1
|
de Souza TG, de Abreu MR, Kuradomi RY, Batlouni SR. Effect of temperature on gonadal differentiation and growth of Leporinus friderici. Anim Reprod 2024; 21:e20230158. [PMID: 39021500 PMCID: PMC11253782 DOI: 10.1590/1984-3143-ar2023-0158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 04/29/2024] [Indexed: 07/20/2024] Open
Abstract
This study aimed to investigate the effect of temperature on gonadal differentiation, growth, survival, and sex ratio of Leporinus friderici reared at 25 °C or 29 °C from 50 to 240 days after eclosion (DAE) in a water recirculation system. A total of 110 fish at 50 DAE (6.7 ± 0.1 cm and 6.1 ± 0.3 g) were equally and randomly distributed in 10 boxes (90 L) (11 fish/box, 5 boxes/temperature). One fish from each experimental unit was randomly sampled at 50, 70, 90, 110, 130, 150, 170, 190, 210 and 240 DAE. Female gonadal differentiation started at 150 DAE (11.4 ± 0.0 cm and 16.4 ± 0.0 g) at 25 °C and at 170 DAE (10.7 ± 0.7 cm and 27.7 ± 8.5 g) at 29 ºC, while testes differentiation only occurred at 29 °C from 190 DAE (12.1 ± 0.0 cm and 38.0 ± 0.0 g). Of 50 fishes sampled in each condition, 17 (12 females and five males) and three (three females) displayed gonadal differentiation at 29 °C and 25 °C, respectively. Final biometric values at 29 °C were twice those obtained at 25 °C, reaching 13.9 ± 0.65 cm and 57.3 ± 10.12 g versus 11.2 ± 0.39 cm and 28.5 ± 2.95 g, respectively. While temperature clearly influenced gonadal differentiation and growth, it had inconclusive effects on sex ratio. The higher temperature (29 °C) has direct implications for the production of this species, as it accelerates growth without causing mortality.
Collapse
Affiliation(s)
| | | | - Rafael Yutaka Kuradomi
- Centro de Aquicultura, Universidade Estadual Paulista, Jaboticabal, SP, Brasil
- Instituto de Ciências Exatas e Tecnologia, Universidade Federal do Amazonas, Itacoatiara, AM, Brasil
| | | |
Collapse
|
2
|
Allahyari M, Motavalizadeh-Kakhky AR, Mehrzad J, Zhiani R, Chamani J. Cellulose nanocrystals derived from chicory plant: an un-competitive inhibitor of aromatase in breast cancer cells via PI3K/AKT/mTOP signalling pathway. J Biomol Struct Dyn 2024; 42:5575-5589. [PMID: 37340682 DOI: 10.1080/07391102.2023.2226751] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 06/13/2023] [Indexed: 06/22/2023]
Abstract
A significant contributing factor in the development of breast cancer is the estrogens. The synthesis of estrogens is primarily facilitated by aromatase (CYP19), a cytochrome P450 enzyme. Notably, aromatase is expressed at a higher level in human breast cancer tissue compared with the normal breast tissue. Therefore, inhibiting aromatase activity is a potential strategy in hormone receptor-positive breast cancer treatment. In this study, Cellulose Nanocrystals (CNCs) were obtained from Chicory plant waste through a sulfuric acid hydrolysis method with the objective of investigating that whether the obtained CNCs could act as an inhibitor of aromatase enzyme, and prevent the conversion of androgens to estrogens. Structural analysis of CNCs was carried out using Fourier transform infrared spectroscopy (FTIR) and X-ray diffraction (XRD), while morphological results were obtained using AFM, TEM, and FE-SEM. Furthermore, the nano-particles were found to be spherical in shape with a diameter range of 35-37 nm and displayed a reasonable negative surface charge. Stable transfection of MCF-7 cells with CYP19 has demonstrated the ability of CNCs to inhibit aromatase activities and prevent cell growth by interfering with the enzyme activities. Spectroscopic results revealed the binding constant of CYP19-CNCs and (CYP19-Androstenedione)-CNCs complexes to be 2.07 × 103 L/gr and 2.06 × 104 L/gr, respectively. Conductometry and CD data reported different interaction behaviors among CYP19 and CYP19-Androstenedione complexes at the presence of CNCs in the system. Moreover, the addition of CNCs to the solution in a successive manner resulted in the enhancement of the secondary structure of the CYP19-androstenedione complex. Additionally, CNCs showed a marked reduction in the viability of cancer cells compared to normal cells by enhancing the expression of Bax and p53 at protein and mRNA levels, and by decreasing mRNA levels of PI3K, AKT, and mTOP, as well as protein levels of PI3Kg-P110 and P-mTOP, in MCF-7 cells after incubation with CNCs at IC50 concentration. These findings confirm the decrease in proliferation of breast cancer cells associated with induction of apoptosis through down-regulation of the PI3K/AKT/mTOP signaling pathway. According to the provided data, the obtained CNCs are capable of inhibiting aromatase enzyme activity, which has significant implications for the treatment of cancer.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Manizheh Allahyari
- Department of Biochemistry, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran
| | - Ali Reza Motavalizadeh-Kakhky
- Department of Chemistry, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran
- New Material Technology and Processing Research Center, Department of Chemistry, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran
| | - Jamshid Mehrzad
- Department of Biochemistry, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran
| | - Rahele Zhiani
- Department of Chemistry, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran
- Advance Research Center of Chemistry Biochemistry& Nanomaterial, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran
| | - Jamshidkhan Chamani
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| |
Collapse
|
3
|
Zhao Y, Wang Y, Gu P, Tuo L, Wang L, Jiang SW. Transgenic mice applications in the study of endometriosis pathogenesis. Front Cell Dev Biol 2024; 12:1376414. [PMID: 38933332 PMCID: PMC11199864 DOI: 10.3389/fcell.2024.1376414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
Endometriosis (EM), characterized by ectopic growth of endometrial tissues and recurrent pelvic pain, is a common disease with severe negative impacts on the life quality of patients. Conventional uterine tissue transplantation-based models have been broadly used to investigate the pathogenic mechanism(s) of EM. Transgenic mice with whole body or uterine/pelvic tissue-specific labelling by the expression of GFP, β-gal or other light-emitting or chromogenic markers enable investigators to analyze the contribution to endometriotic lesions by the donor or recipient side after uterine tissue transplantation. Moreover, when coupled to uterine tissue transplantation, transgenic mice with a specific EM-related gene knocked out or overexpressed make it possible to determine the gene's in vivo role(s) for EM pathogenesis. Furthermore, observations on the rise of de novo endometriotic lesions as well as structural/functional changes in the eutopic endometrium or pelvic tissues after gene manipulation will directly relate the cognate gene to the onset of EM. A major advantage of transgenic EM models is their efficiency for analyzing gene interactions with hormonal, dietetic and/or environmental factors. This review summarizes the features/sources/backgrounds of transgenic mice and their applications to EM studies concerning hormonal regulation, angiogenesis and inflammation. Findings from these studies, the advantages/disadvantages of transgenic EM models, and future expectations are also discussed.
Collapse
Affiliation(s)
- Yali Zhao
- Center of Prenatal Diagnosis, Lianyungang Maternal and Child Health Hospital, Lianyungang, Jiangsu, China
| | - Yao Wang
- Department of Gynecology, Lianyungang Maternal and Child Health Hospital, Lianyungang, Jiangsu, China
| | - Pinlang Gu
- Department of Gynecology, Lianyungang Maternal and Child Health Hospital, Lianyungang, Jiangsu, China
| | - Lingjin Tuo
- Lianyungang Research Institute for Women’s and Children’s Health, Lianyungang Maternal and Child Health Hospital, Lianyungang, Jiangsu, China
| | - Leilei Wang
- Center of Prenatal Diagnosis, Lianyungang Maternal and Child Health Hospital, Lianyungang, Jiangsu, China
| | - Shi-Wen Jiang
- Lianyungang Research Institute for Women’s and Children’s Health, Lianyungang Maternal and Child Health Hospital, Lianyungang, Jiangsu, China
| |
Collapse
|
4
|
Zhang D, Tian T, Han L, Du J, Zhu T, Lei C, Song H, Li S. Expression characteristics of the cyp19a1b aromatase gene and its response to 17β-estradiol treatment in largemouth bass (Micropterus salmoides). FISH PHYSIOLOGY AND BIOCHEMISTRY 2024; 50:575-588. [PMID: 38216846 DOI: 10.1007/s10695-023-01291-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 12/22/2023] [Indexed: 01/14/2024]
Abstract
To investigate the regulatory role of the cyp19a1b aromatase gene in the sexual differentiation of largemouth bass (Micropterus salmoides, LMB), we obtained the full-length cDNA sequence of cyp19a1b using rapid amplification of cDNA ends technique. Tissue expression characteristics and feedback with 17-β-estradiol (E2) were determined using quantitative real-time PCR (qRT-PCR), while gonad development was assessed through histological section observations. The cDNA sequence of LMB cyp19a1b was found to be1950 base pairs (bp) in length, including a 5' untranslated region of 145 bp, a 3' untranslated region of 278 bp, and an open reading frame encoding a protein consisting of 1527 bp that encoded 508 amino acids. The qRT-PCR results indicated that cyp19a1b abundantly expressed in the brain, followed by the gonads, and its expression in the ovaries was significantly higher than that observed in the testes (P < 0.05). After feeding fish with E2 for 30 days, the expression of cyp19a1b in the pseudo-female gonads (XY-F) was significantly higher than that in males (XY-M) (P < 0.05), whereas expression did not differ significantly between XX-F and XY-F fish (P > 0.05). Although the expression of cyp19a1b in XY-F and XX-F fish was not significantly different after 60 days (P>0.05), both exhibited significantly higher levels than that of XY-M fish (P<0.05). Histological sections analysis showed the presence of oogonia in both XY-F and XX-F fish at 30 days, while spermatogonia were observed in XY-M fish. At 60 days, primary oocytes were abundantly observed in both XY-F and XX-F fish, while a few spermatogonia were visible in XY-M fish. At 90 days, the histological sections' results showed that a large number of oocytes were visible in XY-F and XX-F fish. Additionally, the gonads of XY-M fish contained numerous spermatocytes. These results suggest that cyp19a1b plays a pivotal role in the development of ovaries and nervous system development in LMB.
Collapse
Affiliation(s)
- Dongyun Zhang
- Pearl River Fisheries Research Institute, Chinese Academy of Fisheries Sciences, Key Laboratory of Tropical and Subtropical Fishery Resource Application and Cultivation, China Ministry of Agriculture, Guangzhou, 510380, China
- College of Life Science, Huzhou University, Huzhou, 313000, Zhejiang, China
| | - Taihang Tian
- Pearl River Fisheries Research Institute, Chinese Academy of Fisheries Sciences, Key Laboratory of Tropical and Subtropical Fishery Resource Application and Cultivation, China Ministry of Agriculture, Guangzhou, 510380, China
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, People's Republic of China
| | - Linqiang Han
- Guangdong province Liangshi Aquaculture Seed Industry, Foshan, 528100, Guangdong, China
| | - Jinxing Du
- Pearl River Fisheries Research Institute, Chinese Academy of Fisheries Sciences, Key Laboratory of Tropical and Subtropical Fishery Resource Application and Cultivation, China Ministry of Agriculture, Guangzhou, 510380, China
| | - Tao Zhu
- Pearl River Fisheries Research Institute, Chinese Academy of Fisheries Sciences, Key Laboratory of Tropical and Subtropical Fishery Resource Application and Cultivation, China Ministry of Agriculture, Guangzhou, 510380, China
| | - Caixia Lei
- Pearl River Fisheries Research Institute, Chinese Academy of Fisheries Sciences, Key Laboratory of Tropical and Subtropical Fishery Resource Application and Cultivation, China Ministry of Agriculture, Guangzhou, 510380, China
| | - Hongmei Song
- Pearl River Fisheries Research Institute, Chinese Academy of Fisheries Sciences, Key Laboratory of Tropical and Subtropical Fishery Resource Application and Cultivation, China Ministry of Agriculture, Guangzhou, 510380, China.
| | - Shengjie Li
- Pearl River Fisheries Research Institute, Chinese Academy of Fisheries Sciences, Key Laboratory of Tropical and Subtropical Fishery Resource Application and Cultivation, China Ministry of Agriculture, Guangzhou, 510380, China.
| |
Collapse
|
5
|
Zhang M, Rottschäfer V, C M de Lange E. The potential impact of CYP and UGT drug-metabolizing enzymes on brain target site drug exposure. Drug Metab Rev 2024; 56:1-30. [PMID: 38126313 DOI: 10.1080/03602532.2023.2297154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023]
Abstract
Drug metabolism is one of the critical determinants of drug disposition throughout the body. While traditionally associated with the liver, recent research has unveiled the presence and functional significance of drug-metabolizing enzymes (DMEs) within the brain. Specifically, cytochrome P-450 enzymes (CYPs) and UDP-glucuronosyltransferases (UGTs) enzymes have emerged as key players in drug biotransformation within the central nervous system (CNS). This comprehensive review explores the cellular and subcellular distribution of CYPs and UGTs within the CNS, emphasizing regional expression and contrasting profiles between the liver and brain, humans and rats. Moreover, we discuss the impact of species and sex differences on CYPs and UGTs within the CNS. This review also provides an overview of methodologies for identifying and quantifying enzyme activities in the brain. Additionally, we present factors influencing CYPs and UGTs activities in the brain, including genetic polymorphisms, physiological variables, pathophysiological conditions, and environmental factors. Examples of CYP- and UGT-mediated drug metabolism within the brain are presented at the end, illustrating the pivotal role of these enzymes in drug therapy and potential toxicity. In conclusion, this review enhances our understanding of drug metabolism's significance in the brain, with a specific focus on CYPs and UGTs. Insights into the expression, activity, and influential factors of these enzymes within the CNS have crucial implications for drug development, the design of safe drug treatment strategies, and the comprehension of drug actions within the CNS. To that end, CNS pharmacokinetic (PK) models can be improved to further advance drug development and personalized therapy.
