 11/17/2021] [Indexed: 01/21/2023] Open
Abstract
Simple Summary Human breast tissue extracellular matrix (ECM) is a microenvironment essential for the survival and biological activities of mammary epithelial cells. The ECM structural features of human breast tissues remain poorly defined. In this study, we identified the structural and mechanical properties of human normal breast and invasive ductal carcinoma tissue ECM using histological methods and atomic force microscopy. Additionally, a protein hydrogel was generated using human breast tissue ECM and defined for its microstructural features using immunofluorescence imaging and machine learning. Furthermore, we examined the three-dimensional growth of normal mammary epithelial cells or breast cancer cells cultured on the ECM protein hydrogel, where the cells exhibited biological phenotypes like those seen in native breast tissues. Our data provide novel insights into cancer cell biology, tissue microenvironment mimicry and engineering, and native tissue ECM-based biomedical and pharmaceutical applications. Abstract Tissue extracellular matrix (ECM) is a structurally and compositionally unique microenvironment within which native cells can perform their natural biological activities. Cells grown on artificial substrata differ biologically and phenotypically from those grown within their native tissue microenvironment. Studies examining human tissue ECM structures and the biology of human tissue cells in their corresponding tissue ECM are lacking. Such investigations will improve our understanding about human pathophysiological conditions for better clinical care. We report here human normal breast tissue and invasive ductal carcinoma tissue ECM structural features. For the first time, a hydrogel was successfully fabricated using whole protein extracts of human normal breast ECM. Using immunofluorescence staining of type I collagen (Col I) and machine learning of its fibrous patterns in the polymerized human breast ECM hydrogel, we have defined the microstructural characteristics of the hydrogel and compared the microstructures with those of other native ECM hydrogels. Importantly, the ECM hydrogel supported 3D growth and cell-ECM interaction of both normal and cancerous mammary epithelial cells. This work represents further advancement toward full reconstitution of the human breast tissue microenvironment, an accomplishment that will accelerate the use of human pathophysiological tissue-derived matrices for individualized biomedical research and therapeutic development.
Collapse
Affiliation(s)
- Chandler R. Keller
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA; (C.R.K.); (K.F.R.)
| | - Yang Hu
- Department of Crop and Soil Sciences, College of Agricultural, Human, and Natural Resources Sciences, Washington State University, Pullman, WA 99164, USA; (Y.H.); (Z.Z.)
| | - Kelsey F. Ruud
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA; (C.R.K.); (K.F.R.)
| | - Anika E. VanDeen
- School of Mechanical and Materials Engineering, Washington State University, Pullman, WA 99164, USA; (A.E.V.); (R.K.C.)
| | - Steve R. Martinez
- Department of Surgery, The Everett Clinic and Providence Regional Cancer Partnership, Everett, WA 98201, USA;
- Department of Medical Education and Clinical Sciences, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA
| | - Barry T. Kahn
- CellNetix Pathology & Laboratories, Seattle, WA 98104, USA;
- Providence Regional Medical Center, Everett, WA 98201, USA
| | - Zhiwu Zhang
- Department of Crop and Soil Sciences, College of Agricultural, Human, and Natural Resources Sciences, Washington State University, Pullman, WA 99164, USA; (Y.H.); (Z.Z.)
| | - Roland K. Chen
- School of Mechanical and Materials Engineering, Washington State University, Pullman, WA 99164, USA; (A.E.V.); (R.K.C.)
| | - Weimin Li
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA; (C.R.K.); (K.F.R.)
- Correspondence:
| |
Collapse
|
38
|
Basta MD, Paulson H, Walker JL. The local wound environment is a key determinant of the outcome of TGFβ signaling on the fibrotic response of CD44 + leader cells in an ex vivo post-cataract-surgery model. Exp Eye Res 2021; 213:108829. [PMID: 34774488 DOI: 10.1016/j.exer.2021.108829] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 10/22/2021] [Accepted: 11/03/2021] [Indexed: 01/17/2023]
Abstract
The cytokine transforming growth factor beta (TGFβ) has a role in regulating the normal and pathological response to wound healing, yet how it shifts from a pro-repair to a pro-fibrotic function within the wound environment is still unclear. Using a clinically relevant ex vivo post-cataract surgery model that mimics the lens fibrotic disease posterior capsule opacification (PCO), we investigated the influence of two distinct wound environments on shaping the TGFβ-mediated injury response of CD44+ vimentin-rich leader cells. The substantial fibrotic response of this cell population occurred within a rigid wound environment under the control of endogenous TGFβ. However, TGFβ was dispensable for the role of leader cells in wound healing on the endogenous basement membrane wound environment, where repair occurs in the absence of a major fibrotic outcome. A difference between leader cell function in these distinct environments was their cell surface expression of the latent TGFβ activator, αvβ3 integrin. This receptor is exclusively found on this CD44+ cell population when they localize to the leading edge of the rigid wound environment. Providing exogenous TGFβ to bypass any differences in the ability of the leader cells to sustain activation of TGFβ in different environments revealed their inherent ability to induce pro-fibrotic reactions on the basement membrane wound environment. Furthermore, exposure of the leader cells in the rigid wound environment to TGFβ led to an accelerated fibrotic response including the earlier appearance of pro-collagen + cells, alpha smooth muscle actin (αSMA)+ myofibroblasts, and increased fibrotic matrix production. Collectively, these findings show the influence of the local wound environment on the extent and severity of TGFβ-induced fibrotic responses. These findings have important implications for understanding the development of the lens fibrotic disease PCO in response to cataract surgery wounding.
