1
|
Resnikoff HA, Schwarzbauer JE. Increased basal fibronectin is sufficient to promote excess endothelial cell matrix assembly causing localized barrier dysfunction. Mol Biol Cell 2024; 35:ar120. [PMID: 39046775 PMCID: PMC11449387 DOI: 10.1091/mbc.e24-02-0090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/25/2024] Open
Abstract
Endothelial cell behavior is regulated by subendothelial extracellular matrix (ECM). The ECM protein fibronectin (FN) is rare in healthy blood vessels but accumulates in disease accompanied by endothelial dysfunctions. Here, we report that excess assembly of FN disrupts key endothelial functions. We mimicked increased FN expression as in diseased stroma by providing exogenous FN basally in a Transwell insert and found dose-dependent upregulation of subendothelial FN matrix assembly. Taking advantage of discontinuous matrix assembly by endothelial cells, we show correlations between regional increases in FN matrix and disruptions in endothelial cell morphology, VE-cadherin junctions, and the cell cycle, all of which were not changed in FN-deficient regions of the monolayer. These changes affected endothelial barrier function with increased monolayer permeability exposing basal regions of high FN matrix and permitting FN-dependent adhesion of MDA-MB-231 tumor cells from the apical side of the monolayer. FN matrix accumulation was quick and increases in FN matrix preceded all other changes in the endothelium. Therefore, subendothelial accumulation of FN matrix is a cause, not an effect, of endothelial monolayer disorganization and leakiness. Regulating FN accumulation in the subendothelial space could be an important target for controlling progression of fibrosis and related diseases.
Collapse
Affiliation(s)
- Henry A. Resnikoff
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| | | |
Collapse
|
2
|
Wilkie IC. Basement Membranes, Brittlestar Tendons, and Their Mechanical Adaptability. BIOLOGY 2024; 13:375. [PMID: 38927255 PMCID: PMC11200632 DOI: 10.3390/biology13060375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 05/20/2024] [Accepted: 05/21/2024] [Indexed: 06/28/2024]
Abstract
Basement membranes (BMs) are thin layers of extracellular matrix that separate epithelia, endothelia, muscle cells, and nerve cells from adjacent interstitial connective tissue. BMs are ubiquitous in almost all multicellular animals, and their composition is highly conserved across the Metazoa. There is increasing interest in the mechanical functioning of BMs, including the involvement of altered BM stiffness in development and pathology, particularly cancer metastasis, which can be facilitated by BM destabilization. Such BM weakening has been assumed to occur primarily through enzymatic degradation by matrix metalloproteinases. However, emerging evidence indicates that non-enzymatic mechanisms may also contribute. In brittlestars (Echinodermata, Ophiuroidea), the tendons linking the musculature to the endoskeleton consist of extensions of muscle cell BMs. During the process of brittlestar autotomy, in which arms are detached for the purpose of self-defense, muscles break away from the endoskeleton as a consequence of the rapid destabilization and rupture of their BM-derived tendons. This contribution provides a broad overview of current knowledge of the structural organization and biomechanics of non-echinoderm BMs, compares this with the equivalent information on brittlestar tendons, and discusses the possible relationship between the weakening phenomena exhibited by BMs and brittlestar tendons, and the potential translational value of the latter as a model system of BM destabilization.
Collapse
Affiliation(s)
- Iain C Wilkie
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow G12 8QQ, UK
| |
Collapse
|
3
|
Schoenenberger MS, Halfter W, Ferrand A, Halfter K, Tzankov A, Scholl HPN, Henrich PB, Monnier CA. The biophysical and compositional properties of human basement membranes. FEBS J 2024; 291:477-488. [PMID: 37984833 DOI: 10.1111/febs.17007] [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: 04/03/2023] [Revised: 10/14/2023] [Accepted: 11/16/2023] [Indexed: 11/22/2023]
Abstract
Basement membranes are among the most widespread, non-cellular functional materials in metazoan organisms. Despite this ubiquity, the links between their compositional and biophysical properties are often difficult to establish due to their thin and delicate nature. In this article, we examine these features on a molecular level by combining results from proteomics, elastic, and nanomechanical analyses across a selection of human basement membranes. Comparing results between these different membranes connects certain compositional attributes to distinct nanomechanical signatures and further demonstrates to what extent water defines these properties. In all, these data underline BMs as stiff yet highly elastic connective tissue layers and highlight how the interplay between composition, mechanics and hydration yields such exceptionally adaptable materials.
Collapse
Affiliation(s)
| | - Willi Halfter
- Department of Ophthalmology, University of Basel, Switzerland
| | - Alexia Ferrand
- Imaging Core Facility, Biozentrum of the University of Basel, Switzerland
| | - Kathrin Halfter
- Munich Cancer Registry, Institute of Medical Informatics, Biometry and Epidemiology, Maximilian University Munich, Germany
| | - Alexandar Tzankov
- Histopathology and Autopsy, Institute of Medical Genetics and Pathology, University Hospital and University of Basel, Switzerland
| | - Hendrik P N Scholl
- Department of Ophthalmology, University of Basel, Switzerland
- Institute of Molecular and Clinical Ophthalmology Basel (IOB), Switzerland
| | - Paul Bernhard Henrich
- Department of Ophthalmology, University of Basel, Switzerland
- Università della Svizzera Italiana, Lugano, Switzerland
| | | |
Collapse
|
4
|
Chavda ND, Sari B, Asiri FM, Hamill KJ. Laminin N-terminus (LaNt) proteins, laminins and basement membrane regulation. Biochem Soc Trans 2022; 50:1541-1553. [PMID: 36355367 PMCID: PMC9788559 DOI: 10.1042/bst20210240] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/21/2022] [Accepted: 10/24/2022] [Indexed: 10/03/2023]
Abstract
Basement membranes (BMs) are structured regions of the extracellular matrix that provide multiple functions including physical support and acting as a barrier, as a repository for nutrients and growth factors, and as biophysical signalling hubs. At the core of all BMs is the laminin (LM) family of proteins. These large heterotrimeric glycoproteins are essential for tissue integrity, and differences between LM family members represent a key nexus in dictating context and tissue-specific functions. These variations reflect genetic diversity within the family, which allows for multiple structurally and functionally distinct heterotrimers to be produced, each with different architectures and affinities for other matrix proteins and cell surface receptors. The ratios of these LM isoforms also influence the biophysical properties of a BM owing to differences in their relative ability to form polymers or networks. Intriguingly, the LM superfamily is further diversified through the related netrin family of proteins and through alternative splicing leading to the generation of non-LM short proteins known as the laminin N-terminus (LaNt) domain proteins. Both the netrins and LaNt proteins contain structural domains involved in LM-to-LM interaction and network assembly. Emerging findings indicate that one netrin and at least one LaNt protein can potently influence the structure and function of BMs, disrupting the networks, changing physical properties, and thereby influencing tissue function. These findings are altering the way that we think about LM polymerisation and, in the case of the LaNt proteins, suggest a hitherto unappreciated form of LM self-regulation.
Collapse
Affiliation(s)
- Natasha D. Chavda
- Institute of Life Course and Medical Sciences, University of Liverpool, 6 West Derby Street, Liverpool L78TX, U.K
| | - Bilge Sari
- Institute of Life Course and Medical Sciences, University of Liverpool, 6 West Derby Street, Liverpool L78TX, U.K
| | - Fawziah M. Asiri
- Institute of Life Course and Medical Sciences, University of Liverpool, 6 West Derby Street, Liverpool L78TX, U.K
| | - Kevin J. Hamill
- Institute of Life Course and Medical Sciences, University of Liverpool, 6 West Derby Street, Liverpool L78TX, U.K
| |
Collapse
|
5
|
Tao J, Li X, Liang C, Liu Y, Zhou J. Expression of basement membrane genes and their prognostic significance in clear cell renal cell carcinoma patients. Front Oncol 2022; 12:1026331. [PMID: 36353536 PMCID: PMC9637577 DOI: 10.3389/fonc.2022.1026331] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/06/2022] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC) is a malignant tumor with limited treatment options. A recent study confirmed the involvement of basement membrane (BM) genes in the progression of many cancers. Therefore, we studied the role and prognostic significance of BM genes in ccRCC. METHODS Co-expression analysis of ccRCC-related information deposited in The Cancer Genome Atlas database and a BM geneset from a recent study was conducted. The differentially expressed BM genes were validated using quantitative reverse-transcription polymerase chain reaction (qRT-PCR). Least absolute shrinkage and selection operator regression and univariate Cox regression analyses were performed to identify a BM gene signature with prognostic significance for ccRCC. Multivariate Cox regression, time-dependent receiver operating characteristic, Kaplan-Meier, and nomogram analyses were implemented to appraise the prognostic ability of the signature and the findings were further verified using a Gene Expression Omnibus dataset. Additionally, immune cell infiltration and and pathway enrichment analyses were performed using ImmuCellAI and Gene Set Enrichment Analysis (GSEA), respectively. Finally, the DSIGDB dataset was used to screen small-molecule therapeutic drugs that may be useful in treating ccRCC patients. RESULTS We identified 108 BM genes exhibiting different expression levels compared to that in normal kidney tissues, among which 32 genes had prognostic values. The qRT-PCR analyses confirmed that the expression patterns of four of the ten selected genes were the same as the predicted ones. Additionally, we successfully established and validated a ccRCC patient prediction model based on 16 BM genes and observed that the model function is an independent predictor. GSEA revealed that differentially expressed BM genes mainly displayed significant enrichment of tumor and metabolic signaling cascades. The BM gene signature was also associated with immune cell infiltration and checkpoints. Eight small-molecule drugs may have therapeutic effects on ccRCC patients. CONCLUSION This study explored the function of BM genes in ccRCC for the first time. Reliable prognostic biomarkers that affect the survival of ccRCC patients were determined, and a BM gene-based prognostic model was established.
Collapse
Affiliation(s)
- Junyue Tao
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, China
- Institute of Urology, Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China
| | - Xiao Li
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, China
- Institute of Urology, Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China
| | - Chaozhao Liang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, China
- Institute of Urology, Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China
| | - Yi Liu
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, China
- Institute of Urology, Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China
| | - Jun Zhou
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, China
- Institute of Urology, Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China
| |
Collapse
|
6
|
Chen M, Hu R, Cavinato C, Zhuang ZW, Zhang J, Yun S, Fernandez Tussy P, Singh A, Murtada SI, Tanaka K, Liu M, Fernández-Hernando C, Humphrey JD, Schwartz MA. Fibronectin-Integrin α5 Signaling in Vascular Complications of Type 1 Diabetes. Diabetes 2022; 71:2020-2033. [PMID: 35771994 PMCID: PMC9450851 DOI: 10.2337/db21-0958] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 06/20/2022] [Indexed: 11/13/2022]
Abstract
Vascular complications are a major cause of illness and death in patients with type 1 diabetes (T1D). Diabetic vascular basement membranes are enriched in fibronectin (FN), an extracellular matrix protein that amplifies inflammatory signaling in endothelial cells through its main receptor, integrin α5β1. Binding of the integrin α5 cytoplasmic domain to phosphodiesterase 4D5 (PDE4D5), which increases phosphodiesterase catalytic activity and inhibits antiinflammatory cAMP signaling, was found to mediate these effects. Here, we examined mice in which the integrin α5 cytoplasmic domain is replaced by that of α2 (integrin α5/2) or the integrin α5 binding site in PDE4D is mutated (PDE4Dmut). T1D was induced via injection of streptozotocin and hyperlipidemia induced via injection of PCSK9 virus and provision of a high-fat diet. We found that in T1D and hyperlipidemia, the integrin α5/2 mutation reduced atherosclerosis plaque size by ∼50%, with reduced inflammatory cell invasion and metalloproteinase expression. Integrin α5/2 T1D mice also had improved blood-flow recovery from hindlimb ischemia and improved biomechanical properties of the carotid artery. By contrast, the PDE4Dmut had no beneficial effects in T1D. FN signaling through integrin α5 is thus a major contributor to diabetic vascular disease but not through its interaction with PDE4D.