Collapse
Affiliation(s)
- Mengxu Zhang
- Division of Systems Pharmacology and Pharmacy, Predictive Pharmacology Group, Leiden Academic Centre of Drug Research, Leiden University, Leiden, The Netherlands
| | - Vivi Rottschäfer
- Mathematical Institute, Leiden University, Leiden, The Netherlands
- Korteweg-de Vries Institute for Mathematics, University of Amsterdam, Amsterdam, The Netherlands
| | - Elizabeth C M de Lange
- Division of Systems Pharmacology and Pharmacy, Predictive Pharmacology Group, Leiden Academic Centre of Drug Research, Leiden University, Leiden, The Netherlands
| |
Collapse
|
6
|
Shaw K, Lu C, Liu X, Trudeau VL. Arginine vasopressin injection rescues delayed oviposition in cyp19a1b-/- mutant female zebrafish. Front Endocrinol (Lausanne) 2023; 14:1308675. [PMID: 38144569 PMCID: PMC10739748 DOI: 10.3389/fendo.2023.1308675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 11/23/2023] [Indexed: 12/26/2023] Open
Abstract
In zebrafish, estrogens produced in the ovaries via Cyp19a1a activity are required for both sexual differentiation of the ovary during early development as well as maintenance of the ovarian state during adulthood. The importance of Cyp19a1b that is highly expressed in the brain for female reproduction is still under study. We previously reported that female cyp19a1b -/- mutant zebrafish have significantly lower brain estradiol levels and impaired spawning behavior characterized by an increased latency to oviposition during dyadic sexual behavior encounters. In the current study, we provide evidence that the delayed oviposition in female cyp19a1b -/- mutants is linked to impaired arginine vasopressin (Avp) signaling. Droplet digital PCR experiments revealed that levels of the estrogen receptors, avp, and oxytocin (oxt) are lower in the hypothalamus of mutant females compared to wildtype fish. We then used acute intraperitoneal injections of Avp and Oxt, along with mixtures of their respective receptor antagonists, to determine that Avp can uniquely rescue the delayed oviposition in female cyp19a1b -/- mutants. Using immunohistochemistry, we demonstrated that Cyp19a1b-expressing radial glial cell (RGC) fibers surround and are in contact with Avp-immunopositive neurons in the preoptic areas of the brain. This could provide the neuroanatomical proximity for RGC-derived estrogens to diffuse to and activate estrogen receptors and regulate avp expression levels. Together these findings identify a positive link between Cyp19a1b and Avp for female zebrafish sexual behavior. They also suggest that the female cyp19a1b -/- mutant behavioral phenotype is likely a consequence of impaired processing of Avp-dependent social cues important for mate identification and assessment.
Collapse
Affiliation(s)
- Katherine Shaw
- Department of Biology, University of Ottawa, Ottawa, ON, Canada
| | - Chunyu Lu
- Department of Biology, University of Ottawa, Ottawa, ON, Canada
| | - Xiaochun Liu
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou, China
| | | |
Collapse
|
7
|
Shaw K, Therrien M, Lu C, Liu X, Trudeau VL. Mutation of brain aromatase disrupts spawning behavior and reproductive health in female zebrafish. Front Endocrinol (Lausanne) 2023; 14:1225199. [PMID: 37435485 PMCID: PMC10332311 DOI: 10.3389/fendo.2023.1225199] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 06/13/2023] [Indexed: 07/13/2023] Open
Abstract
Aromatase (Cyp19a1) is the steroidogenic enzyme that converts androgens into bioactive estrogens, and hence is in a pivotal position to mediate reproduction and sexual behavior. In teleosts, there are two aromatase paralogs: cyp19a1a that is highly expressed in granulosa and Leydig cells in the gonads with critical function in sexual differentiation of the ovary, and cyp19a1b that is highly expressed in radial glial cells in the brain with unknown roles in reproduction. Cyp19a1 -/- mutant zebrafish lines were used to investigate the importance of the cyp19a1 paralogs for spawning behavior and offspring survival and early development. Mutation of cyp19a1b was found to increase the latency to the first oviposition in females. Mutation of cyp19a1b in females also increased the number of eggs spawned; however, significantly more progeny died during early development resulting in no net increase in female fecundity. This finding suggests a higher metabolic cost of reproduction in cyp19a1b -/- mutant females. In males, the combined mutation of both cyp19a1 paralogs resulted in significantly lower progeny survival rates, indicating a critical function of cyp19a1 during early larval development. These data establish the specific importance of cyp19a1b for female spawning behavior and the importance of the cyp19a1 paralogs for early larval survival.
Collapse
Affiliation(s)
- Katherine Shaw
- Department of Biology, University of Ottawa, Ottawa, ON, Canada
| | - Mylène Therrien
- Department of Biology, University of Ottawa, Ottawa, ON, Canada
| | - Chunyu Lu
- Department of Biology, University of Ottawa, Ottawa, ON, Canada
| | - Xiaochun Liu
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou, China
| | | |
Collapse
|
8
|
Ghosh D. Structures and Functions of Human Placental Aromatase and Steroid Sulfatase, Two Key Enzymes in Estrogen Biosynthesis. Steroids 2023; 196:109249. [PMID: 37207843 DOI: 10.1016/j.steroids.2023.109249] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/12/2023] [Accepted: 05/14/2023] [Indexed: 05/21/2023]
Abstract
Cytochrome P450 aromatase (AROM) and steroid sulfatase (STS) are the two key enzymes for the biosynthesis of estrogens in human, and maintenance of the critical balance between androgens and estrogens. Human AROM, an integral membrane protein of the endoplasmic reticulum, is a member of the cytochrome P450 superfamily. It is the only enzyme to catalyze the conversion of androgens with non-aromatic A-rings to estrogens characterized by the aromatic A-ring. Human STS, also an integral membrane protein of the endoplasmic reticulum, is a Ca2+-dependent enzyme that catalyzes the hydrolysis of sulfate esters of estrone and dehydroepiandrosterone to the unconjugated steroids, the precursors of the most potent forms of estrogens and androgens, namely, 17β-estradiol, 16α,17β-estriol, testosterone and dihydrotestosterone. Expression of these steroidogenic enzymes locally within organs and tissues of the endocrine, reproductive, and central nervous systems is the key for maintaining high levels of the reproductive steroids. The enzymes have been drug targets for the prevention and treatment of diseases associated with steroid hormone excesses, especially in breast, endometrial and prostate malignancies. Both enzymes have been the subjects of vigorous research for the past six decades. In this article, we review the important findings on their structure-function relationships, specifically, the work that began with unravelling of the closely guarded secrets, namely, the 3-D structures, active sites, mechanisms of action, origins of substrate specificity and the basis of membrane integration. Remarkably, these studies were conducted on the enzymes purified in their pristine forms from human placenta, the discarded and their most abundant source. The purification, assay, crystallization, and structure determination methodologies are described. Also reviewed are their functional quaternary organizations, post-translational modifications and the advancements made in the structure-guided inhibitor design efforts. Outstanding questions that still remain open are summarized in closing.
Collapse
Affiliation(s)
- Debashis Ghosh
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, NY 13210.
| |
Collapse
|
9
|
Moradi M, Zhandi M, Sharafi M, Akbari A, Atrabi MJ, Totonchi M. Gene expression profile of placentomes and clinical parameters in the cows with retained placenta. BMC Genomics 2022; 23:760. [PMID: 36411408 PMCID: PMC9677913 DOI: 10.1186/s12864-022-08989-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/04/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Retained placenta (RP) is a prevalent disorder in cattle with many health-related and economic costs for the farm owners. Its etiology has not been clarified yet and there is no definite therapy for this disorder. In this study we conducted RNA-seq, hematologic and histologic experiments to survey the causes of RP development. METHODS Blood samples were collected from 4 RP and 3 healthy cows during periparturtion period for hematological assessments followed by placentome sampling within 30 min after parturition. Cows were grouped as RP and control in case the placenta was retained or otherwise expelled, respectively. Total RNA was extracted from placentome samples followed by RNA-sequencing. RESULTS We showed 240 differentially expressed genes (DEGs) between the RP and control groups. Enrichment analyzes indicated immune system and lipid metabolism as prominent over- and under-represented pathways in RP cows, respectively. Hormonal assessments showed that estradiol-17β (E2) was lower and cortisol tended to be higher in RP cows compared to controls at the day of parturition. Furthermore, histologic experiment showed that villi-crypt junctions remain tighter in RP cows compared to controls and the crypts layer seemed thicker in the placentome of RP cows. Complete blood cell (CBC) parameters were not significantly different between the two groups. CONCLUSION Overall, DEGs derived from expression profiling and these genes contributed to enrichment of immune and lipid metabolism pathways. We suggested that E2 could be involved in development of RP and the concentrations of P4 and CBC counts periparturition might not be a determining factor.
Collapse
Affiliation(s)
- Mehdi Moradi
- grid.46072.370000 0004 0612 7950Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| | - Mahdi Zhandi
- grid.46072.370000 0004 0612 7950Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| | - Mohsen Sharafi
- grid.412266.50000 0001 1781 3962Department of Animal Science, Faculty of Agriculture, Tarbiat Modares University, Tehran, Iran ,grid.419336.a0000 0004 0612 4397Department of Embryology, Reproduction Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACER, Tehran, Iran
| | - Arvand Akbari
- grid.417689.5Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Mohammad Jafari Atrabi
- grid.411984.10000 0001 0482 5331Institute of Pharmacology and Toxicology, University Medical Center, Georg August University, Göttingen, Germany
| | - Mehdi Totonchi
- grid.417689.5Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| |
Collapse
|
10
|
Cabrera Zapata LE, Garcia-Segura LM, Cambiasso MJ, Arevalo MA. Genetics and Epigenetics of the X and Y Chromosomes in the Sexual Differentiation of the Brain. Int J Mol Sci 2022; 23:ijms232012288. [PMID: 36293143 PMCID: PMC9603441 DOI: 10.3390/ijms232012288] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/10/2022] [Accepted: 10/11/2022] [Indexed: 11/27/2022] Open
Abstract
For many decades to date, neuroendocrinologists have delved into the key contribution of gonadal hormones to the generation of sex differences in the developing brain and the expression of sex-specific physiological and behavioral phenotypes in adulthood. However, it was not until recent years that the role of sex chromosomes in the matter started to be seriously explored and unveiled beyond gonadal determination. Now we know that the divergent evolutionary process suffered by X and Y chromosomes has determined that they now encode mostly dissimilar genetic information and are subject to different epigenetic regulations, characteristics that together contribute to generate sex differences between XX and XY cells/individuals from the zygote throughout life. Here we will review and discuss relevant data showing how particular X- and Y-linked genes and epigenetic mechanisms controlling their expression and inheritance are involved, along with or independently of gonadal hormones, in the generation of sex differences in the brain.
Collapse
Affiliation(s)
- Lucas E. Cabrera Zapata
- Instituto de Investigación Médica Mercedes y Martín Ferreyra (INIMEC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Córdoba, Córdoba 5016, Argentina
- Instituto Cajal (IC), Consejo Superior de Investigaciones Científicas (CSIC), 28002 Madrid, Spain
| | | | - María Julia Cambiasso
- Instituto de Investigación Médica Mercedes y Martín Ferreyra (INIMEC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Córdoba, Córdoba 5016, Argentina
- Cátedra de Biología Celular, Facultad de Odontología, Universidad Nacional de Córdoba, Córdoba 5000, Argentina
- Correspondence: (M.J.C.); (M.A.A.)
| | - Maria Angeles Arevalo
- Instituto Cajal (IC), Consejo Superior de Investigaciones Científicas (CSIC), 28002 Madrid, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: (M.J.C.); (M.A.A.)
| |
Collapse
|
11
|
Queathem ED, Fitzgerald M, Welly R, Rowles CC, Schaller K, Bukhary S, Baines CP, Rector RS, Padilla J, Manrique-Acevedo C, Lubahn DB, Vieira-Potter VJ. Suppression of estrogen receptor beta classical genomic activity enhances systemic and adipose-specific response to chronic beta-3 adrenergic receptor (β3AR) stimulation. Front Physiol 2022; 13:920675. [PMID: 36213237 PMCID: PMC9534559 DOI: 10.3389/fphys.2022.920675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 08/15/2022] [Indexed: 11/23/2022] Open
Abstract
White adipose tissue (WAT) dysfunction independently predicts cardiometabolic disease, yet there is a lack of effective adipocyte-targeting therapeutics. B3AR agonists enhance adipocyte mitochondrial function and hold potential in this regard. Based on enhanced sensitivity to B3AR-mediated browning in estrogen receptor (ER)alpha-null mice, we hypothesized that ERβ may enhance the WAT response to the B3AR ligand, CL316,243 (CL). Methods: Male and female wild-type (WT) and ERβ DNA binding domain knock-out (ERβDBDKO) mice fed high-fat diet (HFD) to induce obesity were administered CL (1 mg/kg) daily for 2 weeks. Systemic physiological assessments of body composition (EchoMRI), bioenergetics (metabolic chambers), adipocyte mitochondrial respiration (oroboros) and glucose tolerance were performed, alongside perigonadal (PGAT), subcutaneous (SQAT) and brown adipose tissue (BAT) protein expression assessment (Western blot). Mechanisms were tested in vitro using primary adipocytes isolated from WT mice, and from Esr2-floxed mice in which ERβ was knocked down. Statistical analyses were performed using 2 × 2 analysis of variance (ANOVA) for main effects of genotype (G) and treatment (T), as well as GxT interactions; t-tests were used to determine differences between in vitro treatment conditions (SPSS V24). Results: There were no genotype differences in HFD-induced obesity or systemic rescue effects of CL, yet ERβDBDKO females were more sensitive to CL-induced increases in energy expenditure and WAT UCP1 induction (GxT, p < 0.05), which coincided with greater WAT B3AR protein content among the KO (G, p < 0.05). Among males, who were more insulin resistant to begin with (no genotype differences before treatment), tended to be more sensitive to CL-mediated reduction in insulin resistance. With sexes combined, basal WAT mitochondrial respiration trended toward being lower in the ERβDBDKO mice, but this was completely rescued by CL (p < 0.05). Confirming prior work, CL increased adipose tissue ERβ protein (T, p < 0.05, all), an effect that was enhanced in WAT and BAT the female KO (GxT, p < 0.01). In vitro experiments indicated that an inhibitor of ERβ genomic function (PHTPP) synergized with CL to further increase UCP1 mRNA (p = 0.043), whereas full ERβ protein was required for UCP1 expression (p = 0.042). Conclusion: Full ERβ activity appears requisite and stimulatory for UCP1 expression via a mechanism involving non-classical ERβ signaling. This novel discovery about the role of ERβ in adipocyte metabolism may have important clinical applications.
