Human Xylosyltransferase I—An Important Linker between Acute Senescence and Fibrogenesis
Abstract
:1. Introduction
2. Materials and Methods
2.1. Cell Culture and Hydrogen Peroxide Treatment
2.2. Trypan Blue Dye Exclusion Assay
2.3. Bicinchoninic Acid Assay
2.4. Quantitative Senescence-Associated β-Gal Activity Assay
2.5. Quantitative Reverse Transcription Polymerase Chain Reaction (qRT-PCR)
2.6. XT-I Selective Enzyme Activity Assay by UPLC/ESI-MS/MS
2.7. Immunoblotting
2.8. Immunofluorescence
2.9. Statistical Analysis
3. Results
3.1. H2O2-Treatment of Human Proto-Myofibroblasts Induces Acute Senescence
3.2. Suppression of XT Expression in Acute Senescent Proto-Myofibroblasts
3.3. ECM Remodeling in Acute Senescent Proto-Myofibroblasts
3.4. The SASP of Acute Senescent Proto-Myofibroblasts
4. Discussion
5. Conclusions
Supplementary Materials
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Campisi, J.; Di d’Adda Fagagna, F. Cellular senescence: When bad things happen to good cells. Nat. Rev. Mol. Cell Biol. 2007, 8, 729–740. [Google Scholar] [CrossRef] [PubMed]
- López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The hallmarks of aging. Cell 2013, 153, 1194–1217. [Google Scholar] [CrossRef] [PubMed]
- van Deursen, J.M. The role of senescent cells in ageing. Nature 2014, 509, 439–446. [Google Scholar] [CrossRef]
- Alcorta, D.A.; Xiong, Y.; Phelps, D.; Hannon, G.; Beach, D.; Barrett, J.C. Involvement of the cyclin-dependent kinase inhibitor p16 (INK4a) in replicative senescence of normal human fibroblasts. Proc. Natl. Acad. Sci. USA 1996, 93, 13742–13747. [Google Scholar] [CrossRef]
- Coppé, J.-P.; Desprez, P.-Y.; Krtolica, A.; Campisi, J. The senescence-associated secretory phenotype: The dark side of tumor suppression. Annu. Rev. Pathol. Mech. Dis. 2010, 5, 99–118. [Google Scholar] [CrossRef]
- Lee, B.Y.; Han, J.A.; Im, J.S.; Morrone, A.; Johung, K.; Goodwin, E.C.; Kleijer, W.J.; DiMaio, D.; Hwang, E.S. Senescence-associated beta-galactosidase is lysosomal beta-galactosidase. Aging Cell 2006, 5, 187–195. [Google Scholar] [CrossRef]
- Dimri, G.P.; Testori, A.; Acosta, M.; Campisi, J. Replicative senescence, aging and growth-regulatory transcription factors. Biol. Signals 1996, 5, 154–162. [Google Scholar] [CrossRef]
- Collado, M.; Blasco, M.A.; Serrano, M. Cellular senescence in cancer and aging. Cell 2007, 130, 223–233. [Google Scholar] [CrossRef] [PubMed]
- Muñoz-Espín, D.; Cañamero, M.; Maraver, A.; Gómez-López, G.; Contreras, J.; Murillo-Cuesta, S.; Rodríguez-Baeza, A.; Varela-Nieto, I.; Ruberte, J.; Collado, M.; et al. Programmed cell senescence during mammalian embryonic development. Cell 2013, 155, 1104–1118. [Google Scholar] [CrossRef]
- Storer, M.; Mas, A.; Robert-Moreno, A.; Pecoraro, M.; Ortells, M.C.; Di Giacomo, V.; Yosef, R.; Pilpel, N.; Krizhanovsky, V.; Sharpe, J.; et al. Senescence is a developmental mechanism that contributes to embryonic growth and patterning. Cell 2013, 155, 1119–1130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jun, J.-I.; Lau, L.F. The matricellular protein CCN1 induces fibroblast senescence and restricts fibrosis in cutaneous wound healing. Nat. Cell Biol. 2010, 12, 676–685. [Google Scholar] [CrossRef] [PubMed]
