1
|
Poudel SB, Ruff RR, He Z, Dixit M, Yildirim G, Jayarathne H, Manchanayake DH, Basta-Pljakic J, Duran-Ortiz S, Schaffler MB, Kopchick JJ, Sadagurski M, Yakar S. The impact of inactivation of the GH/IGF axis during aging on healthspan. GeroScience 2024:10.1007/s11357-024-01426-3. [PMID: 39535693 DOI: 10.1007/s11357-024-01426-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024] Open
Abstract
Several mouse lines with congenital growth hormone (GH)/insulin-like growth factor-1 (IGF-1) axis disruption have shown improved health and extended lifespan. The current study investigated how inactivating this axis, specifically during aging, impacts the healthspan. We used a tamoxifen-inducible global GH receptor (GHR) knockout mouse model starting at 12 months and followed the mice until 24 months of age (iGHRKO12-24 mice). We found sex- and tissue-specific effects, with some being pro-aging and others anti-aging. Measuring an array of cytokines in serum revealed that inactivation of the GH/IGF-1 axis at 12 months did not affect systemic inflammation during aging. On the other hand, hypothalamic inflammation was significantly reduced in iGHRKO12-24 mice, evidenced by GFAP+ (glial fibrillary acidic protein, a marker of astrocytes) and Iba-1+ (a marker for microglia). Liver RNAseq analysis indicated feminization of the male transcriptome, with significant changes in the expression of monooxygenase, sulfotransferase, and solute-carrier-transporter gene clusters. Finally, we found impaired bone morphology, more pronounced in male iGHRKO12-24 mice and correlated with GH/IGF-1 inactivation onset age. We conclude that inhibiting the GH/IGF-1 axis during aging only partially preserves the beneficial healthspan effects observed with congenital GH deficiency.
Collapse
Affiliation(s)
- Sher Bahadur Poudel
- David B. Kriser Dental Center, Department of Molecular Pathobiology, New York University College of Dentistry, 345 East 24Th Street, New York, NY, 10010-4086, USA
| | - Ryan R Ruff
- David B. Kriser Dental Center, Department of Epidemiology and Health Promotion, New York University College of Dentistry, New York, NY, 10010-4086, USA
| | - Zhiming He
- David B. Kriser Dental Center, Department of Molecular Pathobiology, New York University College of Dentistry, 345 East 24Th Street, New York, NY, 10010-4086, USA
| | - Manisha Dixit
- David B. Kriser Dental Center, Department of Molecular Pathobiology, New York University College of Dentistry, 345 East 24Th Street, New York, NY, 10010-4086, USA
| | - Godze Yildirim
- David B. Kriser Dental Center, Department of Molecular Pathobiology, New York University College of Dentistry, 345 East 24Th Street, New York, NY, 10010-4086, USA
| | - Hashan Jayarathne
- Department of Biological Sciences, iBio (Integrative Biosciences Center), Integrative Biosciences Center, and Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, 48202, USA
| | - Dulmalika Herath Manchanayake
- Department of Biological Sciences, iBio (Integrative Biosciences Center), Integrative Biosciences Center, and Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, 48202, USA
| | - Jelena Basta-Pljakic
- Department of Biomedical Engineering, City College of New York, New York, NY, 10031, USA
| | - Silvana Duran-Ortiz
- Edison Biotechnology Institute and Department of Biomedical Sciences, Ohio University, Athens, OH, USA
| | - Mitchell B Schaffler
- Department of Biomedical Engineering, City College of New York, New York, NY, 10031, USA
| | - John J Kopchick
- Edison Biotechnology Institute and Department of Biomedical Sciences, Ohio University, Athens, OH, USA
| | - Marianna Sadagurski
- Department of Biological Sciences, iBio (Integrative Biosciences Center), Integrative Biosciences Center, and Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, 48202, USA
| | - Shoshana Yakar
- David B. Kriser Dental Center, Department of Molecular Pathobiology, New York University College of Dentistry, 345 East 24Th Street, New York, NY, 10010-4086, USA.
| |
Collapse
|
2
|
Iglesias-Carres L, Mas-Capdevila A, Bravo FI, Suárez M, Arola-Arnal A, Muguerza B. Sex Differences in the Absorption, Disposition, Metabolism, and Excretion of Grape Seed Proanthocyanidins in Prepubescent Rats. Mol Nutr Food Res 2024; 68:e2400399. [PMID: 39194387 DOI: 10.1002/mnfr.202400399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/25/2024] [Indexed: 08/29/2024]
Abstract
SCOPE The absorption, disposition, metabolism, and excretion (ADME) of phenolic compounds are key factors in determining their bioactivity. The group demonstrates that the ADME of a Grape Seed Proanthocyanidin Extract (GSPE) depends on sex in adult rats and specifically, methylated metabolites are only quantified in brain male adult rats. The aim of this study is to determine whether these differences exist before puberty. METHODS AND RESULTS Prepubescent 4-week-old male and female Wistar rats are administered GSPE at a dose of 1000 mg kg-1. Plasma, liver, mesenteric white adipose tissue (MWAT), brain, and kidneys are extracted excised 2 h after GSPE administration, and the PAs metabolite profile is studied by HPLC-ESI-MS/MS. Moreover, plasma estradiol and brain and liver catechol-O-methyltransferase (COMT) protein levels are also studied. Results showed that there are no differences in plasma and brain among sexes and only differences are observed in liver, MWAT, and kidney with individual metabolites. This agrees with the lack of differences in estradiol and COMT levels among sexes. However, the ADME of PAs metabolites is higher in male rats. CONCLUSIONS The results demonstrate lack of sex-dependence in metabolite profile in prepubescent rats, suggesting that sex differences in the metabolism of GSPE occur due to puberty.
Collapse
Affiliation(s)
- Lisard Iglesias-Carres
- Nutrigenomics Research Group, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, C/ Marcel·lí Domingo 1, Tarragona, 43007, Spain
| | - Anna Mas-Capdevila
- Nutrigenomics Research Group, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, C/ Marcel·lí Domingo 1, Tarragona, 43007, Spain
| | - Francisca I Bravo
- Nutrigenomics Research Group, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, C/ Marcel·lí Domingo 1, Tarragona, 43007, Spain
- Nutrigenomics Research Group, Institut d'Investigació Sanitària Pere Virgili, C/ Marcel·lí Domingo s/n, Tarragona, 43007, Spain
- Center of Environmental, Food and Toxicological Technology (TecnATox), University Rovira i Virgili, C/ Marcel·lí Domingo s/n, Tarragona, 43007, Spain
| | - Manuel Suárez
- Nutrigenomics Research Group, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, C/ Marcel·lí Domingo 1, Tarragona, 43007, Spain
- Nutrigenomics Research Group, Institut d'Investigació Sanitària Pere Virgili, C/ Marcel·lí Domingo s/n, Tarragona, 43007, Spain
- Center of Environmental, Food and Toxicological Technology (TecnATox), University Rovira i Virgili, C/ Marcel·lí Domingo s/n, Tarragona, 43007, Spain
| | - Anna Arola-Arnal
- Nutrigenomics Research Group, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, C/ Marcel·lí Domingo 1, Tarragona, 43007, Spain
- Nutrigenomics Research Group, Institut d'Investigació Sanitària Pere Virgili, C/ Marcel·lí Domingo s/n, Tarragona, 43007, Spain
- Center of Environmental, Food and Toxicological Technology (TecnATox), University Rovira i Virgili, C/ Marcel·lí Domingo s/n, Tarragona, 43007, Spain
| | - Begoña Muguerza
- Nutrigenomics Research Group, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, C/ Marcel·lí Domingo 1, Tarragona, 43007, Spain
- Nutrigenomics Research Group, Institut d'Investigació Sanitària Pere Virgili, C/ Marcel·lí Domingo s/n, Tarragona, 43007, Spain
- Center of Environmental, Food and Toxicological Technology (TecnATox), University Rovira i Virgili, C/ Marcel·lí Domingo s/n, Tarragona, 43007, Spain
| |
Collapse
|
3
|
Hayashi A, Terasaka S, Nukada Y, Kameyama A, Yamane M, Shioi R, Iwashita M, Hashizume K, Morita O. 4″-Sulfation Is the Major Metabolic Pathway of Epigallocatechin-3-gallate in Humans: Characterization of Metabolites, Enzymatic Analysis, and Pharmacokinetic Profiling. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:8264-8273. [PMID: 35786898 PMCID: PMC9284555 DOI: 10.1021/acs.jafc.2c02150] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Epigallocatechin-3-gallate (EGCG), a major green tea polyphenol, has beneficial effects on human health. This study aimed to elucidate the detailed EGCG sulfation process to better understand its phase II metabolism, a process required to maximize its health benefits. Results show that kinetic activity of sulfation in the human liver and intestinal cytosol is 2-fold and 60- to 300-fold higher than that of methylation and glucuronidation, respectively, suggesting sulfation as the key metabolic pathway. Moreover, SULT1A1 and SULT1A3 are responsible for sulfation in the liver and intestine, respectively. Additionally, our human ingestion study revealed that the concentration of EGCG-4″-sulfate in human plasma (Cmax: 177.9 nmol·L-1, AUC: 715.2 nmol·h·L-1) is equivalent to free EGCG (Cmax: 233.5 nmol·L-1, AUC: 664.1 nmol·h·L-1), suggesting that EGCG-4″-sulfate is the key metabolite. These findings indicate that sulfation is a crucial factor for improving EGCG bioavailability, while also advancing the understanding of the bioactivity and toxicity of EGCG.
Collapse
Affiliation(s)
- Akane Hayashi
- Safety
Science Laboratories, Kao Corporation, Tochigi 321-3497, Japan
- . Tel.: +81-285-68-7214
| | - Shimpei Terasaka
- Safety
Science Laboratories, Kao Corporation, Tochigi 321-3497, Japan
| | - Yuko Nukada
- Safety
Science Laboratories, Kao Corporation, Tochigi 321-3497, Japan
| | - Akiyo Kameyama
- Safety
Science Laboratories, Kao Corporation, Tochigi 321-3497, Japan
| | - Masayuki Yamane
- Safety
Science Laboratories, Kao Corporation, Tochigi 321-3497, Japan
| | - Ryuta Shioi
- Biological
Science Laboratories, Kao Corporation, Tochigi 321-3497, Japan
| | - Masazumi Iwashita
- Biological
Science Laboratories, Kao Corporation, Tochigi 321-3497, Japan
| | - Kohjiro Hashizume
- Biological
Science Laboratories, Kao Corporation, Tochigi 321-3497, Japan
| | - Osamu Morita
- Safety
Science Laboratories, Kao Corporation, Tochigi 321-3497, Japan
| |
Collapse
|
4
|
Gao Q, Chu Q, Liu M, Gao S, Xu Q, Zhou B. Functional variants in the promoter region of sulfotransferase 1C1 gene associated with estrogen degradation in gilts. Anim Sci J 2022; 93:e13738. [PMID: 35665986 DOI: 10.1111/asj.13738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 04/07/2022] [Accepted: 04/14/2022] [Indexed: 11/28/2022]
Abstract
Chinese indigenous Mi gilts have clearer estrus expression than European Large White gilts, and sulfotransferase 1C1 (SULT1C1) gene was differentially expressed between them. To investigate the differential expression mechanism of porcine SULT1C1 gene, we cloned its promoter region and predicted its activity. Six deletion expression vectors (P1, P2, P3, P4, P5, and P6) for the promoter of SULT1C1 gene were constructed. Vector P3 (-1084/+261) had the highest expression activity, whereas vector P4 (-642/+261) showed a reduced in promoter activity, which suggests that the core promoter region of SULT1C1 gene is located between -1084 bp and -642 bp. Two single nucleotide polymorphisms (SNPs), c. - 994 G > A (rs345070974) and c. - 946 G > A (rs337902009) were found in Mi and Large White gilts between -1100 and -661 bp, and the expression vectors with four haplotypes (GG, AA, GA, and AG) of two SNPs were constructed. The relative luciferase activity of vector with haplotype GG was the greatest among four vectors. These indicate that c. - 994 G > A and c. - 946 G > A are key mutations for promoter activity of SULT1C1 gene. Porcine SULT1C1 promoter with -994 G allele and -946 G allele significantly improved the gene expression level. It could be involved in different estrus expression between Large White and Mi gilts.
