Dermatan sulfate epimerase 1 expression and mislocalization may interfere with dermatan sulfate synthesis and breast cancer cell growth

Dermatan sulfate epimerase 1 expression and mislocalization may interfere with dermatan sulfate synthesis and breast cancer cell growth

Journal Pre-proof Dermatan sulfate epimerase 1 expression and mislocalization may interfere with dermatan sulfate synthesis and breast cancer cell gro...

55MB Sizes 0 Downloads 47 Views

Journal Pre-proof Dermatan sulfate epimerase 1 expression and mislocalization may interfere with dermatan sulfate synthesis and breast cancer cell growth Eduardo Listik, Everton Galvão Xavier, Maria Aparecida da Silva Pinhal, Leny Toma PII:

S0008-6215(19)30581-6

DOI:

https://doi.org/10.1016/j.carres.2020.107906

Reference:

CAR 107906

To appear in:

Carbohydrate Research

Received Date: 3 October 2019 Revised Date:

10 December 2019

Accepted Date: 3 January 2020

Please cite this article as: E. Listik, Everton.Galvã. Xavier, M.A.d. Silva Pinhal, L. Toma, Dermatan sulfate epimerase 1 expression and mislocalization may interfere with dermatan sulfate synthesis and breast cancer cell growth, Carbohydrate Research (2020), doi: https://doi.org/10.1016/ j.carres.2020.107906. This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. Please note that, during the production process, errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain. © 2020 Published by Elsevier Ltd.

1

Dermatan sulfate epimerase 1 expression and mislocalization may interfere with dermatan sulfate synthesis and breast cancer cell growth Eduardo Listik#, Everton Galvão Xavier#, Maria Aparecida da Silva Pinhal & Leny Toma*

Affiliation: Department of Biochemistry, Universidade Federal de São Paulo, Rua Três de Maio, 100, São Paulo, SP, Brazil. CEP: 04044-020. [email protected]; [email protected]; [email protected]; [email protected].

Corresponding author: Leny Toma, Ph.D. Department of Biochemistry Universidade Federal de São Paulo Rua Três de Maio, 100, 5th floor, CEP: 04044-020 São Paulo, SP, Brazil. Phone: +55-11-5573-6407 Email: [email protected]

#

Authors share equal contribution.

2 1

ABSTRACT

2

Dermatan sulfate (DS) is a glycosaminoglycan (GAG) that is produced through the

3

epimerization of the glucuronic acid on chondroitin sulfate into iduronic acid (IduA) by

4

dermatan sulfate epimerase (DS-epi) 1 and 2. Proteoglycans (PGs) play essential physiological

5

and pathological roles during cellular development, proliferation, differentiation, and cancer

6

metastasis. DS proteoglycans play vital roles during the process of tumorigenesis, due to the

7

increased flexibility of the polysaccharide chain in the presence of IduA residues, which

8

facilitate specific interactions with proteins, such as growth factors, cytokines, and angiogenic

9

factors. Furthermore, DS-epi is highly expressed in many tumors, especially in esophageal

10

squamous cell carcinoma. This study aimed to investigate the expression of DS-epi1 in multiple

11

breast cancer cell lines, including MCF7 (luminal A), MDA-MB-231 (triple-negative) and

12

SKBR3 (human epidermal growth factor receptor 2-positive), and its involvement in cancer

13

progression. A SKBR3 variant, SKBR3m, presented the most erratic cell growth pattern when

14

compared with those for MCF7 and MDA-MB-231. Moreover, SKBR3m cells demonstrated

15

the highest level of DS-epi1 gene expression and higher

16

protein level, MCF7 cells displayed the highest protein level for DS-epi1, whereas MDA-MB-

17

231 cells had the lowest level. DS-epi1 was found in vesicles and in the perinuclear

18

compartment only in SKBR3m cells, suggesting localization in the Golgi apparatus in these

19

cells, in contrast with the cytoplasmic localization observed in MCF7 and MDA-MB-231 cells.

20

The cytoplasm location of DS-epi1 likely compromised the formation of DS chains, but the

21

core protein was detected using a decorin antibody. Golgi-specific labeling confirmed the

22

localization of DS-epi1 in SKBR3m cells at the Golgi apparatus, indicating that the location of

23

the enzyme was a determinant for the synthesis of DS in this cell line, suggesting that DS may

24

play a decisive role in the tumor growth observed in this breast cancer cell line.

35

S-DS content. However, at the

3 25

Keywords: Breast cancer; cancer biology; dermatan sulfate; decorin proteoglycan;

26

Golgi apparatus.

27

1 INTRODUCTION

28

Breast cancer is one of the most prevalent types of tumors among women worldwide,

29

with more than 1.7 million new registered cases and 522,000 deaths every year. The incidence

30

of breast cancer has continued to grow, and more than 3.2 million new cases are expected by

31

2050 [1]. Breast cancer pathology can be classified according to morphologic, histologic, and

32

molecular parameters. Morphologically, breast tumors can be categorized as infiltrating ductal

33

carcinomas of no particular type, infiltrating lobular and tubular carcinomas, mucinous

34

carcinomas, and many others. Histological classifications allow the sub-categorization of

35

breast tumors, using characteristics such as cellular differentiation, nuclear pleomorphism,

36

and mitotic counts. At the molecular level, breast tumors can be classified according to the

37

presence or absence of estrogen receptor (ER), progesterone receptor (PR), or human

38

epidermal growth factor receptor type 2 (HER2). The majority of breast tumors are ER-

39

positive; however, many subgroups can be characterized, such as luminal A and B tumors,

40

which are ER+/PR+ [2-4].

41

Proteoglycans (PGs) are macromolecules that possess a protein core, to which one or

42

more glycosaminoglycan (GAG) chains are covalently attached [5]. Such molecules have

43

essential biological functions during vascularization and cancer metastasis and assist many

44

signaling proteins in the extracellular matrix [6, 7]. During tumoral development and growth,

45

the expression of PGs is markedly modified, frequently upregulated, and many PGs influence

46

or inhibit cell proliferation, depending on the tissue type [8-10].

47

Chondroitin sulfate (CS) is a GAG that is naturally found in the extracellular matrix of

48

connective tissue, either on the cell surface or in intracellular compartments. A disaccharidic

4 49

unit, composed of D-glucuronic acid (GlcA, β-1,4) and N-acetyl-D-galactosamine (GalNAc,

50

β-1,3), can be found in CS [11, 12]. Dermatan sulfate (DS), in contrast, possesses the GlcA

51

epimer iduronic acid (IdoA). Both GAGs are structurally similar, and they appear

52

concomitantly in PGs [7, 13].

