Article

A systems proteomics view of the endogenous human claudin family

LIU, Fei, et al.

Abstract

Claudins are the major transmembrane protein components of tight junctions in human endothelia and epithelia. Tissue-specific expression of claudin members suggests that this protein family is not only essential for sustaining the role of tight junction in cell permeability control but also vital in organizing cell contact signaling by protein-protein interactions. How this family of protein is collectively processed and regulated is key to understanding the role of junctional in preserving cell identity and tissue integrity. The focus of this review is to first provide a brief overview of the functional context, on the basis of the extensive body of claudin biology research that has been thoroughly reviewed, for endogenous human claudin members and then ascertain existing and future proteomics techniques that may be applicable to systematically characterizing the chemical forms and interacting protein partners of this protein family in human. The ability to elucidate claudin-based signaling networks may provide new insight into cell development and differentiation programs that are crucial to tissue stability [...]

Reference

LIU, Fei, et al. A systems proteomics view of the endogenous human claudin protein family. Journal of Proteome Research, 2016, vol. 15, no. 2, p. 339-359

DOI : 10.1021/acs.jproteome.5b00769 PMID : 26680015

Available at: http://archive-ouverte.unige.ch/unige:79118

Disclaimer: layout of this document may differ from the published version.

1 / 1 Subscriber access provided by UNIVERSITE DE GENEVE Review A systems proteomics view of the endogenous human claudin protein family Fei Liu, Michael Koval, Shoba Ranganathan, Susan Fanayan, William S. Hancock, Emma K. Lundberg, Ronald C Beavis, Lydie Lane, Paula Duek, Leon McQuade, Neil L. Kelleher, and Mark S. Baker J. Proteome Res., Just Accepted Manuscript • DOI: 10.1021/acs.jproteome.5b00769 • Publication Date (Web): 18 Dec 2015 Downloaded from http://pubs.acs.org on January 11, 2016

Just Accepted

“Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a free service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are accessible to all readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

Journal of Proteome Research is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties. Page 1 of 58 Journal of Proteome Research

1 2 Title: A systems proteomics view of the endogenous human claudin protein family 3 4 5 Fei Liu,*a Michael Koval, b Shoba Ranganathan, a Susan Fanayan,c William S. Hancock,c,d Emma K. 6 Lundberg, e Ronald C. Beavis, f Lydie Lane, g Paula Duek,g Leon McQuade,h Neil L. Kelleher,i Mark 7 S. Baker c 8 9

10 a 11 Department of Chemistry and Biomolecular Sciences, Macquarie University, Sydney, NSW 2109, 12 Australia b 13 Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine and 14 Department of Cell Biology, Emory University School of Medicine, 205 Whitehead Biomedical 15 Research Building, 615 Michael Street, Atlanta, GA 30322 16 cDepartment of Biomedical Sciences, Macquarie University, Sydney, NSW 2109, Australia 17 dBarnett Institute and Department of Chemistry and Chemical Biology, Northeastern University, 18 Boston, Massachusetts 02115, United States 19 e 20 SciLifeLab, School of Biotechnology, Royal Institute of Technology (KTH), SE-171 21 Solna, Stockholm, Sweden 21 f 22 University of Manitoba, Department of Biochemistry and Medical Genetics, 744 Bannatyne 23 Avenue, Winnipeg, Manitoba, R3E 0W3 Canada 24 gSIB-Swiss Institute of Bioinformatics, CMU - Rue Michel-Servet 1, 1211 Geneva, Switzerland 25 hAustralian Proteome Analysis Facility, Macquarie University, Sydney, NSW 2109, Australia 26 iDepartments of Chemistry and Molecular Biosciences, and the Proteomics Center of Excellence, 27 Northwestern University, 2145 N. Sheridan Road, Evanston, Illinois 60208, United States 28 29 30 31 32 *(F.L.) Tel: (61)298508312. Fax: (61)298508313. Email: [email protected]. 33 34 35 Key words: membrane protein complexes, cellcontact signaling, systems proteomics, membrane 36 proteomics, targeted proteomics, topdown proteomics, chemical proteomics 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 ACS Paragon Plus Environment Journal of Proteome Research Page 2 of 58

1 2 Abstract 3 4 Claudins are the major transmembrane protein components of tight junctions in human endothelia 5 6 and epithelia. Tissuespecific expression of claudin members suggests that this protein family is not 7 8 only essential for sustaining the role of tight junction in cell permeability control but also vital in 9 10 11 organizing cell contact signaling by proteinprotein interactions. How this family of protein is 12 13 collectively processed and regulated is key to understanding the role of junctional proteins in 14 15 preserving cell identity and tissue integrity. The focus of this review is to first provide a brief 16 17 overview of the functional context, on the basis of the extensive body of claudin biology research 18 19 20 that has been thoroughly reviewed, for endogenous human claudin members and then ascertain 21 22 existing and future proteomics techniques that may be applicable to systematically characterizing 23 24 the chemical forms and interacting protein partners of this protein family in human. The ability to 25 26 elucidate claudinbased signaling networks may provide new insight into cell development and 27 28 differentiation programs that are crucial to tissue stability and manipulation. 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 ACS Paragon Plus Environment Page 3 of 58 Journal of Proteome Research

1 2 Introduction 3 4 5 6 Claudins are a family of transmembrane proteins for barrier and pore formation in metazoans, 7 8 especially for vertebrates and tunicates.1 Chordate claudins are the essential architectural proteins of 9 10 29 11 tight junction strands in the apical junctional complex of epithelia and endothelia. These 12 13 junctional strands, containing claudin protein complexes, act mainly as paracellular seals for large 14 15 molecules and a semipermeable barrier to ions in tissues. 5,6,10 Distinct claudin members are 16 17 expressed in a tissue to confer tissue specific permeability and barrier characteristics. 7,11 Claudins 18 19 3,1215 20 also serve as protein scaffolds for assembling signaling complexes at cell junctions. 21 22 Understanding how the claudin family of proteins are expressed, organized and regulated in the cell 23 24 with temporal and spatial resolution is critical to unraveling mechanisms that control paracellular 25 26 barrier function, tissue integrity and stability. 27 28 29 30 31 The seminal discovery of claudins by Furuse and Tsukita in 1998 came 25 years after the initial 32 16,17 33 observation of cellcell contact ultrastructures. Since this seminal work the claudin protein 34 35 family has been the subject of intense investigations to understand their structure and function in 36 37 physiological and pathological contexts. The number of claudin varies between species. For 38 39 40 example, in the case of the puffer fish Takifugu rubripes , up to 56 claudin genes are found by 41 18 42 genome sequence analysis, whilst in Homo sapiens , the claudin family has at least 23 and 43 44 maybe more if all predicted claudin genes are found to be expressed as proteins.19 Because different 45 46 claudins are differentially expressed with tissue specificity and temporal regulation, claudins are 47 48 likely to confer several vital roles particularly in control of paracellular permeability but also in cell 49 50 51 differentiation, morphogenesis, and tissue maintenance. 52 53 54 55 Several reviews have appeared to account in detail advances in claudin physiology and biology of 56 57 many organisms.1,410,14,18,19,20,21 These reviews represent a vast body of research in claudin 58 59 60 ACS Paragon Plus Environment Journal of Proteome Research Page 4 of 58

1 2 physiology in humans and other organisms. The majority of insights into defining how claudins 3 4 function have been obtained using epithelial cell lines where the ramifications of claudin expression 5 6 are measured after manipulation by either cDNA overexpression or RNA silencing. Claudin 7 8 deficient 22 and overexpression 23,24 transgenic mouse models have also proven informative in 9 10 11 identifying functions for specific claudins in regulating epithelial barrier function and other aspects 12 13 of mammalian physiology relevant to human disease. These are valuable approaches. However it is 14 15 difficult to employ these models so that claudin expression is manipulated within the normal 16 17 physiologic range of mRNA and protein expression. The claudin expression profile of many 18 19 20 different tissues and cells, including normal and tumor cells, has been assessed at the mRNA and 21 22 protein level. While these approaches provide some insights into how the claudin proteome is 23 24 organized in different tissues, they have two major limitations. First, claudin mRNA does not 25 26 necessarily correlate with levels of protein expression,25 since processes such as tight junction 27 28 turnover are significantly regulated by proteinprotein interactions 26 and posttranslational 29 30 2730 31 modifications (PTMs) of claudins in both human and animal cell models. Second, direct 32 33 measurements of claudin protein have been largely restricted to antibodydependent approaches 34 35 (e.g. immunoblot) which have significant utility but are difficult to use to measure the stoichiometry 36 37 of claudin composition, which is a critical parameter needed to determine how differential claudin 38 39 40 expression influences epithelial barrier function and other aspects of cell physiology. 41 42 43 44 The approach of “systems proteomics”, encompassing specialty technologies such as membrane 45 46 proteomics, 31,32 targeted proteomics, 3335 bottomup/topdown proteomics, 36 structural 47 48 proteomics, 37,38 and chemical proteomics,39,40 has the potential to provide valuable adjuncts to other 49 50 51 established methods currently used to define roles for claudins in cell and tissue function. 52 53 However, the application of systems proteomics has lagged behind our understanding of functional 54 55 roles of claudins in regulating human cells and tissues. There are several reasons for this. Claudins 56 57 are highly hydrophobic proteins that are more difficult to isolate and analyze by mass spectrometry 58 59 60 ACS Paragon Plus Environment Page 5 of 58 Journal of Proteome Research

1 41 2 (MS) than hydrophilic proteins. Also claudinfunction is highly dependent on cell localization 3 4 that, in turn, is regulated by interactions with other proteins. Claudinprotein interactions range 5 6 from weak, unstable interactions (e.g. claudinclaudin interactions 14,42 ) to interactions that 7 8 effectively immobilize claudins onto the actin cytoskeleton. 43 These are classes of proteinprotein 9 10 11 interactions that are difficult to analyze and will require novel approaches to define. 12 13 14 15 In this review, human claudin genetics and biology is summarized first for a general outlook while 16 17 also guiding the reader to other sources for further details on claudin cell and molecular physiology. 18 19 20 Here the discussion focuses on the chemical characteristics of human claudins with a particular 21 22 emphasis placed on MScentered, systems proteomics approaches to studying human endogenous 23 24 claudins. Some of the investigations on other organisms such as mice, rats, and canine cell/tissue 25 26 models will be discussed only for occasional comparisons. Specialty proteomics technologies, such 27 28 as membrane proteomics, structural proteomics, targeted proteomics, bottomup/topdown 29 30 31 proteomics, and chemical proteomics, are assessed for consideration of their further development 32 33 and integration into investigating human endogenous claudins. Because endogenous claudins are 34 35 low in abundance with complex chemical modification and localization profiles, this will most 36 37 certainly push technology development to new limits. Emerging MS approaches for elucidating 38 39 40 membrane protein complexes in the relevant context of claudin biological function is also addressed 41 42 here. Human claudin PTMs, revealed thus far by MS and/or immunodetection, are discussed to 43 44 highlight how claudin function may be regulated. Existing approaches for characterizing 45 46 endogenous human claudins by MS in conjunction with chemical partition and enrichment 47 48 techniques are also reviewed to underscore the challenges and also opportunities in unraveling the 49 50 51 claudin protein networks holistically with improved temporal and spatial resolution. New chemical 52 53 delineation of the native claudin protein system may lead to novel hypotheses into claudin biology 54 55 and new cell assembly and tissue manipulation capabilities. The advanced technologies developed 56 57 58 59 60 ACS Paragon Plus Environment Journal of Proteome Research Page 6 of 58

1 2 for claudin systems proteomics could also assist further biological investigations into junctional 3 4 signaling networks involved in intercellular communication in metazoans. 4448 5 6 7 8 1. A general overview of the genetic profile and protein family, biological role, and tissue 9 10 11 expression of the human claudins 12 13 14 15 As indicated earlier, recent reviews have already provided insightful accounts of claudin genetics 16 17 and biology, along with their significant interactions with other important junction proteins. 18 19 49 50 20 Information on human claudins from various databases, such as UniProt , Ensembl and the

21 51 22 Human Protein Atlas (HPA) is compiled in Table 1 to provide a general outlook of human 23 24 claudins. Much of the recent claudin research is not yet captured in these databases, although they 25 26 offer an overview of human claudins to help guide navigating the detailed reviews and current 27 28 articles for indepth analysis. 29 30 31 32 33 Genetic analysis 34 35 Including transcript variants, 27 mammalian claudin genes have been reported, and at least 23 are 36 37 found in humans (Table 1, column 1).20,5254 The mammalian CLDN13 gene is absent in humans but 38 39 1 40 present in rodents, while CLDN24 , 25 , 26 , and 27 are putative claudin genes. The official gene 41 55 42 names for claudins, attributed by the HUGO committee, are shown in Table 1 43 44 (column 1 and 2) with other synonyms also in use (e.g. in GeneCards 56 , Table 1, column 3). The 45 46 human claudin genes are spread across 13 (1, 3, 4, 6, 7, 8, 11, 13, 16, 17, 21, 22, and 47 48 X) with generally few or even no introns found for these genes (Table 1, column 4). Chromosomes 49 50 51 3 and 7 have the highest frequency of claudins ( CLDN1, 11 , 16, and 18 on 3; CLDN3 , 52 53 4, 12 , and 15 on chromosome 7). Some claudins also exhibit high pairwise , 54 55 such as CLDN3 and 4, CLDN6 and 9, CLDN8 and 17 , and CLDN22 and 24 . Coordinated gene 56 57 expression is possible and has been observed for CLDN3 and CLDN4 .54 Mammalian claudins are 58 59 60 ACS Paragon Plus Environment Page 7 of 58 Journal of Proteome Research

1 2 classified into the “classic” (CLDN1-10, 14, 15, 17, and 19 ) and “nonclassic” (CLDN11-13, 16, 18, 3 4 20-24) groups on the basis of their phylogenetic relationship,57 and it is also possible to split the 5 6 claudins into eight groups based on their gene structures and phylogenetic distances.1,10 7 8 9 10 11 Table 1. An overview of the human claudin family members from databases. 12 13 Gene Protein IDs Synonyms Chromosome Tissue Tissue-specific e i 14 (UniProt/ (GeneCards) Localization Frequency with RNA FPKM b f 15 Ensembl ) HPA Evidence (max in non- 16 specific tissue) 17 CLDN1 O95832/ SEMP1, ILVASC 3q28 very common 18 163347 (skin: 224.1) 19 g CLDN2 P57739/ SP82 Xq22 common 20 165376 (kidney: 21 114.5) 22 23 CLDN3 O15551/ CPETR2 7q11 very common 24 165215 (colon: 94.8) 25 h 26 CLDN4 O14493/ CPER, CPETR1, 7q11 common 189143 WBSCR8 (colon: 27 160.9) 28 29 CLDN5 O00501/ AWAL, TMVCF 22q11 very common adipose 30 184113 tissue: 31 110.8; lung: 32 32.1 33 (spleen: 34 10.4) 35 36 CLDN6 P56747/ UNQ757, 16p13 very common 37 184697 PRO1488, Skullin (placenta: 38 1.3) 39 CLDN7 O9547 1/ CEPTRL2, 17p13 very common 40 181885 CPETRL2 (colon: 41 297.3) 42 43 CLDN8 P56748/ UNQ779, 21q22 very common 44 156284 PRO1573 (kidney: 45 63.6) 46 47 CLDN9 O95484/ 16p13 very common 48 213937 (pancreas: 49 2.0) 50 51 CLDN10 P78369/ CPETRL3, OSP L 13q32 less common 134873 (salivary 52 gland: 53 150.7) 54 CLDN11 O75508/ OSP, OTM 3q26 less common 55 013297 (testis: 56 167.0) 57 CLDN12 P56749/ 7q21 very common 58 157224 (liver: 31.9) 59 60 ACS Paragon Plus Environment Journal of Proteome Research Page 8 of 58

1 2 CLDN14 O95500/ UNQ777, 21q22 NA kidney: 5.3; 3 159261 PRO1571, liver: 8.4 4 DFNB29 (appendix: 0.5) 5 CLDN15 P56746/ 7q22 very common duodenum: 6 106404 184.0; 7 small 8 intestine:14 9 0.1 10 (spleen: 11 12.6) 12 13 CLDN16 Q9Y5I7/ PCLN1, 3q28 rare kidney: 14 113946 PCLN1, HOMG3 39.0 15 (thyroid 16 gland: 6.8)

17 CLDN17 P56750/ UNQ758 21q22 NA esophagus: 18 156282 PRO1489 6.4 19 (testis: 0.3) 20 21 CLDN18 P56856/ UNQ778 3q22 rare lung: 246.6; 22 066405 PRO1572, stomach: 23 SFTA5, SFTPJ 556.1 24 (heart 25 muscle: 26 7.3) 27 28 CLDN19 Q8N6F1/ HOMG5 1p34 NA kidney: 29 164007 21.9; placenta: 30 11.3 31 (adipose 32 tissue: 0.2) 33 34 CLDN20 P56880/ 6q25 NA 35 171217 (uterus: 36 0.3) 37 CLDN21 a see CLDN24 38 CLDN22 Q8N7P3/ 4q35 NA 39 177300 (adrenal 40 gland: 0.1) 41 CLDN23 Q96B33/ 2310014B08RikC 8p23 NA 42 253958 LDNL, (stomach: hCG1646163 17.6) 43 CLDN24 A6NM45/ also named 4q35 NA 44 185758 CLDN21 (kidney: 45 0.7) 46 CLDN25 C9JDP6/ 11q23 rare 47 228607 (adipose 48 tissue: 0.0) 49 CLDN26 B3SHH9 c TMEM114 d 16p13 NA 50 (testis: 0.4) c d 51 CLDN27 A6NFC5 TMEM235 17q25 NA 52 (testis: 0.1) a 53 Merged with CLDN24: see http://www.ncbi.nlm.nih.gov/gene/53843 .

54 b 55 Ensemble code prefix: ENSG00000. 56 c Not identified as claudin but as TMEM (transmembrane) proteins. 57 58 d Tracked from GeneCards synonyms. 59 60 ACS Paragon Plus Environment Page 9 of 58 Journal of Proteome Research

1 e 54 2 CLDN13 is absent in human. CLDN 6 and 9, as well as CLDN 8 and 17, may be considered as paralogs. 3 f HPA evidence was calculated based on the manual curation of Western blot, tissue profiling and subcellular location 4 5 using a limited number of claudin antibodies that have been submitted to HPA and validated by HPA. Many other 6 7 claudin antibodies are available but not included in the HPA list. Very common = high or medium levels in at least 20 8 9 tissues; Common = high or medium levels in at least 10 tissues; Less common = high or medium levels in more than 3 10 11 but less than 10 tissues; Rare = high or medium levels in only 1 to 3 tissues; NA = no antibodies. 12 g 13 In nucleus but not nucleoli, cell junctions by immunofluorescence analysis.