- Krizhanovsky, V.; Yon, M.; Dickins, R.A.; Hearn, S.; Simon, J.; Miething, C.; Yee, H.; Zender, L.; Lowe, S.W. Senescence of activated stellate cells limits liver fibrosis. Cell 2008, 134, 657–667. [Google Scholar] [CrossRef]
- Childs, B.G.; Durik, M.; Baker, D.J.; van Deursen, J.M. Cellular senescence in aging and age-related disease: From mechanisms to therapy. Nat. Med. 2015, 21, 1424–1435. [Google Scholar] [CrossRef]
- Schafer, M.J.; White, T.A.; Iijima, K.; Haak, A.J.; Ligresti, G.; Atkinson, E.J.; Oberg, A.L.; Birch, J.; Salmonowicz, H.; Zhu, Y.; et al. Cellular senescence mediates fibrotic pulmonary disease. Nat. Commun. 2017, 8, 14532. [Google Scholar] [CrossRef]
- Rosenbloom, J.; Macarak, E.; Piera-Velazquez, S.; Jimenez, S.A. Human fibrotic diseases: Current challenges in fibrosis research. In Fibrosis: Methods and Protocols; Rittie, L., Ed.; Humana Press: New York, NY, USA, 2017; pp. 1–23. ISBN 978-1-4939-7112-1. [Google Scholar]
- Wynn, T.A. Cellular and molecular mechanisms of fibrosis. J. Pathol. 2008, 214, 199–210. [Google Scholar] [CrossRef]
- Desmoulière, A.; Darby, I.A.; Gabbiani, G. Normal and pathologic soft tissue remodeling: Role of the myofibroblast, with special emphasis on liver and kidney fibrosis. Lab. Investig. 2003, 83, 1689–1707. [Google Scholar] [CrossRef]
- Hinz, B. The role of myofibroblasts in wound healing. Curr. Res. Transl. Med. 2016, 64, 171–177. [Google Scholar] [CrossRef]
- Biernacka, A.; Dobaczewski, M.; Frangogiannis, N.G. TGF-β signaling in fibrosis. Growth Factors 2011, 29, 196–202. [Google Scholar] [CrossRef] [PubMed]
- Gressner, A.M.; Krull, N.; Bachem, M.G. Regulation of proteoglycan expression in fibrotic liver and cultured fat-storing cells. Pathol. Res. Pract. 1994, 190, 864–882. [Google Scholar] [CrossRef] [PubMed]
- Kuroda, K.; Shinkai, H. Decorin and glycosaminoglycan synthesis in skin fibroblasts from patients with systemic sclerosis. Arch. Dermatol. Res. 1997, 289, 481–485. [Google Scholar] [CrossRef] [PubMed]
- Bensadoun, E.S.; Burke, A.K.; Hogg, J.C.; Roberts, C.R. Proteoglycan deposition in pulmonary fibrosis. Am. J. Respir. Crit. Care Med. 1996, 154, 1819–1828. [Google Scholar] [CrossRef] [PubMed]
- Hardingham, T.E.; Fosang, A.J. Proteoglycans: Many forms and many functions. FASEB J. 1992, 6, 861–870. [Google Scholar] [CrossRef]
- Gandhi, N.S.; Mancera, R.L. The structure of glycosaminoglycans and their interactions with proteins. Chem. Biol. Drug Des. 2008, 72, 455–482. [Google Scholar] [CrossRef]
- Couchman, J.R.; Pataki, C.A. An introduction to proteoglycans and their localization. J. Histochem. Cytochem. 2012, 60, 885–897. [Google Scholar] [CrossRef]
- Götting, C.; Kuhn, J.; Zahn, R.; Brinkmann, T.; Kleesiek, K. Molecular cloning and expression of human UDP-d-Xylose:proteoglycan core protein beta-d-xylosyltransferase and its first isoform XT-II. J. Mol. Biol. 2000, 304, 517–528. [Google Scholar] [CrossRef] [PubMed]
- Kearns, A.E.; Campbell, S.C.; Westley, J.; Schwartz, N.B. Initiation of chondroitin sulfate biosynthesis: A kinetic analysis of UDP-D-xylose: Core protein beta-D-xylosyltransferase. Biochemistry 1991, 30, 7477–7483. [Google Scholar] [CrossRef] [PubMed]