Collapse
Affiliation(s)
- Minghao Chen
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale School of Medicine, New Haven, CT
- Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT
| | - Rui Hu
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale School of Medicine, New Haven, CT
- Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, People’s Republic of China
| | - Cristina Cavinato
- Department of Biomedical Engineering, Yale University, New Haven, CT
| | - Zhenwu W. Zhuang
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale School of Medicine, New Haven, CT
- Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT
| | - Jiasheng Zhang
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale School of Medicine, New Haven, CT
- Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT
| | - Sanguk Yun
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale School of Medicine, New Haven, CT
- Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT
| | - Pablo Fernandez Tussy
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT
- Departments of Comparative Medicine and Pathology, Yale Center for Molecular and Systems Metabolism, Yale School of Medicine, New Haven, CT
| | - Abhishek Singh
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT
- Departments of Comparative Medicine and Pathology, Yale Center for Molecular and Systems Metabolism, Yale School of Medicine, New Haven, CT
| | - Sae-Il Murtada
- Department of Biomedical Engineering, Yale University, New Haven, CT
| | - Keiichiro Tanaka
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale School of Medicine, New Haven, CT
- Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT
| | - Min Liu
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale School of Medicine, New Haven, CT
- Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, People’s Republic of China
| | - Carlos Fernández-Hernando
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT
- Departments of Comparative Medicine and Pathology, Yale Center for Molecular and Systems Metabolism, Yale School of Medicine, New Haven, CT
| | - Jay D. Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, CT
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT
| | - Martin A. Schwartz
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale School of Medicine, New Haven, CT
- Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT
- Department of Biomedical Engineering, Yale University, New Haven, CT
- Departments of Cell Biology and Biomedical Engineering, Yale University, New Haven, CT
| |
Collapse
|
7
|
Giblin MJ, Ontko CD, Penn JS. Effect of cytokine-induced alterations in extracellular matrix composition on diabetic retinopathy-relevant endothelial cell behaviors. Sci Rep 2022; 12:12955. [PMID: 35902594 PMCID: PMC9334268 DOI: 10.1038/s41598-022-12683-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 05/10/2022] [Indexed: 12/15/2022] Open
Abstract
Retinal vascular basement membrane (BM) thickening is an early structural abnormality of diabetic retinopathy (DR). Recent studies suggest that BM thickening contributes to the DR pathological cascade; however, much remains to be elucidated about the exact mechanisms by which BM thickening develops and subsequently drives other pathogenic events in DR. Therefore, we undertook a systematic analysis to understand how human retinal microvascular endothelial cells (hRMEC) and human retinal pericytes (hRP) change their expression of key extracellular matrix (ECM) constituents when treated with diabetes-relevant stimuli designed to model the three major insults of the diabetic environment: hyperglycemia, dyslipidemia, and inflammation. TNFα and IL-1β caused the most potent and consistent changes in ECM expression in both hRMEC and hRP. We also demonstrate that conditioned media from IL-1β-treated human Müller cells caused dose-dependent, significant increases in collagen IV and agrin expression in hRMEC. After narrowing our focus to inflammation-induced changes, we sought to understand how ECM deposited by hRMEC and hRP under inflammatory conditions affects the behavior of naïve hRMEC. Our data demonstrated that diabetes-relevant alterations in ECM composition alone cause both increased adhesion molecule expression by and increased peripheral blood mononuclear cell (PBMC) adhesion to naïve hRMEC. Taken together, these data demonstrate novel roles for inflammation and pericytes in driving BM pathology and suggest that inflammation-induced ECM alterations may advance other pathogenic behaviors in DR, including leukostasis.
Collapse
Affiliation(s)
- Meredith J Giblin
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, USA.
| | - Cayla D Ontko
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, USA
| | - John S Penn
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, USA
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville, USA
| |
Collapse
|
8
|
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
|
9
|
Kostov K, Blazhev A. Serum Anti-Collagen IV IgM and IgG Antibodies as Indicators of Low Vascular Turnover of Collagen IV in Patients with Long-Term Complications of Type 2 Diabetes. Diagnostics (Basel) 2021; 11:900. [PMID: 34069322 PMCID: PMC8158678 DOI: 10.3390/diagnostics11050900] [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: 04/21/2021] [Revised: 05/13/2021] [Accepted: 05/17/2021] [Indexed: 11/29/2022] Open
Abstract
Thickening of the vascular basement membrane (BM) is a fundamental structural change in the small blood vessels in diabetes. Collagen type IV (CIV) is a major component of the BMs, and monitoring the turnover of this protein in type 2 diabetes (T2D) can provide important information about the mechanisms of vascular damage. The aim of the study was through the use of non-invasive biomarkers of CIV (autoantibodies, derivative peptides, and immune complexes) to investigate vascular turnover of CIV in patients with long-term complications of T2D. We measured serum levels of these biomarkers in 59 T2D patients with micro- and/or macrovascular complications and 20 healthy controls using an ELISA. Matrix metalloproteinases-2 and -9 (MMP-2 and MMP-9) were also tested. In the T2D group, significantly lower levels of CIV markers and significantly higher levels of MMP-2 and MMP-9 were found compared to controls. A significant positive correlation was found between IgM antibody levels against CIV and MMP-2. These findings suggest that vascular metabolism of CIV is decreased in T2D with long-term complications and show that a positive linear relationship exists between MMP-2 levels and CIV turnover in the vascular wall.
Collapse
Affiliation(s)
- Krasimir Kostov
- Department of Pathophysiology, Medical University-Pleven, 1 Kliment Ohridski Str., 5800 Pleven, Bulgaria
| | - Alexander Blazhev
- Department of Biology, Medical University-Pleven, 1 Kliment Ohridski Str., 5800 Pleven, Bulgaria;
| |
Collapse
|
10
|
Asnaghi MA, Barthlott T, Gullotta F, Strusi V, Amovilli A, Hafen K, Srivastava G, Oertle P, Toni R, Wendt D, Holländer GA, Martin I. Thymus Extracellular Matrix-Derived Scaffolds Support Graft-Resident Thymopoiesis and Long-Term In Vitro Culture of Adult Thymic Epithelial Cells. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2010747. [PMID: 34539304 PMCID: PMC8436951 DOI: 10.1002/adfm.202010747] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/28/2021] [Indexed: 05/10/2023]
Abstract
The thymus provides the physiological microenvironment critical for the development of T lymphocytes, the cells that orchestrate the adaptive immune system to generate an antigen-specific response. A diverse population of stroma cells provides surface-bound and soluble molecules that orchestrate the intrathymic maturation and selection of developing T cells. Forming an intricate 3D architecture, thymic epithelial cells (TEC) represent the most abundant and important constituent of the thymic stroma. Effective models for in and ex vivo use of adult TEC are still wanting, limiting the engineering of functional thymic organoids and the understanding of the development of a competent immune system. Here a 3D scaffold is developed based on decellularized thymic tissue capable of supporting in vitro and in vivo thymopoiesis by both fetal and adult TEC. For the first time, direct evidences of feasibility for sustained graft-resident T-cell development using adult TEC as input are provided. Moreover, the scaffold supports prolonged in vitro culture of adult TEC, with a retained expression of the master regulator Foxn1. The success of engineering a thymic scaffold that sustains adult TEC function provides unprecedented opportunities to investigate thymus development and physiology and to design and implement novel strategies for thymus replacement therapies.
Collapse
Affiliation(s)
- M. Adelaide Asnaghi
- Department of BiomedicineUniversity Hospital BaselUniversity of BaselBasel4031Switzerland
| | - Thomas Barthlott
- Department of BiomedicineUniversity Children's Hospital University of BaselBasel4058Switzerland
| | - Fabiana Gullotta
- Department of BiomedicineUniversity Hospital BaselUniversity of BaselBasel4031Switzerland
| | - Valentina Strusi
- Department of BiomedicineUniversity Hospital BaselUniversity of BaselBasel4031Switzerland
| | - Anna Amovilli
- Department of BiomedicineUniversity Hospital BaselUniversity of BaselBasel4031Switzerland
| | - Katrin Hafen
- Department of BiomedicineUniversity Children's Hospital University of BaselBasel4058Switzerland
| | | | - Philipp Oertle
- ARTIDIS AGBasel4057Switzerland
- Biozentrum and the Swiss Nanoscience InstituteUniversity of BaselBasel4056Switzerland
| | - Roberto Toni
- Department of Medicine and Surgery – DIMEC, Unit of BiomedicalBiotechnological and Translational Sciences (S.BI.BI.T.)Laboratory of Regenerative Morphology and Bioartificial Structures (Re.Mo.Bio.S.)University of ParmaParma43121Italy
- Division of Endocrinology Diabetes, and MetabolismTufts Medical Center – Tufts University School of MedicineBostonMA02111USA
| | - David Wendt
- Department of BiomedicineUniversity Hospital BaselUniversity of BaselBasel4031Switzerland
| | - Georg A. Holländer
- Department of BiomedicineUniversity Children's Hospital University of BaselBasel4058Switzerland
- Developmental ImmunologyDepartment of Paediatrics and Weatherall Institute of Molecular MedicineUniversity of OxfordOxfordOX3 9DSUK
| | - Ivan Martin
- Department of BiomedicineUniversity Hospital BaselUniversity of BaselBasel4031Switzerland
- Department of Biomedical EngineeringUniversity Hospital BaselUniversity of BaselAllschwil4123Switzerland
| |
Collapse
|
11
|
Bódi N, Mezei D, Chakraborty P, Szalai Z, Barta BP, Balázs J, Rázga Z, Hermesz E, Bagyánszki M. Diabetes-related intestinal region-specific thickening of ganglionic basement membrane and regionally decreased matrix metalloproteinase 9 expression in myenteric ganglia. World J Diabetes 2021; 12:658-672. [PMID: 33995853 PMCID: PMC8107976 DOI: 10.4239/wjd.v12.i5.658] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/10/2021] [Accepted: 04/22/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The importance of the neuronal microenvironment has been recently highlighted in gut region-specific diabetic enteric neuropathy. Regionally distinct thickening of endothelial basement membrane (BM) of intestinal capillaries supplying the myenteric ganglia coincide with neuronal damage in different intestinal segments. Accelerated synthesis of matrix molecules and reduced degradation of matrix components may also contribute to the imbalance of extracellular matrix dynamics resulting in BM thickening. Among the matrix degrading proteinases, matrix metalloproteinase 9 (MMP9) and its tissue inhibitor (TIMP1) are essential in regulating extracellular matrix remodelling.
AIM To evaluate the intestinal segment-specific effects of diabetes and insulin replacement on ganglionic BM thickness, MMP9 and TIMP1 expression.
METHODS Ten weeks after the onset of hyperglycaemia gut segments were taken from the duodenum and ileum of streptozotocin-induced diabetic, insulin-treated diabetic and sex- and age-matched control rats. The thickness of BM surrounding myenteric ganglia was measured by electron microscopic morphometry. Whole-mount preparations of myenteric plexus were prepared from the different gut regions for MMP9/TIMP1 double-labelling fluorescent immunohistochemistry. Post-embedding immunogold electron microscopy was applied on ultrathin sections to evaluate the MMP9 and TIMP1 expression in myenteric ganglia and their microenvironment from different gut segments and conditions. The MMP9 and TIMP1 messenger ribonucleic acid (mRNA) level was measured by quantitative polymerase chain reaction.
RESULTS Ten weeks after the onset of hyperglycaemia, the ganglionic BM was significantly thickened in the diabetic ileum, while it remained intact in the duodenum. The immediate insulin treatment prevented the diabetes-related thickening of the BM surrounding the ileal myenteric ganglia. Quantification of particle density showed an increasing tendency for MMP9 and a decreasing tendency for TIMP1 from the proximal to the distal small intestine under control conditions. In the diabetic ileum, the number of MMP9-indicating gold particles decreased in myenteric ganglia, endothelial cells of capillaries and intestinal smooth muscle cells, however, it remained unchanged in all duodenal compartments. The MMP9/TIMP1 ratio was also decreased in ileal ganglia only. However, a marked segment-specific induction was revealed in MMP9 and TIMP1 at the mRNA levels.
CONCLUSION These findings support that the regional decrease in MMP9 expression in myenteric ganglia and their microenvironment may contribute to extracellular matrix accumulation, resulting in a region-specific thickening of ganglionic BM.
Collapse
Affiliation(s)
- Nikolett Bódi
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged 6726, Hungary
| | - Diána Mezei
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged 6726, Hungary
| | - Payal Chakraborty
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, Szeged 6726, Hungary
| | - Zita Szalai
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged 6726, Hungary
| | - Bence Pál Barta
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged 6726, Hungary
| | - János Balázs
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged 6726, Hungary
| | - Zsolt Rázga
- Department of Pathology, Faculty of Medicine, University of Szeged, Szeged 6720, Hungary
| | - Edit Hermesz
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, Szeged 6726, Hungary
| | - Mária Bagyánszki
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged 6726, Hungary
| |
Collapse
|
12
|
Cappelli HC, Guarino BD, Kanugula AK, Adapala RK, Perera V, Smith MA, Paruchuri S, Thodeti CK. Transient receptor potential vanilloid 4 channel deletion regulates pathological but not developmental retinal angiogenesis. J Cell Physiol 2021; 236:3770-3779. [PMID: 33078410 PMCID: PMC7920906 DOI: 10.1002/jcp.30116] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 10/07/2020] [Accepted: 10/09/2020] [Indexed: 01/08/2023]
Abstract
Transient receptor potential vanilloid 4 (TRPV4) channels are mechanosensitive ion channels that regulate systemic endothelial cell (EC) functions such as vasodilation, permeability, and angiogenesis. TRPV4 is expressed in retinal ganglion cells, Müller glia, pigment epithelium, microvascular ECs, and modulates cell volume regulation, calcium homeostasis, and survival. TRPV4-mediated physiological or pathological retinal angiogenesis remains poorly understood. Here, we demonstrate that TRPV4 is expressed, functional, and mechanosensitive in retinal ECs. The genetic deletion of TRPV4 did not affect postnatal developmental angiogenesis but increased pathological neovascularization in response to oxygen-induced retinopathy (OIR). Retinal vessels from TRPV4 knockout mice subjected to OIR exhibited neovascular tufts that projected into the vitreous humor and displayed reduced pericyte coverage compared with wild-type mice. These results suggest that TRPV4 is a regulator of retinal angiogenesis, its deletion augments pathological retinal angiogenesis, and that TRPV4 could be a novel target for the development of therapies against neovascular ocular diseases.