14 h 15 In plasma membrane by immunofluorescence analysis. 16 i FPKM value = number of fragments per kilobase gene model and million reads. The FPKM threshold value for 17 18 detection is > 1. 58 FPKM measurements reflect measured tissues represented in the database and are not necessarily 19 20 representative of a complete mRNA expression profile. 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 ACS Paragon Plus Environment Journal of Proteome Research Page 10 of 58

1 2 3 4 5 6 Table 2. Peptide evidence for claudin proteins by MS analysis. For each claudin, peptides with good MS evidence are listed (data compiled from 7 8 www.thegpm.org and www.nextprot.org). 9 10 11 Proteins # of Best Evidence Sequences with Good Evidence (EC = 4) a 12 a b 13 Observations Log(E) Code (amino acid residue numbers in parentheses) 14 CLDN1 323 111 4 VFDSLLNLSSTLQATR(66 81); CLEDDEVQK(107 115); CLEDDEVQKMR(107 117); 15 IVQEFYDPMTPVNAR(144158); FYDPMTPVNARYE(148160); KTTSYPTPRPYPK(189201); 16 TTSYPTPRPYPK(190201); PYPKPAPSSGKDYV(198211); PYPKPAPSSGK(198208) 17 CLDN2 e 100 31 4 DFYSPLVPDSMK(146 157); SNYYDAYQAQPLATR(192 206); SEFNSYSLTGYV(219 230) 18 19

20 c 21 CLDN3 1695 78 4 VYDSLLALPQDLQAAR(6580) ; DFYNPVVPEAQK(145156); DFYNPVVPEAQKR(145157); 22 VVYSAPRSTGPGASLGTGYDR(196216); STGPGASLGTGYDRKDYV(203220); STGPGASLGTGYDRK(203217); STGPGASLGTGYDR(203216); STGPGASLGTGYDRKD(203218) 23 c 24 CLDN4 861 51 4 VYDSLLALPQDLQAAR(6681) ; CTNCLEDESAK(104114); CTNCLEDESAKAK(104116); 25 NIIQDFYNPLVASGQK(142157) CLDN5 169 85 4 GLWMSCVVQSTGHMQCK(134150); VYDSVLALSTEVQAAR(151166); EFYDPSVPVSQK(231242); 26 RPTATGDYDKK(290300) 27 CLDN6 997 59 4 VYDSLLALPQDLQAAR(66 81) c; DFYNPLVAEAQK(146 157); DFYNPLVAEAQKR(146 158); 28 YSTSAPAISRGPSEYPTKNYV(200220); YSTSAPAISRGPSEYPTK(200217); GPSEYPTK(210217) 29 CLDN7 372 69 4 SSYAGDNIITAQAMYK(3348); AGDNIITAQAMYK(3648); GLWMDCVTQSTGMMSCK(4965) d; 30 KMYDSVLALSAALQATR(6581); MYDSVLALSAALQATR(6681); SVLALSAALQATRALMV(6985); 31 VSLVLGFLAMFVATMG(86101); QIVTDFYNPLIPTNIK(143158) 32 CLDN8 18 7 2 33 CLDN9 703 28 4 VYDSLLALPQDLQAAR(6681) c 34 CLDN10 102 24 4 ACVTDSTGVSNCK(5264); ITTEFFDPLFVEQK(142155); YTYNGATSVMSSR(192204); YHGGEDFK(207 35 214) 36 CLDN11 336 362 4 GLWADCVMATGLYHCK(51 66); GLWADCVMATGLYH(51 64); CKPLVDILILPGYVQACR(65 82); 37 PLVDILILPGYVQACR(6782); ILILPGYVQACR(7182); ILPGYVQACR(7382); MGQEPGVAK(108116); 38 FYYTAGSSSPTH(190201); FYYTAGSSSPTHAK(190203) d; FYYTAGSSSPTHA(190202); 39 YYTAGSSSPTHAK(191203); YTAGSSSPTHAK(192203) 40 CLDN12 259 17 4 SSVPNIK(115 121); SRLSAIEIDIPVVSHTT(228 244); SRLSAIEIDIPVVSH(228 242); 41 LSAIEIDIPVVSHTT(230244) 42 43 44 45 46 ACS Paragon Plus Environment 47 48 49 50 51 52 53 54 55 56 57 58 59 60 Page 11 of 58 Journal of Proteome Research

1 2 3 4 CLDN14 14 4 2 5 CLDN15 f 11 8 4 RPYQAPVSVMPVATSDQEGDSSFGK(197 221) 6 CLDN16 f 34 15 4 TCDEYDSILAEHPLK(130 144) 7 CLDN17 5 2 2 8 CLDN18 82 87 4 ESSGFTECR(56 64); GYFTLLGLPAMLQAVR(65 80); AVSYHASGHS VAYK(208 221); SVAYKPGGFK(217 9 226); TEDEVQSYPSKHDYV(247261); TEDEVQSYPSK(247257) 10 CLDN19 g 18 11 4 LYDSLLALDGHIQSAR(6681) 11 CLDN20 2 Not found 1 12 CLDN21 0 See CLDN24 13 CLDN22 10 5 2 14 g d 15 CLDN23 91 15 4 RSSVSTIQVEWPEPDLAPAIK(202 222) 16 CLDN24 54 9 2 17 CLDN25 2 3 2 18 19 a Evidence code: 4 = good evidence; 3 = moderate evidence; 2 = weak evidence; 1 = no evidence. 20 b All sequences are at least 8 amino acid residues in length. c 21 The peptide is common to claudins 3, 4, 6, and 9. d 22 Single nucleotide polymorphismassociated peptide variants found for this peptide. e Claudin 2 is considered missing in neXtProt. 23 f 24 In neXtProt, only peptides by single reaction monitoring were available for claudin 15 and 16. g In neXtProt, additional peptides have been identified for the claudin although not identified as level 4 evidence by GPM. 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 ACS Paragon Plus Environment 47 48 49 50 51 52 53 54 55 56 57 58 59 60 Journal of Proteome Research Page 12 of 58

1 2 Protein family 3 4 5 6 From the protein structure and function perspective, the claudin protein family is included in the 7 8 pfam00822 superfamily of proteins that are characterized by their four helical transmembrane 9 10 1,7,21 11 regions. The pfam00822 superfamily also includes other subfamilies such as the PMP22, EMP, 12 13 MP20 and calcium channel CACNG proteins. Human claudins are in the range of 200300 amino 14 15 acids (20–35 kD) and share the common transmembrane regions made of four helices (TM 1–4) 16 17 with two extracellular loops (ECL1 and ECL2) and one intracellular loop (ICL). 18 19 20 21 22 The general molecular architecture of the claudin protein family, determined by an extensive body 23 24 of genetic, biochemical, and biophysical experiments, is shown in Figure 1a. The four 25 26 transmembrane regions (TM 1–4) are typically 24 amino acids in length, while the αhelix of TM3 27 28 extends beyond the membrane by about 10 amino acids.53,59 The ECL1 is generally around 50 29 30 31 amino acids with charged amino acids for paracellular pore formation. A Prosite signature sequence 32 1,60 33 for the claudin family has been identified as [GN]LWx(2)Cx(7, 9)[STDENQH]C in ECL1. 34 35 A consensus sequence, WX(1722)WX(2)CX(810)C is found in ECL1 with the first Trp 36 37 flanking the end of TM 1. 61 A signature motif, GLW, is found 2 amino acids away from the N 38 39 40 terminus of the conserved CX(810)C motif. Most claudins also have an Arg marking the end of 41 42 the ECL1. Inside the cell, the claudin Nterminus (<10 amino acids), ICL (~12 amino acids), and C 43 44 terminus (25 to 55 amino acids) provide interaction sites with other proteins. Most notably, a PDZ 62 45 46 domain binding motif YV at the Cterminus is completely conserved amongst classic claudins and 47 48 also prevalent amongst the nonclassic ones. 57,63 The YV motif is the main interaction site for 49 50 51 binding other junction associated proteins such as ZO1, 2 and 3, although other residues in 52 63 53 proximity of the Cterminus also contribute to claudin’s protein binding affinity. The first 54 55 mammalian claudin structure (mouse claudin15) at 2.4 Å confirms this general claudin fourhelix 56 57 bundle that spans the full length of the lipid bilayer (Fig. 1b). 64 Subsequently, a binary structure at 58 59 60 ACS Paragon Plus Environment Page 13 of 58 Journal of Proteome Research

1 2 3.7 Å resolution of mouse claudin19 complexed to the Cterminal region of Clostridium 3 4 perfringens enterotoxin further validates the generality of the transmembrane, fourhelix bundle 5 6 scaffold and identifies the specific binding motifs in both the ECL1 and ECL2 loops to the bacterial 7 8 toxin. 65 9 10

11 12 Figure 1. a) A general scheme of the claudin protein structure; b) secondary structural motifs found 13 64 14 in the ECL loops of the Xray structure of mouse claudin15. 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 The claudin protein family expression level by immunoblotting is not necessarily correlated to its 37 38 39 RNA level (Table 1, column 5 and 6). Human claudins 24 to 27 have been tentatively assigned as 40 41 members of the claudin family, and the evidence for these members remains tenuous even at the 42 43 RNA level (Table 1, column 6). Many antibodies have been in use for claudin detection in both 44 45 cells and tissues (Table 1, column 5), although polyclonal antibodies recognize multiple epitopes 46 47 48 and antibody crossreactivity is a significant concern in analyzing claudins. Currently, four human 49 66 50 claudin proteins (claudin8, 9, 20, and 22) are considered “missing” at the MS level (as defined by 51 52 HUPO: the Human Proteome Organization) with evidence for their presence only at the 53 54 transcription level, although for human claudins 8 and 9, protein presence has been suggested by 55 56 immunoblotting and likely requires further validation. 67,68 In terms of good MS evidence, eleven of 57 58 59 the human claudins, namely claudins 1, 2, 3, 4, 5, 6, 7, 10, 11, 12 and 18 have been identified 60 ACS Paragon Plus Environment Journal of Proteome Research Page 14 of 58

1 2 multiple times by more than one unique peptide with good log(E) scores (Table 2). Further, four 3 4 human claudins (15, 16, 19, and 23) have been identified multiple times by just one unique peptide 5 6 with good log(E) scores. In the case of claudin 9, only one nonunique peptide has been identified 7 8 multiple times with good log(E) scores, which is also found in claudins 3, 4, and 6. Overall, of the 9 10 11 23 definitive human claudins (not including the disputed claudins 26 and 27), human claudins 8, 9, 12 13 14, 17, 20, 22, 24 and 25 do not have definitive MSbased protein evidence. Therefore, only about 14 15 half of the human claudin protein family can be considered as identified with certainty by MS at the 16 17 protein level, with the other half either “missing” or uncertain by MS. For human claudins (7, 10, 18 19 20 11, 18, and 19) with isoforms by , no credible MS evidence exists for their 21 22 presence as protein isoforms. One caveat is that several of these may be enriched in tissues that 23 24 have not been probed for claudin expression by MS. For instance, claudin18 has been shown by 25 26 immunologic techniques to be highly expressed at the protein level in stomach and lung. 27 28 29 30 31 32 33 2. A general overview of the tissue and functional profile of the human claudins 34 35 36 37 Claudins are found widely expressed, either as gene transcripts or expressed proteins, in diverse 38 39 69 40 tissues. The vast majority of claudin expression is found in barrier forming epithelial and 41 42 endothelial cells. Not all claudins are concurrently expressed by all tissues, and it is these 43 44 differences in claudin expression that regulate cell function, most notably, paracellular barrier 45 46 function. Reflecting a predominant role in regulating barrier function, claudins frequently are found 47 48 concentrated at the tight junction, however some claudins are also found at other intracellular 49 50 1,14,70 51 locations, such as regions along the basolateral plasma membrane. Nuclear localization of 52 1,10,20,7173 53 claudins has also been observed. Roles for nonjunctional claudins are not well 54 55 understood, although the literature correlating nuclear claudin localization with tumor metastasis 56 57 suggest roles for these claudin pools in regulating cell growth, division and migration.61,7478 58 59 60 ACS Paragon Plus Environment Page 15 of 58 Journal of Proteome Research

1 2 3 4 The most prevalent roles for claudins are in barrier and pore formation, and the details of how 5 6 individual claudins influence tight junctions have been extensively reviewed. 21,57,61,79 The majority 7 8 of claudins are primarily barrier forming, based largely on studies of cell line models where the 9 10 11 effects of increasing and decreasing claudin expression on measures of barrier function was 12 13 assessed, including transepithelial resistance and paracellular flux of small molecular weight 14 15 tracers. By contrast, claudin2, 10, 15, and 17 are primarily poreforming and lead to increased 16 17 paracellular permeability correlating with increased expression as a result of formation of 18 19 64 20 paracellular ion and water channels. The highresolution structure of claudin15 has provided 21 22 several insights into the structural basis for claudin paracellular ion selectivity. Homology modeling 23 24 and structure determination of other claudins will likely provide a valuable approach to further 25 26 define the permeability characteristics of other claudins. Claudins also interact with other junction 27 28 membrane proteins, such as occludin, tricellulin, and junctional adhesion moleculeA (JAMA), to 29 30 10,21,80 31 stabilize and regulate their membrane retention for improved barrier formation. 32 33 34 35 Claudin mutations have been implicated in human diseases affecting ion homeostasis. 61,7477,81 One 36 37 of the best characterized claudinassociated diseases is familial hypomagnesemia with 38 39 8284 40 hypercalciuria and nephrocalcinoisis that is caused by mutations in claudin16 and claudin19. 41 42 The majority of the mutant claudin16 alleles (12 out of 16) show a missense mutation in the highly 43 44 conserved amino acids in ECL1 or ECL2. 83,85 Such mutations have been postulated to alter the 45 46 tertiary structure of the extracellular domains in claudin complexes and thus change the cation 47 48 specificity of claudin pores. Other mutations affect claudin16/claudin19 interactions diminishing 49 50 82 51 the transport and assembly of these claudins into tight junctions. Claudin mutations can be linked 52 53 to diverse, tissue specific pathologic outcomes, which underscores the need to understand the 54 55 structural basis that regulates their function and proteinprotein interactions. 56 57 58 59 60 ACS Paragon Plus Environment Journal of Proteome Research Page 16 of 58

1 2 3 4 3. Post-translational modification of claudins. 5 6 7 8 PTMs greatly expand the claudin chemical diversity found in cells and in turn further diversify 9 10 86 11 claudin function. Potential biological processes that may be differentially regulated through 12 13 claudin PTM include protein interaction partnerships, trafficking, subcellular localization, oligomer 14 15 (hetero or homo) assembly, and finally net claudin homeostasis. 13 Here we discuss several classes 16 17 of claudin PTMs: phosphorylation, palmitoylation, ubiquitination and glycosylation (Figure 2). 18 19 20 Proteolysis is another way by which claudins can be subject to PTM. It is not well established 21 22 whether any proteolytic claudin fragments have biological activity or whether this largely serves to 23 24 decrease the total claudin protein pool. 25 26 27 28 Figure 2. Known or hypothetical PTMs of claudins and the regions in which they are predicted to 29 30 31 occur. Predicted PTMs include phospohorylation (red oval), palmitoylation (yellow line), 32 33 ubiquitination (blue circle), proteolysis (green marks) and glycosylation (grey box). 34 35 N-terminus C-terminus 36 37 TM1 TM2 TM3 TM4 38 39 O O O SUMO or Ub HO 40 O P serine protease HO O 41 HO O S N O cleavage site 42 O 14 H AcHN 43 44 45 46 47 Phosphorylation 48 49 Currently, phosphorylation on Ser/Thr/Tyr residues is the most abundantly observed claudin PTM. 50 51 Theoretical predictions of potential phosphorylation sites indicate that up to 10 phosphorylation 52 53 sites are possible by a variety of protein kinases (e.g. PKA/C, MAPK, WNK, Src, and Eph).86 Most 54 55 56 of these predicted phosphorylation sites are located in the Cterminal tail. One important feature of 57 58 claudin phosphorylation is that the Tyr residue found in the highly conserved PDZ binding motif at 59 60 ACS Paragon Plus Environment Page 17 of 58 Journal of Proteome Research

1 2 the end of the Cterminus can be a phosphorylation site by the Eph receptor tyrosine protein kinase, 3 4 thus providing a means to regulate its binding activity. For example, in the case of claudin4 in 5 6 HT29 colon carcinoma cells, EphA2 activation leads to claudin4 tyrosine phosphorylation, and this 7 8 specific Tyr phosphorylation lowers its association with ZO1 and leads to less claudin4 9 10 87 11 integration into cell junctions and higher paracellular permeability in MDCK cell models. This 12 13 result indicates that EphA2 Tyr phosphorylation near the claudin Cterminus may alter the function 14 15 of the tight junction and promote a regime change in signaling at the tight junction. Tyr 16 17 phosphorylation for claudin5, detected by a general phosphorTyr antibody, has also been linked to 18 19 88 20 increased paracellular permeability in cultured human brain capillary endothelial cells. Similarly, 21 22 Ser/Thr phosphorylation of claudins is known to influence the barrier strength of cell junctions. 23 24 Phosphorylation of T192 of claudin3 by PKA has been found in ovarian cancer cells as a 25 26 mechanism for disruption of normal junctional function. 89 In other instances, Ser/Thr 27 28 phosphorylation of claudin7 can upregulate the caspase pathway to increase the chemosensitivity 29 30 31 of lung cancer cells toward cisplatin or increase paracellular permeability to chloride anions in 32 90,91 92 33 kidney cells. Largescale phosphoproteomics investigations (from PhosphoSitePlus with 34 35 specific journal article references and the Global Proteome Machine 93 have identified multiple 36 37 Tyr/Ser/Thr sites of claudin1–12, 15, 16, 18, and 23 in human T cells, embryonic stem cells and 38 39 90,91,94113 40 cancer cells (summarized in Table 3). However, the specific roles of these claudin PTMs 41 42 remain largely unknown without functional validation from biological experiments. 43 44 45 46 Table 3. Human claudin PTM sites from PhosphoSitePlus or the Global Proteome Machine (GPM) 47 48 databases (p = phosphorylation; ub = ubiquitination108 ). Sites in bold are from GPM without a 49 50 51 specific article reference from PhosphoSitePlus. 52 53 Claudin PTM sites 54 55 1 T195 p; Y199 -p; S205 -p; S206 p; Y210 p 56 2 S192p; Y194p; Y195p; Y198p; S208p; S219p 57 58 3 Y198 p; S199 p; S203 p; T204 p; S209 p; T212 p; Y214 p; Y219 p 59 60 ACS Paragon Plus Environment Journal of Proteome Research Page 18 of 58

1 2 4 Y193 p; S194 p; Y197 p; Y208 p 3 5 Y148p; Y152 -p; Y154 -p; S155p; T207p; Y212p; Y217p 4 5 6 Y152 -p; S201 p; T202 p; S203 p; S208 p; S212 p; Y214 p; Y219 p 6 7 7 K203ub; S204p; S206p; S207p; K208ub; Y210p 8 9 8* Y67 p; S69 p; S74 p; S21 5-p 10 11 9 Y200p 12 13 10 S94 p; T198 p; S199 p; S202 p 14 15 11 Y191p; Y192p; T193 -p; S196p; S197p; S198p 16 12 T111 p; S115 p; S116 p; K121 ub; Y211 p; S216 p; Y220 p; S228 p; S231 p 17 18 15 T84p; S111p; S211 -p; S217 -p; S218 -p 19 20 16 S217 p; S278 -p 21 22 18 Y206p; S210p; Y211p; S214p; S217 -p; Y220p; T229 -p; Y241p; T247p; S253p; Y254p; S256p; 23 24 Y260p 25 26 23 S203p; S204p; S206 -p; S207p; Y223p; Y224p 27 28 29 94 * Phosphorylated claudin8 peptides were reported. 30 31 32 33 34 The effects of phosphorylation are claudin specific, in regards to the specific residue modified, and 35 36 vary even amongst highly homologous claudins. For example, S208 phosphorylation of claudin2 is 37 38 required for its membrane retention and reduced trafficking to lysosomes in MDCKII cells; 39 40 however, this phosphorylation does not alter its binding to ZO1 or ZO2.30 In the case of claudin5, 41 42 43 PKA phosphorylation at T207 in endothelial cells, while conducive to its translocation into the tight 44 114,115 45 junction membrane, leads to increased paracellular permeability. In addition to barrier 46 47 function, claudin phosphorylation is also known to control selective ion permeability. For example, 48 49 reduction of claudin16 phosphorylation at S217 results in impaired Mg 2+ permeability in renal 50 51 MDCK cells. 116 As an additional confounding variable, tyrosines are subject to nitrosylation under 52 53 117 54 conditions of oxidative stress. Evidence for tyrosine nitration of claudins has only been found in 55 118 56 rats at this point, where renal claudin2 nitration was detected in early diabetic rats. 57 58 59 60 ACS Paragon Plus Environment Page 19 of 58 Journal of Proteome Research