Collapse
Affiliation(s)
- Eric D. Queathem
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
- Department of Biochemistry, University of Missouri, Columbia, MO, United States
| | - Maggie Fitzgerald
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - Rebecca Welly
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - Candace C. Rowles
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - Kylie Schaller
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
- Department of Biochemistry, University of Missouri, Columbia, MO, United States
| | - Shahad Bukhary
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - Christopher P. Baines
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States
| | - R. Scott Rector
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
- Internal Medicine-Division of Gastroenterology and Hepatology, University of Missouri, Columbia, MO, United States
- Research Service, Truman VA Memorial Hospital, Columbia, MO, United States
| | - Jaume Padilla
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States
| | - Camila Manrique-Acevedo
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Missouri Columbia School of Medicine, Columbia, MO, United States
| | - Dennis B. Lubahn
- Department of Biochemistry, University of Missouri, Columbia, MO, United States
| | - Victoria J. Vieira-Potter
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| |
Collapse
|
12
|
Källsten L, Almamoun R, Pierozan P, Nylander E, Sdougkou K, Martin JW, Karlsson O. Adult Exposure to Di-N-Butyl Phthalate (DBP) Induces Persistent Effects on Testicular Cell Markers and Testosterone Biosynthesis in Mice. Int J Mol Sci 2022; 23:ijms23158718. [PMID: 35955852 PMCID: PMC9369267 DOI: 10.3390/ijms23158718] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 11/16/2022] Open
Abstract
Studies indicate that phthalates are endocrine disruptors affecting reproductive health. One of the most commonly used phthalates, di-n-butyl phthalate (DBP), has been linked with adverse reproductive health outcomes in men, but the mechanisms behind these effects are still poorly understood. Here, adult male mice were orally exposed to DBP (10 or 100 mg/kg/day) for five weeks, and the testis and adrenal glands were collected one week after the last dose, to examine more persistent effects. Quantification of testosterone, androstenedione, progesterone and corticosterone concentrations by liquid chromatography-mass spectrometry showed that testicular testosterone was significantly decreased in both DBP treatment groups, whereas the other steroids were not significantly altered. Western blot analysis of testis revealed that DBP exposure increased the levels of the steroidogenic enzymes CYP11A1, HSD3β2, and CYP17A1, the oxidative stress marker nitrotyrosine, and the luteinizing hormone receptor (LHR). The analysis further demonstrated increased levels of the germ cell marker DAZL, the Sertoli cell markers vimentin and SOX9, and the Leydig cell marker SULT1E1. Overall, the present work provides more mechanistic understanding of how adult DBP exposure can induce effects on the male reproductive system by affecting several key cells and proteins important for testosterone biosynthesis and spermatogenesis, and for the first time shows that these effects persist at least one week after the last dose. It also demonstrates impairment of testosterone biosynthesis at a lower dose than previously reported.
Collapse
|
13
|
Kim YJ, Park JE, Chung JY, Kim JY, Lee SG, Lee SJ, Yu WJ, Kim HY, Kim HJ, Koh H, Bae HR, Yoo YH, Kim JM. Constitutive expression of cytochrome P450 1B1 endows testicular Leydig cells with susceptibility to 7,12-dimethylbenzanthracene-induced cell death. J Toxicol Sci 2022; 47:317-326. [PMID: 35908932 DOI: 10.2131/jts.47.317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Testicular Leydig cells produce testosterone through the participation of steroidogenic proteins. The CYP1B1 enzyme has been shown to catalyze 7,12-dimethylbenzanthracene (DMBA), a representative polycyclic aromatic hydrocarbon. We hypothesized that exposure to DMBA causes Leydig cell cytotoxicity through activation of CYP1B1. Leydig cells were exposed to various concentrations of DMBA for the induction of CYP1B1 expression and activity. The status of CYP1B1 function was monitored by evaluation of cytotoxicity-mediated cell death. Our data show that exposure to DMBA causes cytotoxicity in Leydig cells by CYP1B1 activation. DMBA evoked a significant increase in the generation of reactive oxygen species (ROS) by which the depolarization of mitochondrial membrane potential (MMP) is initiated and caspase-3 activation is augmented. The knockdown of CYP1B1 expression resulted in the suppression of DMBA-induced apoptosis via reduced p53 activation and caspase-3 activation, suggesting that a final metabolite of DMBA (i.e., DMBA-DE) bioactivated by CYP1B1 induces p53 activation by binding to DNA and subsequently causing apoptosis via caspase-3 activation. This finding provides evidence for constitutive expression of CYP1B1 in Leydig cells, which is a trait that only requires an initiating signal for its activity. Further research on CYP1B1 activation-provoked steroid metabolism in Leydig cells may provide decisive clues for elucidating its innate function.
Collapse
Affiliation(s)
- Yoon-Jae Kim
- Department of Anatomy and Cell Biology, College of Medicine, Dong-A University, Korea
| | - Ji-Eun Park
- Department of Anatomy and Cell Biology, College of Medicine, Dong-A University, Korea
| | - Jin-Yong Chung
- Department of Anatomy and Cell Biology, College of Medicine, Dong-A University, Korea
| | - Ji Young Kim
- Department of Anatomy and Cell Biology, College of Medicine, Dong-A University, Korea
| | - Seung Gee Lee
- Department of Anatomy and Cell Biology, College of Medicine, Dong-A University, Korea
| | - Seung-Jin Lee
- Developmental and Reproductive Toxicology Research Group, Korea Institute of Toxicology, Korea
| | - Wook-Joon Yu
- Developmental and Reproductive Toxicology Research Group, Korea Institute of Toxicology, Korea
| | - Hye Young Kim
- Department of Anatomy and Cell Biology, College of Medicine, Dong-A University, Korea
| | - Hyeon Jun Kim
- Department of Orthopaedic Surgery, College of Medicine, Dong-A University, Korea
| | - Hyungjong Koh
- Department of Pharmacology, College of Medicine, Dong-A University, Korea
| | - Hae-Rahn Bae
- Departmens of Physiology, College of Medicine, Dong-A University, Korea
| | - Young Hyun Yoo
- Department of Anatomy and Cell Biology, College of Medicine, Dong-A University, Korea
| | - Jong-Min Kim
- Department of Anatomy and Cell Biology, College of Medicine, Dong-A University, Korea
| |
Collapse
|
14
|
Da Costa KDA, Malvezzi H, Dobo C, Neme RM, Filippi RZ, Aloia TPA, Prado ER, Meola J, Piccinato CDA. Site-Specific Regulation of Sulfatase and Aromatase Pathways for Estrogen Production in Endometriosis. Front Mol Biosci 2022; 9:854991. [PMID: 35591944 PMCID: PMC9110888 DOI: 10.3389/fmolb.2022.854991] [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: 01/14/2022] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
Endometriosis is a highly prevalent gynecological disease characterized by lesions in different sites. Regulation of specific estrogen pathways may favor the formation of distinct microenvironments and the progression of endometriosis. However, no study has simultaneously evaluated the gene and protein regulation of the main estrogen-synthesizing enzymes in endometriosis. Thus, our goals were to study the relationship between gene and protein expression of aromatase (CYP19A1 or ARO), steroid sulfatase (STS), and hydroxysteroid 17-beta dehydrogenase (HSD17B1) in superficial (SUP), ovarian (OMA), and deep infiltrating (DIE) endometriotic lesion sites as well as in the eutopic endometrium of patients with (EE) and without (control) endometriosis in the same and large cohort of patients. The site-specific expression of these enzymes within different cells (glandular and stromal components) was also explored. The study included 108 patients surgically diagnosed with endometriosis who provided biopsies of EE and endometriotic lesions and 16 disease-free patients who collected normal endometrium tissue. Our results showed that CYP19A1 was detected in all endometriosis tissues and was in higher levels than in control. Unique patterns of the STS and HSD17B1 levels showed that they were most closely regulated in all tissues, with manifestation at greater levels in DIE compared to the other endometriotic lesion sites, OMA and SUP. Gene and protein expression of ARO, STS, and HSD17B1 occurred at different rates in endometriotic sites or EE. The distinctive levels of these estrogen-synthesizing enzymes in each endometriotic site support the hypothesis of a tissue microenvironment that can both influence and be influenced by the expression of different estrogenic pathways, locally affecting the availability of estrogen needed for maintenance and progression of endometriotic lesions.
Collapse
Affiliation(s)
| | | | - Cristine Dobo
- Hospital Israelita Albert Einstein, São Paulo, Brazil
- Department of Clinical Pathology, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Rosa Maria Neme
- Hospital Israelita Albert Einstein, São Paulo, Brazil
- Centro de Endometriose São Paulo, Av. República Do Líbano, São Paulo, Brazil
| | - Renée Zon Filippi
- Hospital Israelita Albert Einstein, São Paulo, Brazil
- Department of Clinical Pathology, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | | | | | - Juliana Meola
- Department of Gynaecology & Obstetrics, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Carla de Azevedo Piccinato
- Hospital Israelita Albert Einstein, São Paulo, Brazil
- Department of Gynaecology & Obstetrics, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
- *Correspondence: Carla de Azevedo Piccinato,
| |
Collapse
|
15
|
Triazoles Synthesis & Applications as Nonsteroidal Aromatase Inhibitors for Hormone-Dependent Breast Cancer Treatment. HETEROATOM CHEMISTRY 2022. [DOI: 10.1155/2022/5349279] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
In the last few years, nonsteroidal aromatase inhibitors (AIs) have been emerged as promising agents for treating hormone-dependent breast cancer in postmenopausal women because of their inhibitory effect on estrogen synthesis. Indeed, these compounds can block the activity of aromatase, the enzyme that intervenes in the last steps of estrogen production pathway. Triazoles are the core structures of nonsteroidal AIs. The nitrogen atom of the triazole moiety plays a fundamental role in the aromatase functionality by interacting with the iron ions of the heme group. In general, AIs possess numerous advantages as they quench the last step of estrogen synthesis without any inhibitory effects on the production of other steroids produced via the same pathway. Some AIs as anastrozole, letrozole, and vorozole have already been approved by the Food and Drug Administration in the treatment of breast cancer. The previously mentioned compounds present severe and adverse effects as polycystic ovary syndrome (PCOS), resistance onset on long-term treatments, and a higher risk of bone fractures. This review focuses intensively on the role of AIs in the treatment of hormone-sensitive types of cancers, especially the role of triazoles as nonsteroidal AIs. Also, the review provides an overview about the chemistry of triazoles along with the different methods by which the
-triazoles and s-triazoles are synthesized.
Collapse
|
16
|
McMillin SL, Minchew EC, Lowe DA, Spangenburg EE. Skeletal muscle wasting: the estrogen side of sexual dimorphism. Am J Physiol Cell Physiol 2022; 322:C24-C37. [PMID: 34788147 PMCID: PMC8721895 DOI: 10.1152/ajpcell.00333.2021] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The importance of defining sex differences across various biological and physiological mechanisms is more pervasive now than it has been over the past 15-20 years. As the muscle biology field pushes to identify small molecules and interventions to prevent, attenuate, or even reverse muscle wasting, we must consider the effect of sex as a biological variable. It should not be assumed that a therapeutic will affect males and females with equal efficacy or equivalent target affinities under conditions where muscle wasting is observed. With that said, it is not surprising to find that we have an unclear or even a poor understanding of the effects of sex or sex hormones on muscle wasting conditions. Although recent investigations are beginning to establish experimental approaches that will allow investigators to assess the impact of sex-specific hormones on muscle wasting, the field still needs rigorous scientific tools that will allow the community to address critical hypotheses centered around sex hormones. The focus of this review is on female sex hormones, specifically estrogens, and the roles that these hormones and their receptors play in skeletal muscle wasting conditions. With the overall review goal of assembling the current knowledge in the area of sexual dimorphism driven by estrogens with an effort to provide insights to interested physiologists on necessary considerations when trying to assess models for potential sex differences in cellular and molecular mechanisms of muscle wasting.
Collapse
Affiliation(s)
- Shawna L. McMillin
- 1Division of Rehabilitation Science, Department of Rehabilitation Medicine, Medical School, University of Minnesota, Minneapolis, Minnesota,2Division of Physical Therapy, Department of Rehabilitation Medicine, Medical School, University of Minnesota, Minneapolis, Minnesota
| | - Everett C. Minchew
- 3Department of Physiology, Brody School of Medicine, East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, North Carolina
| | - Dawn A. Lowe
- 1Division of Rehabilitation Science, Department of Rehabilitation Medicine, Medical School, University of Minnesota, Minneapolis, Minnesota,2Division of Physical Therapy, Department of Rehabilitation Medicine, Medical School, University of Minnesota, Minneapolis, Minnesota
| | - Espen E. Spangenburg
- 3Department of Physiology, Brody School of Medicine, East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, North Carolina
| |
Collapse
|
17
|
Bjune JI, Strømland PP, Jersin RÅ, Mellgren G, Dankel SN. Metabolic and Epigenetic Regulation by Estrogen in Adipocytes. Front Endocrinol (Lausanne) 2022; 13:828780. [PMID: 35273571 PMCID: PMC8901598 DOI: 10.3389/fendo.2022.828780] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 01/24/2022] [Indexed: 12/12/2022] Open
Abstract
Sex hormones contribute to differences between males and females in body fat distribution and associated disease risk. Higher concentrations of estrogens are associated with a more gynoid body shape and with more fat storage on hips and thighs rather than in visceral depots. Estrogen-mediated protection against visceral adiposity is shown in post-menopausal women with lower levels of estrogens and the reduction in central body fat observed after treatment with hormone-replacement therapy. Estrogen exerts its physiological effects via the estrogen receptors (ERα, ERβ and GPR30) in target cells, including adipocytes. Studies in mice indicate that estrogen protects against adipose inflammation and fibrosis also before the onset of obesity. The mechanisms involved in estrogen-dependent body fat distribution are incompletely understood, but involve, e.g., increased mTOR signaling and suppression of autophagy and adipogenesis/lipid storage. Estrogen plays a key role in epigenetic regulation of adipogenic genes by interacting with enzymes that remodel DNA methylation and histone tail post-translational modifications. However, more studies are needed to map the differential epigenetic effects of ER in different adipocyte subtypes, including those in subcutaneous and visceral adipose tissues. We here review recent discoveries of ER-mediated transcriptional and epigenetic regulation in adipocytes, which may explain sexual dimorphisms in body fat distribution and obesity-related disease risk.