Collapse
Affiliation(s)
- Holly C. Cappelli
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272
- School of Biomedical Sciences, Kent State University, Kent, OH 44240
| | - Brianna D. Guarino
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272
| | - Anantha K. Kanugula
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272
| | - Ravi K. Adapala
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272
- School of Biomedical Sciences, Kent State University, Kent, OH 44240
| | - Vidushani Perera
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272
| | - Matthew A. Smith
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272
- Rebbeca D. Considine Research Institute, Akron Children’s Hospital, Akron, OH 44302
| | | | - Charles K. Thodeti
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272
- School of Biomedical Sciences, Kent State University, Kent, OH 44240
| |
Collapse
|
13
|
Pan WW, Gardner TW, Harder JL. Integrative Biology of Diabetic Retinal Disease: Lessons from Diabetic Kidney Disease. J Clin Med 2021; 10:1254. [PMID: 33803590 PMCID: PMC8003049 DOI: 10.3390/jcm10061254] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/09/2021] [Accepted: 03/12/2021] [Indexed: 01/13/2023] Open
Abstract
Diabetic retinal disease (DRD) remains the most common cause of vision loss in adults of working age. Progress on the development of new therapies for DRD has been limited by the complexity of the human eye, which constrains the utility of traditional research techniques, including animal and tissue culture models-a problem shared by those in the field of kidney disease research. By contrast, significant progress in the study of diabetic kidney disease (DKD) has resulted from the successful employment of systems biology approaches. Systems biology is widely used to comprehensively understand complex human diseases through the unbiased integration of genetic, environmental, and phenotypic aspects of the disease with the functional and structural manifestations of the disease. The application of a systems biology approach to DRD may help to clarify the molecular basis of the disease and its progression. Acquiring this type of information might enable the development of personalized treatment approaches, with the goal of discovering new therapies targeted to an individual's specific DRD pathophysiology and phenotype. Furthermore, recent efforts have revealed shared and distinct pathways and molecular targets of DRD and DKD, highlighting the complex pathophysiology of these diseases and raising the possibility of therapeutics beneficial to both organs. The objective of this review is to survey the current understanding of DRD pathophysiology and to demonstrate the investigative approaches currently applied to DKD that could promote a more thorough understanding of the structure, function, and progression of DRD.
Collapse
Affiliation(s)
- Warren W. Pan
- Department of Ophthalmology and Visual Sciences, University of Michigan Medical School, Ann Arbor, MI 48105, USA; (W.W.P.); (T.W.G.)
| | - Thomas W. Gardner
- Department of Ophthalmology and Visual Sciences, University of Michigan Medical School, Ann Arbor, MI 48105, USA; (W.W.P.); (T.W.G.)
- Department of Internal Medicine (Metabolism, Endocrinology and Diabetes), University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Jennifer L. Harder
- Department of Internal Medicine (Nephrology), University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
14
|
Ferrara M, Lugano G, Sandinha MT, Kearns VR, Geraghty B, Steel DHW. Biomechanical properties of retina and choroid: a comprehensive review of techniques and translational relevance. Eye (Lond) 2021; 35:1818-1832. [PMID: 33649576 PMCID: PMC8225810 DOI: 10.1038/s41433-021-01437-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 12/06/2020] [Accepted: 01/26/2021] [Indexed: 02/06/2023] Open
Abstract
Studying the biomechanical properties of biological tissue is crucial to improve our understanding of disease pathogenesis. The biomechanical characteristics of the cornea, sclera and the optic nerve head have been well addressed with an extensive literature and an in-depth understanding of their significance whilst, in comparison, knowledge of the retina and choroid is relatively limited. Knowledge of these tissues is important not only to clarify the underlying pathogenesis of a wide variety of retinal and vitreoretinal diseases, including age-related macular degeneration, hereditary retinal dystrophies and vitreoretinal interface diseases but also to optimise the surgical handling of retinal tissues and, potentially, the design and properties of implantable retinal prostheses and subretinal therapies. Our aim with this article is to comprehensively review existing knowledge of the biomechanical properties of retina, internal limiting membrane (ILM) and the Bruch’s membrane–choroidal complex (BMCC), highlighting the potential implications for clinical and surgical practice. Prior to this we review the testing methodologies that have been used both in vitro, and those starting to be used in vivo to aid understanding of their results and significance.
Collapse
Affiliation(s)
| | - Gaia Lugano
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | | | - Victoria R Kearns
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Brendan Geraghty
- Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK.
| | - David H W Steel
- Sunderland Eye Infirmary, Sunderland, UK. .,Bioscience Institute, Newcastle University, Newcastle Upon Tyne, UK.
| |
Collapse
|
15
|
Khalilgharibi N, Mao Y. To form and function: on the role of basement membrane mechanics in tissue development, homeostasis and disease. Open Biol 2021; 11:200360. [PMID: 33593159 PMCID: PMC8061686 DOI: 10.1098/rsob.200360] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The basement membrane (BM) is a special type of extracellular matrix that lines the basal side of epithelial and endothelial tissues. Functionally, the BM is important for providing physical and biochemical cues to the overlying cells, sculpting the tissue into its correct size and shape. In this review, we focus on recent studies that have unveiled the complex mechanical properties of the BM. We discuss how these properties can change during development, homeostasis and disease via different molecular mechanisms, and the subsequent impact on tissue form and function in a variety of organisms. We also explore how better characterization of BM mechanics can contribute to disease diagnosis and treatment, as well as development of better in silico and in vitro models that not only impact the fields of tissue engineering and regenerative medicine, but can also reduce the use of animals in research.
Collapse
Affiliation(s)
- Nargess Khalilgharibi
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK.,Institute for the Physics of Living Systems, University College London, Gower Street, London WC1E 6BT, UK
| | - Yanlan Mao
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK.,Institute for the Physics of Living Systems, University College London, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
16
|
De Gregorio C, Ezquer F. Sensory neuron cultures derived from adult db/db mice as a simplified model to study type-2 diabetes-associated axonal regeneration defects. Dis Model Mech 2021; 14:dmm.046334. [PMID: 33262160 PMCID: PMC7847260 DOI: 10.1242/dmm.046334] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 11/17/2020] [Indexed: 12/11/2022] Open
Abstract
Diabetic neuropathy (DN) is an early common complication of diabetes mellitus (DM), leading to chronic pain, sensory loss and muscle atrophy. Owing to its multifactorial etiology, neuron in vitro cultures have been proposed as simplified systems for DN studies. However, the most used models currently available do not recreate the chronic and systemic damage suffered by peripheral neurons of type-2 DM (T2DM) individuals. Here, we cultured neurons derived from dorsal root ganglia from 6-month-old diabetic db/db-mice, and evaluated their morphology by the Sholl method as an easy-to-analyze readout of neuronal function. We showed that neurons obtained from diabetic mice exhibited neuritic regeneration defects in basal culture conditions, compared to neurons from non-diabetic mice. Next, we evaluated the morphological response to common neuritogenic factors, including nerve growth factor NGF and Laminin-1 (also called Laminin-111). Neurons derived from diabetic mice exhibited reduced regenerative responses to these factors compared to neurons from non-diabetic mice. Finally, we analyzed the neuronal response to a putative DN therapy based on the secretome of mesenchymal stem cells (MSC). Neurons from diabetic mice treated with the MSC secretome displayed a significant improvement in neuritic regeneration, but still reduced when compared to neurons derived from non-diabetic mice. This in vitro model recapitulates many alterations observed in sensory neurons of T2DM individuals, suggesting the possibility of studying neuronal functions without the need of adding additional toxic factors to culture plates. This model may be useful for evaluating intrinsic neuronal responses in a cell-autonomous manner, and as a throughput screening for the pre-evaluation of new therapies for DN. Summary: Morphological characterization of a model for evaluating neuritic regeneration in vitro in dorsal root ganglion primary neurons derived from type-2 diabetic mice with an advanced stage of diabetic neuropathy.
Collapse
Affiliation(s)
- Cristian De Gregorio
- Center for Regenerative Medicine, School of Medicine Clínica Alemana-Universidad del Desarrollo, Santiago, 7690000 Chile
| | - Fernando Ezquer
- Center for Regenerative Medicine, School of Medicine Clínica Alemana-Universidad del Desarrollo, Santiago, 7690000 Chile
| |
Collapse
|
17
|
Role of the Internal Limiting Membrane in Structural Engraftment and Topographic Spacing of Transplanted Human Stem Cell-Derived Retinal Ganglion Cells. Stem Cell Reports 2020; 16:149-167. [PMID: 33382979 PMCID: PMC7897583 DOI: 10.1016/j.stemcr.2020.12.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 12/11/2022] Open
Abstract
Retinal ganglion cell (RGC) replacement holds potential for restoring vision lost to optic neuropathy. Transplanted RGCs must undergo neuroretinal integration to receive afferent visual signals for processing and efferent transmission. To date, retinal integration following RGC transplantation has been limited. We sought to overcome key barriers to transplanted human stem cell-derived RGC integration. Following co-culture ex vivo on organotypic mouse retinal explants, human RGCs cluster and extend bundled neurites that remain superficial to the neuroretina, hindering afferent synaptogenesis. To enhance integration, we increased the cellular permeability of the internal limiting membrane (ILM). Extracellular matrix digestion using proteolytic enzymes achieved ILM disruption while minimizing retinal toxicity and preserving glial reactivity. ILM disruption is associated with dispersion rather than clustering of co-cultured RGC bodies and neurites, and increased parenchymal neurite ingrowth. The ILM represents a significant obstacle to transplanted RGC connectivity and its circumvention may be necessary for functional RGC replacement.
Collapse
|
18
|
Aschauer J, Pollreisz A, Karst S, Hülsmann M, Hajdu D, Datlinger F, Egner B, Kriechbaum K, Pablik E, Schmidt-Erfurth UM. Longitudinal analysis of microvascular perfusion and neurodegenerative changes in early type 2 diabetic retinal disease. Br J Ophthalmol 2020; 106:528-533. [PMID: 33293271 DOI: 10.1136/bjophthalmol-2020-317322] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 12/12/2022]
Abstract
AIM To prospectively monitor subclinical changes in capillary perfusion and retinal layer thickness in patients with type 2 diabetes and early diabetic retinal disease over 2 years. METHODS In this longitudinal study we performed biannual retinal vascular imaging using optical coherence tomography angiography (RTVue) to analyse the foveal avascular zone (FAZ) area, perimeter, acircularity index (AI) and parafoveal superficial/deep vessel density (VD). Spectral-domain optical coherence tomography (Spectralis) was used to measure the thickness of nine macular layers and the peripapillary nerve fibre layer. RESULTS Among 117 eyes (58 left) of 59 patients (21 female), 105 had no diabetic retinopathy (DR), 6 mild and 6 moderate non-proliferative DR at baseline. We found DR progression in 13 eyes at year 2. The FAZ area (+0.008±0.002 mm2/year, p<0.0001), perimeter (+0.036±0.010 mm/year, p=0.006) and AI (+0.005±0.002/year, p=0.0280) increased significantly. A pronounced decrease was found in the superficial (-1.425±0.290%/year, p<0.0001) but not the deep VD. Inner neuroretinal loss was confined to the ganglion cell (-0.539±0.150 µm/year, p=0.0004) and the inner plexiform layer (-0.361±0.127 µm/year, p=0.0045). In the outer retina, we observed a statistically significant decrease in thickness in the outer plexiform, photoreceptor layer and pigment epithelium of -0.921±0.161 µm/year, -0.325±0.139 µm/year and -0.385±0.084 µm/year, respectively. CONCLUSION Subclinical signs of microangiopathy and neurodegeneration appear in parallel and are highly progressive even in the earliest stages of diabetic retinal disease.Trial registration number EudraCT20156000239634.