Collapse
Affiliation(s)
- Morgan D Basta
- Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Heather Paulson
- Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Janice L Walker
- Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, 19107, USA; Department of Ophthalmology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, 19107, USA.
| |
Collapse
|
39
|
Rubí-Sans G, Nyga A, Rebollo E, Pérez-Amodio S, Otero J, Navajas D, Mateos-Timoneda MA, Engel E. Development of Cell-Derived Matrices for Three-Dimensional In Vitro Cancer Cell Models. ACS APPLIED MATERIALS & INTERFACES 2021; 13:44108-44123. [PMID: 34494824 DOI: 10.1021/acsami.1c13630] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Most morphogenetic and pathological processes are driven by cells responding to the surrounding matrix, such as its composition, architecture, and mechanical properties. Despite increasing evidence for the role of extracellular matrix (ECM) in tissue and disease development, many in vitro substitutes still fail to effectively mimic the native microenvironment. We established a novel method to produce macroscale (>1 cm) mesenchymal cell-derived matrices (CDMs) aimed to mimic the fibrotic tumor microenvironment surrounding epithelial cancer cells. CDMs are produced by human adipose mesenchymal stem cells cultured in sacrificial 3D scaffold templates of fibronectin-coated poly-lactic acid microcarriers (MCs) in the presence of macromolecular crowders. We showed that decellularized CDMs closely mimic the fibrillar protein composition, architecture, and mechanical properties of human fibrotic ECM from cancer masses. CDMs had highly reproducible composition made of collagen types I and III and fibronectin ECM with tunable mechanical properties. Moreover, decellularized and MC-free CDMs were successfully repopulated with cancer cells throughout their 3D structure, and following chemotherapeutic treatment, cancer cells showed greater doxorubicin resistance compared to 3D culture in collagen hydrogels. Collectively, these results support the use of CDMs as a reproducible and tunable tool for developing 3D in vitro cancer models.
Collapse
Affiliation(s)
- Gerard Rubí-Sans
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid 28029, Spain
| | - Agata Nyga
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
| | - Elena Rebollo
- Molecular Imaging Platform, Molecular Biology Institute of Barcelona (IBMB-CSIC), Barcelona 08028, Spain
| | - Soledad Pérez-Amodio
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid 28029, Spain
- IMEM-BRT group, Department of Materials Science, EEBE, Technical University of Catalonia (UPC), Barcelona 08019, Spain
| | - Jorge Otero
- Unitat Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona 08036, Spain
- CIBER de Enfermedades Respiratorias, Madrid 28029, Spain
| | - Daniel Navajas
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
- Unitat Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona 08036, Spain
- CIBER de Enfermedades Respiratorias, Madrid 28029, Spain
| | - Miguel A Mateos-Timoneda
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès (Barcelona) 08195, Spain
| | - Elisabeth Engel
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid 28029, Spain
- IMEM-BRT group, Department of Materials Science, EEBE, Technical University of Catalonia (UPC), Barcelona 08019, Spain
| |
Collapse
|
40
|
Dalton CJ, Lemmon CA. Fibronectin: Molecular Structure, Fibrillar Structure and Mechanochemical Signaling. Cells 2021; 10:2443. [PMID: 34572092 PMCID: PMC8471655 DOI: 10.3390/cells10092443] [Citation(s) in RCA: 138] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/09/2021] [Accepted: 09/10/2021] [Indexed: 12/12/2022] Open
Abstract
The extracellular matrix (ECM) plays a key role as both structural scaffold and regulator of cell signal transduction in tissues. In times of ECM assembly and turnover, cells upregulate assembly of the ECM protein, fibronectin (FN). FN is assembled by cells into viscoelastic fibrils that can bind upward of 40 distinct growth factors and cytokines. These fibrils play a key role in assembling a provisional ECM during embryonic development and wound healing. Fibril assembly is also often upregulated during disease states, including cancer and fibrotic diseases. FN fibrils have unique mechanical properties, which allow them to alter mechanotransduction signals sensed and relayed by cells. Binding of soluble growth factors to FN fibrils alters signal transduction from these proteins, while binding of other ECM proteins, including collagens, elastins, and proteoglycans, to FN fibrils facilitates the maturation and tissue specificity of the ECM. In this review, we will discuss the assembly of FN fibrils from individual FN molecules; the composition, structure, and mechanics of FN fibrils; the interaction of FN fibrils with other ECM proteins and growth factors; the role of FN in transmitting mechanobiology signaling events; and approaches for studying the mechanics of FN fibrils.