Collapse
Affiliation(s)
- Qinxue Gao
- Jiangsu Agri‐animal Husbandry Vocational College Taizhou China
| | - Qingpo Chu
- College of Animal Science and Technology Nanjing Agricultural University Nanjing China
| | - Mingzheng Liu
- College of Animal Science and Technology Nanjing Agricultural University Nanjing China
| | - Siyuan Gao
- College of Animal Science and Technology Nanjing Agricultural University Nanjing China
| | - Qinglei Xu
- College of Animal Science and Technology Nanjing Agricultural University Nanjing China
| | - Bo Zhou
- College of Animal Science and Technology Nanjing Agricultural University Nanjing China
| |
Collapse
|
5
|
Zhang A, Matsushita M, Zhang L, Wang H, Shi X, Gu H, Xia Z, Cui JY. Cadmium exposure modulates the gut-liver axis in an Alzheimer's disease mouse model. Commun Biol 2021; 4:1398. [PMID: 34912029 PMCID: PMC8674298 DOI: 10.1038/s42003-021-02898-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 11/16/2021] [Indexed: 12/17/2022] Open
Abstract
The human Apolipoprotein E4 (ApoE4) variant is the strongest known genetic risk factor for Alzheimer's disease (AD). Cadmium (Cd) has been shown to impair learning and memory at a greater extent in humanized ApoE4 knock-in (ApoE4-KI) mice as compared to ApoE3 (common allele)-KI mice. Here, we determined how cadmium interacts with ApoE4 gene variants to modify the gut-liver axis. Large intestinal content bacterial 16S rDNA sequencing, serum lipid metabolomics, and hepatic transcriptomics were analyzed in ApoE3- and ApoE4-KI mice orally exposed to vehicle, a low dose, or a high dose of Cd in drinking water. ApoE4-KI males had the most prominent changes in their gut microbiota, as well as a predicted down-regulation of many essential microbial pathways involved in nutrient and energy homeostasis. In the host liver, cadmium-exposed ApoE4-KI males had the most differentially regulated pathways; specifically, there was enrichment in several pathways involved in platelet activation and drug metabolism. In conclusion, Cd exposure profoundly modified the gut-liver axis in the most susceptible mouse strain to neurological damage namely the ApoE4-KI males, evidenced by an increase in microbial AD biomarkers, reduction in energy supply-related pathways in gut and blood, and an increase in hepatic pathways involved in inflammation and xenobiotic biotransformation.
Collapse
Affiliation(s)
- Angela Zhang
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| | - Megumi Matsushita
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| | - Liang Zhang
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| | - Hao Wang
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| | - Xiaojian Shi
- Arizona Metabolomics Laboratory, College of Health Solutions, Arizona State University, Phoenix, AZ, USA
| | - Haiwei Gu
- Arizona Metabolomics Laboratory, College of Health Solutions, Arizona State University, Phoenix, AZ, USA
| | - Zhengui Xia
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| | - Julia Yue Cui
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA.
| |
Collapse
|
6
|
Kurogi K, Manabe Y, Liu MC, Suiko M, Sakakibara Y. Molecular cloning and characterization of common marmoset SULT1C subfamily members that catalyze the sulfation of thyroid hormones. Biosci Biotechnol Biochem 2021; 85:2113-2120. [PMID: 34370005 PMCID: PMC8458394 DOI: 10.1093/bbb/zbab141] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 07/29/2021] [Indexed: 11/14/2022]
Abstract
Cytosolic sulfotransferase SULT1C subfamily is one of the most flexible gene subfamilies during mammalian evolution. The physiological functions of SULT1C enzymes still remain to be fully understood. In this study, common marmoset (Callithrix jacchus), a promising primate animal model, was used to investigate the functional relevance of the SULT1C subfamily. Gene database search revealed 3 intact SULT1C genes and a pseudogene in its genome. These 4 genes were named SULT1C1, SULT1C2, SULT1C3P, and SULT1C5, according to the sequence homology and gene location. Since SULT1C5 is the orthologous gene for human SULT1C2P, we propose, here, to revisit the designation of human SULT1C2P to SULT1C5P. Purified recombinant SULT1C enzymes showed sulfating activities toward a variety of xenobiotic compounds and thyroid hormones. Kinetic analysis revealed high catalytic activities of SULT1C1 and SULT1C5 for 3,3'-T2. It appears therefore that SULT1C isoforms may play a role in the thyroid hormone metabolism in common marmoset.
Collapse
Affiliation(s)
- Katsuhisa Kurogi
- Department of Biochemistry and Applied Biosciences, University of Miyazaki, Miyazaki, Japan
| | - Yoko Manabe
- Department of Biochemistry and Applied Biosciences, University of Miyazaki, Miyazaki, Japan
| | - Ming-Cheh Liu
- Department of Pharmacology, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, OH, USA
| | - Masahito Suiko
- Department of Biochemistry and Applied Biosciences, University of Miyazaki, Miyazaki, Japan
| | - Yoichi Sakakibara
- Department of Biochemistry and Applied Biosciences, University of Miyazaki, Miyazaki, Japan
| |
Collapse
|
7
|
Kojima M, Degawa M. Androgen-Dependent Differences in the Amounts of CYP mRNAs in the Pig Kidney. Biol Pharm Bull 2021; 44:1120-1128. [PMID: 34334497 DOI: 10.1248/bpb.b21-00333] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We previously reported androgen-dependent sex and breed differences in the amounts of mRNAs of CYP isoforms in the pig liver. To clarify whether there are such sex and breed differences in the kidney, we examined the amounts of several CYP mRNAs in the kidney using both sexes of 5-month-old Landrace, Meishan and/or their crossbred F1 (LM and ML) pigs. Significant sex differences in the amounts of several CYP mRNAs were found: male < female for CYP2A19 and CYP3A29; and male > female for CYP4A24/25 in all the breeds. Sex differences in the amount of CYP2B22 mRNA (male < female) and in CYP2C33 and CYP2C49 mRNAs (male > female) were also observed in all the breeds except Landrace pigs. Furthermore, a significant sex difference (male < female) in CYP3A46 mRNA was only found in LM and ML pigs. No significant sex differences were found in either Landrace or Meishan pigs for CYP1A1, CYP1A2 and CYP4B1 mRNAs. The amounts of CYP2C33 and CYP4A24/25 mRNAs in males were higher in Meishan pigs than in Landrace pigs. Additional experiments using pigs treated by castration and/or testosterone propionate indicated that sex and breed differences in the amounts of those CYP mRNAs were, at least in part, dependent on the levels of serum testosterone. Furthermore, the effects of androgen on the amounts of CYP mRNAs in the kidney did not necessarily correlate with those in the liver, suggesting that there is a tissue-selective factor responsible for the androgen-related expression of CYP genes.
Collapse
Affiliation(s)
- Misaki Kojima
- Animal Genome Unit, Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization (NARO)
| | - Masakuni Degawa
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka
| |
Collapse
|
8
|
A simple method to measure sulfonation in man using paracetamol as probe drug. Sci Rep 2021; 11:9036. [PMID: 33907224 PMCID: PMC8079418 DOI: 10.1038/s41598-021-88393-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 03/25/2021] [Indexed: 11/26/2022] Open
Abstract
Sulfotransferase enzymes (SULT) catalyse sulfoconjugation of drugs, as well as endogenous mediators, gut microbiota metabolites and environmental xenobiotics. To address the limited evidence on sulfonation activity from clinical research, we developed a clinical metabolic phenotyping method using paracetamol as a probe substrate. Our aim was to estimate sulfonation capability of phenolic compounds and study its intraindividual variability in man. A total of 36 healthy adult volunteers (12 men, 12 women and 12 women on oral contraceptives) received paracetamol in a 1 g-tablet formulation on three separate occasions. Paracetamol and its metabolites were measured in plasma and spot urine samples using liquid chromatography-high resolution mass spectrometry. A metabolic ratio (Paracetamol Sulfonation Index—PSI) was used to estimate phenol SULT activity. PSI showed low intraindividual variability, with a good correlation between values in plasma and spot urine samples. Urinary PSI was independent of factors not related to SULT activity, such as urine pH or eGFR. Gender and oral contraceptive intake had no impact on PSI. Our SULT phenotyping method is a simple non-invasive procedure requiring urine spot samples, using the safe and convenient drug paracetamol as a probe substrate, and with low intraindividual coefficient of variation. Although it will not give us mechanistic information, it will provide us an empirical measure of an individual’s sulfonator status. To the best of our knowledge, our method provides the first standardised in vivo empirical measure of an individual’s phenol sulfonation capability and of its intraindividual variability. EUDRA-CT 2016-001395-29, NCT03182595 June 9, 2017.
Collapse
|
9
|
Melia T, Waxman DJ. Genetic factors contributing to extensive variability of sex-specific hepatic gene expression in Diversity Outbred mice. PLoS One 2020; 15:e0242665. [PMID: 33264334 PMCID: PMC7710091 DOI: 10.1371/journal.pone.0242665] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 11/09/2020] [Indexed: 12/12/2022] Open
Abstract
Sex-specific transcription characterizes hundreds of genes in mouse liver, many implicated in sex-differential drug and lipid metabolism and disease susceptibility. While the regulation of liver sex differences by growth hormone-activated STAT5 is well established, little is known about autosomal genetic factors regulating the sex-specific liver transcriptome. Here we show, using genotyping and expression data from a large population of Diversity Outbred mice, that genetic factors work in tandem with growth hormone to control the individual variability of hundreds of sex-biased genes, including many long non-coding RNA genes. Significant associations between single nucleotide polymorphisms and sex-specific gene expression were identified as expression quantitative trait loci (eQTLs), many of which showed strong sex-dependent associations. Remarkably, autosomal genetic modifiers of sex-specific genes were found to account for more than 200 instances of gain or loss of sex-specificity across eight Diversity Outbred mouse founder strains. Sex-biased STAT5 binding sites and open chromatin regions with strain-specific variants were significantly enriched at eQTL regions regulating correspondingly sex-specific genes, supporting the proposed functional regulatory nature of the eQTL regions identified. Binding of the male-biased, growth hormone-regulated repressor BCL6 was most highly enriched at trans-eQTL regions controlling female-specific genes. Co-regulated gene clusters defined by overlapping eQTLs included sets of highly correlated genes from different chromosomes, further supporting trans-eQTL action. These findings elucidate how an unexpectedly large number of autosomal factors work in tandem with growth hormone signaling pathways to regulate the individual variability associated with sex differences in liver metabolism and disease.
