ArticlePDF Available

A combined genotype of three SNPs in the bovine gene is related to growth performance in Chinese cattle

Authors:

Abstract and Figures

PPARD is involved in multiple biological processes, especially for those associated with energy metabolism. PPARD regulates lipid metabolism through up-regulate expression of genes associating with adipogenesis. This makes PPARD a significant candidate gene for production traits of livestock animals. Association studies between PPARD polymorphisms and production traits have been reported in pigs but are limited for other animals, including cattle. Here, we investigated the expression profile and polymorphism of bovine PPARD as well as their association with growth traits in Chinese cattle. Our results showed that the highest expression of PPARD was detected in kidney, following by adipose, which is consistent with its involvement in energy metabolism. Three SNPs of PPARD were detected and used to undergo selection pressure according the result of Hardy–Weinberg equilibrium analysis (P < 0.05). Moreover, all of these SNPs showed moderate diversity (0.25 < PIC < 0.5), indicating their relatively high selection potential. Association analysis suggested that individuals with the GAAGTT combined genotype of three SNPs detected showed optimal values in all of the growth traits analyzed. These results revealed that the GAAGTT combined genotype of three SNPs detected in the bovine PPARD gene was a significant potential genetic marker for marker-assisted selection in Chinese cattle. However, this should be further verified in larger populations before being applied to breeding.
Content may be subject to copyright.
Arch. Anim. Breed., 60, 357–362, 2017
https://doi.org/10.5194/aab-60-357-2017
© Author(s) 2017. This work is distributed under
the Creative Commons Attribution 3.0 License.
Open Access
Archives Animal Breeding
A combined genotype of three SNPs in the bovine PPARD
gene is related to growth performance in Chinese cattle
Jieping Huang1,2, Qiuzhi Zheng1, Shuzhe Wang1, Qiongqiong Zhang1, Lijun Jiang1,2, Ruijie Hao1,
Fen Li1,2, and Yun Ma1,2
1College of Life Sciences, Xinyang Normal University, Xinyang, Henan, China
2Institute for Conservation and Utilization of Agro-Bioresources in Dabie Mountains, Xinyang, Henan, China
Correspondence to: Yun Ma (mayun_666@126.com)
Received: 26 May 2017 – Accepted: 28 August 2017 – Published: 6 October 2017
Abstract. PPARD is involved in multiple biological processes, especially for those associated with energy
metabolism. PPARD regulates lipid metabolism through up-regulate expression of genes associating with adipo-
genesis. This makes PPARD a significant candidate gene for production traits of livestock animals. Association
studies between PPARD polymorphisms and production traits have been reported in pigs but are limited for other
animals, including cattle. Here, we investigated the expression profile and polymorphism of bovine PPARD as
well as their association with growth traits in Chinese cattle. Our results showed that the highest expression
of PPARD was detected in kidney, following by adipose, which is consistent with its involvement in energy
metabolism. Three SNPs of PPARD were detected and used to undergo selection pressure according the result
of Hardy–Weinberg equilibrium analysis (P< 0.05). Moreover, all of these SNPs showed moderate diversity
(0.25 <PIC <0.5), indicating their relatively high selection potential. Association analysis suggested that in-
dividuals with the GAAGTT combined genotype of three SNPs detected showed optimal values in all of the
growth traits analyzed. These results revealed that the GAAGTT combined genotype of three SNPs detected
in the bovine PPARD gene was a significant potential genetic marker for marker-assisted selection in Chinese
cattle. However, this should be further verified in larger populations before being applied to breeding.
1 Introduction
Peroxisome-proliferator-activated receptors (PPARs) are a
group of transcription factors belonging to the nuclear hor-
mone receptor superfamily (Evans et al., 2004). Many stud-
ies have revealed PPARs take part in numerous biological
processes, including lipid metabolism, the insulin signaling
pathway, glucose metabolism, and adipocyte differentiation
(Youssef and Badr, 2013). To date, three subtypes of PPARs
have been discovered: PPARA,PPARD, and PPARG. Among
these, PPARD is widely expressed in various tissues, includ-
ing kidney, liver, heart, intestine, and adipose (Abbott, 2009).
PPARD regulates lipid metabolism through up-regulate ex-
pression of genes involved in the adipogenesis process (Ved-
hachalam et al., 2007). Recently, studies have suggested that
PPARD is essential for adipogenesis as well (Garbacz et al.,
2015; Barroso et al., 2015; Palomer et al., 2016).
Genetic variation in PPARD is proved to be associated
with human diseases. The PPARD rs2016520 polymorphism
was reported to affect repaglinide response in Han Chinese
patients with type 2 diabetes mellitus (Song et al., 2015).
Furthermore, this mutation was shown to be associated with
brain diseases (Huang et al., 2015) and colorectal cancer
(Rosalesreynoso et al., 2017). For its vital role in various
biological processes, PPARD is a potential gene affecting
production traits of livestock animals. Polymorphisms of
PPARD were shown to affect ear size (Ren et al., 2011) and
litter size (Spötter et al., 2010) in pigs. Recently, functional
SNPs in the 50regulatory region of the porcine PPARD gene
have been reported to be significantly associated with fat
deposition traits (Zhang et al., 2015). However, association
studies between PPARD and production traits in other ani-
mals are limited, including cattle.
Published by Copernicus Publications on behalf of the Leibniz Institute for Farm Animal Biology.
358 J. Huang et al.: A combined genotype of three SNPs
Marker-assisted selection (MAS) has been widely used as
a breeding strategy in livestock (Margawati, 2012). To detect
functional SNPs of PPARD for MAS in cattle, we (i) ana-
lyzed the expression profile of PPARD in different tissues,
(ii) detected SNPs in the bovine PPARD gene by direct se-
quencing using 514 Chinese cattle, and (iii) assessed the re-
lationship between detected SNPs and growth traits in partial
cattle.
