Proteomics Study Reveals That Docosahexaenoic and Arachidonic

1 hour ago - ... ACS Macro Letters · ACS Medicinal Chemistry Letters · ACS Nano · ACS Omega · ACS Pharmacology & Translational Science - New in 2018 ...
0 downloads 0 Views 2MB Size
Subscriber access provided by UNIV OF CAMBRIDGE

Bioactive Constituents, Metabolites, and Functions

Proteomics Study Reveals That Docosahexaenoic and Arachidonic Acids Exert Different in Vitro Anticancer Activities in Colorectal Cancer Cells Ignacio Ortea, Maria Jose Gonzalez-Fernandez, Rebeca P Ramos-Bueno, and Jose Luis Guil Guerrero J. Agric. Food Chem., Just Accepted Manuscript • DOI: 10.1021/acs.jafc.8b00915 • Publication Date (Web): 26 May 2018 Downloaded from http://pubs.acs.org on May 26, 2018

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 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 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.

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 36

Journal of Agricultural and Food Chemistry

1 2

Proteomics Study Reveals That Docosahexaenoic and

3 4

Arachidonic Acids Exert Different in Vitro Anticancer Activities

5

in Colorectal Cancer Cells

6 7

Ignacio Orteaa*, María José González-Fernándezb, Rebeca P. Ramos-Buenob,

8

and José Luis Guil-Guerrerob

9

a

IMIBIC, Hospital Universitario Reina Sofía, Universidad de Córdoba, E14004, Córdoba,

10

Spain. b

11 12

Food Technology Division, Agrifood Campus of International Excellence, ceiA3, University of Almería, E40120, Almería, Spain

13 14

* Corresponding author, [email protected]; Phone +34 957 213 757

15 16

1

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

17 18

ABSTRACT Two PUFAs, DHA and ARA, as well as derivatives such as eicosanoids, regulate

19

different activities, affecting transcription factors and therefore DNA transcription, being a

20

critical step for the functioning of FA-derived signalling. This work has attempted to

21

determine the in vitro anticancer activities of these molecules linked to the gene

22

transcription regulation of HT-29 colorectal cancer cells. We applied 3-(4,5-

23

dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) test along with lactate-

24

dehydrogenase (LDH) and caspase-3 assays; proteome changes were assessed by

25

‘sequential windowed acquisition of all theoretical mass spectra’ (SWATH-MS)

26

quantitative proteomics followed by pathway analysis in order to determine the affected

27

molecular mechanisms. In all assays, DHA inhibited cell proliferation of HT-29 cells to a

28

higher extent than ARA and acted primarily by down-regulating proteasome particles,

29

while ARA presented a dramatic effect on all six DNA replication helicase particles. The

30

results indicated that both DHA and ARA are potential chemopreventive agent candidates.

31 32 33

KEY WORDS ARA; colorectal cancer; DHA; proteomics; PUFA; SWATH

34

2

ACS Paragon Plus Environment

Page 2 of 36

Page 3 of 36

35 36

Journal of Agricultural and Food Chemistry

INTRODUCTION Colorectal cancer (CRC) has one of the highest mortality rates. The fact that large

37

regional differences exist in the incidence and mortality of CRC suggests a close

38

relationship between genetic and environmental factors, with diet and lifestyle being two of

39

the most determinant for CRC incidence. Diet is widely recognised as a critical factor for

40

cancer risk and mortality.1

41

Free fatty acids (FFAs) assume a critical part in many biological roles. Apart from

42

serving as precursors of a wide range of signalling and structural molecules, they are used

43

to obtain energy.2 Although many of the circulating FFAs are bound to albumin, the small

44

fraction of unbound FFAs in the aqueous phase mediate physiological activity and are most

45

sensitive to changes in health and disease; they are also transported across membranes.3

46

Previous data have recognised the importance of certain polyunsaturated FAs (PUFAs)

47

for the prevention of CRC, while the results from epidemiological studies have suggested a

48

negative correlation between both CRC development and PUFA intake.4 Some studies have

49

also found that, in vitro, some PUFAs increase the cytotoxicity of different

50

chemotherapeutics in brain, lung, breast, sarcoma, lymphocytic and colon human cell

51

cultures.5 The degree of in vitro anticancer activity of various FAs has been related to their

52

chemical structure, both of which increase such actions. Montaung et al.6 evaluated the

53

cytotoxic capacity of most of the FAs of interest in a combined manner in prostate cancer

54

cells.

55

n-3 PUFAs have been correlated with CRC preventive effects while n-6 PUFAs have

56

shown a positive association with CRC, although some authors have undermined this

57

archetype by proposing more complex functions for PUFAs.7,8 Because of the nutrition and

3

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

58

health claims made on n-3 PUFAs, they are widely available in a ‘nutraceutical’

59

formulations.9 Although n-6 PUFAs show a positive association with CRC risk,9 some

60

studies have also found that arachidonic acid (ARA, 20:4n6) inhibits the growth of some

61

cancer cell lines, such as Caco-2, but not HT-29.10 In addition, eicosanoids, biologically

62

active molecules derived from ARA, have been proposed as mediators involved in

63

intestinal epithelial homeostasis; other studies have linked hydroxyeicosatetraenoic acids

64

(HETEs), metabolised from ARA by the lipoxygenase (LOX) pathway and cytochrome P-

65

450 isozymes, with cancer-related processes such as cell apoptosis and proliferation,

66

angiogenesis, or metastasis.11-13 But because of the contradictory effects of the

67

manipulation of the ARA cascade,12,14 the mechanisms that mediate these effects remain

68

unclear.

69

Different mechanisms have been suggested to participate in the anticancer activities of

70

n-3 PUFAs, such as apoptosis promotion, inhibition of cell transformation and repression

71

of cell proliferation. Many of them have been associated to the repression of eicosanoids

72

production from n-6 PUFAs.15 Previous studies have tested the inhibition of HT-29 cell

73

proliferation produced by some n-3 FAs. Docosahexaenoic acid (DHA, 22:6n3) strongly

74

and dose-dependently promotes apoptosis, and it enhances many cellular responses that

75

decrease cell proliferation. Because DHA is promptly integrated into cancer cell

76

membranes, considerable research has been conducted on membrane–initiated phenomena.