- Schön, S.; Prante, C.; Bahr, C.; Kuhn, J.; Kleesiek, K.; Götting, C. Cloning and recombinant expression of active full-length xylosyltransferase I (XT-I) and characterization of subcellular localization of XT-I and XT-II. J. Biol. Chem. 2006, 281, 14224–14231. [Google Scholar] [CrossRef] [PubMed]
- Götting, C.; Sollberg, S.; Kuhn, J.; Weilke, C.; Huerkamp, C.; Brinkmann, T.; Krieg, T.; Kleesiek, K. Serum xylosyltransferase: A new biochemical marker of the sclerotic process in systemic sclerosis. J. Investig. Dermatol. 1999, 112, 919–924. [Google Scholar] [CrossRef]
- Pönighaus, C.; Ambrosius, M.; Casanova, J.C.; Prante, C.; Kuhn, J.; Esko, J.D.; Kleesiek, K.; Götting, C. Human xylosyltransferase II is involved in the biosynthesis of the uniform tetrasaccharide linkage region in chondroitin sulfate and heparan sulfate proteoglycans. J. Biol. Chem. 2007, 282, 5201–5206. [Google Scholar] [CrossRef]
- Götting, C.; Kuhn, J.; Kleesiek, K. Human xylosyltransferases in health and disease. Cell. Mol. Life Sci. 2007, 64, 1498–1517. [Google Scholar] [CrossRef]
- Faust, I.; Roch, C.; Kuhn, J.; Prante, C.; Knabbe, C.; Hendig, D. Human xylosyltransferase-I—A new marker for myofibroblast differentiation in skin fibrosis. Biochem. Biophys. Res. Commun. 2013, 436, 449–454. [Google Scholar] [CrossRef]
- Prante, C.; Milting, H.; Kassner, A.; Farr, M.; Ambrosius, M.; Schön, S.; Seidler, D.G.; Banayosy, A.E.; Körfer, R.; Kuhn, J.; et al. Transforming growth factor beta1-regulated xylosyltransferase I activity in human cardiac fibroblasts and its impact for myocardial remodeling. J. Biol. Chem. 2007, 282, 26441–26449. [Google Scholar] [CrossRef]
- Ly, T.-D.; Kleine, A.; Plümers, R.; Fischer, B.; Schmidt, V.; Hendig, D.; Distler, J.H.W.; Kuhn, J.; Knabbe, C.; Faust, I. Cytokine-mediated induction of human xylosyltransferase-I in systemic sclerosis skin fibroblasts. Biochem. Biophys. Res. Commun. 2021, 549, 34–39. [Google Scholar] [CrossRef]
- Ly, T.-D.; Plümers, R.; Fischer, B.; Schmidt, V.; Hendig, D.; Kuhn, J.; Knabbe, C.; Faust, I. Activin A-mediated regulation of XT-I in human skin fibroblasts. Biomolecules 2020, 10, 609. [Google Scholar] [CrossRef]
- Venkatesan, N.; Barré, L.; Bourhim, M.; Magdalou, J.; Mainard, D.; Netter, P.; Fournel-Gigleux, S.; Ouzzine, M. Xylosyltransferase-I regulates glycosaminoglycan synthesis during the pathogenic process of human osteoarthritis. PLoS ONE 2012, 7, e34020. [Google Scholar] [CrossRef] [PubMed]
- Ly, T.-D.; Riedel, L.; Fischer, B.; Schmidt, V.; Hendig, D.; Distler, J.; Kuhn, J.; Knabbe, C.; Faust, I. microRNA-145 mediates xylosyltransferase-I induction in myofibroblasts via suppression of transcription factor KLF4. Biochem. Biophys. Res. Commun. 2020, 523, 1001–1006. [Google Scholar] [CrossRef] [PubMed]
- Riedel, L.; Fischer, B.; Ly, T.-D.; Hendig, D.; Kuhn, J.; Knabbe, C.; Faust, I. microRNA-29b mediates fibrotic induction of human xylosyltransferase-I in human dermal fibroblasts via the Sp1 pathway. Sci. Rep. 2018, 8, 17779. [Google Scholar] [CrossRef]
- Fischer, B.; Ly, T.-D.; Schmidt, V.; Hendig, D.; Kuhn, J.; Knabbe, C.; Faust, I. Xylosyltransferase-deficient human HEK293 cells show a strongly reduced proliferation capacity and viability. Biochem. Biophys. Res. Commun. 2020, 521, 507–513. [Google Scholar] [CrossRef]
