1
|
Papp FR, Katko M, Csiki R, Galgoczi E, Molnar Z, Erdei A, Bodor M, Steiber Z, Ujhelyi B, Nagy EV. Characteristics of Hyaluronan Metabolism During Myofibroblast Differentiation in Orbital Fibroblasts. Invest Ophthalmol Vis Sci 2024; 65:13. [PMID: 39504052 PMCID: PMC11549924 DOI: 10.1167/iovs.65.13.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/12/2024] [Indexed: 11/11/2024] Open
Abstract
Purpose To study the impact of myofibroblast differentiation (MD) on hyaluronan (HA) turnover in orbital fibroblasts (OFs) focusing on the expression of its key enzymes and their potential implications in the pathogenesis of thyroid eye disease (TED). Methods Primary cultures of OFs were established from tissue samples (TED OFs, n = 4; non-TED OFs, n = 5). MD was induced by TGF-β1 (5 ng/mL). Measurements were performed after 24- and 72-hour treatments. The proliferation rate was determined by 5-bromo-2'-deoxyuridine (BrdU) incorporation. HA level and size were measured using an aggrecan-based ELISA-like method and agarose gel electrophoresis, respectively. mRNA expressions of myofibroblast markers and enzymes with a role in HA metabolism were determined using real-time PCR. Results Upregulation of type I collagen alpha1 chain, alpha-smooth muscle actin, and fibronectin indicated that OFs underwent MD after stimulation by TGF-β. After 72 hours, proliferation of untreated cultures declined, but it remained higher in myofibroblasts. Pericellular HA content, but not HA in the supernatant of myofibroblasts, increased compared to untreated cells. TGF-β was a potent stimulator of hyaluronan synthase 1 (HAS1) expression. The expression of hyaluronidase-1 and cell migration-inducing protein (CEMIP) diminished following MD, whereas the expression of transmembrane protein 2, the regulator of HA catabolism through CEMIP, was elevated. The size distribution of HA shifted toward a high-molecular-weight form following treatment with TGF-β. Conclusions OFs undergoing MD are characterized by decreased HA turnover as a consequence of the inhibition of hyaluronidases and HAS1 induction. Our results suggest that hyaluronidases could be potential targets in the treatment of TED.
Collapse
Affiliation(s)
- Fruzsina R. Papp
- Division of Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Doctoral School of Health Sciences, University of Debrecen, Debrecen, Hungary
| | - Monika Katko
- Division of Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Robert Csiki
- Division of Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Doctoral School of Health Sciences, University of Debrecen, Debrecen, Hungary
| | - Erika Galgoczi
- Division of Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsanett Molnar
- Division of Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Annamaria Erdei
- Division of Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Miklos Bodor
- Division of Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zita Steiber
- Department of Ophthalmology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Bernadett Ujhelyi
- Department of Ophthalmology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Endre V. Nagy
- Division of Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
2
|
Galgoczi E, Molnar Z, Katko M, Ujhelyi B, Steiber Z, Nagy EV. Cyclosporin A inhibits PDGF-BB induced hyaluronan synthesis in orbital fibroblasts. Chem Biol Interact 2024; 396:111045. [PMID: 38729283 DOI: 10.1016/j.cbi.2024.111045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 04/11/2024] [Accepted: 05/08/2024] [Indexed: 05/12/2024]
Abstract
Orbital connective tissue changes are contributors to the pathogenesis in thyroid eye disease (TED). Activated fibroblasts respond to immune stimuli with proliferation and increased hyaluronan (HA) production. Cyclosporin A (CsA) was reported to be beneficial in the treatment of TED. PDGF isoforms are increased in orbital tissue of TED patients and enhance HA production. We aimed to study the effect of CsA on HA production and hyaluronan synthase (HAS1, 2 and 3) and hyaluronidase (HYAL1 and 2) mRNA expressions in orbital fibroblasts (OFs). Measurements were performed in the presence or absence of CsA (10 μM) in unstimulated or PDGF-BB (10 ng/ml) stimulated OFs. The HA production of TED OFs (n = 7) and NON-TED OFs (n = 6) were measured by ELISA. The levels of mRNA expressions were examined using RT-PCR. The proliferation rate and metabolic activity were measured by BrdU incorporation and MTT assays, respectively. Treatment with CsA resulted in an average 42% decrease in HA production of OFs (p < 0.0001). CsA decreased the expression levels of HAS2, HAS3 and HYAL2 (p = 0.005, p = 0.005 and p = 0.002, respectively.) PDGF-BB increased HA production (p < 0.001) and HAS2 expression (p = 0.004). CsA could reduce the PDGF-BB-stimulated HA production (p < 0.001) and HAS2 expression (p = 0.005) below the untreated level. In addition, CsA treatment caused a decrease in proliferation potential (p = 0.002) and metabolic activity (p < 0.0001). These findings point to the fact that CsA affects HA metabolism via HAS2, HAS3 and HYAL2 inhibition in OFs. In addition to its well characterized immunosuppressant properties, CsA's beneficial effect in TED may be related to its direct inhibitory effect on basal and growth factor stimulated HA production.