53

GAG biosynthesis begins at the rough endoplasmic reticulum (RER) and proceeds to

54

the Golgi apparatus. In the RER, GAG synthesis begins with the formation of a

55

tetrasaccharidic sequence of GlcA, β1→3, D-galactose (Gal), β1→3, Gal, β1→4, xylose (Xyl),

56

β1→ [14-16]. After the addition of xylose and D-galactose, the synthetic process is transferred

57

to the Golgi apparatus [17, 18]. The fifth added sugar determines whether the GAG will

58

become a CS/DS or heparan sulfate (HS)/heparin (Hep) [19, 20]. The extension of CS occurs

59

through the activity of the chondroitin synthase enzyme. CS chains in mammals are

60

commonly sulfated on C-4 or C-6 positions of GalNAc, generating the isomers chondroitin-4-

61

sulfate or chondroitin-6-sulfate, respectively [21-23]. GlcA/IdoA can also be sulfated on the

62

C-2 position, either in CS or DS [24, 25]. Epimerization proceeds sulfation, during which

63

dermatan epimerase (DS-epi) 1 and 2 act on the C-5 of GlcA to generate an IdoA moiety [26,

64

27].

65

PGs play significant roles in several physiological and pathological cellular processes.

66

The results obtained in the present study may indicate that higher expression levels of DS-

67

epi1 in cells derived from breast tumors could be linked with higher growth rates. However,

68

the cells that express higher levels of DS-epi1 do not show higher DS-epi1 protein levels;

69

instead, DS-epi1 presents with a more specific localization in vesicles. This result led us to

70

question whether the vesicle-localized DS-epi1 found in cells with higher levels of DS-epi1

71

expression is functional.

72

2 MATERIALS AND METHODS

5 73

2.1 Materials

74

Anti-HS-epi (polyclonal rabbit, IgG, NBP2-31872) and anti-DS-epi1 (polyclonal

75

rabbit, IgG, NBP1-87962) antibodies were purchased from Novus Biologicals (Centennial,

76

Colorado, USA). Anti-decorin (monoclonal mouse, IgG, MA5-29240) was purchased from

77

Thermo Fisher (Waltham, Massachusetts, USA). All secondary antibodies, including anti-

78

rabbit Alexa Fluor 488 (cat. A-21206), anti-mouse Alexa Fluor 633 (cat. A-21050),

79

phalloidin-647 (cat. A22287), and lectin HPA from Helix Pomatia-647 (cat. L32454), were

80

purchased from Thermo Fisher. RPMI1640, Dulbecco’s Modified Eagle Medium (DMEM),

81

and fetal bovine serum (FBS) were all obtained from Gibco® (from Thermo Fisher).

82

2.2 Cell culture

83

MDA-MB-231 (ATCC® HTB-26TM), MCF7 (ATCC® HTB-22TM) and SKBR3

84

(ATCC® HTB-30TM) cells were purchased from the American Type Culture Collection

85

(ATCC, Manassas, Virgínia, USA). MDA-MB-231 and MCF7 cells were cultivated in

86

DMEM, whereas SKBR3 cells were cultivated in RPMI1640. SKBR3 cells were used in

87

specific assays: SKBR3m from a high passage (> 50), and SKBR3a from low passage levels

88

(< 5), as it has been previously described that breast cancer cell lines may acquire a more

89

aggressive profile in higher passages [28-31]. All media were supplemented with 10% FBS

90

(v/v) and 1% penicillin/streptomycin (v/v) The cells were grown at 37 °C, in a humidified

91

atmosphere containing 5% CO2.

92

2.3 MTT assay

93

The MTT assay was performed using the tetrazolium salt (MTT) reagent, as

94

previously described [32]. In brief, MDA-MB-231, MCF7, and both types of SKBR3 cells

95

were plated at a 3×103 cell/mL density in a 96-well plate. A 5 mg/mL sterile solution of MTT

96

in phosphate-buffered saline (PBS) was prepared, and 10 µL was added 4 h prior to cell

6 97

incubation for 24, 48, and 72 h. In some assays, a 96-h time point was also analyzed. At each

98

time point, the supernatant was removed from each well, and the formazan precipitate was

99

solubilized with 120 µL dimethylsulfoxide (DMSO). The plate was then briefly agitated, and

100

the absorbance of each well was read using a plate reader at 570 nm. The optical density of

101

blank wells was subtracted from each experimental well, and the results were normalized

102

against those for control wells to which MTT was not added.

103

2.4

104

electrophoresis

35

S-Sulfate metabolic radiolabeling of GAGs and analysis by agarose gel

105

MDA-MB-231, MCF7 and both types of SKBR3 cells were cultivated in either 35-

106

mm dishes or 24-well plates until they reached 60% confluence. Then, they were incubated

107

with Na235SO4 (100 µCi/mL) for 24 h. The medium was replaced with OptiMEM during the

108

radioactive pulse.

109

The medium was collected, and cells were washed twice with 2 mL cold PBS and

110

collected by scraping in 500 µL 3.5 M urea buffer (pH 7.4). PGs in both the collected media

111

and the cell extracts were proteolyzed with proteinase K at a final concentration of 100

112

µg/mL, for 3 h at 40 °C, before being inactivated at 95 °C for 10 min. Alternatively, the PGs

113

from the cell extracts and media were precipitated with three volumes of ethanol, in the

114

presence of 200 µg CS as a carrier, and incubated overnight at -20 °C. The solution was

115

centrifugated at 3,000 rpm for 10 min, and the precipitate was left to dry at a vacuum

116

concentrator. A proteolytic solution, consisting of 0.9 mM alcalase in 100 mM Tris-HCl (pH

117

7.4), was added and incubated for two days at 54 °C, followed by inactivation by boiling for 2

118

h.

119

In some experiments, after proteolysis, GAGs were digested with either chondroitinase

120

AC (0.1 units in 20 mM Tris-HCl, pH 7.2, 2% glycerol), chondroitinase ABC (0.1 units in 50

7 121

mM Tris-HCl, 60 mM sodium acetate, 0.02% of BSA (v/v)), both from Sigma Aldrich (San

122

Luis, Missouri, USA) or water, as a control. All samples were incubated overnight at 37 °C

123

(for chondroitinases). Then, agarose electrophoresis was performed, as previously described

124

[33]. In brief, 5 µL of each sample were applied to agarose gel slabs in 0.05 M 1,3-

125

diaminepropane acetate, pH 9.0. Afterward, GAGs in the gel slabs were precipitated for 1 h

126

with 0.2% cetyltrimethylammonium bromide (v/v), and then air-dried and stained with a

127

solution composed of 0.1% toluidine blue, 50% ethanol, and 1% acetic acid. The gel was

128

destained in a solution composed of 50% ethanol and 1% acetic acid and exposed to a

129

phosphor screen for visualization and quantification using a Cyclone® PhosphorImager. The

130

data, in counts per minute (cpm), were obtained by comparison with a standard radioactive

131

ink measured using a liquid scintillation counter (TRI-CARB 4910TR, Perkin-Elmer,

132

Waltham, Massachusetts, USA). The final value was normalized against total cell protein

133

content.

