Sclerosing Sialadenitis Is Associated With Salivary Gland Hypofunction and a Unique Gene Expression Profile in Sjögren's Syndrome

Hongen Yin, Thomas J F Pranzatelli, Benjamin N French, Nan Zhang, Blake M Warner, John A Chiorini, NIDCD/NIDCR Genomics and Computational Biology Core, Hongen Yin, Thomas J F Pranzatelli, Benjamin N French, Nan Zhang, Blake M Warner, John A Chiorini, NIDCD/NIDCR Genomics and Computational Biology Core

Abstract

Purpose: To develop a novel method to quantify the amount of fibrosis in the salivary gland and to investigate the relationship between fibrosis and specific symptoms associated with Sjögren's syndrome (SS) using this method.

Materials and methods: Paraffin-embedded labial salivary gland (LSG) slides from 20 female SS patients and their clinical and LSG pathology data were obtained from the Sjögren's International Collaborative Clinical Alliance. Relative interstitial fibrosis area (RIFA) in Masson's trichrome-stained LSG sections was quantified from digitally scanned slides and used for correlation analysis. Gene expression levels were assessed by microarray analysis. Core promoter accessibility for RIFA-correlated genes was determined using DNase I hypersensitive sites sequencing analysis.

Results: RIFA was significantly correlated with unstimulated whole saliva flow rate in SS patients. Sixteen genes were significantly and positively correlated with RIFA. In a separate analysis, a group of differentially expressed genes was identified by comparing severe and moderate fibrosis groups. This combined set of genes was distinct from differentially expressed genes identified in lung epithelium from idiopathic pulmonary fibrosis patients compared with controls. Single-cell RNA sequencing analysis of salivary glands suggested most of the RIFA-correlated genes are expressed by fibroblasts in the gland and are in a permissive chromatin state.

Conclusion: RIFA quantification is a novel method for assessing interstitial fibrosis and the impact of fibrosis on SS symptoms. Loss of gland function may be associated with salivary gland fibrosis, which is likely to be driven by a unique set of genes that are mainly expressed by fibroblasts.

Trial registration: ClinicalTrials.gov NCT02327884.

Keywords: Sjögren’s syndrome; salivary gland hypofunction; salivary gland interstitial fibrosis; sclerosing sialadenitis; transcriptomic gene expression profile.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2021 Yin, Pranzatelli, French, Zhang, Warner, Chiorini and NIDCD/NIDCR Genomics and Computational Biology Core.