Collapse
Affiliation(s)
- Erika Galgoczi
- Division of Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Nagyerdei krt. 98, Hungary.
| | - Zsanett Molnar
- Division of Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Nagyerdei krt. 98, Hungary.
| | - Monika Katko
- Division of Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Nagyerdei krt. 98, Hungary.
| | - Bernadett Ujhelyi
- Department of Ophthalmology, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Nagyerdei krt. 98, Hungary.
| | - Zita Steiber
- Department of Ophthalmology, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Nagyerdei krt. 98, Hungary.
| | - Endre V Nagy
- Division of Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Nagyerdei krt. 98, Hungary.
| |
Collapse
|
3
|
Li H, Min J, Yang Y, Suo W, Wang W, Tian J, Qin Y. TMEM2 inhibits the development of Graves' orbitopathy through the JAK-STAT signaling pathway. J Biol Chem 2024; 300:105607. [PMID: 38159864 PMCID: PMC10839445 DOI: 10.1016/j.jbc.2023.105607] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 11/13/2023] [Accepted: 11/30/2023] [Indexed: 01/03/2024] Open
Abstract
A mouse model was used to investigate the role of the hyaluronidase, transmembrane protein 2 (TMEM2), on the progression of Graves' orbital (GO) disease. We established a GO mouse model through immunization with a plasmid expressing the thyroid stimulating hormone receptor. Orbital fibroblasts (OFs) were subsequently isolated from both GO and non-GO mice for comprehensive in vitro analyses. The expression of TMEM2 was assessed using qRT-PCR, Western blot and immunohistochemistry in vivo. Disease pathology was evaluated by H&E staining and Masson's trichrome staining in GO mouse tissues. Our investigation revealed a notable reduction in TMEM2 expression in GO mouse orbital tissues. Through overexpression and knockdown assays, we demonstrated that TMEM2 suppresses inflammatory cytokines and reactive oxygen species production. TMEM2 also inhibits the formation of lipid droplets in OFs and the expression of adipogenic factors. Further incorporating Gene Set Enrichment Analysis of relevant GEO datasets and subsequent in vitro cell experiments, robustly confirmed that TMEM2 overexpression was associated with a pronounced upregulation of the JAK/STAT signaling pathway. In vivo, TMEM2 overexpression reduced inflammatory cell infiltration, adipogenesis, and fibrosis in orbital tissues. These findings highlight the varied regulatory role of TMEM2 in GO pathogenesis. Our study reveals that TMEM2 plays a crucial role in mitigating inflammation, suppressing adipogenesis, and reducing fibrosis in GO. TMEM2 has potential as a therapeutic target and biomarker for treating or alleviating GO. These findings advance our understanding of GO pathophysiology and provide opportunities for targeted interventions to modulate TMEM2 for therapeutic purposes.