134

2.5 RNA extraction and real-time quantitative reverse transcription PCR (RT-

135

qPCR)

136

MDA-MB-231, MCF7, and SKBR3m cells were cultivated in 60-mm dishes until 90%

137

confluence was achieved. The medium was removed, and cells were incubated for 5 min with

138

1 mL TRIzolTM reagent. The lysate solution was transferred to tubes and mixed with 500 µL

139

chloroform by inversion. The tubes were centrifuged for 30 min, at 12,000 rpm, at 4 °C, and

140

the aqueous phase was transferred to a new tube. RNA was precipitated with 500 µL

141

isopropanol and sequentially washed with 1 mL 75% ethanol. The RNA pellet was left to dry

142

in a vacuum concentrator and resuspended in 40 µL RNase-free water.

143

Total RNA was quantified in a Nanodrop (NanoVueTM Plus, GE Healthcare, Chicago,

144

Illinois, USA), and a sample containing 2 µg total RNA and 2 µM oligo(dT)15 primers in 5 µL

145

RNase-free water was incubated at 70 °C for 5 min and then stored at 4 °C to minimize the

8 146

formation of RNA secondary structures. A 15 µL mixture, containing 667 µM dNTP mix, 4

147

mM MgCl2, 10.7 U/µL Improm-II reverse transcriptase, Improm-II reverse transcriptase

148

buffer (1.33×) and 1.33 U/µL Ribolock RNAse Inhibitor (Thermo Fisher Scientific), was

149

added to each sample. The reverse transcription reaction conditions were as follows:

150

annealing for 5 min at 25 °C, extension at 42 °C for 60 min, and enzyme inactivation at 70 °C

151

for 10 min.

152

cDNA was also quantified, and a quantitative PCR (qPCR) reaction mixture was

153

prepared to contain 50 ng/mL cDNA, 500 nM forward primers, 500 nM reverse primers, and

154

5 µL SYBR® Green PCR Master Mix (Applied Biosystems by Thermo Fisher), in a total

155

volume of 20 µL. The primers used in this assay are listed in Table 1. The qPCR conditions

156

included an activation phase for 10 min at 95 °C, and 40 cycles of denaturation (15 s at

157

95 °C), and annealing (1 min at 60 °C), with data collection occurring during the last step.

158

Table 1. Primers used in RT-qPCR.

Gene

Forward and reverse primers (5’-NNN-3’)

F: 5’-TGTGTGCTGTATCCTGAGAACA-3’ Dermatan sulfate epimerase 1 R: 5’-CAAGGCGCATCTTTCACCAC-3’ F: 5’-TTCCAAAGTCTATGCACAGGAGC-3’ Heparan sulfate epimerase R: 5’-TCCACATTGTAGCCTTCAAAAGAC-3’ F: 5’-TGGACCGTTTCAACAGAGAGGCTT 3’ Decorin proteoglycan R: 5’-TGGACCACTCGAAGATGGCATTGA-3’ Biglycan proteoglycan

F: 5’-AAGCTCAACTACCTGCGCATCTCA-3’

9 R: 5’-ATCATCCTGATCTGGTTGTGGCCT-3’ F: 5’-TAGCACTGCCCTTGGAATTTGTGC-3’ Versican proteoglycan R: 5’-AGCGGAGACCAGTGTGAACTTGAT-3’ F: 5’-TCGACAGTCAGCCGCATCTTCTTT-3’ GAPDH R: 5’-ACCAAATCCGTTGACTCCGACCTT-3’

159

2.6 Flow cytometry staining and data collection

160

MDA-MB-231, MCF7, and SKBR3m cells were cultivated in 100-mm dishes until

161

90% confluence was reached. The cells were detached using a 20-min incubation with 500

162

µM ethylenediaminetetraacetic acid (EDTA). The cells were collected by centrifugation,

163

distributed into tubes, and fixed with 200 µL 2% formaldehyde in PBS for 20 min, on ice.

164

The cells were washed sequentially with PBS and 0.1 M glycine and then permeabilized with

165

100 µL 0.01% saponin in PBS for 30 min on ice. After washing with PBS, the cells were

166

incubated with 50 µL antibody solution at 1:100 dilution for 1 h 30 min (anti-DS-epi1, anti-

167

HS-epi, or anti-decorin), diluted in 1% BSA in PBS. After washing with PBS, 50 µL

168

secondary antibody solution was added, containing either anti-rabbit Alexa Fluor 488 (1:200)

169

or anti-mouse Alexa Fluor 633 (1:200), for 40 min. The cells were resuspended in 200 µL

170

PBS and counted with a BD FACSCalibur (BD Biosciences). The fluorescence geometric

171

mean average of the cell population was used to compare flow cytometry data among cell

172

lines, and histograms were plotted for visual assessments.

173

2.7 Immunofluorescence staining and confocal imaging

174

MDA-MB-231, MCF7, and SKBR3m cells were plated at a 2×105 cell/mL density on

175

sterile borosilicate coverslips in a 24-well plate and incubated overnight. The supernatant was

10 176

removed, and cells were washed with cold PBS, followed by fixation with 4%

177

paraformaldehyde that was pre-warmed to 37 °C for 15 min. The coverslips were washed

178

three times with PBS, one time with 0.1 M glycine, and once again with PBS, for 5 min each

179

wash. The cells were then blocked and permeabilized with a solution containing 0.1% Triton

180

X-100 and 3% BSA for 1 h. Anti-DS-epi1 (rabbit IgG, 1:100) or anti-HS-epi (rabbit IgG,

181

1:150) were diluted in 0.05% Triton X-100 with 1% BSA in PBS were added to the cells and

182

incubated overnight at 4 °C. The coverslips were washed three times with PBS for 10 min and

183

incubated with anti-rabbit Alexa Fluor 488 (1:200) and phalloidin-647 (1:200) in 0.05%

184

Triton X-100 with 1% BSA in PBS, for 1 h at room temperature. The cells were washed three

185

times with PBS for 5 min and incubated with 4′,6-Diamidine-2′-phenylindole dihydrochloride

186

(DAPI, 1:2,000 in PBS) for 15 min. The coverslips were washed five times with PBS for five

187

minutes, dipped twice in cold type I water, mounted with FluoromountTM onto clean slides,

188

and sealed with colorless nail polish. For Golgi apparatus staining, the coverslips were

189

incubated with lectin HPA from Helix pomatia 647 (1:100) diluted in 0.05% Triton X-100

190

with 1% BSA in PBS for 1 h after the secondary antibody labeling, followed by washing three

191

times with PBS before DAPI staining and mounting.

192

The Leica TCS SP8 confocal microscope, with four laser beams (405 nm, 488 nm, 552

193

nm, and 638 nm), was used for image acquisition. Two sequential scans were performed at

194

400 Hz, first using the 405 nm laser (bandpass from 405–450 nm) and the 638 nm laser

195

(collection from > 640 nm), followed by a scan using the 488 nm laser (bandpass from 488–

196

550 nm). A control slide that was not stained with the primary antibody was used to set the

197

gain and offset values for each cell/antibody group.