Collapse
Affiliation(s)
- Julia Aschauer
- Department of Ophthalmology and Optometry, Medical University of Vienna, Wien, Wien, Austria
| | - Andreas Pollreisz
- Department of Ophthalmology and Optometry, Medical University of Vienna, Wien, Wien, Austria
| | - Sonja Karst
- Department of Ophthalmology and Optometry, Medical University of Vienna, Wien, Wien, Austria
| | - Martin Hülsmann
- Division of Cardiology, Department of Internal Medicine, Medical University of Vienna, Wien, Wien, Austria
| | - Dorottya Hajdu
- Department of Ophthalmology and Optometry, Medical University of Vienna, Wien, Wien, Austria
| | - Felix Datlinger
- Department of Ophthalmology and Optometry, Medical University of Vienna, Wien, Wien, Austria
| | - Berit Egner
- Department of Ophthalmology and Optometry, Medical University of Vienna, Wien, Wien, Austria
| | - Katharina Kriechbaum
- Department of Ophthalmology and Optometry, Medical University of Vienna, Wien, Wien, Austria
| | | | | |
Collapse
|
19
|
Halfter W, Moes S, Halfter K, Schoenenberger MS, Monnier CA, Kalita J, Asgeirsson D, Binggeli T, Jenoe P, Scholl HPN, Henrich PB. The human Descemet's membrane and lens capsule: Protein composition and biomechanical properties. Exp Eye Res 2020; 201:108326. [PMID: 33147472 DOI: 10.1016/j.exer.2020.108326] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 10/18/2020] [Accepted: 10/20/2020] [Indexed: 02/07/2023]
Abstract
The Descemet's membrane (DM) and the lens capsule (LC) are two ocular basement membranes (BMs) that are essential in maintaining stability and structure of the cornea and lens. In this study, we investigated the proteomes and biomechanical properties of these two materials to uncover common and unique properties. We also screened for possible protein changes during diabetes. LC-MS/MS was used to determine the proteomes of both BMs. Biomechanical measurements were conducted by atomic force microscopy (AFM) in force spectroscopy mode, and complemented with immunofluorescence microscopy. Proteome analysis showed that all six existing collagen IV chains represent 70% of all LC-protein, and are thus the dominant components of the LC. The DM on the other hand is predominantly composed of a single protein, TGF-induced protein, which accounted for around 50% of all DM-protein. Four collagen IV-family members in DM accounted for only 10% of the DM protein. Unlike the retinal vascular BMs, the LC and DM do not undergo significant changes in their protein compositions during diabetes. Nanomechanical measurements showed that the endothelial/epithelial sides of both BMs are stiffer than their respective stromal/anterior-chamber sides, and both endothelial and stromal sides of the DM were stiffer than the epithelial and anterior-chamber sides of the LC. Long-term diabetes did not change the stiffness of the DM and LC. In summary, our analyses show that the protein composition and biomechanical properties of the DM and LC are different, i.e., the LC is softer than DM despite a significantly higher concentration of collagen IV family members. This finding is unexpected, as collagen IV members are presumed to be responsible for BM stiffness. Diabetes had no significant effect on the protein composition and the biomechanical properties of both the DM and LC.
Collapse
Affiliation(s)
- Willi Halfter
- Department of Ophthalmology, University of Basel, Switzerland.
| | - Suzette Moes
- Proteomics Core Facility, Biozentrum, University of Basel, Switzerland.
| | - Kathrin Halfter
- Munich Cancer Registry, Institute of Medical Informatics, Biometry and Epidemiology, Maximilian University Munich, Germany.
| | | | | | - Joanna Kalita
- Biozentrum and the Swiss Nanoscience Institute, University of Basel, Switzerland
| | - Daphne Asgeirsson
- Biozentrum and the Swiss Nanoscience Institute, University of Basel, Switzerland.
| | | | - Paul Jenoe
- Proteomics Core Facility, Biozentrum, University of Basel, Switzerland.
| | - Hendrik P N Scholl
- Department of Ophthalmology, University of Basel, Switzerland; Institute of Molecular and Clinical Ophthalmology Basel (IOB), Switzerland; Wilmer Eye Institute, Johns Hopkins University, Baltimore, MA, USA.
| | - Paul Bernhard Henrich
- Department of Ophthalmology, University of Basel, Switzerland; Università della Svizzera Italiana, Lugano, Switzerland.
| |
Collapse
|
20
|
Roy S, Kim D. Retinal capillary basement membrane thickening: Role in the pathogenesis of diabetic retinopathy. Prog Retin Eye Res 2020; 82:100903. [PMID: 32950677 DOI: 10.1016/j.preteyeres.2020.100903] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 09/08/2020] [Accepted: 09/15/2020] [Indexed: 02/07/2023]
Abstract
Vascular basement membrane (BM) thickening has been hailed over half a century as the most prominent histological lesion in diabetic microangiopathy, and represents an early ultrastructural change in diabetic retinopathy (DR). Although vascular complications of DR have been clinically well established, specific cellular and molecular mechanisms underlying dysfunction of small vessels are not well understood. In DR, small vessels develop insidiously as BM thickening occurs. Studies examining high resolution imaging data have established BM thickening as one of the foremost structural abnormalities of retinal capillaries. This fundamental structural change develops, at least in part, from excess accumulation of BM components. Although BM thickening is closely associated with the development of DR, its contributory role in the pathogenesis of DR is coming to light recently. DR develops over several years before clinical manifestations appear, and it is during this clinically silent period that hyperglycemia induces excess synthesis of BM components, contributes to vascular BM thickening, and promotes structural and functional lesions including cell death and vascular leakage in the diabetic retina. Studies using animal models show promising results in preventing BM thickening with subsequent beneficial effects. Several gene regulatory approaches are being developed to prevent excess synthesis of vascular BM components in an effort to reduce BM thickening. This review highlights current understanding of capillary BM thickening development, role of BM thickening in retinal vascular lesions, and strategies for preventing vascular BM thickening as a potential therapeutic strategy in alleviating characteristic lesions associated with DR.
Collapse
Affiliation(s)
- Sayon Roy
- Boston University School of Medicine, Boston, MA, USA.
| | - Dongjoon Kim
- Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
21
|
Frizziero L, Parrozzani R, Londei D, Pilotto E, Midena E. Quantification of vascular and neuronal changes in the peripapillary retinal area secondary to diabetic retinopathy. Br J Ophthalmol 2020; 105:1577-1583. [PMID: 32933938 DOI: 10.1136/bjophthalmol-2020-316468] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 06/22/2020] [Accepted: 08/31/2020] [Indexed: 12/27/2022]
Abstract
PURPOSE To investigate and quantify peripapillary vascular and neuronal changes secondary to diabetic retinopathy, using spectral-domain optical coherence tomography (OCT) and OCT angiography (OCTA). DESIGN This was a cross-sectional study. METHODS 51 eyes of 51 patients affected by non-proliferative diabetic retinopathy (NPDR) and 19 age-matched healthy control eyes underwent full ophthalmic examination, including OCT and OCTA in the peripapillary area. Vessel area density (VAD), vessel length fraction (VLF) and vessel diameter index (VDI) were quantified in a ring-shaped region of interest of each OCTA image. Capillaries and larger vessels were separately analysed. The thickness of the peripapillary retinal nerve fibre layer (pRNFL) and macular ganglion cell complex (GCC) was also analysed. RESULTS VAD and VLF of peripapillary capillaries were significantly reduced in NPDR eyes, along with the progression of NPDR (p<0.05). VDI was significantly reduced in mild (p=0.0093) and moderate (p=0.0190) NPDR eyes, but not in severe NPDR (p=0.0841). Larger peripapillary vessels showed a significant increase of both VAD and VDI in NPDR eyes. pRNFL and GCC thickness decreased in NPDR eyes, reaching statistical significance only for GCC. No statistically significant correlation was found between perfusion parameters and pRNFL and GCC thickness. CONCLUSIONS Retinal capillary remodelling in NPDR involves the peripapillary vascularisation too, as confirmed by OCTA quantitative parameters. The peripapillary macrovasculature and microvasculature need to be separately evaluated. The lack of direct correlation between peripapillary capillaries changes and the loss of retinal nerve fibres suggests that neuronal damage cannot be simply considered secondary to the microvascular one.
Collapse
Affiliation(s)
| | | | - Davide Londei
- Department of Ophthalmology, University of Padova, Padova, Italy
| | | | - Edoardo Midena
- IRCCS - Fondazione Bietti, Rome, Italy .,Department of Ophthalmology, University of Padova, Padova, Italy
| |
Collapse
|
22
|
Gamidov AA, Baryshev KV, Perevozchikov KA, Surnina ZV. [Atomic force microscopy in the study of retinal structure]. Vestn Oftalmol 2020; 136:251-257. [PMID: 32880147 DOI: 10.17116/oftalma2020136042251] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The review addresses the current state of atomic force microscopy (AFM) usage in ophthalmology. Briefly presented here are the history of the development of AFM, principles and modes of operation, its advantages, disadvantages, as well as a comparison with other types of microscopy. The review describes the capabilities of AFM in visualization of various structures of the eye. A significant part of the review is devoted to the study of the retina, which arouses great interest among researchers. In particular, the possibilities of AFM for visualization at the submicron level of various structures in the retina, such as the internal limiting membrane, membrane cells, Müller cells, retinal pigment epithelium in their normal state and in the presence of a pathology (age-related macular degeneration, diabetes mellitus) were reviewed. In addition, several study papers were analyzed, providing a base for the judgement of the mechanical properties of said structures. An AFM study of the visual pigment rhodopsin helped identify its dimeric structure. The stability of the rhodopsin molecule was proved to be determined by the degree of strength of its individual segments connected to each other. The AFM method is a highly accurate method that helps solve many fundamental and practical problems, particularly in ophthalmology.
Collapse
Affiliation(s)
- A A Gamidov
- Research Institute of Eye Diseases, Moscow, Russia
| | - K V Baryshev
- Research Institute of Eye Diseases, Moscow, Russia
| | | | - Z V Surnina
- Research Institute of Eye Diseases, Moscow, Russia
| |
Collapse
|
23
|
Mutgan AC, Jandl K, Kwapiszewska G. Endothelial Basement Membrane Components and Their Products, Matrikines: Active Drivers of Pulmonary Hypertension? Cells 2020; 9:cells9092029. [PMID: 32899187 PMCID: PMC7563239 DOI: 10.3390/cells9092029] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/27/2020] [Accepted: 08/29/2020] [Indexed: 12/19/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a vascular disease that is characterized by elevated pulmonary arterial pressure (PAP) due to progressive vascular remodeling. Extracellular matrix (ECM) deposition in pulmonary arteries (PA) is one of the key features of vascular remodeling. Emerging evidence indicates that the basement membrane (BM), a specialized cluster of ECM proteins underlying the endothelium, may be actively involved in the progression of vascular remodeling. The BM and its steady turnover are pivotal for maintaining appropriate vascular functions. However, the pathologically elevated turnover of BM components leads to an increased release of biologically active short fragments, which are called matrikines. Both BM components and their matrikines can interfere with pivotal biological processes, such as survival, proliferation, adhesion, and migration and thus may actively contribute to endothelial dysfunction. Therefore, in this review, we summarize the emerging role of the BM and its matrikines on the vascular endothelium and further discuss its implications on lung vascular remodeling in pulmonary hypertension.
Collapse
Affiliation(s)
- Ayse Ceren Mutgan
- Otto Loewi Research Center, Division of Physiology, Medical University of Graz, 8010 Graz, Austria;
| | - Katharina Jandl
- Ludwig Boltzmann Institute for Lung Vascular Research, 8010 Graz, Austria;
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria
| | - Grazyna Kwapiszewska
- Otto Loewi Research Center, Division of Physiology, Medical University of Graz, 8010 Graz, Austria;
- Ludwig Boltzmann Institute for Lung Vascular Research, 8010 Graz, Austria;
- Correspondence:
| |
Collapse
|
24
|
Nguyen NH, Kim D, Roy S. High Glucose Increases Binding of Lysyl Oxidase to Extracellular Matrix Proteins: Implications for Diabetic Retinopathy. Invest Ophthalmol Vis Sci 2020; 61:40. [PMID: 32340032 PMCID: PMC7401919 DOI: 10.1167/iovs.61.4.40] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Purpose To determine whether high glucose (HG) compromises internalization of lysyl oxidase (LOX) through excess binding of LOX with extracellular matrix (ECM) proteins. Methods To determine whether HG promotes binding of LOX with ECM proteins, fibronectin (FN) and collagen IV (Coll IV), total or ECM-only proteins from rat retinal endothelial cells grown in normal (N; 5 mM) or HG (30 mM) medium were analyzed by coimmunoprecipitation and Western blot (WB). In parallel, coimmunostaining was performed to determine changes in LOX binding to FN or Coll IV. To determine the effect of HG on extracellular LOX levels, medium in which cells were grown for 1, 3, 5, and 7 days were assessed for LOX levels. Results WB analysis using total protein showed LOX overexpression and elevated levels of LOX bound to Coll IV or FN in HG condition. Similarly, a significant increase in LOX bound to FN or Coll IV was observed in ECM-only protein. These data were supported by Z-stack confocal microscopy images from coimmunostaining. Furthermore, immunostaining performed on ECM layer revealed increased presence of LOX bound to Coll IV or FN. Additionally, when media from cells grown in HG was monitored, a maximal increase in LOX level was observed by day 3, which declined by day 7. Conclusions Findings indicate that HG promotes binding of LOX to FN and Coll IV extracellularly that results in reduced LOX internalization, attenuation of negative feedback, and upregulation of LOX expression associated with diabetic retinopathy.
