Skip to main content
BMC Genomics logoLink to BMC Genomics
. 2018 Feb 9;19(Suppl 3):0. doi: 10.1186/s12864-018-4478-3

Candidate SNP markers of reproductive potential are predicted by a significant change in the affinity of TATA-binding protein for human gene promoters

Irina V Chadaeva 1,2, Petr M Ponomarenko 3, Dmitry A Rasskazov 1, Ekaterina B Sharypova 1, Elena V Kashina 1, Dmitry A Zhechev 1, Irina A Drachkova 1, Olga V Arkova 1,4, Ludmila K Savinkova 1, Mikhail P Ponomarenko 1,2,, Nikolay A Kolchanov 1,2, Ludmila V Osadchuk 1,5, Alexandr V Osadchuk 1
PMCID: PMC5836831  PMID: 29504899

Abstract

Background

The progress of medicine, science, technology, education, and culture improves, year by year, quality of life and life expectancy of the populace. The modern human has a chance to further improve the quality and duration of his/her life and the lives of his/her loved ones by bringing their lifestyle in line with their sequenced individual genomes. With this in mind, one of genome-based developments at the junction of personalized medicine and bioinformatics will be considered in this work, where we used two Web services: (i) SNP_TATA_Comparator to search for alleles with a single nucleotide polymorphism (SNP) that alters the affinity of TATA-binding protein (TBP) for the TATA boxes of human gene promoters and (ii) PubMed to look for retrospective clinical reviews on changes in physiological indicators of reproductive potential in carriers of these alleles.

Results

A total of 126 SNP markers of female reproductive potential, capable of altering the affinity of TBP for gene promoters, were found using the two above-mentioned Web services. For example, 10 candidate SNP markers of thrombosis (e.g., rs563763767) can cause overproduction of coagulation inducers. In pregnant women, Hughes syndrome provokes thrombosis with a fatal outcome although this syndrome can be diagnosed and eliminated even at the earliest stages of its development. Thus, in women carrying any of the above SNPs, preventive treatment of this syndrome before a planned pregnancy can reduce the risk of death. Similarly, seven SNP markers predicted here (e.g., rs774688955) can elevate the risk of myocardial infarction. In line with Bowles’ lifespan theory, women carrying any of these SNPs may modify their lifestyle to improve their longevity if they can take under advisement that risks of myocardial infarction increase with age of the mother, total number of pregnancies, in multiple pregnancies, pregnancies under the age of 20, hypertension, preeclampsia, menstrual cycle irregularity, and in women smokers.

Conclusions

According to Bowles’ lifespan theory—which links reproductive potential, quality of life, and life expectancy—the above information was compiled for those who would like to reduce risks of diseases corresponding to alleles in own sequenced genomes. Candidate SNP markers can focus the clinical analysis of unannotated SNPs, after which they may become useful for people who would like to bring their lifestyle in line with their sequenced individual genomes.

Electronic supplementary material

The online version of this article (10.1186/s12864-018-4478-3) contains supplementary material, which is available to authorized users.

Keywords: Reproductive potential, Gene, Promoter, TATA box, TATA-binding protein, Single nucleotide polymorphism, SNP marker, Keyword-based search, Prediction, Verification

Background

Incessant progress in medical and biological sciences, advancement of technology, and education year in and year out improve quality of life and life expectancy of the population, creating comfortable conditions for active living. Nonetheless, there are numerous factors that adversely affect human health. They can include, for example, different kinds of environmental pollution, an increase in population density, which leads to the rapid spread of infections and parasitoses, and an increase in psychological stress. This situation not only reduces the quality of life and longevity of the individual but also has a deferred, long-term effect on the next generation, by acting as a mutagen [1]. The accumulating mutational load often worsens health and reduces the subsequent generation’s survival and adaptation to their habitat that ultimately reduces the chances of sustainable population reproduction.

The effects of the above factors limit individual reproductive potential: a concept used in population ecology to assess the evolutionary success of an individual [2] or a population [3]. In the 1970s, Eric Pianka defined reproductive potential as the most important conditional indicator reflecting a population’s ability to reproduce, survive, and develop under optimal ecological conditions [25]. In the context of human society, in the term “reproductive potential,” researchers can also include the mental state and physical state that allow a person to produce healthy offspring when social and physical maturity is achieved. Consequently, reproductive potential depends not only on physiological readiness for reproduction (primarily the reproductive system), but also on the general physical condition (with the exception of existing diseases that are incompatible with the implementation of reproduction) and on socio-economic status. With this in mind, everything is focused on individual ability for reproduction until the next generation becomes reproductive. In particular, not only the phenotype plays a role here, but so does the genotype, where most abilities of a given individual are encoded, both normal and mutational as well as epigenetic ones. It should also be noted that reproductive potential varies throughout the life cycle and does so in different ways for men and women. Ideally, the evaluation of reproductive potential would include not only the direct material and energy costs of reproduction but also the price of the risk associated with future reproductive attempts [5].

Predictive-preventive personalized medicine may help to improve individual reproductive success. Its methods include prediction (based on the analysis of the genome) of the probability of a specific disease, analysis of individual indicators, biomarkers (such as single nucleotide polymorphisms, SNPs [6, 7]), and the development of preventive and therapeutic measures for changing the physiological parameters of the reproductive potential in patients [8]. In particular, the analysis of SNP biomarkers allows a physician not only to make a prognosis for a patient regarding possible diseases that can reduce reproductive potential but also to adjust the prescribed treatment, taking into account individual characteristics and reactions to medicines.

In addition, according to Bowles’ lifespan theory [9], which links reproductive potential, quality of life, and life expectancy of an individual, it is possible timely to prevent diseases, which correspond to the alleles of the decoded genotype.

Within the framework of the biggest modern scientific project “1000 Genomes”, 10545 individual genomes have already been sequenced [10]. The “reference human genome” is publicly available via the Ensembl database [11] using the Web service UCSC Genome Browser [12]. A total of 100,877,027 SNPs have been experimentally identified and stored in the dbSNP database [6]. Database dbWGFP [13] containing 8.58 billion possible human whole-genome SNPs has already been created for accumulation of predictions, experimental data, clinical observations, and any other information relevant for biomedical analysis of individual genomes. For such an analysis, the most valuable biomedical SNP markers—within the framework of personalized medicine—are those that can differ between the individual human genomes of patients having some pathology and the reference human genome [14]. To find such markers, cohorts of patients with a given disease and healthy volunteers (as a control) are compared in a clinical study (e.g., [15]).

As far as human health is concerned, the clinical search for biomedical SNP markers is the only acceptable method. Nevertheless, it is so laborious and expensive that its application to all 8.58 billion potentially possible SNPs [13] and all known human pathologies is rather unlikely. Moreover, both Haldane’s dilemma [16] and Kimura’s theory of neutral evolution [17] independently predict that the absolute majority of SNPs in humans are neutral and do not affect health in any way; thus, it is unclear why it is necessary to verify them clinically. With this in mind, the mainstream clinical search for SNP markers of a given disease is currently limited by the simplest idea about heuristic handmade selection of candidate SNPs for clinical testing among unannotated SNPs on the basis of their location near the human genes that are already clinically associated with this disease (e.g., [18, 19]). Accordingly, computer-based preliminary analysis of unannotated SNPs can eliminate the absolute majority of neutral SNPs to make the clinical cohort-based search for biomedical SNP markers faster, cheaper, and more targeted [20]. There are many public Web services [2138] that facilitate the computer-based search for candidate SNP markers using various similarity measures based on whole-genome data in health [39], after treatment [40], and during a disease [41] or infection [42] to eliminate unannotated SNPs that bear the least resemblance to known biomedical SNP markers (i.e. to eliminate the most probable neutral SNPs). The Central Limit Theorem predicts that the accuracy of this similarity-based elimination of unannotated neutral SNPs increases with the increase in the size and diversity of whole-genome data under study [43].

Now, the best accuracy of this mainstream search corresponds to SNPs in protein-coding regions of genes [44], i.e., SNPs that damage proteins [45] whose defects are uncorrectable by treatment or lifestyle changes. On the contrary, the worst accuracy of this kind of search is seen for regulatory SNPs [11], which alter concentrations of proteins without any damage to the proteins, and such problems are correctable by medication and lifestyle. The best balance between the predictability and biomedical usefulness corresponds to the regulatory SNPs between nucleotide positions -70 and –20 upstream of a transcription start site (TSS) [46, 47] where TATA-binding protein (TBP) binds to the promoter at the very beginning of transcription initiation. This TBP–promoter complex is obligatory for any TSSes because the TBP knockout model animals (TBP−/−) are always inviable since their development cannot proceed past the blastula stage because their maternal supply of TBP is exhausted [48, 49]. Moreover, the TBP–promoter affinity linearly correlates with the transcription magnitude of the human gene containing this promoter [50]. This notion has been repeatedly confirmed experimentally (for review, see [51]). The canonical form of the TBP-binding site (TATA box, synonyms: Hogness box and Goldberg-Hogness box [52]) is the best-studied regulatory element among human gene promoters [47].

In our previous studies, we developed public Web service SNP_TATA_Comparator (http://beehive.bionet.nsc.ru/cgi-bin/mgs/tatascan/start.pl) [53] and applied it to predict candidate SNP markers within TATA boxes of human genes associated with obesity [54], autoimmune diseases [55], chronopathology [56], aggressiveness [57, 58], Alzheimer’s disease [59], and efficacy of anticancer chemotherapy [60] (for review, see [20]). In the present work, we applied our Web service [53] in the same way to human reproductive potential as the most common concept of population ecology dealing with the evolutionary success of either individuals [2] or populations [3].

Results

Tables 1, 2, 3, 4, 5, 6 and 7 present the results obtained by our Web service [53] for the 126 known and candidate reproductive-potential-related SNP markers in the TBP-binding sites of human gene promoters (see Methods: Supplementary Method, Additional file 1).

Table 1.

Known and candidate SNP markers of tumors in reproductive organs

Gene dbSNP [6] rel. 147 or see [Ref] 5′ flank wt
mut
3′ flank KD, nM Known diseases (SNP markers) or hypothetical disease (candidate SNP markers) [Ref] or [this work]
wt
mut
Δ Z α ρ
ESR2 rs35036378 cctctcggtc t
g
ttaaaaggaa 6
8
5 10-3 B ESR2-deficient pT1 breast tumor needing tamoxifen prophylaxis against cancer [61]
rs766797386 ttaaaaggaa g
t
aaggggctta 6
7
3 10-2 C (hypothetically) the same disease [this work]
HSD17B1 rs201739205 aggtgatatc a
c
agcccagagc 13
18
5 10-6 A higher risk of breast cancer [64]
rs201739205 agcaggtgat a
t
tcaagcccag 13
35
18 10-6 A (hypothetically) the same disease [this work]
rs748743528 gcaggtgata t
c
caagcccaga 13
28
13 10-6 A
rs755636251 ggcgaagcag g
t
tgatatcaag 13
11
2 0.05 D (hypothetically) higher risk of breast cancer [68]
PGR rs10895068 gggagataaa g
a
gagccgcgtg 10
6
8 10-6 A endometrial cancer caused by the spurious TATA box and its TSS disbalancing both α and β isoforms of progesterone receptor [65]
rs544843047 agtcgggaga t
c
aaaggagccg 10
22
14 10-6 A (hypothetically) health as the norm without the above-mentioned spurious TATA box [this work]
GSTM3 rs1332018 ccccttatgt c
a
gggtataaag 4
3
= 2 1 E maternal “c” (Wb: TF-binding site damaged, not TATA box), elevates risk of a brain tumor in her child, renal cancer, and Alzheimer’s disease [66, 67]
rs200209906 gtataaagcc c
t,a
ctcccgctca 3.6
4.3
2 1 E (hypothetically) the same disease and low risks of breast cancer in those who never drink alcohol and lesser Hg-resistance during reproduction [this work], [69]
rs750789679 cgggtataaa g
c
cccctcccgc 3.6
4.5
3 10-2 C
rs748231432 cccttatgtc g
c,t
ggtataaagc 3.6
3.0
3 0.05 D (hypothetically) lower risk of a brain tumor in a child whose mother has “c”-allele of rs1332018 [this work], [66]
rs763859166 gggtataaag c
t
ccctcccgct 3.6
2.9
3 10-2 C

Hereinafter, ancestral (wt) and minor (mut) alleles; KD, dissociation constant of TBP–DNA interaction; Δ, a change: overexpression (↑), deficit (↓), norm (=); α = 1 – p, significance {where p value is shown in Fig. 1; α = 1 denotes insignificance}; ρ, heuristic rank of candidate SNP markers varying in alphabetical order from the “best” (A) to the “worst” (E); the CETP gene: 18bp, the 18-bp deletion 5’-gggcggacatacatatac-3’; the F3 gene: 30bp, 17bp, and 18bp as the insertions 5’-agaccttcataagaaataatcctgatccaa-3’, 5’-tgctgcgtactggcaaa-3’, and 5’-acggcgtagagactggga-3’ of 30 bp, 17 bp, and 18 bp in length, respectively; EMSA, electrophoretic mobility shift assay; Hg, mercury; LUC, luciferase reporter assay; TF, transcription factor; Wb: western blot.

Table 2.

Known and candidate SNP markers of tumors in nonreproductive organs

Gene dbSNP [6] rel. 147 or see [Ref] 5′ flank wt
mut
3′ flank KD, nM Known diseases (SNP markers) or hypothetical disease (candidate SNP markers) [Ref] or [this work]
wt
mut
Δ Z α ρ
IL1B rs1143627 ttttgaaagc c
t
ataaaaacag 5
2
15 10-6 A high risks of gastric, liver, and non–small cell lung cancers; gastric ulcer, chronic gastritis, recurrent major depression, obesity, Graves’ disease, pre-eclampsia, (hypothetically) short time-to-delivery in pregnancy and childbirth [7785] [this work]
rs549858786 tgaaagccat a
t
aaaacagcga 5
7
8 10-6 A (hypothetically) lesser risk of the same diseases [60]
CYP2A6 rs28399433 tcaggcagta t
g
aaaggcaaac 2
9
21 10-6 A low risk of lung cancer in smokers (LUC: “-34g” corresponds to 50% of “-34t”), (hypothetically) lesser damage from secondhand smoke in pregnant women who are nonsmokers [86, 87], [this work], [9092]
rs761592914 tttttcaggc a
c
gtataaaggc 2
3
3 10-3 B (hypothetically) the same disease [this work]
CYP2B6 rs34223104 gatgaaattt t
c
ataacagggt 4
10
15 10-6 A TATAWT→USFSNP, TSSWT→TATASNP, and de-novo TSSSNP can cause overexpression of this gene of a bioactivator of immunosuppressive and antitumor prodrug cyclophosphamide [88]
rs563558831 tgaaatttta t
c
aacagggtgc 4
10
13 10-6 A (hypothetically) the same problem [this work]
DHFR rs10168 ctgcacaaat g
a
gggacgaggg 15
9
9 10-6 A resistance to methotrexate treatment of leukemia and (hypothetically) that in the cases of ectopic pregnancy, metastatic choriocarcinoma, and gestational trophoblastic disease [89], [this work], [93]
rs750793297 tgcacaaatg g
t
ggacgagggg 15
13
3 10-2 C (hypothetically) the same diseases [this work]
rs766799008 ctgcacaaat a
g
tggggacgag 15
19
3 10-3 B (hypothetically) greater bioactivity of methotrexate during treatment of leukemia, ectopic pregnancy, metastatic choriocarcinoma, and gestational trophoblastic disease [this work], [60]
rs764508464 ctgcacaaat a
-
tggggacgag 15
37
17 10-6 A
rs754122321 ctcgcctgca c
g
aaatggggac 15
25
9 10-3 B

See “Note” under Table 1

Table 3.

Known and candidate reproductivity-related SNP markers in genes of hormones

Gene dbSNP [6] rel. 147 or see [Ref] 5′ flank wt
mut
3′ flank KD, nM Known diseases (SNP markers) or hypothetical disease (candidate SNP markers) [Ref] or [this work]
wt
mut
Δ Z α ρ
LEP rs201381696 tcgggccgct a
g
taagaggggc 4
12
17 10-6 A hypoleptinemia elevates risk of obesity [54, 106]
rs200487063 tgatcgggcc g
a
ctataagagg 4
2
6 10-6 A (hypothetically) hyperleptinemia elevates risk of hypertension in obesity [this work], [107, 110]
rs34104384 ccgctataag a
t
ggggcgggca 4
3
4 10-2 C
GCG rs183433761 gctggagagt a
g
tataaaagca 0.9
1.6
17 10-6 A resistance to obesity during a high-fat diet [54]
(hypothetically) hypoglucogonemia decreases pregnancy probability, serum insulin in pregnancy, and during late gestational period [this work], [111, 112]
rs757035851 tatataaaag cag
-
tgcgccttgg 0.9
1.1
3 10-3 B
GH1 rs11568827 aggggccagg g
-
tataaaaagg 1.5
1.4
= 1 1 E short stature (EMSA: unknown TF-binding site lost, not TATA box) [107]
(hypothetically) higher risk of GH1 deficiency as clinical syndrome whose symptoms are increased central adiposity, atherogenesis, as well as cerebrovascular and cardiac morbidity (and mortality), and, also, decreased lean body mass, bone mineral density, quality of life [this work], [113]
rs796237787 gaaggggcca g
-
ggtataaaaa
rs768454929 agggtataaa a
c
agggcccaca 1.5
2.6
7 10-6 A
rs761695685 gccagggtat a
g
aaaagggccc 1.5
5.8
19 10-6 A
rs774326004 ccagggtata a
t
aaagggccca 1.5
0.9
7 10-6 A (hypothetically) higher risks of acromegaly [this work], [114]
rs777003420 aaggggccag g
t
gtataaaaag 1.5
1.3
3 0.05 D
INS rs5505 agatcactgt c
t
cttctgccat 53
44
4 10-3 B type 1 diabetes after neonatal diabetes mellitus [108]
(hypothetically) hyperinsulinemia elevates the placental leptin which causes neonatal macrosomia [this work], [115]
rs563207167 tcagccctgc c
t
tgtctcccag 53
44
4 10-3 B
rs11557611 gatcactgtc c
t
ttctgccatg 53
60
2 0.05 D (hypothetically) hypoinsulinemia slows down fetal growth [this work], [116]

See “Note” under Table 1

Table 4.

