Accepted Manuscript Title: A liquid chromatography-tandem mass spectrometry-based method for the simultaneous determination of hydroxy sterols and bile acids Author: Clara John Philipp Werner Anna Worthmann Katrin Wegner Klaus T¨odter Ludger Scheja Sascha Rohn Joerg Heeren Markus Fischer PII: DOI: Reference:

S0021-9673(14)01655-0 http://dx.doi.org/doi:10.1016/j.chroma.2014.10.064 CHROMA 355942

To appear in:

Journal of Chromatography A

Received date: Revised date: Accepted date:

5-9-2014 20-10-2014 21-10-2014

Please cite this article as: C. John, P. Werner, A. Worthmann, K. Wegner, K. T¨odter, L. Scheja, S. Rohn, J. Heeren, M. Fischer, A liquid chromatographytandem mass spectrometry-based method for the simultaneous determination of hydroxy sterols and bile acids, Journal of Chromatography A (2014), http://dx.doi.org/10.1016/j.chroma.2014.10.064 This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. 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.

1

A liquid chromatography-tandem mass spectrometry-based method for the simultaneous de-

2

termination of hydroxy sterols and bile acids

3 4

Clara John1*, Philipp Werner2*, Anna Worthmann1, Katrin Wegner2, Klaus Tödter1, Ludger Scheja1,

5

Sascha Rohn2, Joerg Heeren1# and Markus Fischer2#

6

* These authors contributed equally

ip t

7 8

1

9

Eppendorf, Martinistr. 52, 20246 Hamburg; 2Hamburg School of Food Science, Institute of Food

Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-

Chemistry, University of Hamburg, Grindelallee 117, 20146 Hamburg, Germany

11

#

12

Markus Fischer, Hamburg School of Food Science, Institute of Food Chemistry, University of Ham-

13

burg, Grindelallee 117, 20146 Hamburg, Germany. Phone: +49(0)40-42838-4342, Email: mar-

14

[email protected]

15

Joerg Heeren, University Medical Center Hamburg-Eppendorf, Dept. of Biochemistry and Molecular

16

Cell Biology, Martinistraße 52, 20246 Hamburg, Germany. Phone: +49(0)40-4710-54745, Email:

17

[email protected]

cr

10

M

an

us

Corresponding Authors:

18

Abstract

22

Recently, hydroxy sterols and bile acids have gained growing interest as they are important regulators

23

of energy homoeostasis and inflammation. The high number of different hydroxy sterols and bile acid

24

species requires powerful analytical tools to quantify these structurally and chemically similar ana-

25

lytes. Here, we introduce a liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based

26

method for rapid quantification of 34 sterols (hydroxy sterols, primary, secondary bile acids as well as

27

their taurine and glycine conjugates). Chromatographic baseline separation of isomeric hydroxy sterols

28

and bile acids is obtained using a rugged amide embedded C18 (polar embedded) stationary phase.

29

The current method features a simple extraction protocol validated for blood plasma, urine, gall blad-

30

der, liver, feces, and adipose tissue avoiding solid phase extraction as well as derivatization proce-

31

dures. The total extraction recovery for representative analytes ranged between 58-86 % in plasma, 85

32

% in urine, 79-92 % in liver, 76-98 % in adipose tissue, 93-104 % in feces and 62-79 % in gall blad-

33

der. The validation procedure demonstrated that the calibration curves were linear over the selected

34

concentration ranges for 97 % of the analytes, with calculated coefficients of determination (R2) of

35

greater than 0.99. A feeding study in wild type mice with a standard chow and a cholesterol-enriched

36

Western type diet illustrated that the protocol described here provides a powerful tool to simultane-

Ac ce

pt

ed

19 20 21

Page 1 of 21

ously quantify cholesterol derivatives and bile acids in metabolically active tissues and to follow the

38

enterohepatic circulation.

39

1. Introduction

40

Sterols are apolar lipids formed by four fused rings resulting in an inflexible core. While plants syn-

41

thesize diverse phytosterols, cholesterol is the most abundant sterol synthesized in animal cells. Its

42

chemical structure allows cholesterol to serve as a component of membranes, facilitating membrane

43

fluidity, as well as a precursor of steroid hormones and other signaling molecules. In carnivores, cho-

44

lesterol supply is provided either by diet or by de novo synthesis [1,2].

45

High intracellular cholesterol levels are cytotoxic and a disturbed cholesterol metabolism observed in

46

patients with hypercholesterolemia is causally associated with the development of cardiovascular dis-

47

ease [3]. An important way to get rid of excess cholesterol is the biliary excretion of either non-

48

esterified cholesterol or its hydrophilic derivatives, the bile acids (BA) [4,5]Besides their role as in-

49

termediates in the BA synthesis pathway, hydroxy sterols are well known agonists for the transcription

50

factor liver X receptor and the estrogen receptor α (ERα), thereby regulating systemic lipid and energy

51

metabolism [6] as well as inflammatory processes known to be important for the development and

52

progression of chronic metabolic diseases such as cardiovascular disease, type 2 diabetes mellitus and

53

cancer [7-9]. In a multi-step reaction catalyzed by enzymes located in the liver [5,10-14], hydroxy

54

sterols are converted into primary bile acids (PBA). In humans, cholic acid (CA) and chenodeoxy-

55

cholic acid (CDCA) are the main primary bile acids whereas CA and muricholic acid (MCA) pre-

56

dominate in mice [5]. Further conjugation to taurine (predominant in rodents) or glycine (predominant

57

in humans) increases hydrophilicity. Via bile ducts BA reach the intestine where they facilitate absorp-

58

tion of dietary fats. Secondary bile acids (SBA) arise from action of the gut microbiota [4]. Besides

59

their function in fat absorption, BA have signaling functions, acting as agonists and antagonists of the

60

transcription factor farnesoid X receptor (FXR) and G-protein coupled bile acid receptor 1 (GBAR1

61

also known as TGR5). These receptors mediate bile acid, lipid, and glucose homeostasis as well as

62

energy expenditure [6,15-18]. The multifunctional presence of hydroxy sterols and bile acids in differ-

63

ent biochemical pathways shows that their quantitative observation in different biofluids and organs is

64

of great scientific relevance, especially with respect to the study of chronic inflammatory metabolic

65

diseases. Unfortunately, there are very few analytical methods for the determination of hydroxy sterols

66

and bile acids in one analytical approach. A bottleneck to the analysis of hydroxy sterols and bile acids

67

is the chromatographic separation prior to analysis via mass-spectrometric detection. As many of the

68

analytes are isobaric and very similar in their structure, no selective multiple reaction monitoring-

69

(MRM) transitions can be obtained. This results in the need for chromatographic baseline separation.

70

In the past, this problem has often been solved by determining very similar compounds as a sum pa-

71

rameter or via gas chromatography (GC)-MS-approaches [19]. GC-based applications are able to sepa-

72

rate positional isomeric sterol compounds but need further sample preparation steps including complex

Ac ce

pt

ed

M

an

us

cr

ip t

37

Page 2 of 21

derivatization techniques. As sterols and their derivatives can have very different biochemical func-

74

tions, these procedures are not purposeful for some scientific research questions [20,21].