198

The colocalization assessment between the Golgi staining and DS-epi1 was performed

199

in Huygens Professional 17.10.0 (Scientific Volume Imaging, Hilversum, The Netherlands).

200

Each of the five fields for the tested breast cancer cell lines had a region of interest (ROI)

11 201

generated in the red channel (i.e., the Golgi staining), with an established threshold. The

202

Costes method was applied for the background threshold prior to calculation of the Manders’

203

Overlap Coefficient (MOC), in which higher colocalization between DS-epi1 and the Golgi

204

staining is observed when MOC is nearest 1.

205

2.8 Data analysis and statistical treatment

206

Data were analyzed using GraphPad Prism 8.0 (San Diego, California, USA) and

207

Microsoft Office Excel 2016 (Redmond, Washington, USA). Different statistical tests were

208

applied, including one- and two-way analysis of variance (ANOVA), unpaired Student’s t-

209

test, and multiple Student’s t-tests. Statistically significant comparisons were set to the 5%

210

level, and data were plotted as the mean ± standard error of the mean (SEM).

211

3 RESULTS

212

3.1. SKBR3 cells grow more rapidly than MCF7 or MDA-MB-231

213

The initial aim of this study was to analyze and compare the expression of DS-epi1

214

among three different breast tumor cell lines, MCF7, MDA-MB-231 and SKBR3. However,

215

during routine cell culture, SKBR3 cells clearly demonstrated different growth behavior

216

compared with the other cell lines, which was indicated by the cell MTT assay. This assay

217

was performed using MDA-MB-231, MCF7, and SKBR3 cells at 24-, 48-, 72-, and 96-h time

218

points, as shown in Figure 1. The results clearly indicated that the SKBR3 cell line showed

219

increased OD570, with an exponential growth phase after 48 h, when compared with the other

220

tested cell lines. MDA-MB-231 and MCF7 cells showed similar growth curves, with a slow

221

growth curve that entered a saturation phase after 72 h, in contrast with SKBR3 cells. This

222

experiment was unexpected because MDA-MB-231 cells are classified as triple-negative (ER-

223

/PR-/HER2-) cells, which are commonly associated with increased genetic instability and

224

aggressiveness and a worse prognosis [34].

12 225

3.2. One variant of SKBR3 cells shows an entirely different growth curve and

226

expresses a different pattern of GAGs

227

These results led us to suspect that the SKBR3 cells used in this study (Figure 1) could

228

be modified (SKBR3m), as it was used at high passage levels (> 50). In an attempt to further

229

investigate this characteristic, a recently acquired ATCC vial (SKBR3a) was used, and the

230

cells were compared. The MTT assay was employed to assay the growth behavior of both

231

variants, using three time points, 24, 48 and 72 h (Figure 2). SKBR3m cells displayed erratic

232

growth, showing an exponential pattern, in contrast with the linear growth observed for

233

SKBR3a cells. This result suggested that SKBR3m may represent an aggressive variant of

234

SKBR3 cells.

235

We then investigated the GAG distributions in both SKBR3 cells, as shown in Figure

236

3. Apparent differences in CS and DS distributions were observed between SKBR3a and

237

SKBR3m cells. Although the CS content was low in SKBR3m cells, the DS content was high,

238

which was the opposite pattern of that observed for SKBR3a cells, where CS was expressed at

239

high levels, whereas DS was difficult to detect.

240

Therefore, we hypothesized that the different CS/DS contents might be associated with

241

the apparent difference in the growth curves between SKBR3m and SKBR3a cells, and we

242

proposed to investigate the role played by DS-epi1 in SKBR3m cells. All further

243

investigations utilized SKBR3m cells in all biological assays used to compare these cells with

244

other breast cancer cell lines.

245

3.3 Dermatan Sulfate epimerase 1 is expressed at higher levels in SKBR3m than

246

in other tested cell lines.

247

The expression level of DS-epi1 was investigated in MCF7 (luminal), MDA-MB-231

248

(triple-negative), and SKBR3m (HER2-positive) breast cancer cell lines. qPCR was used to

13 249

examine the expression levels of DS-epi1, the principal DS PGs, decorin, and biglycan

250

(Figure 4), relative to glyceraldehyde 3-phosphate dehydrogenase (GAPDH) expression

251

levels. SKBR3m cells expressed significantly higher levels of DS-epi1 (p < 0.0001) than

252

either MDA-MB-231 (p = 0.003) or MCF7 (p = 0.0002) cells, according to Dunnett’s test. A

253

multiple Student’s t-test approach revealed that MCF7 cells expressed less DS-epi1 and

254

decorin than MDA-MB-231 (p < 0.0001) and SKBR3m (p = 0.0204) cells, indicating a

255

relationship between this PG and epiDS expression. Heparan sulfate epimerase (HS-epi)

256

expression was analyzed as a control and showed an independent pattern compared with

257

either DS-epi1 or decorin, as expected (Figure 4).

258

3.4. SKBR3m cells have lower protein levels of Dermatan Sulfate epimerase 1

259

than the other cell lines

260

The flow cytometry analysis was performed using specific antibodies to quantify the

261

protein levels of DS-epi1, HS-epi and decorin (Figure 5). Unexpectedly, MCF7 cells showed

262

higher protein expression levels of DS-epi1 than SKBR3m and MDA-MB-231 cells.

263

However, no differences in the protein expression levels of HS-epi were observed when

264

comparing the three breast cancer cell lines. MCF7 and SKBR3m cells displayed higher

265

expression levels of decorin than MDA-MB-231 cells.

266

The dot plot graph (Figure 6) allowed us to verify the existence of different cell

267

populations distributed among the studied cell lines, emphasizing a double-positive

268

population (decorin+/DS-epi1+). MCF7 cells contained the most double-positive cells (Q2 =

269

96.4%), followed by SKBR3m (Q2 = 88.8%) and MDA-MB-231 (Q2 = 86.1%) cells. The

270

existence of a decorin+/HS-epi+ population was invariable among the cell lines, with

271

approximately 90% of cells comprising this double-positive population.

14 272

3.5 Dermatan Sulfate epimerase 1 localizes to the Golgi apparatus in SKBR3m

273

cells but little in other cell lines

274

Immunofluorescence assays and confocal microscopy of MDA-MB-231, MCF7, and

275

SKBR3m cells showed the subcellular localizations of DS-epi1 and HS-epi proteins. Cells

276

were also labeled with DAPI and phalloidin-647 to identify the nucleus and the cytoskeleton,

277

respectively, as shown in Figure 7. Immunofluorescence quantification was analyzed by

278

ANOVA, with the Holm Šidák test, and the data revealed that SKBR3m cells presented the

279

lowest DS-epi1 protein contents among the three tested cell lines, whereas MCF7 cells had

280

the highest content (Figure 8), in contrast with the qPCR data.