77

For instance, DHA has been described to take part in the promotion of apoptosis and the

78

inhibition of both in vivo and in vitro proliferation. However, the role of DHA on cell cycle

79

regulation has been scarcely studied on cancer cells, and therefore the mechanisms of

80

action of DHA are not yet entirely understood.16

4

ACS Paragon Plus Environment

Page 4 of 36

Page 5 of 36

Journal of Agricultural and Food Chemistry

81

As for ARA, previous studies have debated whether the increased ARA metabolism

82

found in cancer cells is an initiator or an output of tumour progression. Studies on COX-2

83

and its inhibitors have strongly supported the notion that the induction of COX-2 is a

84

critical step in carcinogenesis and tumour growth. A previous study has also shown that

85

unesterified ARA is an important signal for apoptosis and that the apoptosis promotion by

86

nonsteroidal anti-inflammatory drugs and other inhibitors of ARA metabolism results from

87

ARA accumulation.17. Bishayee and Sethi18 have stated that the ARA pathway, a metabolic

88

process, plays a key role in carcinogenesis. Hence, previous studies have considered the

89

ARA pathway metabolic enzymes phospholipase A2s, cyclooxygenases (COXs) and

90

lipoxygenase (LOX) – together with the metabolic derivatives of these enzymes, such as

91

prostaglandins and leukotrienes – to be new targets for the prevention and treatment of

92

cancer.

93

This work has attempted to determine the in vitro anticancer activities of DHA and

94

ARA, based on the regulation of gene transcription in HT-29 CRC cells. To do so, we

95

performed different cell assays and studied protein abundance changes induced by DHA

96

and ARA in the global proteome of these cells by means of a quantitative proteomics

97

approach based on liquid chromatography–mass spectrometry (LC-MS) called “sequential

98

windowed acquisition of all theoretical fragment ion mass spectra” (SWATH-MS)19

99

followed by pathway analysis.

100 101

MATERIALS AND METHODS

102

Oil Samples

5

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

103

Two commercial oils, DHASCO® and ARASCO®, 40% DHA and 40% ARA,

104

respectively, obtained from the Market Bioscience Corporation (Columbia, MD, USA),

105

were used as a source for obtaining DHA and ARA FFAs. DHASCO® and ARASCO® are

106

produced by combining high oleic sunflower oil with an oil obtained from a unicellular alga

107

(Crypthecodinium cohnii) and a fungi (Mortierella alpina ), respectively.

108

Purification process

109

ARA and DHA as FFAs were purified from 40 mg of DHA- or ARA-ethyl ester

110

according to a previously described chromatography process,20 using mixtures of n-hexane

111

and acetone as eluting solvents in the sequence shown in Supplementary Table S1,

112

optimized for maximizing PUFA purity, and at flow rates of approximately 1–2 mL/min.

113

Eluting fractions were collected in labelled tubes and analysed directly using GLC to

114

determine the purity grade. Subsequently, the ethyl esters were acidified according to

115

Vázquez and Akoh’s approach.21 FFAs were obtained in this manner.

116

GLC analyses

117

A GLC device (Focus, Thermo Electron, Cambridge, UK) combining a flame ionisation

118

detector with a capillary column (30 m x 0.25 mm ID x 0.25 µm) (Omegawax 250, Supelco

119

(Bellefonte, PA, USA)) were used for FA ethyl esters analysis. Temperature of the oven

120

was programmed as follows: 90ºC for 1 min, increase to 100°C in 1 min, then 3 min at

121

100°C, increase of 6ºC/min until reaching 260°C, and finally a step of 260°C during 5 min.

122

The injector temperature was set to 250°C, with a split ratio of 50:1 and an injector volume

123

of 4 µL. The detector temperature was set to 260°C. Nitrogen carrier gas flow was 1

124

mL/min.

6

ACS Paragon Plus Environment

Page 6 of 36

Page 7 of 36

125 126

Journal of Agricultural and Food Chemistry

Cell assays The cell line used for the assays was HT-29 (colon cancer), kindly provided by the

127

Technical Instrumentation Facility of the University of Granada (Granada, Spain), and

128

incubated at 37°C and 5% CO2 in Gibco RPMI 1640 medium 1640 (Thermo Fisher

129

Scientific, Waltham, MA, USA) with penicillin-streptomycin (100 mg/ml), 1-glutamine (2

130

mM), sodium pyruvate (1mM) and 5% foetal bovine serum. Cell cultures and a 3-(4,5-

131

dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) test were conducted as

132

detailed in previous works.22

133

Final concentration of the purified acyl species (acetone-diluted ARA-FFA or DHA-

134

FFA) added to the medium was in the range of 50–600 µM. The final concentration of

135

acetone added to culture medium, was 1% (v/v). Control cells were also cultured with a

136

medium containing 1% acetone. Lactate dehydrogenase (LDH) assay (the In Vitro

137

Toxicology Assay Kit, Lactic Dehydrogenase base, Sigma TOX 7, Sigma-Aldrich, St.

138

Louis, MO, USA) was conducted as for the MTT assay but with a cell density of 5 ×

139

103 cells/well, and measures were performed following the manufacturer’s instructions. The

140

results were obtained by determining the absorbance at 490 nm with a reference filter at

141

690 nm at 48 h and 72 h. For measuring caspase-3 activity we used the Caspase-3 Assay

142

Kit (Sigma CASP-3-C, Sigma-Aldrich, St. Louis, MO, USA) following the instructions

143

from the manufacturer and determining the absorbance (450 nm) at 90 min, with a cell

144

density of 1 x 107.

145

Proteomics analysis

146

Sample preparation

7

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

147

HT-29 cells from three different groups – DHA (600 µM DHA, added to the culture

148

medium at 24h), ARA (600 µM, added at 24h) and control (no acyl species added) – were

149

recovered and lysed. Protein extracts were obtained from six biological replicates for each

150

group, and protein content was solubilized in 0.2% RapiGest SF (Waters, Milford, MA,

151

USA) after precipitation. Protein content in each sample was determined using a nano-

152

fluorimetric assay (Qubit Protein Assay, Thermo Fisher Scientific). 40 µg of protein was

153

digested with trypsin (Promega, Madison, WI, USA) as previously described23 in two steps,

154

incubating at 37ºC for 2 h in the first step and for 15 h in the second step. RePliCal iRT

155

peptides (PolyQuant GmbH, Bad Abbach, Germany) were spiked into each sample in order

156

to calibrate the peptide retention times in the SWATH runs.

157

Creation of the spectral library

158

For building the spectral library, equal mixtures of the six biological replicates from

159

each condition were pooled and analysed using a shotgun data-dependent acquisition

160

(DDA) approach by nano-liquid chromatography coupled to tandem mass spectrometry

161

(nLC-MS/MS) as described elsewhere24. The equipment used was a Sciex Triple TOF

162

5600+ (Redwood City, CA, USA) coupled to an Eksigent Ekspert nLC415 (Sciex)

163

equippled with a reversed phase capillary column (Acclaim PepMap C18, Thermos Fisher

164

Scientific). Detailed LC and mass spectrometry parameters used can be found in the

165

Supplementary Material.

166

DDA runs were searched against a human Swiss-Prot protein database (20,200 entries,

167

appended with the sequences from the RePliCal iRT peptides) using Protein Pilot (Sciex,

168

version 5.0.1), setting a false discovery rate (FDR) threshold of 0.01. SWATH Acquisition

8

ACS Paragon Plus Environment

Page 8 of 36

Page 9 of 36

Journal of Agricultural and Food Chemistry

169

MicroApp (version 2.0, Sciex) was used for building a peptide spectral library containing

170

the peptides identified in the database search with a confidence score above 99%.