75

In this work, our aim was to provide a method for the quantitative determination of 34 sterols in bio-

76

logical matrices using high performance liquid chromatography electrospray ionization triple quadru-

77

pol tandem mass spectrometry (HPLC-ESI-QqQ-MS/MS). In contrast to others [22-26] our method

78

offers the simultaneous determination of hydroxy sterols and bile acids in one single run, in addition to

79

the advantage of a quick and easy-to-use sample preparation and clean-up without further complex

80

derivatization techniques. The chromatographic baseline separation of isomeric hydroxy sterols and

81

bile acids is obtained using a rugged amide embedded C18 (polar embedded) stationary phase. In this

82

paper, the method is validated in 6 biological matrices and additionally, applicability is demonstrated

83

in a mouse feeding study. The method achieved a high dynamic range in different biological matrices

84

and was successfully applied for the determination of sterols in blood plasma, urine, bile, feces, adi-

85

pose tissue and liver.

86

2. Theory

87

All sterols are based on the same steroid with a hydroxyl-group at the 3-position of the A-ring and

88

often only differ in the structural position of mainly polar functional groups. As shown in Figure 1,

89

this results in many isobaric compounds that cannot be determined quantitatively via MRM-MS-

90

applications due to identical precursor- and fragment-masses without prior baseline separation [22].

91

The steroid backbone is mainly of nonpolar character, thus liquid chromatography is performed using

92

reversed phase conditions. On the one hand, this results in good retardation for nearly all steroid com-

93

pounds but selectivity is often poor when it comes to isobaric structures only differing in the position-

94

ing of a polar functional group (Figure 1). On the other hand, the application of mainly polar station-

95

ary phases like hydrophilic interaction liquid chromatography- (HILIC) columns often shows elution

96

with the solvent front or is characterized by poor selectivity. In order to separate isobaric sterols by

97

liquid chromatography, an approach requires a stationary phase with hydrophobic properties such as a

98

C18 reversed phase- (RP) that is combined with polar selectivity. In principle, there are two types of

99

RP-based stationary phases with polar selectivity: polar endcapped and polar embedded phases. For

100

endcapping, accessible peripheral silanol groups of the C18-RP are endcapped with polar groups. For

101

polar embedding, the polar groups are located within the alkyl-chain. Figure 2 shows a schematic rep-

102

resentation of the fundamental construction of a polar embedded stationary phase. Polar carbamate-,

103

urea-, ether- and amide groups are commonly used to increase the polar selectivity that is mainly me-

104

diated by hydrogen bonding interactions [27].

105

3. Materials and methods

106

3.1 Chemicals

107

Methanol (LC-MS-grade) and ammonium acetate (≥ 97 %, p.a.) were purchased from Carl Roth

108

(Karlsruhe, Germany). LC-MS grade water was obtained from Merck Millipore Ultra-pure water puri-

109

fication system (Merck Millipore, Darmstadt, Germany). Formic acid (FA) was of 99+ % purity and

Ac ce

pt

ed

M

an

us

cr

ip t

73

Page 3 of 21

obtained from Acros Organics (Geel, Belgium). The following bile acids: CDCA, UDCA, HDCA, α-

111

MCA, β-MCA, ω-MCA, GLCA, GUDCA, TLCA, TUDCA, THDCA, T-α-MCA, T-β-MCA and d4-

112

GCDCA and following steroid hormones: progesterone, pregnanolone, pregnanedione and THDOC

113

were purchased from Steraloids, Inc. (Newport, RI, USA). All further standards including TCA, GCA,

114

GCDCA, GDCA, TCDCA, DOC, CA, DCA, TDCA, stigmasterol, and dried bovine bile were pur-

115

chased from Sigma-Aldrich Chemie GmbH (Munich, Germany). 7-OH-Chol, 22-S-OH-Chol, 24-S-

116

OH-Chol, d6-25-OH-Chol, 27-OH-Chol-, 25-OH-Chol, d7-Chol, cholesterol and desmosterol were

117

obtained from Avanti Polar Lipids, Inc. (Alabaster, AL, USA). All abbreviations are shown in Table 1.

118

3.2 Standard solutions and calibration

119

Standard solutions of individual bile acids and steroid hormones were prepared by dissolving the re-

120

spective compounds separately in methanol, whereas sterol standards including the deuterated internal

121

standards d4-GCDCA, d6-25-OH-Chol, d7-Chol were prepared in Folch reagent [28]. All solutions

122

were stored at -20 °C. A nine-point calibration curve was constructed. For every calibration point, the

123

internal standard (IS) was used at a final concentration of 10 μM for d4-GCDCA, 10 μM for d6-25-

124

OH-Chol and 100 μM for d7-Chol, respectively. For quantification of bile acids and steroid hormones,

125

calculation of analyte levels was performed based on the internal standard d4-GCDCA. The sterols 7- ,

126

22-S-, 24-S-, 25- and 27-OH-Chol were corrected for d6-25-OH-Chol whereas levels for cholesterol,

127

desmosterol, stigmasterol, sitosterol are based on the internal standard d7-Chol.

128

3.3 Sample preparation

129

3.3.1 Solid phase extraction

130

Solid phase extraction was performed according to the method of Lund and Diczfalusy [29]. To moni-

131

tor the best recovery rates of representative analytes (cholic acid, cholesterol, stigmasterol), eleven

132

different compositions of solvents (compare Suppl. Table 1) used for washing and elution were tested.

133

3.3.2 Extraction

134

EDTA-plasma and urine samples were obtained from healthy volunteers. Samples were stored at

135

-20°C until analysis. 70 μL MeOH and 10 μL IS-mix containing 100 μM d4-GCDCA, 100 μM d6-25-

136

OH-Chol and 1000 μM d7-Chol were added to 20 μL plasma. The samples were vortexed for three

137

times, shaken continuously over a period of 10 minutes and centrifuged (12,000 g, 10 minutes). The

138

supernatants were transferred into glass vials and analyzed by LC-MS/MS. Urine samples (20 µl)

139

spiked with 10 µl of IS were processed similarly as plasma samples.

140

Liver and adipose tissue: 150 mg of liver or adipose tissues obtained from fasted wild type C57BL/6J

141

mice were spiked with 10 μL IS-solution and mixed with 3 mL methanol followed by homogenization

142

with an Ultra-Turrax. After 10 minutes of centrifugation (12,000 g), supernatants were transferred to

143

new glass vials and evaporated in vacuo until dryness. The dried lipid-containing samples were dis-

144

solved in 100 μL methanol.

145

Feces: Fecal samples obtained from mice were lyophilized and then dried samples were pestled using

146

a mortar. 5 mg of the homogenic powder were extracted with 1000 μL MeOH containing IS-mixture

Ac ce

pt

ed

M

an

us

cr

ip t

110

Page 4 of 21

(100 μmol/L d4-GCDCA, 100 μmol/L d6-25-OH-Chol, 1000 μmol/L d7-Chol). After sonication the

148

samples were centrifuged and the supernatants were used for LC-MS/MS analysis.

149

3.4 LC-MS-analysis

150

Liquid chromatography was performed on a Accucore™ Polar Premium HPLC column (2.6 μm, 150

151

mm x 2.1 mm i.d.), equipped with an Accucore™ Polar Premium defender guard column (Thermo

152

Fischer Scientific Inc., Waltham, MA, USA), at 20°C with a flow rate of 300 μL/min using an Agilent

153

1200 Infinity Quaternary LC System (Agilent Technologies, Waldbronn, Germany). The mobile phase

154

A was water, and B was MeOH, both containing 0.1 % formic acid and ammonium acetate at 5 mM.