Collapse
Affiliation(s)
- Tisha Melia
- Department of Biology and Bioinformatics Program, Boston University, Boston, Massachusetts, United States of America
| | - David J. Waxman
- Department of Biology and Bioinformatics Program, Boston University, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
10
|
Wang K, Chan MYC, Xu J, Li PS, Liu X, Lee AYF, Lee SST, Cheung WT. Male-biased fasting-induced changes in hepatic tauro-cholic acid and plasma cholesterol in Sult2a8-haplodeficient mice. Transgenic Res 2020; 29:499-510. [DOI: 10.1007/s11248-020-00215-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 09/10/2020] [Indexed: 12/16/2022]
|
11
|
Del Río JP, Molina S, Hidalgo-Lanussa O, Garcia-Segura LM, Barreto GE. Tibolone as Hormonal Therapy and Neuroprotective Agent. Trends Endocrinol Metab 2020; 31:742-759. [PMID: 32507541 DOI: 10.1016/j.tem.2020.04.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 04/24/2020] [Indexed: 12/11/2022]
Abstract
Tibolone (TIB), a selective tissue estrogenic activity regulator (STEAR) in clinical use by postmenopausal women, activates hormonal receptors in a tissue-specific manner. Estrogenic activity is present mostly in the brain, vagina, and bone, while the inactive forms predominate in the endometrium and breast. Conflicting literature on TIB's actions has been observed. While it has benefits for vasomotor symptoms, bone demineralization, and sexual health, a higher relative risk of hormone-sensitive cancer has been reported. In the brain, TIB can improve mood and cognition, neuroinflammation, and reactive gliosis. This review aims to discuss the systemic effects of TIB on peri- and post-menopausal women and its role in the brain. We suggest that TIB is a hormonal therapy with promising neuroprotective properties.
Collapse
Affiliation(s)
- Juan Pablo Del Río
- Reproductive Health Research Institute, Santiago, Chile; Translational Psychiatry Laboratory, Clínica Psiquiátrica Universitaria, Hospital Clínico, Universidad de Chile, Santiago, Chile; Millennium Nucleus to Improve the Mental Health of Adolescents and Youths (Imhay), Santiago, Chile
| | | | - Oscar Hidalgo-Lanussa
- Departamento de Nutrición y Bioquímica, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Luis Miguel Garcia-Segura
- Instituto Cajal, CSIC, Madrid, Spain; Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - George E Barreto
- Department of Biological Sciences, School of Natural Sciences, University of Limerick, Limerick, Ireland; Health Research Institute, University of Limerick, Limerick, Ireland.
| |
Collapse
|
12
|
Zhang X, Xu Y, Bai Q, Li X, Han J, Hou Y, Ji Y, Zhang Z. Inhibition of LXR signaling by SULT2B1b promotes liver regeneration after partial hepatectomy in mouse models of nonalcoholic fatty liver disease. Am J Physiol Gastrointest Liver Physiol 2020; 319:G87-G96. [PMID: 32475129 DOI: 10.1152/ajpgi.00380.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Hydroxysteroid sulfotransferase 2B1b (SULT2B1b) plays a critical role in hepatic energy homeostasis. Liver X receptors (LXRs) are implicated in multiple physiological functions, including the inhibition of hepatocyte proliferation and regulation of fatty acid and cholesterol metabolism. We have previously reported that SULT2B1b promotes hepatocyte proliferation by inactivating LXR signaling in vivo and in vitro, leading to our hypothesis that SULT2B1b promotes fatty liver regeneration. In the present study, female C57BL/6 and S129 mice were fed a high-fat diet for 8 wk to establish a nonalcoholic fatty liver disease (NAFLD) mouse model. 70% partial hepatectomy (PH) was performed to induce liver regeneration. Our experiments revealed that the SULT2B1b overexpression significantly promotes the regeneration of hepatocytes in NAFLD C57BL/6 mice after PH, increasing liver regrowth by 11% within 1 day, and then by 21%, 33%, and 24% by 2, 3, and 5 days post-PH, respectively. Compared with the wild-type NAFLD S129 mice, SULT2B1 deletion NAFLD S129 mice presented reduced hepatocyte regeneration at postoperative day 2, as verified by decreased liver regrowth (37.4% vs. 46.1%, P < 0.05) and the results of immunohistochemical staining, quantitative real-time polymerase chain reaction, and Western blot analysis. Moreover, LXRα signaling and SULT2B1b expression are highly correlated in the regeneration of NAFLD mouse liver; SULT2B1b overexpression suppresses LXRα signaling, while the LXRα-signaling agonist T0901317 blocks SULT2B1b-induced hepatocyte regeneration in NAFLD mouse liver. Thus, the upregulation of SULT2B1b may promote hepatocyte regeneration via the suppression of LXRα activation in NAFLD mice, providing a potential strategy for improving hepatic-steatosis-related liver regeneration disorders.NEW & NOTEWORTHY This study demonstrates for the first time that hydroxysteroid sulfotransferase 2B1b (SULT2B1b) overexpression promotes the regeneration of fatty liver after partial hepatectomy in mice with nonalcoholic fatty liver disease, while reducing triglyceride accumulation in the regenerative fatty liver. Liver X receptor signaling may be crucial in the SULT2B1b-mediated regeneration of fatty liver. Thus, SULT2B1b may be a potential target for treating hepatic steatosis-related liver regeneration disorders.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Pathology, Fudan University Zhongshan Hospital, Shanghai, China
| | - Yuyin Xu
- Department of Pathology, Fudan University Shanghai Cancer Centre, Shanghai, China.,Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Qianming Bai
- Department of Pathology, Fudan University Shanghai Cancer Centre, Shanghai, China
| | - Xiaobo Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jing Han
- Department of Pathology, Fudan University Zhongshan Hospital, Shanghai, China
| | - Yingyong Hou
- Department of Pathology, Fudan University Zhongshan Hospital, Shanghai, China
| | - Yuan Ji
- Department of Pathology, Fudan University Zhongshan Hospital, Shanghai, China
| | - Zhigang Zhang
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
13
|
Hausner EA, Elmore SA, Yang X. Overview of the Components of Cardiac Metabolism. Drug Metab Dispos 2019; 47:673-688. [PMID: 30967471 PMCID: PMC7333657 DOI: 10.1124/dmd.119.086611] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 03/26/2019] [Indexed: 12/20/2022] Open
Abstract
Metabolism in organs other than the liver and kidneys may play a significant role in how a specific organ responds to chemicals. The heart has metabolic capability for energy production and homeostasis. This homeostatic machinery can also process xenobiotics. Cardiac metabolism includes the expression of numerous organic anion transporters, organic cation transporters, organic carnitine (zwitterion) transporters, and ATP-binding cassette transporters. Expression and distribution of the transporters within the heart may vary, depending on the patient's age, disease, endocrine status, and various other factors. Several cytochrome P450 (P450) enzyme classes have been identified within the heart. The P450 hydroxylases and epoxygenases within the heart produce hydroxyeicosatetraneoic acids and epoxyeicosatrienoic acids, metabolites of arachidonic acid, which are critical in regulating homeostatic processes of the heart. The susceptibility of the cardiac P450 system to induction and inhibition from exogenous materials is an area of expanding knowledge, as are the metabolic processes of glucuronidation and sulfation in the heart. The susceptibility of various transcription factors and signaling pathways of the heart to disruption by xenobiotics is not fully characterized but is an area with implications for disruption of normal postnatal development, as well as modulation of adult cardiac health. There are knowledge gaps in the timelines of physiologic maturation and deterioration of cardiac metabolism. Cross-species characterization of cardiac-specific metabolism is needed for nonclinical work of optimum translational value to predict possible adverse effects, identify sensitive developmental windows for the design and conduct of informative nonclinical and clinical studies, and explore the possibilities of organ-specific therapeutics.
Collapse
Affiliation(s)
- Elizabeth A Hausner
- United States Food and Drug Administration, Center for Drug Evaluation and Research, Silver Spring, Maryland (E.A.H., X.Y.); and National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (S.A.E.)
| | - Susan A Elmore
- United States Food and Drug Administration, Center for Drug Evaluation and Research, Silver Spring, Maryland (E.A.H., X.Y.); and National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (S.A.E.)
| | - Xi Yang
- United States Food and Drug Administration, Center for Drug Evaluation and Research, Silver Spring, Maryland (E.A.H., X.Y.); and National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (S.A.E.)
| |
Collapse
|
14
|
Marinho AT, Miranda JP, Caixas U, Charneira C, Gonçalves-Dias C, Marques MM, Monteiro EC, Antunes AMM, Pereira SA. Singularities of nevirapine metabolism: from sex-dependent differences to idiosyncratic toxicity. Drug Metab Rev 2019; 51:76-90. [PMID: 30712401 DOI: 10.1080/03602532.2019.1577891] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Nevirapine (NVP) is a first-generation non-nucleoside reverse transcriptase inhibitor widely used for the treatment and prophylaxis of human immunodeficiency virus infection. The drug is taken throughout the patient's life and, due to the availability of an extended-release formulation, it is administered once daily. This antiretroviral is one of the scarce examples of drugs with prescription criteria based on sex, in order to prevent adverse reactions. The therapy with NVP has been associated with potentially life-threatening liver and idiosyncratic skin toxicity. Multiple evidence has emerged regarding the formation of electrophilic NVP metabolites as crucial for adverse idiosyncratic reactions. The formation of reactive metabolites that yield covalent adducts with proteins has been demonstrated in patients under NVP-based treatment. Interestingly, several pharmacogenetic- and sex-related factors associated with NVP toxicity can be mechanistically explained by an imbalance toward increased formation of NVP-derived reactive metabolites and/or impaired detoxification capability. Moreover, the haptenation of self-proteins by these reactive species provides a plausible link between NVP bioactivation and immunotoxicity, further supporting the relevance of this toxicokinetics hypothesis. In the current paper, we review the existing knowledge and recent developments on NVP metabolism and their relation to NVP toxicity.