1 2 Palmitoylation 3 4 Similar to phosphorylation, palmitoylation can significantly alter claudin localization, protein 5 6 interactions, trafficking and stability. 86 The TM2 and TM4 of claudins are each flanked by a 7 8 cysteine residue that has a palmitoylation motif. Some claudins have additional cysteine residues 9 10 11 potentially available for palmitoylation in the ICL and Cterminal tail. For example, in MDCK cells 12 13 two cysteine residues in the ICL and two more in the Cterminal tail of claudin14 must be 14 15 palmitoylated as a requirement for efficient plasma membrane localization and tight junction 16 17 assembly.119 However, in transfected HEK cells, palmitoylation of claudin7 is associated with 18 19 20 partitioning into glycosphingolipid membrane microdomains and inhibits integration into tight 21 22 junctions, suggesting a model where palmitoylation controls the relative amounts of claudin7 in the 23 24 basolatersal vs. tight junction pools. 120 Protein Sacylation has been profiled at the proteome 25 26 scale, 121123 and given the difficulties in measuring palmitoylation by standard biochemical 27 28 techniques, this is an area that would be ideally suited for state of the art proteomics analysis. 29 30 31 32 33 Ubiquitination and SUMOylation 34 35 Evidence for human claudin ubiquitination and SUMOylation is scarce but more frequently 36 37 observed in model cell lines of other organisms. Polyubiquitination of claudin5 is observed in 38 39 124 40 claudin5 transfected HeLa cells at K199 to target the protein for proteasomal degradation. This 41 42 results in claudin5 being removed from the plasma membrane and loss of the tight junction strands, 43 44 indicating that with this particular PTM claudin stability can be compromised by a potential 45 46 mechanism of increased turnover. Claudin ubiquitination has been more consistently observed in 47 48 canine, mouse, and rat cell models according to PhosphositePlus. For example, polyubiquitination 49 50 51 of claudin1 in MDCK cells appears to be dependent on LNX1p80 overexpression but the sites of 52 53 ubiquitination are not specified and also do not involve Lys48, a site that is typically ubiquitinated 54 55 for proteasomedependent degradation.125 SUMOylation of claudin2 at K218 is also reported in 56 57 MDCK cells in which claudin2 is removed from the lower lateral membrane but not the tight 58 59 60 ACS Paragon Plus Environment Journal of Proteome Research Page 20 of 58

1 126 2 junction. It is noteworthy that not all claudins contain ubiquitination sites, although the classic 3 4 claudins are thought to be the primary candidates for ubiquitination, and this is supported by Lys 5 6 ubiquintination sites identified by large scale proteomics (Table 3, claudins 7 and 12). The current 7 8 understanding of claudin ubiquitination/SUMOylation on claudin localization, function or stability 9 10 11 is based primarily on studies of nonhuman claudins. It is likely that direct evidence for 12 13 ubiquitination/SUMOylation of human endogenous claudins will be forthcoming. 14 15 16 17 Glycosylation 18 19 20 Experimental evidence for claudin glycosylation is again scarce, perhaps due to the low abundance 21 22 level of these proteins that require specific enrichment prior to such analyses. This lack of data also 23 24 suggests that claudin glycosylation is a rare event. Putative N and Oglycosylation sites on some 25 26 claudins have been identified.127,128 Some of these predicted Oglycosylation sites overlap with 27 28 predicted phosphorylation sites. These overlaps are known as the Yin Yang sites, where the two 29 30 31 modifications can reciprocally occur on the same Ser/Thr residue or occupy adjacent residues. For 32 33 example, in the case of human claudin1, eight potential OGlcNAc modifications sites were 34 35 predicted to be located at T190, T191, S24, S56, S185, S192, S205 and S206. Of these eight 36 37 potential glycosylation sites, four (T191, S192, S205, and S206) were also predicted as Yin Yang 38 39 40 sites. However, there is no compelling evidence suggesting a role for this PTM overlap in claudin 41 42 function. In addition, Nglycosylation sites have been predicted to exist in the ECL1 of claudins1 43 44 and 12.129 Furthermore, claudin26 homolog TMEM114 is also predicted to be Nglycosylated at 45 46 position N54 and N88.129,130 47 48 49 50 51 In this review, we have undertaken prediction of Nglycosylation sites in all claudin proteins using 52 131 53 the NetNGlyc 1.0 Server , where a threshold >0.5 was applied for this analysis. This server 54 55 predicts Nglycosylation sites in human proteins using artificial neural networks that examine the 56 57 sequence context of AsnXaaSer/Thr motifs. The predicted Nglycosylation sites tend to be much 58 59 60 ACS Paragon Plus Environment Page 21 of 58 Journal of Proteome Research

1 2 less abundant compared to those for Oglycosylation. While potential glycosylation sites were 3 4 observed for claudins1, 7, 10, 12, 15, 18, 22, 23 and 24 (all with jury values between 7/9 and 9/9, 5 6 except for claudin7 with a jury value of 5/9), most of the predicted Nglycosylation sites are in the 7 8 helical regions and therefore not likely to be accessible to glycosylation machinery, except for 9 10 11 claudin12 at 47 NLTV, a segment in the ECL1 region. Claudin12 expressed by transfected HeLa 12 13 cells is Nglycosylated as determined by a complex banding pattern observed by immunoblot that 14 15 compresses into a single band when treated with PeptideNGlycosidase F (Koval, unpublished 16 17 results), indicating that this is a functional glycosylation site. Claudin12 transfected COS7 kidney 18 19 132 20 fibroblasts show a similar banding pattern also suggesting it is glycosylated. However, 21 22 immunoblots of claudin12 expressed by endothelial cells and in human brain homogenates show 23 24 predominantly a single band,133 which supports the notion that in an endogenous setting claudin12 25 26 glycosylation is less frequent and instead likely to be a regulated process. Further work is needed to 27 28 determine the extent of claudin glycosylation in a physiologic setting, although most data in the 29 30 31 literature suggests that Nglycosylation of claudins in situ is an exceptionally rare event. 32 33 34 35 Another program called NetOglyc,134 which produces neural network predictions of mucin type 36 37 GalNAc Oglycosylation sites in mammalian proteins, was also used to analyze claudins. Only sites 38 39 40 with scores >0.5 are predicted as glycosylated and marked as positive for Oglycosylation. Using 41 42 this approach, all members of the claudins, except 4, 7, 9 and 24 were predicted to be O 43 44 glycosylated (Table 4). 45 46 47 48 Table 4. Identification of potential Oglycosylation sites in human claudins, using NetOGlyc 4.0 134 . 49 50 51 A threshold of 0.5 was applied for this analysis. 52 53 54 55 Gene/Protein O-glycosylation in the C-terminal tail 56 CLDN1 Y (T195) 57 CLDN2 Y (T204, S206, S207) 58 CLDN3 Y (S196 ,S200 ,T201, S206) 59 60 ACS Paragon Plus Environment Journal of Proteome Research Page 22 of 58

1 2 CLDN4 N 3 CLDN5 Y (S201, T207) 4 CLDN6 Y (S194, S201, T202, S203, S208, S212) 5 CLDN7 N 6 CLDN8 Y (S201, T204, T205, S208, S215) 7 CLDN9 N 8 CLDN10 Y (T198, S199, S202, T215, T216) 9 CLDN11 Y (S196) 10 11 CLDN12 Y (S212) 12 CLDN14 Y (T201, T202, T203, T204, T207, S224) 13 CLDN15 Y (S204, S211) 14 CLDN16 Y (S287, S289) 15 CLDN17 Y (T207, T213, T214) 16 CLDN18 Y (S210, S228) 17 CLDN19 Y (S195, S204) 18 CLDN20 Y (T198, S203) 19 20 CLDN22 Y (T204) 21 CLDN23 Y (S197, S203, S204, S206) 22 CLDN24 N 23 CLDN25 Y (S207) 24 25 26 27 28 Interestingly, all of the predicted Oglycosylation sites are in closeproximity to known or predicted 29 30 phosphorylation sites of claudins in the Cterminal region.72,89,135 This close proximity of the two 31 32 PTM types may suggest potential crosstalk between OGlcNac modification and phosphorylation at 33 34 these positions. The YinOYang server 131 can calculate the Oglycosylation potential for all Ser, Thr, 35 36 136 37 and Tyr residues in a protein sequence and crosscheck these sites against NetPhos 2.0 predictions 38 39 for potential phosphorylation sites to determine potential Yin Yang sites with a high possibility for 40 41 both modifications. Currently the experimental evidence for mammalian claudin glycosylation 42 43 exists only in mice on S241 of claudin16.137 44 45 46 47 48 It should be noted that the above predictions are purely sequencebased and do not incorporate the 49 50 structural information of the individual claudin proteins. Given the importance of glycosylation in 51 52 protein sorting and endocytosis, a more indepth investigation of potential claudin glycosylation in 53 54 human is warranted to help understand whether these modifications play roles in regulating claudin 55 56 57 localization and recycling. 58 59 60 ACS Paragon Plus Environment Page 23 of 58 Journal of Proteome Research

1 2 Proteolysis 3 4 Claudin proteolysis is known to alter the barrier function of intestinal epithelial cells in human and 5 6 animals.138,139 Intestinal barrier integrity is often compromised in autoimmune diseases such as 7 8 coeliac or inflammatory bowel disease, in which gut/intestinal permeability is higher than 9 10 140 11 normal. Such barrier disruption is frequently mediated by proinflammatory cytokines such as 12 13 IFNγ, partly through downstream selective cleavage of some claudins. In particular, in T84 14 15 (intestinal human epithelial) cells, claudin2 is specifically cleaved, by yettobe identified 16 17 proteases, in either the ICL or ECL2 after IFNγ incubation, whilst claudins 1, 3, and 4 in the same 18 19 141 20 cells remain intact. In addition, the basal expression level of all claudin is reduced by IFNγ 21 22 treatment, and a nonspecific Ser protease inhibitor (AEBSF) is able to significantly “rescue” this 23 24 claudin2 expression loss and cleavage. SwissProt bioinformatics analysis has identified a Ser 25 26 protease cleavage site in ECL2 of claudin2 but not in claudins 1, 3 and 4. The transmembrane 27 28 claudin2 cleavage is restricted to the TritonX100 soluble membrane fractions but not in the 29 30 31 TritonX100 insoluble cytoskeletal fraction that contains uncleaved claudin2. This may suggest 32 33 that claudin proteolysis is dependent on claudin localization and thus spatially limited, perhaps due 34 35 to limited access of proteolytic enzymes to the tight junction associated claudin pool. Nonetheless, 36 37 most evidence to date suggests that the net effect of proteolysis is to decrease overall junction 38 39 40 associated claudin protein content, perhaps by inhibiting incorporation of newly synthesized claudin 41 42 into tight junctions. Claudin proteolysis may also contribute to posttranslational control of claudin 43 44 turnover. For instance, claudin2 and claudin4 have half lives of 12 and 4 h respectively, which is 45 46 determined by the Cterminal cytoplasmic tail as demonstrated using chimeric claudin constructs in 47 48 MDCK cells.142 Claudin5 turnover is cell dependent, ranging from 70 min in HUVEC 124 to over 49 50 143 51 3 hours for bovine retinal endothelial cells. 52 53 54 55 4. Mass spectrometry of endogenous claudins 56 57 58 59 60 ACS Paragon Plus Environment Journal of Proteome Research Page 24 of 58

1 2 Mass spectrometry of claudins and their partners from cells or tissues 3 4 More recently, MS analysis, often coupled with affinity enrichment, has seen wider use in 5 6 characterization of endogenous claudins at the protein level given the advantage of the technique in 7 8 identifying lowabundance membrane proteins. An early demonstration of a claudintargeted 9 10 11 enrichment strategy prior to MS employed a GSTfusion affinity column made from Clostridium 12 144,145 13 perfringens enterotoxin (CPE) that binds to the ECL2 of claudins 3, 4, and 7. In these reports, 14 15 affinity enriched claudins from cultured rat cholangiocytes were trypsin digested and subjected to 16 17 first liquid chromatography (LC) and then MS to identify the enriched claudins. Coelution of some 18 19 20 nonCPE binding claudins was also observed by immunoblotting and MS. Relative MS protein 21 22 quantification using SILAC (stable isotope labeling by amino acids in cell culture) also identified 23 24 potential plasma membrane associated claudinbinding proteins as well as claudinbinding proteins 25 26 in the cytosol, nucleus and mitochondria. Endogenous human claudin5 in brain endothelial cells 27 28 has been enriched by cell fractionation for mass spectrometry. 146 Claudin5 partners were also 29 30 31 identified by coimmunoprecipitation and LCMS. It was demonstrated with additional siRNA 32 33 experiments that Gprotein subunit αi2 was a new partner of claudin5 and required for proper 34 35 claudin5 assembly in the tight junction. 36 37 38 39 147 40 Recently, a claudin interactome for Drosophila has been reported. Specific antibodies were 41 42 generated against the claudin Megatrachea in the fly embryos. Subsequent immuoprecipitation, 43 44 followed by MS analysis, identified 142 protein partners of the claudin Megatrachea, including 10 45 46 bona fide members of the septate junctional complex, the invertebrate equivalent to the apical 47 48 junctional complex. Further validation by RNA interference uncovered new protein partners for 49 50 51 Megatrachea interaction including members of the clathrinmediated vesicle proteins. This is 52 53 analogous to human claudin4 internalization by clathrinmediated endocytosis that depends on a 54 55 sorting sequence on the ICL, and also suggest that there are significant parallels between 56 57 invertebrate and vertebrate claudinbased protein complexes and regulation.148 A very recent 58 59 60 ACS Paragon Plus Environment Page 25 of 58 Journal of Proteome Research

1 2 proteomics investigation on proteins proximal to claudin4 and occludin in MDCK II cells also 3 4 significantly expanded the inventory of proteins at the tight junction, particularly in signaling and 5 6 endocytic trafficking proteins. 149 While a comprehensive analysis of the endogenous human 7 8 claudins and their interactomes in various cell types remains absent, these studies serve as important 9 10 11 foundations to further proteomewide investigations for human endogenous claudins. 12 13 14 15 Membrane proteomics of endogenous claudins 16 17 Because endogenous claudins are lowabundance proteins in vertebrate epithelial cells, it is difficult 18 19 150 20 to profile their expression comprehensively without protein enrichment techniques. Nonetheless, 21 22 it is apparent that there are tissue specificities to the expression of claudin members and claudin 23 24 PTMs and that signalling proteins have substrate preferences for some claudin members over 25 26 others. 12 Claudin antibodies are widely available and used for specific immunodetection, however 27 28 these need to be carefully monitored and tested for crossreactivity and nonspecificity. 151 29 30 31 Antibodies enable relative claudin expression levels to be monitored by standard immunoblotting 32 33 methods as well as immunofluorescence imaging. While these approaches are powerful for 34 35 investigating individual claudins, they are limited in that they do not enable direct calculation of 36 37 relative claudin stoichiometry and are difficult to employ to measure PTMs. 38 39 40 41 42 The accurate and systematic expression and PTM profiling of claudins with spatial and temporal 43 44 resolution is currently inaccessible without rigorous methods that can isolate, enrich, and 45 46 distinguish endogenous claudins. Bottomup membrane proteomics methods have been applied 47 48 specifically to enrich membrane junction proteins from intestinal human epithelial cells (T84) or 49 50 152,153 51 mouse liver tissues. A junctiontargeting antibody, as validated by immunofluorescence 52 53 staining, was used to enrich cell junction proteins from T84 that were then immunoblotted to verify 54 55 the presence of tight junction proteins, stained by silver to verify protein size, and digested for 56 57 analysis by mass spectrometry .152 Over nine hundred proteins were identified, half of which were 58 59 60 ACS Paragon Plus Environment Journal of Proteome Research Page 26 of 58

1 2 synaptic or signalling proteins. However, the coverage of known junctional proteins was low. 3 4 Nevertheless, this study provided a starting point for more indepth and adequate analysis of 5 6 membrane junction proteins such as claudins. A subsequent proteomics investigation utilised 7 8 membrane protein enrichment protocols involving cell fractionation followed by repeated guanidine 9 10 11 treatment and NP40 extraction. This optimized analysis was able to identify claudin1 and 3 by MS 12 153 13 from the bile canaliculi fraction of mouse livers. More success in claudin protein identification 14 15 has been reported in rat renal inner medullar collecting duct cells, in which claudin3, 4, 7, 8, and 16 17 10 were found and listed on the IMCD Proteome Database. 154,155 18 19 20 21 22 The low abundance of the plasma membrane proteome, concomitant with the inadequate proteomic 23 24 coverage of the full complement of membrane proteins, has been recognized for some time and 25 26 explains the challenges in proteomic investigations on claudins or other junctional proteins. 27 28 Hydrophobic proteins are difficult to capture into matrices to enable separation and subsequent 29 30 31 identification, which results in hydrophilic proteins being vastly overrepresented in proteomic 32 33 analyses. Methods for the capture and enrichment of hydrophobic membrane proteins addressing 34 35 this imbalance include isolation in sodium bicarbonate,156 aqueous two phase partitioning using 36 37 polyethylene glycol and dextran 157,158 or nonionic detergents and sucrose gradients for highly 38 39 159 40 insoluble membrane proteins. Recently, a membrane proteome analysis of human embryonic 41 160 42 stem cells (hESCs) identified three claudins: claudin3, 6, and 7. Claudin6 was the most 43 44 abundant of the three, which is consistent with genetic analysis showing claudin6 as the highest 45 46 expressed claudin member in hESCs. 69 Although claudin2, 4, and 12 genes were also reported to 47 48 express in many hESCs, their protein products were not identified in this analysis. Compared to a 49 50 161 51 prior hESC proteomics study that did not identify any claudin, methods with membrane protein 52 53 enrichment may help with enhancing claudin and tight junction membrane protein coverage. 54 55 56 57 Top-down proteomics of claudins 58 59 60 ACS Paragon Plus Environment Page 27 of 58 Journal of Proteome Research

1 162166 2 Topdown MS has been in decades of development for characterizing intact proteins. 3 4 Recently, topdown proteomics has emerged as a powerful approach for characterizing proteins in 5 6 their native and intact form (Figure 3).167171 Unlike bottomup proteomics, in which proteins are 7 8 broken into peptide fragments for identification, topdown proteomics reveals the precise complete 9 10 11 “proteoform(s)” of a protein without loss of the chemical information embedded in the structure of 12 170 13 the protein or proteoform. While bottomup proteomics will remain a powerful approach for 14 15 obtaining a comprehensive claudin overview, the high level of homology between claudin family 16 17 members requires further validation for reliable protein identification and characterization. Top 18 19 20 down proteomics in this regard holds the definite promise of capturing the exact chemical details of 21 22 claudins, which are well below 50 kD in size, with high fidelity and thus will be able to uncover 23 24 authentic biological activities encoded by these claudin protein characteristics. In addition, the 25 26 coverage bias in bottomup proteomics toward more abundant proteins is better mitigated in the 27 28 topdown approach. This is particularly relevant to endogenous claudins because generally low 29 30 31 abundance membrane proteins with a high level of homology can be prone to misidentification and 32 172,173 33 irreproducible quantification in proteomics. 34 35 36 37 Advances in topdown proteomics methodology, in particular with improved protein ion excitation 38 39 40 and resolution in mass spectrometry for large protein ion fragmentation, have already enabled large 41 174176 42 scale, robust protein identification in human cells. For example, HeLa S3 cells have been 43 44 fractionated by multiple methods in sequence including solution isoelectric focusing, geleluted 45 46 liquid fraction entrapment electrophoresis, and nanocapillary liquid chromatography. 174 The 47 48 resulting fractions were then analyzed by MS in which whole protein molecules could be ionized 49 50 51 and fragmented in tandem for protein identification. In total over 3,000 protein products from 52 53 1,043 genes were found, displaying a full range of protein diversification by PTMs, RNA splicing 54 55 and proteolysis. Even proteins as large as over 100 kDa and up to 11 transmembrane helices were 56 57 mapped. More importantly, unknown isoforms of endogenous proteins related to senescence were 58 59 60 ACS Paragon Plus Environment Journal of Proteome Research Page 28 of 58