155

The gradient elution started with 40 % A for 2 minutes, linearly increased to 100 % B in 18 minutes,

156

which was kept constant for 20 minutes and brought back to 40 % A in 3 minutes followed by 13 min-

157

utes of re-equilibration. For matrix abundant samples, step 3 was extended for another period of 10

158

minutes. The injection volume for all samples was 5 μL. For detection a QqQ-MS/MS API 4000 Q

159

trap (Applied Biosystems, Darmstadt, Germany) equipped with a turbo ion spray source, operating in

160

positive ion mode was used with the following mass spectrometer settings: ion spray voltage = 4500

161

V; ion source heater = 550°C; source gas 1 = 40 psi; source gas 2 = 50 psi and curtain gas = 20 psi.

162

Analytes were identified by their chromatographic characteristics and their specific fragmentations,

163

using mass transitions between precursor and product ions for each analyte (Table 1). The entire LC-

164

MS/MS-system was controlled using Analyst 1.6.1 software (AB Sciex, Concord, Ontario, Canada).

165

3.5 Method validation

166

Validation of the method was performed either in absence (base calibration) or presence (matrix cali-

167

bration) of the following matrices: plasma, bile, urine, liver, fat and feces. Linearity, recovery, lower

168

limit of quantification (LLoQ), precision and accuracy were assessed in analogy to accepted guidelines

169

[30].

170

Linearity: Linearity was determined by analysis of calibration curves for all commercially available

171

standards of bile acids/ hydroxy sterols. The method was validated using a nine-point calibration curve

172

(n = 5). To asses matrix effects, fixed volumes of each matrix were spiked three times (n = 3) with

173

different volumes of the appropriate mix-standard solution to construct a calibration curve of six cali-

174

bration points. The concentration ranges for baseline calibration are shown in Table 2.

175

Recovery: To evaluate the efficiency of the extraction procedure a known concentration of IS-mixture

176

was added to all matrices before and after extraction (n = 3). The samples were treated as described in

177

section 3.2. The integrated average peak area ratio of each analyte before extraction was compared to

178

the corresponding peak area ratio in samples after extraction. The recovery rate (R) was determined as

179

R (%) = concentrationbefore/concentrationafter x 100.

180

Lower limit of quantification:

181

The lower limit of quantification has been set as the lowest standard on the calibration curve as it met

182

the following conditions: The analyte response was at least 5 times the response compared to a blank

183

response and the analyte peak is identifiable, discrete, and reproducible.

Ac ce

pt

ed

M

an

us

cr

ip t

147

Page 5 of 21

Precision and accuracy:

185

Precision was obtained determining the coefficient of variation of every analyte of 15 samples on 3

186

different concentration levels.

187

Accuracy was obtained determining the concentration of every analyte of 15 samples on 3 different

188

concentration levels. The maximal variance between the value determined and the true value is 15 %

189

or 20 % at the LLOQ.

190

3.6 Experimental animals and diets

191

All animal experiments were approved by the Animal Welfare Officers of University Medical Center

192

Hamburg-Eppendorf and Behörde für Gesundheit und Verbraucherschutz Hamburg. Wild type

193

C57BL/6J mice were bred in the animal facility of Universal Medical Center Hamburg-Eppendorf

194

with a day and night cycle of 12 hours with ad libitum access to food and water. Male age-matched

195

(12 weeks) mice were housed at 22°-24°C in single cages and fed either a standard chow diet or a

196

Western Type diet (WTD) containing 21 % total fat and 0.2 % cholesterol (ssniff EF R/M acc.

197

TD88137 mod.) for one week. At day 6, cages were cleaned and feces were collected on day 7 after a

198

24 hours period. Blood samples were collected in EDTA-coated tubes (Sarstedt, Nümbrecht, Deutsch-

199

land) by cardiac puncture of anaesthetized mice after 4 hours of fasting on day 7. Then, organs were

200

harvested after systemic perfusion with phosphate buffered saline (PBS, Life Technologies, Darm-

201

stadt, Germany) via the left heart ventricle and immediately stored at -80°C.

202

4. Results and discussion

203

As bile acids and hydroxy sterols are important regulators of cellular metabolism and inflammatory

204

processes [6-9] their simultaneous determination is of great interest. Up to our knowledge this is the

205

first LC-MS-approach combining the determination of bile acids and hydroxy sterols.

206

4.1

207

Previously described methods predominantly apply GC to separate positional isomeric sterol com-

208

pounds. While this may result in very good separation results, further sample preparation steps includ-

209

ing complex derivatization techniques are needed [31,32]. The advantage of using polar-embedded-

210

LC-MS-techniques for this analytical challenge is that baseline separation and detection of positional

211

isomeric sterol compounds can be achieved without any preceding derivatization steps. To obtain

212

MRM-transitions, 37 single compound solutions (0.1 mM in methanol enriched with 5 mM NH4Ac)

213

were directly injected into the ESI source in positive ionization mode. NH4Ac supplementation

214

thereby increased signal intensity of the analytes in positive ionization mode. Optimization of the dif-

215

ferent MS-parameters was accomplished by means of the auto tuning mode provided by the Analyst

216

1.6. software. Due to the use of NH4Ac as modifier, ammonia adducts were predominantly detected as

217

most abundant precursor ions (Table 1). Fragmentation resulted in the release of either H2O (m/z 18),

218

NH3 + H2O (m/z 35) or NH3 + 2 H2O (m/z 53) (Table 1, Figure 3). As described above, analyzed

219

compounds share obvious structural similarities and are partially isobaric, and therefore fragmentize in

220

a similar or even identical manner (Figure 3). Therefore, high-resolution separation is obligatory to

Ac ce

LC-MS-analyses

pt

ed

M

an

us

cr

ip t

184

Page 6 of 21

identify single sterol derivatives. The substantial improvement of chromatographic separation of the

222

embedded RP18 column in comparison to a conventional RP18 column is shown in Figure 2 a, b. No-

223

tably, most prominent differences are observed in chromatographic resolution of isobaric muricholates

224

as well as hydroxy sterols (Figure 2 a insets). In general, peak performance was enhanced strongly

225

compared to the RP18 column. Best peak performance was achieved by adding 5 mM NH4Ac and 0.1

226

% FA as modifiers to mobile phases (Figure 2 a-b). In order to combine demands of sensitivity and

227

sufficient data acquisition rates the MRM method was split into three periods.

228

4.2

229

In contrast to methods employing solid phase extraction (SPE) as a classical procedure [22,26,29] we

230

used a simpler liquid-liquid-methanol extraction protocol, as we observed small recovery rates for

231

representative analytes such as cholesterol or cholic acid when performing previously described SPE

232

procedures on normal-phase SPE cartridges (Suppl. Figure 1). Normal-phase SPE was used in order to

233

reduce nonpolar compounds, such as triacylglycerols and cholesterol esters and, in addition, to apply a

234

complementary sample cleanup prior to a reversed phase analytical method. The obtained data shows

235

that a selective sample cleanup via SPE for both, bile acids and hydroxy sterols is not possible in a

236

single SPE run. However, using a simple methanol extraction we obtained good recovery rates for

237

representative analytes, even in the presence of complex matrices (Suppl. Table 2, Figure 5). For re-

238

covery analyses, we added known concentrations of the internal standards to 6 different matrices either

239

before or after extraction, performed as described in Section 3. For 94 % of the representative analytes

240

and the different matrices the recovery was above 75 % (Suppl. Table 3).

241

The obtained results indicate that simple and fast liquid-liquid-extraction protocols can be an efficient

242

alternative to laborious SPE approaches.