Known and candidate reproductivity-related SNP markers in genes of other metabolic proteins

Gene dbSNP [6] rel. 147 or see [Ref] 5′ flank wt
mut
3′ flank KD, nM Known diseases (SNP markers) or hypothetical disease (candidate SNP markers) [Ref] or [this work]
wt
mut
Δ Z α ρ
NOS2 -51t→c [288] gtataaatac t
c
tcttggctgc 2
1
3 10-2 C resistance to malaria and epilepsy (hypothetically) higher risk of gestational diabetes mellitus [288290], [this work], [147]
STAR rs16887226 cagccttcag c
t
gggggacatt 10
10
= 0 1 E hypertensive diabetic patients, (EMSA: unknown TF-binding site lost rather than TATA box) [291]
rs544850971 tcagcggggg a
g
catttaagac 10
12
5 10-2 C (hypothetically) lower risk of the same disease and congenital adrenal hyperplasia [this work], [148]
APOA1 35a→c [292] tgcagacata a
c
ataggccctg 3
4
5 10-6 A fatty liver (hypothetically) high risk of polycystic ovary syndrome in young women [292], [this work], [149]
CETP DEL-51(18 bp) [293] cgtgggggct 18bp
-
gggctccagg 4
7
7 10-6 A hyperalphalipoproteinemia reducing risk of atherosclerosis [293]
rs17231520 ggggctgggc g
a
gacatacata 4
2
10 10-6 A (hypothetically) biomarker of late pregnancy when plasma triglyceride, high-density lipoprotein, and cholesterol concentrations are significantly increased [this work], [150]
rs569033466 atacatatac g
a
ggctccaggc 4
3
4 10-3 B
rs757176551 catatacggg c
g
tccaggctga 4
2
10 10-6 A
SOD1 rs7277748 ggtctggcct a
g
taaagtagtc 2
7
17 10-6 A amyotrophic lateral sclerosis, (hypothetically), asthenospermia, lower female fertility via progesterone deficiency [294], [this work], [151, 152]
TPI1 rs1800202 gcgctctata t
g
aagtgggcag 1
4
17 10-6 B hemolytic anemia, neuromuscular diseases [295, 296]
(hypothtically) higher risk of asthenospermia [this work], [153]
rs781835924 cgcggcgctc t
c
atataagtgg 1
2
10 10-6 B
GJA5 rs10465885 caactaagat g
a
tattaaacac 3
3
= 1 1 E arrhythmia, cardiovascular events (LUC: TF-binding site damaged, not TATA box) [297]
(hypothetically) the same disease and higher risk of heart morphogenesis disorders [this work], [154]
rs587745372 ggcgacagat a
t
cgattaaaaa 6
7
3 10-3 B
rs35594137 gaggagggaa g
a
gcgacagata 6
6
= 0 1 E
arrhythmia, cardiovascular events (LUC: TF-binding site damaged, not TATA box) [298]

See “Note” under Table 1

Table 5.

Known and candidate reproductivity-related SNP markers related to blood proteins

Gene dbSNP [6] rel. 147 or see [Ref] 5′ flank wt
mut
3′ flank KD, nM Known diseases (SNP markers) or hypothetical disease (candidate SNP markers) [Ref] or [this work]
wt
mut
Δ Z α ρ
HBB rs397509430 gggctgggca t
-
atacaacagt 5
29
34 10-6 A malaria resistance and thalassemia [176]
rs33980857 gggctgggca t
a,g,c
atacaacagt 5
21
27 10-6 A
rs34598529 ggctgggcat a
g
aaagtcaggg 5
18
24 10-6 A
rs33931746 gctgggcata a
g,c
aagtcagggc 5
11
14 10-6 A
rs33981098 agggctgggc a
g,c
taaaagtcag 5
9
10 10-6 A
rs34500389 cagggctggg c
a,t,g
ataaaagtca 5
6
3 10-2 C
(hypothetically) the same disease; heterozygotes “wt-mut” are still more viable according to most of clinical indicators in comparison with both homozygotes “wt-wt” and “mut-mut” [this work], [185]
rs63750953 ctgggcataa aa
-
gtcagggcag 5
8
9 10 -6 A
rs281864525 tgggcataaa a
c
gtcagggcag 5
7
7 10 -6 A
rs117785782 ggctgagggt t
c
tgaagtccaa 28
39
7 10 -6 A
HBD rs35518301 caggaccagc a
g
taaaaggcag 4
8
11 10-6 A malaria resistance and thalassemia [176]
(hypothetically) the same disease; heterozygotes “wt-mut” are still more viable according to most of clinical indicators [this work], [185]
rs34166473 aggaccagca t
c
aaaaggcagg 4
8
18 10 -6 A
HBG2 rs745580140 ggagttgctc ta
-
cacaagctct 11
22
10 10 -6 A
ACKR1 rs2814778 ttggctctta t
c
cttggaagca 10
12
4 10-3 B low white-blood-cell count and resistance to malaria, (hypothetically) pre-eclampsia [177, 178], [this work], [186]
MBL2 rs72661131 tctatttcta t
c
atagcctgca 2
4
12 10-6 A variable immunedefici-ency, pre-eclampsia, stroke, [190192]
(hypothetically) the same disease; higher risks of recurrent vulvovaginal infections [this work], [187]
rs562962093 atctatttct a
g
tatagcctgc 2
5
15 10-6 A
rs567653539 tttctatata g
a
cctgcaccca 2
1
12 10-6 A (hypothetically) reduced risks of recurrent vulvovaginal infections
MMP12 rs2276109 gatatcaact a
g
tgagtcactc 11
14
3 10-2 C lower risk of psoriasis, systemic sclerosis, asthma [193195]
(hypothetically), higher risk of ovarian hyper-stimulation syndrome [this work], [188]
rs572527200 gatgatatca a
g
ctatgagtca 11
14
3 10-2 C
F2 rs564528021 agttcaacat t
c
aacccagagg 13
9
7 10-6 A (hypothetically) high risk of pre-eclampsia [this work], [189]
rs752364393 caacattaac c
t
cagaggggtc 13
11
3 10-3 B

See “Note” under Table 1

Table 6.

Known and candidate reproductivity-related SNP markers related to coagulation of blood

Gene dbSNP [6] rel. 147 or see [Ref] 5′ flank wt
mut
3′ flank KD, nM Known diseases (SNP markers) or hypothetical disease (candidate SNP markers) [Ref] or [this work]
wt
mut
Δ Z α ρ
PROC rs528817178 cctttcattc c
t
gcttccacct 27
21
5 10-6 A (hypothetically) higher risk of tumor cell invasion [this work], [214]
rs539608065 ctttcattcc g
a
cttccacctg 27
22
4 10-3 B
rs539731824 ttgtggttat g
a
gattaactcg 10
6
8 10-6 A
rs756414294 ggcgcggcac c
t
agcaccagct 121
27
25 10-6 A
rs777687270 ggcaccagca c
t
cagctgcccg 121
59
13 10-6 A
rs746382956 tgcccgcaga g
a
gtgagcttcc 121
44
19 10-6 A
rs542626506 cacacaggga c
t
agccctttca 27
31
3 10-2 C (hypothetically) high risks of thrombosis, inflammation, and pregnancy loss [this work], [215]
rs61731661 ccctttcatt c
t
cgcttccacc 27
29
5 0.05 D
F8 rs781855957 acggcggcag c
t
ggaagaggga 75
49
8 10-6 A (hypothetically) higher risk of thrombosis [this work] [216]
THBD rs13306848 agggagggcc g
a
ggcacttata 2
2
= 1 1 E thrombosis (LUC: TF site damaged, not TATA) [211]
(hypothetically) higher risks of placental failure and fetal loss [this work], [217]
rs568801899 caatccgagt g
a
tgcggcatca 45
70
6 10-6 A
F3 rs563763767 ccctttatag c
t
gcgcggggca 3
2
6 10-6 A myocardial infarction; thrombosis; [212]
(hypothetically) higher risk of ovarian cancer [this work], [218]
rs779755900 atctcgccgc -
30bp
caactggtag 90
10
43 10-6 A
rs749456955 gatctcgccg c
a
caactggtag 90
75
4 10-3 B
rs746842194 cgatctcgcc -
17bp
gccaactggt 90
31
15 10-6 A
rs754815577 ctcgatctcg -
18bp
ccgccaactg 90
32
17 10-6 A
rs768753666 ggaacccgct c
g
gatctcgccg 90
117
5 10-6 A (hypothetically) lower risk of ovarian cancer
rs774688955 cgccacggaa c
t
ccgctcgatc 90
101
2 0.05 D
F7 -33a→c [213] ccttggaggc a
c
gagaactttg 53
62
3 10-2 C moderate bleeding [213]
(hypothetically) lower risk of ovarian cancer [this work], [218]
rs749691733 agaactttgc c
t
cgtcagtccc 53
66
4 10-3 B
rs367732974 aactttgccc g
a
tcagtcccat 53
47
2 0.05 D (hypothetically) higher risk of ovarian cancer
rs549591993 gcccgtcagt c
a
ccatggggaa 53
25
13 10-6 A
rs777947114 agagaacttt g
a
cccgtcagtc 53
19
19 10-6 A
rs770113559 gtcacccttg g
a
aggcagagaa 53
41
5 10-6 A
rs754814507 cctcccccat c
t
cctctgtcac 53
45
3 10-3 B
F11 rs754739433 tctgggaatt a
g
tttttagtaa 4
5
2 0.05 D (hypothetically) hereditary factor XI deficiency, high risk of spontaneous primary hemorrhage [this work], [219]
rs780731761 ttatttttag t
a
aaaggaaatt 4
7
8 10-6 A
rs747652067 tatttttagt a
g
aaggaaattt 4
7
9 10-6 A
rs374761594 catttgtcta c
t
tgaagcacac 13
10
3 10-3 B (hypothetically) higher risk of angioneurotic edema [this work], [220]
rs759231858 acaccaacca g
t
aataacgaag 13
4
17 10-6 A
rs752308147 ccagaataac g
a
aagctcgata 13
9
6 10-6 A
F9 rs371045754 tggtacaact a
c
atcgacctta 6
10
5 10-6 A (hypothetically) higher risk of hemophilia B [this work], [221]
rs750827465 tttggtacaa c
t
taatcgacct 6
4
7 10-6 A (hypothetically) higher risk of myocardial fibrosis [this work], [222]

See “Note” under Table 1

Table 7.

Candidate SNP markers of reproductivity-related genes

Gene dbSNP [6] rel. 147 or see [Ref] 5′ flank wt
mut
3′ flank KD, nM hypothetical disease (candidate SNP markers) [this work], [Ref]
wt
mut
Δ Z α ρ
AR rs763353257 aagggaagta g
-
gtggaagatt 30
21
6 10-6 A (hypothetically) higher risks of androgenetic alopecia and androgen-induced premature senescence in adult men [251]
rs749306567 aagggaagta g
a
gtggaagatt 30
15
11 10-6 A
rs377711437 cagcactgca g
a
ccacgacccg 75
66
2 0.05 D
MTHFR rs780207553 cacgcactct g
a
ggcctgagct 74
38
12 10-6 A (hypothetically) higher risk of pre-eclampsia [252]
rs749532075 tccctcccca c
t*)
gcactctggg 74
50
7 10-6 A
rs771960561 cctctgttcc c
t
tccccacgca 74
66
3 10-2 B
rs773214376 tgcctctgtt c
t
cctccccacg 74
66
2 0.05 D
rs566478202 ggtgcctctg t
g
tccctcccca 74
85
2 0.05 D (hypothetically) higher risk of adverse pregnancy outcomes [253]
rs752181249 gaggatctac a
c
gccatcagct 27
35
4 10-3 B
DNMT1 rs570287204 gtgggggggg -
gtg
tgtgtgcccg 52
23
11 10-6 A (hypothetically) under stress, higher risk of epigenetic disorders of fetal and newborn brain development causing long-term neurobehavioral problems that may be reversible in adolescence [254, 255]
rs534819409 cgtggggggg g
t
ggcctgagct 52
30
7 10-6 A
rs553454792 gcgtgggggg g
t
gtgtgtgccc 52
23
11 10-6 A
rs558447661 cgtggagctt g
t
gacgagccca 72
29
15 10-6 A
rs535899986 cccagcaaac c
t
gtggagcttg 72
58
5 10-3 B
rs143796354 cacctcccag c
a
aaaccgtgga 72
26
20 10-6 A
rs756103340 gcggcgcgca g
a
cggcagttgg 92
79
3 10-3 B
rs758026532 ccagcaaacc g
t*)
tggagcttgg 72
88
4 10-3 B (hypothetically) higher risks of activation of protooncogenes in cancer [256]
rs772821225 gtctccaata a
c
atgcagctgg 7
8
2 0.05 D
CYP17A1 rs758657961 ctggagttga g
a
ccagcccttg 56
30
11 10-6 A (hypothetically) higher risk of hyperandrogenism in polycystic ovary syndrome [257]
rs373488849 tgccctggag t
c
tgagccagcc 56
70
4 10-3 B (hypothetically) higher risk of fertility impairments [258]
NR5A1 rs147497093 gttcagcaag c
t
acaagagaaa 19
6
17 10-6 A (hypothetically) higher risks of adrenal tumors and endometriosis [259]
rs535432539 cgctgcttcc g
a
cttcgtaagt 31
18
9 10-6 A
rs553326158 gcgctgcttc c
t
gcttcgtaag 31
26
3 10-2 C
rs143242438 caccctcatc c
t
ggtgtgagag 31
21
6 10-6 A

See the footnote of Table 1; *)this SNP includes one more minor neutral allele: “a.”

First, we analyzed all SNPs mapped within [−70; −20] regions upstream of transcription start sites for the human genes containing the known biomedical SNP markers that alter TBP’s binding to promoters of these genes (Tables 1, 2, 3, 4, 5 and 6). Let us first describe in more detail only one human gene in order to briefly review all the others.

Known and candidate reproductivity-related SNP markers of cancers

The human ESR2 gene (estrogen receptor β) contains a known SNP marker (Fig. 1a: rs35036378) of an ESR2-deficient primary pT1 breast tumor, which is needed in tamoxifen-based prophylaxis of cancer [61] as shown in Table 1. The prediction of our Web service [53] is consistent with this independent clinical observation (Fig. 1b: text box “Results”, line “Decision” contains the label “deficiency: significant”).

Fig. 1.

Fig. 1

The result produced by SNP_TATA_Comparator [53] for reproductive potential-related SNP markers in the human ESR2 gene. Legend: a Unannotated SNPs (analyzed in this study) in the region [-70; -20] (where all proven TBP-binding sites (boxed) are located; double-headed arrow, ↔) of the human ESR2 gene promoter retrieved from dbSNP, rel. 147 [6] using the UCSC Genome Browser [12]. Dash-and-double-dot arrows: known and candidate SNP markers of reproductive potential are predicted by a significant change in the affinity of TBP for the human ESR2 gene promoter. b and c The results from our Web service SNP_TATA_Comparator [53] for the two SNP markers of reproductive potential: known marker rs35036378 [61] and candidate marker rs766797386 near the known TBP-binding site (boxed) of the human ESR2 gene promoter. Solid, dotted, and dashed arrows indicate queries in the reference human genome [10] by means of the BioPerl library [265]. Dash-and-dot arrows: estimates of significance of the alteration of gene product abundance in patients carrying the minor allele (mut) relative to the norm (ancestral allele, wt) expressed as a Z-score using package R [266]. Circles indicate the ancestral (wt) and minor (mut) alleles of the SNP marker labeled by its dbSNP ID [6]

Next, near this known biomedical SNP marker rs35036378, we found the unannotated SNP rs766797386, which can also decrease expression of the human ESR2 gene (Fig. 1c) and thus cause an ESR2-deficient primary pT1 tumor requiring prophylaxis by tamoxifen against breast cancer [61]. This result allowed us to suggest rs766797386 as a candidate SNP marker of a higher risk of breast cancer reducing reproductive potential.

Finally, using our secondary keyword search for these two SNP markers (hereinafter: see Methods: Additional file 2: Figure S1. dotted-line box, Additional file 2), we learned (hereinafter: see Table S1, Additional file 3) that cadmium (Cd) elevates the risk of a primary tumor’s becoming malignant [62], whereas mothers undergoing tamoxifen-based treatment should not breastfeed [63].

The human HSD17B1, PGR, and GSTM3 genes encode hydroxysteroid (17-β) dehydrogenase 1, progesterone receptor, and glutathione S-transferase μ3, respectively. Their promoters have the known SNP markers rs201739205, rs10895068, and rs1332018, which elevate risks of breast [64] and endometrial [65] cancers; a brain tumor in a fetus, newborn, or a child [66], respectively; as well as renal cancer and Alzheimer’s disease [67] (Table 1). Near these known biomedical SNP markers, there are four unannotated SNPs rs201739205, rs748743528, rs200209906, and rs750789679, which can similarly alter expression levels of the same genes according to the predictions of our Web service [53] (Table 1). Hence, we proposed them as the candidate SNP markers of the same diseases.

Besides, within the same promoters, we found four other unannotated SNPs rs755636251, rs544843047, rs748231432, and rs763859166, which can cause the opposite alterations in the expression of the corresponding genes (Table 1). Using our primary keyword search (hereinafter: see Methods, Additional file 2: Figure S1. two dashed-line boxes, Additional file 2), we found that both HSD17B1 overexpression and deficiency can elevate the risk of breast cancer [68], whereas GSTM3 deficiency can reduce these risks in people who never drink alcohol [69] (Table 1). In addition, Searles Nielsen and colleagues [66] suggested that another mechanism of GSTM3 overexpression can reduce the risk of a brain tumor in some children, as can rs748231432 and rs763859166 according to our results shown in Table 1.

Finally, using our secondary keyword search, we found eight retrospective clinical reviews [7076]. The most interesting among them, in our opinion, is a report on a nontrivial balance between reproductive potential and the risk of cancers of reproductive organs [70]. It is interesting that only one SNP marker (rs605059; protein-coding region, HSD17B1) of a positive correlation between the lifespan and number of children in women is known so far [71]. It is also noteworthy that one of current theories is that aging is a stepwise reduction in reproductive potential of individuals where one of these steps is under the control of the luteinizing hormone, whose suppression by smoking can reduce the risk of Alzheimer’s disease [9].

The human IL1B, CYP2A6, CYP2B6, and DHFR genes encode interleukin 1β, xenobiotic monooxygenase, 1,4-cineole 2-exo-monooxygenase, and dihydrofolate reductase, respectively. Their promoters contain the known SNP markers (rs1143627 [7785], rs28399433 [86, 87]) of nonreproductive organ cancer, as well as SNP markers (rs34223104 [88] and rs10168 [89]) of bioactivation and resistance to anticancer drugs, as shown in Table 2. Near these known SNP markers, we detected three unannotated SNPs, rs761592914, rs563558831, and rs750793297, which can alter expression levels of the same genes in the same manner (Table 2) and may be candidate SNP markers in this regard.

In addition, in the same gene regions, we found four other unannotated SNPs rs549858786, rs766799008, rs764508464, and rs754122321 that can have the opposite effect on the expression of the corresponding genes (Table 2). Using our primary keyword search, we found four articles [9093] similar to those that were in the case of the known SNPs, where we learned about the correlations between the intensity of physiological and clinical manifestations under study [8589] (Table 2). Finally, our secondary keyword search yielded 12 reviews [93105], among which, the most relevant for us was the notion that Helicobacter pylori infection can cause not only cancer of non-reproductive organs, but can directly reduce human reproductive potential in both men and women [101].

Looking through Tables 1, 2, and Additional file 3: Table S1, one can see that a person increases his/her lifespan and reproductive potential when this person reduces the encounters with cancer risk factors.

Known and candidate reproductivity-related SNP markers of metabolism

Human LEP, GCG, GH1, and INS genes encode hormones leptin, glucagon, somatotropin, and insulin, respectively. There are four known biomedical SNP markers: rs201381696 (obesity [54, 106]), rs183433761 (resistance to obesity during a high-fat diet [54]), rs11568827 (short stature [107]), and rs5505 (type 1 diabetes after neonatal diabetes mellitus [108]) as presented in Table 3.

Near these known SNP markers, 10 candidate SNP markers rs200487063, rs34104384, rs757035851, rs796237787, rs768454929, rs761695685, rs774326004, rs777003420, rs563207167, and rs11557611 were first predicted by our Web service [53] and, then, were characterized by our primary keyword search (Table 3). The most interesting among these predictions [109116], in our opinion, is the candidate SNP marker rs563207167 of neonatal macrosomia whose known clinical marker is hyperinsulinemia [115], which can be caused by the minor allele of this SNP according to our calculations (Table 3).