281

The results also indicated a cytoplasmic localization for DS-epi1 in MDA-MB-231

282

and MCF7 cells, whereas in SKBR3m cells, the localization of this enzyme appears to be

283

perinuclear, with some vesicular stainings in the cytoplasm, suggesting Golgi apparatus

284

localization (Figure 7).

285

A more diffuse and cytoplasmic distribution for HS-epi was observed in MCF7 and

286

MDA-MB-231 cells (Figure 7), whereas localization to the Golgi apparatus was visualized in

287

SKBR3m cells, similar to the localization pattern observed for DS-epi1 (Figure 9).

288

An additional assay employing a Golgi apparatus-specific marker, the lectin HPA

289

from Helix Pomatia 647, was used to verify the specific localization of DS-epi1 in this

290

organelle, whose results can be viewed in Figure 9. This experiment revealed that MCF7 and

291

MDA-MB-231 cells express DS-epi1 primarily in the cytoplasm, whereas DS-epi1 is strictly

292

contained within the Golgi apparatus in SKBR3m cells, which is an organelle involved in the

293

post-translational processing of proteins (Figure 9A).

294

The colocalization analysis between the Golgi staining and DS-epi1 indicated that

295

SKBR3m cells had the highest degree of colocalization of this epimerase in the analyzed

15 296

organelle (0.863 ± 0.007), as shown in Figures 9B and 9C. Although MCF7 and MDA-MB-

297

231 displayed a certain degree of colocalization between both channels, it was 34.8% and

298

18.1% less intense, respectively, in comparison to SKBR3m.

299

3.6 SKBR3m cells, but not MCF7 and MDA-MB-231 cells, synthesize DS

300

After evaluating that DS-epi1 was mainly in the Golgi Apparatus of SKBR3m cells,

301

while in MCF7 and MDA-MB-231, the enzyme was mislocalized to the cytoplasm, it would

302

be worth to investigate the DS synthesis in these cells. To this end, metabolic labeling with

303

Na235SO4 was performed in these cell lines, and the total 35S-GAGs obtained. The GAGs

304

were then incubated with the specific chondroitinase AC and chondroitinase ABC.

305

Chondroitinase AC hydrolyzes β-(1-4) glycosidic bonds between N-acetyl-hexosamine and

306

glucuronic acid. Therefore, it is capable of hydrolyzing chondroitin-4-sulfate, chondroitin-6-

307

sulfate and hyaluronic acid to its unsaturated disaccharides-4-sulfated (∆U-GalNAc4S), -6-

308

sulfated (∆U-GalNAc6S) and non-sulfated (∆U-GlcNAc), respectively. Chondroitinase ABC

309

hydrolyzes β-(1-4) glycosidic bonds between N-acetyl-hexosamine and GlcA or IdoA acid. It

310

hydrolyzes chondroitin-4-sulfate, chondroitin-6-sulfate (∆U-GalNAc4S and ∆U-GalNAc6S),

311

DS (∆U-GalNAc4S) and hyaluronic acid (∆U-GlcNAc). After incubation with these enzymes,

312

the products were subjected to agarose gel electrophoresis, as shown in Figure 10.

313

Agarose gel electrophoresis can retain only larger polymers, not the digested products

314

by the enzymes. Disaccharides are unable to be identified in this experiment. Therefore, DS is

315

hydrolyzed by chondroitinase ABC but is resistant to chondroitinase AC so that this specific

316

GAG can be visualized in this gel. HS is resistant to both chondroitinases AC and ABC, so it

317

is detected in its intact form in this gel, as can be seen in Figure 10. The agarose gel

318

electrophoresis was able to identify a DS band only in SKBR3m cells, which does not occur

319

in MCF7 or MDA-MB-231 cells.

16 320

4 DISCUSSION

321

Breast cancer is a disease that currently affects a large number of people, primarily

322

females, and can be highly lethal, affecting individuals of all age groups [1, 35]. DS-epi1 is

323

the limiting enzyme for DS biosynthesis and acts on the GluA residues in CS chains in the

324

Golgi apparatus, converting them to IdoA [24, 26]. DS is a GAG that plays essential roles in

325

the modulation of oncogenic mechanisms, such as acting as a co-receptor for various growth

326

factors, including hepatocyte growth factor (HGF), fibroblast growth factor type 2 (FGF-2),

327

and platelet-derived growth factor (PDGF), all of which are crucial for the progression of

328

breast tumors. Its function as co-receptor is due to the existence of IdoA residues, which

329

confer greater flexibility to the molecule and favor increased binding specificity to these

330

growth factors [36].

331

We investigated the properties of DS-epi1 in three breast cancer cell lines: MCF7

332

(luminal A, ER+/PR+), MDA-MB-231 (triple-negative) and SKBR3 (HER2+). DS has been

333

shown to play a crucial aspect both in MCF7 and MDA-MB-231. In MDA-MB-231, the cells'

334

ability to synthesize full chains of CS/DS would affect these cells’ viability, motility, and

335

adhesion to matrix substrates [37]. Regarding MCF7, the ability of these cells to interact with

336

DSPGs, such as decorin or biglycan, would influence its viability and how the cells would

337

interact with growth factors, such as the vascular endothelial growth factor-165 (VEGF165)

338

[36]. We, therefore, selected both these cell lines to research the aspects of DS-epi1 but felt

339

the need to add a HER2+ cell line, as HER2 positivity represents 15–20% of breast cancers

340

[38]. SKBR3 was selected as it is one of the most widely used HER2+ cell lines [39].

341

We analyzed the localization of DS-epi1 in these breast cancer cells and assessed its

342

product, DS. We verified that the SKBR3m breast cancer cell line showed higher gene

343

expression levels of DS-epi1 than the other breast cancer cell lines (MCF7 and MDA-MB-

17 344

231). DS-epi1 has been found to be overexpressed in glioblastomas, as assessed by tissue

345

immunohistochemistry [40]. The same study further demonstrated that DS-epi1

346

overexpression was associated with advanced tumor grade and poor survival. DS-epi1

347

knockdown in glioblastoma cells demonstrated that this enzyme was responsible for

348

suppressing cell proliferation, migration, and invasion. In contrast, the induction of DS-epi1

349

overexpression increased malignant phenotypes and tumoral growth in vivo [40]. DS-epi1 is

350

also highly expressed in esophageal squamous cell carcinoma cells, which exhibit a four to

351

five-fold overexpression in enzyme activity compared with healthy tissue. In this tumor type,

352

DS-epi1 was localized both in the stroma surrounding the tumor and in the tumor cells, and

353

high DS-epi1 expression was also shown to be correlated with increased DS production [41].

354

Other studies have demonstrated that the overexpression of DS in esophageal squamous cell

355

carcinoma cells acts synergistically with HGF by stimulating tumoral migration and invasion.

356

This effect may be the result of the co-receptor abilities of DS with HGF and its respective

357

cellular receptor (c-Met), potentiating the growth factor’s effects [42].