243

4.3

244

Validation of the method was performed either in absence (base calibration) or presence (matrix cali-

245

bration) of the following matrices: plasma, bile, urine, liver, adipose tissue, feces. As many published

246

methods apply complex sample clean-ups such as SPE [22,26], it was important to show that the liq-

247

uid-liquid extraction used here leads to reproducible, reasonable, and precise results.

ip t

221

Method validation

Ac ce

248

pt

ed

M

an

us

cr

Sample preparation

249

4.3.1

250

97 % of all analytes showed acceptable linearity (R > 0.99) in a broad dynamic range of 10 - 500- fold

251

(Table 2). Representative calibration curves and corresponding residual plots for UDCA and 7-OH-

252

Cholesterol are shown in Figures 4 a and 4 b, respectively. According to the guidelines of the FDA the

253

coefficient of variation (CV) was < 15 % for ~ ¾ of the analytes in base calibration. Depending on the

254

respective analyte, the lower limit of quantification ranged between 9 nM and 2 µM (Table 2).

255

4.3.2

256

To overcome matrix effects, linearity responses were studied in blank matrices (urine, EDTA-plasma,

257

feces, bile, fat and liver) spiked with mix-standard solution and IS-mixture. The correlation coeffi-

Baseline calibration

Matrix calibration

Page 7 of 21

258

cients of all the calibration curves in the different matrices were at least 0.999 for 82 % of the analytes

259

(see Suppl. Table 3 a-f). CV was between 2.5 % and 15 % for ⅔ of the analytes, which demonstrated a

260

minor influence of matrix effects on our method. The calibration data involving calibration curve

261

equations, curve parameters, and R2 values are presented for each matrix in Suppl. Table 3 a-f.

262

4.3.3

263

The obtained values for precision and accuracy (Table 3) at the LLOQ were below 20 % for 90 % of

264

the analytes. The values for expected concentrations were below 15 % for 90 % the analytes. Thus

265

almost all values met the conditions stated by the FDA [30].

266

4.4

267

As bile acids and hydroxy sterols have recently gained growing interest [4,15,18,33], our method to

268

rapidly quantify cholesterol intermediates of the de novo synthesis and excretion pathways provides a

269

powerful tool to depict and trace the metabolic state of different tissues and to trail enterohepatic cir-

270

culation. As described before [15,33] diet influences the bile acid pool in mice. Therefore, body fluids,

271

feces and organs of wild type C57BL/6J mice fed either a normal chow or a WTD were processed to

272

quantify sterols and its bile acid derivatives. Our observed concentrations and composition of bile

273

acids and hydroxy sterols (Figure 5) are in line with previous studies using GC-MS- or LC-MS-based

274

methods [7,16,17,24,25,33,34]. We did observe that concentrations for bile acids in feces were quite

275

variable. One reason for this is the strong impact of the diverse microbiome on fecal bile acid content

276

and composition [16,35,36]. As shown in Figure 5 (log scale) and Suppl. Figure 2 (linear scale), the

277

diet influences the concentration of specific hydroxylated sterols and bile acids in liver, adipose tissue,

278

feces, and plasma (Figure 5a-d) but not in gall bladder (Suppl. Figure 3). The most prominent change

279

was a decreased concentration of stigmasterol in feces of WTD-fed mice (Figure 5 c). The difference

280

in stigmasterol can be explained by the composition of the diet as the plant based chow diet contains a

281

higher content of phytosterols compared to the milk fat-based WTD. Although the WTD is enriched in

282

cholesterol, we could only detect a significant increase of cholesterol in WAT after WTD feeding

283

(Figure 5 b). As expected, WAT contained the lowest concentrations of bile acids in comparison to the

284

other analyzed organs (Figure 5 a-d). Notably, even in tissues with a low content of bile acids such as

285

the adipose tissue, most bile acid species could be detected. The broad spectrum of matrices applicable

286

to this method results in a comprehensive picture of systemic bile acid and hydroxy sterol metabolism.

287

This opens the perspective to delineate the functional roles of sterols and bile acids in the regulation of

288

metabolism via G-protein coupled receptors and ligand-activated transcription factors. Moreover this

289

method might have the potential to be implemented in clinical screening approaches and patient moni-

290

toring in the future.

291

5

292

Here, we describe a simple, effective and sensitive LC-MS/MS-based protocol to quantify 34 sterols

293

and bile acids using a polar embedded stationary phase. It is highly applicable to different biological

294

compartments such as blood, urine, feces, bile, liver and adipose tissue. The simultaneous quantifica-

ip t

Precision and accuracy

Ac ce

pt

ed

M

an

us

cr

Method application

Conclusion

Page 8 of 21

295

tion of bile acids, sterols and even isobaric hydroxylated cholesterol derivatives provides a powerful

296

tool for the investigation of cholesterol utilizing metabolic pathways in response to environmental or

297

genetic alterations.

298

[7]

[8] [9] [10] [11] [12] [13] [14] [15]

[16]

[17]

ip t

cr

us

[6]

an

[5]

M

[4]

ed

[3]

K. Bloch, The biological synthesis of cholesterol, Science 150 (1965) 19-28. A.A. Kandutsch, A.E. Russell, Preputial gland tumor sterols. 3. A metabolic pathway from lanosterol to cholesterol, Journal of Biological Chemistry 235 (1960) 2256-2261. F.R. Maxfield, I. Tabas, Role of cholesterol and lipid organization in disease, Nature 438 (2005) 612-621. F. Kuipers, V.W. Bloks, A.K. Groen, Beyond intestinal soap-bile acids in metabolic control, Nat Rev Endocrinol (2014). D.W. Russell, The enzymes, regulation, and genetics of bile acid synthesis, Annu Rev Biochem 72 (2003) 137-174. A.C. Calkin, P. Tontonoz, Transcriptional integration of metabolism by the nuclear sterolactivated receptors LXR and FXR, Nat Rev Mol Cell Biol 13 (2012) 213-224. M. Umetani, P. Ghosh, T. Ishikawa, J. Umetani, M. Ahmed, C. Mineo, P.W. Shaul, The Cholesterol Metabolite 27-Hydroxycholesterol Promotes Atherosclerosis via Proinflammatory Processes Mediated by Estrogen Receptor Alpha, Cell Metab 20 (2014) 172182. A. Reboldi, E.V. Dang, J.G. McDonald, L. Guosheng, D.W. Russell, J.G. Cyster, 25Hydroxycholesterol suppresses interleukin-1-driven inflammation downstream of type I interferon, Science 345 (2014) 679-684. E.R. Nelson, S.E. Wardell, J.S. Jasper, S. Park, S. Suchindran, M.K. Howe, N.J. Carver, R.V. Pillai, P.M. Sullivan, V. Sondhi, M. Umetani, J. Geradts, D.P. McDonnell, 27-Hydroxycholesterol links hypercholesterolemia and breast cancer pathophysiology, Science 342 (2013) 1094-1098. W. Song, W.M. Pierce, Jr., R.A. Prough, R.N. Redinger, Characteristics of cholesterol 7 alphahydroxylase and 7 alpha-hydroxycholesterol hydroxylase activities of rodent liver, Biochem Pharmacol 41 (1991) 1439-1447. N.B. Javitt, Biologic role(s) of the 25(R),26-hydroxycholesterol metabolic pathway, Biochim Biophys Acta 1529 (2000) 136-141. E.G. Lund, J.M. Guileyardo, D.W. Russell, cDNA cloning of cholesterol 24-hydroxylase, a mediator of cholesterol homeostasis in the brain, Proc Natl Acad Sci U S A 96 (1999) 72387243. E. Lund, I. Bjorkhem, C. Furster, K. Wikvall, 24-, 25- and 27-hydroxylation of cholesterol by a purified preparation of 27-hydroxylase from pig liver, Biochim Biophys Acta 1166 (1993) 177182. J. Li-Hawkins, E.G. Lund, A.D. Bronson, D.W. Russell, Expression cloning of an oxysterol 7 alpha-hydroxylase selective for 24-hydroxycholesterol, Journal of Biological Chemistry 275 (2000) 16543-16549. M. Watanabe, S.M. Houten, C. Mataki, M.A. Christoffolete, B.W. Kim, H. Sato, N. Messaddeq, J.W. Harney, O. Ezaki, T. Kodama, K. Schoonjans, A.C. Bianco, J. Auwerx, Bile acids induce energy expenditure by promoting intracellular thyroid hormone activation, Nature 439 (2006) 484-489. S.I. Sayin, A. Wahlstrom, J. Felin, S. Jantti, H.U. Marschall, K. Bamberg, B. Angelin, T. Hyotylainen, M. Oresic, F. Backhed, Gut microbiota regulates bile acid metabolism by reducing the levels of tauro-beta-muricholic acid, a naturally occurring FXR antagonist, Cell Metab 17 (2013) 225-235. F. Li, C. Jiang, K.W. Krausz, Y. Li, I. Albert, H. Hao, K.M. Fabre, J.B. Mitchell, A.D. Patterson, F.J. Gonzalez, Microbiome remodelling leads to inhibition of intestinal farnesoid X receptor signalling and decreased obesity, Nat Commun 4 (2013) 2384.