Collapse
Affiliation(s)
- Aline T Marinho
- a CEDOC, Chronic Diseases Research Centre, NOVA Medical School/Faculdade de Ciências Médicas , Universidade NOVA de Lisboa , Lisboa , Portugal
| | - Joana P Miranda
- b Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy , Universidade de Lisboa , Lisboa , Portugal
| | - Umbelina Caixas
- a CEDOC, Chronic Diseases Research Centre, NOVA Medical School/Faculdade de Ciências Médicas , Universidade NOVA de Lisboa , Lisboa , Portugal.,c Centro Hospitalar de Lisboa Central (CHLC) , Lisboa , Portugal
| | - Catarina Charneira
- d Centro de Química Estrutural (CQE) , Instituto Superior Técnico, ULisboa , Lisboa , Portugal
| | - Clara Gonçalves-Dias
- a CEDOC, Chronic Diseases Research Centre, NOVA Medical School/Faculdade de Ciências Médicas , Universidade NOVA de Lisboa , Lisboa , Portugal
| | - M Matilde Marques
- d Centro de Química Estrutural (CQE) , Instituto Superior Técnico, ULisboa , Lisboa , Portugal
| | - Emília C Monteiro
- a CEDOC, Chronic Diseases Research Centre, NOVA Medical School/Faculdade de Ciências Médicas , Universidade NOVA de Lisboa , Lisboa , Portugal
| | - Alexandra M M Antunes
- d Centro de Química Estrutural (CQE) , Instituto Superior Técnico, ULisboa , Lisboa , Portugal
| | - Sofia A Pereira
- a CEDOC, Chronic Diseases Research Centre, NOVA Medical School/Faculdade de Ciências Médicas , Universidade NOVA de Lisboa , Lisboa , Portugal
| |
Collapse
|
15
|
Li Y, Chen Z, Paonessa JD, Meinl W, Bhattacharya A, Glatt H, Vouros P, Zhang Y. Strong impact of sulfotransferases on DNA adduct formation by 4-aminobiphenyl in bladder and liver in mice. Cancer Med 2018; 7:5604-5610. [PMID: 30306738 PMCID: PMC6246946 DOI: 10.1002/cam4.1779] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 08/21/2018] [Accepted: 08/22/2018] [Indexed: 02/03/2023] Open
Abstract
Bladder cancer risk is 3-4 times higher in men than women, but the reason is poorly understood. In mice, male bladder is also more susceptible than female bladder to 4-aminobiphenyl (ABP), a major human bladder carcinogen; however, female liver is more susceptible than male liver to ABP. We investigated the role of sulfotransferase (Sult) in gender-related bladder and liver susceptibility to ABP. Sulfation reactions of aromatic amine bladder carcinogens catalyzed by Sult may generate highly unstable and toxic metabolites. Therefore, liver Sult may decrease bladder exposure to carcinogens by promoting their toxic reactions in the liver. Notably, the expression of several liver Sults is suppressed by androgen in male mice. Here, we show that two Sults are critical for gender-related bladder susceptibility to ABP in mice. We measured tissue level of N-(deoxyguanosin-8-yl)-4-aminobiphenyl (dG-C8-ABP), a principal ABP-DNA adduct, as readout of tissue susceptibility to ABP. We identified Sutl1a1 and to a lesser extent Sult1d1 as Sults that promote dG-C8-ABP formation in hepatic cells. In mice, gender gap in bladder susceptibility to ABP was narrowed by knocking out Sult1a1 and was almost totally eliminated by knocking out both Sutl1a1 and Sult1d1. This was accompanied by dramatic decrease in ABP genotoxicity in the liver (>97%). These results show the strong impact of the Sults on bladder and liver susceptibility to a human carcinogen. Because liver expression of both Sult1a1 and Sutl1d1 is suppressed by androgen in male mice, our results suggest that androgen renders bladder more exposed to ABP in male mice by suppressing Sult-mediated ABP metabolism in liver, which increases bladder delivery of carcinogenic metabolites.
Collapse
Affiliation(s)
- Yun Li
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York.,Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Zhidan Chen
- Barnett Institute and Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts
| | - Joseph D Paonessa
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Walter Meinl
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany
| | - Arup Bhattacharya
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Hansruedi Glatt
- Department of Food Safety, German Federal Institute for Risk Assessment (BfR), Berlin, Germany
| | - Paul Vouros
- Barnett Institute and Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts
| | - Yuesheng Zhang
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York.,Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| |
Collapse
|
16
|
Put "gender glasses" on the effects of phenolic compounds on cardiovascular function and diseases. Eur J Nutr 2018; 57:2677-2691. [PMID: 29696400 DOI: 10.1007/s00394-018-1695-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 04/19/2018] [Indexed: 12/15/2022]
Abstract
INTRODUCTION The influence of sex and gender is particularly relevant in cardiovascular diseases (CVD) as well as in several aspects of drug pharmacodynamics and pharmacokinetics. Anatomical and physiological differences between the sexes may influence the activity of many drugs, including the possibility of their interaction with other drugs, bioactive compounds, foods and beverages. Phenolic compounds could interact with our organism at organ, cellular, and molecular levels triggering a preventive action against chronic diseases, including CVD. RESULTS This article will review the role of sex on the activity of these bioactive molecules, considering the existence of sex differences in oxidative stress. It describes the pharmacokinetics of phenolic compounds, their effects on vessels, on cardiovascular system, and during development, including the role of nuclear receptors and microbiota. CONCLUSIONS Although there is a large gap between the knowledge of the sex differences in the phenolic compounds' activity and safety, and the urgent need for more research, available data underlie the possibility that plant-derived phenolic compounds could differently influence the health of male and female subjects.
Collapse
|
17
|
Feminization of Male Mouse Liver by Persistent Growth Hormone Stimulation: Activation of Sex-Biased Transcriptional Networks and Dynamic Changes in Chromatin States. Mol Cell Biol 2017; 37:MCB.00301-17. [PMID: 28694329 PMCID: PMC5599723 DOI: 10.1128/mcb.00301-17] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 07/03/2017] [Indexed: 12/12/2022] Open
Abstract
Sex-dependent pituitary growth hormone (GH) secretory profiles-pulsatile in males and persistent in females-regulate the sex-biased, STAT5-dependent expression of hundreds of genes in mouse liver, imparting sex differences in hepatic drug/lipid metabolism and disease risk. Here, we examine transcriptional and epigenetic changes induced by continuous GH infusion (cGH) in male mice, which rapidly feminizes the temporal profile of liver STAT5 activity. cGH repressed 86% of male-biased genes and induced 68% of female-biased genes within 4 days; however, several highly female-specific genes showed weak or no feminization, even after 14 days of cGH treatment. Female-biased genes already in an active chromatin state in male liver generally showed early cGH responses; genes in an inactive chromatin state often responded late. Early cGH-responsive genes included those encoding two GH/STAT5-regulated transcriptional repressors: male-biased BCL6, which was repressed, and female-specific CUX2, which was induced. Male-biased genes activated by STAT5 and/or repressed by CUX2 were enriched for early cGH repression. Female-biased BCL6 targets were enriched for early cGH derepression. Changes in sex-specific chromatin accessibility and histone modifications accompanied these cGH-induced sex-biased gene expression changes. Thus, the temporal, sex-biased gene responses to persistent GH stimulation are dictated by GH/STAT5-regulated transcription factors arranged in a hierarchical network and by the dynamics of changes in sex-biased epigenetic states.
Collapse
|
18
|
Marto N, Morello J, Monteiro EC, Pereira SA. Implications of sulfotransferase activity in interindividual variability in drug response: clinical perspective on current knowledge. Drug Metab Rev 2017; 49:357-371. [PMID: 28554218 DOI: 10.1080/03602532.2017.1335749] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The interindividual variability in drug response is a major issue in clinical practice and in drug development. Sulfoconjugation is an important Phase II reaction catalyzed by cytosolic sulfotransferases (SULTs), playing a major role in homeostatic functions, xenobiotic detoxification, and carcinogen bioactivation. SULT display wide interindividual variability, explained only partially by genetic variation, suggesting that other non-genetic, epigenetic, and environmental influences could be major determinants of variability in SULT activity. This review focuses on the factors known to influence SULT variability in expression and activity and the available evidence regarding the impact of SULT variability on drug response.
Collapse
Affiliation(s)
- Natalia Marto
- a CEDOC, Chronic Diseases Research Centre, NOVA Medical School Faculdade de Ciências Médicas, Universidade NOVA de Lisboa , Lisboa , Portugal.,b Department of Internal Medicine , Hospital da Luz , Lisboa , Portugal
| | - Judit Morello
- a CEDOC, Chronic Diseases Research Centre, NOVA Medical School Faculdade de Ciências Médicas, Universidade NOVA de Lisboa , Lisboa , Portugal
| | - Emilia C Monteiro
- a CEDOC, Chronic Diseases Research Centre, NOVA Medical School Faculdade de Ciências Médicas, Universidade NOVA de Lisboa , Lisboa , Portugal
| | - Sofia A Pereira
- a CEDOC, Chronic Diseases Research Centre, NOVA Medical School Faculdade de Ciências Médicas, Universidade NOVA de Lisboa , Lisboa , Portugal
| |
Collapse
|
19
|
Connerney J, Lau-Corona D, Rampersaud A, Waxman DJ. Activation of Male Liver Chromatin Accessibility and STAT5-Dependent Gene Transcription by Plasma Growth Hormone Pulses. Endocrinology 2017; 158:1386-1405. [PMID: 28323953 PMCID: PMC6283433 DOI: 10.1210/en.2017-00060] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 02/10/2017] [Indexed: 02/07/2023]
Abstract
Sex differences in pituitary growth hormone (GH) secretion (pulsatile in males vs near continuous/persistent in females) impart sex-dependent expression to hundreds of genes in adult mouse liver. Signal transducer and activator of transcription (STAT) 5, a GH-activated transcription factor that is essential for liver sexual dimorphism, is dynamically activated in direct response to each male plasma GH pulse. However, the impact of GH-induced STAT5 pulses on liver chromatin accessibility and downstream transcriptional events is unknown. In this study, we investigated the impact of a single pulse of GH given to hypophysectomized mice on local liver chromatin accessibility (DNase hypersensitive site analysis), transcription rates (heterogeneous nuclear RNA analysis), and gene expression (quantitative polymerase chain reaction and RNA sequencing) determined 30, 90, or 240 minutes later. The STAT5-dependent but sex-independent early GH response genes Igf1 and Cish showed rapid, GH pulse-induced increases in chromatin accessibility and gene transcription, reversing the effects of hypophysectomy. Rapid increases in liver chromatin accessibility and transcriptional activity were also induced in hypophysectomized male mice for some (Ces2b, Ugt2b38) but not for other liver STAT5-dependent male-biased genes (Cyp7b1). Moreover, in pituitary-intact male mice, Igf1, Cish, Ces2b, and Ugt2b38 all showed remarkable cycles of chromatin opening and closing, as well as associated cycles of induced gene transcription, which closely followed each endogenous pulse of liver STAT5 activity. Thus, the endogenous rhythms of male plasma GH pulsation dynamically open and then close liver chromatin at discrete, localized regulatory sites in temporal association with transcriptional activation of Igf1, Cish, and a subset of STAT5-dependent male-biased genes.