Collapse
Affiliation(s)
| | - Christopher A. Lemmon
- Department of Biomedical Engineering, Virginia Commonwealth University, 401 W. Main St., Richmond, VA 23284, USA;
| |
Collapse
|
41
|
Amaral DT, Johnson CH, Viviani VR. RNA-Seq analysis of the blue light-emitting Orfelia fultoni (Diptera: Keroplatidae) suggest photoecological adaptations at the molecular level. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2021; 39:100840. [PMID: 34022525 PMCID: PMC8495875 DOI: 10.1016/j.cbd.2021.100840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 04/16/2021] [Accepted: 04/19/2021] [Indexed: 11/21/2022]
Abstract
Bioluminescence in Diptera is found in the Keroplatidae family, within Arachnocampininae and Keroplatinae subfamilies, with reported occurrences in Oceania, Eurasia, and Americas. Larvae of Orfelia fultoni, which inhabit stream banks in the Appalachian Mountains, emit the bluest bioluminescence among insects, using it for prey attraction, similarly to Arachnocampa spp. Although bioluminescence has a similar prey attraction function, the systems of Arachonocampininae and Keroplatinae subfamilies are morphologically/biochemically distinct, indicating different evolutionary origins. To identify the possible coding genes associated with physiological control, ecological adaptations, and origin/evolution of bioluminescence in the Keroplatinae subfamily, we performed the RNA-Seq analysis of O. fultoni larvae during day and night and compared it with the transcriptomes of Arachnocampa luminosa, and reanalyzed the previously published proteomic data of O. fultoni against the RNA-Seq dataset. The abundance of chaperones/heat-shock and hexamerin gene products at night and in luciferase enriched fractions supports their possible association and participation in bioluminescence. The low diversity of copies/families of opsins indicate a simpler visual system in O. fultoni. Noteworthy, gene products associated with silk protein biosynthesis in Orfelia were more similar to Lepidoptera than to the Arachnocampa, indicating that, similarly to the bioluminescent systems, at some point, the biochemical apparatus for web construction may have evolved independently in Orfelia and Arachnocampa.
Collapse
Affiliation(s)
- Danilo T Amaral
- Graduate School of Biotechnology and Environmental Monitoring (UFSCar), Sorocaba, SP, Brazil
| | - Carl H Johnson
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37235, USA
| | - Vadim R Viviani
- Graduate School of Biotechnology and Environmental Monitoring (UFSCar), Sorocaba, SP, Brazil; Graduate School of Evolutive Genetics and Molecular Biology, Federal Univ. São Carlos (UFSCar), São Carlos, SP, Brazil.
| |
Collapse
|
42
|
Spada S, Tocci A, Di Modugno F, Nisticò P. Fibronectin as a multiregulatory molecule crucial in tumor matrisome: from structural and functional features to clinical practice in oncology. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:102. [PMID: 33731188 PMCID: PMC7972229 DOI: 10.1186/s13046-021-01908-8] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 03/09/2021] [Indexed: 12/11/2022]
Abstract
Deciphering extracellular matrix (ECM) composition and architecture may represent a novel approach to identify diagnostic and therapeutic targets in cancer. Among the ECM components, fibronectin and its fibrillary assembly represent the scaffold to build up the entire ECM structure, deeply affecting its features. Herein we focus on this extraordinary protein starting from its complex structure and defining its role in cancer as prognostic and theranostic marker.
Collapse
Affiliation(s)
- Sheila Spada
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Annalisa Tocci
- Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Francesca Di Modugno
- Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, Rome, Italy.
| | - Paola Nisticò
- Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, Rome, Italy.
| |
Collapse
|
43
|
Musiime M, Chang J, Hansen U, Kadler KE, Zeltz C, Gullberg D. Collagen Assembly at the Cell Surface: Dogmas Revisited. Cells 2021; 10:662. [PMID: 33809734 PMCID: PMC8002325 DOI: 10.3390/cells10030662] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/08/2021] [Accepted: 03/12/2021] [Indexed: 12/12/2022] Open
Abstract
With the increased awareness about the importance of the composition, organization, and stiffness of the extracellular matrix (ECM) for tissue homeostasis, there is a renewed need to understand the details of how cells recognize, assemble and remodel the ECM during dynamic tissue reorganization events. Fibronectin (FN) and fibrillar collagens are major proteins in the ECM of interstitial matrices. Whereas FN is abundant in cell culture studies, it is often only transiently expressed in the acute phase of wound healing and tissue regeneration, by contrast fibrillar collagens form a persistent robust scaffold in healing and regenerating tissues. Historically fibrillar collagens in interstitial matrices were seen merely as structural building blocks. Cell anchorage to the collagen matrix was thought to be indirect and occurring via proteins like FN and cell surface-mediated collagen fibrillogenesis was believed to require a FN matrix. The isolation of four collagen-binding integrins have challenged this dogma, and we now know that cells anchor directly to monomeric forms of fibrillar collagens via the α1β1, α2β1, α10β1 and α11β1 integrins. The binding of these integrins to the mature fibrous collagen matrices is more controversial and depends on availability of integrin-binding sites. With increased awareness about the importance of characterizing the total integrin repertoire on cells, including the integrin collagen receptors, the idea of an absolute dependence on FN for cell-mediated collagen fibrillogenesis needs to be re-evaluated. We will summarize data suggesting that collagen-binding integrins in vitro and in vivo are perfectly well suited for nucleating and supporting collagen fibrillogenesis, independent of FN.