pt

[1] [2]

Ac ce

299 300 301 302 303 304 305 306 307 308 309 310 311 312 313 314 315 316 317 318 319 320 321 322 323 324 325 326 327 328 329 330 331 332 333 334 335 336 337 338 339 340 341 342 343 344

Page 9 of 21

[24] [25]

[26] [27] [28] [29] [30] [31] [32] [33] [34] [35] [36]

ip t

cr

[23]

us

[22]

an

[21]

M

[20]

ed

[19]

K. Ryan, V. Tremaroli, C. Clemmensen, P. Kovatcheva-Datchary, A. Myronovych, R. Karns, H. Wilson-Pérez, D. Sandoval, R. Kohli, F. Bäckhed, R. Seeley, FXR is a molecular target for the effects of vertical sleeve gastrectomy, Nature 509 (2014) 183-188. J. Li-Hawkins, M. Gafvels, M. Olin, E.G. Lund, U. Andersson, G. Schuster, I. Bjorkhem, D.W. Russell, G. Eggertsen, Cholic acid mediates negative feedback regulation of bile acid synthesis in mice, Journal of Clinical Investigation 110 (2002) 1191-1200. X. Hu, Y. Bonde, G. Eggertsen, M. Rudling, Muricholic bile acids are potent regulators of bile acid synthesis via a positive feedback mechanism, J Intern Med 275 (2014) 27-38. D.Q. Wang, S. Tazuma, D.E. Cohen, M.C. Carey, Feeding natural hydrophilic bile acids inhibits intestinal cholesterol absorption: studies in the gallstone-susceptible mouse, Am J Physiol Gastrointest Liver Physiol 285 (2003) G494-502. J.G. McDonald, B.M. Thompson, E.C. McCrum, D.W. Russell, Extraction and analysis of sterols in biological matrices by high performance liquid chromatography electrospray ionization mass spectrometry, Methods Enzymol 432 (2007) 145-170. J. Acimovic, A. Lovgren-Sandblom, K. Monostory, D. Rozman, M. Golicnik, D. Lutjohann, I. Bjorkhem, Combined gas chromatographic/mass spectrometric analysis of cholesterol precursors and plant sterols in cultured cells, J Chromatogr B Analyt Technol Biomed Life Sci 877 (2009) 2081-2086. J.C. Garcia-Canaveras, M.T. Donato, C.J. V., A. Lahoz, Targted profiling of circulating and hepatic bile acids in human, mouse, and rat usimng a UPLC-MRM-MS-validated method, Journal of Lipid Research 53 (2012). S.J. Tsai, Y.S. Zhong, J.F. Weng, H.H. Huang, P.Y. Hsieh, Determination of bile acids in pig liver, pig kidney and bovine liver by gas chromatography-chemical ionization tandem mass spectrometry with total ion chromatograms and extraction ion chromatograms, J Chromatogr A 1218 (2011) 524-533. G. Kakiyama, A. Muto, H. Takei, H. Nittono, T. Murai, T. Kurosawa, A.F. Hofmann, W.M. Pandak, J.S. Bajaj, A simple and accurate HPLC method for fecal bile acid profile in healthy and cirrhotic subjects: validation by GC-MS and LC-MS, J Lipid Res 55 (2014) 978-990. J.W. Coym, Comparison of retention on traditional alkyl, polar endcapped, and polar embedded group stationary phases, J Sep Sci 31 (2008) 1712-1718. J. Folch, M. Lees, G.H. Sloane Stanley, A simple method for the isolation and purification of total lipides from animal tissues, Journal of Biological Chemistry 226 (1957) 497-509. E.G. Lund, U. Diczfalusy, Quantitation of receptor ligands by mass spectrometry, Methods Enzymol 364 (2003) 24-37. FDA, Guidance for Industry - Bioanalytical Method Validation, U.S. Departement of Health and Human Services (2001) Center for Drug Evaluation and Research, Center for Veterinary medicine. J.J. Birk, M. Dippold, G.L. Wiesenberg, B. Glaser, Combined quantification of faecal sterols, stanols, stanones and bile acids in soils and terrestrial sediments by gas chromatographymass spectrometry, J Chromatogr A 1242 (2012) 1-10. S. Matysik, G. Schmitz, Application of gas chromatography-triple quadrupole mass spectrometry to the determination of sterol components in biological samples in consideration of the ionization mode, Biochimie 95 (2013) 489-495. Y. Qi, C. Jiang, J. Cheng, K.W. Krausz, T. Li, J.M. Ferrell, F.J. Gonzalez, J.Y. Chiang, Bile acid signaling in lipid metabolism: Metabolomic and lipidomic analysis of lipid and bile acid markers linked to anti-obesity and anti-diabetes in mice, Biochim Biophys Acta (2014). M. Umetani, H. Domoto, A.K. Gormley, I.S. Yuhanna, C.L. Cummins, N.B. Javitt, K.S. Korach, P.W. Shaul, D.J. Mangelsdorf, 27-hydroxycholesterol is an endogenous SERM that inhibits the cardiovascular effects of estrogen, Nature Medicine 13 (2007) 1185-1192. L.A. David, C.F. Maurice, R.N. Carmody, D.B. Gootenberg, J.E. Button, B.E. Wolfe, A.V. Ling, A.S. Devlin, Y. Varma, M.A. Fischbach, S.B. Biddinger, R.J. Dutton, P.J. Turnbaugh, Diet rapidly and reproducibly alters the human gut microbiome, Nature 505 (2014) 559-563. P.J. Turnbaugh, Microbiology: fat, bile and gut microbes, Nature 487 (2012) 47-48.

pt

[18]

Ac ce

345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376 377 378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397