1 2 uncovered, thus demonstrating the discovery power of topdown proteomics in unravelling complex 3 4 biology. This approach has also been expanded to other cell lines such as H1299 with improved 5 6 proteome coverage of over 5,000 proteoforms, including previously unknown lipidanchored and 7 8 hyperphosphorylated proteins. In these HeLa S3 cells, multiple claudin proteoforms have been 9 10 11 identified, including claudin2, 7, 15 and 17. The topdown proteomics approach, combined with 12 13 specific claudin enrichment protocols, will likely further increase claudin coverage with reliable 14 15 PTM information in many other cell types. In addition to a combination of bottomup and topdown 16 17 proteomics techniques to ensure sensitive detection and extensive coverage of lowabundance 18 19 20 membrane proteins in their native states with accurate PTM information, the “middledown”

21 177180 22 approach, in which limited digestion is used in conjunction with topdown techniques, is also 23 24 amenable to extracting chemical information in proteomics. This approach has been in use for 25 26 understanding histone and ribosome PTMs and may very well be suited for the claudin system. 27 28 29 30 31 Figure 3. Topdown and bottomup proteomics: advantages (in bold) and disadvantages (in grey). 32 less bias in purification of low abundance or hydrophobic proteins; 33 higher throughput and coverage; Top-Down high content information on protein ID and PTMs 34 extensive purification needed for larger and less solublizable proteins; high demand on ionisation manipulation and instrumentation; 35 a mixture of a small databases in development 36 number of large peptides Database 37 Purification Mass Analysis (Gel/LC) Spectrometry 38 (ID/PTMs) protein mixture 39 facile purification; 40 higher certainty in peptide mass measurement; Bottom-up extensive databases in operation high bias against low abundance and hydrophic peptides during purification; 41 limited throughput in mass measurment; a mixture of a large higher ambiguilty or information loss in protein ID/PTMs 42 number of small peptides 43 44 45 46 47 Targeted proteomics of claudins 48 49 In targeted proteomics, the mass analysis is focused on one protein or a small set of proteins to 50 51 reduce the mass bias and interference inherent in complex biological samples for improving 52 53 sensitivity and quantification. As such, targeted proteomics of the claudin system represents a more 54 55 56 quantitative option for claudin stoichiometry and PTM characterization with enhanced accuracy and 57 58 precision. One earlier example of such approach was demonstrated in quantifying low abundance 59 60 ACS Paragon Plus Environment Page 29 of 58 Journal of Proteome Research

1 181 2 proteins, such as growth hormones, in very complex and highly dynamic human plasma samples. 3 4 Since then, target proteomics has become widely adopted in both topdown and bottomup 5 6 proteomics for biomarker validation, 182 human plasma proteome analysis, 183185 multiplex protein 7 8 activity assays, 186 PTM profiling, 187,188 and masslinked immunoselective assays. 189 Great progress 9 10 11 has also been made in improving the quantification, throughput and bioinformatics analysis aspects 12 33 13 essential for targeted proteomics to scale up and accommodate broader biological applications. 14 15 35,190194 Recently, targeted proteomics has enabled quantitation of claudin5 expression in the 16 17 plasma membrane fraction of a human hCMEC/D3 cell line (0.879 fmol claudin5/µg protein) as 18 19 195197 20 well as in human brain microvessels (3.39 fmol claudin5/µg protein). Similar levels of 21 22 claudin5 expression (6–8 fmol/ µg protein) are also reported from investigations with mouse brain 23 24 capillaries.41 For the claudin system that has many homologous members, these advances in 25 26 targeted proteomics will continue to facilitate a more reliable analysis of lowabundance claudin 27 28 expression and PTM profiles with temporal and spatial resolution. 29 30 31 32 33 5. Chemical capture and enrichment of endogenous claudins for chemical proteomics 34 35 36 37 Claudin extraction and subcellular fractionation 38 39 40 Existing subcellular or cell compartment fractionation protocols provide spatial segregation and 41 198,199 42 consequentially also enrichment of proteins. However, cytoplasmic aggregates are often 43 44 observed and can render fractionation inefficient. Because every cultured cell line has different 45 46 cytoplasmic and cytoskeletal organisations, optimal conditions for an ideal homogenate before 47 48 fractionation can be highly variant and difficult to obtain. Nonetheless, cellular fractionation has 49 50 51 been used regularly provided that some quality control measures such as marker enzymes or 52 53 morphological analysis are also employed. Subcellular localization of claudins has been 54 55 investigated in monolayers of intestinal Caco2 cells, where knockdown of endogenous protein 56 57 kinase C theta activity decreased membrane and cytoskeletalassociated claudin1 and 4, and 58 59 60 ACS Paragon Plus Environment Journal of Proteome Research Page 30 of 58

1 200 2 increased the cytosolic pool of these claudins as detected by fractionation and immunoblotting. In 3 4 Caco2 cells, depletion of cholesterol by methylßcyclodextrin leads to displacement of claudin3, 5 6 4, and 7 from the cholesterol rich membrane domains along with other TJ proteins such as JAMA 7 8 and occludin. However, claudin1 is not affected by this depletion.201 Overall, claudin extraction 9 10 11 methods have been investigated, albeit more consistently in animal cell models such in MDCK 12 150 13 cells. For example, in MDCK II cells, the extractability of claudins is not affected by cholesterol 14 15 levels, however different detergents, such as Triton X100 or CHAPS, partition different claudins 16 17 into different sizes of aggregates.202 Also in MDCK cells, sodium caprate is shown to increase 18 19 203 20 solubilities of claudin4 and 5 in Triton X100 but not claudin1, 2, and 3. As it is widely 21 22 recognised that the ability of a detergent to extract membrane proteins is celltype dependent and 23 24 different detergents differ in ability to solubilize different membrane proteins,150,204 extraction or 25 26 fractionation of claudins or claudin proteoforms from various cell types and cellular compartments 27 28 will remain extremely challenging and likely require celltype specific optimisation of isolation 29 30 31 protocols prior to mass spectrometry. 32 33 34 35 Covalent tagging of endogenous claudins 36 37 Another related issue is claudin enrichment, which can be partially achieved by subcellular 38 39 40 fractionation. However, multispan membrane proteins such as claudins are, in general, more 41 42 resistant to extraction and identification. While methods on the basis of noncovalent binding are 43 44 helpful, 205 approaches for covalent labelling of endogenous claudins are needed for a reproducible 45 46 capture and enrichment protocol with spatial resolution even at very low expression levels. The 47 48 approach of chemical proteomics with protein tagging, in which proteins are modified by small 49 50 51 molecule probes carrying an affinity anchor, fluorescent reporter, and other functional groups for 52 206210 53 postcapture analysis, may be suited for systemwide profiling of protein or enzyme activities. 54 55 Covalent labelling of claudins in the plasma membrane is frequently achieved by using amine or 56 57 thiol reactive small molecule probes. 211215 58 59 60 ACS Paragon Plus Environment Page 31 of 58 Journal of Proteome Research

1 2 3 4 The thiolbased labelling strategy is used more widely for labelling cysteines in claudins from both 5 6 human and other organisms.216,217 Reactive epitopes such as noncell permeable biotinylated NHS 7 8 esters were used to label HEK293 cells transfected with a mutant claudin5 expression vector in 9 10 212 11 order to quantify the plasma membrane localization level of the claudin5 mutant. Cysteine 12 13 mutants of claudin2 were generated in MDCK I cells for thiollabelling in order to determine the 14 15 protein surface accessibility of claudin2 in cells. 213 A thiol crosslinker was also successfully 16 17 utilized to identify, by MS after covalent labelling, the tetraspanin interacting partners in A431 and 18 19 211 20 A549 cells. Aminereactive labels have also shown the ability to label claudins in cells or

21 218,219 22 animals. Newborn mice with or without claudin1 were subcutaneously injected with an 23 24 aminereactive, biotinylation solution in order to assess the extent of barrier function loss in the 25 26 mice epidermis lacking claudin1. 218 Aminereactive biotinylation agents likewise showed labelling 27 28 of tight junction claudins in the mouse Eph4 cells junctions for permeability visualization upon 29 30 219 31 treatment of fluorescence labelledavidin. These precedents suggest that cell surface claudins can 32 33 be accessible to covalent chemical tagging. 34 35 36 37 Claudins have also been found in other cellular compartments such as endosomes that likely reflect 38 39 69,71 40 intracellular compartments active in tight junction turnover. specifically, surface claudins in 41 42 MDCK II cells have been differentiated from internal claudins by covalent surface biotinylation 43 44 labelling of the cell surface. This differential labelling in these cells was able to demonstrate in 45 46 MDCK cells that the endocytic recycling of claudin1 and 2 was affected in the presence of an 47 48 endocytosis inhibitor, resulting in the intracellular accumulation of these claudins, while other 49 50 215 51 claudins were not affected. Chemical tagging of claudins with spatial resolution may be desirable 52 53 for tracing claudin dynamics in the cell but requires further development of new chemical tags that 54 55 are specific for endogenous claudins. Such improved chemical tools, in conjunction with 56 57 subcellular fractionation and highfidelity methods from advanced proteomics technology, will help 58 59 60 ACS Paragon Plus Environment Journal of Proteome Research Page 32 of 58

1 2 characterize chemically claudin protein forms as potentially the combinatorial protein codes that 3 4 coordinate cell contact signalling and regulate claudin turnover to control tissue identity and 5 6 integrity. 75 7 8 9 10 11 6. Toward molecular identification of claudin-mediated cell-contact signaling platforms 12 13 14 15 Multicellular organisms rely on coordinated cellular processes to maintain tissue stability and 16 17 coordinate cell physiology. Cellcell contact sites represent a critical structure that enables 18 19 20 intercellular communication by organizing the formation of large multiprotein complexes (Figure 21 22 4). For example, intercellular junctions act as sensors for cell contact that regulate signaling by 23 24 forming protein complexes that activate kinases and also regulate by coordinating 25 26 the transit of transcription factors between the plasma membrane and nucleus.220,221,222,15,44,223 27 28 Relevant to claudins, the relatively unstructured intracellular Cterminal region contains several 29 30 31 putative and identified binding motifs that promote proteinprotein interactions, most prominently 32 1 33 the PDZ binding motif . Loss of the Cterminal tail leads to intracellular retention of claudins in the 34 35 ER and proteasome degradation, possibly by improper protein folding or lack of binding to 36 37 trafficking partners. 224226 These serial truncation experiments, however, demonstrate that the PDZ 38 39 40 binding motif is not required for proper claudin trafficking to the plasma membrane and reveal the 41 42 critical role of the juxtamembrane region of about 20 amino acids in localization control of 43 44 claudins. 45 46 47 48 Figure 4. Simplified scheme of the epithelial apical junctional complex, showing a subset of protein 49 50 51 constituents, including tetraspan transmembrane proteins (claudins, occludin), single pass 52 53 transmembrane proteins (JAMA, cadherin), cytosolic scaffold proteins (ZO1, ZO2, catenins), 54 55 signaling proteins (RhoA, MLCK) and the actin cytoskeleton. The Crumbs/PALS/PATJ polarity 56 57 58 59 60 ACS Paragon Plus Environment Page 33 of 58 Journal of Proteome Research

1 2 complex is also shown, which defines the apical/basolateral axis and is directly associated with tight 3 4 junctions via scaffold protein interactions. 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 Many excellent reviews and articles have already emphasized that specific claudins, localized to 34 35 36 various subcellular locations, interact with many other proteins to form specific scaffolds for 37 8,14,46,47,86,227,228 38 signaling complexes in different cell types of various organisms. Some recent 39 40 examples in human cells are highlighted here, while many other related examples are also known in 41 42 other mammals such as rats and mice,132,229231 along with recent proteomic analyses of MDCKII 43 44 45 cells that significantly expand the list of known and new signaling proteins assembled in and around

46 149 47 the tight junction as discussed earlier. 48 49 50 51 HEK293 cells are commonly used as nonpolarised, tight junctionfree cell models for cellcontact 52 53 investigations after transfection with claudins. 232 In transfected HEK293 cells, classic claudins 54 55 56 such as claudin1, 3 and 5 are found mainly at the cellcell contact points and provide multilateral 57 58 interaction with other tight junction proteins such as occludin, tricellulin, and MarvelD3 in order to 59 60 ACS Paragon Plus Environment Journal of Proteome Research Page 34 of 58

1 80,233235 2 secure the TJ strand network for stable barrier function. The dynamic aspect of claudin 3 4 localization in the cell membrane is also demonstrated for claudin2 and 4 in SKCO 15 or Caco2 5 6 BBE (Brush Border Expressing) cells in response to environmental changes such as cytokine 7 8 exposure. 236 This is consistent with the existing notion that claudins can mediate junction 9 10 11 remodeling and recruit other TJ proteins. 12 13 14 15 Claudin expression and dynamics are also known to be regulated by epithelial 16 17 molecule (EpCAM) in epithelial cells.237 Complexes between claudin7 and EpCAM have been 18 19 20 demonstrated by coimmunoprecipitation experiments and shown to recruit proteins into tetraspanin

21 238 22 enriched membrane microdomains. In HEK293 cells expressing EpCAM, the proliferative 23 24 activities of EpCAM require formation of a complex with claudin7 in order to disrupt EpCAM 25 26 oligomerization and activate mitogenic signaling. Mice injected with HEKEpCAMclaudin7 cells 27 28 developed tumors while HEK cells without the EpCAMclaudin7 complex did not induce tumors 29 30 239 31 in the animal. In a transgenic mice model expressing human claudin1, Notch signaling, a 32 33 critical regulator of intestinal epithelial cell differentiation and lineage determination, is up 34 35 regulated by claudin1 overexpression with concurrent upregulation of MMP9 activity and pERK 36 37 signaling.240 In MDAMB231 cells, claudin5 overexpression increases cell motility and co 38 39 40 immunoprecipitates with NWASP and ROCK1, suggesting a possible role of claudin5 in 41 241 42 metastasis. In HEK293 cells, claudin2 expression is regulated by components in Wnt signaling 43 44 whereby both LEF1 and βcatenin expressions enhanced the promoter activity of claudin2.242 In 45 46 human colonic cancer cell line SW480, claudin1 is also identified as a downstream target of 47 48 Wnt/βcatenin signaling, and increased claudin1 expression is observed in all 16 primary colorectal 49 50 51 cancers investigated with significant claudin1 localization detected in cellcell boundaries and the 52 243 53 cytoplasm. 54 55 56 57 58 59 60 ACS Paragon Plus Environment Page 35 of 58 Journal of Proteome Research

1 2 The nonjunctional localization of some claudins has also been linked to disease progression in 3 4 various epithelial cancers, which has been extensively reviewed.7477,89,244246 For example, in 5 6 craniopharyngioma, claudin1 expression is reduced in invasive tumors compared to noninvasive 7 8 ones, and increased cytoplasmic claudin1 localization is detected by immunostaining in tumor cells 9 10 247 11 that border brain tissues and dura. In invasive breast cancer, claudin1 is frequently down 12 248 13 regulated but upregulated in some aggressive subtypes of basallike breast cancer. Higher 14 15 claudin1 protein expression is observed in tumors from older patients, accompanied by more 16 17 frequent cytoplasmic claudin1 localization.249 In melanoma tissue samples, both cytoplasmic and 18 19 20 nuclear claudin1 expression is frequently observed, and nuclear claudin1 expression is drastically

21 250 22 reduced in lymph node metastases. In vitro models demonstrate a positive link between higher 23 24 MMP2 activity and higher cytoplasmic claudin1 expression level, suggesting a role of claudin1 25 26 in melanoma progression. This is not restricted to claudin1, since in proliferating human lung 27 28 adenocarcinoma A549 cells or tissues, claudin2 is found in the nucleus as part of a complex 29 30 73 31 containing ZO1, ZONAB, and cyclin D1. Compared to the wild type, a S208A mutant of claudin 32 33 2 exhibits a higher extent of nuclear localization in its dephosphorylated form, suggesting that 34 35 nuclear claudin2 may serve to retain ZONAB and cyclin D1 in the nucleus to enhance cell 36 37 proliferation. Such proliferation promoting effect is also suggested for claudin1 in osteoblasts. 251 38 39 40 Likewise, both claudin7 transcript and protein expression levels are frequently elevated in 41 42 epithelial ovarian tumors but not in normal ovarian tissues, and in many ovarian cancer cell lines 43 44 both cytoplasmic and cell membrane claudin7 expression are detected by immunostaining. 252 45 46 However, the mechanisms and molecular details behind the change of claudin localization remain 47 48 unclear, particularly when localized to the cytosol or nucleus. 49 50 51 52 53 The diverse localization and binding partners identified using standard biochemical techniques 54 55 suggest that regulation of claudin function is complex. The acquisition of the exact chemical 56 57 characteristics of the specific claudin protein forms, from different cell locations by MS, in addition 58 59 60 ACS Paragon Plus Environment Journal of Proteome Research Page 36 of 58

1 2 to their levels of expression, may help elucidate the details of the signaling context specific to 3 4 cell/tissue types or disease states. Initial attempts at identifying protein partners of claudins are 5 6 listed in Table 5. Those interactions were retrieved from Swissprot and IntAct databases. Reflecting 7 8 the most prominent function of claudins, namely formation of epithelial barriers via tight junctions, 9 10 11 claudinclaudin and claudinscaffold protein interactions are well represented in Table 5. This list 12 13 will certainly be expanded and refined as more claudin specific proteinprotein interactions are 14 15 characterized from an expanded range of host tissues. 16 17 18 19 20 Table 5. Proteinprotein interactions with claudins (compiled from www.swissprot.org and 21 http://www.ebi.ac.uk/intact/ ). 22 23 24 Protein/Protein Interaction Partners Binary Interactions (IntAct annotation; in bold Gene/Protein 25 (UniProtKB/Swiss-Prot manual annotation) when found by two independent experiments) 26 CLDN1 a CLDN3 (but not CLDN2); TJPZO*; MPDZ; BRD4; DRG1; TACSTD2 c 27 INADL; HCV E1/E2 a 28 CLDN2 CLDN3 (but not CLDN1); TJPZO*; KRTAP412; KRT31 ; NOTCH2NL ; TJP1 29 CLDN3 a CLDN1 and CLDN2; TJP ZO* ; 30 CLDN4 TJPZO*; EPHA2 TACSTD2 31 CLDN5 TJP ZO* ; MPDZ 32 33 CLDN6 TJPZO*; b 34 CLDN7 TJPZO*;; EPCAM HGD; RHOXF2; SYNE4; TACSTD2 35 CLDN8 TJP ZO* ; CCDC155; SYNE4 36 CLDN11 TSPAN3 37 CLDN12 ECHS1; SEC13; STRN4 38 39 CLDN15 CLDN15 (linear homooligomers) GEM d 40 CLDN16 CLDN19; Cldn14 41 CLDN19 CLDN16; SRPK2; Cldn14 d 42 aCan form homo and heteropolymers with other claudins. 43 bInteraction requires claudin phosphorylation. 44 cDirect interaction with HCV proteins has not been experimentally validated (see text). 45 dExperiment done with mouse proteins. 46 *TJP1/ZO1; TJP2/ZO2; TJP3/ZO3; these protein partners are common to claudins 1 to 8. 47 48 49 The range of protein partnership for claudins can be illustrated by a protein interaction network 50 51 diagram using human claudin1 as an example (Figure 5). Direct protein interactions with human 52 53 54 claudin1 were obtained by careful mining of the literature. Extended human protein interactions 55 56 between protein partners of claudin1 and other proteins were retrieved from UniProtKB/SwissProt 57 58 and from IntAct (only interactions found by more than one experiment). The domainspecific 59 60 ACS Paragon Plus Environment Page 37 of 58 Journal of Proteome Research