2 Material and methods
2.1 Samples
Samples used in this study were shown in a previous study
(Huang et al., 2017). Briefly, seven tissues were collected
at the slaughter house for reverse transcription polymerase
chain reaction (RT-PCR), including heart, liver, spleen, lung,
kidney, muscle, and adipose of three Jiaxian cattle (bullock,
30 months). A total of 514 individuals from six Chinese cat-
tle breeds – including 141 Jiaxian cattle, 139 Nanyang cat-
tle, 114 Luxi cattle, 30 Qinchuan cattle, 30 Bohai, cattle
and 60 Gaoyuan yak – were used for SNP genotyping. Birth
weight and six growth traits (body weight, body height, body
length, heart girth, hip width, and average daily gain) of Ji-
axian and Nanyang cattle at 6, 12, 18, and 24 months as well
as nine traits (body height, body length, heart girth, abdom-
inal circumference, hip width, sciatic width, height at hip
cross, body weight, and beef performance index) at around
28–30 months of age in 300 Henan cattle (100 Jiaxian, 100
Nanyang, and 100 Luxi) were recorded for association anal-
ysis.
2.2 Expression analysis of PPARD
In order to understand the potential biological effect of
PPARD on cattle, expression levels of PPARD in seven tis-
sues were investigated by RT-PCR. Details of the method are
shown in a previous study (Huang et al., 2017). Total RNA
was reversely transcribed into cDNA using a PrimeScript-
sRT reagent kit with gDNA Eraser (TaKaRa, Japan). RT-PCR
was performed using SYBR Green I with two-step reactions.
Primers of PPARD (NM_001083636.1) and reference genes
(TUBA1A, NM_001166505.1; β-actin, NM_173979.3) for
RT-PCR are shown in Table S1 (Supplement). The relative
expression level of each tissue was presented as mean ±SD.
2.3 SNP detection and genotyping
In order to investigate polymorphism of PPARD
(AC_000180.1) in Chinese cattle, nine pairs of primers
covering CDS and partial upstream regions were synthesized
(Table S1). The methods for SNP detection and genotyping
were as in a previous study (Huang et al., 2017). Pooled
DNA samples were used as a PCR template for SNP de-
tection. For SNPs detected (Table 1), three pairs of specific
Figure 1. Tissue distribution of bovine PPARD mRNA assessed by
RT-PCR. Values shown in this figure are averages of three indepen-
dent experiments. Error bars represent SD (n=3) of relative mRNA
levels. Expression data were normalized using geometric mean of
mRNA levels for two control genes (TUBA1A and β-actin).
Figure 2. Schematic characteristic of SNPs identified in bovine
PPARD and genotyping. From top to bottom: structure of bovine
PPARD, mutant peaks of sequencing, details of SNPs identified, and
electrophoretogram of genotyping.
primers were designed (Table 2). The traditional PCR-RFLP
method was used for SNP genotyping in 514 Chinese cattle
from six breeds. It should be noted that PCR production of
PPARD-MluI primers contained two recognition sites of
MluI, one of which was native and the other was introduced
from primers for genotyping.
2.4 Statistical analysis
The genetic characteristics of each mutation were investi-
gated after genotyping, including allele frequencies, Hardy–
Weinberg equilibrium (HWE), heterozygosity (He), effective
allele numbers (Ne), and polymorphism information con-
tent (PIC). To evaluate the potential relationship between the
PPARD gene and development of cattle, an association study
was performed based on the genotyping results and growth
traits in Nanyang, Jiaxian, and Luxi cattle. Significant analy-
sis was performed by SPSS 19.0 using general linear model.
Results were presented as means ±SE. Other details can be
found in a previous study (Huang et al., 2017).
Arch. Anim. Breed., 60, 357–362, 2017 www.arch-anim-breed.net/60/357/2017/
J. Huang et al.: A combined genotype of three SNPs 359
Table 1. Details of SNPs detected in the bovine PPARD gene.
Label Position Alleles rs number Functional consequence
SNP1 AC_000180.1: 9268142 G> A rs208371564 upstream variant 2KB
SNP2 AC_000180.1: 9341130 A> G rs470835077 intron 2
SNP3 AC_000180.1: 9352706 T> C rs207513597 intron 6
Table 2. Details of primers and restriction enzymes used for genotyping.
Name Primer sequence (50-30)Tm() Size (bp) Used for Restriction Main
enzyme fragments
PPARD
-Hha I
F:GCAGGATATAGTTCCCAGC
R:GACTTGTCATCCCAACCTT
55 137 SNP1 Hha I GG: 117
bp
GA: 137
bp, 117
bp
AA: 137
bp
PPARD
-Pvu II
F:TCCTTCCAGCAGCTACACAG
CT
R:GGGAGACAACTCGCCCAAG
A
57.5 195 SNP2 Pvu II GG: 147
bp
AG: 167
bp, 147
bp
PPARD
-Mlu I
F:ATGGCAGTGGGACACGCG
R:CCACCAGAAATAACCCCCAT
C
63 121 SNP3 Mlu I TT: 107
bp
TC: 121
bp, 107
bp
CC: 121
bp
Note: letters underlined in the primer sequence are the introduced mutant for genotyping.