Collapse
Affiliation(s)
- Moses Musiime
- Department of Biomedicine and Centre for Cancer Biomarkers, University of Bergen, Jonas Lies vei 91, N-5009 Bergen, Norway; (M.M.); (C.Z.)
| | - Joan Chang
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, UK; (J.C.); (K.E.K.)
| | - Uwe Hansen
- Institute for Musculoskeletal Medicine, University Hospital of Münster, 48149 Münster, Germany;
| | - Karl E. Kadler
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, UK; (J.C.); (K.E.K.)
| | - Cédric Zeltz
- Department of Biomedicine and Centre for Cancer Biomarkers, University of Bergen, Jonas Lies vei 91, N-5009 Bergen, Norway; (M.M.); (C.Z.)
| | - Donald Gullberg
- Department of Biomedicine and Centre for Cancer Biomarkers, University of Bergen, Jonas Lies vei 91, N-5009 Bergen, Norway; (M.M.); (C.Z.)
| |
Collapse
|
44
|
Hardcastle AJ, Liskova P, Bykhovskaya Y, McComish BJ, Davidson AE, Inglehearn CF, Li X, Choquet H, Habeeb M, Lucas SEM, Sahebjada S, Pontikos N, Lopez KER, Khawaja AP, Ali M, Dudakova L, Skalicka P, Van Dooren BTH, Geerards AJM, Haudum CW, Faro VL, Tenen A, Simcoe MJ, Patasova K, Yarrand D, Yin J, Siddiqui S, Rice A, Farraj LA, Chen YDI, Rahi JS, Krauss RM, Theusch E, Charlesworth JC, Szczotka-Flynn L, Toomes C, Meester-Smoor MA, Richardson AJ, Mitchell PA, Taylor KD, Melles RB, Aldave AJ, Mills RA, Cao K, Chan E, Daniell MD, Wang JJ, Rotter JI, Hewitt AW, MacGregor S, Klaver CCW, Ramdas WD, Craig JE, Iyengar SK, O'Brart D, Jorgenson E, Baird PN, Rabinowitz YS, Burdon KP, Hammond CJ, Tuft SJ, Hysi PG. A multi-ethnic genome-wide association study implicates collagen matrix integrity and cell differentiation pathways in keratoconus. Commun Biol 2021; 4:266. [PMID: 33649486 PMCID: PMC7921564 DOI: 10.1038/s42003-021-01784-0] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 02/02/2021] [Indexed: 12/14/2022] Open
Abstract
Keratoconus is characterised by reduced rigidity of the cornea with distortion and focal thinning that causes blurred vision, however, the pathogenetic mechanisms are unknown. It can lead to severe visual morbidity in children and young adults and is a common indication for corneal transplantation worldwide. Here we report the first large scale genome-wide association study of keratoconus including 4,669 cases and 116,547 controls. We have identified significant association with 36 genomic loci that, for the first time, implicate both dysregulation of corneal collagen matrix integrity and cell differentiation pathways as primary disease-causing mechanisms. The results also suggest pleiotropy, with some disease mechanisms shared with other corneal diseases, such as Fuchs endothelial corneal dystrophy. The common variants associated with keratoconus explain 12.5% of the genetic variance, which shows potential for the future development of a diagnostic test to detect susceptibility to disease.
Collapse
Affiliation(s)
- Alison J Hardcastle
- UCL Institute of Ophthalmology, London, UK.
