Metabonomics profiling reveals biochemical pathways associated with

1 day ago - Why European Authors Should Submit to Environmental Science & Technology and Environmental Science & Technology Letters. Don't let the ...
0 downloads 0 Views 1MB Size
Subscriber access provided by Kaohsiung Medical University

Article

Metabonomics profiling reveals biochemical pathways associated with pulmonary arterial hypertension in broiler chickens Feng-Jin Shao, Yi-Tian Ying, Xun Tan, Qiao-Yan Zhang, and Wen-Ting Liao J. Proteome Res., Just Accepted Manuscript • DOI: 10.1021/acs.jproteome.8b00316 • Publication Date (Web): 04 Sep 2018 Downloaded from http://pubs.acs.org on September 4, 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 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

Metabonomics profiling reveals biochemical pathways associated with pulmonary arterial hypertension in broiler chickens Feng-Jin Shao†, Yi-Tian Ying†, Xun Tan†*, Qiao-Yan Zhang†, Wen-Ting Liao† †

Department of Veterinary Medicine, Zhejiang University, Hangzhou 310058, P.R. China

* Corresponding Author: Dr. Xun Tan, Department of Veterinary Medicine, Zhejiang University,

Hangzhou

310058,

China.

Phone:

+86-571-88982647. E-mail: [email protected].

ORCID ID: 0000-0002-0897-9052

1

ACS Paragon Plus Environment

+86-571-88982393.

Fax:

Journal of Proteome Research 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ABSTRACT: Pulmonary arterial hypertension (PAH) is the major cause of death in fast growing meat-type chickens (broiler chickens). At present, the underlying mechanisms that give rise to PAH are not fully understood. In order to identify the metabonomics profiles characterizing the process, we conducted a comprehensive gas chromatography-mass spectrometry (GC-MS)-based metabolic profiling of lung tissues from PAH broilers and age-matched controls. PAH was induced by excess salt in drinking water. Medial hypertrophy of pulmonary arteries was present in PAH birds as compared with controls. The metabonomics profiles of lung tissues well distinguished PAH broilers from control subjects. Significant changes in the levels of 41 metabolites were detected in PAH vs. normal birds. Aside from the metabolic alterations indicating a status of oxidative stress and inflammation, evidence of reduced cellular uptake of arginine due to increased lysine biosynthesis and of a shift of arginine metabolism to arginase pathway were observed. In addition, PAH birds showed increased biosynthesis of fatty acids, which may be associated with excessive proliferation of vascular cells during pulmonary vascular remodeling. Furthermore, we observed significant changes in pentose phosphate pathway (PPP) and increased aminomalonic acid in PAH broilers. These results provide additional biochemical insights into the pathogenesis of the PAH. Our data may lead to the development of new strategies to control PAH in broilers.

KEYWORDS: pulmonary arterial hypertension; broiler chicken; lung; metabonomics; oxidative stress; inflammation; nitric oxide; fatty acid; pentose phosphate pathway 2

ACS Paragon Plus Environment

Page 2 of 36

Page 3 of 36 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

INTRODUCTION Fast growing meat-type chickens (broiler chickens) are susceptible to pulmonary arterial hypertension (PAH, also known as ascites syndrome and pulmonary hypertension syndrome), a disease characterized by increased pulmonary arterial pressure leading to right ventricle failure and death. An international survey in commercial broiler flocks showed that PAH affected nearly 5% of broilers 1, making it one of the major causes of death in the broiler industry. A considerable body of evidence suggests that the development of PAH results from increased pulmonary vascular resistance

2-5

. Cumulative evidence attributes the

increased pulmonary vascular resistance to inadequacy of pulmonary vascular capacity to accommodate ever-increasing cardiac output required to meet oxygen demands for rapid growth vasodilators

6, 7

, and to imbalance between vasoconstrictors and

8-15

. Structural remodeling of small pulmonary arteries characterized by

medial hypertrophy and intimal hyperplasia also leads to increased vascular resistance 16-19

. Plexogenic arteriopathy, which might result from dysfunction of endothelial

progenitor cells (EPCs), has been recently implicated in the pathogenesis of PAH

20,

21

. In addition, several lines of evidence suggest that the onset of PAH is associated

with oxidative stress

22-24

and environmental and genetic factors 25. In general, these

studies have provided rich data sets on PAH. However, the exact pathogenic mechanisms underlying the disease still need to be elucidated.