Page 10 of 21

398 ACKNOWLEDGEMENT

400

The authors thank Meike Kröger for excellent technical assistance. J.H. is supported by a grant from

401

the DFG (SFB 841: Liver inflammation: infection, immune regulation and consequences) and by EU

402

FP7 project RESOLVE (FP7-HEALTH-2012-305707).

ip t

399

403

cr

404

pt

ed

M

an

Figure 1 Structural isomerism of cholesterol-derived compounds; (a) Cholesterol, (b) (I) CA, (II) α-MCA, (III) ω-MCA, (IV) β-MCA, (c) (I) 22-OH-Chol (II) 27-OH-Chol, (III) 25-OH-Chol, (IV) 24-OH-Chol Figure 2 Total ion chromatograms (TIC) of a standard mixture containing 34 bile acids and oxidized sterol derivatives separated on a (a) polar embedded C18 column and (b) on a conventional C18 column. The polar embedded stationary phase (scheme a) is equipped with multiple polar groups which are either located within the C18 chain or on remaining free silica groups. The conventional C18 column contains only aliphatic alkyl chains (scheme b). Baseline separation of isobaric compounds such as bile acids (e.g. ω-MCA, α-MCA, β-MCA, and CA) as well as hydroxy sterols (e.g. 22-OH-Chol, 24-OHChol, 25-OH-Chol, 27-OH-Chol) is only achieved using the polar embedded column (compare (XICs) a and b). Figure 3 Electrospray ionization product ion mass spectra of nominated (a) bile acids and (b) sterols. A (I) αMCA, (II) β-MCA, (III) ω-MCA, (IV) CA; B (I) 22-S-OH-Chol, (II) 24-S-OH-Chol, (III) 25-OHChol, (IV) 27-OH-Chol Figure 4 Exemplary calibration curves and corresponding residual plots for (a) UDCA (base calibration), (b) 7OH-Cholesterol (base calibration), (c) UDCA (matrix calibration, feces) and (d) 7-OH-Cholesterol (matrix calibration, feces) Figure 5 Concentration of different oxidized cholesterol derivatives and bile acids in (a) Liver, (b) adipose tissue (AT), (c) feces, (d) plasma of wild type mice fed either a chow diet (open bars) or WTD (filled bars) for one week (data are expressed in mean +/- SEM, n = 5, ***P < 0.001, **P < 0.01, *P < 0.05).

Ac ce

406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429

us

405

430

Page 11 of 21

*Highlights (for review)

Ac

ce pt

ed

M

an

us

cr

ip t

1. Simultaneous quantification of 34 hydroxy sterols and bile acids via LC-ESI-MS/MS. 2. Rapid and simple sample clean-up without complex derivatization techniques or solid phase extraction. 3. Validated for six different biological matrices: plasma, liver, adipose tissue, urine, gall bladder and feces. 4. Chromatographic baseline separation of multiple isobaric compounds using a novel polar embedded stationary phase. 5. Good method application was illustrated taking the example of a mice feeding study.

Page 12 of 21

Ac

ce

pt

ed

M

an

us

cr

i

Figure 1

Page 13 of 21

Ac

ce

pt

ed

M

an

us

cr

i

Figure 2

Page 14 of 21

Ac ce p

te

d

M

an

us

cr

ip t

Figure 3

Page 15 of 21

Ac ce p

te

d

M

an

us

cr

ip t

Figure 4

Page 16 of 21

Ac ce p

te

d

M

an

us

cr

ip t

Figure 5

Page 17 of 21

Tables Table 1 Operating parameters in MRM-mode

C27H46O2

402.65

M + AA

C27H46O2

402.65

M + AA

C27H46O2

402.65

M-H2O

C24H40O4

392.57

M + H+

C27H46O

386.67

M + AA

Cholic acid (CA)

C24H40O5

408.57

M + AA

Deoxycholic acid (DCA) Desmosterol

C24H40O4

392.57

M + AA

C27H44O

384.64

M + AA

Desoxycorticosterone (DOC) Glycochenodeoxycholic acid (GCDCA) Glycocholic acid (GCA) Glycodeoxycholic acid (GDCA) Glycolithocholic acid (GLCA) Glycoursodeoxycholic acid (GUDCA) Hyodeoxycholic acid (HDCA) Pregnanedione

C21H30O3

330.46

M + H+

C26H43NO5

449.62

M + AA

C26H43NO6

465.62

M + AA

pt

ce

Ac C26H43NO5

449.62

M + AA

C26H43NO4

433.62

M + AA

C26H43NO5

449.62

M + AA

C24H40O4

392.57

M + AA

C21H32O2

316.48

M + H+

111 111 111 106 106 106 106 106 106 111 111 111 91 91 91 76 76 76 101 101 101 51 51 51 1 1 1 101 101 101 101 101 101 36 36 36 41 41 41 41 41 41 36 36 36 36 36 36 76 76 76 91 91 91

CE (V) 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10

CXP (V) 19 61 11 17 53 11 9 13 15 33 63 13 19 31 79 5 63 11 37 59 11 27 99 19 13 65 21 57 51 11 33 73 39 25 21 13 29 25 15 27 19 13 21 51 13 23 17 11 9 9 15 19 75 15

10 14 12 10 12 12 12 12 10 12 6 12 10 12 14 12 14 10 12 12 10 10 14 10 10 12 12 14 6 10 16 12 18 12 14 12 12 12 14 12 12 14 12 12 14 12 12 14 12 12 10 8 14 8

ip t

M + AA

EP (V)

cr

402.65

DP (V)

us

C27H46O2

22-S-Hydroxycholesterol (22-OH-Chol) 24-S-Hydroxycholesterol (24-OH-Chol) 25-Hydroxycholesterol (25-OH-Chol) 27-Hydroxycholesterol (27-OH-Chol) 7-Hydroxycholesterol (7-OH-Chol) Chenodeoxycholic acid (CDCA) Cholesterol (Chol)

Fragments (m/z) Q1 420.44 → 367.3 Q2 420.44 → 81.1 Qnt 420.44 → 385.3 Q1 420.39 → 367.3 Q2 420.39 → 69.1 Qnt 420.39 → 385.3 Q1 420.35 → 402.3 Q2 420.35 → 385.3 Qnt 420.35 → 367.3 Q1 420.38 → 161.1 Q2 420.38 → 81.1 Qnt 420.38 → 385.3 Q1 385.32 → 367.2 Q2 385.32 → 159.1 Qnt 385.32 → 91.0 Q1 393.30 → 375.2 Q2 393.30 → 81.1 Qnt 393.30 → 357.3 Q1 404.41 → 147.2 Q2 404.41 → 81.1 Qnt 404.41 → 369.3 Q1 426.34 → 355.2 Q2 426.34 → 91.0 Qnt 426.34 → 373.2 Q1 410.32 → 375.2 Q2 410.32 → 81.1 Qnt 410.32 → 67.0 Q1 402.40 → 81.0 Q2 402.40 → 95.1 Qnt 402.40 → 367.3 Q1 331.20 → 109.0 Q2 331.20 → 97.0 Qnt 331.20 → 79.0 Q1 467.41 → 414.2 Q2 467.41 → 432.2 Qnt 467.41 → 450.2 Q1 483.36 → 412.3 Q2 483.36 → 430.2 Qnt 483.36 → 466.2 Q1 467.39 → 414.3 Q2 467.39 → 432.3 Qnt 467.39 → 450.3 Q1 451.39 → 416.3 Q2 451.39 → 76.0 Qnt 451.39 → 434.3 Q1 467.40 → 414.2 Q2 467.40 → 432.2 Qnt 467.40 → 450.2 Q1 410.24 → 393.2 Q2 410.24 → 375.2 Qnt 410.24 → 357.2 Q1 317.24 → 299.2 Q2 317.24 → 281.2 Qnt 317.24 → 91.0

an

402.65

M+X+ (m/z) M + AA

M

M (g/mol)

ed

Molecular formula C27H46O2

Compound

Q1 = qualifier 1, Q2 = qualifier 2, Qnt = quantifier, M + AA = ammonia - adduct, M + H+ = proton - adduct, DP = declustering potential, Page EP = entrance potential, CE = collision energy, CXP = collision cell exit potential