Collapse
Affiliation(s)
- Hong Li
- Department of Endocrinology, LongHua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Jie Min
- Department of Endocrinology, LongHua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yucheng Yang
- Department of Endocrinology, LongHua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wendong Suo
- Department of Endocrinology, LongHua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei Wang
- Department of Endocrinology, LongHua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiahe Tian
- Department of Endocrinology, LongHua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yujie Qin
- Department of Endocrinology, LongHua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
4
|
Štepánková K, Chudíčková M, Šimková Z, Martinez-Varea N, Kubinová Š, Urdzíková LM, Jendelová P, Kwok JCF. Low oral dose of 4-methylumbelliferone reduces glial scar but is insufficient to induce functional recovery after spinal cord injury. Sci Rep 2023; 13:19183. [PMID: 37932336 PMCID: PMC10628150 DOI: 10.1038/s41598-023-46539-5] [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: 07/16/2023] [Accepted: 11/02/2023] [Indexed: 11/08/2023] Open
Abstract
Spinal cord injury (SCI) induces the upregulation of chondroitin sulfate proteoglycans (CSPGs) at the glial scar and inhibits neuroregeneration. Under normal physiological condition, CSPGs interact with hyaluronan (HA) and other extracellular matrix on the neuronal surface forming a macromolecular structure called perineuronal nets (PNNs) which regulate neuroplasticity. 4-methylumbelliferone (4-MU) is a known inhibitor for HA synthesis but has not been tested in SCI. We first tested the effect of 4-MU in HA reduction in uninjured rats. After 8 weeks of 4-MU administration at a dose of 1.2 g/kg/day, we have not only observed a reduction of HA in the uninjured spinal cords but also a down-regulation of CS glycosaminoglycans (CS-GAGs). In order to assess the effect of 4-MU in chronic SCI, six weeks after Th8 spinal contusion injury, rats were fed with 4-MU or placebo for 8 weeks in combination with daily treadmill rehabilitation for 16 weeks to promote neuroplasticity. 4-MU treatment reduced the HA synthesis by astrocytes around the lesion site and increased sprouting of 5-hydroxytryptamine fibres into ventral horns. However, the current dose was not sufficient to suppress CS-GAG up-regulation induced by SCI. Further adjustment on the dosage will be required to benefit functional recovery after SCI.
Collapse
Affiliation(s)
- Kateřina Štepánková
- Institute of Experimental Medicine, Czech Academy of Sciences, Vídeňská, 1083, Prague, Czech Republic.
- Department of Neuroscience, Charles University, Second Faculty of Medicine, 15006, Prague, Czech Republic.
| | - Milada Chudíčková
- Institute of Experimental Medicine, Czech Academy of Sciences, Vídeňská, 1083, Prague, Czech Republic
| | - Zuzana Šimková
- Institute of Experimental Medicine, Czech Academy of Sciences, Vídeňská, 1083, Prague, Czech Republic
| | - Noelia Martinez-Varea
- Institute of Experimental Medicine, Czech Academy of Sciences, Vídeňská, 1083, Prague, Czech Republic
- Department of Neuroscience, Charles University, Second Faculty of Medicine, 15006, Prague, Czech Republic
| | - Šárka Kubinová
- Institute of Experimental Medicine, Czech Academy of Sciences, Vídeňská, 1083, Prague, Czech Republic
- Institute of Physics, Czech Academy of Sciences, 182 21, Prague, Czech Republic
| | - Lucia Machová Urdzíková
- Institute of Experimental Medicine, Czech Academy of Sciences, Vídeňská, 1083, Prague, Czech Republic.
- Department of Neuroscience, Charles University, Second Faculty of Medicine, 15006, Prague, Czech Republic.
| | - Pavla Jendelová
- Institute of Experimental Medicine, Czech Academy of Sciences, Vídeňská, 1083, Prague, Czech Republic.
- Department of Neuroscience, Charles University, Second Faculty of Medicine, 15006, Prague, Czech Republic.
| | - Jessica C F Kwok
- Institute of Experimental Medicine, Czech Academy of Sciences, Vídeňská, 1083, Prague, Czech Republic.
- Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK.
| |
Collapse
|
5
|
Kocurkova A, Kerberova M, Nesporova K, Lehka K, Sandanusova M, Simek M, Velebny V, Kubala L, Ambrozova G. Endogenously produced hyaluronan contributes to the regulation of peritoneal adhesion development. Biofactors 2023; 49:940-955. [PMID: 37154260 DOI: 10.1002/biof.1957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 04/05/2023] [Indexed: 05/10/2023]
Abstract
Peritoneal adhesions are postsurgical fibrotic complications connected to peritoneal inflammation. The exact mechanism of development is unknown; however, an important role is attributed to activated mesothelial cells (MCs) overproducing macromolecules of extracellular matrix (ECM), including hyaluronic acid (HA). It was suggested that endogenously-produced HA contributes to the regulation of different fibrosis-related pathologies. However, little is known about the role of altered HA production in peritoneal fibrosis. We focused on the consequences of the increased turnover of HA in the murine model of peritoneal adhesions. Changes of HA metabolism were observed in early phases of peritoneal adhesion development in vivo. To study the mechanism, human MCs MeT-5A and murine MCs isolated from the peritoneum of healthy mice were pro-fibrotically activated by transforming growth factor β (TGFβ), and the production of HA was attenuated by two modulators of carbohydrate metabolism, 4-methylumbelliferone (4-MU) and 2-deoxyglucose (2-DG). The attenuation of HA production was mediated by upregulation of HAS2 and downregulation of HYAL2 and connected to the lower expression of pro-fibrotic markers, including fibronectin and α-smooth muscle actin (αSMA). Moreover, the inclination of MCs to form fibrotic clusters was also downregulated, particularly in 2-DG-treated cells. The effects of 2-DG, but not 4-MU, were connected to changes in cellular metabolism. Importantly, the inhibition of AKT phosphorylation was observed after the use of both HA production inhibitors. In summary, we identified endogenous HA as an important regulator of peritoneal fibrosis, not just a passive player during this pathological process.
Collapse
Affiliation(s)
- Anna Kocurkova
- Department of Experimental Biology, Faculty of Sciences, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
- Department of Biophysics of Immune System, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | - Michaela Kerberova
- Department of Biophysics of Immune System, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | | | | | - Miriam Sandanusova
- Department of Experimental Biology, Faculty of Sciences, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
- Department of Biophysics of Immune System, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | - Matej Simek
- Contipro a.s., Dolni Dobrouc, Czech Republic
| | | | - Lukas Kubala
- Department of Experimental Biology, Faculty of Sciences, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
- Department of Biophysics of Immune System, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | - Gabriela Ambrozova
- Department of Biophysics of Immune System, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| |
Collapse
|
6
|
Hundhausen C, Schneckmann R, Ostendorf Y, Rimpler J, von Glinski A, Kohlmorgen C, Pasch N, Rolauer L, von Ameln F, Eckermann O, Altschmied J, Ale-Agha N, Haendeler J, Flögel U, Fischer JW, Grandoch M. Endothelial hyaluronan synthase 3 aggravates acute colitis in an experimental model of inflammatory bowel disease. Matrix Biol 2021; 102:20-36. [PMID: 34464693 DOI: 10.1016/j.matbio.2021.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/27/2021] [Accepted: 08/23/2021] [Indexed: 02/08/2023]
Abstract
The association between hyaluronan (HA) accumulation and increased inflammation in the colon suggests that HA is a potential therapeutic target in inflammatory bowel disease (IBD). However, whether patients with IBD would benefit from interference with HA synthesis is unknown. Here, we used pharmacological and genetic approaches to investigate the impact of systemic and partial blockade of HA synthesis in the Dextran Sodium Sulfate (DSS)-induced colitis model. To systemically inhibit HA production, we used 4-Methylumbelliferone (4-MU), whereas genetic approaches included the generation of mice with global or inducible cell-type specific deficiency in the Hyaluronan synthase 3 (Has3). We found that 4-MU treatment did not ameliorate but exacerbated disease severity characterized by increased body weight loss and enhanced colon tissue destruction compared to control mice without colitis. In contrast, global Has3 deficiency had a profound protective effect as reflected by a low colitis score and reduced infiltration of immune cells into the colon. To get further mechanistic insight into the proinflammatory role of HAS3, we deleted Has3 in a cell-type specific manner. Interestingly, while lack of Has3 expression in intestinal epithelial and smooth muscle cells had no effect or was rather proinflammatory, mice with Has3 deficiency in the endothelium were strongly protected against acute colitis. We conclude that endothelium-derived HAS3 plays a critical role in driving experimental colitis, warranting future studies on cell type-specific therapeutic interference with HA production in human IBD.