Finally, our secondary keyword search produced 31 original articles [105, 117146], e.g., showing that a maternal high-fat diet elevates the risk of hypertrophy in offspring via fetal hyperinsulinemia programmed epigenetically [141]. It is also relevant that bupropion used as an antidepressant against smoking in pregnancy can cause hyperinsulinemia in newborn children [142].

Human genes NOS2, STAR, APOA1, CETP, SOD1, TPI1, and GJA5 code for inducible nitric oxide synthase 2, steroidogenic acute regulatory protein, apolipoprotein A1, cholesteryl ester transfer protein, Cu/Zn superoxide dismutase, triosephosphate isomerase, and connexin 40, respectively. Their promoters contain eight known biomedical SNP markers shown in Table 4.

Around these known biomedical SNP markers, we found six unannotated SNPs rs544850971, rs17231520, rs569033466, rs757176551, rs781835924, and rs587745372, which can alter expression levels of the human genes containing them according to in silico predictions of our Web service [53] (Table 4). Next, we carried out our primary keyword search where [147165] the most interesting finding (in our opinion) is the clinical association between a SOD1 deficiency and asthenospermia [151], as one can see in Table 4. Finally, we performed our secondary keyword search, which yielded 21 literary sources [155175]. For instance, bisphenol A pollution in men can increase the risk of congenital heart morphogenesis disorders in their offspring as Lobmo and colleagues [174] have reported.

As readers can see in Tables 3, 4, and Additional file 3: Table S1, deviations from normal metabolism in parents (e.g., starvation, stress, dietary changes, and polluted environment) can epigenetically program pathologies of the development in their offspring (e.g., [141]). Therefore, a person can increase his/her reproductive potential and lifespan by keeping one’s metabolism normal.

Known and candidate reproductivity-related SNP markers related to blood

Human genes HBB, HBD, HBG2, ACKR1, MBL2, MMP12, and F2 encode subunits β, δ, and γ2 (fetal) of hemoglobin as well as glycoprotein D, mannan-binding lectin, macrophage elastase, and serine protease, respectively. Table 5 shows 10 known SNP markers (rs397509430, rs33980857, rs34598529, rs33931746, rs33981098, rs34500389, and rs35518301) of both malaria resistance and thalassemia [176] as well as rs2814778 (both malaria resistance and low white-blood-cell count [177, 178]), rs72661131 (variable immunodeficiency [179], preeclampsia [180], and stroke [181]), and rs2276109 (lower risks of psoriasis [182], systemic sclerosis [183], and asthma [184]).

Using our Web service [53], we found seven candidate SNP markers rs63750953, rs281864525, rs117785782, rs34166473, rs745580140, rs562962093, and rs572527200, which can alter expression of the human genes containing them, as is the case for the above SNP markers, which can affect the human reproductive potential [185, 186] (Table 5). In addition, using our primary keyword search, we identified three more candidate SNP markers: rs567653539 (reduced risks of recurrent vulvovaginal infections [187]), rs572527200 (high risk of ovarian hyper stimulation syndrome [188]), rs564528021, and rs752364393 (high risk of pre-eclampsia [189]). Finally, we performed our secondary keyword search, which yielded 22 reviews [162, 190210], the most important of which (in our opinion) mentions pre-eclampsia as a leading cause of maternal and fetal mortality and morbidity worldwide [162], as readers can see in Additional file 3: Table S1.

Human genes THBD, PROC, F8, F3, F7, F9, and F11 code for thrombomodulin, and blood coagulation factors XIV, 8, 3, 7, 9, and 11, respectively (Table 6). There are three known SNP markers rs13306848 (thrombosis [211]), rs563763767 (myocardial infarction and thrombosis [212]), and F7:-33a→c (moderate bleeding [213]) located within the promoters of these genes, which are listed in Table 6.

Within 90-bp proximal regions of these promoters, we selected 30 candidate SNP markers of tumor invasion [214], thrombosis, inflammation and pregnancy loss [215217], ovarian cancer [218], hemorrhage [219], angioneurotic edema [220], hemophilia B [221], and myocardial fibrosis [222] (Table 6). We predicted them using our Web service [53] and a primary keyword search, as described above in detail. Finally, our secondary keyword search produced 29 reviews [101, 223250]. The most interesting among them, in our opinion, is the fact that Homo sapiens is the longest-lived species among great apes (Hominidae) in the postreproductive period. Most often, this period in the life of a human is accompanied by various types of dementia and atherosclerosis, whereas cardiomyopathy and myocardial fibrosis predominate in great apes [248].

Looking through Tables 5, 6, and Additional file 3: Table S1, readers can see that by reducing the risk of blood diseases, a person can increase his/her lifespan and reproductive potential.

Candidate SNP markers of reproductivity-related genes

In addition, using a standard keyword search in the PubMed database, we found articles on human reproductive potential. On this basis, we selected a set of 22 human genes—AR, CAT, CLCA4, CYP1B1, CYP17A1, DAZ1, DAZ2, DAZ3, DAZ4, DEFB126, DNMT1, GNRH1, LHCGR, MTHFR, NR5A1, PARP1, PYGO2, SRD5A2, SRY, TACR3, TET1, and TSSK2—whose promoters do not contain known biomedical SNP markers. This gene set represents a wide variety of known reproductivity-related physiological markers, such as enzymes, transcription factors, hormones, and their receptors. Table 7 presents the results obtained using our Web service [53].

None of the SNPs can statistically significantly alter TBP’s affinity for the promoters of human genes CAT, CLCA4, CYP1B1, DAZ1, DAZ2, DAZ3, DAZ4, DEFB126, GNRH1, LHCGR, PARP1, PYGO2, SRD5A2, SRY, TACR3, TET1, and TSSK2 being analyzed (data not shown). Within promoters of five remaining genes (AR, MTHFR, DNMT1, CYP17A1, and NR5A1), in the same way, we found 24 candidate SNP markers (Table 7). Our primary keyword search associated them with androgenetic alopecia and androgen-induced premature senescence in adult men [251], preeclampsia [252], adverse pregnancy outcomes [253], epigenetic disorders of fetal/newborn brain development [254, 255], activation of protooncogenes in cancer [256], hyperandrogenism in polycystic ovary syndrome [257], fertility impairments [258], adrenal tumors and endometriosis [259] (Table 7).

As a cross-validation test, we unexpectedly found the ratio 5:19 of the candidate SNP markers in the reproductivity-related genes (Table 7) decreasing versus increasing TBP-promoter affinity. In contrast, the well-known whole-genome ratio 2:1 of SNPs reducing versus SNPs increasing affinity of the transcription factors for human gene promoters has been identified by two independent teams [260, 261]. According to binomial distribution, this difference between the candidate SNP markers in the reproductivity-related genes (Table 7) and all SNPs of the human genome is statistically significant (α < 0.000005). This statistical significance reflects the stronger pressure of natural selection against underexpression of the reproductivity-related genes. This unexpected finding indicates higher robustness of this specific sort of human genes on a whole-genome scale and is consistent with the commonly accepted meaning of the term “reproductive potential” as a mainstream concept in population ecology, which defines this term as a measure of evolutionary success of either human individuals [2] or populations [3]. This match between our predictions (Table 7) and one of the mainstream biomedical concepts [2, 3] support the plausibility of the candidate SNP markers predicted here.

Verification procedures for the selected candidate SNP markers predicted here

Different public Web services [2138, 53] have their advantages and disadvantages in eliminating unannotated neutral SNPs. To optimize such knowledge, a comparison between the results of these Web services and experimental data as an independent commonly accepted uniform platform seems to be a necessary step for prediction of candidate SNP markers in silico [15, 20, 59]. Keeping this in mind, we selected some of the 126 candidate SNP markers predicted here—rs563763767, rs33981098, rs35518301, rs1143627, rs72661131, rs1800202, and rs7277748—and measured equilibrium dissociation constant KD of TBP–DNA complexes using an electrophoretic mobility shift assay (EMSA) in vitro (see Methods). The results are shown in Fig. 2, for example, panels A and B present electropherograms and their graphical representation in the case of ancestral and minor alleles, respectively, of the candidate SNP marker rs33981098 within the human HBB gene promoter. Here, readers can see that this SNP reduces the TBP–DNA affinity in half: from 44 nM in the norm (wt) to 90 nM in pathology (mut); this finding supports our prediction, namely, the twofold decrease in the estimate of TBP–DNA affinity from 5 to 9 nM (Table 5). Overall, panel C shows the coordinate plane of the predicted (axis X) and the measured (axis Y) ratio of KD;MUT/KD;WT values of minor versus ancestral alleles of each SNP being verified. As one can see in this figure, there is a significant correlation between our predictions in silico and our measurements in vitro in four statistical tests, namely: linear correlation (r), Spearman’s rank correlation (R), Kendall’s rank correlation (τ), and Goodman–Kruskal generalized correlation (γ) test, which confirm one another’s results. Therefore, the correlations between our predictions and experimental data are robust in terms of the variation of statistical criteria that supports the candidate reproductive-potential-related SNP markers predicted here.

Fig. 2.

Fig. 2

Experimental verification of the selected candidate SNP markers by an electrophoretic mobility shift assay (EMSA) in vitro. Legend: a and b Examples of electropherograms in the case of ancestral (panel A: norm, wild-type, wt) and minor (panel b: minor) alleles of the candidate SNP marker rs33981098 within the human HBB gene promoter and the corresponding diagrams of experimental values. c The significant correlations between the ratio of KD values of the equilibrium dissociation constant of the TBP–ODN complex, which were either measured in vitro (Y-axis) or in silico predicted (X-axis). Solid and dashed lines or curves denote the linear regression and boundaries of its 95% confidence interval, calculated using software Statistica (StatsoftTM, USA). Circles denote the ancestral and minor alleles of the candidate SNP markers rs563763767, rs33981098, rs35518301, rs1143627, rs72661131, rs1800202, and rs7277748 being verified; r, R, τ, γ, and α are linear correlation, Spearman’s rank correlation, Kendall’s rank correlation, Goodman–Kruskal generalized correlation, and their significance, respectively.

Besides the conventional EMSA, we used two modern high-performance methods. Figure 3 shows the results of high-resolution spectrometry on SX.20 (Applied Photophysics, UK), where a stopped-flow fluorescence assay in vitro in real-time mode was applied to the selected candidate SNP marker rs1800202 (see Methods). As readers can see in Table 4, we predicted in silico that the KD value of TBP’s binding affinity for this gene’s wild-type promoter (ancestral alleles), 1 nM, can be weakened by the minor allele of this SNP to 4 nM, in agreement with the experimental data: 1 versus 6 nM, respectively (Table 4). This is one more argument in favor of the significance of the candidate reproductive-potential-related SNP markers predicted here.

Fig. 3.

Fig. 3

The kinetics of binding to and bending of the ODN corresponding to the selected SNP marker rs1800202. Legend: a The ancestral allele, ODN 5′-ctcTATATAAgtggg-3′. b The minor allele, ODN 5′-ctcTATAgAAgtggg-3′. ODN concentration was 0.1 μM. TBP concentration was between 0.1 and 1.0 μM as indicated near the corresponding curve of the time series. KD values, a 1 nM and b 6 nM, were obtained as the output of the Dynafit software (Biokin, USA) when we used the corresponding time-series data as input for this software

Finally, we conducted transfection of the human cell line hTERT-BJ1 (human fibroblasts) in culture, using the pGL 4.10 vector carrying a reporter LUC gene whose transcription is initiated by either ancestral or minor alleles of the selected candidate SNP marker rs28399433 of the human CYP2A6 promoter (Table 2). The results are depicted in Fig. 4. As shown in Table 2, the low affinity of TBP for the minor allele of this SNP relative to the norm (ancestral allele) is consistent with the ex vivo underexpression of a reporter LUC gene carrying the minor allele of this SNP within the pGL 4.10 vector. This ex vivo observation independently confirms our prediction that this SNP can reduce the affinity of TBP for the promoter of the human CYP2A6 gene (Table 2).

Fig. 4.

Fig. 4

Cell culture verification of the selected candidate SNP marker rs28399433 in cell line hTERT-BJ1 (human fibroblasts) transfected with the pGL 4.10 vector carrying a reporter LUC gene. Legend: Dark gray bar, the original vector pGL 4.10 (Promega, USA) without any insertions, which served as an independent control; open bars, ancestral allele (wild type, WT); light gray bar, minor allele (rs28399433). The height of the bars and their error bars correspond to the mean estimates and boundaries of their 95% confidence intervals calculated from five independent experiments. All differences are statistically significant at the confidence level of α < 0.05

Thus, three independent experiments indicate that the candidate reproductive-potential-related SNP markers predicted here using our Web-service [53] seem to have statistically significant effects and are not neutral.

Discussion

In this work, we limited our research to SNPs altering TBP’s affinity for human gene promoters (according to predictions made by our Web service [53]) and thereby altering the expression of these genes; this is because the TBP-binding site is the best-studied transcription-regulatory element [47]. Using our Web service [53], we analyzed over 1000 SNPs between nucleotide positions -70 and -20 upstream of more than 50 protein-coding regions documented in the Ensembl database [11] and found only 126 candidate reproductive-potential-related SNP markers (Tables 1,2, 3, 4, 5, 6 and 7). This 8-fold reduction in the number of possible SNPs can make the clinical cohort-based search for such biomedical SNP markers faster, cheaper, and more targeted, indeed.

For clinical verification of the candidate SNP markers predicted here, we heuristically set up their prioritization based on Fisher’s Z-tests as rank ρ-values from the “best” (A) to the “worst” (E) in alphabetical order (Tables 1, 2, 3, 4, 5, 6 and 7). With this in mind, our findings do not mean that all the eliminated SNPs (data not shown) cannot be considered candidate reproductive-potential-related SNP markers. This is because they may alter transcription factor-binding sites without disrupting the TBP-binding site (e.g., rs11568827, rs796237787, and rs16887226). To perform this sort of analysis for any of them, there are many public Web services [2138] whose research capabilities may be enhanced when they are used in combination with our Web service [53].

It is also worth mentioning that 126 candidate SNP markers predicted here are whole-genome landmarks indicative of either elevated or reduced reproductive potential relative to the norm and can be expected to be present in patients as minor alleles of these SNPs [20]. For example, 10 candidate SNP markers of thrombosis (rs563763767, rs781855957, rs13306848, rs568801899, rs779755900, rs749456955, rs746842194, rs754815577, rs768753666, rs774688955) cause overproduction of coagulation inducers (Table 6). In pregnant women, Hughes syndrome provokes thrombosis with a fatal outcome, although this syndrome can be diagnosed and cured even at the earliest stages of its development [230232] (Additional file 3: Table S1). Thus, in women carrying any of the above SNPs, preventive treatment of this syndrome [230232] before a planned pregnancy can reduce the risk of death. Table 6 shows that seven SNPs (rs563763767, rs779755900, rs749456955, rs746842194, rs754815577, rs768753666, rs774688955) among the 10 mentioned above elevate the risk of myocardial infarction. Hence, a woman with some of these SNPs can improve her longevity by bringing her lifestyle in line with the knowledge that the risk of myocardial infarction elevates with total number of pregnancies, the age of the mother, as well as in pregnancy under the age of 20, in multiple pregnancies, in menstrual cycle irregularity, hypertension, preeclampsia, and in women smokers [233236] (Additional file 3: Table S1).

Finally, during our keyword search in the PubMed database, we encountered a large variety of research articles, clinical cases, laboratory data, retrospective reviews, and empirical findings—on human reproductive potential in various life situations—from sociologists, geneticists, legal scholars, clinicians, bioinformaticians, pharmacists, psychologists, pedagogues, physiologists, economists, and other relevant experts such as specialists on management, insurance, environmental protection, health care, and law enforcement (Tables 1, 2, 3, 4, 5, 6 and 7, and Additional file 3: Table S1). This observation means that this vital knowledge is very much in demand for the general population, but it is too scattered for practice use. As one can see in Tables 1, 2, 3, 4, 5, 6 and 7 and Additional file 3: Table S1, 126 candidate reproductive-potential-related SNP markers predicted here may serve as valid whole-genome landmarks near which the above authors can organize their main research on how the evolutionary success of an individual [2] or a population [3] could be enhanced. Consequently, the results of these studies can be directly addressed to people who would like to change their lifestyle in view of the possible risks of diseases. This approach becomes possible within the framework of predictive-preventive personalized medicine based on the sequenced individual genomes.

Conclusions

In keeping with Bowles’ lifespan theory [9], a large body of useful literature can be packaged into readable portions relevant to candidate reproductive-potential-related SNP markers for people who would like to reduce the risks of diseases corresponding to known alleles in own sequenced genome. After clinical validation, these candidate SNP markers may become useful for physicians (to improve treatment of patients) and for the general population (lifestyle choices improving longevity).

Methods

DNA sequences

We analyzed SNPs retrieved from the dbSNP database, v.147 [6] between nucleotide positions -70 and -20 upstream of the protein-coding regions documented by the Ensembl database [11] using the public Web service “UCSC Genome Browser” [12] as shown in Fig. 1a.

Synthetic double-helical deoxyoligonucleotides (ODNs)

The ODNs identical to ancestral and minor alleles of the selected SNPs— rs563763767, rs33981098, rs35518301, rs1143627, rs72661131, rs1800202, and rs7277748—were synthesized and purified (BIOSYN, Novosibirsk, Russia).

Preparation and purification of recombinant full-length human TBP

Recombinant human TBP (full-length native amino acid sequence) was expressed in Escherichia coli BL21 (DE3) cells transformed with the pAR3038-TBP plasmid (a generous gift from Prof. B. Pugh, Pennsylvania State University) as described elsewhere [262] with two modifications: the IPTG concentration was 1.0 instead of 0.1 mM, and the induction time was 3 instead of 1.5 h (for more details, see [263]).

EMSA

The above ODNs were labeled with 32P on both strands by means of T4 polynucleotide kinase (SibEnzyme, Novosibirsk) with subsequent annealing by heating to 95°C (at equimolar concentrations) and slow cooling (no less than 3 h) to room temperature. Equilibrium dissociation constants (KD) for each TBP–ODN complex were measured using a conventional protocol [263] including titration of a fixed amount of the above-mentioned recombinant TBP, 0.3 nM, with the increasing concentrations of each ODN to reach an equilibrium, whose timing was determined independently for each ODN in advance. The binding experiments were conducted at 25°C in a buffer consisting of 20 mM HEPES-KOH pH 7.6, 5 mM MgCl2, 70 mM KCl, 1 mM EDTA, 100 μg/ml BSA, 0.01% of NP-40, and 5% of glycerol. The ТВР–ODN complexes were separated from the unbound ODN using an EMSA, and their abundance levels were measured. The results of these measurements were input into conventional software OriginPro 8, whose output was a KD value expressed in nanomoles per liter, nM.

Stopped-flow fluorescence measurements

The ODNs identical to both ancestral and minor alleles of the selected SNP rs1800202, (i.e., 5′-ctcTATATAAgtggg-3′ and 5′-ctcTATAgAAgtggg-3′, respectively) were labeled at their 5′-termini with fluorescent dyes TAMRA and FAM (BIOSYN, Novosibirsk, Russia). Combining a fixed concentration (0.1 μM) of ODNs with various concentrations (0.1, 0.2, 0.4, 0.6, 0.8, or 1.0 μM) of the above TBP, we analyzed six time-series of the fluorescence expressed in conventional units using high-resolution spectrometer SX.20 (Applied Photophysics, UK). The results of these measurements served as input into the Dynafit software (Biokin, USA), whose output was the above KD values (for more details, see [264]).