Figures

Figure 1
Figure 1
Flow chart of study design. The purpose of this study was to investigate the application of quantification of digitally scanned slides of salivary gland interstitial fibrosis and its correlation with patients’ clinical data and transcriptome changes. We developed a novel quantification method based on measuring RIFA and compared this with the conventional interstitial fibrosis evaluation methodology by microscopic classification (4). Following validation, this method was applied to study the correlation of RIFA with Sjögren’s syndrome (SS) clinical features, such as unstimulated whole saliva flow rate. Genes associated with interstitial fibrosis development could be identified by correlating RIFA with gene expression data. These genes were then compared with the list of differentially expressed genes associated with severe fibrosis using microarray data from the SCS group (severe fibrosis) versus the NSCS group or FLS/SCS group (moderate fibrosis). This transcriptome profile that was associated with sclerosing sialadenitis in SS (RIFA-correlated genes and differentially expressed genes from SCS vs. NSCS or FLS/SCS groups) was compared with differentially expressed genes in IPF. Change in expression in this group of genes was further analyzed for common transcriptional factors binding motifs, cellular source of expression, and core promoter openness.
Figure 2
Figure 2
RIFA can be used to numerically quantify interstitial fibrosis in LSG from Sjögren’s syndrome patients. Masson’s trichrome staining was used to detect interstitial fibrosis in LSGs from Sjögren’s syndrome patients. Interstitial fibrosis areas and whole gland areas were measured using Aperio Scan Scope software. Non-fibrosis areas (outlined in yellow) that were included in the interstitial fibrosis areas (outlined in green) were subtracted to obtain accurate measurement. Area less than 50µm2 (orange arrows as example areas) were not excluded. RIFA was quantified as total interstitial fibrosis area (in mm2) per mm2 total glandular size on the section [(green circled areas – yellow circled areas)/total area].
Figure 3
Figure 3
RIFA assessment corresponds with categorical classification of salivary gland interstitial fibrosis. Left: Representative images to detect interstitial fibrosis in the LSG from four different microscopic groups comprising FLS, NSCS, FLS/SCS and SCS. Right: Comparison of mean (±SEM) RIFA in four different microscopic groups: FLS, NSCS, FLS/SCS and SCS (n = 5/group). Statistical significance was determined with unpaired Student’s t test.
Figure 4
Figure 4
RIFA correlates with unstimulated whole saliva flow rate in Sjögren’s syndrome patients. (A) Correlation of RIFA and USW flow rate in Sjögren’s syndrome (SS) patients was analyzed using linear regression analysis. A significant negative correlation was found between RIFA and UWS flow rate (n = 20). (B) Comparison of mean (± SEM) UWS flow rate in different microscopic groups based on mean (± SEM) of RIFA measurement (FLS, NSCS, FLS/SCS, and SCS).
Figure 5
Figure 5
Differentially expressed genes associated with severe interstitial fibrosis are conserved among RIFA-correlated genes. Venn diagram to identify common differentially expressed genes in SCS group (severe fibrosis) compared with NSCS or FLS/SCS group (moderate fibrosis). Fifteen common upregulated differentially expressed genes from SCS group compared with moderate interstitial fibrosis NSCS group or FLS/SCS group were identified. Of these 15 genes, 8 genes were also significantly correlated with RIFA (highlighted in yellow, refer to Table 1).
Figure 6
Figure 6
RIFA-correlated gene expression is primarily localized to fibroblasts in LSG from Sjögren’s syndrome and non-Sjögren’s patients. (A) Single-cell RNA sequencing analysis was used to quantify expression of RIFA-correlated genes in different cell types within LSGs from Sjögren’s syndrome patients and non-Sjögren’s subjects. Data shown are mean(± SEM) of “percent of total expression” of each gene within each cell type. Data revealed that RIFA-correlated genes were, on average, more highly expressed in fibroblasts than in any other cell type. (B) Assessment of “percent of total expression” of individual genes within each cell type revealed that eight genes were preferentially expressed by fibroblasts, which is reflected in increased average expression of list of genes in fibroblasts. Gene expression levels are represented on a color scale from high (red) to low (blue). The sum of values in a row equals 100% of the expression.
Figure 7
Figure 7
Core promoter openness is significantly higher for RIFA-correlated genes in lung fibroblasts than in whole lung tissue. DNase-seq reads were used to estimate chromatin accessibility of promoters of genes associated with fibrosis, all promoters, and the whole genome (as background) in both lung tissue and lung fibroblasts. Data shown are promoter coverage (mean ± SEM). Student’s t test was used to compare promoter coverage between pairs of groups.
Figure 8
Figure 8
Interconnection of RIFA-correlated genes with epithelial-mesenchymal transition and salivary gland interstitial fibrosis formation in Sjögren’s syndrome. Illustration of the hypothesis of the EMT and interstitial fibrosis formation in Sjögren’s syndrome regulated by the16 RIFA-correlated genes. RIFA-correlated genes are identified in red. Figure is created with BioRender.com.