3

ACS Paragon Plus Environment

Journal of Proteome Research 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 4 of 36

Metabonomics is able to provide a comprehensive view of the changes of specific small-molecule metabolites in biological systems in response to pathophysiological stimuli or genetic modification. As the final downstream products of gene transcription, metabolites reflect the actual functional endpoints of biological events associated with a physiological or pathological state of the cell, tissue, organ or organism. Thus, metabonomics is increasingly being used to investigate disease mechanisms and to detect and identify novel drug targets

26, 27

. Previous studies have

identified altered metabolites in the liver and serum of PAH birds when compared to healthy subjects 28-30. In this study, we aimed at characterizing the specific metabolites in the lung that discriminate PAH birds from healthy controls, to gain new insights into the mechanisms underlying PAH and to identify possible pathways with potential therapeutic value. MATERIALS AND METHODS All experimental procedures were carried out in accordance with the Guidelines of Animal Ethic’s Committee of the Zhejiang University (Approval number ZJU2015-445-12). Animal Management and Sampling

Sixty 1-day-old broiler chickens (Ross-308) were purchased from a local hatchery farm. On day 7, the birds were randomly divided into two groups (n = 30) and one group was subjected to drinking water containing 0.5% (w/v) salt (sodium chloride) to accelerate the occurrence of PAH. During the study standard brooding temperatures 4

ACS Paragon Plus Environment

Page 5 of 36 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

were applied to birds with a 24-h constant light schedule. All chickens had free access to water and were fed a corn/soybean meal-based feed formulated to conform or exceed the minimum National Research Council

31

standards for all ingredients

including 21% CP and 3100 kcal/kg ME. From day 7 on, birds were checked daily for signs of disease and those displaying depression, gasping, cyanosis or/and extended abdomens were humanly killed by cervical dislocation. Hearts were dissected, and right ventricle (RV) and total ventricle (TV) were weighed to calculate RV/TV weight ratio as previously described

20

. A bird having an RV/TV ratio ≥0.28

21

was

considered to suffer from sustained PAH. The whole left lung was removed immediately and cut in the transverse plane at the major rib indentations (costal sulci). One interrib division from the middle of each lung was immediately frozen and stored at −196°C in liquid nitrogen for metabonomics analysis. For histological study, the apical regions of all the sampled lungs were fixed in neutral buffered formalin. Once a PAH bird in the salt-treated group was identified, a bird from the control group was killed, the RV and TV were weighed, and the lung tissue was sampled as described above. Morphometric Analyses The paraffin-embedded lung tissues were cut at 4‒5 µm and stained with Verhoeff’s stain to facilitate the identification of internal and external elastic laminae of muscular pulmonary arteries. The total vessel wall area (TA), i.e., the area within the external elastic lamina, the area within the internal elastic lamina, and mean external and inner 5

ACS Paragon Plus Environment

Journal of Proteome Research 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 6 of 36

diameter were measured using Image-Pro Plus software (Version 6.0, Media Cybernetics, Rockville, MD). The vessel wall area (WA), WA/TA as well as the relative medial thickness (RMT, medial thickness/external radius) was calculated. Only vessels with complete elastic laminas were evaluated. The differences in morphological parameters were analyzed by two-tailed student’s t-test using the SPSS software (version 22; IBM Corp., Armonk, NY). The significance level was set at α =0.05. Metabolite Extraction Lung tissue sample (50 mg) was homogenized in a mixture of 1 mL chloroform/methanol/water solvent (v/v/v =2:5:2) in an ice bath. Following centrifugation at 14000 g for 15 min at 4 °C, a total of 800 µL supernatant was collected. For an additional extraction, 600 µL of ice-cold methanol was added to the pellet followed by centrifugation at 14000 g, 4 °C for 15 min. A 100 µL of combined supernatants and 10 µL of internal standards (0.05 mg/mL of

13

C6-L-leucine and

13

C6-15N-L-isoleucine) were mixed, dried under a stream of dry nitrogen gas, and

dissolved in 30 µL of 20 mg/mL methoxyamine hydrochloride in pyridine. The resulting mixture was incubated at 37 °C for 90 min. A 30 µL BSTFA (N,O-Bis(trimethylsilyl)trifluoroacetamide) with 1% TMCS (trimethylchlorosilane) (Sigma-Aldrich) was added into the mixture and derivatized at 70 °C for 60 min prior to metabonomics analysis. Quality control (QC) sample pooled from all lung samples

6

ACS Paragon Plus Environment

Page 7 of 36 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

were prepared and analyzed with the same procedure as those of the experiment samples. GC-MS Analysis The derivatized samples were analyzed on an Agilent 7890A gas chromatography system coupled to an Agilent 5975C inert MSD system (Agilent Technologies Inc., CA, USA). A Rxi-5Sil MS fused-silica capillary column (30 m × 0.25 mm × 0.25 µm; Restek corporation, Bellefonte, PA, USA) was used to separate the derivatives. Helium (>99.999%) was used as a carrier gas at a constant flow rate of 1 mL/min through the column. Injection volume was 1 µL, and the solvent delay time was 6 min. The initial oven temperature was held at 70 °C for 2 min, ramped to 160 °C at a rate of 6 °C/min, to 240 °C at a rate of 10 °C/min, to 300 °C at a rate of 20 °C/min, and finally held at 300 °C for 6 min. The temperatures of injector, transfer line, and electron impact ion source were set to 250 °C, 260 °C, and 230 °C, respectively. The impact energy was 70 eV, and data was collected in a full scan mode (m/z 50‒600). Data Processing and Statistical Multivariate Data Analysis Data pre-processing, including the peak picking, alignment and deconvolution, and further processing of raw GC-MS data were performed as previously described

32

.