358

Curiously, in our work, MCF7 cells presented higher DS-epi1 protein expression

359

levels than the other cell lines, although this effect was expected for the SKBR3m cell line

360

due to the higher gene expression of this enzyme in this specific cell line. The MCF7 and

361

SKBR3m cell lines both showed significantly increased expression levels of the decorin PG

362

core protein, as detected by flow cytometry.

363

We also examined DS-epi1 localization in SKBR3m, MCF7, and MDA-MB-231 cell

364

lines, using immunofluorescence assays with specific antibodies and confocal microscopy.

365

The SKBR3m cell line was the only cell line to show DS-epi1 content in the perinuclear

366

compartment, suggesting a location to the Golgi apparatus. This localization was further

367

confirmed by the immunofluorescence co-localization assay between the DS-epi1 enzyme and

368

Lectin-HPA (Helix pomatia agglutinin), a specific cellular marker for the Golgi apparatus.

18 369

The MCF7 and MDA-MB-231 cell lines showed cytoplasmic localization of the enzyme,

370

which could explain the increased detection of DS-epi1 by flow cytometry and quantitative

371

immunofluorescence.

372

From the literature, we know that the biosynthesis of GAGs, such as CS, occurs in the

373

endoplasmic reticulum and Golgi apparatus, starting with the addition of a tetrasaccharide and

374

followed by the consecutive addition of hexosamine and a non-nitrogen sugar. For the

375

formation of CS, these sugars correspond with N-acetylgalactosamine and glucuronic acid,

376

respectively. The formation of DS chains involves the conversion of GluA in CS chains to

377

IdoA, through the activity of the DS-epi1 enzyme [21-23]. Active DS-epi1 requires an

378

optimal pH of 5.5 and the presence of divalent cations, especially Mn²⁺ and the weakly

379

activating Ca²⁺ and Mg²⁺ cations. These conditions can be found within the Golgi apparatus,

380

which favors maximal enzyme activity in this organelle [43]. Nevertheless, previous studies

381

have observed that a mere change of 0.2–0.4 pH units in the Golgi luminal pH may promote

382

the mislocalization of several Golgi enzymes, such as α(2,3)-sialyltransferase, to endosomal

383

compartments [44]. Other glycosylation enzymes have also faced similar mislocalization to

384

endosomes or the plasma membrane in similar conditions [45, 46]. MCF7 has been shown to

385

be unable to acidify the Golgi lumen, leading to its alkalinization and mislocalization of

386

several of this organelle’s resident enzymes; a context also investigated in selected colorectal

387

cell lines (e.g., HT-29, SW-48, T84 and Caco-2) [47, 48]. Although there have been no

388

researches regarding how DS-epi1 localization may be affected by alkalinization of the Golgi

389

Apparatus, it is of our belief that certain cancer cell lines which may face it, may lose the

390

epimerization process related to DS synthesis, as suggested here.

391

Among the cell lines examined, SKBR3m was the only cell line containing DS after 35

392

the metabolic labeling of

S-GAGs and characterization with chondroitinase enzymes. As

393

mentioned earlier, DS is implicated in various cancer-promoting processes, such as cell

19 394

proliferation and metastasis. Our data showed increased SKBR3m cell growth, which differed

395

from the other cell lines analyzed, leading us to believe that this phenomenon could be

396

associated with the presence of DS.

397

Our results showed higher gene expression levels for DS-epi1 in SKBR3m cells and

398

the specific subcellular localization of the enzyme to the Golgi apparatus and the presence of

399

35

400

pattern. Our results showed that the localization of DS-epi1 in the Golgi apparatus is of great

401

importance for DS synthesis in breast cancer cell lines, suggesting that in cells where this

402

localization was not observed (e.g., cytoplasmic localization), the formation of DS chains

403

would be compromised. Thus, we suggest that the synthesis of DS could be associated with

404

breast cancer cell growth. The investigation of SKBR3 cells in the present study, which

405

resulted in the identification of the modified cell line, was fundamental to these results. The

406

changes in increased growth, combined with the enhanced synthesis of DS, a compound that

407

has been directly associated with oncogenicity, presents the opportunity to study interactions

408

between DS and critical proteins in the tumor microenvironment (e.g., growth factors,

409

chemokines, and cytokines), and we believe that DS deserves better characterization. The

410

importance of this study is to present a suitable model for breast cancer tumorigenesis and to

411

investigate new therapeutic targets that could be related to the “switch” in DS synthesis

412

observed in this study.

413

CONCLUSION

S-DS in these cells, which also demonstrated a more pronounced cellular proliferation

414

Our results showed that, in SKBR3m cells, the Golgi localization of DS-epi1 was a

415

determinant for DS synthesis. In the MCF7 and MDA-MB-231 cell lines, the cytoplasmic

416

localization of DS-epi1 may be associated with the lack of DS synthesis. However, the

417

decorin PG core protein was still synthesized, especially in SKBR3m and MCF7 cells.

20 418

Moreover, SKBR3m cells were the most viable cell line, and the presence of DS in this cell

419

line may contribute to the proliferative aspects in this particular in vitro model.

420

CONFLICT OF INTEREST

421

422

The authors report no conflict of interest in the content presented in this article.

ACKNOWLEDGMENTS

423

The authors acknowledge Caroline Z. Romera and Elizabeth N. Kanashiro

424

(INFAR/UNIFESP, Brazil) for their confocal technical assistance.

425

FUNDING

426

This work was supported by Fundação de Amparo à Pesquisa do Estado de São

427

Paulo (FAPESP, grant number 2016/18066-6 to LT), Conselho Nacional de Desenvolvimento

428

Científico e Tecnológico (CNPq, fellowship to EGX) and, in part, by the Coordenação de

429

Aperfeiçoamento de Pessoal de Nível Superior — Brasil (CAPES) — Finance Code 001,

430

including a fellowship to EL.

431

REFERENCES

432

[1] C. DeSantis, J. Ma, L. Bryan, A. Jemal, CA: a cancer journal for clinicians, 64 (2014) 52-

433

62.

434

[2] W.F. Anderson, N. Chatterjee, W.B. Ershler, O.W. Brawley, Breast cancer research and

435

treatment, 76 (2002) 27-36.

436

[3] K.A. Cadoo, M.N. Fornier, P.G. Morris, The quarterly journal of nuclear medicine and

437

molecular imaging : official publication of the Italian Association of Nuclear Medicine, 57

438

(2013) 312-321.

21 439

[4] A.D. Theocharis, S.S. Skandalis, T. Neill, H.A. Multhaupt, M. Hubo, H. Frey, S. Gopal,

440

A. Gomes, N. Afratis, H.C. Lim, J.R. Couchman, J. Filmus, R.D. Sanderson, L. Schaefer,

441

R.V. Iozzo, N.K. Karamanos, Biochimica et biophysica acta, 1855 (2015) 276-300.

442

[5] X.L. Jia, S.Y. Li, S.S. Dang, Y.A. Cheng, X. Zhang, W.J. Wang, C.E. Hughes, B.