171

SWATH data acquisition and analysis

172

Two µL of each of the six biological replicates from each group was run in the same LC

173

parameters and MS equipment described above for the DDA runs, but using a SWATH

174

data-independent acquisition (DIA) method instead. Mass spectrometry parameters used in

175

the SWATH method are detailed in the Supplementary Material. Briefly, it consisted on the

176

acquisition of 60 MS/MS events with precursor isolation windows of variable width

177

comprising the 350-1,250 m/z range. The width of each of the 60 m/z windows was

178

adjusted according to the ion frequency found in the previous shotgun runs.

179

Protein quantitative data was extracted from the SWATH raw files as previously

180

described24. Briefly, SWATH Acquisition MicroApp was used for extracting the ion

181

chromatogram traces from the SWATH raw files and using the previously generated

182

spectral library, and the following parameters: ten peptides/protein; seven fragment

183

ions/peptide; extraction windows of 20 min and 20 ppm; peptide FDR of 1% and

184

confidence score threshold of 95%. The iRT peptides spiked in each samples were used for

185

realigning the retention times of the quantified peptides.

186

Pathway analysis

187

The significantly affected pathways were studied using Advaita Bio’s iPathwayGuide

188

(http://www.advaitabio.com/ipathwayguide), which considers over-representation of

189

significant proteins, pathway topological information and position and role of every

9

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

190

protein.25 Pathways obtained from the “Kyoto Encyclopedia of Genes and Genomes”

191

(KEGG) database were used.

192

Statistical analysis

193

For cell assays (MTT, LDH and caspase-3 assays), statistical significance (P < 0.05) was

194

determined by analysis of variance (ANOVA) followed by the assessment of differences

195

using STATGRAPHICS Plus version 5 (Statistical Graphic Corp., Warrenton, VA, USA).

196

Figs. 1 and 2 show mean data for three independent experiments ± standard deviation.

197

Normalisation of the protein abundance signal as measured by SWATH was carried out

198

using MarkerView (v1.2.1, Sciex), and testing for differential abundance was performed at

199

the protein level by applying a Student’s t-test. The qvalue R package was used in order to

200

obtain a FDR q-value estimation to check for multiple testing underestimation of p-

201

values26. For pathway analysis, a p-value was calculated for each reported pathway by

202

iPathwayGuide using the Fisher’s method, and p-values were adjusted for multiple

203

comparisons using the FDR approach.

204 205

RESULTS AND DISCUSSION

206

Antiproliferative actions of DHA and ARA

207

The purity of the assayed PUFAs was found to be 98.1% and 99.2% (FA% of total FA

208

area) for DHA and ARA, respectively. The cell proliferation inhibition exercised by both

209

FFAs on HT-29 CRC cells was evaluated by means of the MTT test, which measures cell

210

survival and proliferation. The effects of DHA and ARA on HT-29 cell proliferation are

211

shown in Fig. 1.A. As depicted in the figure, significant concentration-dependent inhibitory

10

ACS Paragon Plus Environment

Page 10 of 36

Page 11 of 36

Journal of Agricultural and Food Chemistry

212

effects were found from the two studied compounds. The half-maximal inhibitory

213

concentrations (IC50) at 48 h and 72 h for DHA were 259 µM and 245 µM, respectively,

214

while for ARA the same values were 368 µM and 324 µM, respectively.

215

For the physiological relevance of these concentrations, a previous study found the

216

serum concentration of DHA in rats to be 97.3 µg/ml (297 µM) after 4 weeks of DHA

217

supplementation.27 The present IC50 for DHA may thus be relevant to tumour progression

218

in vivo, while uncertainty remains for ARA; it would be necessary to experiment in vivo to

219

determine the concentrations of such PUFAs attainable in serum without producing adverse

220

effects.

221

Das and Das,28 using MTT assay, found a significant cell viability reduction of SH-

222

SY5Y (55% reduction) and C6G cells (54% reduction) after a 24 h incubation with 100 µM

223

DHA. In addition, Ramos-Bueno et al.22 demonstrated that acyl-species ARA and DHA

224

derived from ARASCO® and DHASCO® oils, respectively, had relatively high in vitro

225

anticancer activity on HT-29 cells. Chen et al.,29 using the MTT test, showed that Hep G2-

226

MV2E1-9 cells lost more than 80% viability after a pretreatment with 50 µM ARA.

227

LDH assay

228

An LDH assay was conducted to assess the release of the lactate dehydrogenase enzyme

229

into the culture medium after cell membrane damage caused by both DHA and ARA. The

230

concentrations for performing the tests were 0–600 µM, at which the tested compounds

231

showed different in vitro antiproliferative activities from those detected by the MTT assay.

232

DHA induced more active damage at 300 µM and 48 h exposure than ARA did (Fig.

233

1.B), although at 500 µM and after 72 h exposure, ARA was more effective in this action

11

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

234

than DHA. Overall, LDH values were lower than those obtained in the MTT assay. In this

235

regard, Ramos-Bueno et al.22 indicated that, when comparing the effects measured by LDH

236

and MTT assays, the former were attained at higher levels of cell damage. Such differences

237

could be attributable to the specificity of LDH testing compared to the MTT assay. A

238

previous study showed that LDH leakage occurred only when reactive oxygen species

239

related to cell death appeared, which were responsible for mitochondrial damage.22 Overall,

240

the MTT assay showed cell growth inhibition at lower concentrations of FFA than the LDH

241

test, because MTT reduction is not an indicator of cell death but rather constitutes a

242

measure of relative metabolic activity, as demonstrated in a previous study on HT-29

243

cells.30 Concerning ARA, our results agree with previous findings,29,31 which have shown

244

the same PUFA-stimulated LDH activity in some other cells (e.g. Hep G2-MV2E1-9 and

245

Sertoli).

246

Caspase-3 assay

247

The activation of caspase-3, specifically through self-cleavage, has been shown to be the

248

event that commits cells to apoptotic death.32 As shown in Fig. 2, DHA induced caspase-3

249

activation at 300 µM, while ARA did not, and both PUFAs exercised a similar induction of

250

apoptosis at 600 µM. Several studies have suggested that DHA displays a notable

251

protective effect on CRC, which could be at least partly due to its pro-apoptotic activity.

252

The mechanisms affected by DHA for promoting apoptosis in CRC are still largely

253

unknown, however. Previous works have demonstrated the apoptotic activity exerted by

254

DHA in several cancer cell lines,33,34 while a variety of authors have tested whether DHA

255

induced apoptosis in CRC cells.4,35,36. Habermann et al.,37 using an in vitro model and HT-

256

29 and LT97 cells, proposed a generic mechanism for the apoptosis promoted by DHA that

12

ACS Paragon Plus Environment

Page 12 of 36

Page 13 of 36

Journal of Agricultural and Food Chemistry

257

was based on inducing reactive oxygen species due to DHA double bonds, which led to

258

apoptosis. Narayanan et al.38 found that DHA-regulated genes and transcription factors

259

induced apoptosis in human colon cancer cells. Zhang et al.39 have also proposed that both

260

DHA and ARA trigger the apoptosis of colon cancer cells through a mitochondrial

261

pathway; they found that DHA in particular induced dramatic activities in both caspase-3

262

and caspase-9.