- Fischer, B.; Schmidt, V.; Ly, T.-D.; Kleine, A.; Knabbe, C.; Faust-Hinse, I. First characterization of human dermal fibroblasts showing a decreased xylosyltransferase-I expression induced by the CRISPR/Cas9 system. Int. J. Mol. Sci. 2022, 23, 5045. [Google Scholar] [CrossRef] [PubMed]
- Masur, S.K.; Dewal, H.S.; Dinh, T.T.; Erenburg, I.; Petridou, S. Myofibroblasts differentiate from fibroblasts when plated at low density. Proc. Natl. Acad. Sci. USA 1996, 93, 4219–4223. [Google Scholar] [CrossRef] [Green Version]
- Zdanov, S.; Remacle, J.; Toussaint, O. Establishment of H2O2-induced premature senescence in human fibroblasts concomitant with increased cellular production of H2O2. Ann. N. Y. Acad. Sci. 2006, 1067, 210–216. [Google Scholar] [CrossRef]
- Wang, Z.; Wei, D.; Xiao, H. Methods of cellular senescence induction using oxidative stress. Methods Mol. Biol. 2013, 1048, 135–144. [Google Scholar] [CrossRef] [PubMed]
- Strober, W. Trypan blue exclusion test of cell viability. Curr. Protoc. Immunol. 2015, 111, A3. [Google Scholar] [CrossRef] [PubMed]
- Smith, P.K.; Krohn, R.I.; Hermanson, G.T.; Mallia, A.K.; Gartner, F.H.; Provenzano, M.D.; Fujimoto, E.K.; Goeke, N.M.; Olson, B.J.; Klenk, D.C. Measurement of protein using bicinchoninic acid. Anal. Biochem. 1985, 150, 76–85. [Google Scholar] [CrossRef]
- Ly, T.-D.; Kleine, A.; Fischer, B.; Schmidt, V.; Hendig, D.; Kuhn, J.; Knabbe, C.; Faust, I. Identification of putative non-substrate-based XT-I inhibitors by natural product library screening. Biomolecules 2020, 10, 1467. [Google Scholar] [CrossRef]
- Gary, R.K.; Kindell, S.M. Quantitative assay of senescence-associated beta-galactosidase activity in mammalian cell extracts. Anal. Biochem. 2005, 343, 329–334. [Google Scholar] [CrossRef]
- Fischer, B.; Kuhn, J.; Ly, T.-D.; Schmidt, V.; Kleine, A.; Hendig, D.; Knabbe, C.; Faust, I. Development of a xylosyltransferase-I-selective UPLC MS/MS activity assay using a specific acceptor peptide. Biochimie 2021, 184, 88–94. [Google Scholar] [CrossRef]
- Jun, J.-I.; Lau, L.F. Cellular senescence controls fibrosis in wound healing. Aging 2010, 2, 627–631. [Google Scholar] [CrossRef]
- Gonzalez, A.C.d.O.; Costa, T.F.; Andrade, Z.d.A.; Medrado, A.R.A.P. Wound healing—A literature review. An. Bras. Dermatol. 2016, 91, 614–620. [Google Scholar] [CrossRef]
- van de Water, L.; Varney, S.; Tomasek, J.J. Mechanoregulation of the myofibroblast in wound contraction, scarring, and fibrosis: Opportunities for new therapeutic intervention. Adv. Wound Care 2013, 2, 122–141. [Google Scholar] [CrossRef] [Green Version]
- Bladier, C.; Wolvetang, E.J.; Hutchinson, P.; de Haan, J.B.; Kola, I. Response of a primary human fibroblast cell line to H2O2: Senescence-like growth arrest or apoptosis? Cell Growth Differ. 1997, 8, 589–598. [Google Scholar] [PubMed]
- Kurz, D.J.; Decary, S.; Hong, Y.; Erusalimsky, J.D. Senescence-associated (beta)-galactosidase reflects an increase in lysosomal mass during replicative ageing of human endothelial cells. J. Cell Sci. 2000, 113 Pt 20, 3613–3622. [Google Scholar] [CrossRef]
- Zhan, H.; Suzuki, T.; Aizawa, K.; Miyagawa, K.; Nagai, R. Ataxia telangiectasia mutated (ATM)-mediated DNA damage response in oxidative stress-induced vascular endothelial cell senescence. J. Biol. Chem. 2010, 285, 29662–29670. [Google Scholar] [CrossRef]