Collapse
Affiliation(s)
- Jan-Inge Bjune
- Hormone Laboratory, Department of Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Pouda Panahandeh Strømland
- Hormone Laboratory, Department of Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
| | - Regine Åsen Jersin
- Hormone Laboratory, Department of Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Gunnar Mellgren
- Hormone Laboratory, Department of Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Simon Nitter Dankel
- Hormone Laboratory, Department of Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- *Correspondence: Simon Nitter Dankel,
| |
Collapse
|
18
|
White Adipose Tissue Depots Respond to Chronic Beta-3 Adrenergic Receptor Activation in a Sexually Dimorphic and Depot Divergent Manner. Cells 2021; 10:cells10123453. [PMID: 34943961 PMCID: PMC8700379 DOI: 10.3390/cells10123453] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 11/28/2021] [Accepted: 11/30/2021] [Indexed: 12/24/2022] Open
Abstract
Beta-3 adrenergic receptor activation via exercise or CL316,243 (CL) induces white adipose tissue (WAT) browning, improves glucose tolerance, and reduces visceral adiposity. Our aim was to determine if sex or adipose tissue depot differences exist in response to CL. Daily CL injections were administered to diet-induced obese male and female mice for two weeks, creating four groups: male control, male CL, female control, and female CL. These groups were compared to determine the main and interaction effects of sex (S), CL treatment (T), and WAT depot (D). Glucose tolerance, body composition, and energy intake and expenditure were assessed, along with perigonadal (PGAT) and subcutaneous (SQAT) WAT gene and protein expression. CL consistently improved glucose tolerance and body composition. Female PGAT had greater protein expression of the mitochondrial uncoupling protein 1 (UCP1), while SQAT (S, p < 0.001) was more responsive to CL in increasing UCP1 (S×T, p = 0.011) and the mitochondrial biogenesis induction protein, PPARγ coactivator 1α (PGC1α) (S×T, p = 0.026). Females also displayed greater mitochondrial OXPHOS (S, p < 0.05) and adiponectin protein content (S, p < 0.05). On the other hand, male SQAT was more responsive to CL in increasing protein levels of PGC1α (S×T, p = 0.046) and adiponectin (S, p < 0.05). In both depots and in both sexes, CL significantly increased estrogen receptor beta (ERβ) and glucose-related protein 75 (GRP75) protein content (T, p < 0.05). Thus, CL improves systemic and adipose tissue-specific metabolism in both sexes; however, sex differences exist in the WAT-specific effects of CL. Furthermore, across sexes and depots, CL affects estrogen signaling by upregulating ERβ.
Collapse
|
19
|
Maccallini C, Gallorini M, Sisto F, Akdemir A, Ammazzalorso A, De Filippis B, Fantacuzzi M, Giampietro L, Carradori S, Cataldi A, Amoroso R. New azolyl-derivatives as multitargeting agents against breast cancer and fungal infections: synthesis, biological evaluation and docking study. J Enzyme Inhib Med Chem 2021; 36:1632-1645. [PMID: 34289751 PMCID: PMC8300937 DOI: 10.1080/14756366.2021.1954918] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 01/06/2023] Open
Abstract
Nonsteroidal aromatase inhibitors (NSAIs) are well-established drugs for the therapy of breast cancer. However, they display some serious side effects, and their efficacy can be compromised by the development of chemoresistance. Previously, we have reported different indazole-based carbamates and piperidine-sulphonamides as potent aromatase inhibitors. Starting from the most promising compounds, here we have synthesised new indazole and triazole derivatives and evaluated their biological activity as potential dual agents, targeting both the aromatase and the inducible nitric oxide synthase, being this last dysregulated in breast cancer. Furthermore, selected compounds were evaluated as antiproliferative and cytotoxic agents in the MCF-7 cell line. Moreover, considering the therapeutic diversity of azole-based compounds, all the synthesized compounds were also evaluated as antifungals on different Candida strains. A docking study, as well as molecular dynamics simulation, were carried out to shed light on the binding mode of the most interesting compound into the different target enzymes catalytic sites.
Collapse
Affiliation(s)
- Cristina Maccallini
- Department of Pharmacy, University “G. d’Annunzio” of Chieti -Pescara, Chieti, Italy
| | - Marialucia Gallorini
- Department of Pharmacy, University “G. d’Annunzio” of Chieti -Pescara, Chieti, Italy
| | - Francesca Sisto
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Atilla Akdemir
- Department of Pharmacology, Faculty of Pharmacy, Bezmialem Vakif University, Computer-aided drug discovery laboratory, Istanbul, Turkey
| | | | - Barbara De Filippis
- Department of Pharmacy, University “G. d’Annunzio” of Chieti -Pescara, Chieti, Italy
| | | | - Letizia Giampietro
- Department of Pharmacy, University “G. d’Annunzio” of Chieti -Pescara, Chieti, Italy
| | - Simone Carradori
- Department of Pharmacy, University “G. d’Annunzio” of Chieti -Pescara, Chieti, Italy
- Department of Pharmacology, Faculty of Pharmacy, Bezmialem Vakif University, Computer-aided drug discovery laboratory, Istanbul, Turkey
| | - Amelia Cataldi
- Department of Pharmacy, University “G. d’Annunzio” of Chieti -Pescara, Chieti, Italy
| | - Rosa Amoroso
- Department of Pharmacy, University “G. d’Annunzio” of Chieti -Pescara, Chieti, Italy
| |
Collapse
|
20
|
Saldanha CJ. Glial estradiol synthesis after brain injury. CURRENT OPINION IN ENDOCRINE AND METABOLIC RESEARCH 2021; 21:100298. [PMID: 35274063 PMCID: PMC8903152 DOI: 10.1016/j.coemr.2021.100298] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Glial cells are important contributors to the hormonal milieu of the brain, particularly following damage. In birds and mammals, neural injury induces the expression of aromatase in astroglia at and around the site of damage. This review describes the progression of our understanding about the incidence, regulation, and function of estrogens synthesized in glia. Following a quick discussion of the landmark studies that first demonstrated steroidogenesis in glia, I go on to describe how the inflammatory response following perturbation of the brain results in the transcription of aromatase and the resultant rise in local estradiol. I end with several unanswered questions, the answers to which may reveal the precise manner in which neurosteroids protect the brain from injury, both prior to and immediately following injury.
Collapse
Affiliation(s)
- Colin J Saldanha
- Dept of Neuroscience and Center for Behavioral Neuroscience, American University, 4400 Massachusetts Avenue NW, Washington DC 20016
| |
Collapse
|
21
|
Bolt MJ, Singh P, Obkirchner CE, Powell RT, Mancini MG, Szafran AT, Stossi F, Mancini MA. Endocrine disrupting chemicals differentially alter intranuclear dynamics and transcriptional activation of estrogen receptor-α. iScience 2021; 24:103227. [PMID: 34712924 PMCID: PMC8529556 DOI: 10.1016/j.isci.2021.103227] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 08/30/2021] [Accepted: 09/30/2021] [Indexed: 11/21/2022] Open
Abstract
Transcription is a highly regulated sequence of stochastic processes utilizing many regulators, including nuclear receptors (NR) that respond to stimuli. Endocrine disrupting chemicals (EDCs) in the environment can compete with natural ligands for nuclear receptors to alter transcription. As nuclear dynamics can be tightly linked to transcription, it is important to determine how EDCs affect NR mobility. We use an EPA-assembled set of 45 estrogen receptor-α (ERα) ligands and EDCs in our engineered PRL-Array model to characterize their effect upon transcription using fluorescence in situ hybridization and fluorescence recovery after photobleaching (FRAP). We identified 36 compounds that target ERα-GFP to a transcriptionally active, visible locus. Using a novel method for multi-region FRAP analysis we find a strong negative correlation between ERα mobility and inverse agonists. Our findings indicate that ERα mobility is not solely tied to transcription but affected highly by the chemical class binding the receptor. Development of a new algorithm for multi-foci FRAP analysis ERα agonists can be segregated into fast-moving and slow-moving receptor groups ERα inverse agonists receptor mobility inversely correlates with transcription Steroidal compounds result in a slower moving receptor than other classes
Collapse
Affiliation(s)
- Michael J Bolt
- Center for Advanced Microscopy and Image Informatics, Institute of Biosciences & Technology, Texas A&M University, Houston, TX 77030, USA.,Center for Translational Cancer Research, Institute of Biosciences & Technology, Texas A&M University, Houston, TX 77030, USA
| | - Pankaj Singh
- Center for Advanced Microscopy and Image Informatics, Institute of Biosciences & Technology, Texas A&M University, Houston, TX 77030, USA.,Center for Translational Cancer Research, Institute of Biosciences & Technology, Texas A&M University, Houston, TX 77030, USA
| | - Caroline E Obkirchner
- Center for Advanced Microscopy and Image Informatics, Institute of Biosciences & Technology, Texas A&M University, Houston, TX 77030, USA.,Center for Translational Cancer Research, Institute of Biosciences & Technology, Texas A&M University, Houston, TX 77030, USA
| | - Reid T Powell
- Center for Translational Cancer Research, Institute of Biosciences & Technology, Texas A&M University, Houston, TX 77030, USA
| | - Maureen G Mancini
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Adam T Szafran
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Fabio Stossi
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA.,Center for Advanced Microscopy and Image Informatics, Institute of Biosciences & Technology, Texas A&M University, Houston, TX 77030, USA
| | - Michael A Mancini
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA.,Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX 77030, USA.,Center for Advanced Microscopy and Image Informatics, Institute of Biosciences & Technology, Texas A&M University, Houston, TX 77030, USA.,Center for Translational Cancer Research, Institute of Biosciences & Technology, Texas A&M University, Houston, TX 77030, USA
| |
Collapse
|
22
|
Cai S, Feng Y, Ye J, Deng Y, Cai Z, Zhu X, Liu R, Zhang Y, Zou Z, Tang Z, Han Z, Hon CT, Zhong W, He H. The prognostic roles of CYP19A1 expression in bladder cancer patients of different genders. Transl Androl Urol 2021; 10:3579-3590. [PMID: 34733654 PMCID: PMC8511542 DOI: 10.21037/tau-21-400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 08/02/2021] [Indexed: 12/24/2022] Open
Abstract
Background The incidence of bladder cancer (BCa) in male is approximately three to four times higher than in female, but the oncological outcomes in female patients with BCa are significantly worse than in male patients. Although many biomarkers have been identified in recent decades to predict the prognosis of BCa patients, few of them are able to distinguish the prognosis of BCa patients with gender difference. Aromatase encoded by the CYP19A1 gene catalyzes the conversion of androgens to estrogens. In this study, we investigate the prognosis significance of CYP19A1 expression considering the gender difference in BCa patients from four available public databases. Methods Four available public databases of BCa, including GSE13507, TCGA-BLCA, E-MTAB-4321, and E-MTAB-1803, were utilized in this analysis. The overall survival (OS) and progression-free survival (PFS) in different stages and genders were evaluated using the Kaplan-Meier analysis based on the optimal cut-off values of CYP19A1 expression. Then, Gene Set Enrichment Analysis (GSEA) were further performed to explore the potential biologic pathways by altering CYP19A1 expression in BCa patients. Results The results showed that patients with high CYP19A1 expression had a poorer outcome compared with those with low expression in both BCa cohorts in general. Higher CYP19A1 expression in male patients were significantly associated with shorter survival for either non-muscle-invasive bladder cancer (NMIBC) or muscle-invasive bladder cancer (MIBC). However, female NMIBC patients with high CYP19A1 expression were identified to have a better prognosis, whereas high CYP19A1 expression in female MIBC patients were significantly associated with poorer survival. The result of the GSEA showed that different outcomes in female and male patients with NMIBC were related to the interaction of CYP19A1 and the cell-cycle-related pathways. Conclusions These findings demonstrated that CYP19A1 expression might have a potential role in distinguishing the prognosis of female BCa patients dependent on tumor stage. Our results provide new insights for aromatase-mediated BCa therapy.
Collapse
Affiliation(s)
- Shanghua Cai
- Department of Urology, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Yuanfa Feng
- Department of Urology, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Jianheng Ye
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yulin Deng
- Department of Urology, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Zhiduan Cai
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xuejin Zhu
- Department of Urology, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Ren Liu
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yixun Zhang
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Zhihao Zou
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Zhenfeng Tang
- Department of Urology, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Zhaodong Han
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Chi Tin Hon
- Macau Institute of Systems Engineering, Macau University of Science and Technology, Macau, China
| | - Weide Zhong
- Department of Urology, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China.,Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China.,Department of Urology, Huizhou Municipal Central Hospital, Huizhou, China.,Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, China
| | - Huichan He
- Department of Urology, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
23
|
Regulation of Estradiol Synthesis by Aromatase Interacting Partner in Breast (AIPB). Mol Cell Biol 2021; 41:e0035721. [PMID: 34460330 DOI: 10.1128/mcb.00357-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Estradiol is essential for the development of female sex characteristics and fertility. Postmenopausal women and breast cancer patients have high levels of estradiol. Aromatase catalyzes estradiol synthesis; however, the factors regulating aromatase activity are unknown. We identified a new 22-kDa protein, aromatase interacting partner in breast (AIPB), from the endoplasmic reticulum of human breast tissue. AIPB expression is reduced in tumorigenic breast and further reduced in triple-negative tumors. Like that of aromatase, AIPB expression is induced by nonsteroidal estrogen. We found that AIPB and aromatase interact in nontumorigenic and tumorigenic breast tissues and cells. In tumorigenic cells, conditional AIPB overexpression decreased estradiol, and blocking AIPB availability with an AIPB-binding antibody increased estradiol. Estradiol synthesis is highly increased in AIPB knockdown cells, suggesting that the newly identified AIPB protein is important for aromatase activity and a key modulator of estradiol synthesis. Thus, a change in AIPB protein expression may represent an early event in tumorigenesis and be predictive of an increased risk of developing breast cancer.