- Moorfields Eye Hospital, NHS Foundation Trust, London, UK.
| | - Petra Liskova
- UCL Institute of Ophthalmology, London, UK
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
- Department of Ophthalmology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Yelena Bykhovskaya
- The Cornea Eye Institute, Beverly Hills, CA, USA
- Department of Surgery and Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Bennet J McComish
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | | | - Chris F Inglehearn
- Division of Molecular Medicine, Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | - Xiaohui Li
- Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation (formerly Los Angeles Biomedical Research Institute) at Harbor-UCLA Medical Center; Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Hélène Choquet
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Mahmoud Habeeb
- Department of Ophthalmology, Erasmus Medical Center GD, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center GD, Rotterdam, The Netherlands
| | - Sionne E M Lucas
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Srujana Sahebjada
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, Australia
- Department of Surgery, Ophthalmology, University of Melbourne, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, Australia
| | | | | | - Anthony P Khawaja
- UCL Institute of Ophthalmology, London, UK
- Moorfields Eye Hospital, NHS Foundation Trust, London, UK
- NIHR Biomedical Research Centre, Moorfields Eye Hospital, London, UK
| | - Manir Ali
- Division of Molecular Medicine, Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | - Lubica Dudakova
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Pavlina Skalicka
- Department of Ophthalmology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Bart T H Van Dooren
- Department of Ophthalmology, Erasmus Medical Center GD, Rotterdam, The Netherlands
- Amphia Hospital, Breda, The Netherlands
| | | | - Christoph W Haudum
- Division of Endocrinology and Diabetology, Endocrinology Lab Platform, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Valeria Lo Faro
- Department of Ophthalmology, University Medical Center Groningen (UMCG), Groningen, the Netherlands
- Department of Ophthalmology, Academic Medical Center, Amsterdam, The Netherlands
| | - Abi Tenen
- Vision Eye Institute, Melbourne, VIC, Australia
- School of Primary and Allied Health Care, Monash University, Melbourne, VIC, Australia
- Melbourne Stem Cell Centre, Melbourne, VIC, 3800, Australia
| | - Mark J Simcoe
- Section of Ophthalmology, School of Life Course Sciences, King's College London, London, UK
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Karina Patasova
- Section of Ophthalmology, School of Life Course Sciences, King's College London, London, UK
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Darioush Yarrand
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Jie Yin
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Salina Siddiqui
- Division of Molecular Medicine, Leeds Institute of Medical Research, University of Leeds, Leeds, UK
- Department of Ophthalmology, St James's University Hospital, Leeds, UK
| | - Aine Rice
- Division of Molecular Medicine, Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | - Layal Abi Farraj
- Division of Molecular Medicine, Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | - Yii-Der Ida Chen
- Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation (formerly Los Angeles Biomedical Research Institute) at Harbor-UCLA Medical Center; Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Jugnoo S Rahi
- UCL Great Ormond Street Hospital Institute of Child Health, London, UK
| | | | | | - Jac C Charlesworth
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | | | - Carmel Toomes
- Division of Molecular Medicine, Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | - Magda A Meester-Smoor
- Department of Ophthalmology, Erasmus Medical Center GD, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center GD, Rotterdam, The Netherlands
| | - Andrea J Richardson
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, Australia
| | - Paul A Mitchell
- Centre for Vision Research, Department of Ophthalmology, Westmead Institute for Medical Research, University of Sydney, Westmead, NSW, Australia
| | - Kent D Taylor
- Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation (formerly Los Angeles Biomedical Research Institute) at Harbor-UCLA Medical Center; Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Ronald B Melles
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Anthony J Aldave
- The Jules Stein Institute, University of California Los Angeles, Los Angeles, CA, USA
| | - Richard A Mills
- Department of Ophthalmology, Flinders University, Adelaide, SA, Australia
| | - Ke Cao
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, Australia
- Department of Surgery, Ophthalmology, University of Melbourne, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, Australia
| | - Elsie Chan
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, Australia
- Department of Surgery, Ophthalmology, University of Melbourne, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, Australia
| | - Mark D Daniell
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, Australia
- Department of Surgery, Ophthalmology, University of Melbourne, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, Australia
| | - Jie Jin Wang
- Health Services and Systems Research, Duke-NUS Medical School, Singapore, Singapore
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation (formerly Los Angeles Biomedical Research Institute) at Harbor-UCLA Medical Center; Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Alex W Hewitt
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
- Vision Eye Institute, Melbourne, VIC, Australia
- School of Primary and Allied Health Care, Monash University, Melbourne, VIC, Australia
- Melbourne Stem Cell Centre, Melbourne, VIC, 3800, Australia
| | - Stuart MacGregor
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Caroline C W Klaver
- Department of Ophthalmology, Erasmus Medical Center GD, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center GD, Rotterdam, The Netherlands
| | - Wishal D Ramdas
- Department of Ophthalmology, Erasmus Medical Center GD, Rotterdam, The Netherlands
| | - Jamie E Craig
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
- Department of Ophthalmology, Flinders University, Adelaide, SA, Australia
| | - Sudha K Iyengar
- Department of Ophthalmology, Case Western Reserve University, Cleveland, OH, USA
| | - David O'Brart
- Section of Ophthalmology, School of Life Course Sciences, King's College London, London, UK
- St Thomas Hospital, Guy's and St. Thomas NHS Trust, London, London, UK
| | - Eric Jorgenson
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Paul N Baird
- Department of Surgery, Ophthalmology, University of Melbourne, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, Australia
| | - Yaron S Rabinowitz
- The Cornea Eye Institute, Beverly Hills, CA, USA
- Department of Surgery and Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Kathryn P Burdon
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
- Department of Ophthalmology, Flinders University, Adelaide, SA, Australia
| | - Chris J Hammond
- Section of Ophthalmology, School of Life Course Sciences, King's College London, London, UK
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
- St Thomas Hospital, Guy's and St. Thomas NHS Trust, London, London, UK
| | - Stephen J Tuft
- UCL Institute of Ophthalmology, London, UK.