Cell culture, transfection, and reporter assays

Cell line hTERT-BJ1 (human fibroblasts) was cultivated in a complete medium consisting of Dulbecco’s modified Eagle’s medium/Nutrient mixture F-12 Ham, supplemented with 10% (v/v) of fetal bovine serum (Sigma), penicillin (100 U/mL), and streptomycin (100 μg/mL; BioloT). The culture was maintained at 37°C in a humidified atmosphere containing 5% of CO2 until the desired degree of confluence. The proximal core promoter (177 bp long) containing either the ancestral allele or minor allele of the selected candidate SNP marker rs28399433 (5′-tcaggcagTATAAAggcaaac-3′ or 5′- tcaggcagTAgAAAggcaaac-3′, respectively) was cloned into the pGL 4.10 vector (Promega, USA) and cotransfected with pRL-TK using Screen Fect A (InCella) as described elsewhere [265]. Next, the cells were cultured in 6-well plates for 24 h. Luciferase activity was determined using the Dual-Luciferase Reporter Assay Kit (Promega, USA) All the experiments were conducted five times independently at 80–85% confluence.

DNA sequence analysis in silico

We analyzed DNA sequences between nucleotide positions -70 and -20 upstream of the protein-coding regions in the human genes retrieved from the human reference genome using the standard BioPerl library [266] via our Web service [53] in the case of ancestral alleles of SNPs under study, as described in Fig. 1b. In the case of minor alleles of these SNPs, we created sequences by hand using the above DNA sequences according to the description of these alleles from database dbSNP [6] as described in Fig. 1c. Next, clicking on the “Calculate” button (Fig. 1b, and c), we computed the maximal –ln(KD) value and its standard deviation ± ε of the affinity of TBP for the [–70; -20] region (where all the known sites are located) for both ancestral and minor alleles of the human gene promoter being analyzed. On this basis, using a package R [267], our Web service [54] made its statistical decision whether the analyzed SNP can alter the expression of the human gene under study as described in Additional file 1 [268274]. Earlier, we tested these estimates using independent data from more than a hundred our own experiments [275285] and the experiments of other authors (for review, see [51]). Finally, as soon as we predicted either SNP-caused significant overexpression or SNP-driven significant underexpression of the human genes being analyzed (as clinically relevant physiological markers), we conducted a manual two-step keyword search in NCBI databases [286] as described in detail in Additional file 2 [287].

Additional files

Additional file 1: (220.2KB, pdf)

Supplementary method. A sequence-based statistical estimate of the SNP-caused alteration in the affinity of TATA box binding protein (TBP) for the human gene promoter containing this SNP within its region [-70; -20]. (PDF 220 kb)

Additional file 2: (222KB, pdf)

Supplementary method. Keyword search in the PubMed database. (PDF 221 kb)

Additional file 3: (198.4KB, pdf)

Table S1. Clinically known dependences between reproductive potential and hereditary diseases whose SNP markers were predicted in this work. (PDF 198 kb)

Acknowledgments

We are grateful to Shevchuk Editing (Brooklyn, NY, USA; URL: http://www.shevchuk-editing.com) for English editing.

Funding

The publication costs for this article were funded by grant #14.B25.31.0033 from the government of the Russian Federation, Resolution No. 220 (to Professor E.I. Rogaev as principal investigator, PI). The data compilation was supported by Russian Ministry of Science and Education under 5-100 Excellence Programme (for IVC and MPP). The experiment ex vivo was supported by project #0324-2016-0002 (for EVK, LVO, and AVO), the experiments in vitro were supported by project #0324-2016-0003 (for LKS, IAD, EBS, OVA, and DAZ), the software development was financed by projects #0324-2016-0008 (for DAR and NAK).

Availability of data and materials

Web service SNP_TATA_Comparator is public available (URL=http://beehive.bionet.nsc.ru/cgi-bin/mgs/tatascan/start.pl).

About this supplement

This article has been published as part of BMC Genomics Volume 19 Supplement 3, 2018: Selected articles from Belyaev Conference 2017: genomics. The full contents of the supplement are available online at https://bmcgenomics.biomedcentral.com/articles/supplements/volume-19-supplement-3.

Abbreviations

ACKR1

atypical chemokine receptor 1

APOA1

apolipoprotein A1

AR

androgen receptor

CAT

catalase

CETP

cholesteryl ester transfer protein

CLCA4

chloride channel accessory 4

CYP17A1

cytochrome p450 family 17 subfamily A member 1

CYP1B1

cytochrome P450 family 1 subfamily B member 1

CYP2A6

cytochrome P450 family 2 subfamily A member 6

CYP2B6

cytochrome P450 family 2 subfamily B Member 6

DAZ1 (2, 3, 4)

deleted in azoospermia 1 (2, 3, 4, respectively)

DEFB126

defensin β 126

DHFR

dihydrofolate reductase

DNMT1

DNA methyltransferase 1

EMSA

electrophoretic mobility shift assay

ESR2

estrogen receptor 2

F2 (3, 7, 8, 9, 11)

coagulation factor II (III, VII, VIII, IX, XI, respectively)

GCG

glucagon

GH1

growth hormone 1

GJA5

gap junction protein α5

GNRH1

gonadotropin releasing hormone 1

GSTM3

glutathione S-transferase μ3

HBB

hemoglobin subunit β

HBD

hemoglobin subunit δ

HBG2

hemoglobin subunit γ2

HSD17B1

hydroxysteroid 17-β dehydrogenase 1

IL1B

interleukin 1 β

INS

insulin

Kd

equilibrium dissociation constant

LEP

leptin

LHCGR

luteinizing hormone (choriogonadotropin receptor)

Ln

natural logarithm

MBL2

mannose binding lectin 2

MMP12

matrix metallopeptidase 12

MTHFR

methylenetetrahydrofolate reductase

Mut

minor allele of SNPs. Genes

NOS2

nitric oxide synthase 2

NR5A1

nuclear receptor subfamily 5 group A member 1

PARP1

poly(ADP-ribose) polymerase 1

PGR

progesterone receptor

PROC

protein C (inactivator of coagulation factors Va and VIIIa)

PYGO2

pygopus family PHD finger 2

SNP

single nucleotide polymorphism

SOD1

superoxide dismutase 1

SRD5A2

steroid 5 α-reductase 2

SRY

sex determining region Y

STAR

steroidogenic acute regulatory protein

TACR3

tachykinin receptor 3

TBP

TATA-binding protein

TET1

Tet methylcytosine dioxygenase 1

TF

transcription factor

THBD

thrombomodulin

TPI1

triosephosphate isomerase 1

TSS

transcription start site

TSSK2

testis specific serine kinase 2

WT

wild type (norm)

Authors’ contributions

NAK conceived of and supervised the study. LKS, IAD, EBS, OVA, and DAZ conducted the in vitro experiments. EVK conducted the ex vivo experiments. PMP and DAR designed, developed, maintained, adapted, and tuned the software for sequence analysis. IVC analyzed data in silico. LVO and AVO interpreted the computer-based predictions in comparison with experimental data. MPP wrote the manuscript. All the coauthors read and approved the final version of the manuscript.

Ethics approval and consent to participate

Not applicable

Consent for publication

Not applicable

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Footnotes

Electronic supplementary material

The online version of this article (10.1186/s12864-018-4478-3) contains supplementary material, which is available to authorized users.