Collapse
|
24
|
Chaudhary H, Patel J, Jain NK, Joshi R. The role of polymorphism in various potential genes on polycystic ovary syndrome susceptibility and pathogenesis. J Ovarian Res 2021; 14:125. [PMID: 34563259 PMCID: PMC8466925 DOI: 10.1186/s13048-021-00879-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 09/09/2021] [Indexed: 12/27/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrinopathies affecting the early reproductive age in women, whose pathophysiology perplexes many researchers till today. This syndrome is classically categorized by hyperandrogenism and/or hyperandrogenemia, menstrual and ovulatory dysfunction, bulky multi follicular ovaries on Ultrasonography (USG), and metabolic abnormalities such as hyperinsulinemia, dyslipidemia, obesity. The etiopathogenesis of PCOS is not fully elucidated, but it seems that the hypothalamus-pituitary-ovarian axis, ovarian, and/or adrenal androgen secretion may contribute to developing the syndrome. Infertility and poor reproductive health in women's lives are highly associated with elevated levels of androgens. Studies with ovarian theca cells taken from PCOS women have demonstrated increased androgen production due to augmented ovarian steroidogenesis attributed to mainly altered expression of critical enzymes (Cytochrome P450 enzymes: CYP17, CYP21, CYP19, CYP11A) in the steroid hormone biosynthesis pathway. Despite the heterogeneity of PCOS, candidate gene studies are the widely used technique to delineate the genetic variants and analyze for the correlation of androgen biosynthesis pathway and those affecting the secretion or action of insulin with PCOS etiology. Linkage and association studies have predicted the relationship between genetic variants and PCOS risk among families or populations. Several genes have been proposed as playing a role in the etiopathogenesis of PCOS, and the presence of mutations and/or polymorphisms has been discovered, which suggests that PCOS has a vital heritable component. The following review summarizes the influence of polymorphisms in crucial genes of the steroidogenesis pathway leading to intraovarian hyperandrogenism which can result in PCOS.
Collapse
Affiliation(s)
- Hiral Chaudhary
- Department of Biochemistry and Forensic Science, University School of Sciences, Gujarat University, Ahmedabad, Gujarat 380009 India
| | - Jalpa Patel
- Department of Biochemistry and Forensic Science, University School of Sciences, Gujarat University, Ahmedabad, Gujarat 380009 India
| | - Nayan K. Jain
- Department of Life Science, University School of Sciences, Gujarat University, Ahmedabad, Gujarat 380009 India
| | - Rushikesh Joshi
- Department of Biochemistry and Forensic Science, University School of Sciences, Gujarat University, Ahmedabad, Gujarat 380009 India
| |
Collapse
|
25
|
Reddy HM, Bhattacharya R, Tiwari S, Mishra K, Annapurna P, Jehan Z, Praveena NM, Alex JL, Dhople VM, Singh L, Sivaramakrishnan M, Chaturvedi A, Rangaraj N, Shiju TM, Sreedevi B, Kumar S, Dereddi RR, Rayabandla SM, Jesudasan RA. Y chromosomal noncoding RNAs regulate autosomal gene expression via piRNAs in mouse testis. BMC Biol 2021; 19:198. [PMID: 34503492 PMCID: PMC8428117 DOI: 10.1186/s12915-021-01125-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 08/17/2021] [Indexed: 12/03/2022] Open
Abstract
Background Deciphering the functions of Y chromosome in mammals has been slow owing to the presence of repeats. Some of these repeats transcribe coding RNAs, the roles of which have been studied. Functions of the noncoding transcripts from Y chromosomal repeats however, remain unclear. While a majority of the genes expressed during spermatogenesis are autosomal, mice with different deletions of the long arm of the Y chromosome (Yq) were previously also shown to be characterized by subfertility, sterility and sperm abnormalities, suggesting the presence of effectors of spermatogenesis at this location. Here we report a set of novel noncoding RNAs from mouse Yq and explore their connection to some of the autosomal genes expressed in testis. Results We describe a set of novel mouse male-specific Y long arm (MSYq)-derived long noncoding (lnc) transcripts, named Pirmy and Pirmy-like RNAs. Pirmy shows a large number of splice variants in testis. We also identified Pirmy-like RNAs present in multiple copies at different loci on mouse Y chromosome. Further, we identified eight differentially expressed autosome-encoded sperm proteins in a mutant mouse strain, XYRIIIqdel (2/3 Yq-deleted). Pirmy and Pirmy-like RNAs have homology to 5′/3′UTRs of these deregulated autosomal genes. Several lines of experiments show that these short homologous stretches correspond to piRNAs. Thus, Pirmy and Pirmy-like RNAs act as templates for several piRNAs. In vitro functional assays reveal putative roles for these piRNAs in regulating autosomal genes. Conclusions Our study elucidates a set of autosomal genes that are potentially regulated by MSYq-derived piRNAs in mouse testis. Sperm phenotypes from the Yq-deleted mice seem to be similar to that reported in inter-specific male-sterile hybrids. Taken together, this study provides novel insights into possible role of MSYq-derived ncRNAs in male sterility and speciation. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-021-01125-x.
Collapse
Affiliation(s)
- Hemakumar M Reddy
- Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad, Telangana, 500007, India.,Present address: Brown University BioMed Division, Department of Molecular Biology, Cell Biology and Biochemistry, 185 Meeting Street room 257, Sidney Frank Life Sciences Building, Providence, RI, 02912, USA
| | - Rupa Bhattacharya
- Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad, Telangana, 500007, India.,, Pennington, NJ, 08534, USA
| | - Shrish Tiwari
- Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad, Telangana, 500007, India
| | - Kankadeb Mishra
- Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad, Telangana, 500007, India.,Department of Cell Biology, Memorial Sloan Kettering Cancer Centre, Rockefeller Research Laboratory, 430 East 67th Street, RRL 445, New York, NY, 10065, USA
| | - Pranatharthi Annapurna
- Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad, Telangana, 500007, India.,Departments of Orthopaedic Surgery & Bioengineering, University of Pennsylvania, 376A Stemmler Hall, 36th Street & Hamilton Walk, Philadelphia, PA, 19104, USA
| | - Zeenath Jehan
- Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad, Telangana, 500007, India.,Department of Genetics and Molecular Medicines, Vasavi Medical and Research Centre, 6-1-91 Khairatabad, Hyderabad, 500 004, India
| | | | - Jomini Liza Alex
- Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad, Telangana, 500007, India
| | - Vishnu M Dhople
- Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad, Telangana, 500007, India.,Department of Functional Genomics, Ernst-Moritz-Arndt-University of Greifswald Interfaculty Institute for Genetics and Functional Genomics, Friedrich-Ludwig-Jahn-Straße 15 a, 17487, Greifswald, Germany
| | - Lalji Singh
- Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad, Telangana, 500007, India
| | - Mahadevan Sivaramakrishnan
- Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad, Telangana, 500007, India.,Jubilant Biosystems Ltd., #96, Industrial Suburb, 2nd Stage, Yeshwantpur, Bangalore, Karnataka, 560022, India
| | - Anurag Chaturvedi
- Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad, Telangana, 500007, India.,Environmental Genomics Group, School of Biosciences, University of Birmingham, Birmingham, UK
| | - Nandini Rangaraj
- Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad, Telangana, 500007, India
| | - Thomas Michael Shiju
- Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad, Telangana, 500007, India.,Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, 44120, USA
| | - Badanapuram Sreedevi
- Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad, Telangana, 500007, India
| | - Sachin Kumar
- Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad, Telangana, 500007, India
| | - Ram Reddy Dereddi
- Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad, Telangana, 500007, India.,Institute for Anatomy and Cell Biology, building-307, Heidelberg, Germany
| | - Sunayana M Rayabandla
- Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad, Telangana, 500007, India.,Telangana Social Welfare Residential Degree College for Women, Suryapet, Telangana, 508213, India
| | - Rachel A Jesudasan
- Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad, Telangana, 500007, India. .,Department of Genetics, Osmania University, Hyderabad, Telangana, 500007, India. .,Inter University Centre for Genomics & Gene Technology, Karyavattom Campus, University of Kerala, Trivandrum, Kerala, India.
| |
Collapse
|
26
|
Abaffy T, Matsunami H. 19-hydroxy Steroids in the Aromatase Reaction: Review on Expression and Potential Functions. J Endocr Soc 2021; 5:bvab050. [PMID: 34095690 PMCID: PMC8169043 DOI: 10.1210/jendso/bvab050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Indexed: 12/05/2022] Open
Abstract
Scientific evidence related to the aromatase reaction in various biological processes spanning from mid-1960 to today is abundant; however, as our analytical sensitivity increases, a new look at the old chemical reaction is necessary. Here, we review an irreversible aromatase reaction from the substrate androstenedione. It proceeds in 3 consecutive steps. In the first 2 steps, 19-hydroxy steroids are produced. In the third step, estrone is produced. They can dissociate from the enzyme complex and either accumulate in tissues or enter the blood. In this review, we want to highlight the potential importance of these 19-hydroxy steroids in various physiological and pathological conditions. We focus primarily on 19-hydroxy steroids, and in particular on the 19-hydroxyandrostenedione produced by the incomplete aromatase reaction. Using a PubMed database and the search term “aromatase reaction,” 19-hydroxylation of androgens and steroid measurements, we detail the chemistry of the aromatase reaction and list previous and current methods used to measure 19-hydroxy steroids. We present evidence of the existence of 19-hydroxy steroids in brain tissue, ovaries, testes, adrenal glands, prostate cancer, as well as during pregnancy and parturition and in Cushing’s disease. Based on the available literature, a potential involvement of 19-hydroxy steroids in the brain differentiation process, sperm motility, ovarian function, and hypertension is suggested and warrants future research. We hope that with the advancement of highly specific and sensitive analytical methods, future research into 19-hydroxy steroids will be encouraged, as much remains to be learned and discovered.
Collapse
Affiliation(s)
- Tatjana Abaffy
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27710, USA
| | - Hiroaki Matsunami
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27710, USA
| |
Collapse
|
27
|
Hung SW, Zhang R, Tan Z, Chung JPW, Zhang T, Wang CC. Pharmaceuticals targeting signaling pathways of endometriosis as potential new medical treatment: A review. Med Res Rev 2021; 41:2489-2564. [PMID: 33948974 PMCID: PMC8252000 DOI: 10.1002/med.21802] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 12/23/2020] [Accepted: 03/19/2021] [Indexed: 12/13/2022]
Abstract
Endometriosis (EM) is defined as endometrial tissues found outside the uterus. Growth and development of endometriotic cells in ectopic sites can be promoted via multiple pathways, including MAPK/MEK/ERK, PI3K/Akt/mTOR, NF-κB, Rho/ROCK, reactive oxidative stress, tumor necrosis factor, transforming growth factor-β, Wnt/β-catenin, vascular endothelial growth factor, estrogen, and cytokines. The underlying pathophysiological mechanisms include proliferation, apoptosis, autophagy, migration, invasion, fibrosis, angiogenesis, oxidative stress, inflammation, and immune escape. Current medical treatments for EM are mainly hormonal and symptomatic, and thus the development of new, effective, and safe pharmaceuticals targeting specific molecular and signaling pathways is needed. Here, we systematically reviewed the literature focused on pharmaceuticals that specifically target the molecular and signaling pathways involved in the pathophysiology of EM. Potential drug targets, their upstream and downstream molecules with key aberrant signaling, and the regulatory mechanisms promoting the growth and development of endometriotic cells and tissues were discussed. Hormonal pharmaceuticals, including melatonin, exerts proapoptotic via regulating matrix metallopeptidase activity while nonhormonal pharmaceutical sorafenib exerts antiproliferative effect via MAPK/ERK pathway and antiangiogenesis activity via VEGF/VEGFR pathway. N-acetyl cysteine, curcumin, and ginsenoside exert antioxidant and anti-inflammatory effects via radical scavenging activity. Natural products have high efficacy with minimal side effects; for example, resveratrol and epigallocatechin gallate have multiple targets and provide synergistic efficacy to resolve the complexity of the pathophysiology of EM, showing promising efficacy in treating EM. Although new medical treatments are currently being developed, more detailed pharmacological studies and large sample size clinical trials are needed to confirm the efficacy and safety of these treatments in the near future.
Collapse
Affiliation(s)
- Sze Wan Hung
- Department of Obstetrics and GynaecologyThe Chinese University of Hong KongHong Kong
| | - Ruizhe Zhang
- Department of Obstetrics and GynaecologyThe Chinese University of Hong KongHong Kong
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and GeneticsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou
| | - Zhouyurong Tan
- Department of Obstetrics and GynaecologyThe Chinese University of Hong KongHong Kong
| | | | - Tao Zhang
- Department of Obstetrics and GynaecologyThe Chinese University of Hong KongHong Kong
| | - Chi Chiu Wang
- Department of Obstetrics and GynaecologyThe Chinese University of Hong KongHong Kong
- Reproduction and Development, Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong Kong
- School of Biomedical SciencesThe Chinese University of Hong KongHong Kong
- Chinese University of Hong Kong‐Sichuan University Joint Laboratory in Reproductive MedicineThe Chinese University of Hong KongHong Kong
| |
Collapse
|
28
|
Harthern-Flint SL, Dolfing J, Mrozik W, Meynet P, Eland LE, Sim M, Davenport RJ. Experimental and Genomic Evaluation of the Oestrogen Degrading Bacterium Rhodococcus equi ATCC13557. Front Microbiol 2021; 12:670928. [PMID: 34276604 PMCID: PMC8281962 DOI: 10.3389/fmicb.2021.670928] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 05/27/2021] [Indexed: 12/12/2022] Open
Abstract
Rhodococcus equi ATCC13557 was selected as a model organism to study oestrogen degradation based on its previous ability to degrade 17α-ethinylestradiol (EE2). Biodegradation experiments revealed that R. equi ATCC13557 was unable to metabolise EE2. However, it was able to metabolise E2 with the major metabolite being E1 with no further degradation of E1. However, the conversion of E2 into E1 was incomplete, with 11.2 and 50.6% of E2 degraded in mixed (E1-E2-EE2) and E2-only conditions, respectively. Therefore, the metabolic pathway of E2 degradation by R. equi ATCC13557 may have two possible pathways. The genome of R. equi ATCC13557 was sequenced, assembled, and mapped for the first time. The genome analysis allowed the identification of genes possibly responsible for the observed biodegradation characteristics of R. equi ATCC13557. Several genes within R. equi ATCC13557 are similar, but not identical in sequence, to those identified within the genomes of other oestrogen degrading bacteria, including Pseudomonas putida strain SJTE-1 and Sphingomonas strain KC8. Homologous gene sequences coding for enzymes potentially involved in oestrogen degradation, most commonly a cytochrome P450 monooxygenase (oecB), extradiol dioxygenase (oecC), and 17β-hydroxysteroid dehydrogenase (oecA), were identified within the genome of R. equi ATCC13557. These searches also revealed a gene cluster potentially coding for enzymes involved in steroid/oestrogen degradation; 3-carboxyethylcatechol 2,3-dioxygenase, 2-hydroxymuconic semialdehyde hydrolase, 3-alpha-(or 20-beta)-hydroxysteroid dehydrogenase, 3-(3-hydroxy-phenyl)propionate hydroxylase, cytochrome P450 monooxygenase, and 3-oxosteroid 1-dehydrogenase. Further, the searches revealed steroid hormone metabolism gene clusters from the 9, 10-seco pathway, therefore R. equi ATCC13557 also has the potential to metabolise other steroid hormones such as cholesterol.