3 Results and discussion
3.1 Expression profile of PPARD
The expression profile of PPARD has been widely investi-
gated in rodent and human development, but it is limited in
cattle. In order to understand the potential biological effect
of PPARD on cattle, expression levels of PPARD were in-
vestigated (Fig. 1). Consistent with previous studies, PPARD
was widely expressed in main tissues, suggesting that it was
involved in multiple biological processes. The highest level
of expression was detected in kidney, followed by adipose
tissue (Fig. 1), indicating its significant biological role in
kidney and adipose tissues. Expression levels of the PPARD
gene in the other six tissues were nearly the same, with rel-
atively low values. In fact, the expression pattern of PPARD
was found to be variable in different studies. PPARD was
expressed in kidney with a high level in adult rats (Brais-
sant and Wahli, 1996), adult mice (Girroir et al., 2008), and
adult human (Auboeuf et al., 1997). PPARs were identified as
the genetic sensor responsive to fatty acid ligands (Feige et
al., 2006) and involved in lipid metabolism and the insulin
signaling pathway (Youssef and Badr, 2013). In addition,
chronic kidney disease was attributed to metabolic disorders
mainly through the mechanisms of insulin resistance and re-
sultant hyperinsulinemia (Perlstein et al., 2007). In fat tissue,
only a moderate level was detected in adult rats and humans
(Braissant and Wahli, 1996; Auboeuf et al., 1997). These re-
sults were consistent with our study. However, a moderate to
high level of expression in liver, heart, and lung was detected
in adult rodents and humans (Girroir et al., 2008; Tugwood
et al., 1996; Mukherjee et al., 1997), which was nearly con-
tradictory with our result. Regardless, all of these results un-
derline multiple functions of PPARD in the development of
mammals. Thus, PPARD should be necessary for cattle de-
velopment.
3.2 SNP detection and genetic characteristics of
PPARD in Chinese cattle
In total, three SNPs were detected (Table 1 and Fig. 2),
including AC_000180.1:g.9268142 G > A in the upstream
www.arch-anim-breed.net/60/357/2017/ Arch. Anim. Breed., 60, 357–362, 2017
360 J. Huang et al.: A combined genotype of three SNPs
Table 3. Association analysis between combined genotypes of three SNPs in PPARD and growth traits of 300 adult cattle.
Growth
traits
Combined genotypes
GAAGCC
(10)
GAAGTC
(66)
GAAGTT
(35)
GAGGTC
(45)
GAGGTT
(21)
GGAGTC
(29)
GGAGTT
(14)
GGGGTC
(41)
GGGGTT
(20)
BH
(cm)
129.450 ±
3.028
129.106 ±
1.179
130.843 ±
1.618
130.033 ±
1.427
129.286 ±
2.089
129.448 ±
1.778
127.286 ±
2.559
131.598 ±
1.495
127.450 ±
2.141
BL
(cm)
145.900 ±
4.034
144.068 ±
1.570
147.743 ±
2.156
143.500 ±
1.901
143.619 ±
2.783
143.017 ±
2.369
146.643 ±
3.409
145.415 ±
1.992
141.750 ±
2.852
HG
(cm)
171.400 ±
5.195
174.538 ±
2.022
176.414 ±
2.777
174.156 ±
2.449
168.214 ±
3.585
169.414 ±
3.051
171.357 ±
4.391
175.634 ±
2.566
168.750 ±
3.673
AC
(cm)
200.100 ±
6.774
203.008 ±
2.637ab
205.514 ±
3.621a
204.156 ±
3.193a
192.476 ±
4.674b
199.069 ±
3.978
200.143 ±
5.725
201.415 ±
3.345
190.700 ±
4.790bc
HW
(cm)
45.200 ±
1.897a
45.023 ±
0.738a
45.471 ±
1.014a
45.344 ±
0.894A
43.310 ±
1.309
44.190 ±
1.114
45.286 ±
1.603a
43.341 ±
0.937
41.100 ±
1.341Bb
SW
(cm)
28.500 ±
1.456A
28.659 ±
0.567A
28.586 ±
0.778A
27.489 ±
0.687A
27.548 ±
1.005A
26.914 ±
0.855A
28.107 ±
1.231A
25.549 ±
0.719a
22.600 ±
1.030Bb
HHC
(cm)
129.300 ±
2.691
129.394 ±
1.048
131.357 ±
1.438
129.122 ±
1.269
129.238 ±
1.857
131.621 ±
1.580
128.214 ±
2.274
132.232 ±
1.329
128.575 ±
1.903
BW
(kg)
391.453 ±
32.883
388.720 ±
12.800
413.462 ±
17.577a
399.380 ±
15.501
385.273 ±
2.691
391.784 ±
19.310
378.964 ±
27.791
391.686 ±
16.240
350.251 ±
3.252b
BPI
(kg cm1)
3.017 ±
0.194
3.001 ±
0.076
3.113 ±
0.104a
3.045 ±
0.092
2.906 ±
0.134
2.987 ±
0.114
2.962 ±
0.164
2.956 ±
0.096
2.738 ±
0.137b
BH: body height; BL: body length; HG: heart girth; AC: abdominal circumference; HW: hip width; SW: sciatic width; HHC: height at hip cross; BW: body weight;
BPI: beef performance index.
Lowercase letters mean difference of the value at P<0.05; uppercase letters mean difference of the value at P <0.01.
region (SNP1, rs208371564), AC_000180.1:g.9341130
A > G in intron 2 (SNP2, rs470835077), and
AC_000180.1:g.9352706 T > C in intron 6 (SNP3,
rs207513597). A total of 3801 SNPs of the bovine
PPARD gene can be searched in the SNP database of NCBI
(https://www.ncbi.nlm.nih.gov/snp/), including 710 detected
by cluster and 3091 with no information. No more SNPs
could be found from other studies. Thus, the three SNPs
detected in this study were not further analyzed although
they had been detected by cluster previously.