References

    1. Fox RI. Sjogren’s Syndrome. Lancet (2005) 366(9482):321–31. 10.1016/S0140-6736(05)66990-5
    1. Porcheri C, Mitsiadis TA. Physiology, Pathology and Regeneration of Salivary Glands. Cells (2019) 8(9). 10.3390/cells8090976
    1. Giovelli RA, Santos MC, Serrano EV, Valim V. Clinical Characteristics and Biopsy Accuracy in Suspected Cases of Sjogren’s Syndrome Referred to Labial Salivary Gland Biopsy. BMC Musculoskelet Disord (2015) 16:30. 10.1186/s12891-015-0482-9
    1. Daniels TE, Cox D, Shiboski CH, Schiodt M, Wu A, Lanfranchi H, et al. . Associations Between Salivary Gland Histopathologic Diagnoses and Phenotypic Features of Sjogren’s Syndrome Among 1,726 Registry Participants. Arthritis Rheum (2011) 63(7):2021–30.
    1. Kamisawa T, Nakajima H, Egawa N, Funata N, Tsuruta K, Okamoto A, et al. . IgG4-Related Sclerosing Disease Incorporating Sclerosing Pancreatitis, Cholangitis, Sialadenitis and Retroperitoneal Fibrosis With Lymphadenopathy. Pancreatology (2006) 6(1-2):132–7. 10.1159/000090033
    1. Denton CP, Black CM, Abraham DJ. Mechanisms and Consequences of Fibrosis in Systemic Sclerosis. Nat Clin Pract Rheumatol (2006) 2(3):134–44. 10.1038/ncprheum0115
    1. Colby TV. Pulmonary Pathology in Patients With Systemic Autoimmune Diseases. Clin Chest Med (1998) 19(4):587–612, vii. 10.1016/S0272-5231(05)70105-2
    1. Ito I, Nagai S, Kitaichi M, Nicholson AG, Johkoh T, Noma S, et al. . Pulmonary Manifestations of Primary Sjogren’s Syndrome: A Clinical, Radiologic, and Pathologic Study. Am J Respir Crit Care Med (2005) 171(6):632–8. 10.1164/rccm.200403-417OC
    1. Young A, Koduri G, Batley M, Kulinskaya E, Gough A, Norton S, et al. . Mortality in Rheumatoid Arthritis. Increased in the Early Course of Disease, in Ischaemic Heart Disease and in Pulmonary Fibrosis. Rheumatol (Oxf) (2007) 46(2):350–7. 10.1093/rheumatology/kel253
    1. Bookman AA, Shen H, Cook RJ, Bailey D, McComb RJ, Rutka JA, et al. . Whole Stimulated Salivary Flow: Correlation With the Pathology of Inflammation and Damage in Minor Salivary Gland Biopsy Specimens From Patients With Primary Sjogren’s Syndrome But Not Patients With Sicca. Arthritis Rheum (2011) 63(7):2014–20. 10.1002/art.30295
    1. Leehan KM, Pezant NP, Rasmussen A, Grundahl K, Moore JS, Radfar L, et al. . Minor Salivary Gland Fibrosis in Sjogren’s Syndrome Is Elevated, Associated With Focus Score and Not Solely a Consequence of Aging. Clin Exp Rheumatol (2018) 36 Suppl 112(3):80–8.
    1. Voulgarelis M, Tzioufas AG. Pathogenetic Mechanisms in the Initiation and Perpetuation of Sjogren’s Syndrome. Nat Rev Rheumatol (2010) 6(9):529–37. 10.1038/nrrheum.2010.118
    1. Zeisberg M, Yang C, Martino M, Duncan MB, Rieder F, Tanjore H, et al. . Fibroblasts Derive From Hepatocytes in Liver Fibrosis Via Epithelial to Mesenchymal Transition. J Biol Chem (2007) 282(32):23337–47. 10.1074/jbc.M700194200
    1. Rastaldi MP, Ferrario F, Giardino L, Dell'Antonio G, Grillo C, Grillo P, et al. . Epithelial-Mesenchymal Transition of Tubular Epithelial Cells in Human Renal Biopsies. Kidney Int (2002) 62(1):137–46. 10.1046/j.1523-1755.2002.00430.x
    1. Das V, Bhattacharya S, Chikkaputtaiah C, Hazra S, Pal M. The Basics of Epithelial-Mesenchymal Transition (EMT): A Study From a Structure, Dynamics, and Functional Perspective. J Cell Physiol (2019) 234(2019):14535–55.
    1. Kalluri R, Weinberg RA. The Basics of Epithelial-Mesenchymal Transition. J Clin Invest (2009) 119(6):1420–8. 10.1172/JCI39104