Collapse
Affiliation(s)
| | - Jan Dolfing
- School of Engineering, Newcastle University, Newcastle upon Tyne, United Kingdom.,Faculty Engineering and Environment, Northumbria University, Newcastle upon Tyne, United Kingdom
| | - Wojciech Mrozik
- School of Engineering, Newcastle University, Newcastle upon Tyne, United Kingdom.,Department of Inorganic Chemistry, Faculty of Pharmacy, Medical University of Gdańsk, Gdańsk, Poland
| | - Paola Meynet
- School of Engineering, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Lucy E Eland
- School of Computing Science, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Martin Sim
- School of Computing Science, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Russell J Davenport
- School of Engineering, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
29
|
Maternal stress programs a demasculinization of glutamatergic transmission in stress-related brain regions of aged rats. GeroScience 2021; 44:1047-1069. [PMID: 33983623 PMCID: PMC8116647 DOI: 10.1007/s11357-021-00375-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 04/20/2021] [Indexed: 12/20/2022] Open
Abstract
Brain aging may be programmed by early-life stress. Aging affects males and females differently, but how perinatal stress (PRS) affects brain aging between sexes is unknown. We showed behavioral and neurobiological sex differences in non-stressed control rats that were strongly reduced or inverted in PRS rats. In particular, PRS decreased risk-taking behavior, spatial memory, exploratory behavior, and fine motor behavior in male aged rats. In contrast, female aged PRS rats displayed only increased risk-taking behavior and reduced exploratory behavior. PRS induced large reductions in the expression of glutamate receptors in the ventral and dorsal hippocampus and prefrontal cortex only in male rats. PRS also reduced the expression of synaptic vesicle-associated proteins, glucocorticoid receptors (GR), and mineralocorticoid receptors (MR) in the ventral hippocampus of aged male rats. In contrast, in female aged rats, PRS enhanced the expression of MRs and brain-derived neurotrophic factor (BDNF) in the ventral hippocampus and the expression of glial fibrillary acidic protein (GFAP) and BDNF in the prefrontal cortex. A common PRS effect in both sexes was a reduction in exploratory behavior and metabotropic glutamate (mGlu2/3) receptors in the ventral hippocampus and prefrontal cortex. A multidimensional analysis revealed that PRS induced a demasculinization profile in glutamate-related proteins in the ventral and dorsal hippocampus and prefrontal cortex, as well as a demasculinization profile of stress markers only in the dorsal hippocampus. In contrast, defeminization was observed only in the ventral hippocampus. Measurements of testosterone and 17-β-estradiol in the plasma and aromatase in the dorsal hippocampus were consistent with a demasculinizing action of PRS. These findings confirm that the brains of males and females differentially respond to PRS and aging suggesting that females might be more protected against early stress and age-related inflammation and neurodegeneration. Taken together, these results may contribute to understanding how early environmental factors shape vulnerability to brain aging in both sexes and may lay the groundwork for future studies aimed at identifying new treatment strategies to improve the quality of life of older individuals, which is of particular interest given that there is a high growth of aging in populations around the world.
Collapse
|
30
|
Dommel S, Hoffmann A, Berger C, Kern M, Klöting N, Kannt A, Blüher M. Effects of Whole-Body Adenylyl Cyclase 5 ( Adcy5) Deficiency on Systemic Insulin Sensitivity and Adipose Tissue. Int J Mol Sci 2021; 22:4353. [PMID: 33919448 PMCID: PMC8122634 DOI: 10.3390/ijms22094353] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 04/16/2021] [Indexed: 02/07/2023] Open
Abstract
Genome-wide association studies have identified adenylyl cyclase type 5 (ADCY5) as candidate gene for diabetes-related quantitative traits and an increased risk of type 2 diabetes. Mice with a whole-body deletion of Adcy5 (Adcy5-/-) do not develop obesity, glucose intolerance and insulin resistance, have improved cardiac function and increased longevity. Here, we investigated Adcy5 knockout mice (Adcy5-/-) to test the hypothesis that changes in adipose tissue (AT) may contribute to the reported healthier phenotype. In contrast to previous reports, we found that deletion of Adcy5 did not confer any physiological or biochemical benefits. However, this unexpected finding allowed us to investigate the effects of Adcy5 depletion on AT independently of lower body weight and a metabolically healthier phenotype. Adcy5-/- mice exhibited an increased number of smaller adipocytes, lower mean adipocyte size and a distinct AT gene expression pattern with midline 1 (Mid1) as the most significantly downregulated gene compared to control mice. Our Adcy5-/- model challenges previously described beneficial effects of Adcy5 deficiency and suggests that targeting Adcy5 does not improve insulin sensitivity and may therefore limit the relevance of ADCY5 as potential drug target.
Collapse
Affiliation(s)
- Sebastian Dommel
- Medical Center, Medical Department III—Endocrinology, Nephrology, Rheumatology, University of Leipzig, 04103 Leipzig, Germany; (S.D.); (C.B.); (N.K.)
| | - Anne Hoffmann
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany; (A.H.); (M.K.)
| | - Claudia Berger
- Medical Center, Medical Department III—Endocrinology, Nephrology, Rheumatology, University of Leipzig, 04103 Leipzig, Germany; (S.D.); (C.B.); (N.K.)
| | - Matthias Kern
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany; (A.H.); (M.K.)
| | - Nora Klöting
- Medical Center, Medical Department III—Endocrinology, Nephrology, Rheumatology, University of Leipzig, 04103 Leipzig, Germany; (S.D.); (C.B.); (N.K.)
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany; (A.H.); (M.K.)
| | - Aimo Kannt
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 60596 Frankfurt am Main, Germany;
- Experimental Pharmacology, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
- Sanofi Diabetes Research and Development, 60596 Frankfurt am Main, Germany
| | - Matthias Blüher
- Medical Center, Medical Department III—Endocrinology, Nephrology, Rheumatology, University of Leipzig, 04103 Leipzig, Germany; (S.D.); (C.B.); (N.K.)
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany; (A.H.); (M.K.)
| |
Collapse
|
31
|
Comparison of Gonadal Transcriptomes Uncovers Reproduction-Related Genes with Sexually Dimorphic Expression Patterns in Diodon hystrix. Animals (Basel) 2021; 11:ani11041042. [PMID: 33917262 PMCID: PMC8068034 DOI: 10.3390/ani11041042] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/04/2021] [Accepted: 04/04/2021] [Indexed: 12/16/2022] Open
Abstract
Diodon hystrix is a new and emerging aquaculture species in south China. However, due to the lack of understanding of reproductive regulation, the management of breeding and reproduction under captivity remains a barrier for the commercial aquaculture of D. hystrix. More genetic information is needed to identify genes critical for gonadal development. Here, the first gonadal transcriptomes of D. hystrix were analyzed and 151.89 million clean reads were generated. All reads were assembled into 57,077 unigenes, and 24,574 could be annotated. By comparing the gonad transcriptomes, 11,487 differentially expressed genes were obtained, of which 4599 were upregulated and 6888 were downregulated in the ovaries. Using enrichment analyses, many functional pathways were found to be associated with reproduction regulation. A set of sex-biased genes putatively involved in gonad development and gametogenesis were identified and their sexually dimorphic expression patterns were characterized. The detailed transcriptomic data provide a useful resource for further research on D. hystrix reproductive manipulation.
Collapse
|
32
|
Genomic Structure of the Porcine CYP19 Locus and Expression of the CYP19A3 Paralog. Genes (Basel) 2021; 12:genes12040533. [PMID: 33917597 PMCID: PMC8067493 DOI: 10.3390/genes12040533] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 03/30/2021] [Accepted: 04/05/2021] [Indexed: 11/17/2022] Open
Abstract
Proper, tissue-specific regulation of CYP19, the gene encoding aromatase, the key enzyme of estrogen synthesis, is essential for reproductive processes. Here, we analyzed transcriptional regulation of the porcine CYP19 in female and male gonads and brain by 5'RACE and RT-PCR and comprehensively mapped the pig CYP19 locus by in silico analysis. Our data revealed that the complete locus, including three paralogous copies, CYP19A1, CYP19A2 and CYP19A3, spans approximately 330 kb of the porcine chromosome 1. The locus also harbors the first exon of the Gliomedin gene (GLDN) in reverse orientation. Only transcripts of the CYP19A3 paralog were substantially expressed in gonads and hypothalamus. We identified CYP19A3-associated untranslated exons approximately 160 kb and 50 kb distal from the first codon. The 5´ untranslated regions of transcripts were derived from either a proximal or from one of these distal untranslated exons. Transcripts including only untranslated exons could be amplified from testis, thus suggesting long non-coding transcripts. The data revealed an additional layer of complexity in the regulation of the porcine CYP19 locus. Tissue-specific expression is not only achieved by tissue- and stage-specific expression of the three different CYP19 paralogs, but also by directing the expression of CYP19A3 from different, proximal and distal promoter regions.
Collapse
|
33
|
Drzewiecka EM, Kozlowska W, Zmijewska A, Wydorski PJ, Franczak A. Electromagnetic Field (EMF) Radiation Alters Estrogen Release from the Pig Myometrium during the Peri-Implantation Period. Int J Mol Sci 2021; 22:ijms22062920. [PMID: 33805726 PMCID: PMC7999543 DOI: 10.3390/ijms22062920] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 12/17/2022] Open
Abstract
An electromagnetic field (EMF) may affect the functions of uterine tissues. This study hypothesized that EMF changes the estrogenic activity of pig myometrium during the peri-implantation period. Tissue was collected on days 15-16 of the gestation and incubated in the presence of EMF (50 and 120 Hz, 2 and 4 h). The cytochrome P450 aromatase type 3 (CYP19A3) and hydroxysteroid 17β dehydrogenase type 4 (HSD17B4) mRNA transcript abundance, cytochrome P450arom (aromatase), and 17β hydroxysteroid dehydrogenase 17βHSD) protein abundance and estrone (E1) and estradiol-17β (E2) release were examined using Real-Time PCR, Western blot and radioimmunoassay. Selected myometrial slices were treated with progesterone (P4) to determine whether it functions as a protector against EMF. CYP19A3 mRNA transcript abundance in slices treated with EMF was less at 50 Hz (2 h) and greater at 120 Hz (2 and 4 h). HSD17B4 mRNA transcript was greater in slices treated with EMF at 120 Hz (2 h). Progesterone diminished EMF-related effects on CYP19A3 and HSD17B4. When P4 was added, EMF had suppressive (50 and 120 Hz, 2 h) or enhancing (50 Hz, 4 h) effects on aromatase abundance. The E1 release was lower after 4 h of EMF treatment at 50 Hz and P4 did not protect myometrial E1 release. In conclusion, EMF alters the synthesis and release of E1 and did not affect E2 release in the myometrium during the peri-implantation period.
Collapse
Affiliation(s)
| | | | | | | | - Anita Franczak
- Correspondence: (E.M.D.); (A.F.); Tel.: +48-89-523-32-01 (E.M.D. & A.F.)
| |
Collapse
|
34
|
Melatonin Administration Accelerates Puberty Onset in Mice by Promoting FSH Synthesis. Molecules 2021; 26:molecules26051474. [PMID: 33803091 PMCID: PMC7963190 DOI: 10.3390/molecules26051474] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/02/2021] [Accepted: 03/06/2021] [Indexed: 12/20/2022] Open
Abstract
Although melatonin has been extensively studied in animal reproduction, the mechanism of melatonin in puberty remains elusive. This study was designed to explore the effect of intraperitoneal administration of melatonin on puberty onset in female mice. The injection of melatonin into postnatal days 10 mice at a dose of 15 mg/kg accelerated the puberty onset in mice. Mechanistically, there was no difference in physical growth and serum Leptin levels after melatonin administration. Meanwhile, the serum levels of reproductive hormones involved in hypothalamic-pituitary-ovarian axis, such as FSH and estrogen level in serum were increased. The mRNA levels of GnRH and GnRHr were not affected by melatonin, while the expressions of FSHβ in pituitary and Cyp19a1 in ovary were significantly up-regulated. In addition, melatonin still promoted FSH synthesis after ovariectomy. Furthermore, the enhanced activity of ERK1/2 signaling verified that the expression of FSHβ increased in pituitary. We confirmed that melatonin promoted the FSH synthesis in pituitary, thereby increased serum estrogen levels and ultimately accelerated puberty onset. However, these effects of melatonin may be pharmacological due to the high dose. This study would help us to understand the functions of melatonin in pubertal regulation comprehensively.