263

Proteomics analysis

264

DIA MS strategies are showing great power for the study of abundance differences in

265

complete proteomes. Precision and accuracy of the DIA approach of SWATH-MS19 has

266

been described to match that of selected reaction monitoring, the gold standard in protein

267

quantitation, even when applied to the quantitation of whole proteomes.40

268

Shotgun analysis and spectral library

269

Equal amounts of all samples from each group were pooled and analysed by DDA LC-

270

MS. All the datasets were combined, and 15,406 peptides and 2,140 proteins were

271

identified using the ProteinPilot search engine using a FDR threshold of 1%

272

(Supplementary Table S2). This search result was then used for building a spectral library,

273

which comprised 2,002 proteins.

274

Protein abundance changes

275

Protein quantitative data was extracted from the SWATH raw files as described in

276

Material and Method above. In total, 7,653 peptides and 1,882 proteins were confidently

277

quantified (Supplementary Table S3). To assess the global impact of the overall quantified

278

proteins, principal component analysis (PCA) was performed on the whole protein dataset.

13

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

279

PCA was able to separate the two tested groups from each other and from the control group

280

(Fig. 3a), thus showing a large and different effect of the specific FFA (DHA or ARA) on

281

HT-29 colon cancer cell proteomes.

282

Effects of DHA and ARA on HT-29 colon cancer cell proteomes

283

Supplementary Tables S4 and S5 show the results from the differential abundance

284

testing for both comparisons (the DHA group to control and the ARA group to control,

285

respectively), including a p-value and the corresponding FDR-adjusted q-value for each of

286

the 1,882 quantified proteins. A total of 1,015 proteins showed changes in expression when

287

we addressed the impact of the DHA extract and considered a non-adjusted p-value

288

threshold of 0.05. Notably, due to the p-value distribution (Supplementary Fig. S1a), the q-

289

values obtained from the FDR p-value correction were lower than the corresponding non-

290

adjusted p-values (Supplementary Fig. S1b), which meant that we had more significant

291

tests (significant proteins) if we chose a q-value threshold rather than a p-value threshold in

292

order to consider a protein to be significantly regulated (Supplementary Fig. S1c).

293

For downstream analyses, we considered a restrictive scenario – namely p-value and

294

fold-change thresholds of 0.01 and 2.0, respectively – in order to have more confidence in

295

selecting the proteins that presented real expression changes. When we applied these cut-

296

off values, 284 proteins showed changes in expression, with 45 proteins being up-regulated

297

and 239 down-regulated (Fig. 3b and Supplementary Fig. S6).

298

When we addressed the changes caused by the ARA extract on HT-29 cancer cells and

299

applied the same p-value and fold-change cut-off values, only 73 proteins were found to

300

present changes in expression, with 27 proteins being up-regulated and 46 down-regulated

301

(Fig. 3c and Supplementary Fig. S7). The p-value distribution, q-value to p-value

14

ACS Paragon Plus Environment

Page 14 of 36

Page 15 of 36

Journal of Agricultural and Food Chemistry

302

correspondence, and number of significant proteins according to several cut-off values for

303

ARA to control differential abundance tests are shown in Supplementary Fig. S2. When we

304

compared the 284 and 73 proteins that we found were regulated as a consequence of adding

305

DHA and ARA to the media (respectively), only 43 proteins were commonly regulated in

306

both conditions (Fig. 3d). DHA was responsible for a larger effect on HT-29 cancer cells

307

than ARA, as it is derived from the number of affected proteins and the larger fold changes

308

that are found.

309

Pathway impact analysis

310

After FDR correction of the p-values, three pathways were found to be significantly

311

affected by DHA (Fig. 4), with the Proteasome pathway (KEGG: 03050) presenting the

312

deepest effect. The proteasome is a protein-degrading large complex which participates in

313

different critical cellular functions, including cell cycle regulation, inflammatory and stress

314

responses, apoptosis, signal transduction, cell differentiation and antigen processing. The

315

26S proteasome is the most characteristic conformation, consisting of two 19S regulatory

316

particles (which combine two base particles, PA700 Lid and PA700 Base) and a 20S

317

proteolytic particle, and it is responsible for the degradation of ubiquitinated-labelled

318

proteins.41 The 11S Regulator particle, or PA28, is a second type of proteasome particle. It

319

consists of two different subparticles, PA28-α and PA28-β, which have been described to

320

be greatly up-regulated by IFN-γ and have a role in generating antigenic peptides that are

321

presented in the MHC-I pathway.42 We found up to 18 proteins from the Proteasome

322

pathway to be down-regulated as a consequence of the addition of DHA extract, including

323

members of most of the proteasome regulatory particles (PA700 Lid particle, PA700 Base

324

particle, PA28-αβ and PA28-γ), in addition to the 20S Core particle (Fig. 5). PA28- γ is

15

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

325

localised in the nucleus and has been described to have a role in apoptosis and cell cycle

326

regulation.43 Many proteasome inhibitors, including natural substances such as polyphenol

327

compounds, have been found to hinder cell proliferation, selectively promote apoptosis in

328

proliferating cells, and inhibit angiogenesis.44,45 These qualities make these agents attractive

329

drug candidates for treating different kinds of cancer, and some have even been studied in

330

human clinical trials.44,46 According to our results, DHA-derived FFA could be included

331

among these candidates. Even though the mechanisms of action are not completely

332

understood, various studies have demonstrated that proteasome inhibitors alter the balance

333

of pro- and anti-apoptotic proteins in human cancer cells, generating an accumulation of

334

tumour-suppressor proteins and the induction of apoptosis.47 Some authors have suggested

335

that proteasome inhibitors promote cell death by means of a p53-dependent mechanism, by

336

inducing the expression of some of the genes related to apoptosis, such as PUMA or Bax.48

337

Others, however, have pointed to a p53-independent mechanism of proteasome inhibitors-

338

induced apoptosis. Noxa, one pro-apoptotic protein, has been linked to this apoptosis

339

mechanism, since several proteasome inhibitors have been found to induce it in a p53-

340

independent manner in cancer cells of different origins.49 Our data could not clearly

341

demonstrate a p53-dependent nor independent mechanism of apoptosis. No quantitative

342

information was obtained from our SWATH-MS analysis for the main pro- and anti-

343

apoptotic proteins, PUMA, NOXA, Mcl-1, p21 or Bcl-2. A few other pro-apoptotic factors