Collapse
Affiliation(s)
- Christian Hundhausen
- Institute for Pharmacology and Clinical Pharmacology, University Hospital, Heinrich-Heine-University Düsseldorf, Germany
| | - Rebekka Schneckmann
- Institute for Pharmacology and Clinical Pharmacology, University Hospital, Heinrich-Heine-University Düsseldorf, Germany
| | - Yanina Ostendorf
- Institute for Pharmacology and Clinical Pharmacology, University Hospital, Heinrich-Heine-University Düsseldorf, Germany
| | - Jacqueline Rimpler
- Institute for Pharmacology and Clinical Pharmacology, University Hospital, Heinrich-Heine-University Düsseldorf, Germany
| | - Anette von Glinski
- Institute for Pharmacology and Clinical Pharmacology, University Hospital, Heinrich-Heine-University Düsseldorf, Germany
| | - Christina Kohlmorgen
- Institute for Pharmacology and Clinical Pharmacology, University Hospital, Heinrich-Heine-University Düsseldorf, Germany
| | - Nina Pasch
- Institute for Pharmacology and Clinical Pharmacology, University Hospital, Heinrich-Heine-University Düsseldorf, Germany
| | - Luca Rolauer
- Institute for Pharmacology and Clinical Pharmacology, University Hospital, Heinrich-Heine-University Düsseldorf, Germany
| | - Florian von Ameln
- Environmentally-induced Cardiovascular Degeneration, Clinical Chemistry and Laboratory Diagnostics, Medical Faculty, University Clinics and Heinrich-Heine-University Düsseldorf and IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Olaf Eckermann
- Environmentally-induced Cardiovascular Degeneration, Clinical Chemistry and Laboratory Diagnostics, Medical Faculty, University Clinics and Heinrich-Heine-University Düsseldorf and IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Joachim Altschmied
- Environmentally-induced Cardiovascular Degeneration, Clinical Chemistry and Laboratory Diagnostics, Medical Faculty, University Clinics and Heinrich-Heine-University Düsseldorf and IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Niloofar Ale-Agha
- Environmentally-induced Cardiovascular Degeneration, Clinical Chemistry and Laboratory Diagnostics, Medical Faculty, University Clinics and Heinrich-Heine-University Düsseldorf, Germany
| | - Judith Haendeler
- Environmentally-induced Cardiovascular Degeneration, Clinical Chemistry and Laboratory Diagnostics, Medical Faculty, University Clinics and Heinrich-Heine-University Düsseldorf, Germany
| | - Ulrich Flögel
- Institute for Molecular Cardiology, University Hospital, Heinrich-Heine-University Düsseldorf, Germany
| | - Jens W Fischer
- Institute for Pharmacology and Clinical Pharmacology, University Hospital, Heinrich-Heine-University Düsseldorf, Germany
| | - Maria Grandoch
- Institute for Pharmacology and Clinical Pharmacology, University Hospital, Heinrich-Heine-University Düsseldorf, Germany.