Collapse
|
35
|
Di Nardo G, Zhang C, Marcelli AG, Gilardi G. Molecular and Structural Evolution of Cytochrome P450 Aromatase. Int J Mol Sci 2021; 22:E631. [PMID: 33435208 PMCID: PMC7827799 DOI: 10.3390/ijms22020631] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/06/2021] [Accepted: 01/07/2021] [Indexed: 12/22/2022] Open
Abstract
Aromatase is the cytochrome P450 enzyme converting androgens into estrogen in the last phase of steroidogenesis. As estrogens are crucial in reproductive biology, aromatase is found in vertebrates and the invertebrates of the genus Branchiostoma, where it carries out the aromatization reaction of the A-ring of androgens that produces estrogens. Here, we investigate the molecular evolution of this unique and highly substrate-selective enzyme by means of structural, sequence alignment, and homology modeling, shedding light on its key role in species conservation. The alignments led to the identification of a core structure that, together with key and unique amino acids located in the active site and the substrate recognition sites, has been well conserved during evolution. Structural analysis shows what their roles are and the reason why they have been preserved. Moreover, the residues involved in the interaction with the redox partner and some phosphorylation sites appeared late during evolution. These data reveal how highly substrate-selective cytochrome P450 has evolved, indicating that the driving forces for evolution have been the optimization of the interaction with the redox partner and the introduction of phosphorylation sites that give the possibility of modulating its activity in a rapid way.
Collapse
Affiliation(s)
- Giovanna Di Nardo
- Department of Life Sciences and Systems Biology, University of Torino, via Accademia Albertina 13, 1023 Torino, Italy; (C.Z.); (A.G.M.)
| | | | | | - Gianfranco Gilardi
- Department of Life Sciences and Systems Biology, University of Torino, via Accademia Albertina 13, 1023 Torino, Italy; (C.Z.); (A.G.M.)
| |
Collapse
|
36
|
Beguelini MR, Santiago CS, Guerra LHA, Santos FCA, Góes RM, Morielle-Versute E, Taboga SR. The hormonal control of the uterus of the bat Myotis nigricans during its different reproductive phases: emphasis on progesterone and estradiol. Cell Tissue Res 2021; 384:211-229. [PMID: 33409655 DOI: 10.1007/s00441-020-03342-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 11/05/2020] [Indexed: 10/22/2022]
Abstract
Myotis nigricans is a species of bat from the Vespertilionidae family that is endemic of the Neotropical region. Its insectivorous feeding habit plus its large range of prey species, great geographical dispersion, wide colonies, and anthropomorphized behavior make this species an important ecological agent that acts in the control of nocturnal insects. Reproductively, M. nigricans presents geographic variations, having different patterns of reproduction according to its geographical location. Despite these extremely interesting characteristics, no more detailed study of the hormonal control of the reproduction of this species has been conducted. Therefore, the aim of the present study was to evaluate the variations in serum hormone concentrations and in uterine hormonal control of this bat during its different reproductive phases. Twenty adult females were collected, divided into four (4) sample groups, according to the reproductive status (nonreproductive, initial, and advanced pregnancy and lactating), and submitted to hormone dosage and immunohistochemical analyses. The results demonstrated that the uterus of M. nigricans is strongly regulated by the interaction/cross-talk between serum concentrations of estradiol (E2) and progesterone with their respective hormone receptors. Significant increases in the concentration of E2 and progesterone are needed to regulate the early pregnancy. The persistence of the corpus luteum throughout pregnancy is necessary, since its placenta does not express aromatase. The expressions of ERα and PR appear to be synchronized in order to coordinate a large portion of the processes that occur inside the uterus of M. nigricans during pregnancy and lactation.
Collapse
Affiliation(s)
- Mateus R Beguelini
- Center of Biological and Health Science, UFOB - Universidade Federal Do Oeste da Bahia, Barreiras, Bahia, Brazil.
| | - Cornélio S Santiago
- Center of Biological and Health Science, UFOB - Universidade Federal Do Oeste da Bahia, Barreiras, Bahia, Brazil
| | - Luiz H A Guerra
- Department of Biology, UNESP - University Estadual Paulista, São José Do Rio Preto, São Paulo, Brazil
| | - Fernanda C A Santos
- Department of Histology and Embryology, UFG - Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Rejane M Góes
- Department of Biology, UNESP - University Estadual Paulista, São José Do Rio Preto, São Paulo, Brazil
| | - Eliana Morielle-Versute
- Department of Zoology and Botany, UNESP - University Estadual Paulista, São José Do Rio Preto, São Paulo, Brazil
| | - Sebastião R Taboga
- Department of Biology, UNESP - University Estadual Paulista, São José Do Rio Preto, São Paulo, Brazil
| |
Collapse
|
37
|
Kiesel L, Eichbaum C, Baumeier A, Eichbaum M. Obesity Epidemic-The Underestimated Risk of Endometrial Cancer. Cancers (Basel) 2020; 12:E3860. [PMID: 33371216 PMCID: PMC7767192 DOI: 10.3390/cancers12123860] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/13/2020] [Accepted: 12/15/2020] [Indexed: 12/24/2022] Open
Abstract
Endometrial cancer (EC) is the most frequently observed malignant gynecologic disease in developed countries. There is a strong association between the established risk factor obesity and the incidence of EC. Furthermore, the rate of women with a body mass index (BMI) > 30 kg/m2 is increasing worldwide, correspondingly leading to a higher prevalence of EC. Understanding the adipose tissue as an endocrine organ, elementary pathophysiological pathways of tumorigenesis have been revealed. This includes the fundamental role of hyperglycemia, insulin resistance, and hyperestrogenemia, as well as interactions with a chronic proinflammatory microenvironment. Therapeutic options potentially include metformin or bariatric surgery. Moreover, changes in individual lifestyle such as weight reduction, physical activity, and an awareness of healthy nutrition are effective in preventing the disease.
Collapse
Affiliation(s)
- Ludwig Kiesel
- Department of Gynecology and Obstetrics, University of Münster Medical School, Albert-Schweitzer-Campus 1, 48149 Münster, Germany;
| | - Christine Eichbaum
- Department of Gynecology and Obstetrics, University of Frankfurt Medical School, Theodor-Stern-Kai 7, 60596 Frankfurt, Germany;
| | - Ariane Baumeier
- Department of Gynecology and Obstetrics, University of Münster Medical School, Albert-Schweitzer-Campus 1, 48149 Münster, Germany;
| | - Michael Eichbaum
- Department of Gynecology and Obstetrics, Helios Dr. Horst-Schmidt-Kliniken Wiesbaden, Ludwig-Erhard-Str. 100, 65199 Wiesbaden, Germany
| |
Collapse
|
38
|
Jin K, Zuo Q, Song J, Zhang Y, Chen G, Li B. CYP19A1 (aromatase) dominates female gonadal differentiation in chicken (Gallus gallus) embryos sexual differentiation. Biosci Rep 2020; 40:BSR20201576. [PMID: 32990306 PMCID: PMC7560524 DOI: 10.1042/bsr20201576] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 06/30/2020] [Accepted: 09/28/2020] [Indexed: 01/30/2023] Open
Abstract
Cytochrome P450 Family 19 SubFamily A member 1 (CYP19A1) gene encodes an aromatase which regulates the sexual differentiation in vertebrates by initiating and maintaining 17β-Estradiol (E2) synthesis. Here, we described the spatiotemporal expression pattern of CYP19A1 and its functional role in the embryonic gonad development in amphoteric chickens (Gallus gallus). Results showed that CYP19A1 exhibited a sexually dimorphic expression pattern in female gonads early at embryonic day 5.5 (HH 28) and robustly expressed within the cytoplasm in ovarian medullas. Most importantly, we induced the gonadal sex reversal by ectopically delivering the aromatase inhibitor (AI) or estradiol (E2) into chicken embryos. To further explore the role of CYP19A1 in chicken embryonic sexual differentiation, we successfully developed an effective method to deliver lentiviral particles with CYP19A1 manipulation into chicken embryos via embryonic intravascular injection. The analysis of interference and overexpression of CYP19A1 provided solid evidences that CYP19A1 is both necessary and sufficient to initiate sex differentiation toward female in chicken embryos. Collectively, this work demonstrates that CYP19A1 is a crucial sex differentiation gene in the embryonic development, which provides a foundation for understanding the mechanism of sex determination and differentiation in chickens.
Collapse
Affiliation(s)
- Kai Jin
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu 225009, China
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safty of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Qisheng Zuo
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu 225009, China
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safty of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Jiuzhou Song
- Animal and Avian Sciences, University of Maryland, College Park, MD 20741, U.S.A
| | - Yani Zhang
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu 225009, China
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safty of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Guohong Chen
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu 225009, China
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safty of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Bichun Li
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu 225009, China
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safty of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
39
|
Brooks DC, Coon V JS, Ercan CM, Xu X, Dong H, Levine JE, Bulun SE, Zhao H. Brain Aromatase and the Regulation of Sexual Activity in Male Mice. Endocrinology 2020; 161:5895007. [PMID: 32910181 PMCID: PMC7485274 DOI: 10.1210/endocr/bqaa137] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 08/17/2020] [Indexed: 12/22/2022]
Abstract
The biologically active estrogen estradiol has important roles in adult brain physiology and sexual behavior. A single gene, Cyp19a1, encodes aromatase, the enzyme that catalyzes the conversion of testosterone to estradiol in the testis and brain of male mice. Estradiol formation was shown to regulate sexual activity in various species, but the relative contributions to sexual behavior of estrogen that arises in the brain versus from the gonads remained unclear. To determine the role of brain aromatase in regulating male sexual activity, we generated a brain-specific aromatase knockout (bArKO) mouse. A newly generated whole-body total aromatase knockout mouse of the same genetic background served as a positive control. Here we demonstrate that local aromatase expression and estrogen production in the brain is partially required for male sexual behavior and sex hormone homeostasis. Male bArKO mice exhibited decreased sexual activity in the presence of strikingly elevated circulating testosterone. In castrated adult bArKO mice, administration of testosterone only partially restored sexual behavior; full sexual behavior, however, was achieved only when both estradiol and testosterone were administered together. Thus, aromatase in the brain is, in part, necessary for testosterone-dependent male sexual activity. We also found that brain aromatase is required for negative feedback regulation of circulating testosterone of testicular origin. Our findings suggest testosterone activates male sexual behavior in part via conversion to estradiol in the brain. These studies provide foundational evidence that sexual behavior may be modified through inhibition or enhancement of brain aromatase enzyme activity and/or utilization of selective estrogen receptor modulators.
Collapse
Affiliation(s)
- David C Brooks
- Division of Reproductive Science in Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - John S Coon V
- Division of Reproductive Science in Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Cihangir M Ercan
- Division of Reproductive Science in Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Xia Xu
- Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, Maryland
| | - Hongxin Dong
- Department of Psychiatry & Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Jon E Levine
- Wisconsin National Primate Research Center, Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | - Serdar E Bulun
- Division of Reproductive Science in Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Hong Zhao
- Division of Reproductive Science in Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Correspondence: Hong Zhao, M.D., Ph.D., Division of Reproductive Science in Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine at Northwestern University, 303 E. Superior Street, Suite 10–111, Chicago, Illinois 60611–2914. E-mail:
| |
Collapse
|
40
|
Abstract
Molecular mechanisms by which sex steroids interact with P. aeruginosa to modulate its virulence have yet to be reported. Our work provides the first characterization of a steroid-induced membrane stress mechanism promoting P. aeruginosa virulence, which includes the release of proinflammatory outer membrane vesicles, resulting in inflammation, host tissue damage, and reduced bacterial clearance. We further demonstrate that at nanomolar (physiological) concentrations, male and female sex steroids promote virulence in clinical strains of P. aeruginosa based on their dynamic membrane fluidic properties. This work provides, for the first-time, mechanistic insight to better understand and predict the P. aeruginosa related response to sex steroids and explain the interindividual patient variability observed in respiratory diseases such as cystic fibrosis that are complicated by gender differences and chronic P. aeruginosa infection. Estrogen, a major female sex steroid hormone, has been shown to promote the selection of mucoid Pseudomonas aeruginosa in the airways of patients with chronic respiratory diseases, including cystic fibrosis. This results in long-term persistence, poorer clinical outcomes, and limited therapeutic options. In this study, we demonstrate that at physiological concentrations, sex steroids, including testosterone and estriol, induce membrane stress responses in P. aeruginosa. This is characterized by increased virulence and consequent inflammation and release of proinflammatory outer membrane vesicles promoting in vivo persistence of the bacteria. The steroid-induced P. aeruginosa response correlates with the molecular polarity of the hormones and membrane fluidic properties of the bacteria. This novel mechanism of interaction between sex steroids and P. aeruginosa explicates the reported increased disease severity observed in females with cystic fibrosis and provides evidence for the therapeutic potential of the modulation of sex steroids to achieve better clinical outcomes in patients with hormone-responsive strains.
Collapse
|
41
|
Romano S, Kaufman OH, Marlow FL. Loss of dmrt1 restores zebrafish female fates in the absence of cyp19a1a but not rbpms2a/b. Development 2020; 147:dev.190942. [PMID: 32895289 DOI: 10.1242/dev.190942] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 08/19/2020] [Indexed: 12/18/2022]
Abstract
Sex determination and differentiation is a complex process regulated by multiple factors, including factors from the germline or surrounding somatic tissue. In zebrafish, sex-determination involves establishment of a bipotential ovary that undergoes sex-specific differentiation and maintenance to form the functional adult gonad. However, the relationships among these factors are not fully understood. Here, we identify potential Rbpms2 targets and apply genetic epistasis experiments to decipher the genetic hierarchy of regulators of sex-specific differentiation. We provide genetic evidence that the crucial female factor rbpms2 is epistatic to the male factor dmrt1 in terms of adult sex. Moreover, the role of Rbpms2 in promoting female fates extends beyond repression of Dmrt1, as Rbpms2 is essential for female differentiation even in the absence of Dmrt1. In contrast, female fates can be restored in mutants lacking both cyp19a1a and dmrt1, and prolonged in bmp15 mutants in the absence of dmrt1. Taken together, this work indicates that cyp19a1a-mediated suppression of dmrt1 establishes a bipotential ovary and initiates female fate acquisition. Then, after female fate specification, Cyp19a1a regulates subsequent oocyte maturation and sustains female fates independently of Dmrt1 repression.