Collapse
Affiliation(s)
- Jeannette Connerney
- Department of Biology and Bioinformatics Program, Boston University, Boston, Massachusetts 02215
| | - Dana Lau-Corona
- Department of Biology and Bioinformatics Program, Boston University, Boston, Massachusetts 02215
| | - Andy Rampersaud
- Department of Biology and Bioinformatics Program, Boston University, Boston, Massachusetts 02215
| | - David J Waxman
- Department of Biology and Bioinformatics Program, Boston University, Boston, Massachusetts 02215
| |
Collapse
|
20
|
Feng L, Yuen YL, Xu J, Liu X, Chan MYC, Wang K, Fong WP, Cheung WT, Lee SST. Identification and characterization of a novel PPARα-regulated and 7α-hydroxyl bile acid-preferring cytosolic sulfotransferase mL-STL (Sult2a8). J Lipid Res 2017; 58:1114-1131. [PMID: 28442498 DOI: 10.1194/jlr.m074302] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 04/19/2017] [Indexed: 12/25/2022] Open
Abstract
PPARα has been known to play a pivotal role in orchestrating lipid, glucose, and amino acid metabolism via transcriptional regulation of its target gene expression during energy deprivation. Recent evidence has also suggested that PPARα is involved in bile acid metabolism, but how PPARα modulates the homeostasis of bile acids during fasting is still not clear. In a mechanistic study aiming to dissect the spectrum of PPARα target genes involved in metabolic response to fasting, we identified a novel mouse gene (herein named mL-STL for mouse liver-sulfotransferase-like) that shared extensive homology with the Sult2a subfamily of a superfamily of cytosolic sulfotransferases, implying its potential function in sulfonation. The mL-STL gene expressed predominantly in liver in fed state, but PPARα was required to sustain its expression during fasting, suggesting a critical role of PPARα in regulating the mL-STL-mediated sulfonation during fasting. Functional studies using recombinant His-tagged mL-STL protein revealed its narrow sulfonating activities toward 7α-hydroxyl primary bile acids, including cholic acid, chenodeoxycholic acid, and α-muricholic acid, and thus suggesting that mL-STL may be the major hepatic bile acid sulfonating enzyme in mice. Together, these studies identified a novel PPARα-dependent gene and uncovered a new role of PPARα as being an essential regulator in bile acid biotransformation via sulfonation during fasting.
Collapse
Affiliation(s)
- Lu Feng
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR
| | - Yee-Lok Yuen
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR
| | - Jian Xu
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR
| | - Xing Liu
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR
| | - Martin Yan-Chun Chan
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR
| | - Kai Wang
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR
| | - Wing-Ping Fong
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR
| | - Wing-Tai Cheung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR
| | - Susanna Sau-Tuen Lee
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR
| |
Collapse
|
21
|
Soukup ST, Müller DR, Kurrat A, Diel P, Kulling SE. Influence of testosterone on phase II metabolism and availability of soy isoflavones in male Wistar rats. Arch Toxicol 2017; 91:1649-1661. [PMID: 27743010 DOI: 10.1007/s00204-016-1853-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 09/15/2016] [Indexed: 01/16/2023]
Abstract
Genistein and daidzein are the main isoflavones in soy. Their potential beneficial or adverse effects in males like the prevention of prostate cancer or the impact on reproductive functions are controversially discussed. Major determinants of their bioactivity are the absorption and biotransformation of isoflavones. In this study, we focused on the influence of testosterone on plasma availability and phase II metabolism of isoflavones. Male Wistar rats, receiving an isoflavones rich diet, were randomized into three groups: Two groups were orchiectomized (ORX) at postnatal day (PND) 80 and treated for 11 days with testosterone propionate (TP) (ORX TP group) or a vehicle (ORX group) after a 7 days lasting hormonal decline. The third group served as control and remained intact. Rats were sacrificed at PND 98. ORX rats had reduced isoflavones plasma levels. Differently regulated mRNA expressions of transporters relevant for transport of phase II metabolites in liver and kidney may be responsible for this reduction, more precisely Slc10a1 and Slc21a1 in kidney as well as Slc22a8 in liver. While main phase II metabolites in intact rats were disulfates and sulfoglucuronides, the amount of sulfate conjugates was significantly diminished by ORX. In accordance with that, mRNA expression of different sulfotransferases was reduced in liver by ORX. The observed effects could be almost restored by TP treatment. In conclusion, testosterone, and likely further androgens, has a huge impact on phase II metabolism and availability of isoflavones by influencing the expression of different sulfotransferases and transporters.
Collapse
Affiliation(s)
- Sebastian T Soukup
- Department of Safety and Quality of Fruit and Vegetables, Max Rubner-Institut, Haid-und-Neu-Straße 9, 76131, Karlsruhe, Germany
| | - Dennis R Müller
- Department of Molecular and Cellular Sports Medicine, German Sport University Cologne, Cologne, Germany
| | - Anne Kurrat
- Department of Molecular and Cellular Sports Medicine, German Sport University Cologne, Cologne, Germany
| | - Patrick Diel
- Department of Molecular and Cellular Sports Medicine, German Sport University Cologne, Cologne, Germany
| | - Sabine E Kulling
- Department of Safety and Quality of Fruit and Vegetables, Max Rubner-Institut, Haid-und-Neu-Straße 9, 76131, Karlsruhe, Germany.
| |
Collapse
|
22
|
Guo Y, Cui JY, Lu H, Klaassen CD. Effect of nine diets on mRNAs of phase-II conjugation enzymes in livers of mice. Xenobiotica 2016; 47:645-654. [PMID: 27686132 DOI: 10.1080/00498254.2016.1213926] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
1. Phase-II enzymes are important in metabolizing many xenobiotics including prescription drugs and chemical carcinogens. Whereas it is known that diet can alter the expression of phase-II conjugation enzymes, the previous studies are limited in using only two or three diets and examining only a few enzymes. 2. Adult male C57BL6 mice were fed one of nine diets for 3 weeks. Of the 87 genes encoding major hepatic phase-II enzymes, approximately one-half (43) were altered by at least one diet. Diet restriction altered the hepatic expression of the most genes encoding phase-II enzymes (27), followed by lab chow (15), atherogenic diet (13), high-fat diet (10), western diet (7), high-fructose diet (5), and essential fatty acid-deficient diet (3), whereas the low n-3 fatty acid diet had no effect on the hepatic expression of these phase-II enzymes. 3. This comprehensive study provides detailed information on which conjugation enzymes are changed by these diets, and these data can be used to further investigate the mechanism for these changes in messenger RNAs, and whether these changes result in alterations in enzyme activity and drug action.
Collapse
Affiliation(s)
- Ying Guo
- a Department of Internal Medicine , University of Kansas Medical Center , Kansas City, KS , USA.,b Department of Clinical Pharmacology , Xiangya Hospital, Central South University , Changsha , P.R. China , and
| | - Julia Yue Cui
- a Department of Internal Medicine , University of Kansas Medical Center , Kansas City, KS , USA
| | - Hong Lu
- a Department of Internal Medicine , University of Kansas Medical Center , Kansas City, KS , USA.,c Department of Pharmacology , SUNY Upstate Medical University , Syracuse, NY , USA
| | - Curtis D Klaassen
- a Department of Internal Medicine , University of Kansas Medical Center , Kansas City, KS , USA
| |
Collapse
|
23
|
Crosstalk of HNF4 α with extracellular and intracellular signaling pathways in the regulation of hepatic metabolism of drugs and lipids. Acta Pharm Sin B 2016; 6:393-408. [PMID: 27709008 PMCID: PMC5045537 DOI: 10.1016/j.apsb.2016.07.003] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 05/05/2016] [Accepted: 05/11/2016] [Indexed: 12/15/2022] Open
Abstract
The liver is essential for survival due to its critical role in the regulation of metabolic homeostasis. Metabolism of xenobiotics, such as environmental chemicals and drugs by the liver protects us from toxic effects of these xenobiotics, whereas metabolism of cholesterol, bile acids (BAs), lipids, and glucose provide key building blocks and nutrients to promote the growth or maintain the survival of the organism. As a well-established master regulator of liver development and function, hepatocyte nuclear factor 4 alpha (HNF4α) plays a critical role in regulating a large number of key genes essential for the metabolism of xenobiotics, metabolic wastes, and nutrients. The expression and activity of HNF4α is regulated by diverse hormonal and signaling pathways such as growth hormone, glucocorticoids, thyroid hormone, insulin, transforming growth factor-β, estrogen, and cytokines. HNF4α appears to play a central role in orchestrating the transduction of extracellular hormonal signaling and intracellular stress/nutritional signaling onto transcriptional changes in the liver. There have been a few reviews on the regulation of drug metabolism, lipid metabolism, cell proliferation, and inflammation by HNF4α. However, the knowledge on how the expression and transcriptional activity of HNF4α is modulated remains scattered. Herein I provide comprehensive review on the regulation of expression and transcriptional activity of HNF4α, and how HNF4α crosstalks with diverse extracellular and intracellular signaling pathways to regulate genes essential in liver pathophysiology.
Collapse
|
24
|
Selwyn FP, Cheng SL, Bammler TK, Prasad B, Vrana M, Klaassen C, Cui JY. Developmental Regulation of Drug-Processing Genes in Livers of Germ-Free Mice. Toxicol Sci 2015; 147:84-103. [PMID: 26032512 DOI: 10.1093/toxsci/kfv110] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Very little is known about the effect of gut microbiota on the ontogeny of drug-processing genes (DPGs) in liver. In this study, livers were harvested from conventional (CV) and germ-free (GF) male and female mice from 1 to 90 days of age. RNA-Seq in livers of 90-day-old male mice showed that xenobiotic metabolism was the most downregulated pathway within the mRNA transcriptome in absence of intestinal bacteria. In male livers, the mRNAs of 67 critical DPGs partitioned into 4 developmental patterns (real-time-quantitative polymerase chain reaction): Pattern-1 gradually increased to adult levels in livers of CV mice and were downregulated in livers of GF mice, as exemplified by the major drug-metabolizing enzymes cytochrome 3a (Cyp3a) family, which are prototypical pregnane X receptor (PXR)-target genes. Genes in Pattern-2 include Cyp1a2 (aryl hydrocarbon receptor-target gene), Cyp2c family, and Cyp2e1, which were all upregulated mainly at 90 days of age; as well as the peroxisome proliferator-activated receptor α (PPARα)-target genes Cyp4a family and Aldh3a2, which were upregulated not only in 90-days adult age, but also between neonatal and adolescent ages (from 1 to 30 days of age). Genes in Pattern-3 were enriched predominantly in livers of 15-day-old mice, among which the sterol-efflux transporter dimers Abcg5/Abcg8 were downregulated in GF mice. Genes in Pattern-4 were neonatal-enriched, among which the transporter Octn1 mRNA tended to be lower in GF mice at younger ages but higher in adult GF mice as compared with age-matched CV mice. Protein assays confirmed the downregulation of the PXR-target gene Cyp3a protein (Western-blot and liquid chromatography tandem mass spectroscopy), and decreased Cyp3a enzyme activities in male GF livers. Increased microsomal-Cyp4a proteins and nuclear-PPARα were also observed in male GF livers. Interestingly, in contrast to male livers, the mRNAs of Cyp2c or Cyp4a were not readily upregulated in female GF livers approaching adult age, suggesting the maturation of female-specific hormones interferes with the interactions between intestinal microbiota and DPG ontogeny. In conclusion, intestinal microbiota markedly impacts the ontogeny of many hepatic DPGs in a gender-specific manner.