Then, genetic characteristics of SNPs were investigated
based on the genotyping result (Table S2). We noted that the
AA genotype of SNP2 was absent in all of the populations in
this study. At the same time, the A allele was not rare in Chi-
nese cattle. Therefore, we speculated that individuals with
the AA genotype of SNP2 died during the embryonic stage
or were culled because of disease at an early age. Amazingly,
approximately half of the breeds were not in agreement with
the HWE (P <0.05) at each of these SNP loci, suggesting
that they might undergo selection pressure. All of the three
SNP loci showed moderate diversity (0.25< PIC< 0.5), indi-
cating their relatively high selection potential. Further selec-
tion could be implied if a positive effect were found among
these SNPs in cattle breeds investigated.
3.3 Association study between PPARD and growth traits
Potential genomic mutations of the PPARD gene might be
related to growth traits of cattle. First, relationship between
PPARD and growth traits were investigated in 173 Henan cat-
tle based on a single SNP locus (Table S3). Several signif-
icant differences were identified without regularity. Gener-
ally, the phenotypic value should change along with the vari-
ation in genotype (in the order of wild type, heterozygous
type, and homozygous mutant type) with a specific trend.
Moreover, this trend should be the same among different
breeds and ages. However, significant differences detected in
Table S3 did not conform to such trends and showed disor-
der. This might be due to the low sample size, or else multiple
loci affect the same traits with different weight. Therefore, it
was hard to estimate the real association between these SNPs
and growth traits in cattle.
We speculated that coordination among multiple SNPs
loci might contribute to development or be linked with
Arch. Anim. Breed., 60, 357–362, 2017 www.arch-anim-breed.net/60/357/2017/
J. Huang et al.: A combined genotype of three SNPs 361
growth traits of cattle. Based on such a hypothesis, associ-
ation analysis between combined genotypes of these three
SNPs and growth traits of 300 adult cattle was performed.
Combined genotypes with less than 10 individuals were re-
moved. In total, nine combined genotypes were used for
analysis (Table 3). Interestingly, all traits showed the high-
est values in the GAAGTT combined genotype. Among
these, abdominal circumference, hip width, sciatic width,
body weight, and beef performance index showed signifi-
cant differences. Obviously, individuals with the GAAGTT
combined genotype of these three SNPs showed optimal
growth performance. In fact, association analysis based on
combined genotype has been widely used in studies on the
relationship between genetic variation and diseases in hu-
mans (Kamitani et al., 1995; Boulet et al., 2008; Stelma
et al., 2016) and traits in livestock animals (Garaulet et
al., 2012). However, results from this analytical method
need further verification from multiple points. The bovine
PPARD gene is identified in chromosome 23 (9.27–9.36 Mb).
By searching the quantitative trait locus (QTL) database
of cattle (http://www.animalgenome.org/cgi-bin/QTLdb/BT/
browse) for those QTLs associated with growth trait, four
QTLs were obtained, including a QTL (5.9–16.3 Mb) for
body weight of adult cattle (McClure et al., 2010), a QTL
(0.6–17.5 Mb) for body weight before slaughter (Elo et al.,
1999), a QTL (7.2–21.1 Mb) for body weight at weaning
(McClure et al., 2010), and a QTL (7.2–21.1 Mb) for body
weight at 12 months (McClure et al., 2010). These QTLs fur-
ther suggested that PPARD was a potential significant candi-
date gene for production traits of cattle. Three SNPs iden-
tified in this study were in the non-coding region. In recent
years, transcripts (non-coding RNAs) from non-coding re-
gion have been shown to regulate the transcription of the
origin genes and then affect the biological function of the
origin genes. However, the non-coding region might provide
the binding site for some enzymes relating to transcription.
Thus, mutations in the non-coding region could play a role
in the regulation mechanism. However, SNPs may only be
markers associated with production traits and do not affect
any biological process.
4 Conclusions
The bovine PPARD gene is expressed widely in the main
tissues of adult cattle. Three SNPs of PPARD were iden-
tified in Chinese cattle. The GAAGTT combined genotype
of these three SNPs showed optimal growth performance,
which could be a potential marker for MAS of cattle. How-
ever, further identification should be performed in larger pop-
ulations before being applied to breeding of cattle.
Data availability. The original data are available upon request
from the corresponding author.
The Supplement related to this article is available online
at https://doi.org/10.5194/aab-60-357-2017-supplement.
Author contributions. YM and FL designed experiments and
collected samples. QZ, SW, QZ, LJ and RH carried out the experi-
ment; JH analyzed the data and wrote the manuscript.
Competing interests. The authors declare that they have no con-
flict of interest.
Acknowledgements. This study was supported by the National
Natural Science Foundation of China (no. 31672403), the Chinese
National High Technology Research and Development Program
(no. 2013AA102505-4), the Technology Innovation Teams in
Universities of Henan Province (no. 14IRTSTHN012), and the
Nanhu Scholars Program for Young Scholars of XYNU.
Edited by: Steffen Maak
Reviewed by: three anonymous referees
References
Abbott, B. D.: Review of the expression of peroxisome proliferator-
activated receptors alpha (PPAR alpha), beta (PPAR beta), and
gamma (PPAR gamma) in rodent and human development, Re-
prod. Toxicol., 27, 246–257, 2009.
Auboeuf, D., Rieusset, J., Fajas, L., Vallier, P., Frering, V., Riou,
J.P., Staels, B., Auwerx, J., Laville, M., and Vidal, H.: Tissue
distribution and quantification of the expression of mRNAs of
peroxisome proliferator–activated receptors and liver X receptor-
αin humans: no alteration in adipose tissue of obese and NIDDM
patients, Diabetes, 46, 1319–1327, 1997.