344

and tumour-suppressor proteins were found to be slightly up-regulated, such as AIFM1

345

(apoptosis-inducing factor 1, fold-change 1.65) and P5i11 (tumour protein p53-inducible

346

protein 11, fold-change 1.40). A few other anti-apoptotic proteins were found to be down-

347

regulated, including API5 (apoptosis inhibitor 5, fold-change 0.70), which has been

348

described as a suppressor of E2-promoter binding factor-dependent apoptosis in vivo and in 16

ACS Paragon Plus Environment

Page 16 of 36

Page 17 of 36

Journal of Agricultural and Food Chemistry

349

vitro in multiple tissues,50 and TNF-α induced protein 8 (TFIP8, fold-change 0.61), which

350

is an inhibitor of TNF-mediated apoptosis.51

351

Notably, we found PAWR (PRKC apoptosis WT1 regulator protein), a protein described

352

as pro-apoptotic in certain cancer cells such as prostate,52 to be down-regulated as an effect

353

of the DHA treatment on HT-29 cells (fold-change 0.52). But when we analysed the

354

differentially expressed proteins in the context of the p53 signalling pathway in

355

iPathwayGuide, we found that three key proteins were regulated by DHA (Supplementary

356

Fig. S3): TSP1 (thrombospondin 1, gene THBS1), which mediates anti-angiogenic

357

properties, presented a fold-change of 2.05; CDC2 (gene CDK1, cyclin dependent kinase

358

1), which regulates cell cycle and whose inhibitors are being explored as chemotherapeutic

359

agents in cancer,53 presented a fold-change of 0.24; and p53R2 (RIR2, ribonucleotide

360

reductase regulatory subunit M2, gene RRM2), involved in DNA synthesis, presented a

361

fold-change of 0.33. Interestingly, none of the proteins related to apoptosis within the p53

362

signalling pathway were affected by DHA (Supplementary Fig. S3). This behaviour could

363

point to a p53-independent mechanism of apoptosis as a consequence of treating HT-29

364

cells with DHA, although more research would be required to maintain this argument.

365

When we added ARA-FFA to the HT-29 cells, only one protein in the p53 signalling

366

pathway, p53R2 (gene RRM2), resulted in regulation (down-regulation).

367

According to our pathway analysis, two other pathways, Epstein Barr virus (EBV)

368

infection (KEGG: 05169) and DNA replication (KEGG: 03030), were also found to be

369

significantly affected by the addition of DHA, although the effect was not as evident as for

370

the Proteasome pathway, which presented much higher p-values. EBV is a ubiquitous

371

human herpesvirus that has been associated with oncogenesis in Burkitt lymphoma,

17

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

372

nasopharyngeal carcinoma, Hodgkin’s lymphoma, gastric adenocarcinoma and breast

373

cancer.54 Sixteen proteins from the EBV infection pathway were affected by DHA in our

374

study, with 14 proteins down-regulated, most related to DNA transcription factors

375

(Supplementary Fig. S4a). EBV makes cancer cells more aggressive,54 so we hypothesise

376

that the down-regulation of the proteins related to this pathway (which we found as a

377

consequence of adding DHA) could have had the opposite effect – that is, reducing

378

colorectal cancer cell viability. For DNA replication, DHA was found to produce the under-

379

expression of five out of the six helicase (also called the mini-chromosome maintenance

380

[MCM] complex) particles: Mcm2, Mcm3, Mcm4, Mcm5 and Mcm7 (Supplementary Fig.

381

S4b).

382

DNA replication was the top-ranked pathway when assessing the effect of ARA extract;

383

in that case, all six helicase particles (Mcm2, Mcm3, Mcm4, Mcm5, Mcm6 and Mcm7)

384

were found to be down-regulated (Fig. 6), presenting larger fold-changes than for DHA.

385

DNA helicase is the protein complex that unwinds the duplex parental DNA at the

386

replication fork, ahead of the DNA synthetic machinery.55 DNA replication procedure is

387

likewise linked to cell-cycle and to DNA processes in order to support genomic integrity,56

388

and therefore the down-regulation of the helicase proteins that our results showed was

389

expected to transfer an inhibitory effect to these processes. In fact, the Cell Cycle pathway

390

(KEGG: 04110) – which also occurred due to the down-regulation of the six MCM

391

particles – and the Pyrimidine Metabolism (KEGG: 00240) pathway were the two other

392

significant pathways affected by the addition of ARA (Supplementary Fig. S5), although

393

the Pyrimidine Metabolism pathway presented a much higher p-value than the DNA

394

replication and Cell Cycle pathways. Interestingly, neither the Proteasome pathway nor the

18

ACS Paragon Plus Environment

Page 18 of 36

Page 19 of 36

Journal of Agricultural and Food Chemistry

395

EBV pathway were significantly affected by ARA, since none of the proteins included in

396

these pathways were found to have been differentially expressed.

397

We have established that both ARA-FFA and DHA-FFA exercise different in vitro

398

anticancer activities on CRC. At 300 µM, DHA induced a stronger decrease of colorectal

399

cancer cell proliferation than ARA did, but DHA also induced fewer cytotoxic effects.

400

Pathway analyses of the SWATH-MS-generated proteomics data revealed that both FAs

401

had an impact on the cell cycle by means of down-regulating the protein particles that form

402

the helicase complex, but DHA also affected the proteasome system, since many proteins

403

from the different proteasome subunits were also under-expressed.

404

The present study shows that DHA and ARA are significantly inhibiting the growth of

405

cancer HT-29 cells depending on time and concentration, which suggests that both of them

406

may be helpful for hindering the development of aggressive adenocarcinoma.

407

Based on these findings, further studies will need to be conducted to determine if

408

a combination of PUFAs with regular anticancer molecules would represent a reasonable

409

approach to CRC treatment. The use of next-generation proteomics and other ‘omics’

410

approaches will allow for analysing the effects of natural compounds on global patterns of

411

gene expression and protein composition of the cell, thus improving the knowledge that we

412

have about the biological roles of nutraceuticals.

413 414 415 416

Supporting Information description The LC-MS raw files associated with this article are available at www.proteomexchange.org (Proteome Xchange Consortium), dataset id PXD008653. The

19

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

417

following supporting information and material can be found in the Web edition of the

418

Journal:

419

Supplementary material.pdf, including the detailed LC and mass spectrometry parameters

420

used for both data-dependent acquisition and SWATH methods, and figures S1

421

(Differential abundance test results for DHA vs. control), S2 (Differential abundance test

422

results for ARA vs. control), S3 (HT-29 cells p53 signaling pathway showing protein

423

changes observed as a result of treatment with DHA), S4 (HT-29 cells Epstein Barr virus

424

infection and DNA replication pathways showing the effect of DHA treatment), and S5

425

(HT-29 cells cell cycle and pyrimidine metabolism pathways showing the effect of ARA

426

treatment), and Table S1 (gradient used in the chromatographic purification of both free

427

fatty acids, DHA and ARA).