1 2 annotations of these protein interactions with claudin1 are listed in Table 6. As not all the 3 4 interactions described in literature are yet imported in UniProtKB/SwissProt and Intact, this 5 6 interaction network may be more complex. 7 8 9 10 11 Figure 5. A diagram of the protein interaction network of claudin1. The diagram was made using 12 253 13 Cytoscape. In orange are the intrinsic membrane proteins with at least one transmembrane 14 15 domain. In yellow are the peripheral membrane proteins. These two groups contain plasma 16 17 membrane proteins and endomembrane system proteins (endosome, endoplasmic reticulum, Golgi 18 19 20 apparatus, and cytoplasmic vesicles). Membrane proteins from mitochondria or nucleus are not 21 22 included in these two groups. In green are cytosolic, nuclear and mitochondrial proteins. In blue are 23 24 secreted proteins (with a signal peptide and no transmembrane domains). The pink border indicates 25 26 proteins found in tight junctions. Red edges connect CLDN1 (diamond) with its direct interacting 27 28 partners: TJP1/ZO1, TJP2/ZO2, TJP3/ZO3, MPDZ, INADL, TACSTD2, SRC, KRT76, LNX1, 29 30 31 CD81, CD9, OCLN, MARVELD2, MARVELD3, CLDN3, CLDN7, MMP14, MMP2, EFNB1 32 33 (Rectangles). Interactions with nonhuman proteins are not included in the network but briefly 34 35 described in the text. 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 ACS Paragon Plus Environment Journal of Proteome Research Page 38 of 58

1 2 3 4 Table 6. Domainspecific annotations of protein interactions with claudin1. 5 6 Protein/Protein Claudin 1-domains 7 Interaction Partners 8 Cytosolic tail TJP1 /ZO 1254 9 TJP2/ZO2254 10 TJP3/ZO3254 11 255 12 MPDZ/MUPP1 256 13 INADL/PATJ 14 TACSTD2 257 15 SRC 258 16 KRT76 259 17 LNX1 125,260

18 261 cis (transmembrane) CD81 19 CD9 211 20 262 21 OCL 263 22 MARVELD2 23 MARVELD3 80 24 CLDN3 233,264 25 CLDN1 265 26 CLDN7 237 27 trans CLDN1 265 28 junction/extracellular 264 29 CLDN3 266 30 MMP14 31 MMP2 266 32 EFNB1 267

33 34 35 While the human claudin1 diagram does not include its nonhuman protein partners, pathogenic 36 37 38 proteins have been found to interact with human claudin1. For example, human claudin1 along 39 40 with occludin and the tetraspanin receptor CD81, mediate the initial step of hepatitis C virus (HCV) 41 42 infection.268 During this process CD81 directly interacts with the envelope glycoproteins HCVE1 43 44 and HCVE2.269 The direct physical interaction between claudin1 and the HCVE1/2 glycoproteins 45 46 has not been definitively established but inferred by the ability of the particles expressing the HCV 47 48 270,271 49 glycoproteins to bind claudin1expressing cells. A direct interaction between claudin1 and 50 51 the premembrane (prM) protein from the dengue virus has been suggested to be important for 52 53 dengue virus entry.272 Interestingly, some of the protein partners of human claudin1 were also 54 55 shown to interact with viral proteins: MPDZ with the adenovirus type 9 E4 protein;273 LNX1 with 56 57 274 275 58 the endogenous retrovirus K protein Np9; and SRC with the hepatitis E virus ORF3 protein. 59 60 ACS Paragon Plus Environment Page 39 of 58 Journal of Proteome Research

1 2 Human claudin1 is also involved in cell infection by nonviral pathogens. A recent study suggests 3 4 that the intestinal epithelial damage caused by the protozoan Entamoeba histolytica is initiated by 5 6 the interaction of the virulence complex EhCPADH112 (formed by CP112/EhCP112 and 7 8 ADH112/EhADH112) with claudin1, occludin, TJP1/ZO1 and TJP2/ZO2 followed by their 9 10 276 11 degradation. 12 13 14 15 In addition to standard biophysical techniques, MS is a particularly useful technique for acquiring 16 17 complementary information on stoichiometry and topology of protein complexes.277283 Although 18 19 20 lower in resolution as compared with techniques with atomic resolution, MSbased information is 21 22 crucial to understanding dynamic protein interactions and networks that govern biological 23 24 function. 280 Integrated approaches that combine PTM analysis by MSbased proteomics, protein 25 26 crossinglinking, protein complexes characterization by MS, and structural modeling may offer new 27 28 structural understanding of endogenous protein partnerships or interactomes. 278,279,282 Examples of 29 30 31 complex multicomponent systems that have been successfully analyzed by MScoupled 32 33 quantitative proteomics include large protein assemblies such as the human initiation factor (eIF)3 34 35 complex and the yeast RNA exosome. Recent advances in ionization and dissociation methods, 36 37 including surfaceinduced collision and ionmobility MS procedures, also greatly improve the 38 39 277,281 40 prospect of mass spectrometry in characterizing intact protein complexes in their native states. 41 42 These new technological capacities, in conjunction with enrichment techniques, may begin to help 43 44 shed light on the supramolecular structures of claudin protein complexes that define the signaling 45 46 context at and beyond cell junctions. 47 48 49 50 51 Conclusion 52 53 The predominant role for claudins in regulating epithelial and endothelial barrier function 54 55 necessitates that they need to be able to interact with a multiplicity of proteins in diverse ways, 56 57 including trans (intercellular) interactions with extracellular domains of proteins across intercellular 58 59 60 ACS Paragon Plus Environment Journal of Proteome Research Page 40 of 58

1 2 junctions, cis (in the plane of the plasma membrane) interactions with transmembrane proteins and 3 4 binding to scaffold proteins that regulate their organization and stability. 284 Thus, claudins have 5 6 two roles in regulating tight junction permeability, a direct role by forming different classes of 7 8 paracellular channels and a more indirect role by participating in the formation of the large multi 9 10 11 protein complexes associated with claudincontaining tight junction strands. Although considerable 12 13 progress has been made to understand how claudin diversity affects paracellular channel selectivity, 14 15 precisely defining claudin binding partners at a quantitative level is still at an early stage of 16 17 research. The ensemble of proteins linked to claudins in tight junctions affects paracellular flux 18 19 20 through control of strand morphology, localization and stability. Understanding how protein 21 22 cofactors mediate intercellular interactions, crosslinking of claudins with the actin cytoskeleton 23 24 and integration with signal transduction pathways will provide insights into how tissue barrier 25 26 function is regulated in normal and diseased organs, as well as potential therapeutic targets for the 27 28 manipulation of paracellular flux. 29 30 31 32 33 The regulatory role conferred by the claudin family of proteins in tight junctions is complemented 34 35 by roles in morphogenesis, tissue maintenance, growth control, and cell migration. All of these 36 37 processes require constant protein processing and modification. Individual claudin members have 38 39 40 been investigated biochemically or in cell models/tissues for their expression and localization 41 42 profile, and some claudin members have been characterized by MS in addition to antibodybased 43 44 detection. Many claudin PTMs, such as Cterminal phosphorylation and palmitoylation have been 45 46 identified. There are no doubt many more claudin phosphorylation sites remaining to be identified 47 48 and future studies are needed to provide added evidence in support of PTMs such as ubiquitination, 49 50 51 SUMOylation, palmitoylation, and glycosylation. 52 53 54 55 More importantly, it remains an enormous challenge to acquire a direct and accurate profile of 56 57 claudins, PTMs and interacting partners, considering that claudins are lowabundance, membrane 58 59 60 ACS Paragon Plus Environment Page 41 of 58 Journal of Proteome Research

1 2 bound, and heavily modified transmembrane proteins. While the function of tight junction 3 4 associated claudins is largely to regulate the paracellular barrier, nonjunctional claudins in the 5 6 lateral plasma membrane, cytosol or nucleus likely have distinct roles in cell regulation. 7 8 Complementary to antibodybased detection that will continue to be a major method of 9 10 11 investigation, advanced chemical capture, enrichment, and analysis tools, tailored for highprecision 12 13 MSbased proteomic profiling in a multipronged and integrated manner, could provide unbiased 14 15 approaches to identify claudin PTM and interacting proteins. Defining claudin PTMs and 16 17 interacting proteins in a quantitative and stoichiometric manner has the potential to determine 18 19 20 whether the spectrum of claudin expression not only controls paracellular flux but also integrates 21 22 the flow of information by acting in effect as a cell “identity code” that is recognized across 23 24 intercellular junctions. Understanding this context in which claudins affect cells could enable a 25 26 nuanced approach to therapeutic design by either modulating claudin barrier function (e.g. for the 27 28 purposes of drug delivery) or affecting other nonbarrier cell functions regulated by claudins, such 29 30 31 as cell proliferation and migration. By defining the different molecular contexts of claudin 32 33 expression, systems proteomics is a valuable approach that will make it possible to consider 34 35 specifically targeting junctional vs. nonjunctional roles for claudins. 36 37 38 39 40 41 42 Acknowledgment 43 44 We thank the Australian Research Council (LIEF150100161 to F. L. and S. R.), the National 45 46 Institute of Health (R01HL116958 to M. K.), the National Health and Medical Research Council of 47 48 Australia (NHMRC APP1010303 to M. B.), and the New South Wale Cancer Council (RG1004 & 49 50 51 RG0816 to M. B.) for financial support. 52 53 54 55 56 57 58 59 60 ACS Paragon Plus Environment Journal of Proteome Research Page 42 of 58

1 2 3 4 1 Gunzel, D. & Yu, A. S. Claudins and the modulation of tight junction permeability. Physiol 5 Rev 93 , 525569, doi:10.1152/physrev.00019.2012 (2013). 6 2 Schneeberger, E. E. & Lynch, R. D. The tight junction: a multifunctional complex. Am J 7 Physiol Cell Physiol 286 , C12131228, doi:10.1152/ajpcell.00558.2003 (2004). 8 3 Tsukita, S., Furuse, M. & Itoh, M. Structural and signalling molecules come together at tight 9 10 junctions. Curr Opin Cell Biol 11 , 628633 (1999). 11 4 Furuse, M. Molecular Basis of the Core Structure of Tight Junctions. Cold Spring Harbor 12 Perspectives in Biology 2, a002907a002907, doi:10.1101/cshperspect.a002907 (2009). 13 5 Tsukita, S. & Furuse, M. The structure and function of claudins, cell adhesion molecules at 14 tight junctions. Ann N Y Acad Sci 915 , 129135 (2000). 15 6 Tsukita, S., Furuse, M. & Itoh, M. Multifunctional strands in tight junctions. Nat Rev Mol 16 Cell Biol 2, 285293, doi:10.1038/35067088 (2001). 17 7 Van Itallie, C. M. & Anderson, J. M. Claudins and Epithelial Paracellular Transport. Annual 18 Review of Physiology 68 , 403429, doi:10.1146/annurev.physiol.68.040104.131404 (2006). 19 20 8 Koval, M. Claudinskey pieces in the tight junction puzzle. Cell communication & 21 adhesion 13 , 127138, doi:10.1080/15419060600726209 (2006). 22 9 Van Itallie, C. M. & Anderson, J. M. Architecture of tight junctions and principles of 23 molecular composition. Seminars in cell & developmental biology 36 , 157165, 24 doi:10.1016/j.semcdb.2014.08.011 (2014). 25 10 Gunzel, D. & Fromm, M. Claudins and other tight junction proteins. Comprehensive 26 Physiology 2, 18191852, doi:10.1002/cphy.c110045 (2012). 27 11 Van Itallie, C. M. & Anderson, J. M. The role of claudins in determining paracellular charge 28 selectivity. Proceedings of the American Thoracic Society 1, 3841, 29 30 doi:10.1513/pats.2306013 (2004). 31 12 Matter, K. & Balda, M. S. Signalling to and from tight junctions. Nat Rev Mol Cell Biol 4, 32 225236, doi:10.1038/nrm1055 (2003). 33 13 Findley, M. K. & Koval, M. Regulation and roles for claudinfamily tight junction proteins. 34 IUBMB Life 61 , 431437, doi:10.1002/iub.175 (2009). 35 14 Van Itallie, C. M. & Anderson, J. M. Claudin interactions in and out of the tight junction. 36 Tissue Barriers 1, e25247, doi:10.4161/tisb.25247 (2013). 37 15 Zihni, C., Balda, M. S. & Matter, K. Signalling at tight junctions during epithelial 38 differentiation and microbial pathogenesis. J Cell Sci 127 , 34013413, 39 40 doi:10.1242/jcs.145029 (2014). 41 16 Farquhar, M. G. & Palade, G. E. Junctional complexes in various epithelia. J Cell Biol 17 , 42 375412 (1963). 43 17 Furuse, M., Fujita, K., Hiiragi, T., Fujimoto, K. & Tsukita, S. Claudin1 and 2: novel 44 integral membrane proteins localizing at tight junctions with no sequence similarity to 45 occludin. J Cell Biol 141 , 15391550 (1998). 46 18 Loh, Y. H., Christoffels, A., Brenner, S., Hunziker, W. & Venkatesh, B. Extensive 47 expansion of the claudin gene family in the teleost fish, Fugu rubripes. Genome Res 14 , 48 12481257, doi:10.1101/gr.2400004 (2004). 49 50 19 LalNag, M. & Morin, P. J. The claudins. Genome Biol 10 , 235, doi:10.1186/gb2009108 51 235 (2009). 52 20 Angelow, S., Ahlstrom, R. & Yu, A. S. L. Biology of claudins. AJP: Renal Physiology 295 , 53 F867F876, doi:10.1152/ajprenal.90264.2008 (2008). 54 21 Anderson, J. M. & Van Itallie, C. M. Physiology and Function of the Tight Junction. Cold 55 Spring Harbor Perspectives in Biology 1, a002584a002584, 56 doi:10.1101/cshperspect.a002584 (2009). 57 22 Tamura, A. & Tsukita, S. Paracellular barrier and channel functions of TJ claudins in 58 organizing biological systems: advances in the field of barriology revealed in knockout 59 60 ACS Paragon Plus Environment Page 43 of 58 Journal of Proteome Research

1 2 mice. Seminars in cell & developmental biology 36 , 177185, 3 doi:10.1016/j.semcdb.2014.09.019 (2014). 4 23 Troy, T. C. & Turksen, K. The targeted overexpression of a Claudin mutant in the epidermis 5 of transgenic mice elicits striking epidermal and hair follicle abnormalities. Molecular 6 biotechnology 36 , 166174 (2007). 7 24 Pope, J. L. et al. Claudin1 overexpression in intestinal epithelial cells enhances 8 susceptibility to adenamatous polyposis colimediated colon tumorigenesis. Molecular 9 10 cancer 13 , 167, doi:10.1186/1476459813167 (2014). 11 25 Daugherty, B. L. et al. Developmental regulation of claudin localization by fetal alveolar 12 epithelial cells. American journal of physiology. Lung cellular and molecular physiology 13 287 , L12661273, doi:10.1152/ajplung.00423.2003 (2004). 14 26 Shen, L., Weber, C. R. & Turner, J. R. The tight junction protein complex undergoes rapid 15 and continuous molecular remodeling at steady state. J Cell Biol 181 , 683695, 16 doi:10.1083/jcb.200711165 (2008). 17 27 Camire, R. B. et al. Biphasic modulation of paracellular claudin5 expression in mouse brain 18 endothelial cells is mediated through the phosphoinositide3kinase/AKT pathway. J 19 20 Pharmacol Exp Ther 351 , 654662, doi:10.1124/jpet.114.218339 (2014). 21 28 GarciaHernandez, V. et al. EGF regulates claudin2 and 4 expression through Src and 22 STAT3 in MDCK cells. Journal of cellular physiology 230 , 105115, doi:10.1002/jcp.24687 23 (2015). 24 29 Twiss, F. et al. HGF signaling regulates Claudin3 dynamics through its Cterminal tyrosine 25 residues. Tissue Barriers 1, e27425, doi:10.4161/tisb.27425 (2013). 26 30 Van Itallie, C. M. et al. Phosphorylation of claudin2 on serine 208 promotes membrane 27 retention and reduces trafficking to lysosomes. Journal of Cell Science 125 , 49024912, 28 doi:10.1242/jcs.111237 (2012). 29 30 31 Wu, C. C. & Yates, J. R., 3rd. The application of mass spectrometry to membrane 31 proteomics. Nature biotechnology 21 , 262267, doi:10.1038/nbt0303262 (2003). 32 32 Tan, S., Tan, H. T. & Chung, M. C. Membrane proteins and membrane proteomics. 33 Proteomics 8, 39243932, doi:10.1002/pmic.200800597 (2008). 34 33 Deutsch, E. W., Lam, H. & Aebersold, R. PeptideAtlas: a resource for target selection for 35 emerging targeted proteomics workflows. EMBO reports 9, 429434, 36 doi:10.1038/embor.2008.56 (2008). 37 34 Surinova, S. et al. Automated selected reaction monitoring data analysis workflow for large 38 scale targeted proteomic studies. Nature protocols 8, 16021619, 39 40 doi:10.1038/nprot.2013.091 (2013). 41 35 Toprak, U. H. et al. Conserved peptide fragmentation as a benchmarking tool for mass 42 spectrometers and a discriminating feature for targeted proteomics. Mol Cell Proteomics , 43 doi:10.1074/mcp.O113.036475 (2014). 44 36 Valaskovic, G. A. & Kelleher, N. L. Miniaturized formats for efficient mass spectrometry 45 based proteomics and therapeutic development. Current topics in medicinal chemistry 2, 1 46 12 (2002). 47 37 Sali, A., Glaeser, R., Earnest, T. & Baumeister, W. From words to literature in structural 48 proteomics. Nature 422 , 216225, doi:10.1038/nature01513 (2003). 49 50 38 Heck, A. J. & Van Den Heuvel, R. H. Investigation of intact protein complexes by mass 51 spectrometry. Mass Spectrom Rev 23 , 368389, doi:10.1002/mas.10081 (2004). 52 39 Greenbaum, D. et al. Chemical approaches for functionally probing the proteome. Mol Cell 53 Proteomics 1, 6068 (2002). 54 40 Adam, G. C., Sorensen, E. J. & Cravatt, B. F. Chemical strategies for functional proteomics. 55 Mol Cell Proteomics 1, 781790 (2002). 56 41 Uchida, Y. et al. A study protocol for quantitative targeted absolute proteomics (QTAP) by 57 LCMS/MS: application for interstrain differences in protein expression levels of 58 transporters, receptors, claudin5, and marker proteins at the bloodbrain barrier in ddY, 59 60 ACS Paragon Plus Environment Journal of Proteome Research Page 44 of 58