The resulting data were normalized against total peak abundances and imported to SIMCA software 13.0 package (version 13.0; Umetrics, Umeå, Sweden), where the data were pre-processed by unit variance (UV) scaling and mean centering. Principal component analysis (PCA) and orthogonal partial least squares-discriminant analysis 7

ACS Paragon Plus Environment

Journal of Proteome Research 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 8 of 36

(OPLS-DA) were applied to pre-processed metabolite concentration data to discriminate spectra of PAH group from control group. In order to avoid model over-fitting, a default 7-round cross-validation in SIMCA software was performed throughout to determine the optimal number of principal components. Potential differential metabolites were identified according to the variable importance in the projection (VIP) values obtained from the OPLS-DA model and the p-value from a two-tailed Student’s t-test of the normalized peak area, and those variables with VIP >1.0 and P  5. Hence, nine metabolic pathways were detected as potential metabolic pathways for PAH and control group (Figure 4). Among these pathways, six biological modules were involved in amino acid metabolism, including D-glutamine and D-glutamate metabolism, alanine, aspartate and glutamate metabolism, glycine, serine and threonine metabolism, lysine biosynthesis, arginine and proline metabolism, and glutathione metabolism. The other three biological modules included arachidonic acid metabolism, pentose phosphate pathway (PPP), and biosynthesis of unsaturated fatty acids. The glycine, serine and threonine metabolism and arginine and proline metabolism were significantly altered (p  5 are labeled. –log (p) is from the original P-value calculated from the enrichment analysis. The x-axis represents the pathway impact, and y-axis represents the pathway enrichment. Larger sizes and darker colors represent higher pathway impact values and higher pathway enrichment.

DISCUSSION To the best of our knowledge, this is the first study to identify lung metabolic abnormalities associated with the pathophysiological mechanism of PAH in broilers. Previous study by Espinha et al.

36

provided evidence that gender has no effect on

cardiorespiratory function or metabolic rate in broilers under resting conditions; however, it appears that their metabolic state changes with age36, 37. To minimize the effect of age on metabolic variables, birds in a narrow age range (between day 10 and 14) were selected for investigation in this work. In addition, PAH birds and controls were individually matched on age. GC-MS analysis revealed a wide range of discriminant metabolites between PAH birds and non-PAH controls, which are related to amino acid metabolism, energy metabolism (PPP, biosynthesis of unsaturated fatty acids) and inflammatory response-related metabolism (arachidonic acid metabolism). These results suggest that PAH pathogenesis may be largely mediated at the level of metabolism.

18

ACS Paragon Plus Environment

Page 19 of 36 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

Insufficient production of nitric oxide (NO), a potent vasodilator in the lung circulation, has been recognized as the major contributor to the pathobiology of PAH in broilers

12, 13, 17, 38

. Nevertheless, the underlying mechanisms still remain unclear.

NO is produced by nitric oxide synthase (NOS) from the amino acid arginine. Thus, any factor that affects the bioavailability of arginine for NOS can lead to reduced synthesis of NO. In the present study, we found that PAH broilers had significantly increased lysine in their lungs, which is in line with a previous study on a rat model of monocrotaline (MCT)-induced PAH cellular uptake of arginine

39

. Lysine is known to competitively inhibit

40, 41

. Given that the availability of arginine is one of the

rate-limiting factors in cellular NO production

42

, and that the transport systems for

lysine and arginine in birds are similar to those reported in mammals

43

, our results

allow us to argue that reduced NO production in the lungs of PAH birds is associated with increased lysine. Indeed, there is evidence that lysine perfusion reduces NO production and increases pulmonary vascular resistance in the lungs isolated from a rat model of lung injury 44. Aside from change in lysine that might lead to reduced NO production, shunting of arginine into a metabolic pathway involving arginase was also evident as arginase products ornithine and urea were significant increased in our PAH broilers. Arginase is known to compete with NOS for arginine, and increased arginase activity may lead to deficiency of arginine available for NOS

45

. Supporting our findings in PAH

broilers, increased breakdown of arginine by arginase was also observed in human 19

ACS Paragon Plus Environment

Journal of Proteome Research 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

PAH

Page 20 of 36

46

. Inhibition of arginase has been shown to attenuate many of the

pathophysiological symptoms associated with hypoxic pulmonary hypertension

47, 48

.

In context, diversion of arginine to the arginase pathway should also be taken into account for reduce NO production in PAH broilers. Clearly further studies are warranted to test whether PAH birds have increased lung arginase activity. Previous studies on human patients with severe PAH suggest an abnormality in glucose metabolism characterized by reduced glycolysis in the lung

49

. However, in

PAH broilers, we did not determine a significant alteration in glycolysis. Indeed, metabonomics analysis suggested a clear signature of deregulated PPP, as evidenced by a significant decrease in ribose-5-phosphate. Threitol, an end product of xylose metabolism connected to the PPP through the glucuronate pathway, was also down-regulated in our PAH birds. PPP branches out from glycolysis at the first committed step of glucose metabolism, which is required for the synthesis of ribonucleotides and is a major source of nicotinamide adenine dinucleotide phosphate (NADPH). NADPH is essential for anabolic reactions and redox homeostasis maintenance 50. Thus, decrease in PPP flux may explain the increased oxidative stress in PAH 22, 51. Glutathione (GSH), which is synthesized from its precursor amino acids glycine, cysteine, and glutamic acid, plays a key role in cellular response against oxidative stress. In this work, we did not detect an alteration of GSH in PAH lungs. However, the levels of 2-aminobutyric acid and 2-hydroxybutyric acid, which are generated as 20