Collapse
Affiliation(s)
| | | | - Theo K Bammler
- *Department of Environmental & Occupational Health Sciences and
| | - Bhagwat Prasad
- Department of Pharmaceutics, University of Washington, Seattle, Washington 98195
| | - Marc Vrana
- Department of Pharmaceutics, University of Washington, Seattle, Washington 98195
| | - Curtis Klaassen
- *Department of Environmental & Occupational Health Sciences and
| | - Julia Yue Cui
- *Department of Environmental & Occupational Health Sciences and
| |
Collapse
|
25
|
Lopes BA, Emerenciano M, Gonçalves BAA, Vieira TM, Rossini A, Pombo-de-Oliveira MS. Polymorphisms in CYP1B1, CYP3A5, GSTT1, and SULT1A1 Are Associated with Early Age Acute Leukemia. PLoS One 2015; 10:e0127308. [PMID: 25992585 PMCID: PMC4436276 DOI: 10.1371/journal.pone.0127308] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 04/14/2015] [Indexed: 01/01/2023] Open
Abstract
Based on observational studies, early age leukemia (EAL) was associated with maternal hormone exposure during pregnancy. We studied the association between genetic polymorphisms of estrogen metabolism and EAL. Using data from the Brazilian Collaborative Study Group of Infant Acute Leukemia (2000–2012), 350 cases and 404 age-matched controls and 134 mothers of cases and controls were genotyped to explore polymorphisms in genes of the estrogen metabolism pathway: CYP1B1 (c.1294C>G, rs1056836), CYP3A4 (c.-392A>G, rs2740574), CYP3A5 (c.219-237G>A, rs776746), GSTM1/GSTT1 deletions, and SULT1A1 (c.638G>A, rs9282861; and c.667A>G, rs1801030). Logistic regression was used to calculate the odds ratios (OR) with 95% confidence intervals (CIs), and unconditional logistic regression was used to estimate adjusted odds ratios (aORs) by ethnicity. Because of multiple testing, p values < 0.01 were significant after Bonferroni correction. SULT1A1 (c.638G>A) was associated to infant acute lymphoblastic leukemia and acute myeloid leukemia (AML) risk in males (additive model: aOR = 0.52; 95% CI: 0.29–0.95, p = 0.03; dominant model: aOR = 2.18; 95% CI: 1.17–4.05, p = 0.01, respectively). CYP1B1 polymorphism was associated with a decreased risk of AML either for non-white or female children (additive model: OR = 0.24; 95% CI: 0.08–0.76, p < 0.01; additive model: aOR = 0.27; 95% CI: 0.08–0.89, p = 0.03, respectively). Since polymorphisms of Cytochrome P450 genes presented gender-specific risk associations, we also investigated their expression. CYP1B1 was not expressed in 57.1% of EAL cases, and its expression varied by genotype, gender, and leukemia subtype. Maternal-fetal GSTT1 null genotype was associated with risk of EAL. This study shows that polymorphisms in genes of estrogen metabolism confer genetic susceptibility to EAL, mainly in males, and maternal susceptibility genes modify the risk for developing EAL in newborns.
Collapse
Affiliation(s)
- Bruno Almeida Lopes
- Pediatric Hematology-Oncology Program, Research Center, Instituto Nacional de Câncer, Rio de Janeiro, RJ, Brazil
| | - Mariana Emerenciano
- Pediatric Hematology-Oncology Program, Research Center, Instituto Nacional de Câncer, Rio de Janeiro, RJ, Brazil
| | - Bruno Alves Aguiar Gonçalves
- Pediatric Hematology-Oncology Program, Research Center, Instituto Nacional de Câncer, Rio de Janeiro, RJ, Brazil
| | - Tállita Meciany Vieira
- Pediatric Hematology-Oncology Program, Research Center, Instituto Nacional de Câncer, Rio de Janeiro, RJ, Brazil
| | - Ana Rossini
- Department of Biochemistry, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Maria S. Pombo-de-Oliveira
- Pediatric Hematology-Oncology Program, Research Center, Instituto Nacional de Câncer, Rio de Janeiro, RJ, Brazil
- * E-mail:
| |
Collapse
|
26
|
Ruiz ML, Mottino AD, Catania VA, Vore M. Hormonal regulation of hepatic drug biotransformation and transport systems. Compr Physiol 2014; 3:1721-40. [PMID: 24265243 DOI: 10.1002/cphy.c130018] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The human body is constantly exposed to many xenobiotics including environmental pollutants, food additives, therapeutic drugs, etc. The liver is considered the primary site for drug metabolism and elimination pathways, consisting in uptake, phase I and II reactions, and efflux processes, usually acting in this same order. Modulation of biotransformation and disposition of drugs of clinical application has important therapeutic and toxicological implications. We here provide a compilation and analysis of relevant, more recent literature reporting hormonal regulation of hepatic drug biotransformation and transport systems. We provide additional information on the effect of hormones that tentatively explain differences between sexes. A brief discussion on discrepancies between experimental models and species, as well as a link between gender-related differences and the hormonal mechanism explaining such differences, is also presented. Finally, we include a comment on the pathophysiological, toxicological, and pharmacological relevance of these regulations.
Collapse
Affiliation(s)
- María L Ruiz
- Institute of Experimental Physiology, National University of Rosario, Rosario, Argentina
| | | | | | | |
Collapse
|
27
|
Sidharthan NP, Butcher NJ, Mitchell DJ, Minchin RF. Expression of the orphan cytosolic sulfotransferase SULT4A1 and its major splice variant in human tissues and cells: dimerization, degradation and polyubiquitination. PLoS One 2014; 9:e101520. [PMID: 24988429 PMCID: PMC4079325 DOI: 10.1371/journal.pone.0101520] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 06/08/2014] [Indexed: 11/19/2022] Open
Abstract
The cytosolic sulfotransferase SULT4A1 is highly conserved between mammalian species but its function remains unknown. Polymorphisms in the SULT4A1 gene have been linked to susceptibility to schizophrenia. There are 2 major SULT4A1 transcripts in humans, one that encodes full length protein (wild-type) and one that encodes a truncated protein (variant). Here, we investigated the expression of SULT4A1 in human tissues by RT-PCR and found the wild-type mRNA to be expressed mainly in the brain, gastrointestinal tract and prostate while the splice variant was more widely expressed. In human cell-lines, the wild-type transcript was found in neuronal cells, but the variant transcript was expressed in nearly all other lines examined. Western blot analysis only identified SULT4A1 protein in cells that expressed the wild-type mRNA. No variant protein was detected in cells that expressed the variant mRNA. Ectopically expressed full length SULT4A1 protein was stable while the truncated protein was not, having a half-life of approximately 3 hr. SULT4A1 was also shown to homodimerize, consistent with other SULTs that contain the consensus dimerization motif. Mutation of the dimerization motif resulted in a monomeric form of SULT4A1 that was rapidly degraded by polyubiquitination on the lysine located within the dimerization motif. These results show that SULT4A1 is widely expressed in human tissues, but mostly as a splice variant that produces a rapidly degraded protein. Dimerization protects the protein from degradation. Since many other cytosolic sulfotransferases possess the conserved lysine within the dimerization motif, homodimerization may serve, in part, to stabilize these enzymes in vivo.
Collapse
Affiliation(s)
- Neelima P. Sidharthan
- Laboratory for Molecular and Cellular Pharmacology, School of Biomedical Sciences, University of Queensland, Brisbane, Australia
| | - Neville J. Butcher
- Laboratory for Molecular and Cellular Pharmacology, School of Biomedical Sciences, University of Queensland, Brisbane, Australia
| | - Deanne J. Mitchell
- Laboratory for Molecular and Cellular Pharmacology, School of Biomedical Sciences, University of Queensland, Brisbane, Australia
| | - Rodney F. Minchin
- Laboratory for Molecular and Cellular Pharmacology, School of Biomedical Sciences, University of Queensland, Brisbane, Australia
- * E-mail:
| |
Collapse
|
28
|
Li W, Ning M, Koh KH, Kim H, Jeong H. 17β-Estradiol induces sulfotransferase 2A1 expression through estrogen receptor α. Drug Metab Dispos 2014; 42:796-802. [PMID: 24492894 PMCID: PMC3965899 DOI: 10.1124/dmd.113.055178] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 02/03/2014] [Indexed: 01/02/2023] Open
Abstract
Sulfotransferase (SULT) 2A1 catalyzes sulfonation of drugs and endogenous compounds and plays an important role in xenobiotic metabolism as well as in the maintenance of steroid and lipid homeostasis. A recent study showed that 17β-estradiol (E2) increases the mRNA levels of SULT2A1 in human hepatocytes. Here we report the underlying molecular mechanisms. E2 enhanced SULT2A1 expression in human hepatocytes and HepG2-ER cells (HepG2 stably expressing ERα). SULT2A1 induction by E2 was abrogated by antiestrogen ICI 182,780, indicating a key role of ERα in the induction. Results from deletion and mutation assays of SULT2A1 promoter revealed three cis-elements located within -257/+140 region of SULT2A1 that are potentially responsible for the induction. Chromatin immunoprecipitation assay verified the recruitment of ERα to the promoter region. Electrophoretic mobility shift assays revealed that AP-1 proteins bind to one of the cis-elements. Interestingly, SULT2A1 promoter assays using ERα mutants revealed that the DNA-binding domain of ERα is indispensable for SULT2A1 induction by E2, suggesting that direct ERα binding to the SULT2A1 promoter is also necessary for the induction. Taken together, our results indicate that E2 enhances SULT2A1 expression by both the classical and nonclassical mechanisms of ERα action.
Collapse
Affiliation(s)
- Wei Li
- Department of Pharmacy Practice (W.L., K.H.K., H.K., H.J.) and Department of Biopharmaceutical Sciences (M.N., H.J.), College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois; and Medical College, Yangzhou University, Yangzhou, Jiangsu, China (W.L.)
| | | | | | | | | |
Collapse
|
29
|
Kojima M, Degawa M. Sex Differences in the Constitutive Gene Expression of Sulfotransferases and UDP-glucuronosyltransferases in the Pig Liver: Androgen-mediated Regulation. Drug Metab Pharmacokinet 2014; 29:192-7. [DOI: 10.2133/dmpk.dmpk-13-rg-086] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
30
|
Fu ZD, Klaassen CD. Short-term calorie restriction feminizes the mRNA profiles of drug metabolizing enzymes and transporters in livers of mice. Toxicol Appl Pharmacol 2013; 274:137-46. [PMID: 24240088 DOI: 10.1016/j.taap.2013.11.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 10/15/2013] [Accepted: 11/04/2013] [Indexed: 01/22/2023]
Abstract
Calorie restriction (CR) is one of the most effective anti-aging interventions in mammals. A modern theory suggests that aging results from a decline in detoxification capabilities and thus accumulation of damaged macromolecules. The present study aimed to determine how short-term CR alters mRNA profiles of genes that encode metabolism and detoxification machinery in the liver. Male C57BL/6 mice were fed CR (0, 15, 30, or 40%) diets for one month, followed by mRNA quantification of 98 xenobiotic processing genes (XPGs) in the liver, including 7 uptake transporters, 39 phase-I enzymes, 37 phase-II enzymes, 10 efflux transporters, and 5 transcription factors. In general, 15% CR did not alter mRNAs of most XPGs, whereas 30 and 40% CR altered over half of the XPGs (32 increased and 29 decreased). CR up-regulated some phase-I enzymes (fold increase), such as Cyp4a14 (12), Por (2.3), Nqo1 (1.4), Fmo2 (5.4), and Fmo3 (346), and numerous number of phase-II enzymes, such as Sult1a1 (1.2), Sult1d1 (2.0), Sult1e1 (33), Sult3a1 (2.2), Gsta4 (1.3), Gstm2 (1.3), Gstm3 (1.7), and Mgst3 (2.2). CR feminized the mRNA profiles of 32 XPGs in livers of male mice. For instance, CR decreased the male-predominantly expressed Oatp1a1 (97%) and increased the female-predominantly expressed Oatp1a4 (11). In conclusion, short-term CR alters the mRNA levels of over half of the 98 XPGs quantified in livers of male mice, and over half of these alterations appear to be due to feminization of the liver.