1 2 FVB, and C57BL/6J mice. Fluids and barriers of the CNS 10 , 21, doi:10.1186/20458118 3 1021 (2013). 4 42 Koval, M. Differential pathways of claudin oligomerization and integration into tight 5 junctions. Tissue Barriers 1, e24518, doi:10.4161/tisb.24518 (2013). 6 43 Ivanov, A. I., Parkos, C. A. & Nusrat, A. Cytoskeletal regulation of epithelial barrier 7 function during inflammation. The American journal of pathology 177 , 512524, 8 doi:10.2353/ajpath.2010.100168 (2010). 9 10 44 McCrea, P. D., Gu, D. & Balda, M. S. Junctional music that the nucleus hears: cellcell 11 contact signaling and the modulation of gene activity. Cold Spring Harb Perspect Biol 1, 12 a002923, doi:10.1101/cshperspect.a002923 (2009). 13 45 Matter, K. & Balda, M. S. SnapShot: Epithelial tight junctions. Cell 157 , 992992 e991, 14 doi:10.1016/j.cell.2014.04.027 (2014). 15 46 GonzalezMariscal, L., Tapia, R. & Chamorro, D. Crosstalk of tight junction components 16 with signaling pathways. Biochimica et biophysica acta 1778 , 729756, 17 doi:10.1016/j.bbamem.2007.08.018 (2008). 18 47 Balda, M. S. & Matter, K. Tight junctions and the regulation of gene expression. Biochimica 19 20 et biophysica acta 1788 , 761767, doi:10.1016/j.bbamem.2008.11.024 (2009). 21 48 Capaldo, C. T. & Nusrat, A. Claudin switching: Physiological plasticity of the Tight 22 Junction. Seminars in cell & developmental biology 42 , 2229, 23 doi:10.1016/j.semcdb.2015.04.003 (2015). 24 49 UniProt: a hub for protein information. Nucleic acids research 43 , D204212, 25 doi:10.1093/nar/gku989 (2015). 26 50 Cunningham, F. et al. Ensembl 2015. Nucleic acids research 43 , D662669, 27 doi:10.1093/nar/gku1010 (2015). 28 51 Uhlen, M. et al. Towards a knowledgebased Human Protein Atlas. Nature biotechnology 29 30 28 , 12481250, doi:10.1038/nbt12101248 (2010). 31 52 Morita, K., Furuse, M., Fujimoto, K. & Tsukita, S. Claudin multigene family encoding four 32 transmembrane domain protein components of tight junction strands. Proc Natl Acad Sci U 33 S A 96 , 511516 (1999). 34 53 Mineta, K. et al. Predicted expansion of the claudin multigene family. FEBS Letters 585 , 35 606612, doi:10.1016/j.febslet.2011.01.028 (2011). 36 54 Baltzegar, D. A., Reading, B. J., Brune, E. S. & Borski, R. J. Phylogenetic revision of the 37 claudin gene family. Marine Genomics 11 , 1726, doi:10.1016/j.margen.2013.05.001 38 (2013). 39 40 55 Gray, K. A., Yates, B., Seal, R. L., Wright, M. W. & Bruford, E. A. Genenames.org: the 41 HGNC resources in 2015. Nucleic Acids Res 43 , D10791085, doi:10.1093/nar/gku1071 42 (2015). 43 56 Safran, M. et al. GeneCards Version 3: the human gene integrator. Database : the journal of 44 biological databases and curation 2010 , baq020, doi:10.1093/database/baq020 (2010). 45 57 Krause, G. et al. Structure and function of claudins. Biochimica et Biophysica Acta (BBA) - 46 Biomembranes 1778 , 631645, doi:10.1016/j.bbamem.2007.10.018 (2008). 47 58 Mortazavi, A., Williams, B. A., McCue, K., Schaeffer, L. & Wold, B. Mapping and 48 quantifying mammalian transcriptomes by RNASeq. Nat Methods 5, 621628, 49 50 doi:10.1038/nmeth.1226 (2008). 51 59 Kollmar, R., Nakamura, S. K., Kappler, J. A. & Hudspeth, A. J. Expression and phylogeny 52 of claudins in vertebrate primordia. Proceedings of the National Academy of Sciences 98 , 53 1019610201, doi:10.1073/pnas.171325898 (2001). 54 60 Sigrist, C. J. A. et al. PROSITE, a protein domain database for functional characterization 55 and annotation. Nucleic Acids Research 38 , D161D166, doi:10.1093/nar/gkp885 (2009). 56 61 Turksen, K. Barriers built on claudins. Journal of Cell Science 117 , 24352447, 57 doi:10.1242/jcs.01235 (2004). 58 59 60 ACS Paragon Plus Environment Page 45 of 58 Journal of Proteome Research

1 2 62 Harris, B. Z. & Lim, W. A. Mechanism and role of PDZ domains in signaling complex 3 assembly. J Cell Sci 114 , 32193231 (2001). 4 63 Nomme, J. et al. Structural Basis of a Key Factor Regulating the Affinity between the 5 Zonula Occludens First PDZ Domain and Claudins. J Biol Chem 290 , 1659516606, 6 doi:10.1074/jbc.M115.646695 (2015). 7 64 Suzuki, H. et al. Crystal structure of a claudin provides insight into the architecture of tight 8 junctions. Science 344 , 304307, doi:10.1126/science.1248571 (2014). 9 10 65 Saitoh, Y. et al. Tight junctions. Structural insight into tight junction disassembly by 11 Clostridium perfringens enterotoxin. Science 347 , 775778, doi:10.1126/science.1261833 12 (2015). 13 66 Horvatovich, P. et al. Quest for Missing Proteins: Update 2015 on ChromosomeCentric 14 Human Proteome Project. Journal of proteome research , doi:10.1021/pr5013009 (2015). 15 67 Uhlen, M. et al. Antibodybased protein profiling of the human . Mol Cell 16 Proteomics 11 , M111 013458, doi:10.1074/mcp.M111.013458 (2012). 17 68 Zheng, A. et al. Claudin6 and claudin9 function as additional coreceptors for hepatitis C 18 virus. J Virol 81 , 1246512471, doi:10.1128/jvi.0145707 (2007). 19 20 69 Hewitt, K. J., Agarwal, R. & Morin, P. J. The claudin gene family: expression in normal and 21 neoplastic tissues. BMC Cancer 6, 186, doi:10.1186/147124076186 (2006). 22 70 Yoshida, Y., Ban, Y. & Kinoshita, S. Tight junction transmembrane protein claudin subtype 23 expression and distribution in human corneal and conjunctival . Investigative 24 ophthalmology & visual science 50 , 21032108, doi:10.1167/iovs.083046 (2009). 25 71 Dhawan, P. et al. Claudin1 regulates cellular transformation and metastatic behavior in 26 colon cancer. J Clin Invest 115 , 17651776, doi:10.1172/jci24543 (2005). 27 72 French, A. D. et al. PKC and PKA phosphorylation affect the subcellular localization of 28 claudin1 in melanoma cells. Int J Med Sci 6, 93101 (2009). 29 30 73 Ikari, A. et al. Nuclear distribution of claudin2 increases cell proliferation in human lung 31 adenocarcinoma cells. Biochimica et biophysica acta 1843 , 20792088, 32 doi:10.1016/j.bbamcr.2014.05.017 (2014). 33 74 Morin, P. J. Claudin proteins in human cancer: promising new targets for diagnosis and 34 therapy. Cancer Res 65 , 96039606, doi:10.1158/00085472.can052782 (2005). 35 75 Gupta, I. R. & Ryan, A. K. Claudins: unlocking the code to tight junction function during 36 embryogenesis and in disease. Clinical Genetics 77 , 314325, doi:10.1111/j.1399 37 0004.2010.01397.x (2010). 38 76 Kwon, M. J. Emerging roles of claudins in human cancer. Int J Mol Sci 14 , 1814818180, 39 40 doi:10.3390/ijms140918148 (2013). 41 77 Singh, A. B., Sharma, A. & Dhawan, P. Claudin family of proteins and cancer: an overview. 42 J Oncol 2010 , 541957, doi:10.1155/2010/541957 (2010). 43 78 Singh, A. B. & Dhawan, P. Claudins and cancer: Fall of the soldiers entrusted to protect the 44 gate and keep the barrier intact. Seminars in cell & developmental biology , 45 doi:10.1016/j.semcdb.2015.05.001 (2015). 46 79 Anderson, J. M., Van Itallie, C. M. & Fanning, A. S. Setting up a selective barrier at the 47 apical junction complex. Curr Opin Cell Biol 16 , 140145, doi:10.1016/j.ceb.2004.01.005 48 (2004). 49 50 80 Cording, J. et al. In tight junctions, claudins regulate the interactions between occludin, 51 tricellulin and marvelD3, which, inversely, modulate claudin oligomerization. J Cell Sci 52 126 , 554564, doi:10.1242/jcs.114306 (2013). 53 81 Krug, S. M., Schulzke, J. D. & Fromm, M. Tight junction, selective permeability, and 54 related diseases. Seminars in cell & developmental biology 36 , 166176, 55 doi:10.1016/j.semcdb.2014.09.002 (2014). 56 82 Hou, J., Rajagopal, M. & Yu, A. S. Claudins and the kidney. Annu Rev Physiol 75 , 479501, 57 doi:10.1146/annurevphysiol030212183705 (2013). 58 59 60 ACS Paragon Plus Environment Journal of Proteome Research Page 46 of 58

1 2 83 Konrad, M. et al. CLDN16 genotype predicts renal decline in familial hypomagnesemia 3 with hypercalciuria and nephrocalcinosis. Journal of the American Society of Nephrology : 4 JASN 19 , 171181, doi:10.1681/asn.2007060709 (2008). 5 84 Konrad, M. et al. Mutations in the tightjunction gene claudin 19 (CLDN19) are associated 6 with renal magnesium wasting, renal failure, and severe ocular involvement. American 7 journal of human genetics 79 , 949957, doi:10.1086/508617 (2006). 8 85 Simon, D. B. et al. Paracellin1, a renal tight junction protein required for paracellular 9 10 Mg2+ resorption. Science 285 , 103106 (1999). 11 86 GonzálezMariscal, L., Garay, E. & Quirós, M. Regulation of Claudins by Posttranslational 12 Modifications and CellSignaling Cascades. 65 , 113150, doi:10.1016/s1063 13 5823(10)650065 (2010). 14 87 Tanaka, M. EphA2 Phosphorylates the Cytoplasmic Tail of Claudin4 and Mediates 15 Paracellular Permeability. Journal of Biological Chemistry 280, 4237542382, 16 doi:10.1074/jbc.M503786200 (2005). 17 88 Shen, W. et al. Tyrosine phosphorylation of VEcadherin and claudin5 is associated with 18 TGFbeta1induced permeability of centrally derived vascular endothelium. Eur J Cell Biol 19 20 90 , 323332, doi:10.1016/j.ejcb.2010.10.013 (2011). 21 89 D'Souza, T., Agarwal, R. & Morin, P. J. Phosphorylation of claudin3 at threonine 192 by 22 cAMPdependent protein kinase regulates tight junction barrier function in ovarian cancer 23 cells. J Biol Chem 280 , 2623326240, doi:10.1074/jbc.M502003200 (2005). 24 90 Hoggard, J. et al. Claudin7 increases chemosensitivity to cisplatin through the upregulation 25 of caspase pathway in human NCIH522 lung cancer cells. Cancer Sci 104 , 611618, 26 doi:10.1111/cas.12135 (2013). 27 91 Tatum, R. et al. WNK4 phosphorylates ser(206) of claudin7 and promotes paracellular Cl( 28 ) permeability. FEBS Lett 581 , 38873891, doi:10.1016/j.febslet.2007.07.014 (2007). 29 30 92 Hornbeck, P. V. et al. PhosphoSitePlus, 2014: mutations, PTMs and recalibrations. Nucleic 31 acids research 43 , D512520, doi:10.1093/nar/gku1267 (2015). 32 93 Fenyo, D., Eriksson, J. & Beavis, R. Mass spectrometric protein identification using the 33 global proteome machine. Methods in molecular biology (Clifton, N.J.) 673 , 189202, 34 doi:10.1007/9781607618423_11 (2010). 35 94 Mayya, V. et al. Quantitative phosphoproteomic analysis of T cell receptor signaling reveals 36 systemwide modulation of proteinprotein interactions. Sci Signal 2, ra46, 37 doi:10.1126/scisignal.2000007 (2009). 38 95 Rigbolt, K. T. et al. Systemwide temporal characterization of the proteome and 39 40 phosphoproteome of human embryonic stem cell differentiation. Sci Signal 4, rs3, 41 doi:10.1126/scisignal.2001570 (2011). 42 96 Bai, Y. et al. Phosphoproteomics identifies driver tyrosine kinases in sarcoma cell lines and 43 tumors. Cancer Res 72 , 25012511, doi:10.1158/00085472.can113015 (2012). 44 97 Shiromizu, T. et al. Identification of missing proteins in the neXtProt database and 45 unregistered phosphopeptides in the PhosphoSitePlus database as part of the Chromosome 46 centric Human Proteome Project. Journal of proteome research 12 , 24142421, 47 doi:papers2://publication/doi/10.1021/pr300825v (2013). 48 98 Kim, J.Y. et al. Dissection of TBK1 signaling via phosphoproteomics in lung cancer cells. 49 50 PNAS 110 , 1241412419, doi:papers2://publication/doi/10.1073/pnas.1220674110 (2013). 51 99 Phanstiel, D. H. et al. Proteomic and phosphoproteomic comparison of human ES and iPS 52 cells. Nature Methods 8, 821827, doi:papers2://publication/doi/10.1038/nmeth.1699 53 (2011). 54 100 Gu, T.L. et al. Survey of tyrosine kinase signaling reveals ROS kinase fusions in human 55 cholangiocarcinoma. PLoS ONE 6, e15640, 56 doi:papers2://publication/doi/10.1371/journal.pone.0015640 (2011). 57 58 59 60 ACS Paragon Plus Environment Page 47 of 58 Journal of Proteome Research

1 2 101 Ding, V. M. Y. et al. Tyrosine phosphorylation profiling in FGF2 stimulated human 3 embryonic stem cells. PLoS ONE 6, e17538, 4 doi:papers2://publication/doi/10.1371/journal.pone.0017538 (2011). 5 102 Moritz, A. et al. AktRSKS6 kinase signaling networks activated by oncogenic receptor 6 tyrosine kinases. Science Signaling 3, ra64, 7 doi:papers2://publication/doi/10.1126/scisignal.2000998 (2010). 8 103 Han, G. et al. Phosphoproteome analysis of human liver tissue by longgradient nanoflow 9 10 LC coupled with multiple stage MS analysis. Electrophoresis 31 , 10801089, 11 doi:papers2://publication/doi/10.1002/elps.200900493 (2010). 12 104 Van Hoof, D. et al. Phosphorylation dynamics during early differentiation of human 13 embryonic stem cells. Cell Stem Cell 5, 214226, 14 doi:papers2://publication/doi/10.1016/j.stem.2009.05.021 (2009). 15 105 Guo, A. et al. Signaling networks assembled by oncogenic EGFR and cMet. Proceedings of 16 the National Academy of Sciences 105 , 692697, 17 doi:papers2://publication/doi/10.1073/pnas.0707270105 (2008). 18 106 Rikova, K. et al. Global survey of phosphotyrosine signaling identifies oncogenic kinases in 19 20 lung cancer. Cell 131 , 11901203, doi:10.1016/j.cell.2007.11.025 (2007). 21 107 Herskowitz, J. H. et al. Phosphoproteomic analysis reveals sitespecific changes in GFAP 22 and NDRG2 phosphorylation in frontotemporal lobar degeneration. Journal of proteome 23 research 9, 63686379, doi:10.1021/pr100666c (2010). 24 108 Kim, W. et al. Systematic and quantitative assessment of the ubiquitinmodified proteome. 25 Molecular cell 44 , 325340, doi:10.1016/j.molcel.2011.08.025 (2011). 26 109 Aono, S. & Hirai, Y. Phosphorylation of claudin4 is required for tight junction formation in 27 a human keratinocyte cell line. Exp Cell Res 314 , 33263339, 28 doi:10.1016/j.yexcr.2008.08.012 (2008). 29 30 110 Imami, K., Sugiyama, N., Kyono, Y., Tomita, M. & Ishihama, Y. Automated 31 phosphoproteome analysis for cultured cancer cells by twodimensional nanoLCMS using a 32 calcined titania/C18 biphasic column. Analytical sciences : the international journal of the 33 Japan Society for Analytical Chemistry 24 , 161166 (2008). 34 111 Depontieu, F. R. et al. Identification of tumorassociated, MHC class IIrestricted 35 phosphopeptides as targets for immunotherapy. Proc Natl Acad Sci U S A 106 , 12073 36 12078, doi:10.1073/pnas.0903852106 (2009). 37 112 Ikari, A. et al. Tight junctional localization of claudin16 is regulated by syntaxin 8 in renal 38 tubular epithelial cells. J Biol Chem 289 , 1311213123, doi:10.1074/jbc.M113.541193 39 40 (2014). 41 113 Kettenbach, A. N. et al. Quantitative phosphoproteomics identifies substrates and functional 42 modules of Aurora and Pololike kinase activities in mitotic cells. Sci Signal 4, rs5, 43 doi:10.1126/scisignal.2001497 (2011). 44 114 Ishizaki, T. et al. Cyclic AMP induces phosphorylation of claudin5 immunoprecipitates and 45 expression of claudin5 gene in blood–brainbarrier endothelial cells via protein kinase A 46 dependent and independent pathways. Experimental Cell Research 290 , 275288, 47 doi:10.1016/s00144827(03)003549 (2003). 48 115 Soma, T. et al. Thr207 of claudin5 is involved in sizeselective loosening of the endothelial 49 50 barrier by cyclic AMP. Experimental Cell Research 300 , 202212, 51 doi:10.1016/j.yexcr.2004.07.012 (2004). 52 116 Ikari, A. Phosphorylation of paracellin1 at Ser217 by protein kinase A is essential for 53 localization in tight junctions. Journal of Cell Science 119 , 17811789, 54 doi:10.1242/jcs.02901 (2006). 55 117 Overgaard, C. E., Daugherty, B. L., Mitchell, L. A. & Koval, M. Claudins: control of barrier 56 function and regulation in response to oxidant stress. Antioxidants & redox signaling 15 , 57 11791193, doi:10.1089/ars.2011.3893 (2011). 58 59 60 ACS Paragon Plus Environment Journal of Proteome Research Page 48 of 58

1 2 118 MolinaJijon, E., RodriguezMunoz, R., Namorado Mdel, C., PedrazaChaverri, J. & Reyes, 3 J. L. Oxidative stress induces claudin2 nitration in experimental type 1 diabetic 4 nephropathy. Free radical biology & medicine 72 , 162175, 5 doi:10.1016/j.freeradbiomed.2014.03.040 (2014). 6 119 Van Itallie, C. M. Palmitoylation of claudins is required for efficient tightjunction 7 localization. Journal of Cell Science 118 , 14271436, doi:10.1242/jcs.01735 (2005). 8 120 Heiler, S., Mu, W., Zoller, M. & Thuma, F. The importance of claudin7 palmitoylation on 9 10 membrane subdomain localization and metastasispromoting activities. Cell communication 11 and signaling : CCS 13 , 29, doi:10.1186/s129640150105y (2015). 12 121 Yang, W., Di Vizio, D., Kirchner, M., Steen, H. & Freeman, M. R. Proteome scale 13 characterization of human Sacylated proteins in lipid raftenriched and nonraft 14 membranes. Mol Cell Proteomics 9, 5470, doi:10.1074/mcp.M800448MCP200 (2010). 15 122 Dowal, L., Yang, W., Freeman, M. R., Steen, H. & Flaumenhaft, R. Proteomic analysis of 16 palmitoylated platelet proteins. Blood 118 , e6273, doi:10.1182/blood201105353078 17 (2011). 18 123 Martin, B. R., Wang, C., Adibekian, A., Tully, S. E. & Cravatt, B. F. Global profiling of 19 20 dynamic protein palmitoylation. Nat Methods 9, 8489, doi:10.1038/nmeth.1769 (2012). 21 124 Mandel, I. et al. The ubiquitinproteasome pathway regulates claudin 5 degradation. J Cell 22 Biochem 113 , 24152423, doi:10.1002/jcb.24118 (2012). 23 125 Takahashi, S. et al. The E3 ubiquitin ligase LNX1p80 promotes the removal of claudins 24 from tight junctions in MDCK cells. Journal of Cell Science 122 , 985994, 25 doi:10.1242/jcs.040055 (2009). 26 126 Van Itallie, C. M., Mitic, L. L. & Anderson, J. M. SUMOylation of claudin2. Annals of the 27 New York Academy of Sciences 1258 , 6064, doi:10.1111/j.17496632.2012.06541.x (2012). 28 127 Ahmad, W. et al. Claudin1 required for HCV virus entry has high potential for 29 30 phosphorylation and Oglycosylation. Virology Journal 8, 229, doi:10.1186/1743422x8 31 229 (2011). 32 128 Butt, A. M. et al. Role of post translational modifications and novel crosstalk between 33 phosphorylation and ObetaGlcNAc modifications in human claudin1, 3 and 4. 34 Molecular Biology Reports 39 , 13591369, doi:10.1007/s1103301108707 (2011). 35 129 Price, M. G., Davis, C. F., Deng, F. & Burgess, D. L. The alphaamino3hydroxyl5 36 methyl4isoxazolepropionate receptor trafficking regulator "stargazin" is related to the 37 claudin family of proteins by Its ability to mediate cellcell adhesion. J Biol Chem 280 , 38 1971119720, doi:10.1074/jbc.M500623200 (2005). 39 40 130 Maher, G. J. et al. The cataractassociated protein TMEM114, and TMEM235, are 41 glycosylated transmembrane proteins that are distinct from claudin family members. FEBS 42 Lett 585 , 21872192, doi:10.1016/j.febslet.2011.05.060 (2011). 43 131 Gupta, R. & Brunak, S. Prediction of glycosylation across the human proteome and the 44 correlation to protein function. Pac Symp Biocomput , 310322 (2002). 45 132 Fujita, H. et al. Differential expression and subcellular localization of claudin7, 8, 12, 13, 46 and 15 along the mouse intestine. The journal of histochemistry and cytochemistry : official 47 journal of the Histochemistry Society 54 , 933944, doi:10.1369/jhc.6A6944.2006 (2006). 48 133 Schrade, A. et al. Expression and localization of claudins3 and 12 in transformed human 49 50 brain endothelium. Fluids and barriers of the CNS 9, 6, doi:10.1186/2045811896 (2012). 51 134 Steentoft, C. et al. Precision mapping of the human OGalNAc glycoproteome through 52 SimpleCell technology. The EMBO journal 32 , 14781488, doi:10.1038/emboj.2013.79 53 (2013). 54 135 Awan, F. M. et al. Insilico analysis of claudin5 reveals novel putative sites for post 55 translational modifications: Insights into potential molecular determinants of bloodbrain 56 barrier breach during HIV1 infiltration. Infection, genetics and evolution : journal of 57 molecular epidemiology and evolutionary genetics in infectious diseases 27 , 355365, 58 doi:10.1016/j.meegid.2014.07.022 (2014). 59 60 ACS Paragon Plus Environment Page 49 of 58 Journal of Proteome Research