428

Supplementary Table S2.xlsx. 2,140 proteins that were identified using the ProteinPilot

429

search engine and a FDR threshold of 1%.

430

Supplementary Table S3.xlsx. SWATH-based protein quantification data for all the

431

samples.

432

Supplementary Table S4.xlsx. Differential abundance test for DHA vs. control.

433

Supplementary Table S5.xlsx. Differential abundance test for ARA vs. control.

434

Supplementary Table S6.xlsx. Differential expressed proteins for DHA vs. control.

435

Supplementary Table S7.xlsx. Differential expressed proteins for ARA vs. control.

436 437

References

20

ACS Paragon Plus Environment

Page 20 of 36

Page 21 of 36

438

Journal of Agricultural and Food Chemistry

1. Azrad, M.; Turgeon, C.; Demark-Wahnefried, W. Current evidence linking

439

polyunsaturated fatty acids with cancer risk and progression. Front. Oncol. 2013,

440

3,224.

441

2. Hardy, S.; El-Assaad, W.; Przybytkowski, E.; Joly, E.; Prentki, M.; Langelier, Y.

442

Saturated Fatty Acid-induced Apoptosis in MDA-MB-231 Breast Cancer Cells. J. Biol.

443

Chem. 2003, 278, 31861-31870.

444

3. Kampf, J.P.; Kleinfeld, A.M. Is membrane transport of FFA mediated by lipid, protein,

445

or both? An unknown protein mediates free fatty acid transport across the adipocyte

446

plasma membrane. Physiology (Bethesda) 2007, 22, 7-14.

447

4. Calviello, G.; Resci, F.; Serini, S.; Piccioni, E.; Toesca, A.; Boninsegna, A.; Monego, G.;

448

Ranelletti, F.; Palozza. P. Docosahexaenoic acid induces proteasome-dependent

449

degradation of b-catenin, down-regulation of survivin and apoptosis in human

450

colorectal cancer cells not expressing COX-2. Carcinogenesis 2007, 28, 1202–1209.

451

5. Antal, O.; Hackler,L.; Shen, J.; Mán, I.; Hideghéty, K.; Kitajka, K.; Puskás, L.G.

452

Combination of unsaturated fatty acids and ionizing radiation on human glioma cells:

453

cellular, biochemical and gene expression analysis. Lipids Health Dis. 2014, 13, 142.

454

6. Motaung, E.; Prinsloo, S. E.; van Aswegen, C. H.; du Toit, P. J.; Becker, P. J.; du Plessis,

455

D. J. Cytotoxicity of combined essential fatty acids on a human prostate cancer cell

456

line. Prostaglandins, Leukotrienes Essent. Fatty Acids 1999, 61, 331-337.

457

7. Zhang, C.; Yu, H.; Ni, X.; Shen, S.; Das, U.N. Growth Inhibitory Effect of

458

Polyunsaturated Fatty Acids (PUFAs) on Colon Cancer Cells via Their Growth

459

Inhibitory Metabolites and Fatty Acid Composition Changes. PLoS ONE 2015, 10,

460

e0123256.

21

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

461

8. Michalak, A.; Mosinska, P.; Fichna, J. Polyunsaturated Fatty Acids and Their

462

Derivatives: Therapeutic Value for Inflammatory, Functional Gastrointestinal

463

Disorders, and Colorectal Cancer. Front. Pharmacol. 2016, 7, 459.

464 465

9. Cockbain, A.J.; Toogood, G.J.; Hull, M.A. Omega-3 polyunsaturated fatty acids for the treatment and prevention of colorectal cancer. Gut 2012, 61, 135-149.

466

10. Dommels, Y. E.; Haring, M. M.; Keestra, N. G.; Alink, G. M.; van Bladeren, P. J.; van

467

Ommen, B. The role of cyclooxygenase in n-6 and n-3 polyunsaturated fatty acid

468

mediated effects on cell proliferation, PGE 2 synthesis and cytotoxicity in human

469

colorectal carcinoma cell lines. Carcinogenesis 2003, 24, 385-392.

470 471 472 473 474 475 476 477

11. Moreno, J.J. New aspects of the role of hydroxyeicosatetraenoic acids in cell growth and cancer development. Biochem. Pharmacol. 2009, 77, 1-10. 12. Ferrer, R.; Moreno, J.J. Role of eicosanoids on intestinal epithelial homeostasis. Biochem. Pharmacol. 2010, 80, 431-438. 13. Moreno, J.J. Eicosanoid receptors: Targets for the treatment of disrupted intestinal epithelial homeostasis. Eur. J. Pharmacol. 2017, 796, 7-19. 14. Cabral, M.; Martin-Venegas, R.; Moreno, J.J. Leukotriene D4-induced Caco-2 cell proliferation is mediated by prostaglandin E2 synthesis. Physiol. Rep. 2015, 3, e12417.

478

15. Kang, K.S.; Wang, P.; Yamabe, N.; Fukui, M.; Jay, T.; Zhu, B.T. Docosahexaenoic

479

Acid Induces Apoptosis in MCF-7 Cells In Vitro and In Vivo via Reactive Oxygen

480

Species Formation and Caspase 8 Activation. PLoS ONE 2010, 5, e10296.

481

16. Newell, M.; Baker, K.; Postovit, L.M.; Field, C.J. A Critical Review on the Effect of

482

Docosahexaenoic Acid (DHA) on Cancer Cell Cycle Progression. Int. J. Mol. Sci.

483

2017, 18, 1784.

22

ACS Paragon Plus Environment

Page 22 of 36

Page 23 of 36

484

Journal of Agricultural and Food Chemistry

17. Cao, Y.; Pearman, A. T.; Zimmerman, G. A.; McIntyre, T. M.; Prescott, S. M.

485

Intracellular unesterified arachidonic acid signals apoptosis. Proc. Natl. Acad. Sci. U.

486

S. A. 2000, 97, 11280-11285.

487 488 489

18. Bishayee, A.; Sethi, G. Bioactive natural products in cancer prevention and therapy: Progress and promise. Semin. Cancer Biol. 2016, 40-41, 1-3. 19. Gillet, L.C.; Navarro, P.; Tate, S.; Röst, H.; Selevsek, N.; Reiter, L.; Bonner, R.;

490

Aebersold, R. Targeted data extraction of the MS/MS spectra generated by data-

491

independent acquisition: a new concept for consistent and accurate proteome analysis.

492

Mol. Cell. Proteomics 2012, 11, O111.016717.

493

20. Guil-Guerrero, J.L.; Campra-Madrid, P.; Belarbi, E.H. (2000). γ-Linolenic acid

494

purification from seed oil sources by argentated silica gel chromatography column.

495

Process Biochem. 2000, 36, 341-354.