Barroso, E., Rodríguez-Rodríguez, R., Chacón, M.R., Maymó-
Masip, E., Ferrer, L., Salvadó, L., Salmerón, E., Wabistch,
M., Palomer, X., Vendrell, J., Wahli, W., and Vázquez-Carrera,
M.: PPARβ/δameliorates fructose-induced insulin resistance in
adipocytes by preventing Nrf2 activation, Biochim. Biophys.
Acta., 1852, 1049–1058, 2015.
Boulet, S., Kleyman, M., Kim, J. Y., Kamya, P., Sharafi, S., Simic,
N., Bruneau, J., Routy, J. P., Tsoukas, C. M., and Bernard, N. F.:
A combined genotype of KIR3DL1 high expressing alleles and
HLA-B* 57 is associated with a reduced risk of HIV infection,
AIDS, 22, 1487–1491, 2008.
Braissant, O., Foufelle, F., Scotto, C., Dauça, M., and Wahli, W.:
Differential expression of peroxisome proliferator-activated re-
ceptors (PPARs): tissue distribution of PPAR-alpha, -beta, and
-gamma in the adult rat, Endocrinology, 137, 354–366, 1996.
Elo, K. T., Vilkki, J., de Koning, D. J., and Mäki-Tanila, A. V.: A
quantitative trait locus for live weight maps to bovine chromo-
some 23, Mamm. Genome, 10, 831–835, 1999.
Evans, R. M., Barish, G. D., and Wang, Y. X.: PPARs and the com-
plex journey to obesity, Nat. Med., 10, 355–361, 2004.
Feige, J. N., Gelman, L., Michalik, L., Desvergne, B., and Wahli,
W.: From molecular action to physiological outputs: peroxisome
www.arch-anim-breed.net/60/357/2017/ Arch. Anim. Breed., 60, 357–362, 2017
362 J. Huang et al.: A combined genotype of three SNPs
proliferator-activated receptors are nuclear receptors at the cross-
roads of key cellular functions, Prog. Lipid Res., 45, 120–159,
2006.
Garaulet, M., Tardido, A. E., Lee, Y. C., Smith, C. E., Parnell, L.
D., and Ordovás, J. M.: SIRT1 and CLOCK 3111T>C combined
genotype is associated with evening preference and weight loss
resistance in a behavioral therapy treatment for obesity, Int. J.
Obes., 36, 1436–1441, 2012.
Garbacz, W. G., Huang, J. T., Higgins, L. G., Wahli, W., and Palmer,
C. A. N.: PPARαis required for PPARδaction in regulation of
body weight and hepatic steatosis in mice, PPAR Res., 1, 927057,
doi:10.1155/2015/927057, 2015.
Girroir, E. E., Hollingshead, H. E., He, P., Zhu, B., Perdew, G. H.,
and Peters, J. M.: Quantitative expression patterns of peroxisome
proliferator-activated receptor-beta/delta (pparbeta/delta) protein
in mice, Biochem. Biophys. Res. Commun., 371, 456–461, 2008.
Huang, J., Chen, N., Li, X., An, S., Zhao, M., Sun, T., Hao, R., and
Ma, Y.: Two novel SNPs of PPARγsignificantly affect wean-
ing growth traits of Nanyang cattle, Animal Biotechnol., 1–7,
doi:10.1080/10495398.2017.1304950, 2017.
Huang, Y., Nie, S., Zhou, S., Li, K., Sun, J., Zhao, J., Fei, B.,
Wang, Z., Ye, H., Hong, Q., Gao, X., and Duan, S.: PPARD
rs2016520 polymorphism and circulating lipid levels connect
with brain diseases in Han Chinese and suggest sex-dependent
effects, Biomed. Pharmacother., 70, 7–11, 2015.
Kamitani, A., Rakugi, H., Higaki, J., Ohishi, M., Shi, S. J., Takami,
S., Nakata, Y., Higashino, Y., Fujii, K., and Mikami, H.: En-
hanced predictability of myocardial infarction in Japanese by
combined genotype analysis, Hypertension, 25, 950–953, 1995.
Margawati, E.: A global strategy of using molecular genetic infor-
mation to improve genetics in livestock, Reprod. Domest. Anim.,
47, 7–9, 2012.
McClure, M. C., Morsci, N. S., Schnabel, R. D., Kim, J. M., Yao, P.,
Rolf, M. M., McKay, S. D., Gregg, S. J., Chapple, R. H., North-
cutt, S. L., and Taylor, J. F.: A genome scan for quantitative trait
loci influencing carcass, post-natal growth and reproductive traits
in commercial Angus cattle, Anim. Genet., 41, 597–607, 2010.
Mukherjee, R., Jow, L., Croston, G. E., and Paterniti, J. R.: Identifi-
cation, characterization, and tissue distribution of human perox-
isome proliferator-activated receptor (PPAR) isoforms PPARy2
versus PPARy1 and activation with retinoid X receptor agonists
and antagonists, J. Biol. Chem., 272, 8071–8076, 1997.
Palomer, X., Barroso, E., Zarei, M., Botteri, G., and Vázquez-
Carrera, M.: PPARβ/δand lipid metabolism in the heart,
Biochim. Biophys. Acta., 1861, 1–10, 2016.
Perlstein, T. S., Gerhard-Herman, M., Hollenberg, N. K., Williams,
G. H., and Thomas, A: Insulin induces renal vasodilation, in-
creases plasma renin activity, and sensitizes the renal vasculature
to angiotensin receptor blockade in healthy subjects[J], J. Am.
Soc. Nephrol., 18, 944–951, 2007.
Ren, J., Duan, Y., Qiao, R., Yao, F., Zhang, Z., Yang, B., Guo,
Y., Xiao, S., Wei, R., Ouyang, Z., Ding, N., Ai, H., and
Huang, L.: A missense mutation in PPARD causes a major
QTL effect on ear size in pigs, PloS Genet., 7, e1002043,
doi:10.1371/journal.pgen.1002043, 2011.