Collapse
Affiliation(s)
| | | | - Sayon Roy
- , , Massachusetts.,, , Massachusetts
| |
Collapse
|
25
|
Spadaro A, Rao M, Lorenti M, Romano MR, Augello A, Eandi CM, Platania CBM, Drago F, Bucolo C. New Brilliant Blue G Derivative as Pharmacological Tool in Retinal Surgery. Front Pharmacol 2020; 11:708. [PMID: 32523529 PMCID: PMC7261835 DOI: 10.3389/fphar.2020.00708] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 04/29/2020] [Indexed: 11/13/2022] Open
Abstract
Our study was aimed at assessing the retinal binding of a new synthetic Brilliant Blue G (BBG) derivative (pure benzyl-Brilliant Blue G; PBB) ophthalmic formulation, to improve vitreoretinal surgery procedure. Protein affinity of the new molecule was evaluated in vitro (cell-free assay) and in silico. Furthermore, an ex vivo model of vitreoretinal surgery was developed by using porcine eyes to assess the pharmacological profile of PBB, compared to commercial formulations based on BBG and methyl-BBG (Me-BBG). PBB showed a higher affinity for proteins (p < 0.05), compared to BBG and Me-BBG. In vitro and in silico studies demonstrated that the high selectivity of PBB could be related to high lipophilicity and binding affinity to fibronectin, the main component of the retinal internal limiting membrane (ILM). The PBB staining capabilities were evaluated in porcine eyes in comparison with BBG and Me-BBG. Forty microliters of each formulation were slowly placed over the retinal surface and removed after 30 s. After that, ILM peeling was carried out, and the retina collected. BBG, Me-BBG, and PBB quantification in ILM and retina tissues was carried out by HPLC analysis. PBB levels in the ILM were significantly (p < 0.05) higher compared to BBG and Me-BBG formulations. On the contrary, PBB showed a much lower (p < 0.05) distribution in retina (52 ng/mg tissue) compared to BBG and Me-BBG, in particular PBB levels were significantly (p < 0.05) lower. Therefore, the new synthetic Brilliant Blue derivative (PBB) showed a great ILM selectivity in comparison to underneath retinal layers. In conclusion, these findings had high translational impact with a tangible improving in ex vivo model of retinal surgery, suggesting a future use during surgical practice.
Collapse
Affiliation(s)
- Angelo Spadaro
- Department of Drug Sciences, University of Catania, Catania, Italy
| | - Marco Rao
- Department of Drug Sciences, University of Catania, Catania, Italy
| | - Miriam Lorenti
- Department of Drug Sciences, University of Catania, Catania, Italy
| | | | - Antonio Augello
- Section of Hygiene and Food of Animal Origin (SIAOA - UFCM), Department of Veterinary Prevention, Azienda Sanitaria Provinciale (ASP - CT), Catania, Italy
| | - Chiara Maria Eandi
- Department of Ophthalmology, University of Lausanne, Hôpital Ophtalmique Jules-Gonin, Lausanne, Switzerland.,Department of Surgical Science, University of Torino, Torino, Italy
| | - Chiara Bianca Maria Platania
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania, Italy
| | - Filippo Drago
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania, Italy.,Center for Research in Ocular Pharmacology-CERFO, University of Catania, Catania, Italy
| | - Claudio Bucolo
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania, Italy.,Center for Research in Ocular Pharmacology-CERFO, University of Catania, Catania, Italy
| |
Collapse
|
26
|
Gellen B, Thorin-Trescases N, Thorin E, Gand E, Sosner P, Brishoual S, Rigalleau V, Montaigne D, Javaugue V, Pucheu Y, Gatault P, Piguel X, Hadjadj S, Saulnier PJ. Serum tenascin-C is independently associated with increased major adverse cardiovascular events and death in individuals with type 2 diabetes: a French prospective cohort. Diabetologia 2020; 63:915-923. [PMID: 32040670 DOI: 10.1007/s00125-020-05108-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 01/13/2020] [Indexed: 12/25/2022]
Abstract
AIMS/HYPOTHESIS Tenascin-C (TN-C) is an extracellular matrix glycoprotein highly expressed in inflammatory and cardiovascular (CV) diseases. Serum TN-C has not yet been specifically studied in individuals with type 2 diabetes, a condition associated with chronic low-grade inflammation and increased CV disease risk. In this study, we hypothesised that elevated serum TN-C at enrolment in participants with type 2 diabetes would be associated with increased risk of death and major adverse CV events (MACE) during follow-up. METHODS We used a prospective, monocentric cohort of consecutive type 2 diabetes participants (the SURDIAGENE [SUivi Rénal, DIAbète de type 2 et GENEtique] cohort) with all-cause death as a primary endpoint and MACE (CV death, non-fatal myocardial infarction or stroke) as a secondary endpoint. We used a proportional hazard model after adjustment for traditional risk factors and the relative integrated discrimination improvement (rIDI) to assess the incremental predictive value of TN-C for these risk factors. RESULTS We monitored 1321 individuals (58% men, mean age 64 ± 11 years) for a median of 89 months. During follow-up, 442 individuals died and 497 had MACE. Multivariate Cox analysis showed that serum TN-C concentrations were associated with an increased risk of death (HR per 1 SD: 1.27 [95% CI 1.17, 1.38]; p < 0.0001) and MACE (HR per 1 SD: 1.23 [95% CI 1.13, 1.34]; p < 0.0001). Using TN-C concentrations on top of traditional risk factors, prediction of the risk of all-cause death (rIDI: 8.2%; p = 0.0006) and MACE (rIDI: 6.7%; p = 0.0014) improved significantly, but modestly. CONCLUSIONS/INTERPRETATION In individuals with type 2 diabetes, increased serum TN-C concentrations were independently associated with death and MACE. Therefore, including TN-C as a prognostic biomarker could improve risk stratification in these individuals.
Collapse
Affiliation(s)
- Barnabas Gellen
- ELSAN, Polyclinique de Poitiers, 1 Rue de la Providence, F-86000, Poitiers, France.
| | | | - Eric Thorin
- Research Center, Montreal Heart Institute, Montreal, QC, Canada
- Department of Surgery, Faculty of Medicine, Montreal Heart Institute, Université de Montréal, Montreal, QC, Canada
| | - Elise Gand
- INSERM, Centre d'Investigation Clinique CIC1402, Université de Poitiers, CHU de Poitiers, Poitiers, France
| | - Philippe Sosner
- INSERM, Centre d'Investigation Clinique CIC1402, Université de Poitiers, CHU de Poitiers, Poitiers, France
- Laboratoire MOVE (EA 6314), Université de Poitiers, Poitiers, France
- Centre Médico-Sportif Mon Stade, Paris, France
| | - Sonia Brishoual
- INSERM, Centre d'Investigation Clinique CIC1402, Université de Poitiers, CHU de Poitiers, Poitiers, France
| | - Vincent Rigalleau
- Endocrinology - Diabetology - Nutrition, CHU de Bordeaux, Hôpital Haut-Lévêque, Pessac, France
| | - David Montaigne
- Department of Clinical Physiology - Echocardiography, CHU Lille, Lille, France
- INSERM U1011, EGID, Institut Pasteur de Lille, University of Lille, Lille, France
| | - Vincent Javaugue
- INSERM, Centre d'Investigation Clinique CIC1402, Université de Poitiers, CHU de Poitiers, Poitiers, France
- Nephrology, CHU de Poitiers, Poitiers, France
| | | | - Philippe Gatault
- Transplantation, Immunology and Inflammation (T2I) - EA4245, CHRU de Tours, Nephrology-Hypertension, Dialysis and Renal Transplantation, FHU SUPORT, Université de Tours, Tours, France
| | - Xavier Piguel
- Endocrinology-Diabetology, CHU de Poitiers, Poitiers, France
| | - Samy Hadjadj
- Transplantation, Immunology and Inflammation (T2I) - EA4245, CHRU de Tours, Nephrology-Hypertension, Dialysis and Renal Transplantation, FHU SUPORT, Université de Tours, Tours, France
- L'Institut du Thorax, INSERM, CNRS, UNIV Nantes, CHU Nantes, Nantes, France
| | - Pierre-Jean Saulnier
- INSERM, Centre d'Investigation Clinique CIC1402, Université de Poitiers, CHU de Poitiers, Poitiers, France
| | | |
Collapse
|
27
|
Duan P, Chen S, Zeng Y, Xu H, Liu Y. Osteopontin Upregulates Col IV Expression by Repressing miR-29a in Human Retinal Capillary Endothelial Cells. MOLECULAR THERAPY-NUCLEIC ACIDS 2020; 20:242-251. [PMID: 32182570 PMCID: PMC7078126 DOI: 10.1016/j.omtn.2020.02.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 01/23/2020] [Accepted: 02/05/2020] [Indexed: 01/04/2023]
Abstract
Abnormal synthesis of extracellular matrix (ECM), especially collagen type IV (Col IV), in human retinal capillary endothelial cells (HRCECs) and resultant basement membrane (BM) thickening is the most prominent and characteristic feature of early diabetic retinopathy (DR). Osteopontin (OPN) has been shown to play an important role in the pathogenesis of DR and specifically, found to be critically involved in diabetic nephropathy, as it can upregulate many factors, like collagen IV. However, the precise role of OPN in the pathogenesis of DR and the underlying mechanisms remain unclear. In this study, 51 differentially expressed microRNAs (miRNAs; 42 miRNAs upregulated and 9 miRNAs downregulated) were first identified in retina of streptozotocin (STZ)-induced diabetic mice with DR. Among these miRNAs, we identified miRNA (miR)-29a as a prominent miRNA that targeted and directly downregulated Col IV expression through database prediction and dual-luciferase reporter assay, which was further confirmed in HRCECs using miR-29a mimic, miR-29a inhibitor, and pre-miR-29a transfection. Furthermore, OPN upregulated Col IV expression via a miR-29a-repressed pathway in HRCECs. Taken together, these results provided a miR-29a-repressing mechanism through which OPN plays roles in abnormal synthesis of Col IV in HRCECs and resultant BM thickening, contributing to the pathogenesis of DR.
Collapse
Affiliation(s)
- Ping Duan
- Southwest Hospital, Southwest Eye Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China; Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Chongqing 400038, China
| | - Siyu Chen
- Southwest Hospital, Southwest Eye Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China; Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Chongqing 400038, China
| | - Yuxiao Zeng
- Southwest Hospital, Southwest Eye Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China; Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Chongqing 400038, China
| | - Haiwei Xu
- Southwest Hospital, Southwest Eye Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China; Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Chongqing 400038, China.
| | - Yong Liu
- Southwest Hospital, Southwest Eye Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China; Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Chongqing 400038, China.
| |
Collapse
|
28
|
Chen L, Wei Y, Zhou X, Zhang Z, Chi W, Gong L, Jiang X, Zhang S. Morphologic, Biomechanical, and Compositional Features of the Internal Limiting Membrane in Pathologic Myopic Foveoschisis. Invest Ophthalmol Vis Sci 2019; 59:5569-5578. [PMID: 30480705 DOI: 10.1167/iovs.18-24676] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose To investigate alterations in the morphologic, compositional, and biomechanical properties of the internal limiting membrane (ILM) in pathologic myopic foveoschisis (MF) eyes. Methods ILM specimens were peeled from 61 eyes with MF and 56 eyes with stage III/IV idiopathic macular hole (IMH) as a control. Samples were analyzed for transmission electron microscopy (TEM), scanning electron microscopy, immunofluorescence, Western blotting, and atomic force microscopy. ILM characteristics were compared between the two groups. Results TEM findings revealed that thickness of the MF ILMs significantly decreased compared with that of IMH ILMs (0.753 ± 0.215 vs. 1.894 ± 0.247 μm; P < 0.0001). The vitreal side stiffness of the MF ILMs was markedly higher than that of the IMH ILMs (3.520 ± 0.803 vs. 0.879 ± 0.230 MPa, P < 0.0001). Comparing with the IMH group, collagen IV exhibited decreased concentration and different immunofluorescence distribution in ILMs of MF eyes, so also protein α3 (IV), α4 (IV), and α5 (IV). The immunofluorescence staining results showed that astrocytes were observed in none of the IMH eyes and in 12 of 16 MF eyes (75%, P < 0.0001). Conclusions These alterations in the MF ILMs appear to be associated with Müller cell and astrocyte reactive gliosis. The present findings contribute to a more in-depth understanding of the pathogenesis of MF.