- Moorfields Eye Hospital, NHS Foundation Trust, London, UK.
| | - Pirro G Hysi
- Section of Ophthalmology, School of Life Course Sciences, King's College London, London, UK.
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK.
- UCL Great Ormond Street Hospital Institute of Child Health, London, UK.
| |
Collapse
|
45
|
Efthymiou G, Radwanska A, Grapa AI, Beghelli-de la Forest Divonne S, Grall D, Schaub S, Hattab M, Pisano S, Poet M, Pisani DF, Counillon L, Descombes X, Blanc-Féraud L, Van Obberghen-Schilling E. Fibronectin Extra Domains tune cellular responses and confer topographically distinct features to fibril networks. J Cell Sci 2021; 134:jcs.252957. [PMID: 33526715 DOI: 10.1242/jcs.252957] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 01/15/2021] [Indexed: 12/25/2022] Open
Abstract
Cellular fibronectin (FN; also known as FN1) variants harboring one or two alternatively spliced so-called extra domains (EDB and EDA) play a central bioregulatory role during development, repair processes and fibrosis. Yet, how the extra domains impact fibrillar assembly and function of the molecule remains unclear. Leveraging a unique biological toolset and image analysis pipeline for direct comparison of the variants, we demonstrate that the presence of one or both extra domains impacts FN assembly, function and physical properties of the matrix. When presented to FN-null fibroblasts, extra domain-containing variants differentially regulate pH homeostasis, survival and TGF-β signaling by tuning the magnitude of cellular responses, rather than triggering independent molecular switches. Numerical analyses of fiber topologies highlight significant differences in variant-specific structural features and provide a first step for the development of a generative model of FN networks to unravel assembly mechanisms and investigate the physical and functional versatility of extracellular matrix landscapes.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
| | - Agata Radwanska
- Université Côte d'Azur, CNRS, INSERM, iBV, Nice 06108, France
| | - Anca-Ioana Grapa
- Université Côte d'Azur, CNRS, INSERM, iBV, Nice 06108, France.,Université Côte d'Azur, Inria, CNRS, i3S, Nice 06902, France
| | | | - Dominique Grall
- Université Côte d'Azur, CNRS, INSERM, iBV, Nice 06108, France
| | | | - Maurice Hattab
- Université Côte d'Azur, CNRS, INSERM, iBV, Nice 06108, France
| | - Sabrina Pisano
- Université Côte d'Azur, Inserm, CNRS, IRCAN, Nice 06107, France
| | - Mallorie Poet
- Université Côte d'Azur, Inserm, CNRS, IRCAN, Nice 06107, France
| | | | | | | | | | | |
Collapse
|
46
|
Garrison CM, Schwarzbauer JE. Fibronectin fibril alignment is established upon initiation of extracellular matrix assembly. Mol Biol Cell 2021; 32:739-752. [PMID: 33625865 PMCID: PMC8108514 DOI: 10.1091/mbc.e20-08-0533] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The physical structure of the extracellular matrix (ECM) is tissue-specific and fundamental to normal tissue function. Proper alignment of ECM fibers is essential for the functioning of a variety of tissues. While matrix assembly in general has been intensively investigated, little is known about the mechanisms required for formation of aligned ECM fibrils. We investigated the initiation of fibronectin (FN) matrix assembly using fibroblasts that assemble parallel ECM fibrils and found that matrix assembly sites, where FN fibrillogenesis is initiated, were oriented in parallel at the cell poles. We show that these polarized matrix assembly sites progress into fibrillar adhesions and ultimately into aligned FN fibrils. Cells that assemble an unaligned meshwork matrix form matrix assembly sites around the cell periphery, but the distribution of matrix assembly sites in these cells could be modulated through micropatterning or mechanical stretch. While an elongated cell shape corresponds with a polarized matrix assembly site distribution, these two features are not absolutely linked, since we discovered that transforming growth factor beta (TGF-β1) enhances matrix assembly site polarity and assembly of aligned fibrils independent of cell elongation. We conclude that the ultimate orientation of FN fibrils is determined by the alignment and distribution of matrix assembly sites that form during the initial stages of cell–FN interactions.