1 2 136 Blom, N., Gammeltoft, S. & Brunak, S. Sequence and structurebased prediction of 3 eukaryotic protein phosphorylation sites. Journal of molecular biology 294 , 13511362, 4 doi:10.1006/jmbi.1999.3310 (1999). 5 137 Trinidad, J. C. et al. Global identification and characterization of both OGlcNAcylation and 6 phosphorylation at the murine synapse. Mol Cell Proteomics 11 , 215229, 7 doi:10.1074/mcp.O112.018366 (2012). 8 138 Buzza, M. S. et al. Membraneanchored serine protease matriptase regulates epithelial 9 10 barrier formation and permeability in the intestine. Proc Natl Acad Sci U S A 107 , 4200 11 4205, doi:10.1073/pnas.0903923107 (2010). 12 139 Groschwitz, K. R. et al. Mast cells regulate homeostatic intestinal epithelial migration and 13 barrier function by a chymase/Mcpt4dependent mechanism. Proc Natl Acad Sci U S A 106 , 14 2238122386, doi:10.1073/pnas.0906372106 (2009). 15 140 Zeissig, S. et al. Changes in expression and distribution of claudin 2, 5 and 8 lead to 16 discontinuous tight junctions and barrier dysfunction in active Crohn's disease. Gut 56 , 61 17 72, doi:10.1136/gut.2006.094375 (2007). 18 141 Willemsen, L. E. M., Hoetjes, J. P., Van Deventer, S. J. H. & Van Tol, E. A. F. Abrogation 19 20 of IFNgamma mediated epithelial barrier disruption by serine protease inhibition. Clinical 21 and Experimental Immunology 142 , 275284, doi:10.1111/j.13652249.2005.02906.x 22 (2005). 23 142 Van Itallie, C. M., Colegio, O. R. & Anderson, J. M. The cytoplasmic tails of claudins can 24 influence tight junction barrier properties through effects on protein stability. The Journal of 25 membrane biology 199 , 2938 (2004). 26 143 Murakami, T., Felinski, E. A. & Antonetti, D. A. Occludin phosphorylation and 27 ubiquitination regulate tight junction trafficking and vascular endothelial growth factor 28 induced permeability. J Biol Chem 284 , 2103621046, doi:10.1074/jbc.M109.016766 29 30 (2009). 31 144 Ebihara, C. et al. Preparation of a claudintargeting molecule using a Cterminal fragment of 32 Clostridium perfringens enterotoxin. J Pharmacol Exp Ther 316 , 255260, 33 doi:10.1124/jpet.105.093351 (2006). 34 145 Lohrberg, D. et al. A strategy for enrichment of claudins based on their affinity to 35 Clostridium perfringens enterotoxin. BMC Molecular Biology 10 , 61, doi:10.1186/1471 36 21991061 (2009). 37 146 Luissint, A.C. et al. Guanine nucleotidebinding protein Gαi2: a new partner of claudin5 38 that regulates tight junction integrity in human brain endothelial cells. Journal of Cerebral 39 40 Blood Flow & Metabolism 32 , 860873, doi:10.1038/jcbfm.2011.202 (2012). 41 147 Jaspers, M. H. J. et al. The Claudin Megatrachea Protein Complex. Journal of Biological 42 Chemistry 287 , 3675636765, doi:10.1074/jbc.M112.399410 (2012). 43 148 Ivanov, A. I. Endocytosis of Epithelial Apical Junctional Proteins by a Clathrinmediated 44 Pathway into a Unique Storage Compartment. Molecular Biology of the Cell 15 , 176188, 45 doi:10.1091/mbc.E03050319 (2004). 46 149 Fredriksson, K. et al. Proteomic analysis of proteins surrounding occludin and claudin4 47 reveals their proximity to signaling and trafficking networks. PLoS One 10 , e0117074, 48 doi:10.1371/journal.pone.0117074 (2015). 49 50 150 Mitic, L. L. Expression, solubilization, and biochemical characterization of the tight 51 junction transmembrane protein claudin4. Protein Science 12 , 218227, 52 doi:10.1110/ps.0233903 (2003). 53 151 Tsukita, S., Yamazaki, Y., Katsuno, T. & Tamura, A. Tight junctionbased epithelial 54 microenvironment and cell proliferation. Oncogene 27 , 69306938, 55 doi:10.1038/onc.2008.344 (2008). 56 152 Tang, V. W. Proteomic and bioinformatic analysis of epithelial tight junction reveals an 57 unexpected cluster of synaptic molecules. Biology Direct 1, 37, doi:10.1186/174561501 58 37 (2006). 59 60 ACS Paragon Plus Environment Journal of Proteome Research Page 50 of 58

1 2 153 Yamazaki, Y., Okawa, K., Yano, T. & Tsukita, S. Optimized proteomic analysis on gels of 3 cellcell adhering junctional membrane proteins. Biochemistry 47 , 53785386, 4 doi:10.1021/bi8002567 (2008). 5 154 Pisitkun, T. et al. Highthroughput identification of IMCD proteins using LCMS/MS. 6 Physiological genomics 25 , 263276, doi:10.1152/physiolgenomics.00214.2005 (2006). 7 155 Hoffert, J. D., Wang, G., Pisitkun, T., Shen, R. F. & Knepper, M. A. An automated platform 8 for analysis of phosphoproteomic datasets: application to kidney collecting duct 9 10 phosphoproteins. Journal of proteome research 6, 35013508, doi:10.1021/pr0701153 11 (2007). 12 156 Fujiki, Y., Hubbard, A. L., Fowler, S. & Lazarow, P. B. Isolation of intracellular membranes 13 by means of sodium carbonate treatment: application to endoplasmic reticulum. J Cell Biol 14 93 , 97102 (1982). 15 157 Fischer, F., Wolters, D., Rogner, M. & Poetsch, A. Toward the complete membrane 16 proteome: high coverage of integral membrane proteins through transmembrane peptide 17 detection. Mol Cell Proteomics 5, 444453, doi:10.1074/mcp.M500234MCP200 (2006). 18 158 Franzel, B., Fischer, F., Trotschel, C., Poetsch, A. & Wolters, D. The twophase partitioning 19 20 systema powerful technique to purify integral membrane proteins of Corynebacterium 21 glutamicum for quantitative shotgun analysis. Proteomics 9, 22632272, 22 doi:10.1002/pmic.200800766 (2009). 23 159 Mathias, R. A. et al. Triton X114 phase separation in the isolation and purification of 24 mouse liver microsomal membrane proteins. Methods 54 , 396406, 25 doi:10.1016/j.ymeth.2011.01.006 (2011). 26 160 McQuade, L. R., Schmidt, U., Pascovici, D., Stojanov, T. & Baker, M. S. Improved 27 membrane proteomics coverage of human embryonic stem cells by peptide IPGIEF. 28 Journal of proteome research 8, 56425649, doi:10.1021/pr900597s (2009). 29 30 161 Schulz, T. C. et al. A largescale proteomic analysis of human embryonic stem cells. BMC 31 genomics 8, 478, doi:10.1186/147121648478 (2007). 32 162 Han, X., Jin, M., Breuker, K. & McLafferty, F. W. Extending topdown mass spectrometry 33 to proteins with masses greater than 200 kilodaltons. Science 314 , 109112, 34 doi:10.1126/science.1128868 (2006). 35 163 Fenn, J. B., Mann, M., Meng, C. K., Wong, S. F. & Whitehouse, C. M. Electrospray 36 ionization for mass spectrometry of large biomolecules. Science 246 , 6471 (1989). 37 164 Loo, J. A., Edmonds, C. G. & Smith, R. D. Primary sequence information from intact 38 proteins by electrospray ionization tandem mass spectrometry. Science 248 , 201204 (1990). 39 40 165 Demirev, P. A., Ramirez, J. & Fenselau, C. Tandem mass spectrometry of intact proteins for 41 characterization of biomarkers from Bacillus cereus T spores. Anal Chem 73 , 57255731 42 (2001). 43 166 Bogdanov, B. & Smith, R. D. Proteomics by FTICR mass spectrometry: top down and 44 bottom up. Mass Spectrom Rev 24 , 168200, doi:10.1002/mas.20015 (2005). 45 167 Kelleher, N. L. TopDown Proteomics. Analytical Chemistry 66 , 197203 (2004). 46 168 Mann, M. & Kelleher, N. L. Precision proteomics: The case for high resolution and high 47 mass accuracy. Proceedings of the National Academy of Sciences 105 , 1813218138, 48 doi:10.1073/pnas.0800788105 (2008). 49 50 169 Savaryn, J. P., Catherman, A. D., Thomas, P. M., Abecassis, M. M. & Kelleher, N. L. The 51 emergence of topdown proteomics in clinical research. Genome Med 5, 53, 52 doi:10.1186/gm457 (2013). 53 170 Smith, L. M. et al. Proteoform: a single term describing protein complexity. Nature Methods 54 10 , 186187, doi:10.1038/nmeth.2369 (2013). 55 171 Yates, J. R. & Kelleher, N. L. Top Down Proteomics. Analytical Chemistry 85 , 61516151, 56 doi:10.1021/ac401484r (2013). 57 58 59 60 ACS Paragon Plus Environment Page 51 of 58 Journal of Proteome Research

1 2 172 Nesvizhskii, A. I. & Aebersold, R. Interpretation of shotgun proteomic data: the protein 3 inference problem. Mol Cell Proteomics 4, 14191440, doi:10.1074/mcp.R500012MCP200 4 (2005). 5 173 Nesvizhskii, A. I., Vitek, O. & Aebersold, R. Analysis and validation of proteomic data 6 generated by tandem mass spectrometry. Nat Methods 4, 787797, doi:10.1038/nmeth1088 7 (2007). 8 174 Tran, J. C. et al. Mapping intact protein isoforms in discovery mode using topdown 9 10 proteomics. Nature 480 , 254258, doi:10.1038/nature10575 (2011). 11 175 Catherman, A. D. et al. Largescale Topdown Proteomics of the Human Proteome: 12 Membrane Proteins, Mitochondria, and Senescence. Mol Cell Proteomics 12 , 34653473, 13 doi:10.1074/mcp.M113.030114 (2013). 14 176 Kelleher, N. L. A cellbased approach to the human proteome project. Journal of the 15 American Society for Mass Spectrometry 23 , 16171624, doi:10.1007/s1336101204699 16 (2012). 17 177 Moradian, A., Kalli, A., Sweredoski, M. J. & Hess, S. The topdown, middledown, and 18 bottomup mass spectrometry approaches for characterization of histone variants and their 19 20 posttranslational modifications. Proteomics 14 , 489497, doi:10.1002/pmic.201300256 21 (2014). 22 178 Wu, C. et al. A protease for 'middledown' proteomics. Nat Methods 9, 822824, 23 doi:10.1038/nmeth.2074 (2012). 24 179 Cannon, J. et al. Highthroughput middledown analysis using an orbitrap. Journal of 25 proteome research 9, 38863890, doi:10.1021/pr1000994 (2010). 26 180 Liu, X. et al. De Novo Protein Sequencing by Combining TopDown and BottomUp 27 Tandem Mass Spectra. Journal of proteome research , doi:10.1021/pr401300m (2014). 28 181 Wu, S. L. et al. Targeted proteomics of lowlevel proteins in human plasma by LC/MSn: 29 30 using human growth hormone as a model system. Journal of proteome research 1, 459465 31 (2002). 32 182 Immler, D., Greven, S. & Reinemer, P. Targeted proteomics in biomarker validation: 33 detection and quantification of proteins using a multidimensional peptide separation 34 strategy. Proteomics 6, 29472958, doi:10.1002/pmic.200500659 (2006). 35 183 Whiteaker, J. R. et al. A targeted proteomicsbased pipeline for verification of biomarkers in 36 plasma. Nature biotechnology 29 , 625634, doi:10.1038/nbt.1900 (2011). 37 184 Omenn, G. S., Baker, M. S. & Aebersold, R. Recent Workshops of the HUPO Human 38 Plasma Proteome Project (HPPP): a bridge with the HUPO CardioVascular Initiative and 39 40 the emergence of SRM targeted proteomics. Proteomics 11 , 34393443, 41 doi:10.1002/pmic.201100382 (2011). 42 185 Anderson, N. L. The Human Plasma Proteome: History, Character, and Diagnostic 43 Prospects. Molecular & Cellular Proteomics 1, 845867, doi:10.1074/mcp.R200007 44 MCP200 (2002). 45 186 Everley, R. A., Kunz, R. C., McAllister, F. E. & Gygi, S. P. Increasing throughput in 46 targeted proteomics assays: 54plex quantitation in a single mass spectrometry run. Anal 47 Chem 85 , 53405346, doi:10.1021/ac400845e (2013). 48 187 Peng, Y. et al. Topdown targeted proteomics for deep sequencing of tropomyosin isoforms. 49 50 Journal of proteome research 12 , 187198, doi:10.1021/pr301054n (2013). 51 188 Li, M. et al. Oncogeneinduced cellular senescence elicits an antiWarburg effect. 52 Proteomics 13 , 25852596, doi:10.1002/pmic.201200298 (2013). 53 189 Madian, A. G., Rochelle, N. S. & Regnier, F. E. Masslinked immunoselective assays in 54 targeted proteomics. Anal Chem 85 , 737748, doi:10.1021/ac302071k (2013). 55 190 Qeli, E. et al. Improved prediction of peptide detectability for targeted proteomics using a 56 rankbased algorithm and organismspecific data. Journal of proteomics 108C , 269283, 57 doi:10.1016/j.jprot.2014.05.011 (2014). 58 59 60 ACS Paragon Plus Environment Journal of Proteome Research Page 52 of 58

1 2 191 Reker, D. & Malmstrom, L. Bioinformatic challenges in targeted proteomics. Journal of 3 proteome research 11 , 43934402, doi:10.1021/pr300276f (2012). 4 192 Maiolica, A., Junger, M. A., Ezkurdia, I. & Aebersold, R. Targeted proteome investigation 5 via selected reaction monitoring mass spectrometry. Journal of proteomics 75 , 34953513, 6 doi:10.1016/j.jprot.2012.04.048 (2012). 7 193 Qiu, X., Zhang, H. & Lai, Y. Quantitative targeted proteomics for membrane transporter 8 proteins: method and application. The AAPS journal 16 , 714726, doi:10.1208/s12248014 9 10 96076 (2014). 11 194 Gillet, L. C. et al. Targeted data extraction of the MS/MS spectra generated by data 12 independent acquisition: a new concept for consistent and accurate proteome analysis. Mol 13 Cell Proteomics 11 , O111.016717, doi:10.1074/mcp.O111.016717 (2012). 14 195 Uchida, Y. et al. Quantitative targeted absolute proteomics of human bloodbrain barrier 15 transporters and receptors. Journal of neurochemistry 117 , 333345, doi:10.1111/j.1471 16 4159.2011.07208.x (2011). 17 196 Shawahna, R. et al. Transcriptomic and quantitative proteomic analysis of transporters and 18 drug metabolizing enzymes in freshly isolated human brain microvessels. Molecular 19 20 pharmaceutics 8, 13321341, doi:10.1021/mp200129p (2011). 21 197 Ohtsuki, S. et al. Quantitative targeted absolute proteomic analysis of transporters, receptors 22 and junction proteins for validation of human cerebral microvascular endothelial cell line 23 hCMEC/D3 as a human bloodbrain barrier model. Molecular pharmaceutics 10 , 289296, 24 doi:10.1021/mp3004308 (2013). 25 198 Pasquali, C., Fialka, I. & Huber, L. A. Subcellular fractionation, electromigration analysis 26 and mapping of organelles. J Chromatogr B Biomed Sci Appl 722 , 89102 (1999). 27 199 Huber, L. A. Organelle Proteomics: Implications for Subcellular Fractionation in 28 Proteomics. Circulation Research 92 , 962968, doi:10.1161/01.res.0000071748.48338.25 29 30 (2003). 31 200 Banan, A. Isoform of Protein Kinase C Alters Barrier Function in Intestinal Epithelium 32 through Modulation of Distinct Claudin Isotypes: A Novel Mechanism for Regulation of 33 Permeability. Journal of Pharmacology and Experimental Therapeutics 313 , 962982, 34 doi:10.1124/jpet.105.083428 (2005). 35 201 Lambert, D., O'Neill, C. A. & Padfield, P. J. Methylbetacyclodextrin increases 36 permeability of Caco2 cell monolayers by displacing specific claudins from cholesterol rich 37 domains associated with tight junctions. Cellular physiology and biochemistry : 38 international journal of experimental cellular physiology, biochemistry, and pharmacology 39 40 20 , 495506, doi:10.1159/000107533 (2007). 41 202 Lynch, R. D. et al. Cholesterol depletion alters detergentspecific solubility profiles of 42 selected tight junction proteins and the phosphorylation of occludin. Exp Cell Res 313 , 43 25972610, doi:10.1016/j.yexcr.2007.05.009 (2007). 44 203 Sugibayashi, K., Onuki, Y. & Takayama, K. Displacement of tight junction proteins from 45 detergentresistant membrane domains by treatment with sodium caprate. European journal 46 of pharmaceutical sciences : official journal of the European Federation for 47 Pharmaceutical Sciences 36 , 246253, doi:10.1016/j.ejps.2008.09.011 (2009). 48 204 Schuck, S., Honsho, M., Ekroos, K., Shevchenko, A. & Simons, K. Resistance of cell 49 50 membranes to different detergents. Proc Natl Acad Sci U S A 100 , 57955800, 51 doi:10.1073/pnas.0631579100 (2003). 52 205 Nagase, S., Doyama, R., Yagi, K. & Kondoh, M. Recent advances in claudintargeting 53 technology. Biol Pharm Bull 36 , 708714 (2013). 54 206 Speers, A. E. & Cravatt, B. F. A tandem orthogonal proteolysis strategy for highcontent 55 chemical proteomics. J Am Chem Soc 127 , 1001810019, doi:10.1021/ja0532842 (2005). 56 207 Sadaghiani, A. M., Verhelst, S. H. & Bogyo, M. Tagging and detection strategies for 57 activitybased proteomics. Curr Opin Chem Biol 11 , 2028, doi:10.1016/j.cbpa.2006.11.030 58 (2007). 59 60 ACS Paragon Plus Environment Page 53 of 58 Journal of Proteome Research