Rosalesreynoso, M. A., Wencechavez, L. I., Arredondovaldez, A.
R., Dumois-Petersen, S., Barros-Núñez, P., Gallegos-Arreola,
M. P., Flores-Martínez, S. E., and Sánchez-Corona, J.: Protec-
tive role of +294 T/C (rs2016520) polymorphism of PPARD in
Mexican patients with colorectal cancer, Genet. Mol. Res., 16,
doi:10.4238/gmr16019324, 2017.
Song, J. F., Wang, T., Zhu, J., Zhou, X. Y., Lu, Q., Guo, H., Zhang,
F., Wang, Y., Li, W., Wang, D. D., Cui, Y. W., Lv, D. M., and
Yin, X. X.: PPARD rs2016520 polymorphism affects repaglinide
response in Chinese Han patients with type 2 diabetes mellitus,
Clin. Exp. Pharmacol. Physiol., 42, 27–32, 2015.
Spötter, A., Hamann, H., Müller, S., and Distl, O.: Effect of poly-
morphisms in four candidate genes for fertility on litter size in a
German pig line, Reprod. Domest. Anim., 45, 579–584, 2010.
Stelma, F., Jansen, L., Sinnige, M. J., van Dort, K. A., Takken-
berg, R. B., Janssen, H. L., Reesink, H. W., and Kootstra, N. A.:
HLA-C and KIR combined genotype as new response marker
for HBeAg-positive chronic hepatitis B patients treated with
interferon-based combination therapy, J. Viral. Hepat., 23, 652–
659, https://doi.org/10.1111/jvh.12525, 2016.
Tugwood, J. D., Aldridge, T. C., Lambe, K. G., MacDonald, N.,
and Woodyatt, N. J.: Peroxisome proliferator-activated receptors:
structures and function, Ann. N. Y. Acad. Sci., 804, 252–265,
1996.
Vedhachalam, C., Duong, P. T., Nickel, M., Nguyen, D.,
Dhanasekaran, P., Saito, H., Rothblat, G. H., Lund-Katz, S., and
Phillips, M. C.: Mechanism of ATP-binding cassette transporter
A1-mediated cellular lipid efflux to apolipoprotein AI and for-
mation of high density lipoprotein particles, J. Biol. Chem. 282,
25123–25130, 2007.
Youssef, J. and Badr, M. Z.: PPARs: history and advances, Methods
Mol. Biol., 952, 1–6, 2013.
Zhang, Y., Gao, T., Hu, S., Lin, B., Yan, D., Xu, Z., Zhang, Z., Mao,
Y., Mao, H., Wang, L., Wang, G., Xiong, Y., and Zuo, B.: The
functional SNPs in the 5’regulatory region of the porcine PPARD
gene have significant association with fat deposition traits, PloS
One, 10, e0143734, doi:10.1371/journal.pone.0143734, 2015.
Arch. Anim. Breed., 60, 357–362, 2017 www.arch-anim-breed.net/60/357/2017/
... A total of 673 Chinese indigenous cattle were used in this study, including Luxi cattle (LX, N = 113, Shandong Province), Qinchuan cattle (QC, N = 286, Shaanxi Province), Nanyang cattle (NY, N = 135, Henan Province), and Jiaxian Red cattle (JX, N = 139, Henan Province) (Huang et al., 2017. All individuals selected were between 2 and 5 years old, healthy, and unrelated. ...
Article
Full-text available
Methionine sulfoxide reductase B3 (MSRB3) is instrumental in ossification and fat deposition, which regulate the growth and development of cattle directly. The purpose of this study was aimed to explore insertions and deletions (indels) in MSRB3 gene and investigate their association with growth traits in four indigenous cattle breeds (Luxi cattle, Qinchuan cattle, Nanyang cattle, and Jiaxian Red cattle). Four indels were identified by sequencing with DNA pool. Association analysis showed that three of them were associated with growth traits (P<0.05). For P1, the DD (deletion and deletion) genotype was significantly associated with body length of Nanyang cattle; for P6, II (insertion and insertion) and/or DD genotypes were significantly associated with enhanced growth traits of Qinchuan cattle; for P7, II genotype was significantly associated with hucklebone width of Luxi cattle. Our results demonstrated that the polymorphisms in bovine MSRB3 gene were significantly associated with growth traits, which could be candidate loci for marker-assisted selection (MAS) in cattle breeding.
Article
The palatal organ is a filter-feeding related organ and occupies a considerable proportion of the head of bighead carp (Hypophthalmichthys nobilis), a large cyprinid fish intensive aquaculture in Asia. In this study, we performed RNA-seq of the palatal organ during growth periods of two (M2), six (M6) and 15 (M15) months of age after hatching. The numbers of differentially expressed genes (DEGs) were 1384, 481 and 1837 for M2 VS M6, M6 VS M15 and M2 VS M15 respectively. The following signaling pathways of energy metabolism and cytoskeleton function were enriched, including ECM-receptor interaction, Cardiac muscle contraction, Steroid biosynthesis and PPAR signaling pathway. Several members of collagen family (col1a1, col2a1, col6a2, col6a3, col9a2), Laminin gamma 1 (lamc1), integrin alpha 1 (itga1), Fatty acid binding protein 2 (fads2) and lipoprotein lipase (lpl), and Protein tyrosine kinase 7 (Ptk7) are candidate genes for growth and development of basic tissues of the palatal organ. Furthermore, taste-related genes such as fgfrl1, fgf8a, fsta and notch1a were also identified, which may be involved in the development of taste buds of the palatal organ. The transcriptome data obtained in this study provide insights into the understanding functions and development mechanisms of palatal organ, and potential candidate genes that may be related to the genetic modulation of head size of bighead carp.