Collapse
Affiliation(s)
- Carly M Garrison
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| | | |
Collapse
|
47
|
Ouellette JN, Drifka CR, Pointer KB, Liu Y, Lieberthal TJ, Kao WJ, Kuo JS, Loeffler AG, Eliceiri KW. Navigating the Collagen Jungle: The Biomedical Potential of Fiber Organization in Cancer. Bioengineering (Basel) 2021; 8:17. [PMID: 33494220 PMCID: PMC7909776 DOI: 10.3390/bioengineering8020017] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 01/10/2021] [Accepted: 01/13/2021] [Indexed: 02/07/2023] Open
Abstract
Recent research has highlighted the importance of key tumor microenvironment features, notably the collagen-rich extracellular matrix (ECM) in characterizing tumor invasion and progression. This led to great interest from both basic researchers and clinicians, including pathologists, to include collagen fiber evaluation as part of the investigation of cancer development and progression. Fibrillar collagen is the most abundant in the normal extracellular matrix, and was revealed to be upregulated in many cancers. Recent studies suggested an emerging theme across multiple cancer types in which specific collagen fiber organization patterns differ between benign and malignant tissue and also appear to be associated with disease stage, prognosis, treatment response, and other clinical features. There is great potential for developing image-based collagen fiber biomarkers for clinical applications, but its adoption in standard clinical practice is dependent on further translational and clinical evaluations. Here, we offer a comprehensive review of the current literature of fibrillar collagen structure and organization as a candidate cancer biomarker, and new perspectives on the challenges and next steps for researchers and clinicians seeking to exploit this information in biomedical research and clinical workflows.
Collapse
Affiliation(s)
- Jonathan N. Ouellette
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA; (J.N.O.); (C.R.D.); (T.J.L.); (W.J.K.)
- Laboratory for Optical and Computational Instrumentation, Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, WI 53706, USA; (K.B.P.); (Y.L.)
| | - Cole R. Drifka
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA; (J.N.O.); (C.R.D.); (T.J.L.); (W.J.K.)
- Laboratory for Optical and Computational Instrumentation, Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, WI 53706, USA; (K.B.P.); (Y.L.)
| | - Kelli B. Pointer
- Laboratory for Optical and Computational Instrumentation, Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, WI 53706, USA; (K.B.P.); (Y.L.)
| | - Yuming Liu
- Laboratory for Optical and Computational Instrumentation, Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, WI 53706, USA; (K.B.P.); (Y.L.)
| | - Tyler J Lieberthal
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA; (J.N.O.); (C.R.D.); (T.J.L.); (W.J.K.)
| | - W John Kao
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA; (J.N.O.); (C.R.D.); (T.J.L.); (W.J.K.)
- Department of Industrial and Manufacturing Systems Engineering, Faculty of Engineering, University of Hong Kong, Pokfulam, Hong Kong
| | - John S. Kuo
- Department of Neurosurgery, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA;
| | - Agnes G. Loeffler
- Department of Pathology, MetroHealth Medical Center, Cleveland, OH 44109, USA;
| | - Kevin W. Eliceiri
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA; (J.N.O.); (C.R.D.); (T.J.L.); (W.J.K.)
- Laboratory for Optical and Computational Instrumentation, Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, WI 53706, USA; (K.B.P.); (Y.L.)
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI 53705, USA
- Morgridge Institute for Research, Madison, WI 53715, USA
| |
Collapse
|
48
|
The extracellular matrix: A key player in the pathogenesis of hematologic malignancies. Blood Rev 2020; 48:100787. [PMID: 33317863 DOI: 10.1016/j.blre.2020.100787] [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] [Received: 06/07/2020] [Revised: 09/10/2020] [Accepted: 11/05/2020] [Indexed: 12/26/2022]
Abstract
Hematopoietic stem and progenitor cells located in the bone marrow lay the foundation for multiple lineages of mature hematologic cells. Bone marrow niches are architecturally complex with specific cellular, physiochemical, and biomechanical factors. Increasing evidence suggests that the bone marrow microenvironment contributes to the pathogenesis of hematological neoplasms. Numerous studies have deciphered the role of genetic mutations and chromosomal translocations in the development hematologic malignancies. Significant progress has also been made in understanding how the cellular components and cytokine interactions within the bone marrow microenvironment promote the evolution of hematologic cancers. Although the extracellular matrix is known to be a key player in the pathogenesis of various diseases, it's role in the progression of hematologic malignancies is less understood. In this review, we discuss the interactions between the extracellular matrix and malignant cells, and provide an overview of the role of extracellular matrix remodeling in sustaining hematologic malignancies.