- Nakayama, F.; Hagiwara, A.; Yamamoto, T.; Akashi, M. Hydrogen peroxide as a potential mediator of the transcriptional regulation of heparan sulphate biosynthesis in keratinocytes. Cell. Mol. Biol. Lett. 2008, 13, 475–492. [Google Scholar] [CrossRef]
- Jin, C.L.; Oh, J.-H.; Han, M.; Shin, M.K.; Yao, C.; Park, C.-H.; Jin, Z.H.; Chung, J.H. UV irradiation-induced production of monoglycosylated biglycan through downregulation of xylosyltransferase 1 in cultured human dermal fibroblasts. J. Dermatol. Sci. 2015, 79, 20–29. [Google Scholar] [CrossRef]
- Salminen, A.; Kauppinen, A.; Kaarniranta, K. Emerging role of NF-κB signaling in the induction of senescence-associated secretory phenotype (SASP). Cell. Signal. 2012, 24, 835–845. [Google Scholar] [CrossRef]
- Karin, M.; Shaulian, E. AP-1: Linking hydrogen peroxide and oxidative stress to the control of cell proliferation and death. IUBMB Life 2001, 52, 17–24. [Google Scholar] [CrossRef]
- Khair, M.; Bourhim, M.; Barré, L.; Li, D.; Netter, P.; Magdalou, J.; Fournel-Gigleux, S.; Ouzzine, M. Regulation of xylosyltransferase I gene expression by interleukin 1β in human primary chondrocyte cells: Mechanism and impact on proteoglycan synthesis. J. Biol. Chem. 2013, 288, 1774–1784. [Google Scholar] [CrossRef]
- Müller, B.; Prante, C.; Kleesiek, K.; Götting, C. Identification and characterization of the human xylosyltransferase I gene promoter region. J. Biol. Chem. 2009, 284, 30775–30782. [Google Scholar] [CrossRef]
- Hwang, H.S.; Lee, M.H.; Kim, H.A. Fibronectin fragment inhibits xylosyltransferase-1 expression by regulating Sp1/Sp3- dependent transcription in articular chondrocytes. Osteoarthr. Cartil. 2019, 27, 833–843. [Google Scholar] [CrossRef]
- Qin, Z.; Robichaud, P.; He, T.; Fisher, G.J.; Voorhees, J.J.; Quan, T. Oxidant exposure induces cysteine-rich protein 61 (CCN1) via c-Jun/AP-1 to reduce collagen expression in human dermal fibroblasts. PLoS ONE 2014, 9, e115402. [Google Scholar] [CrossRef]
- Qin, Z.; Fisher, G.J.; Quan, T. Cysteine-rich protein 61 (CCN1) domain-specific stimulation of matrix metalloproteinase-1 expression through αVβ3 integrin in human skin fibroblasts. J. Biol. Chem. 2013, 288, 12386–12394. [Google Scholar] [CrossRef]
- Yamaguchi, Y.; Mann, D.M.; Ruoslahti, E. Negative regulation of transforming growth factor-beta by the proteoglycan decorin. Nature 1990, 346, 281–284. [Google Scholar] [CrossRef] [PubMed]
- McCawley, L.J.; Matrisian, L.M. Matrix metalloproteinases: They’re not just for matrix anymore! Curr. Opin. Cell Biol. 2001, 13, 534–540. [Google Scholar] [CrossRef] [PubMed]
- Mavrogonatou, E.; Papadopoulou, A.; Fotopoulou, A.; Tsimelis, S.; Bassiony, H.; Yiacoumettis, A.M.; Panagiotou, P.N.; Pratsinis, H.; Kletsas, D. Down-regulation of the proteoglycan decorin fills in the tumor-promoting phenotype of ionizing radiation-induced senescent human breast stromal fibroblasts. Cancers 2021, 13, 1987. [Google Scholar] [CrossRef]
- Chen, X.; Johns, D.C.; Geiman, D.E.; Marban, E.; Dang, D.T.; Hamlin, G.; Sun, R.; Yang, V.W. Krüppel-like factor 4 (gut-enriched Krüppel-like factor) inhibits cell proliferation by blocking G1/S progression of the cell cycle. J. Biol. Chem. 2001, 276, 30423–30428. [Google Scholar] [CrossRef] [PubMed]