    1. Pedroza M, To S, Smith J, Agarwal SK. Cadherin-11 Contributes to Liver Fibrosis Induced by Carbon Tetrachloride. PloS One (2019) 14(7):e0218971. 10.1371/journal.pone.0218971
    1. Hardy K, Wu F, Tu W, Zafar A, Boulding T, McCuaig R, et al. . Identification of Chromatin Accessibility Domains in Human Breast Cancer Stem Cells. Nucleus (2016) 7(1):50–67. 10.1080/19491034.2016.1150392
    1. Yin H, Kalra L, Lai Z, Guimaro MC, Aber L, Warner BM, et al. . Inhibition of Bone Morphogenetic Protein 6 Receptors Ameliorates Sjogren’s Syndrome in Mice. Sci Rep (2020) 10(1):2967. 10.1038/s41598-020-59443-z
    1. Shiboski CH, Shiboski SC, Seror R, Criswell LA, Labetoulle M, Lietman TM, et al. . 2016 American College of Rheumatology/European League Against Rheumatism Classification Criteria for Primary Sjogren’s Syndrome: A Consensus and Data-Driven Methodology Involving Three International Patient Cohorts. Arthritis Rheumatol (2017) 69(1):35–45.
    1. Yin H, Kosa P, Liu X, Swaim WD, Lai Z, Cabrera–Perez J, et al. . Matriptase Deletion Initiates a Sjogren’s Syndrome-Like Disease in Mice. PloS One (2014) 9(2):e82852. 10.1371/journal.pone.0082852
    1. Talbot LJ, Bhattacharya SD, Kuo PC. Epithelial-Mesenchymal Transition, the Tumor Microenvironment, and Metastatic Behavior of Epithelial Malignancies. Int J Biochem Mol Biol (2012) 3(2):117–36.
    1. Huang NI, Perez P, Kato T, Mikami YU, Okuda K, Gilmore RC, et al. . Integrated Single-Cell Atlases Reveal an Oral SARS-Cov-2 Infection and Transmission Axis. medRxiv (2020).
    1. Vukmirovic M, Herazo-Maya JD, Blackmon J, Skodric–Trifunovic V, Jovanovic D, Pavlovic S, et al. . Identification and Validation of Differentially Expressed Transcripts by RNA-Sequencing of Formalin-Fixed, Paraffin-Embedded (FFPE) Lung Tissue From Patients With Idiopathic Pulmonary Fibrosis. BMC Pulm Med (2017) 17(1):15. 10.1186/s12890-016-0356-4
    1. Plones T, Engel–Riedel W, Stoelben E, Limmroth C, Schildgen O, Schildgen V, et al. . Molecular Pathology and Personalized Medicine: The Dawn of a New Era in Companion Diagnostics-Practical Considerations About Companion Diagnostics for Non-Small-Cell-Lung-Cancer. J Pers Med (2016) 6(1). 10.3390/jpm6010003
    1. Potts SJ. Digital Pathology in Drug Discovery and Development: Multisite Integration. Drug Discov Today (2009) 14(19-20):935–41. 10.1016/j.drudis.2009.06.013
    1. Romualdi C, Giuliani A, Millino C, Celegato B, Benigni R, Lanfranchi G, et al. . Correlation Between Gene Expression and Clinical Data Through Linear and Nonlinear Principal Components Analyses: Muscular Dystrophies as Case Studies. OMICS (2009) 13(3):173–84. 10.1089/omi.2009.0003
    1. Teman CJ, Wilson AR, Perkins SL, Hickman K, Prchal JT, Salama ME, et al. . Quantification of Fibrosis and Osteosclerosis in Myeloproliferative Neoplasms: A Computer-Assisted Image Study. Leuk Res (2010) 34(7):871–6. 10.1016/j.leukres.2010.01.005
    1. Wei J, Bhattacharyya S, Tourtellotte WG, Varga J. Fibrosis in Systemic Sclerosis: Emerging Concepts and Implications for Targeted Therapy. Autoimmun Rev (2011) 10(5):267–75. 10.1016/j.autrev.2010.09.015
    1. Dillon LA, Okrah K, Hughitt VK, Suresh R, Li Y, Fernandes MC, et al. . Transcriptomic Profiling of Gene Expression and RNA Processing During Leishmania Major Differentiation. Nucleic Acids Res (2015) 43(14):6799–813. 10.1093/nar/gkv656