ACS Paragon Plus Environment

Page 21 of 36 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

byproducts when cystathionine is cleaved to cysteine in times of high GSH demand under oxidative stress

52

, were markedly elevated. It is suggested that increased

2-aminobutyric acid compensates GSH level for consumption by reactive oxidative species (ROS) and promotes GSH synthesis

52

. In this context, the increases in

2-aminobutyric acid and 2-hydroxybutyric acid likely reflect increased oxidative stress in PAH lungs. We also determined decreased levels of glutamic acid as well as O-phosphoserine, the precursor of cysteine, in PAH birds, which may be a consequence of enhanced demand of GSH biosynthesis. In addition, PAH group demonstrated a substantial increase in aminomalonic acid. The origin of aminomalonic acid is associated with free radical damage to proteins, mainly glycine and cysteine

53

. In mammals, increased aminomalonic acid was

observed in cardiovascular diseases with an oxidative stress component, including human PAH

54

. Taken together, our results are consistent with the concept that

oxidative stress is involved in the development of PAH. In the present study, PAH broilers exhibited significant alterations in lipid metabolic activity in their lungs as evident by increased amounts of fatty acids. This metabolic shift is likely associated with excessive proliferation of vascular cells during pulmonary artery remodeling, since hyperproliferating cells have high rates of de novo fatty acid synthesis critical for membrane formation and energy production 55. It has been shown that the expression and activity of fatty acid synthase, an enzyme important for fatty acid synthesis, are increased in proliferating pulmonary smooth 21

ACS Paragon Plus Environment

Journal of Proteome Research 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 22 of 36

muscle cells and endothelial cells, and in the lungs of pulmonary hypertensive rats; accordingly, inhibition of fatty acid synthesis reverses pulmonary vascular remodeling and endothelial dysfunction associated with PAH

56, 57

. Although there is a lack of

direct evidence that fatty acids are involved in pulmonary vascular remodeling in PAH broilers, enhanced PAH mortality in birds fed diets containing higher levels of either saturated or unsaturated fats has been previously observed

58

. Further studies

are required to determine whether the fatty acid synthase plays a role in broiler PAH. Growing evidence suggests that inflammation plays a significant role in broiler PAH

59

. In the present study, increased level of arachidonic acid was found in the

lungs of PAH broilers when compared with healthy subjects. Arachidonic acid pathway is a central regulator of inflammatory response. Arachidonic acid is synthesized from α-linolenic acid derived from linoleic acid, then is converted by lipooxygenase into leukotrienes (LTs) or by cyclooxygenases into prostaglandin H2 (PGH2), which is further processed by a series of specific isomerase and synthase enzymes to produce various eicosanoids including prostaglandin D2, prostaglandin J2, and prostaglandin E2. The formation of leukotrienes and prostaglandins from AA constitutes an inflammatory microenvironment that attracts and stimulates leukocytes 60

. To this end, arachidonic acid might be considered as a biomarker of inflammation

and a therapeutic target of PAH. CONCLUSIONS

22

ACS Paragon Plus Environment

Page 23 of 36 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

In summary, this study investigated the metabolic signatures of the PAH broiler lung using an untargeted metabonomics platform. Results from this global metabolic profiling study revealed number of metabolic alterations that might account for impaired NO production, pulmonary vascular remodeling, oxidative stress and inflammation in PAH birds. We propose, for the first time, the occurrence of PAH in broilers is associated with deregulated PPP. The results of this study may lead to the development of new strategies to treat PAH in broilers.

AUTHOR INFORMATION

Corresponding Author: Dr. Xun Tan, Department of Veterinary Medicine, Zhejiang University, 866 Yuhantang Road, Hangzhou 310058, China; Phone: +86-571-88982393; Fax: +86-571-88982647; E-mail: [email protected]. ORCID ID: 0000-0002-0897-9052 Author Contributions The manuscript was written through contributions of all authors. Feng-Jin Shao reared the chickens, collected samples and did lab analyses. Yi-Tian Ying contributed to data statistical analysis and manuscript writing. Xun Tan conceived the idea, supervised the experiment, lab analyses and data statistical analysis and wrote the manuscript. Qiao-Yan Zhang contributed to chicken management and sample