Collapse
|
49
|
Sarkar S, Saha P, Seth RK, Mondal A, Bose D, Kimono D, Albadrani M, Mukherjee A, Porter DE, Scott GI, Xiao S, Brooks B, Ferry J, Nagarkatti M, Nagarkatti P, Chatterjee S. Higher intestinal and circulatory lactate associated NOX2 activation leads to an ectopic fibrotic pathology following microcystin co-exposure in murine fatty liver disease. Comp Biochem Physiol C Toxicol Pharmacol 2020; 238:108854. [PMID: 32781293 PMCID: PMC7541568 DOI: 10.1016/j.cbpc.2020.108854] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 06/14/2020] [Accepted: 07/03/2020] [Indexed: 12/11/2022]
Abstract
Clinical studies implicated an increased risk of intestinal fibrosis in patients with nonalcoholic fatty liver disease (NAFLD). Our previous studies have shown that microcystin-LR (MC-LR) exposure led to altered gut microbiome and increased abundance of lactate producing bacteria and intestinal inflammation in underlying NAFLD. This led us to further investigate the effects of the MC-LR, a PP2A inhibitor in activating the TGF-β fibrotic pathway in the intestines that might be mediated by increased lactate induced redox enzyme NOX2. Exposure to MC-LR led to higher lactate levels in circulation and in the intestinal content. The higher lactate levels were associated with NOX2 activation in vivo that led to increased Smad2/3-Smad4 co-localization and high alpha-smooth muscle actin (α-SMA) immunoreactivity in the intestines. Mechanistically, primary mouse intestinal epithelial cells treated with lactate and MC-LR separately led to higher NOX2 activation, phosphorylation of TGFβR1 receptor and subsequent Smad 2/3-Smad4 co-localization inhibitable by apocynin (NOX2 inhibitor), FBA (a peroxynitrite scavenger) and DMPO (a nitrone spin trap), catalase and superoxide dismutase. Inhibition of NOX2-induced redox signaling also showed a significant decrease in collagen protein thus suggesting a strong redox signaling induced activation of an ectopic fibrotic manifestation in the intestines. In conclusion, the present study provides mechanistic insight into the role of microcystin in dysbiosis-linked lactate production and subsequently advances our knowledge in lactate-induced NOX2 exacerbation of the cell differentiation and fibrosis in the NAFLD intestines.
Collapse
Affiliation(s)
- Sutapa Sarkar
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, USA
| | - Punnag Saha
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, USA
| | - Ratanesh K Seth
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, USA
| | - Ayan Mondal
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, USA
| | - Dipro Bose
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, USA
| | - Diana Kimono
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, USA
| | - Muayad Albadrani
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, USA
| | | | - Dwayne E Porter
- NIEHS Center for Oceans and Human Health on Climate Change Interactions, Department of Environmental Health Sciences, University of South Carolina, USA
| | - Geoff I Scott
- NIEHS Center for Oceans and Human Health on Climate Change Interactions, Department of Environmental Health Sciences, University of South Carolina, USA
| | - Shuo Xiao
- NIEHS Center for Oceans and Human Health on Climate Change Interactions, Department of Environmental Health Sciences, University of South Carolina, USA
| | - Bryan Brooks
- Department of Environmental Science, Baylor University, USA
| | - John Ferry
- Department of Chemistry and Biochemistry, University of South Carolina, USA
| | - Mitzi Nagarkatti
- Pathology, Microbiology and Immunology(,) University of South Carolina School of Medicine, USA
| | - Prakash Nagarkatti
- Pathology, Microbiology and Immunology(,) University of South Carolina School of Medicine, USA
| | - Saurabh Chatterjee
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, USA.
| |
Collapse
|
50
|
Wu C, Zeng MH, Liao G, Qian K, Li H. Neuropilin-1 Interacts with Fibronectin-1 to Promote Epithelial-Mesenchymal Transition Progress in Gastric Cancer. Onco Targets Ther 2020; 13:10677-10687. [PMID: 33116644 PMCID: PMC7585825 DOI: 10.2147/ott.s275327] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 09/28/2020] [Indexed: 12/15/2022] Open
Abstract
Introduction Neuropilin-1 (NRP1) binds to many ligands and co-receptors and affects cell survival and migration, which is essential for tumor progression. However, there are still largely unknowns about how NRP1 affects the epithelial-mesenchymal transition (EMT)-related malignant progression in gastric cancer. Methods We used TCGA to analyze the expression of NRP1 in gastric cancer and its impact on patient survival. In in vitro experiments, transwell, wound healing and colony formation assays were used to evaluate the effects of NRP1 and ginsenoside Rg3 on the invasion, migration and proliferation of gastric cancer cells. In in vivo experiments, we evaluated the overexpression and knockdown of NRP1 and the effect of ginsenoside Rg3 on tumor growth. Results We found that NRP1 is highly expressed in advanced gastric cancer and associated with poor prognosis. Knockdown of NRP1 expression can inhibit the proliferation and metastasis of gastric cancer cells. Mechanically. NRP1 interacts with fibronectin-1 (FN1) to promote the malignant progression of gastric cancer cells through ECM remodeling. In addition, we found that ginsenoside Rg3 can block the interaction of NRP1 and FN1 and inhibit the progression of gastric cancer. Conclusion Our study suggested that the interaction of NRP1 and FN1 is crucial for the malignant progression of gastric cancer. This may provide a new perspective and potential treatment methods for the treatment of gastric cancer.
Collapse
Affiliation(s)
- Chao Wu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Meng-Hua Zeng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Gang Liao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Kun Qian
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Hui Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, People's Republic of China
| |
Collapse
|