    1. Gerhard GS, Legendre C, Still CD, Chu X, Petrick A, DiStefano JK, et al. . Transcriptomic Profiling of Obesity-Related Nonalcoholic Steatohepatitis Reveals a Core Set of Fibrosis-Specific Genes. J Endocr Soc (2018) 2(7):710–26. 10.1210/js.2018-00122
    1. Wynn TA, Ramalingam TR. Mechanisms of Fibrosis: Therapeutic Translation for Fibrotic Disease. Nat Med (2012) 18(7):1028–40. 10.1038/nm.2807
    1. Gulino A, Di Marcotullio L, Screpanti I. The Multiple Functions of Numb. Exp Cell Res (2010) 316(6):900–6. 10.1016/j.yexcr.2009.11.017
    1. Milite C, Feoli A, Feoli M, Milite D, Rescigno A, Cianciulli AL, et al. . The Emerging Role of Lysine Methyltransferase SETD8 in Human Diseases. Clin Epigenet (2016) 8:102. 10.1186/s13148-016-0268-4
    1. Srivastava SP, Koya D, Kanasaki K. MicroRNAs in Kidney Fibrosis and Diabetic Nephropathy: Roles on EMT and Endmt. BioMed Res Int (2013) 2013:125469. 10.1155/2013/125469
    1. Milani S, Herbst H, Schuppan D, Stein H, Surrenti C. Transforming Growth Factors Beta 1 and Beta 2 Are Differentially Expressed in Fibrotic Liver Disease. Am J Pathol (1991) 139(6):1221–9.
    1. Yu X, Gu P, Huang Z, Fang X, Jiang Y, Luo Q, et al. . Reduced Expression of BMP3 Contributes to the Development of Pulmonary Fibrosis and Predicts the Unfavorable Prognosis in IIP Patients. Oncotarget (2017) 8(46):80531–44. 10.18632/oncotarget.20083
    1. Maslinska M, Paradowska-Gorycka A, Manczak M, Kostyra-Grabczak K, Kwiatkowska B. The Level of TGF-Beta in Sera of Patients With Primary Sjogren’s Syndrome. Reumatologia (2019) 57(6):309–14. 10.5114/reum.2019.91276
    1. Koski H, Konttinen YT, Gu XH, Hietanen J, Malmstrom M. Transforming Growth Factor Beta 2 in Labial Salivary Glands in Sjogren’s Syndrome. Ann Rheum Dis (1995) 54(9):744–7. 10.1136/ard.54.9.744
    1. Hall BE, Zheng C, Swaim WD, Cho A, Nagineni CN, Eckhaus MA, et al. . Conditional Overexpression of TGF-beta1 Disrupts Mouse Salivary Gland Development and Function. Lab Invest (2010) 90(4):543–55. 10.1038/labinvest.2010.5
    1. Borges S, Doppler H, Perez EA, Andorfer CA, Sun Z, Anastasiadis PZ, et al. . Pharmacologic Reversion of Epigenetic Silencing of the PRKD1 Promoter Blocks Breast Tumor Cell Invasion and Metastasis. Breast Cancer Res (2013) 15(2):R66. 10.1186/bcr3460
    1. Hassane S, Leonhard WN, van der Wal A, Hawinkels LJ, Lantinga-van Leeuwen IS, ten Dijke P, et al. . Elevated TGFbeta-Smad Signalling in Experimental Pkd1 Models and Human Patients With Polycystic Kidney Disease. J Pathol (2010) 222(1):21–31. 10.1002/path.2734
    1. Wang Y, Chen S, Chen J, Xie X, Gao S, Zhang C, et al. . Germline Genetic Patterns Underlying Familial Rheumatoid Arthritis, Systemic Lupus Erythematosus and Primary Sjogren’s Syndrome Highlight T Cell-Initiated Autoimmunity. Ann Rheum Dis (2020) 79(2):268–75. 10.1136/annrheumdis-2019-215533
    1. Liu F, Wang S. Molecular Cues for Development and Regeneration of Salivary Glands. Histol Histopathol (2014) 29(3):305–12.
    1. Kim MH, Jung SY, Song KH, Park JI, Ahn J, Kim EH, et al. . A New FGFR Inhibitor Disrupts the TGF-Beta1-Induced Fibrotic Process. J Cell Mol Med (2020) 24(1):830–40. 10.1111/jcmm.14793
    1. Li L, Zhang S, Wei L, Wang Z, Ma W, Liu F, et al. . FGF2 and FGFR2 in Patients With Idiopathic Pulmonary Fibrosis and Lung Cancer. Oncol Lett (2018) 16(2):2490–4. 10.3892/ol.2018.8903