23

ACS Paragon Plus Environment

Journal of Proteome Research 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

collection. Wen-Ting Liao contributed to sample collection. All authors have given approval to the final version of the manuscript. CONFLICT OF INTEREST DISCLOSURE The authors declare no competing financial interest. ACKNOWLEDGMENT This work was supported by Zhejiang Provincial Natural Science Foundation of China (project no. LR12C18001). We would also like to thank Shanghai ProfLeader Biotech Co, Ltd for assistance with the GC-MS analysis. SUPPORTING INFORMATION Table S1. The detailed results of potential metabolic pathways for PAH group vs. control group. Figures S1‒9: KEGG metabolic pathways relevant to PAH. Figure S1. Glutathione metabolism pathway. Figure S2. Glycine, serine and threonine metabolism pathway. Figure S3. Biosynthesis of unsaturated fatty acids. Figure S4. Arginine and proline metabolism. Figure S5. D-Glutamine and D-glutamate metabolism. Figure S6. Alanine, aspartate and glutamate metabolism. Figure S7. Arachidonic acid metabolism pathway. Figure S8. Lysine biosynthesis pathway. Figure S9. Pentose phosphate pathway ABBREVIATIONS

24

ACS Paragon Plus Environment

Page 24 of 36

Page 25 of 36 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

PAH: pulmonary arterial hypertension; RV/TV: right/total ventricle; TA: vessel wall area; WA: wall area; RMT: relative medial thickness; GC-MS: gas chromatography–mass spectrometry; TIC: total ion chromatogram; PCA: principal component analysis; OPLS-DA: orthogonal partial least squares-discriminant analysis; PPP: pentose phosphate pathway; NO: nitric oxide; NOS: nitric oxide synthase; GSH: glutathione REFERENCES 1.

Maxwell, M. H.; Robertson, G. W., UK survey of broiler ascites and sudden

death syndromes in 1993. Br Poult Sci 1998, 39, (2), 203-15.

2.

Chapman, M. E.; Wideman, R. J., Pulmonary wedge pressures confirm

pulmonary hypertension in broilers is initiated by an excessive pulmonary arterial (precapillary) resistance. Poult Sci 2001, 80, (4), 468-73.

3.

Lorenzoni, A. G.; Anthony, N. B.; Wideman, R. J., Transpulmonary pressure

gradient verifies pulmonary hypertension is initiated by increased arterial resistance in broilers. Poult Sci 2008, 87, (1), 125-32.

4.

Wideman, R. J.; Eanes, M. L.; Hamal, K. R.; Anthony, N. B., Pulmonary

vascular pressure profiles in broilers selected for susceptibility to pulmonary hypertension syndrome: age and sex comparisons. Poult Sci 2010, 89, (9), 1815-24.

25

ACS Paragon Plus Environment

Journal of Proteome Research 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

5.

Page 26 of 36

Wideman, R. J.; Kirby, Y. K., A pulmonary artery clamp model for inducing

pulmonary hypertension syndrome (ascites) in broilers. Poult Sci 1995, 74, (5), 805-12.

6.

Wideman, R. F.; Chapman, M. E.; Hamal, K. R.; Bowen, O. T.; Lorenzoni, A.

G.; Erf, G. F.; Anthony, N. B., An inadequate pulmonary vascular capacity and susceptibility to pulmonary arterial hypertension in broilers. Poult Sci 2007, 86, (5), 984-98.

7.

Wideman, R. J.; Erf, G. F.; Chapman, M. E.; Wang, W.; Anthony, N. B.;

Xiaofang, L., Intravenous micro-particle injections and pulmonary hypertension in broiler chickens: acute post-injection mortality and ascites susceptibility. Poult Sci 2002, 81, (8), 1203-17.

8.

Alvarez-Medina,

D.

I.;

Hernandez,

A.;

Orozco,

C.,

Endothelial

hyperpolarizing factor increases acetylcholine-induced vasodilatation in pulmonary hypertensive broilers arterial rings. Res Vet Sci 2012, 92, (1), 1-6.

9.

Chapman, M. E.; Wideman, R. J., Evaluation of the serotonin receptor

blocker methiothepin in broilers injected intravenously with lipopolysaccharide and microparticles. Poult Sci 2006, 85, (12), 2222-30.

10.

Hamal, K. R.; Wideman, R. F.; Anthony, N. B.; Erf, G. F., Differential

expression of vasoactive mediators in microparticle-challenged lungs of chickens that

26

ACS Paragon Plus Environment

Page 27 of 36 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

differ in susceptibility to pulmonary arterial hypertension. Am J Physiol Regul Integr Comp Physiol 2010, 298, (1), R235-42.

11.

Li, Y.; Zeng, J. Y.; Tang, Z. X.; Li, Y. G.; Guo, J. Y.; Pan, J. Q.,

5-Hydroxytryptamine levels in the pulmonary arterioles of broilers with induced pulmonary hypertension and its relationship to pulmonary vascular remodelling. Avian Pathol 2013, 42, (4), 335-41.

12.

Tan, X.; Sun, W. D.; Li, J. C.; Pan, J. Q.; Liu, Y. J.; Wang, J. Y.; Wang, X.

L., L-arginine prevents reduced expression of endothelial nitric oxide synthase (NOS) in pulmonary arterioles of broilers exposed to cool temperatures. Vet J 2007, 173, (1), 151-7.

13.