496

21. Vázquez, L.; Akoh, C.C. Concentration of Stearidonic Acid in Free Fatty Acid and

497

Fatty Acid Ethyl Ester Forms from Modified Soybean Oil by Winterization. J. Am. Oil

498

Chem. Soc. 2011, 88, 1775-1785.

499

22. Ramos-Bueno, R.P.; González-Fernández, M.J.; Guil-Guerrero, J.L. Various

500

acylglycerols from common oils exert different antitumor activities on colorectal

501

cancer cells. Nutr. Cancer 2016, 68, 518-529.

502

23. Vowinckel, J.; Capuano, F.; Campbell, K., Deery, M. J.; Lilley, K. S.; Ralser, M. The

503

beauty of being (label)-free: sample preparation methods for SWATH-MS and next-

504

generation targeted proteomics. F1000Research 2014, 2, 272.

23

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

505

24. Ortea, I.; Ruiz-Sánchez, I.; Cañete, R.; Caballero-Villarraso, J.; Cañete M. D.

506

Identification of candidate serum biomarkers of childhood-onset growth hormone

507

deficiency using SWATH-MS and feature selection. J. Proteomics 2018, 175, 105-113.

508

25. Tarca, A. L.; Draghici, S.; Khatri, P.; Hassan, S. S.; Mittal, P.; Kim, J. S.; Kim, C. J.;

509

Kusanovic, J. P.; Romero, R. A novel signaling pathway impact analysis.

510

Bioinformatics 2009, 25, 75-82.

511

26. Storey, J. D. Bass, A. J.; Dabney, A.; Robinson D. (2015). qvalue: Q-value estimation

512

for false discovery rate control. R package version 2.10.0.

513

http://github.com/jdstorey/qvalue (accessed at Feb 2018).

514

27. Mizota, A.; Sato, E.; Taniai, M.; Adachi-Usami, E.; Nishikawa, M. Protective effects of

515

dietary docosahexaenoic acid against kainate-induced retinal degeneration in rats.

516

Invest. Ophthalmol. Vis. Sci. 2001, 42, 216-221.

517

28. Das, M.; Das, S. Identification of cytotoxic mediators and their putative role in the

518

signaling pathways during docosahexaenoic acid (DHA)-induced apoptosis of cancer

519

cells. Apoptosis 2016, 21, 1408-1421.

520

29. Chen, Q.; Galleano, M.; Cederbaum, A. I. Cytotoxicity and Apoptosis Produced by

521

Arachidonic Acid in Hep G2 Cells Overexpressing Human Cytochrome P4502E1. J.

522

Biol. Chem. 1997,272, 14532–14541.

523

30. Fotakis, G.; Timbrell, J.A. In vitro cytotoxicity assays: Comparison of LDH, neutral

524

red, MTT and protein assay in hepatoma cell lines following exposure to cadmium

525

chloride. Toxicol. Lett. 2006, 160, 171-177.

526

31. Meroni, S.B.; Riera, M.F.; Pellizzari, E.H.; Shteingart, H.F.; Cigorraga, S.B. Possible

527

role of arachidonic acid in the regulation of lactate production in rat Sertoli cells. Int. J.

528

Androl. 2003, 26, 310-317. 24

ACS Paragon Plus Environment

Page 24 of 36

Page 25 of 36

529

Journal of Agricultural and Food Chemistry

32. Akbar, M.; Kim, H. Y. Protective effects of docosahexaenoic acid in staurosporine-

530

induced apoptosis: involvement of phosphatidylinositol-3 kinase pathway. J.

531

Neurochem. 2002, 82, 655-665.

532

33. Chen, Z. Y.; Istfan, N. W. Docosahexaenoic acid is a potent inducer of apoptosis in HT-

533

29 colon cancer cells. Prostaglandins, Leukotrienes Essent. Fatty Acids 2000, 63, 301-

534

308.

535

34. Giros, A.; Grzybowski, M.; Sohn, V. R.; Pons, E.; Fernandez-Morales, J.; Xicola, R.

536

M.; Sethi, P.; et al. Regulation of colorectal cancer cell apoptosis by the n-3

537

polyunsaturated fatty acids Docosahexaenoic and Eicosapentaenoic. Cancer Prev. Res.

538

2009, 2, 732-742.

539

35. Kato, T.; Kolenic, N.; Pardini, R. S. Docosahexaenoic acid (DHA), a primary tumor

540

suppressive omega-3 fatty acid, inhibits growth of colorectal cancer independent of

541

p53 mutational status. Nutr. Cancer 2007, 58, 178-187.

542

36. Fasano, E.; Serini, S.; Piccioni, E.; Toesca, A.; Monego, G.; Cittadini, A.R.; Ranelletti,

543

F.O.; Calviello, G. DHA induces apoptosis by altering the expression and cellular

544

location of GRP78 in colon cancer cell lines. Biochim. Biophys. Acta 2012, 1822,

545

1762–1772.

546

37. Habermann, N.; Schön, A.; Lund, E.K.; Glei, M. Fish fatty acids alter markers of

547

apoptosis in colorectal adenoma and adenocarcinoma cell lines but fish consumption

548

has no impact on apoptosis-induction ex vivo. Apoptosis 2010, 15, 621-630.

549

38. Narayanan, A.B.; Narayanan, N.K.; Reddy, B.S. Docosahexaenoic acid regulated genes

550

and transcription factors inducing apoptosis in human colon cancer cells. Int. J. Oncol.

551

2001, 19, 1255-1262.

25

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

552

39. Zhang, Ch.; Yu. H.; Shen, Y.; Ni, X.; Shen, S.; Das, U.N. Polyunsaturated fatty acids

553

trigger apoptosis of colon cancer cells through a mitochondrial pathway. Arch. Med.

554

Sci. 2014, 5, 1081-1094.

555

40. Huang, Q.; Yang, L.; Luo, J.; Guo, L.; Wang, Z.; Yang, X.; Jin, W.; Famg, Y.; Ye, J.;

556

Shan, B.; Zhang, Y. SWATH enables precise label-free quantification on proteome

557

scale. Proteomics 2015,15, 1215-1223.

558

41. Adams, G. M.; Falke, S.; Goldberg, A. L.; Slaughter, C. A.; DeMartino, G. N.; Gogol,

559

E. P. Structural and functional effects of PA700 and modulator protein on proteasomes.

560

J. Mol. Biol. 1997,273, 646-657.

561 562 563 564 565 566 567

42. Rechsteiner, M.; Realini, C.; Ustrell, V. The proteasome activator 11 S REG (PA28) and class I antigen presentation. Biochem. J. 2000, 345, 1-15. 43. Rechsteiner, M.; Hill, C. P. Mobilizing the proteolytic machine: cell biological roles of preoasome activators and inhibitors. Trends Cell Biol. 2005, 15, 27-33. 44. Kisselev, A. F.; Goldberg, A. L. Proteasome inhibitors: from research tools to drug candidates. Chem. Biol. 2001, 8, 739-758. 45. Nam, S.; Smith, D.M.; Dou, Q. P. Ester bound-containing tea polyphenols potently

568

inhibit proteasome activity in vitro and in vivo. J. Biol. Chem. 2001, 276, 13322-

569

13330.