Collapse
Affiliation(s)
- Zidong Donna Fu
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Curtis D Klaassen
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| |
Collapse
|
31
|
Marinho AT, Rodrigues PM, Caixas U, Antunes AMM, Branco T, Harjivan SG, Marques MM, Monteiro EC, Pereira SA. Differences in nevirapine biotransformation as a factor for its sex-dependent dimorphic profile of adverse drug reactions. J Antimicrob Chemother 2013; 69:476-82. [PMID: 24051761 DOI: 10.1093/jac/dkt359] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVES Nevirapine is widely used for the treatment of HIV-1 infection; however, its chronic use has been associated with severe liver and skin toxicity. Women are at increased risk for these toxic events, but the reasons for the sex-related differences are unclear. Disparities in the biotransformation of nevirapine and the generation of toxic metabolites between men and women might be the underlying cause. The present work aimed to explore sex differences in nevirapine biotransformation as a potential factor in nevirapine-induced toxicity. METHODS All included subjects were adults who had been receiving 400 mg of nevirapine once daily for at least 1 month. Blood samples were collected and the levels of nevirapine and its phase I metabolites were quantified by HPLC. Anthropometric and clinical data, and nevirapine metabolite profiles, were assessed for sex-related differences. RESULTS A total of 52 patients were included (63% were men). Body weight was lower in women (P = 0.028) and female sex was associated with higher alkaline phosphatase (P = 0.036) and lactate dehydrogenase (P = 0.037) levels. The plasma concentrations of nevirapine (P = 0.030) and the metabolite 3-hydroxy-nevirapine (P = 0.035), as well as the proportions of the metabolites 12-hydroxy-nevirapine (P = 0.037) and 3-hydroxy-nevirapine (P = 0.001), were higher in women, when adjusted for body weight. CONCLUSIONS There was a sex-dependent variation in nevirapine biotransformation, particularly in the generation of the 12-hydroxy-nevirapine and 3-hydroxy-nevirapine metabolites. These data are consistent with the sex-dependent formation of toxic reactive metabolites, which may contribute to the sex-dependent dimorphic profile of nevirapine toxicity.
Collapse
Affiliation(s)
- Aline T Marinho
- Centro de Estudos de Doenças Crónicas (CEDOC), NOVA Medical School, Universidade Nova de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Runge-Morris M, Kocarek TA. Expression of the sulfotransferase 1C family: implications for xenobiotic toxicity. Drug Metab Rev 2013; 45:450-9. [PMID: 24028175 DOI: 10.3109/03602532.2013.835634] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The SULT1C enzymes are a relatively under-studied branch of the cytosolic sulfotransferase (SULT) multigene family. Concrete information about SULT1C tissue-specific expression, substrate preference, role in physiology and regulation is just emerging in the literature. The role of SULT1Cs in normal physiology is uncertain, but SULT1C-catalyzed sulfonation of thyroid hormones may be a mechanism to titrate the pre-receptor levels of biologically active thyroid hormone in target tissues. Both rat and human cytosolic SULT1Cs are most noted for their ability to bioactivate potent procarcinogens such as N-hydroxy-2-acetylaminofluorene. This implicates a possible role for the SULT1Cs as modulators of environmental carcinogen exposure and determinants of neoplastic transformation. In humans, the SULT1Cs are likely to function physiologically in cell proliferation and organogenesis pathways during development, as SULT1Cs appear to be preferentially expressed during fetal life. In recent years, the SULT1C nomenclature as presented in the literature has undergone major changes in response to updated genomic information. The purpose of this review is to summarize the current literature on the SULT1Cs and to clarify perspectives on SULT1C species differences, tissue-specific expression, nomenclature and role in pathophysiology. The ultimate goal is to understand the undiscovered impact of SULT1C expression on hormone homeostasis and xenobiotic toxicity during human development and as a prelude to disease development later in life.
Collapse
Affiliation(s)
- Melissa Runge-Morris
- Institute of Environmental Health Sciences, Wayne State University , Detroit, MI , USA
| | | |
Collapse
|
33
|
Lu H, Gunewardena S, Cui JY, Yoo B, Zhong XB, Klaassen CD. RNA-sequencing quantification of hepatic ontogeny and tissue distribution of mRNAs of phase II enzymes in mice. Drug Metab Dispos 2013; 41:844-57. [PMID: 23382457 PMCID: PMC3608454 DOI: 10.1124/dmd.112.050211] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Accepted: 01/31/2013] [Indexed: 01/30/2023] Open
Abstract
Phase II conjugating enzymes play key roles in the metabolism of xenobiotics. In the present study, RNA sequencing was used to elucidate hepatic ontogeny and tissue distribution of mRNA expression of all major known Phase II enzymes, including enzymes involved in glucuronidation, sulfation, glutathione conjugation, acetylation, methylation, and amino acid conjugation, as well as enzymes for the synthesis of Phase II cosubstrates, in male C57BL/6J mice. Livers from male C57BL/6J mice were collected at 12 ages from prenatal to adulthood. Many of these Phase II enzymes were expressed at much higher levels in adult livers than in perinatal livers, such as Ugt1a6b, -2a3, -2b1, -2b5, -2b36, -3a1, and -3a2; Gsta1, -m1, -p1, -p2, and -z1; mGst1; Nat8; Comt; Nnmt; Baat; Ugdh; and Gclc. In contrast, hepatic mRNA expression of a few Phase II enzymes decreased during postnatal liver development, such as mGst2, mGst3, Gclm, and Mat2a. Hepatic expression of certain Phase II enzymes peaked during the adolescent stage, such as Ugt1a1, Sult1a1, Sult1c2, Sult1d1, Sult2as, Sult5a1, Tpmt, Glyat, Ugp2, and Mat1a. In adult mice, the total transcripts for Phase II enzymes were comparable in liver, kidney, and small intestine; however, individual Phase II enzymes displayed marked tissue specificity among the three organs. In conclusion, this study unveils for the first time developmental changes in mRNA abundance of all major known Phase II enzymes in mouse liver, as well as their tissue-specific expression in key drug-metabolizing organs. The age- and tissue-specific expression of Phase II enzymes indicate that the detoxification of xenobiotics is highly regulated by age and cell type.
Collapse
Affiliation(s)
- Hong Lu
- Department of Pharmacology, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY 13210, USA.
| | | | | | | | | | | |
Collapse
|
34
|
Wen X, Donepudi AC, Thomas PE, Slitt AL, King RS, Aleksunes LM. Regulation of hepatic phase II metabolism in pregnant mice. J Pharmacol Exp Ther 2013; 344:244-52. [PMID: 23055538 PMCID: PMC3533409 DOI: 10.1124/jpet.112.199034] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Accepted: 10/05/2012] [Indexed: 02/06/2023] Open
Abstract
Phase II enzymes, including Ugts, Sults, and Gsts, are critical for the disposition and detoxification of endo- and xenobiotics. In this study, the mRNA and protein expression of major phase II enzymes, as well as key regulatory transcription factors, were quantified in livers of time-matched pregnant and virgin control C57BL/6 mice on gestation days (GD) 7, 11, 14, 17, and postnatal days (PND) 1, 15, and 30. Compared with virgin controls, the mRNA expression of Ugt1a1, 1a6, 1a9, 2a3, 2b1, 2b34, and 2b35 decreased 40 to 80% in pregnant dams. Protein expression of Ugt1a6 also decreased and corresponded with reduced in vitro glucuronidation of bisphenol A in S9 fractions from livers of pregnant mice. Similar to Ugts levels, Gsta1 and a4 mRNAs were reduced in pregnant dams in mid to late gestation; however no change in protein expression was observed. Conversely, Sult1a1, 2a1/2, and 3a1 mRNAs increased 100 to 500% at various time points in pregnant and lactating mice and corresponded with enhanced in vitro sulfation of acetaminophen in liver S9 fractions. Coinciding with maximal decreases in Ugts as well as increases in Sults, the expression of transcription factors CAR, PPARα, and PXR and their target genes were downregulated, whereas ERα mRNA was upregulated. Collectively, these data demonstrate altered regulation of hepatic phase II metabolism in mice during pregnancy and suggest that CAR, PPARα, PXR, and ERα signaling pathways may be candidate signaling pathways responsible for these changes.
Collapse
Affiliation(s)
- Xia Wen
- Dept. of Pharmacology and Toxicology, Rutgers University Ernest Mario School of Pharmacy, 170 Frelinghuysen Rd. Piscataway, NJ 08854, USA
| | | | | | | | | | | |
Collapse
|
35
|
Aleksunes LM, Klaassen CD. Coordinated regulation of hepatic phase I and II drug-metabolizing genes and transporters using AhR-, CAR-, PXR-, PPARα-, and Nrf2-null mice. Drug Metab Dispos 2012; 40:1366-79. [PMID: 22496397 PMCID: PMC3382842 DOI: 10.1124/dmd.112.045112] [Citation(s) in RCA: 199] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Accepted: 04/11/2012] [Indexed: 02/06/2023] Open
Abstract
The transcription factors aryl hydrocarbon receptor (AhR), constitutive androstane receptor (CAR), pregnane X receptor (PXR), peroxisome proliferator-activated receptor α (PPARα), and nuclear factor erythroid 2-related factor 2 (Nrf2) regulate genes encoding drug-metabolizing enzymes and transporters in livers of mice after chemical activation. However, the specificity of their transcriptional regulation has not been determined systematically in vivo. The purpose of this study was to identify genes encoding drug-metabolizing enzymes and transporters altered by chemical activators in a transcription factor-dependent manner using wild-type and transcription factor-null mice. Chemical activators were administered intraperitoneally to mice once daily for 4 days. Livers were collected 24 h after the final dose, and total RNA was isolated for mRNA quantification of cytochromes P450, NAD(P)H quinone oxidoreductase 1 (Nqo1), aldehyde dehydrogenases (Aldhs), glutathione transferases (Gsts), sulfotransferases (Sults), UDP-glucuronosyltransferases (Ugts), organic anion-transporting polypeptides (Oatps), and multidrug resistance-associated proteins (Mrps). Pharmacological activation of each transcription factor leads to mRNA induction of drug metabolic and transport genes in livers of male and female wild-type mice, but no change in null mice: AhR (Cyp1a2, Nqo1, Aldh7a1, Ugt1a1, Ugt1a6, Ugt1a9, Ugt2b35, Sult5a1, Gstm3, and Mrp4), CAR (Cyp2b10, Aldh1a1, Aldh1a7, Ugt1a1, Ugt2b34, Sult1e1, Sult3a1, Sult5a1, Papps2, Gstt1, Gsta1, Gsta4, Gstm1-4, and Mrp2-4), PXR (Cyp3a11, Ugt1a1, Ugt1a5, Ugt1a9, Gsta1, Gstm1-m3, Oatp1a4, and Mrp3), PPARα (Cyp4a14, Aldh1a1, mGst3, Gstm4, and Mrp4), and Nrf2 (Nqo1, Aldh1a1, Gsta1, Gsta4, Gstm1-m4, mGst3, and Mrp3-4). Taken together, these data reveal transcription factor specificity and overlap in regulating hepatic drug disposition genes by chemical activators. Coordinated regulation of phase I, phase II, and transport genes by activators of transcription factors can have implications in development of pharmaceuticals as well as risk assessment of environmental contaminants.