Wideman, R. J.; Kirby, Y. K.; Ismail, M.; Bottje, W. G.; Moore, R. W.;

Vardeman, R. C., Supplemental L-arginine attenuates pulmonary hypertension syndrome (ascites) in broilers. Poult Sci 1995, 74, (2), 323-30.

14.

Yang, Y.; Qiao, J.; Wu, Z.; Chen, Y.; Gao, M.; Ou, D.; Wang, H.,

Endothelin-1 receptor antagonist BQ123 prevents pulmonary artery hypertension induced by low ambient temperature in broilers. Biol Pharm Bull 2005, 28, (12), 2201-5.

15.

Li, J. C.; Pan, J. Q.; Huang, G. Q.; Tan, X.; Sun, W. D.; Liu, Y. J.; Wang, X.

L., Expression of PDGF-beta receptor in broilers with pulmonary hypertension

27

ACS Paragon Plus Environment

Journal of Proteome Research 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

induced

by cold temperature and its association with pulmonary vascular

remodeling. Res Vet Sci 2010, 88, (1), 116-21.

16.

Li, Y.; Zeng, J. Y.; Tang, Z. X.; Li, Y. G.; Guo, J. Y.; Pan, J. Q.,

5-Hydroxytryptamine levels in the pulmonary arterioles of broilers with induced pulmonary hypertension and its relationship to pulmonary vascular remodelling. Avian Pathol 2013, 42, (4), 335-41.

17.

Tan, X.; Pan, J. Q.; Li, J. C.; Liu, Y. J.; Sun, W. D.; Wang, X. L., L-Arginine

inhibiting pulmonary vascular remodelling is associated with promotion of apoptosis in pulmonary arterioles smooth muscle cells in broilers. Res Vet Sci 2005, 79, (3), 203-9.

18.

Tan, X.; Liu, Y. J.; Li, J. C.; Pan, J. Q.; Sun, W. D.; Wang, X. L., Activation

of PKCα and pulmonary vascular remodelling in broilers. Res Vet Sci 2005, 79, (2), 131-7.

19.

Tan, X.; Chai, J.; Bi, S. C.; Li, J. J.; Li, W. W.; Zhou, J. Y., Involvement of

matrix metalloproteinase-2 in medial hypertrophy of pulmonary arterioles in broiler chickens with pulmonary arterial hypertension. Vet J 2012, 193, (2), 420-5.

20.

Tan, X.; Juan, F. G.; Shah, A. Q., Involvement of endothelial progenitor cells

in the formation of plexiform lesions in broiler chickens: possible role of local immune/inflammatory response. J Zhejiang Univ Sci B 2017, 18, (1), 59-69.

28

ACS Paragon Plus Environment

Page 28 of 36

Page 29 of 36 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

21.

Tan, X.; Shao, F. J.; Fan, G. J.; Ying, Y. T., Expression of angiogenic factors

and plexiform lesions in the lungs of broiler and layer chickens: A comparison. Poult Sci 2018, 97, (5), 1526-1535.

22.

Iqbal, M.; Cawthon, D.; Wideman, R. J.; Bottje, W. G., Lung mitochondrial

dysfunction in pulmonary hypertension syndrome. II. Oxidative stress and inability to improve function with repeated additions of adenosine diphosphate. Poult Sci 2001, 80, (5), 656-65.

23.

Iqbal, M.; Cawthon, D.; Beers, K.; Wideman, R. J.; Bottje, W. G.,

Antioxidant enzyme activities and mitochondrial fatty acids in pulmonary hypertension syndrome (PHS) in broilers. Poult Sci 2002, 81, (2), 252-60.

24.

Pan, J. Q.; Tan, X.; Li, J. C.; Sun, W. D.; Wang, X. L., Effects of early feed

restriction and cold temperature on lipid peroxidation, pulmonary vascular remodelling and ascites morbidity in broilers under normal and cold temperature. Br Poult Sci 2005, 46, (3), 374-81.

25.

Wideman, R. F.; Rhoads, D. D.; Erf, G. F.; Anthony, N. B., Pulmonary

arterial hypertension (ascites syndrome) in broilers: a review. Poult Sci 2013, 92, (1), 64-83.

26.

Dai, C.; Arceo, J.; Arnold, J.; Sreekumar, A.; Dovichi, N. J.; Li, J.; Littlepage,

L. E., Metabolomics of oncogene-specific metabolic reprogramming during breast cancer. Cancer Metab 2018, 6, 5. 29

ACS Paragon Plus Environment

Journal of Proteome Research 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

27.

Page 30 of 36

Beatty, A.; Fink, L. S.; Singh, T.; Strigun, A.; Peter, E.; Ferrer, C. M.;

Nicolas, E.; Cai, K. Q.; Moran, T. P.; Reginato, M. J.; Rennefahrt, U.; Peterson, J. R., Metabolite Profiling Reveals the Glutathione Biosynthetic Pathway as a Therapeutic Target in Triple-Negative Breast Cancer. Mol Cancer Ther 2018, 17, (1), 264-275.

28.