Article
Full-text available
Peroxisome-proliferator-activated receptor gamma (PPARγ) is a key transcription factor that controls adipocyte differentiation and energy in mammals. Therefore, PPARγ is a potential factor influencing animal growth traits. This study primarily evaluates PPARγ as candidate gene for growth traits of cattle and identifies potential molecular marker for cattle breeding. Per previous studies, PPARγ mRNA was mainly expressed at extremely high levels in adipose tissues as shown by quantitative real-time polymerase chain reaction analysis. Three novel SNPs of the bovine PPARγ gene were identified in 514 individuals from six Chinese cattle breeds: SNP1 (AC_000179.1 g.57386668 C > G) in intron 2 and SNP2 (AC_000179.1 g.57431964 C > T) and SNP3 (AC_000179.1 g.57431994 T > C) in exon 7. The present study also investigated genetic characteristics of these SNP loci in six populations. Association analysis showed that SNP1 and SNP3 loci significantly affect weaning growth traits, especially body weight of Nanyang cattle. These results revealed that SNP1 and SNP3 are potential molecular markers for cattle breeding.
Article
Full-text available
PPARD encodes for peroxisome proliferator-activated receptor delta, which plays a significant role in controlling lipid metabolism, atherosclerosis, inflammation, cancer growth, progression, and apoptosis. Accumulated evidence suggests that the polymorphism rs2016520 in PPARD is associated with lipid metabolism, obesity, metabolic syndrome, and type 2 diabetes mellitus. The aim of this study was to determine whether the single nucleotide polymorphism +294T/C (rs2016520) in PPARD is associated with colorectal cancer (CRC) in the Mexican population. Genomic DNA from 178 CRC patients and 97 healthy blood donors was analyzed. The polymorphism was identified by the polymerase chain reaction-restriction fragment length polymorphism method. Results demonstrated that patients with the T/C genotype for the +294T/C (rs2016520) polymorphism present a protective role against CRC [odds ratio (OR) = 0.39; 95% confidence interval (CI) = 0.22-0.69; P = 0.0008]. This association was also evident for the T/C genotype in the stratified analysis by tumor-node-metastasis stages I+II (OR = 0.26, P = 0.0332) and III+IV (OR = 0.44, P = 0.0067). However, in the stratified analysis by tumor location, we observed an increased risk of rectal cancer (OR = 7.57, P = 0.0403) vs colon cancer (OR = 4.87, P = 0.234) in patients carrying the C/C genotype and under the dominant and recessive models of inheritance. In conclusion, for the first time, the association between the +294T/C (rs2016520) polymorphism and colorectal cancer has been studied in Mexican patients. Our results reveal that variations in PPARD may play a significant role in genetic susceptibility to colorectal cancer.
Article
Full-text available
Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear hormone receptor superfamily that can be activated by various xenobiotics and natural fatty acids. These transcription factors primarily regulate genes involved in lipid metabolism and also play a role in adipocyte differentiation. We present the expression patterns of the PPAR subtypes in the adult rat, determined by in situ hybridization using specific probes for PPAR-alpha, -beta and -gamma, and by immunohistochemistry using a polyclonal antibody that recognizes the three rat PPAR subtypes. In numerous cell types from either ectodermal, mesodermal, or endodermal origin, PPARs are coexpressed, with relative levels varying between them from one cell type to the other. PPAR-alpha is highly expressed in hepatocytes, cardiomyocytes, enterocytes, and the proximal tubule cells of kidney. PPAR-beta is expressed ubiquitously and often at higher levels than PPAR-alpha and -gamma. PPAR-gamma is expressed predominantly in adipose tissue and the immune system. Our results suggest new potential directions to investigate the functions of the different PPAR subtypes.
Article
Full-text available
Peroxisome proliferator-activated receptor delta (PPARD) is a key regulator of lipid metabolism, insulin sensitivity, cell proliferation and differentiation. In this study, we identified two Single Nucleotide Polymorphisms (SNPs, g.1015 A>G and g.1018 T>C) constituting four haplotypes (GT, GC, AC and AT) in the 5' regulatory region of porcine PPARD gene. Functional analysis of the four haplotypes showed that the transcriptional activity of the PPARD promoter fragment carrying haplotype AC was significantly lower than that of the other haplotypes in 3T3-L1, C2C12 and PK-15 cells, and haplotype AC had the lowest binding capacities to the nuclear extracts. Transcription factor 7-like 2 (TCF7L2) enhanced the transcription activities of promoter fragments of PPARD gene carrying haplotypes GT, GC and AT in C2C12 and 3T3-L1 cells, and increased the protein expression of PPARD gene in C2C12 myoblasts. TCF7L2 differentially bound to the four haplotypes, and the binding capacity of TCF7L2 to haplotype AC was the lowest. There were significant associations between -655A/G and fat deposition traits in three pig populations including the Large White × Meishan F2 pigs, France and American Large White pigs. Pigs with genotype GG had significantly higher expression of PPARD at both mRNA and protein level than those with genotype AG. These results strongly suggested that the SNPs in 5' regulatory region of PPARD genes had significant impact on pig fat deposition traits.