Collapse
Affiliation(s)
- Lu Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yantao Wei
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xuezhi Zhou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Zhaotian Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Wei Chi
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Li Gong
- Instrumental Analysis and Research Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xintong Jiang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Shaochong Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| |
Collapse
|
29
|
Zhao H, Darden J, Chappell JC. Establishment and characterization of an embryonic pericyte cell line. Microcirculation 2018; 25:e12461. [PMID: 29770525 DOI: 10.1111/micc.12461] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 05/07/2018] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Pericytes are specialized perivascular cells embedded within the basement membrane. These cells envelope the abluminal surface of endothelial cells and promote microvessel homeostasis. Recent discoveries of unique pericyte functions, particularly in neural tissues, underscore the need for overcoming existing challenges in establishing a functionally validated pericyte cell line. Here, we present methodologies for addressing these challenges as well as an embryonic pericyte cell line for use with in vitro and ex vivo experimental models. METHODS We isolated an enriched population of NG2:DsRed+ pericytes from E12.5 mice. This pericyte cell line was compared to MEFs with respect to gene expression, cell morphology and migration, and engagement with endothelial cells during junction stabilization and angiogenesis. RESULTS NG2+ pericytes displayed gene expression patterns, cell morphology, and 2D migration behaviors distinct from MEFs. In three different vessel formation models, pericytes from this line migrated to and incorporated into developing vessels. When co-cultured with HUVECs, these pericytes stimulated more robust VE-Cadherin junctions between HUVECs as compared to MEFs, as well as contributed to HUVEC organization into primitive vascular structures. CONCLUSIONS Our data support use of this pericyte cell line in a broad range of models to further understand pericyte functionality during normal and pathological conditions.
Collapse
Affiliation(s)
- Huaning Zhao
- Center for Heart and Regenerative Medicine, Virginia Tech Carilion Research Institute, Roanoke, VA, USA.,Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Jordan Darden
- Center for Heart and Regenerative Medicine, Virginia Tech Carilion Research Institute, Roanoke, VA, USA.,Graduate Program in Translational Biology, Medicine, and Health, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - John C Chappell
- Center for Heart and Regenerative Medicine, Virginia Tech Carilion Research Institute, Roanoke, VA, USA.,Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA.,Graduate Program in Translational Biology, Medicine, and Health, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA.,Department of Basic Science Education, Virginia Tech Carilion School of Medicine, Roanoke, VA, USA
| |
Collapse
|
30
|
Abstract
The basement membrane is a thin but dense, sheet-like specialized type of extracellular matrix that has remarkably diverse functions tailored to individual tissues and organs. Tightly controlled spatial and temporal changes in its composition and structure contribute to the diversity of basement membrane functions. These different basement membranes undergo dynamic transformations throughout animal life, most notably during development. Numerous developmental mechanisms are regulated or mediated by basement membranes, often by a combination of molecular and mechanical processes. A particularly important process involves cell transmigration through a basement membrane because of its link to cell invasion in disease. While developmental and disease processes share some similarities, what clearly distinguishes the two is dysregulation of cells and extracellular matrices in disease. With its relevance to many developmental and disease processes, the basement membrane is a vitally important area of research that may provide novel insights into biological mechanisms and development of innovative therapeutic approaches. Here we present a review of developmental and disease dynamics of basement membranes in Caenorhabditis elegans, Drosophila, and vertebrates.
Collapse
|
31
|
Abstract
Edema is typically presented as a secondary effect from injury, illness, disease, or medication, and its impact on patient wellness is nested within the underlying etiology. Therefore, it is often thought of more as an amplifier to current preexisting conditions. Edema, however, can be an independent risk factor for patient deterioration. Improper management of edema is costly not only to the patient, but also to treatment and care facilities, as mismanagement of edema results in increased lengths of hospital stay. Direct tissue trauma, disease, or inappropriate resuscitation and/or ventilation strategies result in edema formation through physical disruption and chemical messenger-based structural modifications of the microvascular barrier. Derangements in microvascular barrier function limit tissue oxygenation, nutrient flow, and cellular waste removal. Recent studies have sought to elucidate cellular signaling and structural alterations that result in vascular hyperpermeability in a variety of critical care conditions to include hemorrhage, burn trauma, and sepsis. These studies and many others have highlighted how multiple mechanisms alter paracellular and/or transcellular pathways promoting hyperpermeability. Roles for endothelial glycocalyx, extracellular matrix and basement membrane, vesiculo-vacuolar organelles, cellular junction and cytoskeletal proteins, and vascular pericytes have been described, demonstrating the complexity of microvascular barrier regulation. Understanding these basic mechanisms inside and out of microvessels aid in developing better treatment strategies to mitigate the harmful effects of excessive edema formation.
Collapse
|
32
|
Halfter W, Moes S, Asgeirsson DO, Halfter K, Oertle P, Melo Herraiz E, Plodinec M, Jenoe P, Henrich PB. Diabetes-related changes in the protein composition and the biomechanical properties of human retinal vascular basement membranes. PLoS One 2017; 12:e0189857. [PMID: 29284024 PMCID: PMC5746242 DOI: 10.1371/journal.pone.0189857] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Accepted: 12/04/2017] [Indexed: 12/18/2022] Open
Abstract
Basement membranes (BMs) are specialized sheets of extracellular matrix that outline epithelial cell layers, muscle fibers, blood vessels, and peripheral nerves. A well-documented histological hallmark of progressing diabetes is a major increase in vascular BM thickness. In order to investigate whether this structural change is accompanied by a change in the protein composition, we compared the proteomes of retinal vascular BMs from diabetic and non-diabetic donors by using LC-MS/MS. Data analysis showed that seventeen extracellular matrix (ECM)-associated proteins were more abundant in diabetic than non-diabetic vascular BMs. Four ECM proteins were more abundant in non-diabetic than in diabetic BMs. Most of the over-expressed proteins implicate a complement-mediated chronic inflammatory process in the diabetic retinal vasculature. We also found an up-regulation of norrin, a protein that is known to promote vascular proliferation, possibly contributing to the vascular remodeling during diabetes. Many of the over-expressed proteins were localized to microvascular aneurisms. Further, the overall stoichiometry of proteins was changed, such that the relative abundance of collagens in BMs from diabetic patients was higher than normal. Biomechanical measurements of vascular BM flat mounts using AFM showed that their outer surface was softer than normal.
Collapse
Affiliation(s)
- Willi Halfter
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - Suzette Moes
- Proteomics Core Facility, Biocenter of the University of Basel, Basel, Switzerland
| | - Daphne O. Asgeirsson
- Biocenter and the Swiss Nanoscience Institute, University of Basel, Basel, Switzerland
| | - Kathrin Halfter
- Institute of Medical Informatics, Biometry and Epidemiology, Maximilian University Munich, Munich, Germany
| | - Philipp Oertle
- Biocenter and the Swiss Nanoscience Institute, University of Basel, Basel, Switzerland
| | - Esther Melo Herraiz
- Biocenter and the Swiss Nanoscience Institute, University of Basel, Basel, Switzerland
| | - Marija Plodinec
- Biocenter and the Swiss Nanoscience Institute, University of Basel, Basel, Switzerland
| | - Paul Jenoe
- Proteomics Core Facility, Biocenter of the University of Basel, Basel, Switzerland
| | | |
Collapse
|
33
|
Fehér J, Taurone S, Spoletini M, Biró Z, Varsányi B, Scuderi G, Orlando MP, Turchetta R, Micera A, Artico M. Ultrastructure of neurovascular changes in human diabetic retinopathy. Int J Immunopathol Pharmacol 2017; 31:394632017748841. [PMID: 29251013 PMCID: PMC5849217 DOI: 10.1177/0394632017748841] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
The previous concept regarding diabetic retinopathy assigned a primary role to
hyperglycemia-induced microvascular alterations, while neuronal and glial
abnormalities were considered to be secondary to either ischemia or exudation.
The aim of this study was to reveal the potential role of neuronal and glial
cells in initial and advanced alterations of the retinopathy in human type 2
diabetes. Electron microscopy and histochemical studies were performed on 38
surgically removed human eyes (28 obtained from diabetic patients and 10 from
non-diabetic patients). Morphometric analysis of basement membrane material and
lipids was performed. An accumulation of metabolic by-products was found in the
capillary wall with aging: this aspect was significantly more pronounced in
diabetics. Müller glial cells were found to contribute to alterations of the
capillary wall and to occlusion, as well as to the development of proliferative
retinopathy and cystoid degeneration of the retina. Our results showed
morphological evidence regarding the role of neuronal and glial cells in the
pathology of diabetic retinopathy, prior and in addition to microangiopathy.
These morphological findings support a neurovascular pathogenesis at the origin
of diabetic retinopathy, thus the current treatment approach should be completed
by neuroprotective measures.
Collapse
Affiliation(s)
- János Fehér
- 1 Ophthalmic Neuroscience Program, Nutripharma Hungaria Ltd, Budapest, Hungary
| | | | | | - Zsolt Biró
- 4 Department of Ophthalmology, University of Pécs, Pécs, Hungary
| | - Balázs Varsányi
- 4 Department of Ophthalmology, University of Pécs, Pécs, Hungary
| | - Gianluca Scuderi
- 5 Ophthalmology Unit, NESMOS Department, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | | | - Rosaria Turchetta
- 3 Department of Sensory Organs, Sapienza University of Rome, Rome, Italy
| | | | - Marco Artico
- 3 Department of Sensory Organs, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
34
|
Reinhard J, Roll L, Faissner A. Tenascins in Retinal and Optic Nerve Neurodegeneration. Front Integr Neurosci 2017; 11:30. [PMID: 29109681 PMCID: PMC5660115 DOI: 10.3389/fnint.2017.00030] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 10/03/2017] [Indexed: 02/04/2023] Open
Abstract
Tenascins represent key constituents of the extracellular matrix (ECM) with major impact on central nervous system (CNS) development. In this regard, several studies indicate that they play a crucial role in axonal growth and guidance, synaptogenesis and boundary formation. These functions are not only important during development, but also for regeneration under several pathological conditions. Additionally, tenascin-C (Tnc) represents a key modulator of the immune system and inflammatory processes. In the present review article, we focus on the function of Tnc and tenascin-R (Tnr) in the diseased CNS, specifically after retinal and optic nerve damage and degeneration. We summarize the current view on both tenascins in diseases such as glaucoma, retinal ischemia, age-related macular degeneration (AMD) or diabetic retinopathy. In this context, we discuss their expression profile, possible functional relevance, remodeling of the interacting matrisome and tenascin receptors, especially under pathological conditions.
Collapse
Affiliation(s)
- Jacqueline Reinhard
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Bochum, Germany
| | - Lars Roll
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Bochum, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
35
|
Mondrinos MJ, Yi YS, Wu NK, Ding X, Huh D. Native extracellular matrix-derived semipermeable, optically transparent, and inexpensive membrane inserts for microfluidic cell culture. LAB ON A CHIP 2017; 17:3146-3158. [PMID: 28809418 PMCID: PMC5782796 DOI: 10.1039/c7lc00317j] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Semipermeable cell culture membranes are commonly used in multilayered microfluidic devices to mimic the basement membrane in vivo and to create compartmentalized microenvironments for physiological cell growth and differentiation. However, existing membranes are predominantly made up of synthetic polymers, providing limited capacity to replicate cellular interactions with native extracellular matrices that play a crucial role in the induction of physiological phenotypes. Here we describe a new type of cell culture membranes engineered from native extracellular matrix (ECM) materials that are thin, semipermeable, optically transparent, and amenable to integration into microfluidic cell culture devices. Facile and cost-effective fabrication of these membranes was achieved by controlled sequential steps of vitrification that transformed three-dimensional (3D) ECM hydrogels into structurally stable thin films. By modulating the composition of the ECM, our technique provided a means to tune key membrane properties such as optical transparency, stiffness, and porosity. For microfluidic cell culture, we constructed a multilayered microdevice consisting of two parallel chambers separated by a thin membrane insert derived from different types of ECM. This study showed that our ECM membranes supported attachment and growth of various types of cells (epithelial, endothelial, and mesenchymal cells) under perfusion culture conditions. Our data also revealed the promotive effects of the membranes on adhesion-associated intracellular signaling that mediates cell-ECM interactions. Moreover, we demonstrated the use of these membranes for constructing compartmentalized microfluidic cell culture systems to induce physiological tissue differentiation or to replicate interfaces between different tissue types. Our approach provides a robust platform to produce and engineer biologically active cell culture substrates that serve as promising alternatives to conventional synthetic membrane inserts. This strategy may contribute to the development of physiologically relevant in vitro cell culture models for a wide range of applications.