Shi, S.; Shen, Y.; Zhao, Z.; Hou, Z.; Yang, Y.; Zhou, H.; Zou, J.; Guo, Y.,

Integrative analysis of transcriptomic and metabolomic profiling of ascites syndrome in broiler chickens induced by low temperature. Mol Biosyst 2014, 10, (11), 2984-93.

29.

Shi, S.; Shen, Y.; Zhang, S.; Zhao, Z.; Hou, Z.; Zhou, H.; Zou, J.; Guo, Y.,

Combinatory evaluation of transcriptome and metabolome profiles of low temperature-induced resistant ascites syndrome in broiler chickens. Sci Rep 2017, 7, (1), 2389.

30. that

Shen, Y.; Shi, S.; Tong, H.; Guo, Y.; Zou, J., Metabolomics analysis reveals bile

acids

and

phospholipids

contribute

to

variable

responses

to

low-temperature-induced ascites syndrome. Mol Biosyst 2014, 10, (6), 1557-67.

31.

National Research Council. Nutrient Requirements of Poultry, 9th ed.;

National Academy Press: Washington, DC, 1994; pp26-32.

32.

Gao, X.; Pujos-Guillot, E.; Sebedio, J. L., Development of a quantitative

metabolomic approach to study clinical human fecal water metabolome based on trimethylsilylation derivatization and GC/MS analysis. Anal Chem 2010, 82, (15), 6447-56. 30

ACS Paragon Plus Environment

Page 31 of 36 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

33.

Xiang, R. P.; Sun, W. D.; Zhang, K. C.; Li, J. C.; Wang, J. Y.; Wang, X. L.,

Sodium chloride-induced acute and chronic pulmonary hypertension syndrome in broiler chickens. Poult Sci 2004, 83, (5), 732-6.

34.

Wideman, R. F.; Fedde, M. R.; Tackett, C. D.; Weigle, G. E.,

Cardio-pulmonary function in preascitic (hypoxemic) or normal broilers inhaling ambient air or 100% oxygen. Poult Sci 2000, 79, (3), 415-25.

35.

Julian, R. J., The effect of increased sodium in the drinking water on right

ventricular hypertrophy, right ventricular failure and ascites in broiler chickens. Avian Pathol 1987, 16, (1), 61-71.

36.

Espinha, L. P.; Souza, F. A.; Capalbo, A. C.; Bícego, K. C.; Macari, M.;

Gargaglioni, L. H., Age and gender influence the cardiorespiratory function and metabolic rate of broiler chicks during normocapnia and hypercapnia. Respir Physiol Neurobiol 2014, 200, 50-6.

37.

Poureslami, R.; Raes, K.; Turchini, G. M.; Huyghebaert, G.; De Smet, S.,

Effect of diet, sex and age on fatty acid metabolism in broiler chickens: n-3 and n-6 PUFA. Br J Nutr 2010, 104, (2), 189-97.

38.

Martinez-Lemus, L. A.; Hester, R. K.; Becker, E. J.; Jeffrey, J. S.; Odom, T.

W., Pulmonary artery endothelium-dependent vasodilation is impaired in a chicken model of pulmonary hypertension. Am J Physiol 1999, 277, (1 Pt 2), R190-7.

31

ACS Paragon Plus Environment

Journal of Proteome Research 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

39.

Page 32 of 36

Rafikova, O.; Meadows, M. L.; Kinchen, J. M.; Mohney, R. P.; Maltepe, E.;

Desai, A. A.; Yuan, J. X.; Garcia, J. G.; Fineman, J. R.; Rafikov, R.; Black, S. M., Metabolic changes precede the development of pulmonary hypertension in the monocrotaline exposed rat lung. PLoS One 2016, 11, (3), e0150480.

40.

Stathopulos, P. B.; Lu, X.; Shen, J.; Scott, J. A.; Hammond, J. R.;

McCormack, D. G.; Arnold, J. M.; Feng, Q., Increased L-arginine uptake and inducible nitric oxide synthase activity in aortas of rats with heart failure. Am J Physiol Heart Circ Physiol 2001, 280, (2), H859-67.

41.

Simmons, W. W.; Closs, E. I.; Cunningham, J. M.; Smith, T. W.; Kelly, R.

A., Cytokines and insulin induce cationic amino acid transporter (CAT) expression in cardiac myocytes. Regulation of L-arginine transport and NO production by CAT-1, CAT-2A, and CAT-2B. J Biol Chem 1996, 271, (20), 11694-702.

42.

Mori, M.; Gotoh, T., Regulation of nitric oxide production by arginine

metabolic enzymes. Biochem Biophys Res Commun 2000, 275, (3), 715-9.

43.

Kirsch, S.; Humphrey, B. D., Functional characterization of the chicken

cationic amino acid transporter-2 isoforms. Comp Biochem Physiol B Biochem Mol Biol 2010, 156, (4), 279-86.

44.

Carter, B. J.; Chicoine, L. G.; Nelin, L. D., L-lysine decreases nitric oxide

production

and

increases

vascular

resistance

in

lungs

isolated

lipopolysaccharide-treated neonatal pigs. Pediatr Res 2004, 55, (6), 979-87. 32

ACS Paragon Plus Environment

from

Page 33 of 36 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

45.