References

  • 1.Axelsson J, Bonde JP, Giwercman YL, Rylander L, Giwercman A. Gene-environment interaction and male reproductive function. Asian J Androl. 2010;12:298–307. doi: 10.1038/aja.2010.16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Pianka ER. Natural selection of optimal reproductive tactics. Amer Zool. 1976;16:775–784. doi: 10.1093/icb/16.4.775. [DOI] [Google Scholar]
  • 3.Chapman RN. Animal ecology with special reference to insects. NY: McGraw-Hill Book Co; 1931. [Google Scholar]
  • 4.Chapman RN. The quantitative analysis of environmental factors. Ecology. 1928;9:111–122. doi: 10.2307/1929348. [DOI] [Google Scholar]
  • 5.Pianka ER. On r and К selection. Amer Natur. 1970;104:592–597. doi: 10.1086/282697. [DOI] [Google Scholar]
  • 6.Sherry ST, Ward MH, Kholodov M, Baker J, Phan L, Smigielski EM, et al. dbSNP: the NCBI database of genetic variation. Nucleic Acids Res. 2001;29:308–311. doi: 10.1093/nar/29.1.308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Deplancke B, Alpern D, Gardeux V. The genetics of transcription factor DNA binding variation. Cell. 2016;166:538–554. doi: 10.1016/j.cell.2016.07.012. [DOI] [PubMed] [Google Scholar]
  • 8.Ortega VE, Meyers DA. Pharmacogenetics: implications of race and ethnicity on defining genetic profiles for personalized medicine. J Allergy Clin Immunol. 2014;133:16–26. doi: 10.1016/j.jaci.2013.10.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Bowles JT. The evolution of aging: a new approach to an old problem of biology. Med Hypotheses. 1998;51:179–221. doi: 10.1016/S0306-9877(98)90079-2. [DOI] [PubMed] [Google Scholar]
  • 10.Telenti A, Pierce LC, Biggs WH, di Iulio J, Wong EH, Fabani MM, et al. Deep sequencing of 10,000 human genomes. Proc Natl Acad Sci U S A. 2016;113:11901–11906. doi: 10.1073/pnas.1613365113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Zerbino DR, Wilder SP, Johnson N, Juettemann T, Flicek PR. The Ensembl regulatory build. Genome Biol. 2015;16:56. doi: 10.1186/s13059-015-0621-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Haeussler M, Raney BJ, Hinrichs AS, Clawson H, Zweig AS, Karolchik D, et al. Navigating protected genomics data with UCSC Genome Browser in a Box. Bioinformatics. 2015;31:764–766. doi: 10.1093/bioinformatics/btu712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Wu J, Wu M, Li L, Liu Z, Zeng W, Jiang R. dbWGFP: a database and web server of human whole-genome single nucleotide variants and their functional predictions. Database (Oxford) 2016;2016:baw024. doi: 10.1093/database/baw024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Trovato GM. Sustainable medical research by effective and comprehensive medical skills: overcoming the frontiers by predictive, preventive and personalized medicine. EPMA J. 2014;5:14. doi: 10.1186/1878-5085-5-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Yoo SS, Jin C, Jung DK, Choi YY, Choi JE, Lee WK, et al. Putative functional variants of XRCC1 identified by RegulomeDB were not associated with lung cancer risk in a Korean population. Cancer Genet. 2015;208:19–24. doi: 10.1016/j.cancergen.2014.11.004. [DOI] [PubMed] [Google Scholar]
  • 16.Haldane JBS. The Cost of Natural Selection. J Genet. 1957;55:511–524. doi: 10.1007/BF02984069. [DOI] [Google Scholar]
  • 17.Kimura M. Evolutionary rate at the molecular level. Nature. 1968;217:624–626. doi: 10.1038/217624a0. [DOI] [PubMed] [Google Scholar]
  • 18.Varzari A, Bruch K, Deyneko IV, Chan A, Epplen JT, Hoffjan S. Analysis of polymorphisms in RIG-I-like receptor genes in German multiple sclerosis patients. J Neuroimmunol. 2014;277:140–144. doi: 10.1016/j.jneuroim.2014.09.015. [DOI] [PubMed] [Google Scholar]
  • 19.Varzari A, Deyneko IV, Tudor E, Turcan S. Polymorphisms of glutathione S-transferase and methylenetetrahydrofolate reductase genes in Moldavian patients with ulcerative colitis: Genotype-phenotype correlation. Meta Gene. 2016;7:76–82. doi: 10.1016/j.mgene.2015.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Ponomarenko M, Rasskazov D, Chadaeva I, Sharypova E, Ponomarenko P, Arkova O, et al. SNP_TATA_Comparator: genomewide landmarks for preventive personalized medicine. Front Biosci (Schol Ed) 2017;9:276–306. doi: 10.2741/s488. [DOI] [PubMed] [Google Scholar]
  • 21.Johnson AD, Handsaker RE, Pulit SL, Nizzari MM, O’Donnell CJ, de Bakker PI. SNAP: a web-based tool for identification and annotation of proxy SNPs using HapMap. Bioinformatics. 2008;24:2938–2939. doi: 10.1093/bioinformatics/btn564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Barenboim M, Manke T. ChroMoS: an integrated web tool for SNP classification, prioritization and functional interpretation. Bioinformatics. 2013;29:2197–2198. doi: 10.1093/bioinformatics/btt356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Riva A. Large-scale computational identification of regulatory SNPs with rSNP-MAPPER. BMC Genomics. 2012;13:S7. doi: 10.1186/1471-2164-13-S4-S7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Macintyre G, Bailey J, Haviv I, Kowalczyk A. is-rSNP: a novel technique for in silico regulatory SNP detection. Bioinformatics. 2010;26:i524–i530. doi: 10.1093/bioinformatics/btq378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Ponomarenko JV, Furman DP, Frolov AS, Podkolodny NL, Orlova GV, Ponomarenko MP, et al. ACTIVITY: a database on DNA/RNA sites activity adapted to apply sequence-activity relationships from one system to another. Nucleic Acids Res. 2001;29:284–287. doi: 10.1093/nar/29.1.284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Chen CC, Xiao S, Xie D, Cao X, Song CX, Wang T, et al. Understanding variation in transcription factor binding by modeling transcription factor genome-epigenome interactions. PLoS Comput Biol. 2013;9:e1003367. doi: 10.1371/journal.pcbi.1003367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Deyneko IV, Bredohl B, Wesely D, Kalybaeva YM, Kel AE, Blocker H, et al. FeatureScan: revealing property-dependent similarity of nucleotide sequences. Nucleic Acids Res. 2006;34:W591–W595. doi: 10.1093/nar/gkl337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Chen CY, Chang IS, Hsiung CA, Wasserman WW. On the identification of potential regulatory variants within genome wide association candidate SNP sets. BMC Med Genomics. 2014;7:34. doi: 10.1186/1755-8794-7-34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Rasskazov DA, Antontseva EV, Bryzgalov LO, Matveeva MY, Kashina EV, Ponomarenko PM, et al. rSNP_Guide-based evaluation of SNPs in promoters of the human APC and MLH1 genes associated with colon cancer. Russ J Genet App Res. 2014;4:245–253. doi: 10.1134/S2079059714040108. [DOI] [Google Scholar]
  • 30.Boyle AP, Hong EL, Hariharan M, Cheng Y, Schaub MA, Kasowski M, et al. Annotation of functional variation in personal genomes using RegulomeDB. Genome Res. 2012;22:1790–1797. doi: 10.1101/gr.137323.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Andersen MC, Engstrom PG, Lithwick S, Arenillas D, Eriksson P, Lenhard B, et al. In silico detection of sequence variations modifying transcriptional regulation. PLoS Comput Biol. 2008;4:e5. doi: 10.1371/journal.pcbi.0040005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Ponomarenko JV, Orlova GV, Frolov AS, Gelfand MS, Ponomarenko MP. SELEX_DB: a database on in vitro selected oligomers adapted for recognizing natural sites and for analyzing both SNPs and site-directed mutagenesis data. Nucleic Acids Res. 2002;30:195–199. doi: 10.1093/nar/30.1.195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Saccone SF, Bolze R, Thomas P, Quan J, Mehta G, Deelman E, et al. SPOT: a web-based tool for using biological databases to prioritize SNPs after a genome-wide association study. Nucleic Acids Res. 2010;38:W201–W209. doi: 10.1093/nar/gkq513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Podkolodnyy NL, Afonnikov DA, YuYu V, Bryzgalov LO, Ivanisenko VA, Demenkov PS, et al. Program complex SNP-MED for analysis of single-nucleotide polymorphism (SNP) effects on the function of genes associated with socially significant diseases. Russ J Genet App Res. 2014;4:159–167. doi: 10.1134/S2079059714030034. [DOI] [Google Scholar]
  • 35.Fu Y, Liu Z, Lou S, Bedford J, Mu XJ, Yip KY, et al. FunSeq2: a framework for prioritizing noncoding regulatory variants in cancer. Genome Biol. 2014;15:480. doi: 10.1186/s13059-014-0480-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Deyneko IV, Kalybaeva YM, Kel AE, Blocker H. Human-chimpanzee promoter comparisons: property-conserved evolution? Genomics. 2010;96:129–133. doi: 10.1016/j.ygeno.2010.06.003. [DOI] [PubMed] [Google Scholar]
  • 37.Coetzee SG, Rhie SK, Berman BP, Coetzee GA, Noushmehr H. FunciSNP: an R/bioconductor tool integrating functional non-coding data sets with genetic association studies to identify candidate regulatory SNPs. Nucleic Acids Res. 2012;40:e139. doi: 10.1093/nar/gks542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Bryzgalov LO, Antontseva EV, Matveeva MY, Shilov AG, Kashina EV, Mordvinov VA, et al. Detection of regulatory SNPs in human genome using ChIP-seq ENCODE data. PLoS One. 2013;8:e78833. doi: 10.1371/journal.pone.0078833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Ni Y, Hall AW, Battenhouse A, Iyer VR. Simultaneous SNP identification and assessment of allele-specific bias from ChIP-seq data. BMC Genet. 2012;13:46. doi: 10.1186/1471-2156-13-46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Hein M, Graver S. Tumor cell response to bevacizumab single agent therapy in vitro. Cancer Cell Int. 2013;13:94. doi: 10.1186/1475-2867-13-94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Hu J, Locasale JW, Bielas JH, O'Sullivan J, Sheahan K, Cantley LC, et al. Heterogeneity of tumor-induced gene expression changes in the human metabolic network. Nat Biotechnol. 2013;31:522–529. doi: 10.1038/nbt.2530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Leschner S, Deyneko IV, Lienenklaus S, Wolf K, Bloecker H, Bumann D, et al. Identification of tumor-specific Salmonella Typhimurium promoters and their regulatory logic. Nucleic Acids Res. 2012;40:2984–2994. doi: 10.1093/nar/gkr1041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Ponomarenko MP, Ponomarenko JV, Frolov AS, Podkolodnaya OA, Vorobyev DG, Kolchanov NA, et al. Oligonucleotide frequency matrices addressed to recognizing functional DNA sites. Bioinformatics. 1999;15:631–643. doi: 10.1093/bioinformatics/15.7.631. [DOI] [PubMed] [Google Scholar]
  • 44.Amberger J, Bocchini CA, Scott AF, Hamosh A. McKusick's Online Mendelian Inheritance in Man (OMIM) Nucleic Acids Res. 2009;37:D793–D796. doi: 10.1093/nar/gkn665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Mitsuyasu H, Izuhara K, Mao XQ, Gao PS, Arinobu Y, Enomoto T, et al. Ile50Val variant of IL4R-alpha upregulates IgE synthesis and associates with atopic asthma. Nat Genet. 1998;19:119–120. doi: 10.1038/472. [DOI] [PubMed] [Google Scholar]
  • 46.Savinkova LK, Ponomarenko MP, Ponomarenko PM, Drachkova IA, Lysova MV, Arshinova TV, et al. TATA box polymorphisms in human gene promoters and associated hereditary pathologies. Biochemistry (Mosc) 2009;74:117–129. doi: 10.1134/S0006297909020011. [DOI] [PubMed] [Google Scholar]
  • 47.Ponomarenko M, Mironova V, Gunbin K, Savinkova L. Hogness Box. In: Maloy S, Hughes K, editors. Brenner's Encyclopedia of Genetics. vol. 3. 2nd ed. San Diego: Academic Press, Elsevier Inc. 2013. p. 491–4
  • 48.Martianov I, Viville S, Davidson I. RNA polymerase II transcription in murine cells lacking the TATA binding protein. Science. 2002;298:1036–1039. doi: 10.1126/science.1076327. [DOI] [PubMed] [Google Scholar]
  • 49.Muller F, Lakatos L, Dantonel J, Strahle U, Tora L. TBP is not universally required for zygotic RNA polymerase II transcription in zebrafish. Curr Biol. 2001;11:282–287. doi: 10.1016/S0960-9822(01)00076-8. [DOI] [PubMed] [Google Scholar]
  • 50.Mogno I, Vallania F, Mitra RD, Cohen BA. TATA is a modular component of synthetic promoters. Genome Res. 2010;20:1391–1397. doi: 10.1101/gr.106732.110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Ponomarenko PM, Suslov VV, Savinkova LK, Ponomarenko MP, Kolchanov NA. A precise equation of equilibrium of four steps of TBP binding with the TATA box for prognosis of phenotypic manifestation of mutations. Biofizika. 2010;55:400–414. [PubMed] [Google Scholar]
  • 52.Lifton R, Goldberg M, Karp R, Hogness D. The organization of the histone genes in Drosophila melanogaster: Functional and evolutionary implications. Cold Spring Harb Symp Quant Biol. 1978;42:1047–1051. doi: 10.1101/SQB.1978.042.01.105. [DOI] [PubMed] [Google Scholar]
  • 53.Ponomarenko M, Rasskazov D, Arkova O, Ponomarenko P, Suslov V, Savinkova L, et al. How to use SNP_TATA_Comparator to find a significant change in gene expression caused by the regulatory SNP of this gene’s promoter via a change in affinity of the TATA-binding protein for this promoter. Biomed Res Int. 2015;2015:359835. doi: 10.1155/2015/359835. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Arkova OV, Ponomarenko MP, Rasskazov DA, Drachkova IA, Arshinova TV, Ponomarenko PM, et al. Obesity-related known and candidate SNP markers can significantly change affinity of TATA-binding protein for human gene promoters. BMC Genomics. 2015;16:S5. doi: 10.1186/1471-2164-16-S13-S5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Ponomarenko MP, Arkova O, Rasskazov D, Ponomarenko P, Savinkova L, Kolchanov N. Candidate SNP markers of gender-biased autoimmune complications of monogenic diseases are predicted by a significant change in the affinity of TATA-binding protein for human gene promoters. Front Immunol. 2016;7:130. doi: 10.3389/fimmu.2016.00130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Ponomarenko P, Rasskazov D, Suslov V, Sharypova E, Savinkova L, Podkolodnaya O, et al. Candidate SNP markers of chronopathologies are predicted by a significant change in the affinity of TATA-binding protein for human gene promoters. Biomed Res Int. 2016;2016:8642703. doi: 10.1155/2016/8642703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Chadaeva IV, Ponomarenko MP, Rasskazov DA, Sharypova EB, Kashina EV, Matveeva MY, et al. Candidate SNP markers of aggressiveness-related complications and comorbidities of genetic diseases are predicted by a significant change in the affinity of TATA-binding protein for human gene promoters. BMC Genomics. 2016;17:995. doi: 10.1186/s12864-016-3353-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Chadaeva I, Rasskazov D, Sharypova E, Savinkova L, Ponomarenko P, Ponomarenko M. Candidate SNP markers of social dominance, which may affect the affinity of the TATA-binding protein for human gene promoters. Russ J Genet Appl Res. 2017;7:523–537. doi: 10.1134/S2079059717050045. [DOI] [Google Scholar]
  • 59.Ponomarenko P, Chadaeva I, Rasskazov D, Sharypova E, Kashina E, Drachkova I, et al. Candidate SNP markers of familial and sporadic Alzheimer’s diseases are predicted by a significant change in the affinity of TATA-binding protein for human gene promoters. Front Aging Neurosci. 2017;9:231. doi: 10.3389/fnagi.2017.00231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Turnaev II, Rasskazov DA, Arkova OV, Ponomarenko MP, Ponomarenko PM, Savinkova LK, et al. Hypothetical SNP markers that significantly affect the affinity of the TATA-binding protein to VEGFA, ERBB2, IGF1R, FLT1, KDR, and MET oncogene promoters as chemotherapy targets. Mol Biol (Mosk) 2016;50:161–173. doi: 10.1134/S0026893316010209. [DOI] [PubMed] [Google Scholar]
  • 61.Philips S, Richter A, Oesterreich S, Rae JM, Flockhart DA, Perumal NB, et al. Functional characterization of a genetic polymorphism in the promoter of the ESR2 gene. Horm Cancer. 2012;3:37–43. doi: 10.1007/s12672-011-0086-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Strumylaite L, Bogusevicius A, Ryselis S, Pranys D, Poskiene L, Kregzdyte R, et al. Association between cadmium and breast cancer. Medicina (Kaunas) 2008;44:415–420. [PubMed] [Google Scholar]
  • 63.Helewa M, Levesque P, Provencher D, Lea RH, Rosolowich V, Shapiro HM, et al. Breast cancer, pregnancy, and breastfeeding. J Obstet Gynaecol Can. 2002;24:164–180. doi: 10.1016/S1701-2163(16)30298-5. [DOI] [PubMed] [Google Scholar]
  • 64.Peltoketo H, Piao Y, Mannermaa A, Ponder BA, Isomaa V, Poutanen M, et al. A point mutation in the putative TATA box, detected in nondiseased individuals and patients with hereditary breast cancer, decreases promoter activity of the 17 β-hydroxysteroid dehydrogenase type 1 gene 2 (EDH17B2) in vitro. Genomics. 1994;23:250–252. doi: 10.1006/geno.1994.1487. [DOI] [PubMed] [Google Scholar]
  • 65.De Vivo I, Huggins GS, Hankinson SE, Lescault PJ, Boezen M, Colditz GA, et al. A functional polymorphism in the promoter of the progesterone receptor gene associated with endometrial cancer risk. Proc Natl Acad Sci USA. 2002;99:12263–12268. doi: 10.1073/pnas.192172299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Searles Nielsen S, Mueller BA, Preston-Martin S, Farin FM, Holly EA, et al. Childhood brain tumors and maternal cured meat consumption in pregnancy: differential effect by glutathione S-transferases. Cancer Epidemiol Biomarkers Prev. 2011;20:2413–2419. doi: 10.1158/1055-9965.EPI-11-0196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Tan X, Wang Y, Han Y, Chang W, Su T, Hou J, et al. Genetic variation in the GSTM3 promoter confer risk and prognosis of renal cell carcinoma by reducing gene expression. Br J Cancer. 2013;109:3105–3115. doi: 10.1038/bjc.2013.669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.He W, Gauri M, Li T, Wang R, Lin SX. Current knowledge of the multifunctional 17β-hydroxysteroid dehydrogenase type 1 (HSD17B1) Gene. 2016;588:54–61. doi: 10.1016/j.gene.2016.04.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Mitrunen K, Hirvonen A. Molecular epidemiology of sporadic breast cancer. The role of polymorphic genes involved in oestrogen biosynthesis and metabolism. Mutat Res. 2003;544:9–41. doi: 10.1016/S1383-5742(03)00016-4. [DOI] [PubMed] [Google Scholar]
  • 70.Jasienska G. Reproduction and lifespan: Trade-offs, overall energy budgets, intergenerational costs, and costs neglected by research. Am J Hum Biol. 2009;21:524–532. doi: 10.1002/ajhb.20931. [DOI] [PubMed] [Google Scholar]
  • 71.Scarabino D, Scacchi R, Pinto A, Corbo RM. Genetic basis of the relationship between reproduction and longevity: a study on common variants of three genes in steroid hormone metabolism--CYP17, HSD17B1, and COMT. Rejuvenation Res. 2015;18:464–472. doi: 10.1089/rej.2015.1665. [DOI] [PubMed] [Google Scholar]
  • 72.Kalogera E, Dowdy SC, Bakkum-Gamez JN. Preserving fertility in young patients with endometrial cancer: current perspectives. Int J Womens Health. 2014;6:691–701. doi: 10.2147/IJWH.S47232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Hambrecht A, Jandial R, Neman J. Emerging role of brain metastases in the prognosis of breast cancer patients. Breast Cancer (Dove Med Press) 2011;3:79–91. doi: 10.2147/BCTT.S19967. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Wright DD, Whitney J. Multiple hamartoma syndrome (Cowden's syndrome): case report and literature review. Gen Dent. 2006;54:417–419. [PubMed] [Google Scholar]
  • 75.Molokwu JC, Prizment AE, Folsom AR. Reproductive characteristics and risk of kidney cancer: Iowa Women's Health Study. Maturitas. 2007;58:156–163. doi: 10.1016/j.maturitas.2007.07.003. [DOI] [PubMed] [Google Scholar]
  • 76.Schnoller TJ, Jentzmik F, Al Ghazal A, Zengerling F, de Petriconi R, Hefty R, et al. Renal masses in pregnancy. Diagnostics and therapeutic management. Urologe A. 2011;50:1064–1067. doi: 10.1007/s00120-011-2685-5. [DOI] [PubMed] [Google Scholar]