443

Caterson, World journal of gastroenterology, 18 (2012) 3962-3976.

444

[6] J.R. Couchman, C.A. Pataki, The journal of histochemistry and cytochemistry : official

445

journal of the Histochemistry Society, 60 (2012) 885-897.

446

[7] A. Malmstrom, B. Bartolini, M.A. Thelin, B. Pacheco, M. Maccarana, The journal of

447

histochemistry and cytochemistry : official journal of the Histochemistry Society, 60 (2012)

448

916-925.

449

[8] R.V. Iozzo, R.D. Sanderson, Journal of cellular and molecular medicine, 15 (2011) 1013-

450

1031.

451

[9] C.M. Willis, M. Kluppel, PloS one, 9 (2014) e103966.

452

[10] T.Y. Eshchenko, V.I. Rykova, A.E. Chernakov, S.V. Sidorov, E.V. Grigorieva,

453

Biochemistry. Biokhimiia, 72 (2007) 1016-1020.

454

[11] P. du Souich, A.G. Garcia, J. Verges, E. Montell, Journal of cellular and molecular

455

medicine, 13 (2009) 1451-1463.

456

[12] K. Sugahara, H. Kitagawa, Current opinion in structural biology, 10 (2000) 518-527.

457

[13] T. Miller, M.C. Goude, T.C. McDevitt, J.S. Temenoff, Acta biomaterialia, 10 (2014)

458

1705-1719.

22 459

[14] K. Sugahara, T. Mikami, T. Uyama, S. Mizuguchi, K. Nomura, H. Kitagawa, Current

460

opinion in structural biology, 13 (2003) 612-620.

461

[15] T. Uyama, H. Kitagawa, K. Sugahara, in: J.P. Kamerling (Ed.) Comprehensive

462

Glycoscience, Elsevier, Amsterdam, 2007, pp. 79-104.

463

[16] J.E. Silbert, G. Sugumaran, IUBMB life, 54 (2002) 177-186.

464

[17] G. Dick, L.K. Akslen-Hoel, F. Grondahl, I. Kjos, K. Prydz, The journal of histochemistry

465

and cytochemistry : official journal of the Histochemistry Society, 60 (2012) 926-935.

466

[18] C. Herzog, I. Lippmann, K. Grobe, A.D. Zamfir, F. Echtermeyer, D.G. Seidler,

467

Molecular bioSystems, 7 (2011) 2872-2881.

468

[19] M.A. Pinhal, B. Smith, S. Olson, J. Aikawa, K. Kimata, J.D. Esko, Proceedings of the

469

National Academy of Sciences of the United States of America, 98 (2001) 12984-12989.

470

[20] K. Cuellar, H. Chuong, S.M. Hubbell, M.E. Hinsdale, The Journal of biological

471

chemistry, 282 (2007) 5195-5200.

472

[21] C. Malavaki, S. Mizumoto, N. Karamanos, K. Sugahara, Connective tissue research, 49

473

(2008) 133-139.

474

[22] K. Sugahara, T. Mikami, Current opinion in structural biology, 17 (2007) 536-545.

475

[23] L. Djerbal, H. Lortat-Jacob, J. Kwok, Glycoconjugate journal, 34 (2017) 363-376.

476

[24] B. Pacheco, A. Malmstrom, M. Maccarana, The Journal of biological chemistry, 284

477

(2009) 9788-9795.

478

[25] J.F. Lensen, J. van der Vlag, E.M. Versteeg, J.F. Wetzels, L.P. van den Heuvel, J.H.

479

Berden, T.H. van Kuppevelt, A.L. Rops, PloS one, 10 (2015) e0134946.

23 480

[26] M. Maccarana, B. Olander, J. Malmstrom, K. Tiedemann, R. Aebersold, U. Lindahl, J.P.

481

Li, A. Malmstrom, The Journal of biological chemistry, 281 (2006) 11560-11568.

482

[27] M. Maccarana, S. Kalamajski, M. Kongsgaard, S.P. Magnusson, A. Oldberg, A.

483

Malmstrom, Molecular and cellular biology, 29 (2009) 5517-5528.

484

[28] G. David, B. Van der Schueren, M. Bernfield, Journal of the National Cancer Institute,

485

67 (1981) 719-728.

486

[29] A.J. Butt, J.L. Martin, K.A. Dickson, F. McDougall, S.M. Firth, R.C. Baxter, The Journal

487

of clinical endocrinology and metabolism, 89 (2004) 1950-1956.

488

[30] P. Krief, C. Saint-Ruf, M. Bracke, C. Boucheix, C. Billard, M. Billard, R. Cassingena, C.

489

Jasmin, M. Mareel, B. Azzarone, International journal of cancer, 43 (1989) 658-664.

490

[31] L. Wang, Z.Y. Wang, Oncology reports, 23 (2010) 1109-1117.

491

[32] D.M. Morgan, Methods in molecular biology, 79 (1998) 179-183.

492

[33] C.P. Dietrich, S.M. Dietrich, Analytical biochemistry, 70 (1976) 645-647.

493

[34] H. Pan, W. Zhou, W. He, X. Liu, Q. Ding, L. Ling, X. Zha, S. Wang, International

494

journal of molecular medicine, 30 (2012) 337-343.

495

[35] J. Ferlay, I. Soerjomataram, R. Dikshit, S. Eser, C. Mathers, M. Rebelo, D.M. Parkin, D.

496

Forman, F. Bray, International journal of cancer, 136 (2015) E359-386.

497

[36] E.M. Kozma, G. Wisowski, M. Latocha, D. Kusz, K. Olczyk, Experimental biology and

498

medicine, 239 (2014) 1575-1588.

499

[37] M. Viola, K. Bruggemann, E. Karousou, I. Caon, E. Carava, D. Vigetti, B. Greve, C.

500

Stock, G. De Luca, A. Passi, M. Gotte, Glycoconjugate journal, 34 (2017) 411-420.

24 501

[38] S. Loibl, L. Gianni, Lancet, 389 (2017) 2415-2429.

502

[39] M. Lacroix, G. Leclercq, Breast cancer research and treatment, 83 (2004) 249-289.

503

[40] W.C. Liao, C.K. Liao, Y.H. Tsai, T.J. Tseng, L.C. Chuang, C.T. Lan, H.M. Chang, C.H.

504

Liu, PloS one, 13 (2018) e0198364.

505

[41] M.A. Thelin, K.J. Svensson, X. Shi, M. Bagher, J. Axelsson, A. Isinger-Ekstrand, T.H.

506

van Kuppevelt, J. Johansson, M. Nilbert, J. Zaia, M. Belting, M. Maccarana, A. Malmstrom,

507

Cancer research, 72 (2012) 1943-1952.

508

[42] J. Villena, E. Brandan, Journal of cellular physiology, 198 (2004) 169-178.

509

[43] A. Malmstrom, The Biochemical journal, 198 (1981) 669-675.