Collapse
MESH Headings
- Aldehyde Dehydrogenase/genetics
- Aldehyde Dehydrogenase/metabolism
- Animals
- Constitutive Androstane Receptor
- Cytochrome P-450 Enzyme System/genetics
- Cytochrome P-450 Enzyme System/metabolism
- Female
- Gene Expression Regulation, Enzymologic
- Glucuronosyltransferase/genetics
- Glucuronosyltransferase/metabolism
- Glutathione Transferase/genetics
- Glutathione Transferase/metabolism
- Inactivation, Metabolic
- Liver/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Multidrug Resistance-Associated Proteins
- NAD(P)H Dehydrogenase (Quinone)/genetics
- NAD(P)H Dehydrogenase (Quinone)/metabolism
- NF-E2-Related Factor 2/genetics
- NF-E2-Related Factor 2/metabolism
- Organic Anion Transporters/genetics
- Organic Anion Transporters/metabolism
- PPAR alpha/genetics
- PPAR alpha/metabolism
- Pregnane X Receptor
- RNA, Messenger/genetics
- Receptors, Aryl Hydrocarbon/genetics
- Receptors, Aryl Hydrocarbon/metabolism
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Receptors, Steroid/genetics
- Receptors, Steroid/metabolism
- Sulfotransferases/genetics
- Sulfotransferases/metabolism
- Transcription Factors/genetics
- Transcription Factors/metabolism
Collapse
Affiliation(s)
- Lauren M Aleksunes
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160-7417, USA
| | | |
Collapse
|
36
|
Sivils JC, Ancrum TM, Bain LJ. LOSS of Mrp1 alters detoxification enzyme expression in a tissue- and hormonal-status-specific manner. J Appl Toxicol 2012; 33:766-73. [PMID: 22522787 DOI: 10.1002/jat.2727] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Revised: 12/29/2011] [Accepted: 12/29/2011] [Indexed: 12/24/2022]
Abstract
The multidrug resistance-associated protein1 (MRP1/ABCC1) is a member of the ABCC transporter subfamily that mediates the efflux of pharmaceuticals, xenobiotics and steroid hormones, typically as glutathione, glucuronide or sulfate conjugates. Since loss of one transporter can be compensated by increasing the expression of other transporters and conjugation enzymes, we sought to examine compensatory changes in phase I, II and III enzyme expression in extrahepatic tissues, including the kidney, lungs and small intestine of intact or castrated Mrp1(-/-) male mice. In the kidney, the expression of several P450s, sulfotransferase 1a1 (Sult), glucuronosyltransferases (Ugt) and Mrps2-4, were significantly changed owing to castration alone. The only time genotype mattered was between the castrated FVB and Mrp1 knockout mice. In contrast, expression of the Ugts, Sult 1a1 and Mrp3 in the lungs was significantly downregulated in the Mrp1 knockout mice, so based exclusively on genotype. In the small intestine, there were interactions between steroid hormone levels and genotype, as the expression differences were only found in mice lacking Mrp1, and were changed between intact and castrated animals. The mechanism behind this pattern of expression may be to due to Nrf2 regulation, as its expression mirrors that of the phase II and phase III enzymes. These results indicate that compensatory responses owing to the loss of Mrp1 vary dramatically, depending on the particular tissue. This information will aid in the understanding of how drug uptake, disposition and elimination can be influenced by both hormone status and the presence and magnitude of transporter expression.
Collapse
Affiliation(s)
- Jeffrey C Sivils
- Department of Biological Sciences, University of Texas at El Paso, 500 W. University Avenue, El Paso, TX 79910, USA
| | | | | |
Collapse
|
37
|
Sugamori KS, Brenneman D, Sanchez O, Doll MA, Hein DW, Pierce WM, Grant DM. Reduced 4-aminobiphenyl-induced liver tumorigenicity but not DNA damage in arylamine N-acetyltransferase null mice. Cancer Lett 2011; 318:206-13. [PMID: 22193722 DOI: 10.1016/j.canlet.2011.12.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Revised: 12/08/2011] [Accepted: 12/08/2011] [Indexed: 01/23/2023]
Abstract
The aromatic amine 4-aminobiphenyl (ABP) is a liver procarcinogen in mice, requiring enzymatic bioactivation to exert its tumorigenic effect. To assess the role of arylamine N-acetyltransferase (NAT)-dependent acetylation capacity in the risk for ABP-induced liver tumors, we compared 1-year liver tumor incidence following the postnatal exposure of wild-type and NAT-deficient Nat1/2(-/-) mice to ABP. At an ABP exposure of 1200 nmol, male Nat1/2(-/-) mice had a liver tumor incidence of 36% compared to 69% in wild-type males, and at 600 nmol there was a complete absence of tumors compared to 60% in wild-type mice. Only one female wild-type mouse had a tumor using this exposure protocol. However, levels of N-deoxyguanosin-8-yl-ABP-DNA adducts did not correlate with either the strain or sex differences in tumor incidence. These results suggest that female sex and NAT deficiency reduce risk for ABP-induced liver tumors, but by mechanisms unrelated to differences in DNA-damaging events.
Collapse
Affiliation(s)
- Kim S Sugamori
- Department of Pharmacology & Toxicology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
38
|
Conley AJ, Plant TM, Abbott DH, Moeller BC, Stanley SD. Adrenal androgen concentrations increase during infancy in male rhesus macaques (Macaca mulatta). Am J Physiol Endocrinol Metab 2011; 301:E1229-35. [PMID: 21900126 PMCID: PMC3274962 DOI: 10.1152/ajpendo.00200.2011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This study investigated adrenal androgens (AA), gonadotropins, and cortisol in castrated and gonad-intact male rhesus macaques from birth through infancy. Blood samples were collected longitudinally from castrated (n = 6; weekly, 1-40 wk) and intact (n = 4; every other week, 1-17 wk) males. Plasma concentrations of AA were determined by liquid chromatography-tandem mass spectrometry, and plasma concentrations of cortisol and gonadotropins were determined by RIA. Dehydroepiandrosterone sulfate (DHEAS) concentrations increased almost threefold (to 8 wk), dehydroepiandrosterone (DHEA) increased more than eightfold (to 11 wk), and androstenedione doubled (to 15 wk) in five castrated infant males and declined continuously thereafter. A sixth castrated male had markedly different temporal patterns and concentrations (many times more than 2 SDs from the cohort mean) of AA and gonadotropins from first sampling (3 wk) and was excluded from analysis. Cortisol increased over 16 wk but correlated poorly with DHEAS. Luteinizing and follicle-stimulating hormones increased to peaks at 3 and 7 wk, respectively. Testis-intact males exhibited similar profiles, but with earlier peaks of DHEAS (5 wk) and DHEA and androstenedione (7 wk). Peak concentrations of DHEAS were lower and those of DHEA and androstenedione were higher in intact than castrated infants. Testosterone was undetectable in castrated males and >0.5 ng/ml in intact males but was not correlated with DHEA or DHEAS. These are the first data documenting a transient increase in AA secretion during infancy in an Old World primate and are consistent with the previously documented time course of zona reticularis development that accompanies increases in androgen synthetic capacity of the adrenal. The rhesus is a promising model for androgen secretion from the human adrenal cortex.
Collapse
Affiliation(s)
- A J Conley
- School of Veterinary Medicine, University of California-Davis, Davis, California 95616, USA.
| | | | | | | | | |
Collapse
|
39
|
Yang CH, Tang L, Lv C, Ye L, Xia BJ, Hu M, Liu ZQ. Sulfation of selected mono-hydroxyflavones by sulfotransferases in vitro: a species and gender comparison. ACTA ACUST UNITED AC 2011; 63:967-70. [PMID: 21635263 DOI: 10.1111/j.2042-7158.2011.01298.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVES Sulfation via sulfotransferases is an important metabolic pathway contributing to the low bioavailability of flavonoids. This study aims to characterize the sulfation of mono-hydroxyflavones (MHFs) to obtain useful information on structure-metabolizing relationships in animal species and gender differences. METHODS Three representative MHFs, namely, 7-, 6- and 4'-MHF, were studied by incubating each MHF at different concentrations with various liver S9 fractions (mouse, rat, dog and human). KEY FINDINGS One mono-sulfate was identified for each MHF. 7-MHF and 4'-MHF usually have greater sulfations than 6-MHF. Regardless of whether the S9 fraction came from a male or female, there was a difference in sulfation in the species observed for all MHFs; the highest activity of sulfotransferases was in dog S9. Furthermore, gender differences affect sulfation of MHFs significantly. In rats, all sulfations for the three MHFs were higher in males than that in females while the opposite was observed in mice. CONCLUSIONS Regiospecific, species and gender dependence exist in the sulfonation of all selected MHFs.
Collapse
Affiliation(s)
- Cai Hua Yang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | | | | | | | | | | | | |
Collapse
|
40
|
Viitaniemi HM, Leder EH. Sex-Biased Protein Expression in Threespine Stickleback, Gasterosteus aculeatus. J Proteome Res 2011; 10:4033-40. [DOI: 10.1021/pr200234a] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
| | - Erica Helen Leder
- Section of Genetics and Physiology, Department of Biology, University of Turku, Finland
| |
Collapse
|
41
|
Baik M, Yu JH, Hennighausen L. Growth hormone-STAT5 regulation of growth, hepatocellular carcinoma, and liver metabolism. Ann N Y Acad Sci 2011; 1229:29-37. [PMID: 21793836 PMCID: PMC3427656 DOI: 10.1111/j.1749-6632.2011.06100.x] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The liver is a primary target of growth hormone (GH). GH signals are mediated by the transcription factor signal transducer and activator of transcription 5 (STAT5). Here, we focus on recent discoveries about the role of GH-STAT5 signaling in hepatic physiology and pathophysiology. We discuss roles of the GH-STAT5 axis in body growth, lipid metabolism, and the cell cycle pertaining to hepatosteatosis, fibrosis, and hepatocellular carcinoma. Finally, we discuss recent discoveries about the role of GH-STAT5 in sex-specific gene expression and bile acid, steroid, and drug metabolism.
Collapse
Affiliation(s)
- Myunggi Baik
- Deptartment of Molecular Biotechnology, WCU-RNNM, Chonnam National University, Gwangju, Republic of Korea
| | - Ji Hoon Yu
- Laboratory of Genetics and Physiology, National Institutes of Health, Bethesda, Maryland
| | - Lothar Hennighausen
- Laboratory of Genetics and Physiology, National Institutes of Health, Bethesda, Maryland
- Deptartment of Molecular Biotechnology, WCU-RNNM, Chonnam National University, Gwangju, Republic of Korea
| |
Collapse
|