    1. Strutz F, Zeisberg M, Renziehausen A, Raschke B, Becker V, van Kooten C, et al. . TGF-Beta 1 Induces Proliferation in Human Renal Fibroblasts Via Induction of Basic Fibroblast Growth Factor (FGF-2). Kidney Int (2001) 59(2):579–92. 10.1046/j.1523-1755.2001.059002579.x
    1. Cha B, Geng X, Mahamud MR, Fu AJ, Mukherjee A, Kim Y, et al. . Mechanotransduction Activates Canonical Wnt/beta-catenin Signaling to Promote Lymphatic Vascular Patterning and the Development of Lymphatic and Lymphovenous Valves. Genes Dev (2016) 30(12):1454–69. 10.1101/gad.282400.116
    1. DiRocco DP, Kobayashi A, Taketo MM, McMahon AP, Humphreys BD, et al. . Wnt4/beta-Catenin Signaling in Medullary Kidney Myofibroblasts. J Am Soc Nephrol (2013) 24(9):1399–412. 10.1681/ASN.2012050512
    1. Kaur H, Phillips-Mason PJ, Burden-Gulley SM, Kerstetter-Fogle AE, Basilion JP, Sloan AE, et al. . Cadherin-11, A Marker of the Mesenchymal Phenotype, Regulates Glioblastoma Cell Migration and Survival In Vivo. Mol Cancer Res (2012) 10(3):293–304. 10.1158/1541-7786.MCR-11-0457
    1. Fernandez IE, Eickelberg O. New Cellular and Molecular Mechanisms of Lung Injury and Fibrosis in Idiopathic Pulmonary Fibrosis. Lancet (2012) 380(9842):680–8. 10.1016/S0140-6736(12)61144-1
    1. O’Connor KS, Parnell G, Patrick E, Ahlenstiel G, Suppiah V, van der Poorten D, et al. . Hepatic Metallothionein Expression in Chronic Hepatitis C Virus Infection Is IFNL3 Genotype-Dependent. Genes Immun (2014) 15(2):88–94. 10.1038/gene.2013.66
    1. Phillips RJ, Burdick MD, Hong K, Lutz MA, Murray LA, Xue YY, et al. . Circulating Fibrocytes Traffic to the Lungs in Response to CXCL12 and Mediate Fibrosis. J Clin Invest (2004) 114(3):438–46. 10.1172/JCI200420997
    1. Taube JH, Herschkowitz JI, Komurov K, Zhou AY, Gupta S, Yang J, et al. . Core Epithelial-to-Mesenchymal Transition Interactome Gene-Expression Signature Is Associated With Claudin-Low and Metaplastic Breast Cancer Subtypes. Proc Natl Acad Sci USA (2010) 107(35):15449–54. 10.1073/pnas.1015095107
    1. Dai W, Zheng H, Cheung AK, Tang CS, Ko JM, Wong BW, et al. . Whole-Exome Sequencing Identifies MST1R as a Genetic Susceptibility Gene in Nasopharyngeal Carcinoma. Proc Natl Acad Sci USA (2016) 113(12):3317–22. 10.1073/pnas.1523436113
    1. Guan S, Zhou J. Frizzled-7 Mediates TGF-beta-Induced Pulmonary Fibrosis by Transmitting non-Canonical Wnt Signaling. Exp Cell Res (2017) 359(1):226–34. 10.1016/j.yexcr.2017.07.025
    1. Miliani de Marval PL, Gimenez-Conti IB, LaCava M, Martinez LA, Conti CJ, Rodriguez ML, et al. . Transgenic Expression of Cyclin-Dependent Kinase 4 Results in Epidermal Hyperplasia, Hypertrophy, and Severe Dermal Fibrosis. Am J Pathol (2001) 159(1):369–79. 10.1016/S0002-9440(10)61703-8
    1. Michael DG, Pranzatelli TJF, Warner BM, Yin H, Chiorini JA. Integrated Epigenetic Mapping of Human and Mouse Salivary Gene Regulation. J Dent Res (2019) 98(2):209–17. 10.1177/0022034518806518
    1. Chou RH, Lin SC, Wen HC, Wu CW, Chang WS. Epigenetic Activation of Human Kallikrein 13 Enhances Malignancy of Lung Adenocarcinoma by Promoting N-Cadherin Expression and Laminin Degradation. Biochem Biophys Res Commun (2011) 409(3):442–7. 10.1016/j.bbrc.2011.05.022
    1. Kelsey SH, Johnson S, Song S, Chakraborty P, Jolly MK, Toneff MJ, et al. . Gene Expression and Chromatin Accessibility During Progressive EMT and MET Linked to Dynamic CTCF Engagement. BioRxiv (2020). 10.1101/2020.05.11.089110

Source: PubMed

3
Subskrybuj