Collapse
Affiliation(s)
- Mark J Mondrinos
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, PA, USA.
| | | | | | | | | |
Collapse
|
36
|
Carreon TA, Edwards G, Wang H, Bhattacharya SK. Segmental outflow of aqueous humor in mouse and human. Exp Eye Res 2017; 158:59-66. [PMID: 27498226 PMCID: PMC5290258 DOI: 10.1016/j.exer.2016.08.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Revised: 07/08/2016] [Accepted: 08/01/2016] [Indexed: 12/28/2022]
Abstract
The main and only modifiable risk factor in glaucoma, the group of usually late onset progressive and irreversible blinding optic neuropathies, is elevated intraocular pressure (IOP). The increase in IOP is due to impeded aqueous humor (AH) outflow through the conventional pathway. The aberrant increased resistance at the trabecular meshwork (TM), the filter-like region in the anterior eye chamber is the major contributory factor in causing the impeded outflow. In normal as well as in glaucoma eyes the regions of the TM are divided into areas of high and low flow. The collector channels and distal outflow regions are now increasingly being recognized as potential players in contributing to impede AH outflow. Structural and molecular make-up contributing to the segmental blockage to outflow is likely to provide greater insight. Establishing segmental blockage to outflow in model systems of glaucoma such as the mouse in parallel to human eyes will expand our repertoire of tools for investigation. Further study into this area of interest has the potential to ultimately lead to the development of new therapeutics focused on lowering IOP by targeting the various components of segmental blockage of outflow in the TM and in the distal outflow region.
Collapse
Affiliation(s)
- Teresia A Carreon
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA; Department of Biochemistry and Molecular Biology, University of Miami, Miami, FL, 33136, USA
| | - Genea Edwards
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA; Department of Biochemistry and Molecular Biology, University of Miami, Miami, FL, 33136, USA
| | - Haiyan Wang
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA; Shanghai First People's Hospital Affiliated to Jiaotong University, Shanghai, 200080, China
| | - Sanjoy K Bhattacharya
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA; Department of Biochemistry and Molecular Biology, University of Miami, Miami, FL, 33136, USA.
| |
Collapse
|
37
|
Abstract
Retinal fibrosis, characterized by dysregulation of extracellular matrix (ECM) protein deposition by retinal endothelial cells, pigment epithelial cells, and other resident cell-types, is a unifying feature of several common retinal diseases. Fibronectin is an early constituent of newly deposited ECM and serves as a template for assembly of other ECM proteins, including collagens. Under physiologic conditions, fibronectin is found in all layers of Bruch's membrane. Proliferative vitreoretinopathy (PVR), a complication of retinal surgery, is characterized by ECM accumulation. Among the earliest histologic manifestations of diabetic retinopathy (DR) is capillary basement membrane thickening, which occurs due to perturbations in ECM homeostasis. Neovascularization, the hallmark of late stage DR as well as exudative age-related macular degeneration (AMD), involves ECM assembly as a scaffold for the aberrant new vessel architecture. Rodent models of retinal injury demonstrate a key role for fibronectin in complications characteristic of PVR, including retinal detachment. In mouse models of DR, reducing fibronectin gene expression has been shown to arrest the accumulation of ECM in the capillary basement membrane. Alterations in matrix metalloproteinase activity thought to be important in the pathogenesis of AMD impact the turnover of fibronectin matrix as well as collagens. Growth factors involved in PVR, AMD, and DR, such as PDGF and TGFβ, are known to stimulate fibronectin matrix assembly. A deeper understanding of how pathologic ECM deposition contributes to disease progression may help to identify novel targets for therapeutic intervention.
Collapse
Affiliation(s)
- Charles G Miller
- Department of Ophthalmology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ 08854-5635, USA
| | - Greg Budoff
- Department of Ophthalmology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ 08854-5635, USA
| | - Jonathan L Prenner
- Department of Ophthalmology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ 08854-5635, USA
- NJ Retina, New Brunswick, NJ 08901-2066, USA
| | - Jean E Schwarzbauer
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544-1014, USA
| |
Collapse
|
38
|
Del Amo EM, Rimpelä AK, Heikkinen E, Kari OK, Ramsay E, Lajunen T, Schmitt M, Pelkonen L, Bhattacharya M, Richardson D, Subrizi A, Turunen T, Reinisalo M, Itkonen J, Toropainen E, Casteleijn M, Kidron H, Antopolsky M, Vellonen KS, Ruponen M, Urtti A. Pharmacokinetic aspects of retinal drug delivery. Prog Retin Eye Res 2016; 57:134-185. [PMID: 28028001 DOI: 10.1016/j.preteyeres.2016.12.001] [Citation(s) in RCA: 410] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 11/25/2016] [Accepted: 12/01/2016] [Indexed: 12/14/2022]
Abstract
Drug delivery to the posterior eye segment is an important challenge in ophthalmology, because many diseases affect the retina and choroid leading to impaired vision or blindness. Currently, intravitreal injections are the method of choice to administer drugs to the retina, but this approach is applicable only in selected cases (e.g. anti-VEGF antibodies and soluble receptors). There are two basic approaches that can be adopted to improve retinal drug delivery: prolonged and/or retina targeted delivery of intravitreal drugs and use of other routes of drug administration, such as periocular, suprachoroidal, sub-retinal, systemic, or topical. Properties of the administration route, drug and delivery system determine the efficacy and safety of these approaches. Pharmacokinetic and pharmacodynamic factors determine the required dosing rates and doses that are needed for drug action. In addition, tolerability factors limit the use of many materials in ocular drug delivery. This review article provides a critical discussion of retinal drug delivery, particularly from the pharmacokinetic point of view. This article does not include an extensive review of drug delivery technologies, because they have already been reviewed several times recently. Instead, we aim to provide a systematic and quantitative view on the pharmacokinetic factors in drug delivery to the posterior eye segment. This review is based on the literature and unpublished data from the authors' laboratory.
Collapse
Affiliation(s)
- Eva M Del Amo
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Anna-Kaisa Rimpelä
- Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland
| | - Emma Heikkinen
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Otto K Kari
- Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland
| | - Eva Ramsay
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Tatu Lajunen
- Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland
| | - Mechthild Schmitt
- Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland
| | - Laura Pelkonen
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Madhushree Bhattacharya
- Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland
| | - Dominique Richardson
- Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland
| | - Astrid Subrizi
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Tiina Turunen
- Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland
| | - Mika Reinisalo
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Jaakko Itkonen
- Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland
| | - Elisa Toropainen
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Marco Casteleijn
- Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland
| | - Heidi Kidron
- Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland
| | - Maxim Antopolsky
- Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland
| | | | - Marika Ruponen
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Arto Urtti
- Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland; School of Pharmacy, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
39
|
Schwarz C, Aldrich BT, Burckart KA, Schmidt GA, Zimmerman MB, Reed CR, Greiner MA, Sander EA. Descemet membrane adhesion strength is greater in diabetics with advanced disease compared to healthy donor corneas. Exp Eye Res 2016; 153:152-158. [PMID: 27777123 DOI: 10.1016/j.exer.2016.10.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 10/19/2016] [Accepted: 10/20/2016] [Indexed: 11/27/2022]
Abstract
Descemet membrane endothelial keratoplasty (DMEK) is an increasingly popular surgical procedure for treating ocular diseases that require a corneal transplant. Previous studies have found that tissue tearing during surgical preparation is more likely elevated in eyes from donors with a history of diabetes mellitus. To quantify these potential differences, we established an experimental technique for quantifying the force required to separate the endothelium-Descemet membrane complex (EDM) from stroma in human donor corneal tissue, and we assessed differences in adhesion strength between diabetic and non-diabetic donor corneas. Transplant suitable corneas were obtained from 23 donors 50-75 years old with an average preservation to assay time of 11.5 days. Corneas were classified from a medical records review as non-diabetic (ND, n = 9), diabetic without evidence of advanced disease (NAD, n = 8), or diabetic with evidence of advanced disease (AD, n = 10). Corneas were sectioned into 3 mm wide strips and the EDM peeled from the stroma. Using the force-extension data obtained from mechanical peel testing, EDM elastic peel tension (TE), elastic stiffness (SE), average delamination tension (TD), and maximum tension (TMAX) were calculated. Mean TE, SE, TD, and TMAX values for ND corneas were 0.78 ± 0.07 mN/mm, 0.37 ± 0.05 mN/mm/mm, 0.78 ± 0.08 mN/mm, and 0.94 ± 0.17 mN/mm, respectively. NAD values did not differ significantly. However, AD values for TE (1.01 ± 0.18 mN/mm), TD (1.09 ± 0.21 mN/mm), and TMAX (1.37 ± 0.24 mN/mm) were greater than ND and NAD corneas (P < 0.05). SE did not differ significantly between groups. These findings provide proof of the concept that chronic hyperglycemia from diabetes mellitus results in a phenotypically more adhesive interface between Descemet membrane and the posterior stroma in donor corneal tissue. Results of this study provide a foundation for further investigations into the impact of diabetes on the posterior cornea, eye banking, and keratoplasty.
Collapse
Affiliation(s)
- Chaid Schwarz
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA
| | - Benjamin T Aldrich
- Department of Ophthalmology and Visual Sciences, University of Iowa Carver College of Medicine, Iowa City, IA, USA; Iowa Lions Eye Bank, Coralville, IA, USA
| | | | | | - M Bridget Zimmerman
- College of Public Health, Department of Biostatistics, University of Iowa, Iowa City, IA, USA
| | | | - Mark A Greiner
- Department of Ophthalmology and Visual Sciences, University of Iowa Carver College of Medicine, Iowa City, IA, USA; Iowa Lions Eye Bank, Coralville, IA, USA; Cornea Research Center, Stephen A. Wynn Institute for Vision Research, Iowa City, IA, USA.
| | - Edward A Sander
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
40
|
Rodriguez-Teja M, Breit C, Clarke M, Talar K, Wang K, Mohammad MA, Pickwell S, Etchandy G, Stasiuk GJ, Sturge J. How to Study Basement Membrane Stiffness as a Biophysical Trigger in Prostate Cancer and Other Age-related Pathologies or Metabolic Diseases. J Vis Exp 2016. [PMID: 27684203 PMCID: PMC5092048 DOI: 10.3791/54230] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Here we describe a protocol that can be used to study the biophysical microenvironment related to increased thickness and stiffness of the basement membrane (BM) during age-related pathologies and metabolic disorders (e.g. cancer, diabetes, microvascular disease, retinopathy, nephropathy and neuropathy). The premise of the model is non-enzymatic crosslinking of reconstituted BM (rBM) matrix by treatment with glycolaldehyde (GLA) to promote advanced glycation endproduct (AGE) generation via the Maillard reaction. Examples of laboratory techniques that can be used to confirm AGE generation, non-enzymatic crosslinking and increased stiffness in GLA treated rBM are outlined. These include preparation of native rBM (treated with phosphate-buffered saline, PBS) and stiff rBM (treated with GLA) for determination of: its AGE content by photometric analysis and immunofluorescent microscopy, its non-enzymatic crosslinking by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS PAGE) as well as confocal microscopy, and its increased stiffness using rheometry. The procedure described here can be used to increase the rigidity (elastic moduli, E) of rBM up to 3.2-fold, consistent with measurements made in healthy versus diseased human prostate tissue. To recreate the biophysical microenvironment associated with the aging and diseased prostate gland three prostate cell types were introduced on to native rBM and stiff rBM: RWPE-1, prostate epithelial cells (PECs) derived from a normal prostate gland; BPH-1, PECs derived from a prostate gland affected by benign prostatic hyperplasia (BPH); and PC3, metastatic cells derived from a secondary bone tumor originating from prostate cancer. Multiple parameters can be measured, including the size, shape and invasive characteristics of the 3D glandular acini formed by RWPE-1 and BPH-1 on native versus stiff rBM, and average cell length, migratory velocity and persistence of cell movement of 3D spheroids formed by PC3 cells under the same conditions. Cell signaling pathways and the subcellular localization of proteins can also be assessed.