510

[44] A. Rivinoja, A. Hassinen, N. Kokkonen, A. Kauppila, S. Kellokumpu, Journal of cellular

511

physiology, 220 (2009) 144-154.

512

[45] M.A. Axelsson, N.G. Karlsson, D.M. Steel, J. Ouwendijk, T. Nilsson, G.C. Hansson,

513

Glycobiology, 11 (2001) 633-644.

514

[46] B.E. Schaub, B. Berger, E.G. Berger, J. Rohrer, Molecular biology of the cell, 17 (2006)

515

5153-5162.

516

[47] A. Rivinoja, N. Kokkonen, I. Kellokumpu, S. Kellokumpu, Journal of cellular

517

physiology, 208 (2006) 167-174.

518

[48] S. Kellokumpu, R. Sormunen, I. Kellokumpu, FEBS letters, 516 (2002) 217-224.

519

25 520

FIGURE CAPTIONS

521

Figure 1. SKBR3m cells grow faster than MDA-MB-231 and MCF7 cells. The MTT assay

522

was performed at 24, 48, 72, and 96 h. The sample size was n = 16. Data were analyzed by

523

two-way ANOVA with Dunnett’s test. In this particular graph, data is displayed as mean ±

524

standard deviation (SD). * p < 0.05 vs. MDA-MB-231 and MCF7; **** p < 0.0001 vs. MDA-

525

MB-231 and MCF7.

526

Figure 2. The variant SKBR3m grows faster than SKBR3a. The MTT assay was

527

performed at 24, 48, and 72 h. For each time point, an unpaired Student’s t-test was applied to

528

compare OD570 scores between variants. The sample size was n = 24. In this particular graph,

529

data is displayed as mean ± SD. A linear fit was obtained for the variant SKBR3a (R2 =

530

0.928), and an exponential curve was fitted for SKBR3m (R2 = 0.934). * p < 0.05;**** p <

531

0.0001 vs. SKBR3 variants.

532

Figure 3. SKBR3m, and not SKBR3a, synthesizes DS and displays a GAG profile in the

533

cellular medium. (A) For each SKBR3 variant, the

534

extract (C) or medium (M), and after electrophoresis, the CS, DS, and HS contents were

535

analyzed. (B) Each GAG content was quantified and normalized against the total protein

536

quantity in the cell. No GAGs were detected in the medium from SKBR3a cells.

537

Figure 4. SKBR3m cells have a higher gene expression of DS-epi1 and decorin, in

538

comparison to MCF7 and MDA-MB-231 cells. All genes were assessed by RT-qPCR, in

539

which the sample size was n = 5–6. Data were analyzed by two-way ANOVA, with Dunnett’s

540

test. **** p < 0.0001 SKBR3m vs. MCF7; #### p < 0.0001 SBR3 vs. MDA-MB-231; and

541

$$$$ p < 0.0001 MCF7 vs. MDA-MB-231.

542

Figure 5. Concerning the protein expression, MCF7 displays higher DS-epi1 expression,

543

although both MCF7 and SKBR3m cells have high expression of decorin. The flow

35

S-GAGs were assessed in the cell

26 544

cytometry assay was used to analyze the expression profiles of DS-epi1, HS-epi, and decorin

545

proteoglycan. The sample size was n = 4–8. (A) Data were analyzed by two-way ANOVA,

546

with the Holm Šidák test. * p < 0.05; **** p < 0.0001 vs. MDA-MB-231. (B) The histograms

547

show cell population distributions in the flow cytometry experiment, in which unfilled

548

histograms are negative controls from unstained samples.

549

Figure 6. MCF7 and SKBR3m have a higher population of cells that both express DS-

550

epi1 and decorin. The flow cytometry assay analyzed DS-epi1, HS-epi, and decorin

551

proteoglycan expression. Decorin proteoglycan was stained with Alexa 633 and is shown in

552

the y-axis, and either DS-epi1 or HS-epi was labeled with Alexa 488 and is displayed on the

553

x-axis. In red, a representative experimental sample is shown for each cell line; and in blue, a

554

negative control.

555

Figure 7. SKBR3m has a more perinuclear localization of DS-epi1, as opposed to the

556

cytoplasmic display in MDA-MB-231 and MCF7. The immunofluorescence assay was

557

performed in MDA-MB-231 (a and d), MCF7 (b and e) and SKBR3m (c and f) cells

558

assessing DS-epi-1 (a, b or c) or HS-epi (d, e, or f) protein content. Cells were stained with

559

DAPI (1) or F-actin (3), in addition to DS-epi1 or HS-epi (2) labeling. The composite image

560

(4) is also exhibited. The scale bar represents 200 µm, and 7 to 9 fields were analyzed.

561

Figure 8. Although SKBR3m has a more precise localization of DS-epi1, MCF7 displays

562

a higher quantity of it. The data from the immunofluorescence assay was quantified for DS-

563

epi1 and HS-epi content. The sample size was n = 7–9. Data were analyzed by two-way

564

ANOVA, with the Holm Šidák test. ** p < 0.01 vs. MDA-MB-231; ### p < 0.001 vs. MCF7.

565

Figure 9. SKBR3m cells have a higher Golgi colocalization of DS-epi1 than MCF7 and

566

MDA-MB-231. (A) Cells were imaged for DS-epi1 (2) and a Golgi apparatus cellular marker,

567

(lectin HPA, 3). The cells were also stained with DAPI (1). All channels were merged (4), and

27 568

the scale bar represents 50 µm. (B) The representatives' histograms display the colocalization

569

between pixels from the red channel (lectin HPA) and the green channel (DS-epi1) for each

570

tested cell line. (C) The Manders’ Overlap Coefficient (MOC) indicates the degree of

571

colocalization between DS-epi1 with the lectin HPA (i.e., the Golgi marker) for each tested

572

cell line. Data were analyzed with multiple Student’s t-tests. * p < 0.05, **** p < 0.0001 vs.

573

SKBR3m.

574

Figure. 10. Only SKBR3m synthesize DS when compared to MDA-MB-231 and MCF7

575

cells. The assay was performed by labeling GAGs with 35S, purifying them with ethanol, and

576

proteolytically degrading the PG core protein with alcalase. The products were then digested

577

with chondroitinase AC or ABC and separated by electrophoretic mobility in agarose gels in

578

PDA buffer. Only SKBR3m cells displayed DS content, differing from the other cell lines that

579

only contained HS and CS (not shown).

HIGHLIGHTS • • • •

A SKBR3 cell line from a high passage (SKBR3m) is highly proliferative in comparison to MCF7 and MDA-MB-231 cells; The SKBR3m cells were the only to synthesize dermatan sulfate; Dermatan sulfate epimerase 1 (DS-epi1) was found in the Golgi Apparatus only in SKBR3m cells; MCF7 and MDA-MB-231 cells presented DS-epi1 mislocalized.

Declaration of interests ☒ The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper. ☐The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: