Identification of Pathways, Gene Networks, and Paralogous Gene

Jul 16, 2012 - Identification of Pathways, Gene Networks, and Paralogous Gene Families in Daphnia pulex .... Identification of Metabolic Pathways in D...
0 downloads 0 Views 714KB Size
Subscriber access provided by UNIV OF CALGARY

Article

Identification of pathways, gene networks and paralogous gene families in Daphnia pulex responding to exposure to the toxic cyanobacterium Microcystis aeruginosa. Jana Asselman, Dieter IM De Coninck, Stephen Glaholt, John Colbourne, Colin Janssen, Joseph Shaw, and Karel De Schamphelaere Environ. Sci. Technol., Just Accepted Manuscript • DOI: 10.1021/es301100j • Publication Date (Web): 16 Jul 2012 Downloaded from http://pubs.acs.org on July 22, 2012

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

Environmental Science & Technology 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 25

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

Environmental Science & Technology

254x142mm (96 x 96 DPI)

ACS Paragon Plus Environment

Environmental Science & Technology

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

1

Identification of pathways, gene networks and paralogous gene families in

2

Daphnia pulex responding to exposure to the toxic cyanobacterium

3

Microcystis aeruginosa.

4 5

Jana Asselman1,*, Dieter IM De Coninck1, Stephen Glaholt2, John K Colbourne3, Colin R Janssen1, Joseph R

6

Shaw2,3, Karel AC De Schamphelaere1

7

1

Laboratory of Environmental Toxicology and Aquatic Ecology, Ghent University, Belgium

8

2

The School of Public and Environmental Affairs, Indiana University, Bloomington, Indiana USA

9

3

The Center for Genomics and Bioinformatics, Indiana University, Bloomington, Indiana USA

10

*

Corresponding author

11

Email: [email protected]

12

Postal Address:

13

Laboratory of Environmental Toxicology and Aquatic Ecology

14

J. Plateaustraat 22

15

B-9000 Gent

16

Belgium

17

Tel: +32 9 264 37 10

18

Fax: +32 9 264 37 66

19 20

21

ACS Paragon Plus Environment

Page 2 of 25

Page 3 of 25

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

Environmental Science & Technology

22

Abstract

23

Although cyanobacteria produce a wide range of natural toxins that impact aquatic organisms, food webs and

24

water quality, the mechanisms of toxicity are still insufficiently understood. Here, we implemented a whole-

25

genome expression microarray to identify pathways, gene networks and paralogous gene families responsive to

26

Microcystis stress in Daphnia pulex. Therefore, neonates of a sensitive isolate were given a diet contaminated

27

with Microcystis to contrast with those given a control diet for sixteen days. The microarray revealed 2247

28

differentially expressed (DE) genes (7.6% of the array) in response to Microcystis, of which 17% are lineage

29

specific( i.e., these genes have no detectable homology to any other gene in currently available databases) and

30

49% are gene duplicates (paralogs). We identified four pathways/gene networks and eight paralogous gene

31

families affected by Microcystis. Differential regulation of the ribosome, including 3 paralogous gene families

32

encoding 40S, 60S and mitochondrial ribosomal proteins, suggests an impact of Microcystis on protein synthesis

33

of D. pulex. In addition, differential regulation of the oxidative phosphorylation pathway (including the NADH

34

ubquinone oxidoreductase gene family) and the trypsin paralogous gene family (a major component of the

35

digestive system in D. pulex) could explain why fitness is reduced based on energy budget considerations.

36 37 38

ACS Paragon Plus Environment

Environmental Science & Technology

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

Introduction

40

Cyanobacterial blooms in water reservoirs have important ecological, economical and human health

41

consequences (1). These blue-green algal blooms have been studied since the end of the 19th century (1), yet

42

gained prevalence during recent decades because of their links to climate change and anthropogenic

43

eutrophication of water reservoirs (2). They produce a wide range of natural toxins (3-7) that have the potential

44

to impact aquatic ecosystems, human health and ecosystem services, which now fuels an increased public and

45

scientific awareness (8-11).

46

The most common and best studied cyanobacterium is Microcystis aeruginosa (12). This cyanobacterium is

47

primarily known for its production of microcystins (a group of toxic cyclic heptapeptides) and aeruginosins - a

48

group of toxins with a peptide-like structure without any standard L-amino acids (13-14). Microcystins are

49

inhibitors of eukaryotic protein serine/threonine phosphatases (13), while aeruginosins inhibit serine proteases at

50

the protein level (14-15).

51

Our goal was to investigate the effects of cyanobacterial stress from M. aeruginosa on the transcriptome of the

52

freshwater microcrustacean Daphnia pulex. Daphnia is a keystone species in aquatic food webs and a prominent

53

model organism for the study of ecotoxicology, evolutionary biology and ecology (16). The recently described

54

draft genome sequence has also made available a large suite of tools to investigate gene functions (17).

55

The effects of Microcystis stress on Daphnia at physiological and life-history level have been studied since the

56

1980s (18-20) and have primarily been related to three different factors: lack of essential nutrients such as

57

essential fatty acids or lipids (18, 21, 22), deter feeding (19, 21), or toxin production (19, 21, 23). Current

58

literature (21, 23) remains undecided whether the effect of Microcystis on Daphnia can be contributed to only

59

one of these factors or a combination of them.

60

We described the transcriptional stress response of D. pulex feeding on M. aeruginosa by using a comprehensive

61

transcriptome microarray. We aimed to identify pathways or gene networks that characterize the response of

62

Microcystis stress. We chose to study the effects of exposure to live cells of M. aeruginosa and not the effects of

63

individual purified cyanobacterial toxins because the effects of purified toxins are known to be different than

64

those of live cyanobacteria cells (21, 24-28). Moreover, studying the responses to feeding on Microcystis

65

provides a more realistic understanding of its impact in the environment.

66

We therefore fed Daphnia pulex neonates with a diet ratio containing a mixture of M. aeruginosa and a normal

67

green algae. The former diet ratio has been shown to invoke a 50% decline in reproductive output in the D. pulex

68

isolate used here (11). Therefore, measured gene expression responses to the toxic diet mixture are tied to effects

ACS Paragon Plus Environment

Page 4 of 25

Page 5 of 25

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

Environmental Science & Technology

69

on the animal’s fitness. An additional aim of this study was to demonstrate how researchers can continue to

70

ecologically annotate the Daphnia pulex genome, which is characterized by a very high number of lineage

71

specific genes (i.e they have no detectable sequence homology to genes in any of the current genome databases

72

(17)), based on the knowledge of the response of such genes to specific environmental conditions (here

73

Microcystis exposure).

74 75

Material and Methods

76

Exposure Experiments

77

Animals

78

Dahpnia pulex were obtained from isoclonal laboratory cultures of an isolate, originating from the BassHaunt

79

Lake, Dorset region, Ontario, Canada. D. pulex were housed in 3L borosilicate beakers (20 per beaker), held at a

80

constant temperature (20 ± 1°C) and photoperiod (16:8 light-dark). Animals were cultured in no N, no P

81

COMBO medium (29) which was renewed weekly. They were fed daily Ankistrodesmus falcatus at a rate of 1.5

82

mg C L-1. For all experiments, neonates (< 24 h old) were isolated from unexposed maintenance cultures. These

83

cultures were synchronized with respect to time to maturity of the adults producing neonates for experiments.

84

Culturing of cyanobacteria

85

The cyanobacterial strain used was a microcystin producing Microcystis aeruginosa strain (UTEX LB2385).

86

Microcystis was cultured in modified COMBO medium (29) under standard and sterile conditions in 6L

87

culturing flasks. Cultures were incubated at 20±1°C under constant light conditions with gentle aeration and

88

allowed to grow until mid-late log phase. Cyanobacteria were concentrated by centrifugation and cleaned by

89

resuspension and centrifugation using reconstituted water (modified COMBO medium) three times before use.

90

Density of the cultures was determined with a counting chamber. Toxicity of the strain was verified by HPLC

91

analysis, in which subsamples of the concentrated cyanobacteria suspension were analyzed for

92

cylindrospermopsin, anatoxin-a and 4 different types of microcystins, i.e. microcystin-RR, YR, LR, LA, LF and

93

LW, which differing in two variable amino acids residues (30) (Environmental Analysis Laboratory, Lake

94

Superior State University, Sault Ste. Marie, MI, USA). An administered volume of this M. aeruginosa

95

suspension per day, i.e. 50% of the diet based on dry weight, resulted in an exposure to 0.069 µg microcystin-YR

96

and 0.050 µg microcystin-LR per day.