Pernow, J.; Jung, C., Arginase as a potential target in the treatment of

cardiovascular disease: reversal of arginine steal? Cardiovasc Res 2013, 98, (3), 334-43.

46.

Kao, C. C.; Wedes, S. H.; Hsu, J. W.; Bohren, K. M.; Comhair, S. A.; Jahoor,

F.; Erzurum, S. C., Arginine metabolic endotypes in pulmonary arterial hypertension. Pulm Circ 2015, 5, (1), 124-34.

47.

Cowburn, A. S.; Crosby, A.; Macias, D.; Branco, C.; Colaco, R. D.;

Southwood, M.; Toshner, M.; Crotty, A. L.; Morrell, N. W.; Chilvers, E. R.; Johnson, R. S., HIF2alpha-arginase axis is essential for the development of pulmonary hypertension. Proc Natl Acad Sci U S A 2016, 113, (31), 8801-6.

48.

Chu, Y.; XiangLi, X.; Niu, H.; Wang, H.; Jia, P.; Gong, W.; Wu, D.; Qin, W.;

Xing, C., Arginase inhibitor attenuates pulmonary artery hypertension induced by hypoxia. Mol Cell Biochem 2016, 412, (1-2), 91-9.

49.

Zhao, Y.; Peng, J.; Lu, C.; Hsin, M.; Mura, M.; Wu, L.; Chu, L.; Zamel, R.;

Machuca, T.; Waddell, T.; Liu, M.; Keshavjee, S.; Granton, J.; de Perrot, M., Metabolomic heterogeneity of pulmonary arterial hypertension. PLoS One 2014, 9, (2), e88727.

50.

Patra, K. C.; Hay, N., The pentose phosphate pathway and cancer. Trends

Biochem Sci 2014, 39, (8), 347-54.

33

ACS Paragon Plus Environment

Journal of Proteome Research 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

51.

Tan, X.; Hu, S. H.; Wang, X. L., The effect of dietary L-carnitine

supplementation on pulmonary hypertension syndrome mortality in broilers exposed to low temperatures. J Anim Physiol Anim Nutr (Berl) 2008, 92, (2), 203-10.

52.

Irino, Y.; Toh, R.; Nagao, M.; Mori, T.; Honjo, T.; Shinohara, M.; Tsuda, S.;

Nakajima, H.; Satomi-Kobayashi, S.; Shinke, T.; Tanaka, H.; Ishida, T.; Miyata, O.; Hirata, K. I., 2-Aminobutyric acid modulates glutathione homeostasis in the myocardium. Sci Rep 2016, 6, 36749.

53.

Copley, S. D.; Frank, E.; Kirsch, W. M.; Koch, T. H., Detection and possible

origins of aminomalonic acid in protein hydrolysates. Anal Biochem 1992, 201, (1), 152-7.

54.

Bujak, R.; Mateo, J.; Blanco, I.; Izquierdo-Garcia, J. L.; Dudzik, D.;

Markuszewski, M. J.; Peinado, V. I.; Laclaustra, M.; Barbera, J. A.; Barbas, C.; Ruiz-Cabello, J., New biochemical insights into the mechanisms of pulmonary arterial hypertension in humans. PLoS One 2016, 11, (8), e0160505.

55.

Baenke, F.; Peck, B.; Miess, H.; Schulze, A., Hooked on fat: the role of lipid

synthesis in cancer metabolism and tumour development. Dis Model Mech 2013, 6, (6), 1353-63.

56.

Singh, N.; Manhas, A.; Kaur, G.; Jagavelu, K.; Hanif, K., Inhibition of fatty

acid synthase is protective in pulmonary hypertension. Br J Pharmacol 2016, 173, (12), 2030-45. 34

ACS Paragon Plus Environment

Page 34 of 36

Page 35 of 36 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

57.

Singh, N.; Singh, H.; Jagavelu, K.; Wahajuddin, M.; Hanif, K., Fatty acid

synthase modulates proliferation, metabolic functions and angiogenesis in hypoxic pulmonary artery endothelial cells. Eur J Pharmacol 2017, 815, 462-469.

58.

Khajali, F.; Fahimi, S., Influence of dietary fat source and supplementary

alpha-tocopheryl acetate on pulmonary hypertension and lipid peroxidation in broilers. J Anim Physiol Anim Nutr (Berl) 2010, 94, (6), 767-72.

59.

Yang, F.; Cao, H.; Xiao, Q.; Guo, X.; Zhuang, Y.; Zhang, C.; Wang, T.; Lin,

H.; Song, Y.; Hu, G.; Liu, P., Transcriptome analysis and gene identification in the pulmonary artery of broilers with ascites syndrome. PLoS One 2016, 11, (6), e0156045.

60.

Salmon, J. A.; Higgs, G. A., Prostaglandins and leukotrienes as inflammatory

mediators. Br Med Bull 1987, 43, (2), 285-96.

35

ACS Paragon Plus Environment

Journal of Proteome Research 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

For TOC only

36

ACS Paragon Plus Environment

Page 36 of 36