18 of 21

Table 1 Operating parameters in MRM-mode

C21H30O2

314.46

M + H+

Stigmasterol

C29H48O

412.69

M + AA

M + AA

Q2 Qnt Q1

430.41 → 55.0 430.41 → 395.3 517.33 → 500.3

M + AA

Q2 Qnt Q1

517.33 → 482.2 517.33 → 464.3 533.33 → 462.2

M + AA

Q2 Qnt Q1

M + AA

Q2 Qnt Q1

M + AA

Q2 Qnt Q1

C26H45NO6S

Taurohyodeoxycholic acid (THDCA)

C26H45NO6S

Taurolithocholic acid (TLCA)

C26H45NO5S

Tauroursodeoxycholic acid (TUDCA)

C26H45NO6S

Tauro-αmuricholic acid (T-α-MCA)

C26H45NO7S

Tauro-βmuricholic acid (T-β-MCA)

C26H45NO7S

Tetrahydrodesoxycorticosterone (THDOC) Ursodeoxycholic acid (UDCA)

C21H34O4

ω-Muricholic acid (ω-MCA)

499.70

483.71

499.70

515.70

515.70

334.49

Ac

α-Muricholic acid (α-MCA) β-Muricholic acid (β-MCA)

499.70

71 71 76

10 10 10

19 27 31

14 14 14

533.33 → 498.2 533.33 → 516.2 517.35 → 464.3

76 76 71

10 10 10

21 17 29

14 16 14

517.35 → 482.2 517.35 → 500.2 517.32 → 464.2

71 71 71

10 10 10

23 15 23

14 16 14

517.32 → 482.3 517.32 → 500.2 501.35 → 484.2

71 71 76

10 10 10

19 13 13

14 14 14

M + AA

Q2 Qnt Q1

501.35 → 126.0 501.35 → 466.2 517.32 → 500.2

76 76 76

10 10 10

49 23 13

8 14 16

M + 2*AA

Q2 Qnt Q1

517.32 → 482.2 517.32 → 464.2 550.26 → 221.0

76 76 46

10 10 10

17 25 27

14 14 18

M + AA

Q2 Qnt Q1

550.26 → 73.0 550.26 → 533.3 533.25 → 516.2

46 46 76

10 10 10

89 9 15

12 16 16

Q2 Qnt Q1

533.25 → 498.2 533.25 → 480.3 352.27 → 317.1

76 76 41

10 10 10

19 25 19

14 14 8

Q2 Qnt

352.27 → 299.1 352.27 → 335.2

41 41

10 10

21 13

8 10

C24H40O4

392.57

Q1

410.18 → 81.0

21

10

65

12

Q2 Qnt Q1 Q2 Qnt Q1

410.18 → 91.1 410.18 → 357.2 426.31 → 355.3 426.31 → 391.3 426.31 → 373.3 426.29 → 373.2

21 21 41 41 41 56

10 10 10 10 10 10

99 19 25 13 17 19

14 10 10 12 10 10

Q2 Qnt Q1 Q2 Qnt

426.29 → 355.2 426.29 → 391.2 426.30 → 355.2 426.30 → 391.2 426.30 → 373.2

56 56 41 41 41

10 10 10 10 10

25 15 23 13 17

10 12 10 12 10

M + AA

M + AA

C24H40O5

408.57

M + AA

C24H40O5

408.57

M + AA

C24H40O5

408.57

M + AA

us

Taurodeoxycholic acid (TDCA)

515.70

8 10 8 18 18 16 6 8 12 16

an

C26H45NO7S

23 11 19 37 37 33 35 77 11 13

ed

Taurocholic acid (TCA)

499.70

CXP (V)

10 10 10

pt

C26H45NO6S

46 46 46 41 41 41 126

EP (V) CE (V) 10 10 10 10 10 10 10

126 126 71

ce

Taurochenodeoxycholic acid (TCDCA)

DP (V)

ip t

Progesterone

Fragments (m/z) Q1 336.34 → 283.2 Q2 336.34 → 319.2 Qnt 336.34 → 301.2 Q1 316.28 → 109.1 Q2 316.28 → 315.2 Qnt 316.28 → 97.0 Q1 430.41 → 83.1

cr

318.49

M+X+ (m/z) M + AA

M (g/mol)

M

Pregnanolone

Molecular formula C21H34O2

Compound

Q1 = qualifier 1, Q2 = qualifier 2, Qnt = quantifier, M + AA = ammonia - adduct, M + H+ = proton - adduct, DP = declustering potential, EP = entrance potential, CE = collision energy, CXP = collision cell exit potential

Page 19 of 21

22-S-OH-Chol 24-S-/25-OH-Chol 27-OH-Chol 7-OH-Chol CDCA/DCA Chol CA Desmosterol DOC GCDCA GCA GDCA GLCA GUDCA HDCA Pregnanedione Pregnanolone Progesterone Stigmasterol TCDCA TCA TDCA THDCA TLCA TUDCA T-α/β-MCA THDOC UDCA α-MCA β-MCA ω-MCA

0.999 0.997 0.996 0.999 0.999 0.991 0.999 0.958 0.999 0.998 0.999 0.999 0.999 0.998 0.994 0.995 0.998 0.999 0.997 0.999 0.999 0.999 0.999 0.998 0.998 0.999 0.996 0.996 0.999 0.999 0.999

y = 3,417x - 645 y = 1,888x - 80 y = 6,193x - 1,180 y = 964,882x - 27,753 y = 49,161x - 5,374 y = 3,831x + 2,253 y = 1E+06x - 56,934 y = 1,971x + 4,652 y = 1E+07x + 1E+06 y = 602,142x - 62,201 y = 1E+06x - 25,293 y =685,171x + 45,121 y = 515,926x - 3,676 y = 466,848x - 16,405 y = 12,264x + 6,158 y = 2.97E+06x + 79,433 y = 923,334x - 11,104 y = 3E06x + 15,618 y = 587x + 4,535 y = 1E+06x - 42,278 y = 1E+06x - 29,816 y = 803,720x - 21,728 y = 386,660x - 5,493 y = 3E+06x - 10,327 y = 694,933x - 20,462 y = 802,232x - 4,972 y = 2E+06x + 24,953 y = 124,576x + 3,094 y = 1E+06x - 19,493 y = 2E+06x - 970 y = 646,768x + 12,653

0.300 0.300 0.200 0.100 0.360 0.600 0.180 1.000 0.180 0.180 0.060 0.180 0.018 0.060 0.300 0.180 0.030 0.018 2.000 0.060 0.060 0.060 0.030 0.018 0.060 0.036 0.060 0.180 0.030 0.009 0.060

ed

pt

1,455 1,149 1,687 139,694 11,172 3,049 116,475 2,502 1,692,780 66,166 68,529 86,557 61,955 54,745 7,244 374,076 134,904 39,710 708 155,666 112,036 54,846 31,955 307,453 80,598 122,764 224,181 20,241 192,283 89,104 61,145