  • 77.El-Omar EM, Carrington M, Chow WH, McColl KE, Bream JH, Young HA, et al. Interleukin-1 polymorphisms associated with increased risk of gastric cancer. Nature. 2000;404:398–402. doi: 10.1038/35006081. [DOI] [PubMed] [Google Scholar]
  • 78.Wang Y, Kato N, Hoshida Y, Yoshida H, Taniguchi H, Goto T, et al. Interleukin-1β gene polymorphisms associated with hepatocellular carcinoma in hepatitis C virus infection. Hepatology. 2003;37:65–71. doi: 10.1053/jhep.2003.50017. [DOI] [PubMed] [Google Scholar]
  • 79.Martinez-Carrillo DN, Garza-Gonzalez E, Betancourt-Linares R, Monico-Manzano T, Antunez-Rivera C, Roman-Roman A, et al. Association of IL1B -511C/-31T haplotype and Helicobacter pylori vacA genotypes with gastric ulcer and chronic gastritis. BMC Gastroenterol. 2010;10:126. doi: 10.1186/1471-230X-10-126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Wu KS, Zhou X, Zheng F, Xu XQ, Lin YH, Yang J. Influence of interleukin-1 β genetic polymorphism, smoking and alcohol drinking on the risk of non-small cell lung cancer. Clin Chim Acta. 2010;411:1441–1446. doi: 10.1016/j.cca.2010.05.035. [DOI] [PubMed] [Google Scholar]
  • 81.Borkowska P, Kucia K, Rzezniczek S, Paul-Samojedny M, Kowalczyk M, Owczarek A, et al. Interleukin-1β promoter (-31T/C and -511C/T) polymorphisms in major recurrent depression. J Mol Neurosci. 2011;44:12–16. doi: 10.1007/s12031-011-9507-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Strandberg L, Mellstrom D, Ljunggren O, Grundberg E, Karlsson MK, Holmberg AH, et al. IL6 and IL1B polymorphisms are associated with fat mass in older men: the MrOS Study Sweden. Obesity (Silver Spring) 2008;16:710–713. doi: 10.1038/oby.2007.95. [DOI] [PubMed] [Google Scholar]
  • 83.Hayashi F, Watanabe M, Nanba T, Inoue N, Akamizu T, Iwatani Y. Association of the -31C/T functional polymorphism in the interleukin-1β gene with the intractability of Graves' disease and the proportion of T helper type 17 cells. Clin Exp Immunol. 2009;158:281–286. doi: 10.1111/j.1365-2249.2009.04034.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Wang X, Jiang F, Liang Y, Xu L, Li H, Liu Y, et al. Interleukin-1β-31C/T and -511T/C polymorphisms were associated with preeclampsia in Chinese Han population. PLoS One. 2014;9:e106919. doi: 10.1371/journal.pone.0106919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Ramos BR, Mendes ND, Tanikawa AA, Amador MA, dos Santos NP, dos Santos SE, et al. Ancestry informative markers and selected single nucleotide polymorphisms in immunoregulatory genes on preterm labor and preterm premature rupture of membranes: a case control study. BMC Pregnancy Childbirth. 2016;16:30. doi: 10.1186/s12884-016-0823-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Pitarque M, von Richter O, Oke B, Berkkan H, Oscarson M, Ingelman-Sundberg M. Identification of a single nucleotide polymorphism in the TATA box of the CYP2A6 gene: impairment of its promoter activity. Biochem Biophys Res Commun. 2001;284:455–460. doi: 10.1006/bbrc.2001.4990. [DOI] [PubMed] [Google Scholar]
  • 87.Pianezza ML, Sellers EM, Tyndale RF. Nicotine metabolism defect reduces smoking. Nature. 1998;393:750. doi: 10.1038/31623. [DOI] [PubMed] [Google Scholar]
  • 88.Zukunft J, Lang T, Richter T, Hirsch-Ernst KI, Nussler AK, Klein K, et al. A natural CYP2B6 TATA box polymorphism (-82T--> C) leading to enhanced transcription and relocation of the transcriptional start site. Mol Pharmacol. 2005;67:1772–1782. doi: 10.1124/mol.104.008086. [DOI] [PubMed] [Google Scholar]
  • 89.Al-Shakfa F, Dulucq S, Brukner I, Milacic I, Ansari M, Beaulieu P, et al. DNA variants in region for noncoding interfering transcript of dihydrofolate reductase gene and outcome in childhood acute lymphoblastic leukemia. Clin Cancer Res. 2009;15:6931–6938. doi: 10.1158/1078-0432.CCR-09-0641. [DOI] [PubMed] [Google Scholar]
  • 90.Xie C, Wen X, Ding P, Liu T, He Y, Niu Z, et al. Influence of CYP2A6*4 genotypes on maternal serum cotinine among Chinese nonsmoking pregnant women. Nicotine Tob Res. 2014;16:406–412. doi: 10.1093/ntr/ntt164. [DOI] [PubMed] [Google Scholar]
  • 91.Rao Y, Hoffmann E, Zia M, Bodin L, Zeman M, Sellers EM, et al. Duplications and defects in the CYP2A6 gene: identification, genotyping, and in vivo effects on smoking. Mol Pharmacol. 2000;58:747–755. doi: 10.1124/mol.58.4.747. [DOI] [PubMed] [Google Scholar]
  • 92.Shimizu M, Koyama T, Kishimoto I, Yamazaki H. Dataset for genotyping validation of cytochrome P450 2A6 whole-gene deletion (CYP2A6*4) by real-time polymerase chain reaction platforms. Data Brief. 2015;5:642–645. doi: 10.1016/j.dib.2015.10.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Skubisz MM, Tong S. Of leaves and butterflies: how methotrexate came to be the savior of women. Obstet Gynecol. 2011;118:1169–1173. doi: 10.1097/AOG.0b013e31822fcc0d. [DOI] [PubMed] [Google Scholar]
  • 94.Kralíkova E, Bajerova J, Raslova N, Rames J, Himmerova V. Smoking and pregnancy: prevalence, knowledge, anthropometry, risk communication. Prague Med Rep. 2005;106:195–200. [PubMed] [Google Scholar]
  • 95.Chow EJ, Stratton KL, Leisenring WM, Oeffinger KC, Sklar CA, Donaldson SS, et al. Pregnancy after chemotherapy in male and female survivors of childhood cancer treated between 1970 and 1999: a report from the Childhood Cancer Survivor Study cohort. Lancet Oncol. 2016;17:567–576. doi: 10.1016/S1470-2045(16)00086-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Dooley MA, Nair R. Therapy insight: preserving fertility in cyclophosphamide-treated patients with rheumatic disease. Nat Clin Pract Rheumatol. 2008;4:250–7. [DOI] [PubMed]
  • 97.Leader A, Lishner M, Michaeli J, Revel A. Fertility considerations and preservation in haemato-oncology patients undergoing treatment. Br J Haematol. 2011;153:291–308. [DOI] [PubMed]
  • 98.Yiu ZZ, Warren RB, Mrowietz U, Griffiths CE. Safety of conventional systemic therapies for psoriasis on reproductive potential and outcomes. J Dermatolog Treat. 2015;26:329–334. doi: 10.3109/09546634.2014.991673. [DOI] [PubMed] [Google Scholar]
  • 99.Schwartz AG, Ray RM, Cote ML, Abrams J, Sokol RJ, Hendrix SL, et al. Hormone use, reproductive history, and risk of lung cancer: The Women's Health Initiative Studies. J Thorac Oncol. 2015;10:1004–1013. doi: 10.1097/JTO.0000000000000558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Zhong GC, Liu Y, Chen N, Hao FB, Wang K, Cheng JH, et al. Reproductive factors, menopausal hormone therapies and primary liver cancer risk: a systematic review and dose-response meta-analysis of observational studies. Hum Reprod Update. 2016;23:126–138. doi: 10.1093/humupd/dmw037. [DOI] [PubMed] [Google Scholar]
  • 101.Moretti E, Figura N, Collodel G, Ponzetto A. Can Helicobacter pylori infection influence human reproduction? World J Gastroenterol. 2014;2:5567–5574. doi: 10.3748/wjg.v20.i19.5567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Laurberg P, Andersen SL. Graves'-Basedow disease in pregnancy. New trends in the management and guidance to reduce the risk of birth defects caused by antithyroid drugs. Nuklearmedizin. 2015;54:106–111. [PubMed] [Google Scholar]
  • 103.Sichel DA, Cohen LS, Robertson LM, Ruttenberg A, Rosenbaum JF. Prophylactic estrogen in recurrent postpartum affective disorder. Biol Psychiatry. 1995;38:814–818. doi: 10.1016/0006-3223(95)00063-1. [DOI] [PubMed] [Google Scholar]
  • 104.Luke B. Adverse effects of female obesity and interaction with race on reproductive potential. Fertil Steril. 2017;107:868–877. doi: 10.1016/j.fertnstert.2017.02.114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Leisegang K, Bouic PJ, Menkveld R, Henkel RR. Obesity is associated with increased seminal insulin and leptin alongside reduced fertility parameters in a controlled male cohort. Reprod Biol Endocrinol. 2014;12:34. doi: 10.1186/1477-7827-12-34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM. Positional cloning of the mouse obese gene and its human homologue. Nature. 1994;372:425–432. doi: 10.1038/372425a0. [DOI] [PubMed] [Google Scholar]
  • 107.Horan M, Millar DS, Hedderich J, Lewis G, Newsway V, Mo N, et al. Human growth hormone 1 (GH1) gene expression: complex haplotype-dependent influence of polymorphic variation in the proximal promoter and locus control region. Hum Mutat. 2003;21:408–423. doi: 10.1002/humu.10167. [DOI] [PubMed] [Google Scholar]
  • 108.Landrum MJ, Lee JM, Riley GR, Jang W, Rubinstein WS, Church DM, et al. ClinVar: public archive of relationships among sequence variation and human phenotype. Nucleic Acids Res. 2014;42:D980–D985. doi: 10.1093/nar/gkt1113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Tumer N, Erdos B, Matheny M, Cudykier I, Scarpace PJ. Leptin antagonist reverses hypertension caused by leptin overexpression, but fails to normalize obesity-related hypertension. J Hypertens. 2007;25:2471–2478. doi: 10.1097/HJH.0b013e3282e9a9fd. [DOI] [PubMed] [Google Scholar]
  • 110.Beltowski J. Central vs. peripheral leptin excess in the pathogenesis of obesity-associated hypertension. J Hypertens. 2008;26:827–828. doi: 10.1097/HJH.0b013e3282f47688. [DOI] [PubMed] [Google Scholar]
  • 111.Takagi Y, Kinoshita K, Ozaki N, Seino Y, Murata Y, Oshida Y, et al. Mice deficient in proglucagon-derived peptides exhibit glucose intolerance on a high-fat diet but are resistant to obesity. PLoS One. 2015;10:e0138322. doi: 10.1371/journal.pone.0138322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Sugiyama C, Yamamoto M, Kotani T, Kikkawa F, Murata Y, Hayashi Y. Fertility and pregnancy-associated ß-cell proliferation in mice deficient in proglucagon-derived peptides. PLoS One. 2012;7:e43745. doi: 10.1371/journal.pone.0043745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Saggese G, Baroncelli GI, Vanacore T, Fiore L, Ruggieri S, Federico G. Indications and strategies for continuing GH treatment during transition from late adolescence to early adulthood in patients with GH deficiency: the impact on bone mass. J Endocrinol Invest. 2004;27:596–602. doi: 10.1007/BF03347486. [DOI] [PubMed] [Google Scholar]
  • 114.Bielohuby M, Roemmler J, Manolopoulou J, Johnsen I, Sawitzky M, Schopohl J, et al. Chronic growth hormone excess is associated with increased aldosterone: a study in patients with acromegaly and in growth hormone transgenic mice. Exp Biol Med (Maywood) 2009;234:1002–1009. doi: 10.3181/0901-RM-34. [DOI] [PubMed] [Google Scholar]
  • 115.Lepercq J, Cauzac M, Lahlou N, Timsit J, Girard J, Auwerx J, et al. Overexpression of placental leptin in diabetic pregnancy: a critical role for insulin. Diabetes. 1998;47:847–850. doi: 10.2337/diabetes.47.5.847. [DOI] [PubMed] [Google Scholar]
  • 116.Bassett JM, Hanson C. Prevention of hypoinsulinemia modifies catecholamine effects in fetal sheep. Am J Physiol Regul Integr Comp Physiol. 2000;278:R1171–R1181. doi: 10.1152/ajpregu.2000.278.5.R1171. [DOI] [PubMed] [Google Scholar]
  • 117.Smith GD, Jackson LM, Foster DL. Leptin regulation of reproductive function and fertility. Theriogenology. 2002;57:73–86. doi: 10.1016/S0093-691X(01)00658-6. [DOI] [PubMed] [Google Scholar]
  • 118.Holness MJ, Munns MJ, Sugden MC. Current concepts concerning the role of leptin in reproductive function. Mol Cell Endocrinol. 1999;157:11–20. doi: 10.1016/S0303-7207(99)00126-4. [DOI] [PubMed] [Google Scholar]
  • 119.Bluher S, Mantzoros CS. Leptin in reproduction. Curr Opin Endocrinol Diabetes Obes. 2007;14:458–464. doi: 10.1097/MED.0b013e3282f1cfdc. [DOI] [PubMed] [Google Scholar]
  • 120.Popovic V, Casanueva FF. Leptin, nutrition and reproduction: new insights. Hormones (Athens). 2002;1:204–217. doi: 10.14310/horm.2002.1169. [DOI] [PubMed] [Google Scholar]
  • 121.Song Y, Yu Y, Wang D, Chai S, Liu D, Xiao X, et al. Maternal high-fat diet feeding during pregnancy and lactation augments lung inflammation and remodeling in the offspring. Respir Physiol Neurobiol. 2015;207:1–6. doi: 10.1016/j.resp.2014.12.003. [DOI] [PubMed] [Google Scholar]
  • 122.Khorram O, Keen-Rinehart E, Chuang TD, Ross MG, Desai M. Maternal undernutrition induces premature reproductive senescence in adult female rat offspring. Fertil Steril. 2015;103:291–298. doi: 10.1016/j.fertnstert.2014.09.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Valleau JC, Sullivan EL. The impact of leptin on perinatal development and psychopathology. J Chem Neuroanat. 2014;61-62:221–232. doi: 10.1016/j.jchemneu.2014.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.El Orabi H, Ghalia AA, Khalifa A, Mahfouz H, El Shalkani A, Shoieb N. Serum leptin as an additional possible pathogenic factor in polycystic ovary syndrome. Clin Biochem. 1999;32:71–75. doi: 10.1016/S0009-9120(98)00091-5. [DOI] [PubMed] [Google Scholar]
  • 125.Anderson CM, Ren J. Leptin, leptin resistance and endothelial dysfunction in pre-eclampsia. Cell Mol Biol (Noisy-le-Grand) 2002;48:OL323–OL329. [PubMed] [Google Scholar]
  • 126.Kazemian E, Sotoudeh G, Dorosty-Motlagh AR, Eshraghian MR, Bagheri M. Maternal obesity and energy intake as risk factors of pregnancy-induced hypertension among Iranian women. J Health Popul Nutr. 2014;32:486–493. [PMC free article] [PubMed] [Google Scholar]
  • 127.Luque-Ramirez M, Escobar-Morreale HF. Polycystic ovary syndrome as a paradigm for prehypertension, prediabetes, and preobesity. Curr Hypertens Rep. 2014;16:500. doi: 10.1007/s11906-014-0500-6. [DOI] [PubMed] [Google Scholar]
  • 128.Taylor PD, Samuelsson AM, Poston L. Maternal obesity and the developmental programming of hypertension: a role for leptin. Acta Physiol (Oxf) 2014;210:508–523. doi: 10.1111/apha.12223. [DOI] [PubMed] [Google Scholar]
  • 129.Perry IJ, Whincup PH, Shaper AG. Environmental factors in the development of essential hypertension. Br Med Bull. 1994;50:246–259. doi: 10.1093/oxfordjournals.bmb.a072890. [DOI] [PubMed] [Google Scholar]
  • 130.Bloom SR, Johnston DI. Failure of glucagon release in infants of diabetic mothers. Br Med J. 1972;4:453–454. doi: 10.1136/bmj.4.5838.453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Lee PA, Houk CP. Gonadotropin-releasing hormone analog therapy for central precocious puberty and other childhood disorders affecting growth and puberty. Treat Endocrinol. 2006;5:287–296. doi: 10.2165/00024677-200605050-00003. [DOI] [PubMed] [Google Scholar]
  • 132.Chandrashekar V, Zaczek D, Bartke A. The consequences of altered somatotropic system on reproduction. Biol Reprod. 2004;71:17–27. doi: 10.1095/biolreprod.103.027060. [DOI] [PubMed] [Google Scholar]
  • 133.Spiliotis BE. Growth hormone insufficiency and its impact on ovarian function. Ann N Y Acad Sci. 2003;997:77–84. doi: 10.1196/annals.1290.009. [DOI] [PubMed] [Google Scholar]
  • 134.Juul A, Hjortskov N, Jepsen LT, Nielsen B, Halkjaer-Kristensen J, Vahl N, et al. Growth hormone deficiency and hyperthermia during exercise: a controlled study of sixteen GH-deficient patients. J Clin Endocrinol Metab. 1995;80:3335–3340. doi: 10.1210/jcem.80.11.7593447. [DOI] [PubMed] [Google Scholar]
  • 135.Grynberg M, Salenave S, Young J, Chanson P. Female gonadal function before and after treatment of acromegaly. J Clin Endocrinol Metab. 2010;95:4518–4525. doi: 10.1210/jc.2009-2815. [DOI] [PubMed] [Google Scholar]
  • 136.Karkanias GB, Morales JC, Li CS. Deficits in reproductive behavior in diabetic female rats are due to hypoinsulinemia rather than hyperglycemia. Horm Behav. 1997;32:19–29. doi: 10.1006/hbeh.1997.1401. [DOI] [PubMed] [Google Scholar]
  • 137.Shvyrkova NA, Aleksandrova EA, Kupets TV. The behavioral development of rats with prenatal hypoinsulinemia. Zh Vyssh Nerv Deiat Im I P Pavlova. 1995;45:1190–1197. [PubMed] [Google Scholar]
  • 138.Chaffin DG, Clark RM, McCracken D, Philipps AF. Effect of hypoinsulinemia on growth in the fetal rabbit. Biol Neonate. 1995;67:186–193. doi: 10.1159/000244162. [DOI] [PubMed] [Google Scholar]
  • 139.Baaziz N, Curry DL. Synthesis-secretion coupling of insulin: effect of pregnancy and lactation. Pancreas. 1993;8:316–324. doi: 10.1097/00006676-199305000-00006. [DOI] [PubMed] [Google Scholar]
  • 140.Felig P, Lynch V. Starvation in human pregnancy: hypoglycemia, hypoinsulinemia, and hyperketonemia. Science. 1970;170:990–992. doi: 10.1126/science.170.3961.990. [DOI] [PubMed] [Google Scholar]
  • 141.Fernandez-Twinn DS, Blackmore HL, Siggens L, Giussani DA, Cross CM, Foo R, et al. The programming of cardiac hypertrophy in the offspring by maternal obesity is associated with hyperinsulinemia, AKT, ERK, and mTOR activation. Endocrinology. 2012;153:5961–5971. doi: 10.1210/en.2012-1508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Gisslen T, Nathan B, Thompson T, Rao R. Hyperinsulinism associated with gestational exposure to bupropion in a newborn infant. J Pediatr Endocrinol Metab. 2011;24:819–822. doi: 10.1515/JPEM.2011.317. [DOI] [PubMed] [Google Scholar]
  • 143.Norman RJ, Masters S, Hague W. Hyperinsulinemia is common in family members of women with polycystic ovary syndrome. Fertil Steril. 1996;66:942–947. doi: 10.1016/S0015-0282(16)58687-7. [DOI] [PubMed] [Google Scholar]
  • 144.Othman NH, Ab Rahman AF. Obstetric and birth outcomes in pregnant women with epilepsy: A hospital-based study. Ann Indian Acad Neurol. 2013;16:534–537. doi: 10.4103/0972-2327.120458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Katz O, Levy A, Wiznitzer A, Sheiner E. Pregnancy and perinatal outcome in epileptic women: a population-based study. J Matern Fetal Neonatal Med. 2006;19:21–25. doi: 10.1080/14767050500434096. [DOI] [PubMed] [Google Scholar]
  • 146.Zhidkova IA, Karlov VA, IuB K'k, Zlatorunskaia MA, Braginskaia SG, Kaznacheeva TV, et al. Reproductive potential of women with epilepsy. Zh Nevrol Psikhiatr Im S S Korsakova. 2009;109:31–36. [PubMed] [Google Scholar]
  • 147.Wojcik M, Zieleniak A, Zurawska-Klis M, Cypryk K, Wozniak LA. Increased expression of immune-related genes in leukocytes of patients with diagnosed gestational diabetes mellitus (GDM) Exp Biol Med (Maywood) 2016;241:457–465. doi: 10.1177/1535370215615699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Caron KM, Soo SC, Wetsel WC, Stocco DM, Clark BJ, Parker KL. Targeted disruption of the mouse gene encoding steroidogenic acute regulatory protein provides insights into congenital lipoid adrenal hyperplasia. Proc Natl Acad Sci USA. 1997;94:11540–11545. doi: 10.1073/pnas.94.21.11540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Roe A, Hillman J, Butts S, Smith M, Rader D, Playford M, et al. Decreased cholesterol efflux capacity and atherogenic lipid profile in young women with PCOS. J Clin Endocrinol Metab. 2014;99:e841–e847. doi: 10.1210/jc.2013-3918. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Silliman K, Tall AR, Kretchmer N, Forte TM. Unusual high-density lipoprotein subclass distribution during late pregnancy. Metabolism. 1993;42:1592–1599. doi: 10.1016/0026-0495(93)90156-I. [DOI] [PubMed] [Google Scholar]
  • 151.Garratt M, Bathgate R, de Graaf SP, Brooks RC. Copper-zinc superoxide dismutase deficiency impairs sperm motility and in vivo fertility. Reproduction. 2013;146:297–304. doi: 10.1530/REP-13-0229. [DOI] [PubMed] [Google Scholar]
  • 152.Noda Y, Ota K, Shirasawa T, Shimizu T. Copper/zinc superoxide dismutase insufficiency impairs progesterone secretion and fertility in female mice. Biol Reprod. 2012;86:1–8. doi: 10.1095/biolreprod.111.092999. [DOI] [PubMed] [Google Scholar]