Article
Full-text available
Peroxisome proliferator activated receptors alpha (PPARí µí»¼) and delta (PPARí µí»¿) belong to the nuclear receptor superfamily. PPARí µí»¼ is a target of well established lipid-lowering drugs. PPARí µí»¿ (also known as PPARí µí»½/í µí»¿) has been investigated as a promising antidiabetic drug target; however, the evidence in the literature on PPARí µí»¿ effect on hepatic lipid metabolism is inconsistent. Mice conditionally expressing human PPARí µí»¿ demonstrated pronounced weight loss and promoted hepatic steatosis when treated with GW501516 (PPARí µí»¿-agonist) when compared to wild type mice. This effect was completely absent in mice with either a dominant negative form of PPARí µí»¿ or deletion of the DNA binding domain of PPARí µí»¿. This confirmed the absolute requirement for PPARí µí»¿ in the physiological actions of GW501516 and confirmed the potential utility against the human form of this receptor. Surprisingly the genetic deletion of PPARí µí»¼ also abrogated the effect of GW501516 in terms of both weight loss and hepatic lipid accumulation. Also the levels of the PPARí µí»¼ endogenous agonist 16:0/18:1-GPC were shown to be modulated by PPARí µí»¿ in wild type mice. Our results show that both PPARí µí»¿ and PPARí µí»¼ receptors are essential for GW501516-driven adipose tissue reduction and subsequently hepatic steatosis, with PPARí µí»¼ working downstream of PPARí µí»¿.
Article
Full-text available
We studied whether PPARβ/δ deficiency modifies the effects of high fructose intake (30% fructose in drinking water) on glucose tolerance and adipose tissue dysfunction, focusing on the CD36-dependent pathway that enhances adipose tissue inflammation and impairs insulin signaling. Fructose intake for 8weeks significantly increased body and liver weight, and hepatic triglyceride accumulation in PPARβ/δ-deficient mice but not in wild-type mice. Feeding PPARβ/δ-deficient mice with fructose exacerbated glucose intolerance and led to macrophage infiltration, inflammation enhanced mRNA and protein levels of CD36, and activation of the JNK pathway in white adipose tissue compared to those of water-fed PPARβ/δ-deficient mice. Cultured adipocytes exposed to fructose also exhibited increased CD36 protein levels and this increase was prevented by the PPARβ/δ activator GW501516. Interestingly, the levels of the nuclear factor E2-related factor 2 (Nrf2), a transcription factor reported to up-regulate Cd36 expression and to impair insulin signaling, were increased in fructose-exposed adipocytes whereas co-incubation with GW501516 abolished this increase. In agreement with Nrf2 playing a role in the fructose-induced CD36 protein level increases, the Nrf2 inhibitor trigonelline prevented the increase and the reduction in insulin-stimulated AKT phosphorylation caused by fructose in adipocytes. Protein levels of the well-known Nrf2 target gene NAD(P)H:quinone oxidoreductase 1 (Nqo1) were increased in water-fed PPARβ/δ-null mice, suggesting that PPARβ/δ deficiency increases Nrf2 activity; and this increase was exacerbated in fructose-fed PPARβ/δ-deficient mice. These findings indicate that the combination of high fructose intake and PPARβ/δ deficiency increases CD36 protein levels via Nrf2, a process that promotes chronic inflammation and insulin resistance in adipose tissue. Copyright © 2015. Published by Elsevier B.V.
Article
Current treatment for chronic hepatitis B infection (CHB) consists of interferon-based therapy. However, for unknown reasons, a large proportion of patients with CHB do not respond to this treatment. Hence, there is a pressing need to establish response markers to select patients who will benefit from therapy and to spare potential nonresponders from unnecessary side effects of antiviral therapy. Here, we assessed whether HLA-C and KIR genotypes were associated with treatment outcome for CHB. Twelve SNPs in or near the HLA-C gene were genotyped in 86 CHB patients (41 HBeAg positive; 45 HBeAg negative) treated with peginterferon alfa-2a + adefovir. Genotyping of killer immunoglobin-like receptors (KIRs) was performed by SSP-PCR. One SNP in HLA-C (rs2308557) was significantly associated with combined response in HBeAg-positive CHB patients (P = 0.003). This SNP is linked to the HLA-C group C1 or C2 classification, which controls KIR binding. The combination of KIR2DL1 with its ligand HLA-C2 was observed significantly more often in HBeAg-positive patients with a combined response (13/14) than in nonresponders (11/27, P = 0.001). Patients with the KIR2DL1/C2 genotype had significantly higher baseline ALT levels (136 vs 50 U/L, P = 0.002) than patients without this combination. Furthermore, KIR2DL1-C2 predicted response independent of HBV genotype and ALT at baseline. HLA-C and KIR genotype is strongly associated with response in HBeAg-positive CHB patients treated with interferon-based therapy. In combination with other known response markers, HLA-C/KIR genotype could enable the selection of patients more likely to respond to interferon-based therapy.
Article
Cardiac lipid metabolism is focus of attention due to its involvement in the development of cardiac disorders. Both a reduction and an increase in fatty acid utilization make the heart more prone to the development of lipotoxic cardiac dysfunction. The ligand-activated transcription factor peroxisome proliferator-activated receptor (PPAR)β/δ modulates different aspects of cardiac fatty acid metabolism, and targeting this nuclear receptor can improve heart diseases caused by altered fatty acid metabolism. In addition, PPARβ/δ regulates glucose metabolism, the cardiac levels of endogenous antioxidants, mitochondrial biogenesis, cardiomyocyte apoptosis, the insulin signaling pathway and lipid-induced myocardial inflammatory responses. As a result, PPARβ/δ ligands can improve cardiac function and ameliorate the pathological progression of cardiac hypertrophy, heart failure, cardiac oxidative damage, ischemia-reperfusion injury, lipotoxic cardiac dysfunction and lipid-induced cardiac inflammation. Most of these findings have been observed in preclinical studies and it remains to be established to what extent these intriguing observations can be translated into clinical practice. This article is part of a Special Issue entitled: Heart Lipid Metabolism edited by G.D. Lopaschuk.