1 2 208 Heal, W. P., Dang, T. H. & Tate, E. W. Activitybased probes: discovering new biology and 3 new drug targets. Chem Soc Rev 40 , 246257, doi:10.1039/c0cs00004c (2011). 4 209 Jing, C. & Cornish, V. W. Chemical tags for labeling proteins inside living cells. Acc Chem 5 Res 44 , 784792, doi:10.1021/ar200099f (2011). 6 210 Hayashi, T. & Hamachi, I. Traceless affinity labeling of endogenous proteins for functional 7 analysis in living cells. Acc Chem Res 45 , 14601469, doi:10.1021/ar200334r (2012). 8 211 Kovalenko, O. V., Yang, X. H. & Hemler, M. E. A novel cysteine crosslinking method 9 10 reveals a direct association between claudin1 and tetraspanin CD9. Mol Cell Proteomics 6, 11 18551867, doi:10.1074/mcp.M700183MCP200 (2007). 12 212 Piontek, J. et al. Formation of tight junction: determinants of homophilic interaction 13 between classic claudins. The FASEB Journal 22 , 146158, doi:10.1096/fj.078319com 14 (2007). 15 213 Angelow, S. & Yu, A. S. L. StructureFunction Studies of Claudin Extracellular Domains 16 by Cysteinescanning Mutagenesis. Journal of Biological Chemistry 284 , 2920529217, 17 doi:10.1074/jbc.M109.043752 (2009). 18 214 Cukierman, L., Meertens, L., Bertaux, C., Kajumo, F. & Dragic, T. Residues in a Highly 19 20 Conserved Claudin1 Motif Are Required for Hepatitis C Virus Entry and Mediate the 21 Formation of CellCell Contacts. Journal of Virology 83 , 54775484, doi:10.1128/jvi.02262 22 08 (2009). 23 215 Dukes, J. D., Whitley, P. & Chalmers, A. D. The PIKfyve Inhibitor YM201636 Blocks the 24 Continuous Recycling of the Tight Junction Proteins Claudin1 and Claudin2 in MDCK 25 cells. PLoS ONE 7, e28659, doi:10.1371/journal.pone.0028659 (2012). 26 216 Giron, P., Dayon, L. & Sanchez, J. C. Cysteine tagging for MSbased proteomics. Mass 27 Spectrom Rev 30 , 366395, doi:10.1002/mas.20285 (2011). 28 217 Dabrowski, S. et al. Redoxsensitive structure and function of the first extracellular loop of 29 30 the cellcell contact protein claudin1: lessons from molecular structure to animals. 31 Antioxidants & redox signaling 22 , 114, doi:10.1089/ars.2013.5706 (2015). 32 218 Furuse, M. et al. Claudinbased tight junctions are crucial for the mammalian epidermal 33 barrier: a lesson from claudin1deficient mice. J Cell Biol 156 , 10991111, 34 doi:10.1083/jcb.200110122 (2002). 35 219 Umeda, K. et al. ZO1 and ZO2 independently determine where claudins are polymerized 36 in tightjunction strand formation. Cell 126 , 741754, doi:10.1016/j.cell.2006.06.043 (2006). 37 220 Fagotto, F. & Gumbiner, B. M. Cell contactdependent signaling. Developmental biology 38 180 , 445454, doi:10.1006/dbio.1996.0318 (1996). 39 40 221 Gumbiner, B. M. Cell adhesion: the molecular basis of tissue architecture and 41 morphogenesis. Cell 84 , 345357 (1996). 42 222 Brucher, B. L. & Jamall, I. S. CellCell Communication in the Tumor Microenvironment, 43 Carcinogenesis, and Anticancer Treatment. Cellular physiology and biochemistry : 44 international journal of experimental cellular physiology, biochemistry, and pharmacology 45 34 , 213243, doi:10.1159/000362978 (2014). 46 223 Guillemot, L., Paschoud, S., Pulimeno, P., Foglia, A. & Citi, S. The cytoplasmic plaque of 47 tight junctions: A scaffolding and signalling center. Biochimica et Biophysica Acta (BBA) - 48 Biomembranes 1778 , 601613, 49 50 doi:papers2://publication/doi/10.1016/j.bbamem.2007.09.032 (2008). 51 224 Arabzadeh, A., Troy, T. C. & Turksen, K. Role of the Cldn6 Cytoplasmic Tail Domain in 52 Membrane Targeting and Epidermal Differentiation In Vivo. Molecular and Cellular 53 Biology 26 , 58765887, doi:10.1128/mcb.0234205 (2006). 54 225 Muller, D. Familial hypomagnesemia with hypercalciuria and nephrocalcinosis: blocking 55 endocytosis restores surface expression of a novel Claudin16 mutant that lacks the entire C 56 terminal cytosolic tail. Human Molecular Genetics 15 , 10491058, doi:10.1093/hmg/ddl020 57 (2006). 58 59 60 ACS Paragon Plus Environment Journal of Proteome Research Page 54 of 58

1 2 226 Ruffer, C. The Cterminal cytoplasmic tail of claudins 1 and 5 but not its PDZbinding motif 3 is required for apical localization at epithelial and endothelial tight junctions. European 4 Journal of Cell Biology 83 , 135144, doi:10.1078/0171933500366 (2004). 5 227 Nagasawa, K. et al. Possible involvement of gap junctions in the barrier function of tight 6 junctions of brain and lung endothelial cells. Journal of cellular physiology 208 , 123132, 7 doi:10.1002/jcp.20647 (2006). 8 228 Kojima, T. et al. Cx32 but not Cx26 is associated with tight junctions in primary cultures of 9 10 rat hepatocytes. Exp Cell Res 263 , 193201, doi:10.1006/excr.2000.5103 (2001). 11 229 Blackman, B., Russell, T., Nordeen, S. K., Medina, D. & Neville, M. C. Claudin 7 12 expression and localization in the normal murine mammary gland and murine mammary 13 tumors. Breast cancer research : BCR 7, R248255, doi:10.1186/bcr988 (2005). 14 230 Liebner, S. et al. Wnt/betacatenin signaling controls development of the bloodbrain 15 barrier. J Cell Biol 183 , 409417, doi:10.1083/jcb.200806024 (2008). 16 231 Beeman, N., Webb, P. G. & Baumgartner, H. K. Occludin is required for apoptosis when 17 claudinclaudin interactions are disrupted. Cell death & disease 3, e273, 18 doi:10.1038/cddis.2012.14 (2012). 19 20 232 Inai, T. The coculture method to examine interactions between claudin isoforms in tight 21 junctionfree HEK293 cells and tight junctionbearing MDCK II cells. Methods in 22 molecular biology (Clifton, N.J.) 762 , 101114, doi:10.1007/9781617791857_8 (2011). 23 233 Milatz, S. et al. Probing the cisarrangement of prototype tight junction proteins claudin1 24 and claudin3. The Biochemical journal , doi:10.1042/BJ20150148 (2015). 25 234 Rossa, J. et al. Claudin3 and claudin5 protein folding and assembly into the tight junction 26 are controlled by nonconserved residues in the transmembrane 3 (TM3) and extracellular 27 loop 2 (ECL2) segments. J Biol Chem 289 , 76417653, doi:10.1074/jbc.M113.531012 28 (2014). 29 30 235 Rossa, J. et al. Molecular and structural transmembrane determinants critical for embedding 31 claudin5 into tight junctions reveal a distinct fourhelix bundle arrangement. The 32 Biochemical journal 464 , 4960, doi:10.1042/BJ20140431 (2014). 33 236 Capaldo, C. T. et al. Proinflammatory cytokineinduced tight junction remodeling through 34 dynamic selfassembly of claudins. Mol Biol Cell 25 , 27102719, doi:10.1091/mbc.E1402 35 0773 (2014). 36 237 Wu, C. J., Mannan, P., Lu, M. & Udey, M. C. Epithelial cell adhesion molecule (EpCAM) 37 regulates claudin dynamics and tight junctions. J Biol Chem 288 , 1225312268, 38 doi:10.1074/jbc.M113.457499 (2013). 39 40 238 Kuhn, S. et al. A complex of EpCAM, claudin7, CD44 variant isoforms, and tetraspanins 41 promotes colorectal cancer progression. Molecular cancer research : MCR 5, 553567, 42 doi:10.1158/15417786.MCR060384 (2007). 43 239 Nubel, T. et al. Claudin7 Regulates EpCAMMediated Functions in Tumor Progression. 44 Molecular Cancer Research 7, 285299, doi:10.1158/15417786.mcr080200 (2009). 45 240 Pope, J. L. et al. Claudin1 regulates intestinal epithelial homeostasis through the 46 modulation of Notchsignalling. Gut 63 , 622634, doi:10.1136/gutjnl2012304241 (2014). 47 241 EscuderoEsparza, A., Jiang, W. G. & Martin, T. A. Claudin5 is involved in breast cancer 48 cell motility through the NWASP and ROCK signalling pathways. Journal of experimental 49 50 & clinical cancer research : CR 31 , 43, doi:10.1186/175699663143 (2012). 51 242 Mankertz, J. et al. Functional crosstalk between Wnt signaling and Cdxrelated 52 transcriptional activation in the regulation of the claudin2 promoter activity. Biochemical 53 and biophysical research communications 314 , 10011007, doi:10.1016/j.bbrc.2003.12.185 54 (2004). 55 243 Miwa, N. et al. Involvement of claudin1 in the betacatenin/Tcf signaling pathway and its 56 frequent upregulation in human colorectal cancers. Oncology research 12 , 469476 (2001). 57 244 Oku, N., Sasabe, E., Ueta, E., Yamamoto, T. & Osaki, T. Tight junction protein claudin1 58 enhances the invasive activity of oral squamous cell carcinoma cells by promoting cleavage 59 60 ACS Paragon Plus Environment Page 55 of 58 Journal of Proteome Research

1 2 of laminin5 gamma2 chain via matrix metalloproteinase (MMP)2 and membranetype 3 MMP1. Cancer Res 66 , 52515257, doi:10.1158/00085472.can054478 (2006). 4 245 Tokes, A. M. et al. Claudin1, 3 and 4 proteins and mRNA expression in benign and 5 malignant breast lesions: a research study. Breast cancer research : BCR 7, R296305, 6 doi:10.1186/bcr983 (2005). 7 246 Swisshelm, K., Macek, R. & Kubbies, M. Role of claudins in tumorigenesis. Advanced drug 8 delivery reviews 57 , 919928, doi:10.1016/j.addr.2005.01.006 (2005). 9 10 247 Stache, C. et al. Tight junction protein claudin1 is differentially expressed in 11 craniopharyngioma subtypes and indicates invasive tumor growth. Neuro-oncology 16 , 256 12 264, doi:10.1093/neuonc/not195 (2014). 13 248 Blanchard, A. A. et al. Claudins 1, 3, and 4 protein expression in ER negative breast cancer 14 correlates with markers of the basal phenotype. Virchows Archiv : an international journal 15 of pathology 454 , 647656, doi:10.1007/s0042800907706 (2009). 16 249 Blanchard, A. A. et al. Claudin 1 expression in basallike breast cancer is related to patient 17 age. BMC Cancer 13 , 268, doi:10.1186/1471240713268 (2013). 18 250 Leotlela, P. D. et al. Claudin1 overexpression in melanoma is regulated by PKC and 19 20 contributes to melanoma cell motility. Oncogene 26 , 38463856, 21 doi:10.1038/sj.onc.1210155 (2007). 22 251 Alshbool, F. Z. & Mohan, S. Differential expression of claudin family members during 23 osteoblast and osteoclast differentiation: Cldn1 is a novel positive regulator of 24 osteoblastogenesis. PLoS One 9, e114357, doi:10.1371/journal.pone.0114357 (2014). 25 252 Dahiya, N., Becker, K. G., Wood, W. H., Zhang, Y. & Morin, P. J. Claudin7 Is Frequently 26 Overexpressed in Ovarian Cancer and Promotes Invasion. PLoS One 6, 27 doi:10.1371/journal.pone.0022119 (2011). 28 253 Shannon, P. et al. Cytoscape: a software environment for integrated models of biomolecular 29 30 interaction networks. Genome Res 13 , 24982504, doi:10.1101/gr.1239303 (2003). 31 254 Itoh, M. et al. Direct binding of three tight junctionassociated MAGUKs, ZO1, ZO2, and 32 ZO3, with the COOH termini of claudins. J Cell Biol 147 , 13511363 (1999). 33 255 Hamazaki, Y., Itoh, M., Sasaki, H., Furuse, M. & Tsukita, S. MultiPDZ domain protein 1 34 (MUPP1) is concentrated at tight junctions through its possible interaction with claudin1 35 and junctional adhesion molecule. J Biol Chem 277 , 455461, doi:10.1074/jbc.M109005200 36 (2002). 37 256 Roh, M. H., Liu, C. J., Laurinec, S. & Margolis, B. The carboxyl terminus of zona 38 occludens3 binds and recruits a mammalian homologue of discs lost to tight junctions. J 39 40 Biol Chem 277 , 2750127509, doi:10.1074/jbc.M201177200 (2002). 41 257 Nakatsukasa, M. et al. Tumorassociated calcium signal transducer 2 is required for the 42 proper subcellular localization of claudin 1 and 7: implications in the pathogenesis of 43 gelatinous droplike corneal dystrophy. The American journal of pathology 177 , 13441355, 44 doi:10.2353/ajpath.2010.100149 (2010). 45 258 Singh, A. B., Sharma, A. & Dhawan, P. Claudin1 expression confers resistance to anoikis 46 in colon cancer cells in a Srcdependent manner. Carcinogenesis 33 , 25382547, 47 doi:10.1093/carcin/bgs275 (2012). 48 259 DiTommaso, T. et al. Keratin 76 is required for tight junction function and maintenance of 49 50 the skin barrier. PLoS Genet 10 , e1004706, doi:10.1371/journal.pgen.1004706 (2014). 51 260 Guo, Z. et al. Proteomics strategy to identify substrates of LNX, a PDZ domaincontaining 52 E3 ubiquitin ligase. Journal of proteome research 11 , 48474862, doi:10.1021/pr300674c 53 (2012). 54 261 Harris, H. J. et al. CD81 and claudin 1 coreceptor association: role in hepatitis C virus entry. 55 J Virol 82 , 50075020, doi:10.1128/jvi.0228607 (2008). 56 262 Raleigh, D. R. et al. Occludin S408 phosphorylation regulates tight junction protein 57 interactions and barrier function. J Cell Biol 193 , 565582, doi:10.1083/jcb.201010065 58 (2011). 59 60 ACS Paragon Plus Environment Journal of Proteome Research Page 56 of 58

1 2 263 Cording, J. et al. Redox Regulation of Cell Contacts by Tricellulin and Occludin: Redox 3 Sensitive Cysteine Sites in Tricellulin Regulate Both Tri and Bicellular Junctions in Tissue 4 Barriers as Shown in Hypoxia and Ischemia. Antioxidants & redox signaling 23 , 10351049, 5 doi:10.1089/ars.2014.6162 (2015). 6 264 Furuse, M., Sasaki, H. & Tsukita, S. Manner of interaction of heterogeneous claudin species 7 within and between tight junction strands. J Cell Biol 147 , 891903 (1999). 8 265 Lim, T. S., Vedula, S. R., Kausalya, P. J., Hunziker, W. & Lim, C. T. Singlemolecular 9 10 level study of claudin1mediated adhesion. Langmuir : the ACS journal of surfaces and 11 colloids 24 , 490495, doi:10.1021/la702436x (2008). 12 266 Miyamori, H. et al. Claudin promotes activation of promatrix metalloproteinase2 mediated 13 by membranetype matrix metalloproteinases. J Biol Chem 276 , 2820428211, 14 doi:10.1074/jbc.M103083200 (2001). 15 267 Tanaka, M., Kamata, R. & Sakai, R. Phosphorylation of ephrinB1 via the interaction with 16 claudin following cellcell contact formation. The EMBO journal 24 , 37003711, 17 doi:10.1038/sj.emboj.7600831 (2005). 18 268 Douam, F., Lavillette, D. & Cosset, F. L. The mechanism of HCV entry into host cells. 19 20 Progress in molecular biology and translational science 129 , 63107, 21 doi:10.1016/bs.pmbts.2014.10.003 (2015). 22 269 Cocquerel, L., Kuo, C. C., Dubuisson, J. & Levy, S. CD81dependent binding of hepatitis C 23 virus E1E2 heterodimers. J Virol 77 , 1067710683 (2003). 24 270 Douam, F. et al. Critical interaction between E1 and E2 glycoproteins determines binding 25 and fusion properties of hepatitis C virus during cell entry. Hepatology (Baltimore, Md.) 59 , 26 776788, doi:10.1002/hep.26733 (2014). 27 271 Hopcraft, S. E. & Evans, M. J. Selection of a hepatitis C virus with altered entry factor 28 requirements reveals a genetic interaction between the E1 glycoprotein and claudins. 29 30 Hepatology (Baltimore, Md.) 62 , 10591069, doi:10.1002/hep.27815 (2015). 31 272 Gao, F. et al. Novel binding between premembrane protein and claudin1 is required for 32 efficient dengue virus entry. Biochemical and biophysical research communications 391 , 33 952957, doi:10.1016/j.bbrc.2009.11.172 (2010). 34 273 Lee, S. S., Glaunsinger, B., Mantovani, F., Banks, L. & Javier, R. T. MultiPDZ domain 35 protein MUPP1 is a cellular target for both adenovirus E4ORF1 and highrisk 36 papillomavirus type 18 E6 oncoproteins. J Virol 74 , 96809693 (2000). 37 274 Armbruester, V. et al. Np9 protein of human endogenous retrovirus K interacts with ligand 38 of numb protein X. J Virol 78 , 1031010319, doi:10.1128/jvi.78.19.1031010319.2004 39 40 (2004). 41 275 Korkaya, H. et al. The ORF3 protein of hepatitis E virus binds to Src homology 3 domains 42 and activates MAPK. J Biol Chem 276 , 4238942400, doi:10.1074/jbc.M101546200 (2001). 43 276 Betanzos, A. et al. The EhCPADH112 complex of Entamoeba histolytica interacts with tight 44 junction proteins occludin and claudin1 to produce epithelial damage. PLoS One 8, e65100, 45 doi:10.1371/journal.pone.0065100 (2013). 46 277 Zhou, M. & Wysocki, V. H. Surface induced dissociation: dissecting noncovalent protein 47 complexes in the gas phase. Acc Chem Res 47 , 10101018, doi:10.1021/ar400223t (2014). 48 278 Stengel, F., Aebersold, R. & Robinson, C. V. Joining forces: integrating proteomics and 49 50 crosslinking with the mass spectrometry of intact complexes. Mol Cell Proteomics 11 , 51 R111 014027, doi:10.1074/mcp.R111.014027 (2012). 52 279 Lasker, K. et al. Integrative structure modeling of macromolecular assemblies from 53 proteomics data. Mol Cell Proteomics 9, 16891702, doi:10.1074/mcp.R110.000067 (2010). 54 280 Benesch, J. L. & Ruotolo, B. T. Mass spectrometry: come of age for structural and 55 dynamical biology. Curr Opin Struct Biol 21 , 641649, doi:10.1016/j.sbi.2011.08.002 56 (2011). 57 58 59 60 ACS Paragon Plus Environment Page 57 of 58 Journal of Proteome Research

1 2 281 Barrera, N. P. & Robinson, C. V. Advances in the mass spectrometry of membrane proteins: 3 from individual proteins to intact complexes. Annu Rev Biochem 80 , 247271, 4 doi:10.1146/annurevbiochem062309093307 (2011). 5 282 Zhou, M. & Robinson, C. V. When proteomics meets structural biology. Trends Biochem 6 Sci 35 , 522529, doi:10.1016/j.tibs.2010.04.007 (2010). 7 283 Heck, A. J. Native mass spectrometry: a bridge between interactomics and structural 8 biology. Nat Methods 5, 927933, doi:10.1038/nmeth.1265 (2008). 9 10 284 Krause, G., Protze, J. & Piontek, J. Assembly and function of claudins: Structurefunction 11 relationships based on homology models and crystal structures. Seminars in cell & 12 developmental biology , doi:10.1016/j.semcdb.2015.04.010 (2015). 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 ACS Paragon Plus Environment Journal of Proteome Research Page 58 of 58

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 For TOC only--high resolution 34 152x120mm (300 x 300 DPI) 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 ACS Paragon Plus Environment