  • 153.Ijiri TW, Vadnais ML, Lin AM, Huang AP, Cao W, Merdiushev T, et al. Male mice express spermatogenic cell-specific triosephosphate isomerase isozymes. Mol Reprod Dev. 2013;80:862–870. doi: 10.1002/mrd.22217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Kirchhoff S, Kim JS, Hagendorff A, Thonnissen E, Kruger O, Lamers WH, et al. Abnormal cardiac conduction and morphogenesis in connexin40 and connexin43 double-deficient mice. Circ Res. 2000;87:399–405. doi: 10.1161/01.RES.87.5.399. [DOI] [PubMed] [Google Scholar]
  • 155.Anyaegbunam AM, Scarpelli S, Mikhail MS. Chronic hypertension in gestational diabetes: influence on pregnancy outcome. Gynecol Obstet Invest. 1995;39:167–170. doi: 10.1159/000292402. [DOI] [PubMed] [Google Scholar]
  • 156.Hagstrom H, Hoijer J, Ludvigsson JF, Bottai M, Ekbom A, Hultcrantz R, et al. Adverse outcomes of pregnancy in women with non-alcoholic fatty liver disease. Liver Int. 2016;36:268–274. doi: 10.1111/liv.12902. [DOI] [PubMed] [Google Scholar]
  • 157.Cheng N, Xiang T, Wu X, Li M, Xie Y, Zhang L. Acute fatty liver of pregnancy: a retrospective study of 32 cases in South China. J Matern Fetal Neonatal Med. 2014;27:1693–1697. doi: 10.3109/14767058.2013.871704. [DOI] [PubMed] [Google Scholar]
  • 158.Matteo M, Serviddio G, Massenzio F, Scillitani G, Castellana L, Picca G, et al. Reduced percentage of natural killer cells associated with impaired cytokine network in the secretory endometrium of infertile women with polycystic ovary syndrome. Fertil Steril. 2010;94:2222–2227. doi: 10.1016/j.fertnstert.2010.01.049. [DOI] [PubMed] [Google Scholar]
  • 159.Huber-Buchholz MM, Carey DG, Norman RJ. Restoration of reproductive potential by lifestyle modification in obese polycystic ovary syndrome: role of insulin sensitivity and luteinizing hormone. J Clin Endocrinol Metab. 1999;84:1470–1474. doi: 10.1210/jcem.84.4.5596. [DOI] [PubMed] [Google Scholar]
  • 160.Gunderson EP, Quesenberry CP Jr, Ning X, Jacobs DR Jr, Gross M, Goff DC Jr, et al. Lactation duration and midlife atherosclerosis. Obstet Gynecol. 2015;126:381-390 [DOI] [PMC free article] [PubMed]
  • 161.Skilton MR, Serusclat A, Begg LM, Moulin P, Bonnet F. Parity and carotid atherosclerosis in men and women: insights into the roles of childbearing and child-rearing. Stroke. 2009;40:1152–1157. doi: 10.1161/STROKEAHA.108.535807. [DOI] [PubMed] [Google Scholar]
  • 162.Belo L, Santos-Silva A, Quintanilha A, Rebelo I. Similarities between pre-eclampsia and atherosclerosis: a protective effect of physical exercise? Curr Med Chem. 2008;15:2223–2229. doi: 10.2174/092986708785747553. [DOI] [PubMed] [Google Scholar]
  • 163.Napoli C, Pignalosa O, Gallo L, Graziano G, Carotenuto F, Fiorito C, et al. Childhood infection and endothelial dysfunction: a potential synergistic link in atherosclerosis. Recenti Prog Med. 2005;96:474–477. [PubMed] [Google Scholar]
  • 164.Martínez HR, Marioni SS, Escamilla Ocanas CE, Gonzalez Garza MT, Moreno-Cuevas JE. Amyotrophic lateral sclerosis in pregnancy: clinical outcome during the post-partum period after stem cell transplantation into the frontal motor cortex. Cytotherapy. 2014;16:402–405. doi: 10.1016/j.jcyt.2013.11.002. [DOI] [PubMed] [Google Scholar]
  • 165.Palomba S, Santagni S, La Sala GB. Progesterone administration for luteal phase deficiency in human reproduction: an old or new issue? J Ovarian Res. 2015;8:77. doi: 10.1186/s13048-015-0205-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Daya S. Efficacy of progesterone support for pregnancy in women with recurrent miscarriage. A meta-analysis of controlled trials. Br J Obstet Gynaecol. 1989;96:275–280. doi: 10.1111/j.1471-0528.1989.tb02386.x. [DOI] [PubMed] [Google Scholar]
  • 167.Nakata K, Yamashita N, Noda Y, Ohsawa I. Stimulation of human damaged sperm motility with hydrogen molecule. Med Gas Res. 2015;5:2. doi: 10.1186/s13618-014-0023-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Frazer KL, Porter S, Goss C. The genetics and implications of neuromuscular diseases in pregnancy. J Perinat Neonatal Nurs. 2013;27:205–214. doi: 10.1097/JPN.0b013e318299c338. [DOI] [PubMed] [Google Scholar]
  • 169.del-Rio-Vellosillo M, Garcia-Medina JJ. Anesthetic considerations in HELLP syndrome. Acta Anaesthesiol Scand. 2016;60:144–157. doi: 10.1111/aas.12639. [DOI] [PubMed] [Google Scholar]
  • 170.Flores JR, Marquez MF. Arrhythmias in pregnancy. How and when to treat? Arch Cardiol Mex. 2007;77:S2-24–S2-31. [PubMed] [Google Scholar]
  • 171.Frassica JJ, Orav EJ, Walsh EP, Lipshultz SE. Arrhythmias in children prenatally exposed to cocaine. Arch Pediatr Adolesc Med. 1994;148:1163–1169. doi: 10.1001/archpedi.1994.02170110049008. [DOI] [PubMed] [Google Scholar]
  • 172.Weissgerber TL, Turner ST, Mosley TH, Jr, Kardia SL, Hanis CL, Milic NM, et al. Hypertension in pregnancy and future cardiovascular event risk in siblings. J Am Soc Nephrol. 2016;27:894–902. doi: 10.1681/ASN.2015010086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Matthiesen NB, Henriksen TB, Gaynor JW, Agergaard P, Bach CC, Hjortdal VE, et al. Congenital heart defects and indices of fetal cerebral growth in a nationwide cohort of 924 422 liveborn infants. Circulation. 2016;133:566–575. doi: 10.1161/CIRCULATIONAHA.115.019089. [DOI] [PubMed] [Google Scholar]
  • 174.Lombo M, Fernandez-Diez C, Gonzalez-Rojo S, Navarro C, Robles V, Herraez MP. Transgenerational inheritance of heart disorders caused by paternal bisphenol A exposure. Environ Pollut. 2015;206:667–678. doi: 10.1016/j.envpol.2015.08.016. [DOI] [PubMed] [Google Scholar]
  • 175.Donofrio MT, Duplessis AJ, Limperopoulos C. Impact of congenital heart disease on fetal brain development and injury. Curr Opin Pediatr. 2011;23:502–511. doi: 10.1097/MOP.0b013e32834aa583. [DOI] [PubMed] [Google Scholar]
  • 176.Martiney JA, Cerami A, Inhibition SAF. of hemozoin formation in Plasmodium falciparum trophozoite extracts by heme analogs: possible implication in the resistance to malaria conferred by the β-thalassemia trait. Mol Med. 1996;2:236–246. [PMC free article] [PubMed] [Google Scholar]
  • 177.Nalls MA, Wilson JG, Patterson NJ, Tandon A, Zmuda JM, Huntsman S, et al. Admixture mapping of white cell count: genetic locus responsible for lower white blood cell count in the Health ABC and Jackson Heart studies. Am J Hum Genet. 2008;82:81–87. doi: 10.1016/j.ajhg.2007.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Michon P, Woolley I, Wood EM, Kastens W, Zimmerman PA, Adams JH. Duffy-null promoter heterozygosity reduces DARC expression and abrogates adhesion of the P. vivax ligand required for blood-stage infection. FEBS Lett. 2001;495:111–114. doi: 10.1016/S0014-5793(01)02370-5. [DOI] [PubMed] [Google Scholar]
  • 179.Boldt AB, Culpi L, Tsuneto LT, de Souza IR, Kun JF, Petzl-Erler ML. Diversity of the MBL2 gene in various Brazilian populations and the case of selection at the mannose-binding lectin locus. Hum Immunol. 2006;67:722–734. doi: 10.1016/j.humimm.2006.05.009. [DOI] [PubMed] [Google Scholar]
  • 180.Sziller I, Babula O, Hupuczi P, Nagy B, Rigo B, Szabo G, et al. Mannose-binding lectin (MBL) codon 54 gene polymorphism protects against development of pre-eclampsia, HELLP syndrome and pre-eclampsia-associated intrauterine growth restriction. Mol Hum Reprod. 2007;13:281–285. doi: 10.1093/molehr/gam003. [DOI] [PubMed] [Google Scholar]
  • 181.Cervera A, Planas AM, Justicia C, Urra X, Jensenius JC, Torres F, et al. Genetically-defined deficiency of mannose-binding lectin is associated with protection after experimental stroke in mice and outcome in human stroke. PLoS One. 2010;5:e8433. doi: 10.1371/journal.pone.0008433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Starodubtseva NL, Sobolev VV, Soboleva AG, Nikolaev AA, Bruskin SA. Genes expression of metalloproteinases (MMP-1, MMP-2, MMP-9, and MMP-12) associated with psoriasis. Russ J Genet. 2011;47:1117–1123. doi: 10.1134/S102279541109016X. [DOI] [PubMed] [Google Scholar]
  • 183.Manetti M, Ibba-Manneschi L, Fatini C, Guiducci S, Cuomo G, Bonino C, et al. Association of a functional polymorphism in the matrix metalloproteinase-12 promoter region with systemic sclerosis in an Italian population. J Rheumatol. 2010;37:1852–1857. doi: 10.3899/jrheum.100237. [DOI] [PubMed] [Google Scholar]
  • 184.Hunninghake GM, Cho MH, Tesfaigzi Y, Soto-Quiros ME, Avila L, Lasky-Su J, et al. MMP12, lung function, and COPD in high-risk populations. N Engl J Med. 2009;361:2599–2608. doi: 10.1056/NEJMoa0904006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Cao A, Galanello R. β-thalassemia. Genet Med. 2010;12:61–76. doi: 10.1097/GIM.0b013e3181cd68ed. [DOI] [PubMed] [Google Scholar]
  • 186.Velzing-Aarts FV, van der Dijs FP, Muskiet FA, Duits AJ. The association of pre-eclampsia with the Duffy negative phenotype in women of West African descent. BJOG. 2002;109:453–455. doi: 10.1111/j.1471-0528.2002.01181.x. [DOI] [PubMed] [Google Scholar]
  • 187.Kalia N, Singh J, Sharma S, Arora H, Kaur M. Genetic and phenotypic screening of mannose-binding lectin in relation to risk of recurrent vulvovaginal infections in women of North India: a prospective cohort study. Front Microbiol. 2017;8:75. doi: 10.3389/fmicb.2017.00075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Pencovich N, Hantisteanu S, Hallak M, Fainaru O. Gonadotropin stimulation in mice leads to ovarian accumulation of immature myeloid cells and altered expression of proangiogenic genes. Eur J Obstet Gynecol Reprod Biol. 2014;179:75–82. doi: 10.1016/j.ejogrb.2014.05.025. [DOI] [PubMed] [Google Scholar]
  • 189.Huang QT, Chen JH, Hang LL, Liu SS, Zhong M. Activation of PAR-1/NADPH oxidase/ROS signaling pathways is crucial for the thrombin-induced sFlt-1 production in extravillous trophoblasts: possible involvement in the pathogenesis of preeclampsia. Cell Physiol Biochem. 2015;35:1654–1662. doi: 10.1159/000373979. [DOI] [PubMed] [Google Scholar]
  • 190.Psihogios V, Rodda C, Reid E, Clark M, Clarke C, Bowden D. Reproductive health in individuals with homozygous β-thalassemia: knowledge, attitudes, and behavior. Fertil Steril. 2002;77:119–127. doi: 10.1016/S0015-0282(01)02933-8. [DOI] [PubMed] [Google Scholar]
  • 191.Le Gouez A, Benachi A, Mercier FJ. Fever and pregnancy. Anaesth Crit Care Pain Med. 2016;35:S5–12. doi: 10.1016/j.accpm.2016.06.007. [DOI] [PubMed] [Google Scholar]
  • 192.Hsu WY, Wu CH, Hsieh CT, Lo HC, Lin JS, Kao MD. Low body weight gain, low white blood cell count and high serum ferritin as markers of poor nutrition and increased risk for preterm delivery. Asia Pac J Clin Nutr. 2013;22:90–99. doi: 10.6133/apjcn.2013.22.1.05. [DOI] [PubMed] [Google Scholar]
  • 193.Wikstrand MH, Hard AL, Niklasson A, Smith L, Lofqvist C, Hellstrom A. Maternal and neonatal factors associated with poor early weight gain and later retinopathy of prematurity. Acta Paediatr. 2011;100:1528–1533. doi: 10.1111/j.1651-2227.2011.02394.x. [DOI] [PubMed] [Google Scholar]
  • 194.Mendola P, Mumford SL, Mannisto TI, Holston A, Reddy UM, Laughon SK. Controlled direct effects of preeclampsia on neonatal health after accounting for mediation by preterm birth. Epidemiology. 2015;26:17–26. doi: 10.1097/EDE.0000000000000213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Allen R, Rogozinska E, Sivarajasingam P, Khan KS, Thangaratinam S. Effect of diet- and lifestyle-based metabolic risk-modifying interventions on preeclampsia: a meta-analysis. Acta Obstet Gynecol Scand. 2014;93:973–985. doi: 10.1111/aogs.12467. [DOI] [PubMed] [Google Scholar]
  • 196.Rayman MP, Searle E, Kelly L, Johnsen S, Bodman-Smith K, Bath SC, et al. Effect of selenium on markers of risk of pre-eclampsia in UK pregnant women: a randomised, controlled pilot trial. Br J Nutr. 2014;112:99–111. doi: 10.1017/S0007114514000531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Hux VJ, Roberts JM. A potential role for allostatic load in preeclampsia. Matern Child Health J. 2015;19:591–597. doi: 10.1007/s10995-014-1543-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Silva LM, Coolman M, Steegers EA, Jaddoe VW, Moll HA, Hofman A, et al. Low socioeconomic status is a risk factor for preeclampsia: the Generation R Study. J Hypertens. 2008;26:1200–1208. doi: 10.1097/HJH.0b013e3282fcc36e. [DOI] [PubMed] [Google Scholar]
  • 199.Melchiorre K, Thilaganathan B. Maternal cardiac function in preeclampsia. Curr Opin Obstet Gynecol. 2011;23:440–447. doi: 10.1097/GCO.0b013e32834cb7a4. [DOI] [PubMed] [Google Scholar]
  • 200.England L, Zhang J. Smoking and risk of preeclampsia: a systematic review. Front Biosci. 2007;12:2471–2483. doi: 10.2741/2248. [DOI] [PubMed] [Google Scholar]
  • 201.Conde-Agudelo A, Belizan JM. Risk factors for pre-eclampsia in a large cohort of Latin American and Caribbean women. BJOG. 2000;107:75–83. doi: 10.1111/j.1471-0528.2000.tb11582.x. [DOI] [PubMed] [Google Scholar]
  • 202.Spinillo A, Capuzzo E, Colonna L, Piazzi G, Nicola S, Baltaro F. The effect of work activity in pregnancy on the risk of severe preeclampsia. Aust N Z J Obstet Gynaecol. 1995;35:380–385. doi: 10.1111/j.1479-828X.1995.tb02146.x. [DOI] [PubMed] [Google Scholar]
  • 203.Gundlapalli AV, Scalchunes C, Boyle M, Hill HR. Fertility, pregnancies and outcomes reported by females with common variable immune deficiency and hypogammaglobulinemia: results from an internet-based survey. J Clin Immunol. 2015;35:125–134. doi: 10.1007/s10875-014-0123-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Brinker KA, Silk HJ. Common variable immune deficiency and treatment with intravenous immunoglobulin during pregnancy. Ann Allergy Asthma Immunol. 2012;108:464–465. doi: 10.1016/j.anai.2012.04.014. [DOI] [PubMed] [Google Scholar]
  • 205.Bereczki D., Jr Pregnancy and acute ischemic stroke. Orv Hetil. 2016;157:763–766. doi: 10.1556/650.2016.30421. [DOI] [PubMed] [Google Scholar]
  • 206.Savitz DA, Danilack VA, Elston B, Lipkind HS. Pregnancy-induced hypertension and diabetes and the risk of cardiovascular disease, stroke, and diabetes hospitalization in the year following delivery. Am J Epidemiol. 2014;180:41–44. doi: 10.1093/aje/kwu118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Reilly CR, Cuesta-Fernandez A, Kayaleh OR. Successful gestation and delivery using clopidogrel for secondary stroke prophylaxis: a case report and literature review. Arch Gynecol Obstet. 2014;290:591–594. doi: 10.1007/s00404-014-3269-6. [DOI] [PubMed] [Google Scholar]
  • 208.Luo L, Chen D, Qu Y, Wu J, Li X, Mu D. Association between hypoxia and perinatal arterial ischemic stroke: a meta-analysis. PLoS One. 2014;9:e90106. doi: 10.1371/journal.pone.0090106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Mann JR, McDermott S, Pan C, Hardin JW. Maternal hypertension and intrapartum fever are associated with increased risk of ischemic stroke during infancy. Dev Med Child Neurol. 2013;55:58–64. doi: 10.1111/j.1469-8749.2012.04409.x. [DOI] [PubMed] [Google Scholar]
  • 210.Tate J, Bushnell C. Pregnancy and stroke risk in women. Womens Health (Lond) 2011;7:363–374. doi: 10.2217/WHE.11.19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Le Flem L, Picard V, Emmerich J, Gandrille S, Fiessinger JN, Aiach M, et al. Mutations in promoter region of thrombomodulin and venous thromboembolic disease. Arterioscler Thromb Vasc Biol. 1999;19:1098–1104. doi: 10.1161/01.ATV.19.4.1098. [DOI] [PubMed] [Google Scholar]
  • 212.Arnaud E, Barbalat V, Nicaud V, Cambien F, Evans A, Morrison C, et al. Polymorphisms in the 5' regulatory region of the tissue factor gene and the risk of myocardial infarction and venous thromboembolism: the ECTIM and PATHROS studies. Etude Cas-Témoins de l'Infarctus du Myocarde. Paris Thrombosis case-control Study. Arterioscler Thromb Vasc Biol. 2000;20:892–898. doi: 10.1161/01.ATV.20.3.892. [DOI] [PubMed] [Google Scholar]
  • 213.Kavlie A, Hiltunen L, Rasi V, Prydz H. Two novel mutations in the human coagulation factor VII promoter. Thromb Haemost. 2003;90:194–205. doi: 10.1160/TH02-09-0050. [DOI] [PubMed] [Google Scholar]
  • 214.Suzuki K, Hayashi T. Protein C and its inhibitor in malignancy. Semin Thromb Hemost. 2007;33:667–672. doi: 10.1055/s-2007-991534. [DOI] [PubMed] [Google Scholar]
  • 215.Lay AJ, Liang Z, Rosen ED, Castellino FJ. Mice with a severe deficiency in protein C display prothrombotic and proinflammatory phenotypes and compromised maternal reproductive capabilities. J Clin Invest. 2005;115:1552–1561. doi: 10.1172/JCI24030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Schambeck CM, Grossmann R, Zonnur S, Berger M, Teuchert K, Spahn A, et al. High factor VIII (FVIII) levels in venous thromboembolism: role of unbound FVIII. Thromb Haemost. 2004;92:42–46. doi: 10.1160/TH04-02-0063. [DOI] [PubMed] [Google Scholar]
  • 217.Sood R, Sholl L, Isermann B, Zogg M, Coughlin SR, Weiler H. Maternal Par4 and platelets contribute to defective placenta formation in mouse embryos lacking thrombomodulin. Blood. 2008;112:585–591. doi: 10.1182/blood-2007-09-111302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Cocco E, Varughese J, Buza N, Bellone S, Lin KY, Bellone M, et al. Tissue factor expression in ovarian cancer: implications for immunotherapy with hI-con1, a factor VII-IgGF(c) chimeric protein targeting tissue factor. Clin Exp Metastasis. 2011;28:689–700. doi: 10.1007/s10585-011-9401-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Khealani B, Farhat Z, Mozaffar T. Factor XI deficiency-related spontaneous primary intraventricular hemorrhage. South Med J. 2000;93:1017–1018. [PubMed] [Google Scholar]
  • 220.Witzke G, Bork K, Benes P, Bockers M. Hereditary angioneurotic oedema and blood-coagulation: interaction between C1-esterase-inhibitor and the activation factors of the proteolytic enzyme systems. Klin Wochenschr. 1983;61:1131–1135. doi: 10.1007/BF01530840. [DOI] [PubMed] [Google Scholar]
  • 221.Lin HF, Maeda N, Smithies O, Straight DL, Stafford DW. A coagulation factor IX-deficient mouse model for human hemophilia B. Blood. 1997;90:3962–3966. [PubMed] [Google Scholar]
  • 222.Ameri A, Kurachi S, Sueishi K, Kuwahara M, Kurachi K. Myocardial fibrosis in mice with overexpression of human blood coagulation factor IX. Blood. 2003;101:1871–1873. doi: 10.1182/blood-2002-05-1581. [DOI] [PubMed] [Google Scholar]
  • 223.Dumas G, Dufresne M, Asselin E, Girouard J, Carrier C, Reyes-Moreno C. CD40 pathway activation reveals dual function for macrophages in human endometrial cancer cell survival and invasion. Cancer Immunol Immunother. 2013;62:273–283. doi: 10.1007/s00262-012-1333-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Lyons TR, Borges VF, Betts CB, Guo Q, Kapoor P, Martinson HA, et al. Cyclooxygenase-2-dependent lymphangiogenesis promotes nodal metastasis of postpartum breast cancer. J Clin Invest. 2014;124:3901–3912. doi: 10.1172/JCI73777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Maller O, Hansen KC, Lyons TR, Acerbi I, Weaver VM, Prekeris R, et al. Collagen architecture in pregnancy-induced protection from breast cancer. J Cell Sci. 2013;126:4108–4110. doi: 10.1242/jcs.121590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Rana K, Thaper D, Prabha V. Is there a role for Serratia marcescens in male infertility: An experimental study? Microb Pathog. 2017;105:13–18. doi: 10.1016/j.micpath.2017.01.061. [DOI] [PubMed] [Google Scholar]
  • 227.Moretti E, Figura N, Campagna MS, Gonnelli S, Iacoponi F, Collodel G. Sperm parameters and semen levels of inflammatory cytokines in Helicobacter pylori-infected men. Urology. 2015;86:41–46. doi: 10.1016/j.urology.2015.02.068. [DOI] [PubMed] [Google Scholar]
  • 228.Burnazian RA, Oganesian MN, Burshtein GE. The biochemical indices of the sperm in chronic inflammatory diseases of the accessory genital glands. Urol Nefrol (Mosk). 1992:36–8. [PubMed]