570 571 572 573

46. Adams, J.; Elliot, P. J. New agents in cancer clinical trials. Oncogene 2000, 19, 66876692. 47. Crawford, L.J.; Walker, B.; Irvine, A.E. Proteasome inhibitors in cancer therapy. J. Cell Commun. Signal. 2011, 5, 101–110.

26

ACS Paragon Plus Environment

Page 26 of 36

Page 27 of 36

574

Journal of Agricultural and Food Chemistry

48. Hock, A.K.; Vigneron, A.M.; Carter, S.; Ludwig, R.L.; Vousden, K.H. Regulation of

575

p53 stability and function by the deubiquitinating enzyme USP42. EMBO J. 2011, 30,

576

4921–4930.

577 578

49. Pandit, B.; Gartel, A.L. Proteasome Inhibitors Induce p53-Independent Apoptosis in Human Cancer Cells. Am. J. Pathol. 2011, 178, 355–360.

579

50. Morris, E.J.; Michaud, W.A.; Ji, J.Y.; Moon, N.S.; Rocco, J.W.; Dyson, N.J. Functional

580

identification of Api5 as a suppressor of E2F-dependent apoptosis in vivo. PLoS Genet.

581

2006, 2(11), e196.

582

51. You, Z.; Ouyang, H.; Lopatin, D.; Polver, P.J.; Wang, C.Y. Nuclear factor-kappa B-

583

inducible death effector domain-containing protein suppresses tumor necrosis factor-

584

mediated apoptosis by inhibiting caspase-8 activity. J. Biol. Chem. 2001, 276, 26398-

585

26404.

586

52. Chakraborty, M.; Qiu, S.G.; Vasudevan, K.M.; Rangnekar, V.M. Par-4 drives

587

trafficking and activation of Fas and FasI to induce prostate cancer cell apoptosis and

588

tumor regression. Cancer Res. 2011, 61, 7255-7263.

589 590 591

53. Wang, Q.; Su, L.; Liu N.; Zhang, L.; Xu, W.; Fang, H. Cyclin dependent kinase 1 inhibitors: a review of recent progress. Curr. Med. Chem. 2011, 18, 2025-2043. 54. Hu, H.; Luo, M.-L.; Desmedt, C.; Nabavi, S.; Yadegarynia, S.; Hong, A.;

592

Konstantinopoulos, P.A.; et al. Epstein-Barr virus infection of mammary epithelial

593

cells promotes malignant transformation. EBioMedicine 2016, 9, 148-160.

594

55. Stillman, B. Smart machines at the DNA replication fork. Cell, 78, 725-728.

595

56. Waga, S.; Stillman, B. The DNA replication fork in eukaryotic cells. Annu. Rev.

596

Biochem. 1998, 67, 721-751.

597 27

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

598 599

Funding sources Mass spectrometry analyses were performed at the IMIBIC Proteomics Unit, member

600

of ProteoRed (Plataforma de Recursos Biomoleculares y Bioinformáticos) and supported

601

by grant PT13/0001, funded by Instituto de Salud Carlos III (ISCIII) and FEDER.

602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617

28

ACS Paragon Plus Environment

Page 28 of 36

Page 29 of 36

618

Journal of Agricultural and Food Chemistry

FIGURE CAPTIONS

619

Figure 1. Effect of DHA (92.1%) and ARA (98.8%) on proliferation and cell membrane

620

integrity of HT-29 cancer cells treated with 1% acetone (control) or different concentrations

621

of FFAs (50, 100, 150, 250, 300, 500 and 600 µM) for 48 h and 72 h. The data represent

622

the means of three complete independent experiments ± SD (error bars). (a) DHA and

623

ARA-FFAs inhibit the viability of HT-29 cancer cells. Dose-response and IC50 values were

624

determined by MTT assay using an enzyme-linked immunosorbent assay (ELISA)

625

microplate reader. (b) Cell membrane damage; dose-dependent LDH activity plot.

626

Figure 2. Caspase-3 activity test. Bar histogram displaying caspase-3 activity in HT-29

627

colon cancer cells after 90 min incubation with DHA and ARA at 300 and 600 µM,

628

compared with untreated cells (control). The data represent the means of three complete

629

independent experiments ± SD (error bars).

630

Figure 3. Quantitative proteomics results. (a) Principal component analysis (PCA) of all

631

1,882 quantified proteins. Using principal components 1 and 2, samples from the two tested

632

groups were separated from each other and from the control group. (b) Volcano plot for

633

DHA vs control, and (c) ARA vs control, showing p-value < 0.01 and fold-change ≥ 2.0 (or

634

≤ 0.5) thresholds. Proteins above these cut-offs are shown in red and were considered

635

significant proteins for the subsequent downstream analyses. The most extreme proteins are

636

labelled with the protein name. (d) Venn diagram showing the number of significant

637

proteins specifically for each comparison and common to both comparisons.

638

Figure 4. Pathway impact analysis. Pathways with significant impacts and their

639

associated corrected p-values for the differentially expressed proteins for the comparisons

640

of DHA vs control and ARA vs control.

29

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

641

Figure 5. Effect of DHA extract on HT-29 cancer cell Proteasome pathway (KEGG:

642

03050). (a) Proteasome pathway diagram overlaid with the gene’s measured fold change.

643

(b) Gene perturbation bar plot for the Proteasome pathway; differentially expressed genes

644

are represented with negative values in blue; positive values are in red.

645

Figure 6. Effect of ARA extract on HT-29 cancer cell DNA replication pathway

646

(KEGG: 03030). (a) DNA replication pathway diagram overlaid with the gene’s measured

647

fold change. (b) Gene perturbation bar plot for the DNA replication pathway; differentially

648

expressed genes are represented with negative values in blue; positive values are in red.

649 650 651 652 653 654 655 656 657 658 659 660 661 30

ACS Paragon Plus Environment

Page 30 of 36

Page 31 of 36

662

Journal of Agricultural and Food Chemistry

Figure 1.

663

664 665 666

Figure 2.

667 668 669

31

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

670

Figure 3.

671 672

32

ACS Paragon Plus Environment

Page 32 of 36

Page 33 of 36

673

Journal of Agricultural and Food Chemistry

Figure 4.

674 675 676 677 678 679 680 681 682 683 684 685 686 687 688

33

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

689

Figure 5.

690 691 692 693 694 695

34

ACS Paragon Plus Environment

Page 34 of 36

Page 35 of 36

696

Journal of Agricultural and Food Chemistry

Figure 6.

697 698 699 700 701 702 703 704 705 706

35

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

707

Graphic for table of contents

708 709

710 711 712 713 714 715 716 717 718 719 720 36

ACS Paragon Plus Environment

Page 36 of 36