Collapse
Affiliation(s)
| | - Claudia Breit
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology
| | - Mitchell Clarke
- School of Biological, Biomedical & Environmental Sciences, University of Hull
| | - Kamil Talar
- School of Biological, Biomedical & Environmental Sciences, University of Hull
| | - Kai Wang
- School of Biological, Biomedical & Environmental Sciences, University of Hull
| | - Mohammad A Mohammad
- School of Biological, Biomedical & Environmental Sciences, University of Hull
| | - Sage Pickwell
- School of Biological, Biomedical & Environmental Sciences, University of Hull
| | - Guillermina Etchandy
- Departamento de Genética, Facultad de Medicina, Universidad de la República (UDELAR)
| | - Graeme J Stasiuk
- School of Biological, Biomedical & Environmental Sciences, University of Hull
| | - Justin Sturge
- School of Biological, Biomedical & Environmental Sciences, University of Hull;
| |
Collapse
|
41
|
Yang X, Scott HA, Monickaraj F, Xu J, Ardekani S, Nitta CF, Cabrera A, McGuire PG, Mohideen U, Das A, Ghosh K. Basement membrane stiffening promotes retinal endothelial activation associated with diabetes. FASEB J 2016; 30:601-11. [PMID: 26443820 PMCID: PMC6188223 DOI: 10.1096/fj.15-277962] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 09/21/2015] [Indexed: 12/21/2022]
Abstract
Endothelial activation is a hallmark of the high-glucose (HG)-induced retinal inflammation associated with diabetic retinopathy (DR). However, precisely how HG induces retinal endothelial activation is not fully understood. We hypothesized that HG-induced up-regulation of lysyl oxidase (LOX), a collagen-cross-linking enzyme, in retinal capillary endothelial cells (ECs) enhances subendothelial basement membrane (BM) stiffness, which, in turn, promotes retinal EC activation. Diabetic C57BL/6 mice exhibiting a 70 and 50% increase in retinal intercellular adhesion molecule (ICAM)-1 expression and leukocyte accumulation, respectively, demonstrated a 2-fold increase in the levels of BM collagen IV and LOX, key determinants of capillary BM stiffness. Using atomic force microscopy, we confirmed that HG significantly enhances LOX-dependent subendothelial matrix stiffness in vitro, which correlated with an ∼2.5-fold increase in endothelial ICAM-1 expression, a 4-fold greater monocyte-EC adhesion, and an ∼2-fold alteration in endothelial NO (decrease) and NF-κB activation (increase). Inhibition of LOX-dependent subendothelial matrix stiffening alone suppressed HG-induced retinal EC activation. Finally, using synthetic matrices of tunable stiffness, we demonstrated that subendothelial matrix stiffening is necessary and sufficient to promote EC activation. These findings implicate BM stiffening as a critical determinant of HG-induced retinal EC activation and provide a rationale for examining BM stiffness and underlying mechanotransduction pathways as therapeutic targets for diabetic retinopathy.
Collapse
Affiliation(s)
- Xiao Yang
- *Department of Bioengineering and Department of Physics and Astronomy, University of California, Riverside, Riverside, California, USA; Department of Surgery and Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, New Mexico, USA; and New Mexico Veterans Affairs Health Care System, Albuquerque, New Mexico, USA
| | - Harry A Scott
- *Department of Bioengineering and Department of Physics and Astronomy, University of California, Riverside, Riverside, California, USA; Department of Surgery and Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, New Mexico, USA; and New Mexico Veterans Affairs Health Care System, Albuquerque, New Mexico, USA
| | - Finny Monickaraj
- *Department of Bioengineering and Department of Physics and Astronomy, University of California, Riverside, Riverside, California, USA; Department of Surgery and Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, New Mexico, USA; and New Mexico Veterans Affairs Health Care System, Albuquerque, New Mexico, USA
| | - Jun Xu
- *Department of Bioengineering and Department of Physics and Astronomy, University of California, Riverside, Riverside, California, USA; Department of Surgery and Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, New Mexico, USA; and New Mexico Veterans Affairs Health Care System, Albuquerque, New Mexico, USA
| | - Soroush Ardekani
- *Department of Bioengineering and Department of Physics and Astronomy, University of California, Riverside, Riverside, California, USA; Department of Surgery and Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, New Mexico, USA; and New Mexico Veterans Affairs Health Care System, Albuquerque, New Mexico, USA
| | - Carolina F Nitta
- *Department of Bioengineering and Department of Physics and Astronomy, University of California, Riverside, Riverside, California, USA; Department of Surgery and Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, New Mexico, USA; and New Mexico Veterans Affairs Health Care System, Albuquerque, New Mexico, USA
| | - Andrea Cabrera
- *Department of Bioengineering and Department of Physics and Astronomy, University of California, Riverside, Riverside, California, USA; Department of Surgery and Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, New Mexico, USA; and New Mexico Veterans Affairs Health Care System, Albuquerque, New Mexico, USA
| | - Paul G McGuire
- *Department of Bioengineering and Department of Physics and Astronomy, University of California, Riverside, Riverside, California, USA; Department of Surgery and Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, New Mexico, USA; and New Mexico Veterans Affairs Health Care System, Albuquerque, New Mexico, USA
| | - Umar Mohideen
- *Department of Bioengineering and Department of Physics and Astronomy, University of California, Riverside, Riverside, California, USA; Department of Surgery and Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, New Mexico, USA; and New Mexico Veterans Affairs Health Care System, Albuquerque, New Mexico, USA
| | - Arup Das
- *Department of Bioengineering and Department of Physics and Astronomy, University of California, Riverside, Riverside, California, USA; Department of Surgery and Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, New Mexico, USA; and New Mexico Veterans Affairs Health Care System, Albuquerque, New Mexico, USA
| | - Kaustabh Ghosh
- *Department of Bioengineering and Department of Physics and Astronomy, University of California, Riverside, Riverside, California, USA; Department of Surgery and Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, New Mexico, USA; and New Mexico Veterans Affairs Health Care System, Albuquerque, New Mexico, USA
| |
Collapse
|
42
|
Ulusoy S, Ozkan G, Menteşe A, Guvercin B, Caner Karahan S, Yavuz A, Altay DU. A new predictor of mortality in hemodialysis patients; Tenascin-C. Life Sci 2015; 141:54-60. [DOI: 10.1016/j.lfs.2015.09.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 09/15/2015] [Accepted: 09/18/2015] [Indexed: 12/16/2022]
|
43
|
Halfter W, Oertle P, Monnier CA, Camenzind L, Reyes-Lua M, Hu H, Candiello J, Labilloy A, Balasubramani M, Henrich PB, Plodinec M. New concepts in basement membrane biology. FEBS J 2015; 282:4466-79. [PMID: 26299746 DOI: 10.1111/febs.13495] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 07/13/2015] [Accepted: 08/18/2015] [Indexed: 12/24/2022]
Abstract
Basement membranes (BMs) are thin sheets of extracellular matrix that outline epithelia, muscle fibers, blood vessels and peripheral nerves. The current view of BM structure and functions is based mainly on transmission electron microscopy imaging, in vitro protein binding assays, and phenotype analysis of human patients, mutant mice and invertebrata. Recently, MS-based protein analysis, biomechanical testing and cell adhesion assays with in vivo derived BMs have led to new and unexpected insights. Proteomic analysis combined with ultrastructural studies showed that many BMs undergo compositional and structural changes with advancing age. Atomic force microscopy measurements in combination with phenotype analysis have revealed an altered mechanical stiffness that correlates with specific BM pathologies in mutant mice and human patients. Atomic force microscopy-based height measurements strongly suggest that BMs are more than two-fold thicker than previously estimated, providing greater freedom for modelling the large protein polymers within BMs. In addition, data gathered using BMs extracted from mutant mice showed that laminin has a crucial role in BM stability. Finally, recent evidence demonstrate that BMs are bi-functionally organized, leading to the proposition that BM-sidedness contributes to the alternating epithelial and stromal tissue arrangements that are found in all metazoan species. We propose that BMs are ancient structures with tissue-organizing functions and were essential in the evolution of metazoan species.
Collapse
Affiliation(s)
- Willi Halfter
- Department of Ophthalmology, University Hospital Basel, Switzerland
| | - Philipp Oertle
- Biozentrum and the Swiss Nanoscience Institute, University of Basel, Switzerland
| | - Christophe A Monnier
- Biozentrum and the Swiss Nanoscience Institute, University of Basel, Switzerland
| | - Leon Camenzind
- Biozentrum and the Swiss Nanoscience Institute, University of Basel, Switzerland
| | - Magaly Reyes-Lua
- Department of Ophthalmology, University Hospital Basel, Switzerland
| | - Huaiyu Hu
- Department of Neurobiology and Physiology, Upstate University Hospital, SUNY University, Syracuse, NY, USA
| | | | | | | | | | - Marija Plodinec
- Biozentrum and the Swiss Nanoscience Institute, University of Basel, Switzerland.,Department of Pathology, University Hospital Basel, Switzerland
| |
Collapse
|
44
|
Roy S, Bae E, Amin S, Kim D. Extracellular matrix, gap junctions, and retinal vascular homeostasis in diabetic retinopathy. Exp Eye Res 2015; 133:58-68. [PMID: 25819455 DOI: 10.1016/j.exer.2014.08.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 08/19/2014] [Accepted: 08/20/2014] [Indexed: 12/15/2022]
Abstract
The vascular basement membrane (BM) contains extracellular matrix (ECM) proteins that assemble in a highly organized manner to form a supportive substratum for cell attachment facilitating myriad functions that are vital to cell survival and overall retinal homeostasis. The BM provides a microenvironment in which bidirectional signaling through integrins regulates cell attachment, turnover, and functionality. In diabetic retinopathy, the BM undergoes profound structural and functional changes, and recent studies have brought to light the implications of such changes. Thickened vascular BM in the retinal capillaries actively participate in the development and progression of characteristic changes associated with diabetic retinopathy. High glucose (HG)-induced compromised cell-cell communication via gap junctions (GJ) in retinal vascular cells may disrupt homeostasis in the retinal microenvironment. In this review, the role of altered ECM synthesis, compromised GJ activity, and disturbed retinal homeostasis in the development of retinal vascular lesions in diabetic retinopathy are discussed.
Collapse
Affiliation(s)
- Sayon Roy
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA; Department of Ophthalmology, Boston University School of Medicine, Boston, MA, USA.
| | - Edward Bae
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA; Department of Ophthalmology, Boston University School of Medicine, Boston, MA, USA
| | - Shruti Amin
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA; Department of Ophthalmology, Boston University School of Medicine, Boston, MA, USA
| | - Dongjoon Kim
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA; Department of Ophthalmology, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
45
|
Panagiotoglou T, Tsilimbaris M, Ginis H, Karyotakis N, Georgiou V, Koutentakis P, Pallikaris I. Ocular rigidity and outflow facility in nonproliferative diabetic retinopathy. J Diabetes Res 2015; 2015:141598. [PMID: 25954761 PMCID: PMC4411434 DOI: 10.1155/2015/141598] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2014] [Revised: 02/19/2015] [Accepted: 03/05/2015] [Indexed: 11/17/2022] Open
Abstract
PURPOSE To compare ocular rigidity (OR) and outflow facility (C) in patients with nonproliferative diabetic retinopathy (NPDR) and control subjects. METHODS Twenty-four patients with NPDR (NPDR group) and 24 controls (control group) undergoing cataract surgery were enrolled. NPDR group was further divided into patients with mild NPDR (NPDR1-group) and patients with moderate and/or severe NPDR (NPDR2-group). After cannulation of the anterior chamber, a computer-controlled device was used to infuse saline and increase the intraocular pressure (IOP) in a stepping procedure from 15 to 40 mmHg. Ocular rigidity and outflow facility coefficients were estimated from IOP and volume recordings. RESULTS Ocular rigidity was 0.0205 μL(-1) in NPDR group and 0.0202 μL(-1) in control group (P = 0.942). In NPDR1-group, OR was 0.017 μL(-1) and in NPDR2-group it was 0.025 μL(-1) (P = 0.192). Outflow facility was 0.120 μL/min/mmHg in NPDR-group compared to 0.153 μL/min/mmHg in the control group at an IOP of 35 mmHg (P = 0.151). There was no difference in C between NPDR1-group and NPDR2-group (P = 0.709). CONCLUSIONS No statistically significant differences in ocular rigidity and outflow facility could be documented between diabetic patients and controls. No difference in OR and C was detected between mild NPDR and severe NPDR.
Collapse
Affiliation(s)
- Theonitsa Panagiotoglou
- Department of Ophthalmology, University Hospital of Heraklion, 71110 Heraklion, Greece
- Department of Ophthalmology, Venizeleio General Hospital of Heraklion, 71409 Heraklion, Greece
| | - Miltiadis Tsilimbaris
- Department of Ophthalmology, University Hospital of Heraklion, 71110 Heraklion, Greece
- Institute of Vision and Optics, School of Medicine, University of Crete, 71110 Heraklion, Greece
- *Miltiadis Tsilimbaris:
| | - Harilaos Ginis
- Institute of Vision and Optics, School of Medicine, University of Crete, 71110 Heraklion, Greece
| | - Nikos Karyotakis
- Institute of Vision and Optics, School of Medicine, University of Crete, 71110 Heraklion, Greece
| | - Vaggelis Georgiou
- Department of Social Medicine, School of Medicine, University of Crete, 71110 Heraklion, Greece
| | - Pavlos Koutentakis
- Department of Ophthalmology, Venizeleio General Hospital of Heraklion, 71409 Heraklion, Greece
| | - Ioannis Pallikaris
- Department of Ophthalmology, University Hospital of Heraklion, 71110 Heraklion, Greece
- Institute of Vision and Optics, School of Medicine, University of Crete, 71110 Heraklion, Greece
| |
Collapse
|