  • 229.I B. Inflammatory gynecological diseases as a social problem in women of reproductive age. Akush Ginekol (Sofiia) 1999;38:33–35. [PubMed] [Google Scholar]
  • 230.Sun Y, Cui M, Zhu W, Xu W, Li N. A case report of a pregnancy-related death caused by primary antiphospholipid antibody syndrome. Int Med Case Rep J. 2014;7:159–163. doi: 10.2147/IMCRJ.S71321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Lopez-Pedrera C, Buendía P, Barbarroja N, Siendones E, Velasco F, Cuadrado MJ. Antiphospholipid-mediated thrombosis: interplay between anticardiolipin antibodies and vascular cells. Clin Appl Thromb Hemost. 2006;12:41–45. doi: 10.1177/107602960601200107. [DOI] [PubMed] [Google Scholar]
  • 232.Hughes G. Hughes syndrome (antiphospholipid syndrome) and myocardial infarction. Int J Clin Pract. 2010;64:1183–1184. doi: 10.1111/j.1742-1241.2010.02340.x. [DOI] [PubMed] [Google Scholar]
  • 233.Soderberg LH, Johansen PP, Herning M, Berg SK. Women's experiences of sexual health after first-time myocardial infarction. J Clin Nurs. 2013;22:3532–3540. doi: 10.1111/jocn.12382. [DOI] [PubMed] [Google Scholar]
  • 234.Rosa GM, Bauckneht M, Ferrero S, Leone Roberti Maggiore U, Brunelli C. Acute myocardial infarction in pregnancy. G Ital Cardiol (Rome) 2013;14:126–134. doi: 10.1714/1218.13525. [DOI] [PubMed] [Google Scholar]
  • 235.Bush N, Nelson-Piercy C, Spark P, Kurinczuk JJ, Brocklehurst P, Knight M, et al. Myocardial infarction in pregnancy and postpartum in the UK. Eur J Prev Cardiol. 2013;20:12–20. doi: 10.1177/1741826711432117. [DOI] [PubMed] [Google Scholar]
  • 236.La Vecchia C, Decarli A, Franceschi S, Gentile A, Negri E, Parazzini F. Menstrual and reproductive factors and the risk of myocardial infarction in women under fifty-five years of age. Am J Obstet Gynecol. 1987;157:1108–1112. doi: 10.1016/S0002-9378(87)80271-5. [DOI] [PubMed] [Google Scholar]
  • 237.Letourneau J, Chan J, Salem W, Chan SW, Shah M, Ebbel E, et al. Fertility sparing surgery for localized ovarian cancers maintains an ability to conceive, but is associated with diminished reproductive potential. J Surg Oncol. 2015;112:26–30. doi: 10.1002/jso.23942. [DOI] [PubMed] [Google Scholar]
  • 238.Crawford NM, Pritchard DA, Herring AH, Steiner AZ. Prospective evaluation of the impact of intermenstrual bleeding on natural fertility. Fertil Steril. 2016;105:1294–1300. doi: 10.1016/j.fertnstert.2016.01.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239.Papp Z, Toth-Pal E, Papp C, Sziller I, Silhavy M, Gavai M, et al. Bilateral hypogastric artery ligation for control of pelvic hemorrhage, reduction of blood flow and preservation of reproductive potential. Experience with 117 cases. Orv Hetil. 2005;146:1279–1285. [PubMed] [Google Scholar]
  • 240.Fotopoulou C, Dudenhausen JW. Uterine compression sutures for preserving fertility in severe postpartum haemorrhage: an overview 13 years after the first description. J Obstet Gynaecol. 2010;30:339–349. doi: 10.3109/01443611003650233. [DOI] [PubMed] [Google Scholar]
  • 241.Adams Hillard PJ. Menstruation in adolescents: what's normal, what's not. Ann N Y Acad Sci. 2008;1135:29–35. doi: 10.1196/annals.1429.022. [DOI] [PubMed] [Google Scholar]
  • 242.den Tonkelaar I, Oddens BJ. Preferred frequency and characteristics of menstrual bleeding in relation to reproductive status, oral contraceptive use, and hormone replacement therapy use. Contraception. 1999;59:357–362. doi: 10.1016/S0010-7824(99)00043-8. [DOI] [PubMed] [Google Scholar]
  • 243.Koeberle P, Pequenot-Jeannin C, Douge C, Barale F. Angioneurotic edema and anesthesia: preparation and perioperative monitoring. Ann Fr Anesth Reanim. 1994;13:417–420. doi: 10.1016/S0750-7658(94)80052-9. [DOI] [PubMed] [Google Scholar]
  • 244.Hardy F, Ngwingtin L, Bazin C, Babinet P. Hereditary angioneurotic edema and pregnancy. J Gynecol Obstet Biol Reprod (Paris) 1990;19:65–68. [PubMed] [Google Scholar]
  • 245.Hopkinson RB, Sutcliffe AJ. Hereditary angioneurotic oedema. Anaesthesia. 1979;34:183–186. doi: 10.1111/j.1365-2044.1979.tb06276.x. [DOI] [PubMed] [Google Scholar]
  • 246.Yu F, Zheng A, Qian J, Li Y, Wu L, Yang J, et al. Prenatal nicotine exposure results in the myocardial fibrosis in the adult male offspring rats. Exp Toxicol Pathol. 2016;68:445–450. doi: 10.1016/j.etp.2016.07.002. [DOI] [PubMed] [Google Scholar]
  • 247.Chen X, Tang Y, Gao M, Qin S, Zhou J, Li X. Prenatal exposure to lipopolysaccharide results in myocardial fibrosis in rat offspring. Int J Mol Sci. 2015;16:10986–10996. doi: 10.3390/ijms160510986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248.Lowenstine LJ, McManamon R, Terio KA. Comparative pathology of aging great apes: Bonobos, Chimpanzees, Gorillas, and Orangutans. Vet Pathol. 2016;53:250–276. doi: 10.1177/0300985815612154. [DOI] [PubMed] [Google Scholar]
  • 249.James AH, Hoots K. The optimal mode of delivery for the haemophilia carrier expecting an affected infant is caesarean delivery. Haemophilia. 2010;16:420–424. doi: 10.1111/j.1365-2516.2009.02142.x. [DOI] [PubMed] [Google Scholar]
  • 250.Kadir RA, Sabin CA, Goldman E, Pollard D, Economides DL, Lee CA. Reproductive choices of women in families with haemophilia. Haemophilia. 2000;6:33–40. doi: 10.1046/j.1365-2516.2000.00353.x. [DOI] [PubMed] [Google Scholar]
  • 251.Yang YC, Fu HC, Wu CY, Wei KT, Huang KE, Kang HY. Androgen receptor accelerates premature senescence of human dermal papilla cells in association with DNA damage. PLoS One. 2013;8:e79434. doi: 10.1371/journal.pone.0079434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252.Perez-Sepulveda A, Espana-Perrot PP, Fernandez XB, Ahumada V, Bustos V, Arraztoa JA. Levels of key enzymes of methionine-homocysteine metabolism in preeclampsia. Biomed Res Int. 2013;2013:731962. doi: 10.1155/2013/731962. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253.Mikael LG, Pancer J, Jiang X, Wu Q, Caudill M, Rozen R. Low dietary folate and methylenetetrahydrofolate reductase deficiency may lead to pregnancy complications through modulation of ApoAI and IFN-γ in spleen and placenta, and through reduction of methylation potential. Mol Nutr Food Res. 2013;57:661–670. doi: 10.1002/mnfr.201200152. [DOI] [PubMed] [Google Scholar]
  • 254.Zhou R, Chen F, Chang F, Bai Y, Chen L. Persistent overexpression of DNA methyltransferase 1 attenuating GABAergic inhibition in basolateral amygdala accounts for anxiety in rat offspring exposed perinatally to low-dose bisphenol A. J Psychiatr Res. 2013;47:1535–1544. doi: 10.1016/j.jpsychires.2013.05.013. [DOI] [PubMed] [Google Scholar]
  • 255.Matrisciano F, Tueting P, Dalal I, Kadriu B, Grayson DR, Davis JM, et al. Epigenetic modifications of GABAergic interneurons are associated with the schizophrenia-like phenotype induced by prenatal stress in mice. Neuropharmacology. 2013;68:184–194. doi: 10.1016/j.neuropharm.2012.04.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 256.Cannuyer J, Van Tongelen A, Loriot A, De Smet C. A gene expression signature identifying transient DNMT1 depletion as a causal factor of cancer-germline gene activation in melanoma. Clin Epigenetics. 2015;7:114. doi: 10.1186/s13148-015-0147-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257.Xu B, Gao L, Cui Y, Gao L, Dai X, Li M, et al. SET protein up-regulated testosterone production in the cultured preantral follicles. Reprod Biol Endocrinol. 2013;11:9. doi: 10.1186/1477-7827-11-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258.Marsh CA, Auchus RJ. Fertility in patients with genetic deficiencies of cytochrome P450c17 (CYP17A1): combined 17-hydroxylase/17,20-lyase deficiency and isolated 17,20-lyase deficiency. Fertil Steril. 2014;101:317–322. doi: 10.1016/j.fertnstert.2013.11.011. [DOI] [PubMed] [Google Scholar]
  • 259.Ferraz-de-Souza B, Lin L, Achermann JC. Steroidogenic factor-1 (SF-1, NR5A1) and human disease. Mol Cell Endocrinol. 2011;336:198–205. doi: 10.1016/j.mce.2010.11.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 260.Kasowski M, Grubert F, Heffelfinger C, Hariharan M, Asabere A, Waszak SM, et al. Variation in transcription factor binding among humans. Science. 2010;328:232–235. doi: 10.1126/science.1183621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261.1000 Genomes Project Consortium. Abecasis GR, Auton A, Brooks LD, MA DP, Durbin RM, et al. An integrated map of genetic variation from 1,092 human genomes. Nature. 2012;491:56–65. doi: 10.1038/nature11632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262.Pugh BF. Purification of the human TATA-binding protein, TBP. Methods Mol Biol. 1995;37:359–367. doi: 10.1385/0-89603-288-4:359. [DOI] [PubMed] [Google Scholar]
  • 263.Savinkova L, Drachkova I, Arshinova T, Ponomarenko P, Ponomarenko M, Kolchanov N. An experimental verification of the predicted effects of promoter TATA box polymorphisms associated with human diseases on interactions between the TATA boxes and TATA-binding protein. PLoS One. 2013;8:e54626. doi: 10.1371/journal.pone.0054626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 264.Arkova O, Kuznetsov N, Fedorova O, Savinkova L. A real-time study of the interaction of TBP with a TATA box-containing duplex identical to an ancestral or minor allele of human gene LEP or TPI. J Biomol Struct Dyn. 2016; 10.1080/07391102.2016.1241190. [DOI] [PubMed]
  • 265.Wolfe A, Kim HH, Tobet S, Stafford DE, Radovick S. Identification of a discrete promoter region of the human GnRH gene that is sufficient for directing neuron-specific expression: a role for POU homeodomain transcription factors. Mol Endocrinol. 2002;16:435–449. doi: 10.1210/mend.16.3.0780. [DOI] [PubMed] [Google Scholar]
  • 266.Stajich JE, Block D, Boulez K, Brenner SE, Chervitz SA, Dagdigian C, et al. The Bioperl toolkit: Perl modules for the life sciences. Genome Res. 2002;12:1611–1618. doi: 10.1101/gr.361602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267.Waardenberg AJ, Basset SD, Bouveret R, Harvey RP. CompGO: an R package for comparing and visualizing Gene Ontology enrichment differences between DNA binding experiments. BMC Bioinformatics. 2015;16:275. doi: 10.1186/s12859-015-0701-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 268.Ponomarenko PM, Savinkova LK, Drachkova IA, Lysova MV, Arshinova TV, Ponomarenko MP, et al. A step-by-step model of TBP/TATA box binding allows predicting human hereditary diseases by single nucleotide polymorphism. Dokl. Biochem. Biophys. 2008;419:88–92. doi: 10.1134/S1607672908020117. [DOI] [PubMed] [Google Scholar]
  • 269.Hahn S, Buratowski S, Sharp PA, Guarente L. Yeast TATA-binding protein TFIID binds to TATA elements with both consensus and nonconsensus DNA sequences. Proc Natl Acad Sci USA. 1989;86:5718–5722. doi: 10.1073/pnas.86.15.5718. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270.Ponomarenko JV, Ponomarenko MP, Frolov AS, Vorobyev DG, Overton GC, Kolchanov NA. Conformational and physicochemical DNA features specific for transcription factor binding sites. Bioinformatics. 1999;15:654–668. doi: 10.1093/bioinformatics/15.7.654. [DOI] [PubMed] [Google Scholar]
  • 271.Karas H, Knuppel R, Schulz W, Sklenar H, Wingender E. Combining structural analysis of DNA with search routines for the detection of transcription regulatory elements. Comput Appl Biosci. 1996;12:441–446. doi: 10.1093/bioinformatics/12.5.441. [DOI] [PubMed] [Google Scholar]
  • 272.Bucher P. Weight matrix descriptions of four eukaryotic RNA polymerase II promoter elements derived from 502 unrelated promoter sequences. J Mol Biol. 1990;212:563–578. doi: 10.1016/0022-2836(90)90223-9. [DOI] [PubMed] [Google Scholar]
  • 273.Ponomarenko MP, Ponomarenko JV, Frolov AS, Podkolodny NL, Savinkova LK, Kolchanov NA, et al. Identification of sequence-dependent DNA features correlating to activity of DNA sites interacting with proteins. Bioinformatics. 1999;15:687–703. doi: 10.1093/bioinformatics/15.7.687. [DOI] [PubMed] [Google Scholar]
  • 274.Sokolenko AA, Sandomirskii II, Savinkova LK. Interaction of yeast TATA-binding protein with short promotor segments. Mol Biol (Mosk) 1996;30:279–285. [PubMed] [Google Scholar]
  • 275.Drachkova I, Savinkova L, Arshinova T, Ponomarenko M, Peltek S, Kolchanov N. The mechanism by which TATA box polymorphisms associated with human hereditary diseases influence interactions with the ТАТА-binding protein. Hum Mutat. 2014;35:601–608. doi: 10.1002/humu.22535. [DOI] [PubMed] [Google Scholar]
  • 276.Drachkova IA, Ponomarenko PM, Arshinova TV, Ponomarenko МP, Suslov VV, Savinkova LK, et al. In vitro examining the existing prognoses how TBP binds to TATA with SNP associated with human diseases. Health. 2011;3:577–583. doi: 10.4236/health.2011.39099. [DOI] [Google Scholar]
  • 277.Ponomarenko PM, Ponomarenko MP, Drachkova IA, Lysova MV, Arshinova TV, Savinkova LK, et al. Prediction of the affinity of the TATA-binding protein to TATA boxes with single nucleotide polymorphisms. Mol Biol (Mosk) 2009;43:327–336. doi: 10.1134/S0026893309030157. [DOI] [PubMed] [Google Scholar]
  • 278.Arkova OV, Kuznetsov NA, Fedorova OS, Kolchanov NA, Savinkova LK. Real-time interaction between ТВР and the TATA box of the human triosephosphate isomerase gene promoter in the norm and pathology. Acta Naturae. 2014;6:36–40. [PMC free article] [PubMed] [Google Scholar]
  • 279.Gunbin KV, Ponomarenko MP, Suslov VV, Gusev F, Fedonin GG, Rogaev EI. Evolution of brain active gene promoters in human lineage towards the increased plasticity of gene regulation. Mol Neurobiol. 2017; 10.1007/s12035-017-0427-4. [DOI] [PubMed]
  • 280.Ponomarenko PM, Ponomarenko MP. Sequence-based prediction of transcription up-regulation by auxin in plants. J Bioinform Comput Biol. 2015;13:1540009. doi: 10.1142/S0219720015400090. [DOI] [PubMed] [Google Scholar]
  • 281.Drachkova IA, Shekhovtsov SV, Peltek SE, Ponomarenko PM, Arshinova TV, Ponomarenko MP, et al. Surface plasmon resonance study of the interaction between the human TATA box binding protein and the TATA element of the NOS2A gene promoter. Vavilov. J Genet Breed. 2012;16:391–396. [Google Scholar]
  • 282.Suslov VV, Ponomarenko PM, Ponomarenko MP, Drachkova IA, Arshinova TV, Savinkova LK, et al. TATA box polymorphisms in genes of commercial and laboratory animals and plants associated with selectively valuable traits. Russ J Genet. 2010;46:394–403. doi: 10.1134/S1022795410040022. [DOI] [PubMed] [Google Scholar]
  • 283.Suslov VV, Ponomarenko PM, Efimov VM, Savinkova LK, Ponomarenko MP, Kolchanov NA. SNPs in the HIV-1 TATA box and the AIDS pandemic. J Bioinform Comput Biol. 2010;8:607–625. doi: 10.1142/S0219720010004677. [DOI] [PubMed] [Google Scholar]
  • 284.Mironova VV, Omelyanchuk NA, Ponomarenko PM, Ponomarenko MP, Kolchanov NA. Specific/nonspecific binding of TBP to promoter DNA of the auxin response factor genes in plants correlated with ARFs function on gene transcription (activator/repressor) Dokl Biochem Biophys. 2010;433:191–196. doi: 10.1134/S1607672910040125. [DOI] [PubMed] [Google Scholar]
  • 285.Ponomarenko MP, Suslov VV, Gunbin KV, Ponomarenko PM, Vishnevsky OV, Kolchanov NA. Identification of the relationship between the variability of the expression of signaling pathway genes in the human brain and the affinity of TATA-binding protein to their promoters. Russ J Genet Appl Res. 2015;5:626–634. doi: 10.1134/S2079059715060088. [DOI] [Google Scholar]
  • 286.NCBI Resource Coordinators Database resources of the National Center for Biotechnology Information. Nucleic Acids Res. 2015;43:D6–17. doi: 10.1093/nar/gku1130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 287.Missala I, Kassner U, Steinhagen-Thiessen E. A systematic literature review of the association of lipoprotein(a) and autoimmune diseases and atherosclerosis. Int J Rheumatol. 2012;2012:480784. doi: 10.1155/2012/480784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 288.Burgner D, Rockett K, Ackerman H, Hull J, Usen S, Pinder M, et al. Haplotypic relationship between SNP and microsatellite markers at the NOS2A locus in two populations. Genes Immun. 2003;4:506-14 [DOI] [PubMed]
  • 289.Casal AJ, Sinclair VJ, Capponi AM, Nicod J, Huynh-Do U, Ferrari P. A novel mutation in the steroidogenic acute regulatory protein gene promoter leading to reduced promoter activity. J Mol Endocrinol. 2006;37:71–80. doi: 10.1677/jme.1.02082. [DOI] [PubMed] [Google Scholar]
  • 290.Matsunaga A, Sasaki J, Han H, Huang W, Kugi M, Koga T, et al. Compound heterozygosity for an apolipoprotein A1 gene promoter mutation and a structural nonsense mutation with apolipoprotein A1 deficiency. Arterioscler Thromb Vasc Biol. 1999;19:348–355. doi: 10.1161/01.ATV.19.2.348. [DOI] [PubMed] [Google Scholar]
  • 291.Plengpanich W, Le Goff W, Poolsuk S, Julia Z, Guerin M, Khovidhunkit W. CETP deficiency due to a novel mutation in the CETP gene promoter and its effect on cholesterol efflux and selective uptake into hepatocytes. Atherosclerosis. 2011;216:370–373. doi: 10.1016/j.atherosclerosis.2011.01.051. [DOI] [PubMed] [Google Scholar]
  • 292.Niemann S, Broom WJ, Brown RH., Jr Analysis of a genetic defect in the TATA box of the SOD1 gene in a patient with familial amyotrophic lateral sclerosis. Muscle Nerve. 2007;36:704–707. doi: 10.1002/mus.20855. [DOI] [PubMed] [Google Scholar]
  • 293.Watanabe M, Zingg BC, Mohrenweiser HW. Molecular analysis of a series of alleles in humans with reduced activity at the triosephosphate isomerase locus. Am J Hum Genet. 1996;58:308–316. [PMC free article] [PubMed] [Google Scholar]
  • 294.Vives-Corrons JL, Rubinson-Skala H, Mateo M, Estella J, Feliu E, Dreyfus JC. Triosephosphate isomerase deficiency with hemolytic anemia and severe neuromuscular disease: familial and biochemical studies of a case found in Spain. Hum Genet. 1978;42:171–180. doi: 10.1007/BF00283637. [DOI] [PubMed] [Google Scholar]
  • 295.Wirka RC, Gore S, van Wagoner DR, Arking DE, Lubitz SA, Lunetta KL, et al. A common connexin-40 gene promoter variant affects connexin-40 expression in human atria and is associated with atrial fibrillation. Circ Arrhythm Electrophysiol. 2011;4:87–93. doi: 10.1161/CIRCEP.110.959726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 296.Firouzi M, Ramanna H, Kok B, Jongsma HJ, Koeleman BP, Doevendans PA, et al. Association of human connexin40 gene polymorphisms with atrial vulnerability as a risk factor for idiopathic atrial fibrillation. Circ. Res. 2004;95:e29–e33. doi: 10.1161/01.RES.0000141134.64811.0a. [DOI] [PubMed] [Google Scholar]
  • 297.Clark IA, Rockett KA, Burgner D. Genes, nitric oxide and malaria in African children. Trends Parasitol. 2003;19:335–337. doi: 10.1016/S1471-4922(03)00147-8. [DOI] [PubMed] [Google Scholar]
  • 298.Gonzalez-Martinez JA, Moddel G, Ying Z, Prayson RA, Bingaman WE, Najm IM. Neuronal nitric oxide synthase expression in resected epileptic dysplastic neocortex. J Neurosurg. 2009;110:343–349. doi: 10.3171/2008.6.17608. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Additional file 1: (220.2KB, pdf)

Supplementary method. A sequence-based statistical estimate of the SNP-caused alteration in the affinity of TATA box binding protein (TBP) for the human gene promoter containing this SNP within its region [-70; -20]. (PDF 220 kb)

Additional file 2: (222KB, pdf)

Supplementary method. Keyword search in the PubMed database. (PDF 221 kb)

Additional file 3: (198.4KB, pdf)

Table S1. Clinically known dependences between reproductive potential and hereditary diseases whose SNP markers were predicted in this work. (PDF 198 kb)

Data Availability Statement

Web service SNP_TATA_Comparator is public available (URL=http://beehive.bionet.nsc.ru/cgi-bin/mgs/tatascan/start.pl).


Articles from BMC Genomics are provided here courtesy of BMC

RESOURCES