SD (µM)

CV (%) 0.43 0.61 0.27 0.14 0.23 0.80 0.11 1.27 0.11 0.11 0.06 0.13 0.12 0.12 0.59 0.13 0.15 0.01 1.21 0.12 0.11 0.07 0.08 0.12 0.12 0.15 0.12 0.16 0.14 0.05 0.09

21.7 20.5 28.7 13.0 9.7 20.3 9.0 26.8 9.6 9.3 6.3 10.7 16.0 11.8 11.9 10.7 17.1 10.8 30.8 12.6 10.9 6.9 9.7 16.1 11.7 10.2 11.9 13.8 16.1 13.1 9.4

Ac

ce

RSD (counts)

cr

LLoQ (µM)

us

Regression equation

an

R

M

Analytes

ip t

Table 2 Calibration curves and figures of merit of the baseline calibration

LLoQ = lower limit of quantification (µM) , RSD = residual standard deviation (counts), SD = standard deviation for the procedure (µM), CV = coefficient of variation (%)

Page 20 of 21

Table 3 Accuracy and precision for three concentration levels

Prec (%)

ACC (%) Prec (%)

ACC (%) Prec (%)

ACC (%) Prec (%)

ACC (%) Prec (%)

ip t

GCDCA C (µM) (%) 0,20 20,00 0,60 4,70 3,00 2,70 0,20 18,20 0,60 12,80 3,00 2,50

CA C (µM) (%) 0,20 19,20 0,60 17,10 3,00 3,30 0,20 3,70 0,60 3,90 3,00 0,40

Desmosterol C (µM) (%) 3,00 15,20 6,00 20,80 10,00 13,20 3,00 12,30 6,00 7,10 10,00 9,90

C 1 (LLOQ) C2 C3 C 1 (LLOQ) C2 C3

GCA C (µM) (%) 0,10 19,70 0,60 6,90 3,00 3,50 0,10 13,70 0,60 6,00 3,00 3,50

GDCA C (µM) (%) 0,20 19,40 0,60 10,50 3,00 6,60 0,20 16,80 0,60 6,10 3,00 5,90

GLCA C (µM) (%) 0,02 4,70 0,30 9,70 3,00 10,40 0,02 16,30 0,30 9,70 3,00 10,50

GUDCA C (µM) (%) 0,10 21,40 0,60 6,40 3,00 7,00 0,10 18,10 0,60 3,60 3,00 6,20

HDCA C (µM) (%) 0,90 12,50 9,00 6,80 15,00 4,20 0,90 20,00 3,00 6,10 15,00 4,20

C 1 (LLOQ) C2 C3 C 1 (LLOQ) C2 C3

Pregnanedione C (µM) (%) 0,20 18,40 0,60 11,60 3,00 3,50 0,20 11,90 0,60 13,00 3,00 2,80

Pregnanolone C (µM) (%) 0,03 17,20 0,60 8,60 3,00 8,00 0,03 18,90 0,60 11,50 3,00 8,50

Progesterone C (µM) (%) 0,20 11,90 0,60 7,80 3,00 5,80 0,20 11,50 0,60 13,40 3,00 5,60

Stigmasterol C (µM) (%) 2,00 16,10 6,00 6,70 10,00 7,20 2,00 55,60 6,00 15,90 10,00 15,50

TCDCA C (µM) (%) 0,10 16,10 0,60 6,70 3,00 7,20 0,10 16,70 0,60 8,00 3,00 8,10

C 1 (LLOQ) C2 C3 C 1 (LLOQ) C2 C3

TCA C (µM) (%) 0,10 19,00 0,60 7,30 3,00 5,90 0,10 17,70 0,60 6,30 3,00 5,80

TDCA C (µM) (%) 0,10 19,90 0,60 8,70 3,00 3,50 0,10 15,20 0,60 7,20 3,00 3,60

THDCA C (µM) (%) 0,03 16,10 0,60 6,40 3,00 3,70 0,03 19,00 0,60 5,30 3,00 3,70

TLCA C (µM) (%) 0,02 18,40 0,30 12,50 3,00 9,10 0,02 17,70 0,30 7,10 3,00 9,30

TUDCA C (µM) (%) 0,10 18,30 0,60 12,30 3,00 2,90 0,10 11,00 0,60 17,30 3,00 2,60

C 1 (LLOQ) C2 C3 C 1 (LLOQ) C2 C3

T-α/β-MCA C (µM) (%) 0,04 9,30 0,60 5,50 6,00 5,10 0,04 7,50 0,60 2,10 6,00 0,80

THDOC C (µM) (%) 0,10 22,00 0,60 19,00 3,00 3,60 0,10 17,10 0,60 17,30 3,00 3,10

UDCA C (µM) (%) 0,20 9,40 1,80 7,90 0,30 7,90 0,20 17,00 1,80 3,70 0,30 4,80

α-MCA C (µM) (%) 0,03 11,10 0,60 8,90 3,00 8,30 0,03 14,20 0,60 6,40 3,00 8,30

β-MCA C (µM) (%) 0,01 17,50 0,20 9,50 1,50 7,00 0,01 13,80 0,20 4,90 1,50 7,20

C 1 (LLOQ) C2 C3 C 1 (LLOQ) C2 C3

ω-MCA C (µM) (%) 0,10 18,70 C = concentration, ACC = accuracy, Prec = precision 0,30 6,30 3,00 5,10 0,10 17,90 0,30 5,30 3,00 5,20

an

us

cr

C 1 (LLOQ) C2 C3 C 1 (LLOQ) C2 C3

(%) 6,00 16,20 12,40 17,00 10,40 11,40

M

ACC (%)

DOC C (µM) (%) 0,20 11,00 0,60 4,80 3,00 5,00 0,20 14,30 0,60 4,40 3,00 5,00

Chol C (µM) 0,60 2,00 10,00 0,60 2,00 10,00

ed

Prec (%)

CDCA/DCA C (µM) (%) 0,20 19,80 0,60 3,70 3,00 4,40 0,20 14,30 0,60 1,10 3,00 5,10

pt

ACC (%)

7-OH-Chol C (µM) (%) 0,10 16,90 1,00 8,50 5,00 5,10 0,10 15,60 1,00 13,30 5,00 5,00

ce

Prec (%)

C 1 (LLOQ) C2 C3 C 1 (LLOQ) C2 C3

27-OH-Chol C (µM) (%) 0,20 19,00 1,00 19,20 2,00 23,30 0,20 39,80 1,00 23,00 2,00 28,80

Ac

ACC (%)

22-S-OH-Chol 24-S-/25-OH-Chol C (µM) (%) C (µM) (%) 0,30 10,10 0,60 18,00 3,00 12,00 3,80 12,20 5,00 13,50 6,30 11,00 0,30 29,10 0,60 25,20 3,00 16,20 3,80 11,90 5,00 14,10 6,30 11,10

Page 21 of 21

A liquid chromatography-tandem mass spectrometry-based method for the simultaneous determination of hydroxy sterols and bile acids.

Recently, hydroxy sterols and bile acids have gained growing interest as they are important regulators of energy homoeostasis and inflammation. The hi...
884KB Sizes 0 Downloads 9 Views