| |
Collapse
|
7
|
Glycocalyx disruption enhances motility, proliferation and collagen synthesis in diabetic fibroblasts. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:118955. [PMID: 33421533 DOI: 10.1016/j.bbamcr.2021.118955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 12/24/2020] [Accepted: 01/04/2021] [Indexed: 11/22/2022]
Abstract
Impaired wound healing represents one of the most debilitating side effects of Diabetes mellitus. Though the role of fibroblasts in wound healing is well-known, the extent to which their function is altered in the context of diabetes remains incompletely understood. Here, we address this question by comparing the phenotypes of healthy dermal fibroblasts (HDFs) and diabetic dermal fibroblasts (DDFs). We show that DDFs are more elongated but less motile and less contractile than HDFs. Reduced motility of DDFs is attributed to formation of larger focal adhesions stabilized by a bulky glycocalyx, associated with increased expression of the cell surface glycoprotein mucin 16 (MUC 16). Disruption of the glycocalyx not only restored DDF motility to levels comparable to that of HDFs, but also led to increased proliferation and collagen synthesis. Collectively, our results illustrate the influence of glycocalyx disruption on mechanics of diabetic fibroblasts relevant to cell motility.
Collapse
|
8
|
Chistyakov DV, Nikolskaya AI, Goriainov SV, Astakhova AA, Sergeeva MG. Inhibitor of Hyaluronic Acid Synthesis 4-Methylumbelliferone as an Anti-Inflammatory Modulator of LPS-Mediated Astrocyte Responses. Int J Mol Sci 2020; 21:E8203. [PMID: 33147798 PMCID: PMC7662953 DOI: 10.3390/ijms21218203] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 10/25/2020] [Accepted: 10/30/2020] [Indexed: 02/07/2023] Open
Abstract
Astrocytes are glial cells that play an important role in neuroinflammation. Astrocytes respond to many pro-inflammatory stimuli, including lipopolysaccharide (LPS), an agonist of Toll-like receptor 4 (TLR4). Regulatory specificities of inflammatory signaling pathways are still largely unknown due to the ectodermal origin of astrocytes. Recently, we have shown that hyaluronic acid (HA) may form part of astrocyte inflammatory responses. Therefore, we tested 4-methylumbelliferone (4-MU), a specific inhibitor of HA synthesis, as a possible regulator of LPS-mediated responses. Rat primary astrocytes were treated with LPS with and without 4-MU and gene expression levels of inflammatory (interleukins 1β, (IL-1β), 6, (IL-6), tumor necrosis factor alpha TNFα,) and resolution interleukin 10 (IL-10) markers were evaluated via real-time PCR and western blot. The release of cytokines and HA was determined by ELISA. Oxylipin profiles were measured by ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) analysis. Our data show that 4-MU (i) has anti-inflammatory effects in the course of TLR4 activation, decreasing the cytokines level TNFα, IL-6 and IL-1β and increasing IL-10, (ii) downregulates prostaglandin synthesis but not via cyclooxygenases COX-1 and COX-2 pathways, (iii) modulates HA synthesis and decreases LPS-induced HA synthase mRNA expression (HAS-1, HAS-2) but does not have an influence on HAS-3, HYAL1 and HYAL2 mRNAs; (iv) the effects of 4-MU are predominantly revealed via JNK but not p38, ERK mitogen-activated protein kinases (MAPKs) or nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB) pathways. For the first time, it is shown that 4-MU possesses the useful potential to regulate an inflammatory astrocyte response.
Collapse
Affiliation(s)
- Dmitry V. Chistyakov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia; (A.A.A.); (M.G.S.)
- SREC PFUR, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia;
| | - Arina I. Nikolskaya
- Faculty of Bioengineering and Bioinformatics, Moscow Lomonosov State University, 119234 Moscow, Russia;
| | - Sergei V. Goriainov
- SREC PFUR, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia;
| | - Alina A. Astakhova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia; (A.A.A.); (M.G.S.)
| | - Marina G. Sergeeva
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia; (A.A.A.); (M.G.S.)
| |
Collapse
|