ACS Paragon Plus Environment

Environmental Science & Technology

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

97

Exposure experiment

98

Animals were exposed in 1L polyethylene beakers (18 neonates per beaker) for a period of sixteen days under a

99

constant photoperiod (16:8 light dark) and constant temperature of 20 ± 1°C. Both control and exposed

100

treatments consisted of four biological replicates, i.e. 4 beakers. All animals were fed with a diet in which the

101

final feeding concentration was 1.5 mg C L-1 on dry weight basis. The diet of the exposed animals contained

102

50% of Microcystis aeruginosa and 50% of Ankistrodesmus falcatus, control diet consisted of 100% A. falcatus.

103

This ratio was based on De Schamphelaere et al. (11), where it resulted in a decline of 50% in reproduction in

104

exposed animals. This diet ratio resulted in an administered volume of 2.67 mL of this M. aeruginosa suspension

105

per day. During the experiment, pH of the media was monitored on regular intervals. The experiment was

106

repeated twice. First, animals were isolated for gene expression analysis. Second, animals were isolated for

107

microcystin measurements.

108

Analysis of microcystin levels in exposed animals

109

Wet weight of the animals was determined on a microbalance at the end of the exposure. Afterwards animals

110

were frozen and stored at -80°C until microcystin analysis. The sample preparation was conducted as follows: all

111

animals were macerated with a pipet tip and then subjected to 5 freeze-thaw cycles (freezing at min 80°C, then

112

thawing at room temperature). Next, 250 µL of 70% methanol (analytical grade, Sigma Aldrich, MO, USA) was

113

added and samples were left to stand for 24 hours at room temperature. This was followed by a sonication of the

114

samples. Finally, samples were filtered through a 0.2 µm syringe filter and filtrate was stored at -20°C until

115

ELISA analysis. Microcystins levels were analyzed using the ELISA technique provided in the QuantiPlateTM

116

Microcystin Kit (Envirologix, Maine, USA) following manufacturer’s protocol.

117

mRNA Extraction, Labeling and Hybridization

118

RNA was extracted with the RNeasy kit and Qiashredder (Qiagen, Venlo, Netherlands) following

119

manufacturer’s protocol. All animals (18 in total) from one beaker were pooled into one sample and will further

120

be referred to as one biological replicate. DNA contamination was removed by a DNAse treatment (Qiagen,

121

Venlo, Netherlands). RNA quantity and quality were determined with the spectrophotometer (Nanodrop, Thermo

122

Fisher Scientific, Wilmington, DE, USA) and with the Bioanalyzer 2100 (Agilent Technologies, Santa Clara,

123

CA, USA) respectively. Samples were stored at -80°C until RNA amplification.

124

The microarray protocol follows detailed instructions by Lopez and Colbourne, 2011 (31). Samples were

125

amplified using a T7-based RNA amplification technique. One microgram of total RNA was amplified with the

ACS Paragon Plus Environment

Page 6 of 25

Page 7 of 25

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

Environmental Science & Technology

126

MessageAmp II aRNA Amplification kit (Ambion, Applied Biosystems, Carlsbad, CA, USA) following

127

manufacturer’s protocol. Quantity and quality of the amplified RNA were determined with the

128

spectrophotometer and Bioanalyzer 2100.

129

Double stranded cDNA was synthetized with SuperScript Double-Stranded cDNA Synthesis Kit (Invitrogen,

130

Carlsbad, CA, USA) following clean up (alkaline hydrolysis and Qiaquick columns, Qiagen). Concentration and

131

integrity of the cDNA were determined with Nanodrop spectrophotometer and Bioanalyzer 2100.

132

Samples were labeled with Dual-Color DNA Labeling Kit (Roche Nimblegen, Madison, WI, USA) following

133

manufacturers protocol. Quantity and quality of the samples were again determined with the spectrophotometer

134

and Bioanalyzer 2100.

135

The microarray design (Table S1) consisted of four arrays, each containing two samples, i.e. a control and a

136

Micrcosytis exposed sample. Different biological replicates were used for each array and dye swaps were

137

conducted. All 8 labeled samples were pooled according to the design (i.e. one control biological replicate was

138

pooled with one Microcystis exposed replicate), resulting in 4 pools to be hybridized to 4 arrays. Each pool was

139

dried and resuspended in hybridization buffer according to Roche NimbleGen’s User Guide for Expression

140

Analysis for Cy-labeled cDNA derived from Eukaryote systems. Subsequent hybridization of each of these

141

pools on the respective arrays followed the same protocol (31) and was executed with the NimbleGen

142

Hybridization Kit (Roche Nimblegen, Madison, WI, USA). After hybridization the slides were washed with

143

NimbleGen Hybridization Wash Buffers (Roche Nimblegen, Madison, WI, USA). The microarray itself is a

144

transcriptome array developed by the Centre for Genomics and Bioinformatics (Indiana University,

145

Bloomington, IN, USA) and is in the National Center for Biotechnology Information (NCBI) Gene Expression

146

Omnibus under the accession number (GEO: GPL11278). Finally, arrays were scanned with the NimbleGen MS

147

200 Microarray Scanner to measure fluorescence and images were processed with NimbleScan 2.6 Software

148

(GEO: GSE36635).

149

Image Analysis and Data Processing

150

Microarray images were analyzed with the statistical software package R (32) and bioconductor (33). We used

151

the LIMMA (34) package with additions and modifications according to Colbourne et al. (17). All signal

152

distributions were quantile normalized across arrays, samples and replicates. Differential expression of a gene

153

was determined based on the mean M-value of probes that represent the gene in question. The M-value for a

154

gene was defined as the log2 ratio of the expression in the exposed animals and the expression of the animals in

155

the control treatment. Linear models were constructed with lmFit and empirical Bayes Statistics were

ACS Paragon Plus Environment

Environmental Science & Technology

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

156

implemented with eBayes function. Benjamin-Hochberg method (35) was implemented to adjust p-values for

157

multiple testing.

158

Analysis of Gene lists

159

The analysis of the gene lists were combined with annotation information on each gene available through

160

wFleabase.org (36), KEGG database (37) and KOG (clusters of eukaryotic orthologous groups) database (38) in

161

R. Annotation information from wFleabase.org including KOG annotation, and enzyme classification numbers,

162

was downloaded in batch and combined with gene expression lists in R. Annotation information from the KEGG

163

database was obtained with KAAS (39), for which all protein sequences of the draft genome sequence were

164

uploaded to the KAAS server. All results were stored in a txt file for further use in R (Files S1-S3). Hence, the

165

gene lists were analyzed in three different steps: KOG grouping analysis, pathway analysis and analysis of

166

paralogous gene families. To assess the impact of duplicated genes, both KOG and KEGG analysis were

167

executed once with and once without duplicated genes (i.e. only single copy genes were considered in the latter

168

analysis). Duplicated genes were excluded based on their grouping into a paralog family as defined on

169

wFleabase.org,

170

predictions/dpulex1_gnomon_paralog_mcl2ids.tab). KOG analysis was executed based on KOG classification as

171

defined by the Joint Genome Institute (http://genome.jgi-psf.org/cgi-bin/kogBrowser?db=Dappu1) where p-

172

value was calculated with a Fisher exact Test (40) and correct for multiple testing with the Benjamin-Hochberg

173

method (35). Pathway analysis with KEGG reference pathway maps revealed differential expression of

174

pathways, where p-value was calculated with a Fisher Exact Test (40) and corrected with Benjamin-Hochberg

175

method (35) for multiple testing. KEGGSOAP package (37) was used in R to query KEGG databases for full

176

pathway annotation. Pathway analysis was executed with both annotated enzyme number classification and

177

Kegg Orthology classification as input identifiers. A global metabolic pathway map was created within KEGG

178

through KEGGSOAP from R. In addition, gene lists were analyzed with Ingenuity Pathway Analysis

179

(Ingenuity® Systems, www.ingenuity.com). The input identifier for Ingenuity Pathway Analysis was the

180

UniProt accession number for each gene. Genes from the dataset that had an absolute M-value larger than 1 and

181

a q-value