- Dang, D.T.; Chen, X.; Feng, J.; Torbenson, M.; Dang, L.H.; Yang, V.W. Overexpression of Krüppel-like factor 4 in the human colon cancer cell line RKO leads to reduced tumorigenecity. Oncogene 2003, 22, 3424–3430. [Google Scholar] [CrossRef]
- Yoon, H.S.; Chen, X.; Yang, V.W. Kruppel-like factor 4 mediates p53-dependent G1/S cell cycle arrest in response to DNA damage. J. Biol. Chem. 2003, 278, 2101–2105. [Google Scholar] [CrossRef]
- Zhang, W.; Geiman, D.E.; Shields, J.M.; Dang, D.T.; Mahatan, C.S.; Kaestner, K.H.; Biggs, J.R.; Kraft, A.S.; Yang, V.W. The gut-enriched Kruppel-like factor (Kruppel-like factor 4) mediates the transactivating effect of p53 on the p21WAF1/Cip1 promoter. J. Biol. Chem. 2000, 275, 18391–18398. [Google Scholar] [CrossRef]
- Diegelmann, R.F.; Evans, M.C. Wound healing: An overview of acute, fibrotic and delayed healing. Front. Biosci. 2004, 9, 283–289. [Google Scholar] [CrossRef]
Gene | Primers | TA [°C] | Efficiency |
---|---|---|---|
hCDKN1A | 5′-GCTTCATGCCAGCTACTTCC-3′ 5′-CCCTTCAAAGTGCCATCTGT-3′ | 66 | 2.00 |
hCDKN2A | 5′-ACCAGAGGCAGTAACCATGC-3′ 5′-AAGTTTCCCGAGGTTTCTCAG-3′ | 66 | 2.00 |
hCOL1A1 | 5′-GATGTGCCACTCTGACT-3′ 5′-GGGTTCTTGCTGATG-3′ | 63 | 1.74 |
hCXCL8 | 5′-GAACTGAGAGTGATTGAGAGTGGA-3′ 5′-CTCTTCAAAAACTTCTCCACAACC-3′ | 63 | 1.88 |
hDCN | 5′-CCTTCCGCTGTCAATG-3′ 5′-GCAGGTCTAGCAGAGTTG-3′ | 63 | 1.76 |
hFN | 5′-CCCAGGGAAGATGTAGA-3′ 5′-CTCTTCCCGAACCTTATG-3′ | 63 | 2.00 |
hGAPDH | 5′-AGGTCGGAGTCAACGGAT-3′ 5′-TCCTGGAAGATGGTGATG-3′ | 59 | 1.83 |
hIL1B | 5′-ACAGATGAAGTGCTCCTTCCA-3′ 5′-GTCGGAGATTCGTAGCTGGAT-3′ | 63 | 1.94 |
hMMP1 | 5′-AGAAACACAAGAGCAAGATGTG-3′ 5′-TGGCGTGTAATTTTCAATCCTGT-3′ | 63 | 1.85 |
hSDHA | 5′-AACTCGCTCTTGGACCTG-3′ 5′-GAGTCGCAGTTCCGATGT-3′ | 66 | 2.00 |
hTGFB1 | 5′- GCGATACCTCAGCAACC-3′ 5′- ACGCAGCAGTTCTTCTCC-3′ | 59 | 1.95 |
hXYLT1 | 5′-TGTGACCTTCTCCACAGACG-3′ 5′-CCACGATGTGCTTGTACTGG-3′ | 63 | 2.00 |
hXYLT2 | 5′-ACACAGATGACCCGCTTGTGG-3′ 5′-TTGGTGACCCGCAGGTTGTTG-3′ | 63 | 1.95 |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Schmidt, V.; Ohmes, J.; Ly, T.-D.; Fischer, B.; Kleine, A.; Knabbe, C.; Faust-Hinse, I. Human Xylosyltransferase I—An Important Linker between Acute Senescence and Fibrogenesis. Biomedicines 2023, 11, 460. https://doi.org/10.3390/biomedicines11020460
Schmidt V, Ohmes J, Ly T-D, Fischer B, Kleine A, Knabbe C, Faust-Hinse I. Human Xylosyltransferase I—An Important Linker between Acute Senescence and Fibrogenesis. Biomedicines. 2023; 11(2):460. https://doi.org/10.3390/biomedicines11020460
Chicago/Turabian StyleSchmidt, Vanessa, Justus Ohmes, Thanh-Diep Ly, Bastian Fischer, Anika Kleine, Cornelius Knabbe, and Isabel Faust-Hinse. 2023. "Human Xylosyltransferase I—An Important Linker between Acute Senescence and Fibrogenesis" Biomedicines 11, no. 2: 460. https://doi.org/10.3390/biomedicines11020460
APA StyleSchmidt, V., Ohmes, J., Ly, T. -D., Fischer, B., Kleine, A., Knabbe, C., & Faust-Hinse, I. (2023). Human Xylosyltransferase I—An Important Linker between Acute Senescence and Fibrogenesis. Biomedicines, 11(2), 460. https://doi.org/10.3390/biomedicines11020460