Collapse
Affiliation(s)
- Shannon Romano
- Department of Cell, Developmental and Regenerative Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1020, New York, NY 10029-6574, USA
| | - Odelya H Kaufman
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Florence L Marlow
- Department of Cell, Developmental and Regenerative Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1020, New York, NY 10029-6574, USA .,Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
42
|
Küpeli Akkol E, Genç Y, Karpuz B, Sobarzo-Sánchez E, Capasso R. Coumarins and Coumarin-Related Compounds in Pharmacotherapy of Cancer. Cancers (Basel) 2020; 12:cancers12071959. [PMID: 32707666 PMCID: PMC7409047 DOI: 10.3390/cancers12071959] [Citation(s) in RCA: 173] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 07/14/2020] [Accepted: 07/17/2020] [Indexed: 12/19/2022] Open
Abstract
Cancer is one of the most common causes of disease-related deaths worldwide. Despite the discovery of many chemotherapeutic drugs that inhibit uncontrolled cell division processes for the treatment of various cancers, serious side effects of these drugs are a crucial disadvantage. In addition, multi-drug resistance is another important problem in anticancer treatment. Due to problems such as cytotoxicity and drug resistance, many investigations are being conducted to discover and develop effective anticancer drugs. In recent years, researchers have focused on the anticancer activity coumarins, due to their high biological activity and low toxicity. Coumarins are commonly used in the treatment of prostate cancer, renal cell carcinoma and leukemia, and they also have the ability to counteract the side effects caused by radiotherapy. Both natural and synthetic coumarin derivatives draw attention due to their photochemotherapy and therapeutic applications in cancer. In this review, a compilation of various research reports on coumarins with anticancer activity and investigation and a review of structure-activity relationship studies on coumarin core are presented. Determination of important structural features around the coumarin core may help researchers to design and develop new analogues with a strong anticancer effect and reduce the potential side effects of existing therapeutics.
Collapse
Affiliation(s)
- Esra Küpeli Akkol
- Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, Etiler 06330, Ankara, Turkey;
- Correspondence: (E.K.A.); (R.C.); Tel.: +90-312-2023185 (E.K.A); +39-081-678664 (R.C.)
| | - Yasin Genç
- Department of Pharmacognosy, Faculty of Pharmacy, Hacettepe University, Sıhhiye 06100, Ankara, Turkey;
| | - Büşra Karpuz
- Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, Etiler 06330, Ankara, Turkey;
| | - Eduardo Sobarzo-Sánchez
- Instituto de Investigación e Innovación en Salud, Facultad de Ciencias de la Salud, Universidad Central de Chile, 8330507 Santiago, Chile;
- Department of Organic Chemistry, Faculty of Pharmacy, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Raffaele Capasso
- Department of Agricultural Sciences, University of Naples Federico II, 80055 Portici (Naples), Italy
- Correspondence: (E.K.A.); (R.C.); Tel.: +90-312-2023185 (E.K.A); +39-081-678664 (R.C.)
| |
Collapse
|
43
|
The Role of Dysfunctional Adipose Tissue in Pancreatic Cancer: A Molecular Perspective. Cancers (Basel) 2020; 12:cancers12071849. [PMID: 32659999 PMCID: PMC7408631 DOI: 10.3390/cancers12071849] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 07/04/2020] [Accepted: 07/06/2020] [Indexed: 12/24/2022] Open
Abstract
Pancreatic cancer (PC) is a lethal malignancy with rising incidence and limited therapeutic options. Obesity is a well-established risk factor for PC development. Moreover, it negatively affects outcome in PC patients. Excessive fat accumulation in obese, over- and normal-weight individuals induces metabolic and inflammatory changes of adipose tissue microenvironment leading to a dysfunctional adipose “organ”. This may drive the association between abnormal fat accumulation and pancreatic cancer. In this review, we describe several molecular mechanisms that underpin this association at both local and systemic levels. We focus on the role of adipose tissue-derived circulating factors including adipokines, hormones and pro-inflammatory cytokines, as well as on the impact of the local adipose tissue in promoting PC. A discussion on potential therapeutic interventions, interfering with pro-tumorigenic effects of dysfunctional adipose tissue in PC, is included. Considering the raise of global obesity, research efforts to uncover the molecular basis of the relationship between pancreatic cancer and adipose tissue dysfunction may provide novel insights for the prevention of this deadly disease. In addition, these efforts may uncover novel targets for personalized interventional strategies aimed at improving the currently unsatisfactory PC therapeutic options.
Collapse
|
44
|
Bai DP, Chen Y, Hu YQ, He WF, Shi YZ, Fan QM, Luo RT, Li A. Transcriptome analysis of genes related to gonad differentiation and development in Muscovy ducks. BMC Genomics 2020; 21:438. [PMID: 32590948 PMCID: PMC7318502 DOI: 10.1186/s12864-020-06852-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 06/19/2020] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Sex-related genes play a crucial role in gonadal differentiation into testes or ovaries. However, the genetic control of gonadal differentiation in Muscovy ducks remains unknown. Therefore, the objective of our study was to screen new candidate genes associated with ovarian and testicular development. RESULTS In this study, 24 males before gonadal differentiation (MB), 24 females before gonadal differentiation (FB), 24 males after gonadal differentiation (MA) and 24 females after gonadal differentiation (FA) were selected from Putian Muscovy ducks, forming 4 groups. RNA-Seq revealed 101.76 Gb of clean reads and 2800 differentially expressed genes (DEGs), including 46 in MB vs FB, 609 in MA vs FA, 1027 in FA vs FB, and 1118 in MA vs MB. A total of 146 signalling pathways were enriched by KEGG analysis, among which 20, 108, 108 and 116 signalling pathways were obtained in MB vs FB, MA vs MB, MA vs FA and FA vs FB, respectively. In further GO and KEGG analyses, a total of 21 candidate genes related to gonad differentiation and development in Muscovy ducks were screened. Among these, 9 genes were involved in the differentiation and development of the testes, and 12 genes were involved in the differentiation and development of the ovaries. In addition, RNA-Seq data revealed 2744 novel genes. CONCLUSIONS RNA-Seq data revealed 21 genes related to gonadal differentiation and development in Muscovy ducks. We further identified 12 genes, namely, WNT5B, HTRA3, RSPO3, BMP3, HNRNPK, NIPBL, CREB3L4, DKK3, UBE2R2, UBPL3KCMF1, ANXA2, and OSR1, involved in the differentiation and development of ovaries. Moreover, 9 genes, namely, TTN, ATP5A1, DMRT1, DMRT3, AMH, MAP3K1, PIK3R1, AGT and ADAMTSL1, were related to the differentiation and development of testes. Moreover, after gonadal differentiation, DMRT3, AMH, PIK3R1, ADAMTSL1, AGT and TTN were specifically highly expressed in males. WNT5B, ANXA2 and OSR1 were specifically highly expressed in females. These results provide valuable information for studies on the sex control of Muscovy ducks and reveal novel candidate genes for the differentiation and development of testes and ovaries.
Collapse
Affiliation(s)
- Ding-Ping Bai
- College of Animal Sciences, Fujian Agricultural and Forestry University, Fuzhou, 350002 China
| | - Yue Chen
- College of Animal Sciences, Fujian Agricultural and Forestry University, Fuzhou, 350002 China
| | - Yu-Qiong Hu
- College of Animal Sciences, Fujian Agricultural and Forestry University, Fuzhou, 350002 China
| | - Wen-Feng He
- College of Animal Sciences, Fujian Agricultural and Forestry University, Fuzhou, 350002 China
| | - Yu-Zhu Shi
- College of Animal Sciences, Fujian Agricultural and Forestry University, Fuzhou, 350002 China
| | - Qin-Ming Fan
- College of Animal Sciences, Fujian Agricultural and Forestry University, Fuzhou, 350002 China
| | - Ru-Tang Luo
- College of Animal Sciences, Fujian Agricultural and Forestry University, Fuzhou, 350002 China
| | - Ang Li
- College of Animal Sciences, Fujian Agricultural and Forestry University, Fuzhou, 350002 China
| |
Collapse
|
45
|
Caciolla J, Spinello A, Martini S, Bisi A, Zaffaroni N, Gobbi S, Magistrato A. Targeting Orthosteric and Allosteric Pockets of Aromatase via Dual-Mode Novel Azole Inhibitors. ACS Med Chem Lett 2020; 11:732-739. [PMID: 32435378 DOI: 10.1021/acsmedchemlett.9b00591] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 03/23/2020] [Indexed: 12/19/2022] Open
Abstract
Breast cancer (BC) is the most diffused cancer type in women and the second leading cause of death among the female population. Effective strategies to fight estrogen responsive (ER+) BC, which represents 70% of all BC cases, rely on estrogen deprivation, via the inhibition of the aromatase enzyme, or the modulation of its cognate estrogen receptor. Current clinical therapies significantly increased patient survival time. Nevertheless, the onset of resistance in metastatic BC patients undergoing prolonged treatments is becoming a current clinical challenge, urgently demanding to devise innovative strategies. In this context, here we designed, synthesized, and performed in vitro inhibitory tests on the aromatase enzyme and distinct ER+/ER- BC cell line types of novel azole bridged xanthones. These compounds are active in the low μM range and behave as dual-mode inhibitors, targeting both the orthosteric and the allosteric sites of the enzyme placed along one access channel. Classical and quantum-classical molecular dynamics simulations of the new compounds, as compared with selected steroidal and nonsteroidal inhibitors, provide a rationale to the observed inhibitory potency and supply the guidelines to boost the activity of inhibitors able to exploit coordination to iron and occupation of the access channel to modulate estrogen production.
Collapse
Affiliation(s)
- Jessica Caciolla
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, via Belmeloro 6, 40126 Bologna, Italy
| | - Angelo Spinello
- CNR-IOM Democritos c/o International School for Advanced Studies (SISSA), Via Bonomea 265, 34136 Trieste, Italy
| | - Silvia Martini
- Fondazione IRCSS Istituto Nazionale dei Tumori, via Amadeo 42, 20113 Milano, Italy
| | - Alessandra Bisi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, via Belmeloro 6, 40126 Bologna, Italy
| | - Nadia Zaffaroni
- Fondazione IRCSS Istituto Nazionale dei Tumori, via Amadeo 42, 20113 Milano, Italy
| | - Silvia Gobbi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, via Belmeloro 6, 40126 Bologna, Italy
| | - Alessandra Magistrato
- CNR-IOM Democritos c/o International School for Advanced Studies (SISSA), Via Bonomea 265, 34136 Trieste, Italy
| |
Collapse
|
46
|
Praveen VP, Ladjouze A, Sauter KS, Pulickal A, Katharopoulos E, Trippel M, Perren A, Pandey AV, Flück CE. Novel CYP19A1 Mutations Extend the Genotype-Phenotype Correlation and Reveal the Impact on Ovarian Function. J Endocr Soc 2020; 4:bvaa030. [PMID: 32318648 PMCID: PMC7159065 DOI: 10.1210/jendso/bvaa030] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 03/04/2020] [Indexed: 02/06/2023] Open
Abstract
Context The steroidogenic enzyme aromatase (CYP19A1) is required for estrogen biosynthesis from androgen precursors in the ovary and extragonadal tissues. The role of aromatase, and thus estrogens, is best illustrated by genetic variations of the CYP19A1 gene leading to aromatase deficiency or excess. Objective The objective of this work is to characterize novel CYP19A1 variants. Design setting and patients Variants causing aromatase deficiency were suspected in four 46,XX children of African and Indian origin by careful clinical phenotyping. Sequencing of the CYP19A1 gene identified novel variants. Minigene experiments, aromatase activity assay, and computational, and histological analysis were used to characterize the variants. Main outcome measure and results CYP19A1 variants were found in all patients: a deletion in intron 9 leading to p.P423_H503del, a delins variant at p.P154, and point variants p.V161D, p.R264C, p.R375C. Except for R264C, all variants showed a loss of function. Protein structure and dynamics studies were in line with functional assays. The 2 female patients with delins variants manifested with ambiguous genitalia at birth. Histologic investigation revealed normal ovarian tissue on one side and a streak gonad on the other. Two female patients presented with abnormal pubertal development and polycystic ovaries. Conclusion In girls, aromatase deficiency usually manifests at birth, but diagnosis may also be made because of abnormal pubertal development or ovarian torsion due to (poly)cystic ovaries. The ovary harboring CYP19A1 variants may present as streak gonad or appears normal at birth, but is then at very high risk to produce cysts with aging and is therefore prone to ovarian torsion.
Collapse
Affiliation(s)
| | | | - Kay-Sara Sauter
- Department of Pediatrics, Division of Pediatric Endocrinology, Diabetology and Metabolism University Children's Hospital Bern, Switzerland, and Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Annie Pulickal
- Department of Endocrinology, Amrita Institute of Medical Sciences, Kochi, Kerala, India
| | - Efstathios Katharopoulos
- Department of Pediatrics, Division of Pediatric Endocrinology, Diabetology and Metabolism University Children's Hospital Bern, Switzerland, and Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Mafalda Trippel
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Aurel Perren
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Amit V Pandey
- Department of Pediatrics, Division of Pediatric Endocrinology, Diabetology and Metabolism University Children's Hospital Bern, Switzerland, and Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Christa E Flück
- Department of Pediatrics, Division of Pediatric Endocrinology, Diabetology and Metabolism University Children's Hospital Bern, Switzerland, and Department of Biomedical Research, University of Bern, Bern, Switzerland
| |
Collapse
|
47
|
Maldonado-Domínguez M, Srnec M. Understanding and Predicting Post H-Atom Abstraction Selectivity through Reactive Mode Composition Factor Analysis. J Am Chem Soc 2020; 142:3947-3958. [PMID: 32000494 DOI: 10.1021/jacs.9b12800] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The selective functionalization of C-H bonds is one of the Grails of synthetic chemistry. In this work, we demonstrate that the selectivity toward fast hydroxylation or radical diffusion (known as the OH-rebound and dissociation mechanisms) following H-atom abstraction (HAA) from a substrate C-H bond by high-valent iron-oxo oxidants is already encoded in the HAA step when the post-HAA barriers are much lower than the preceding one. By applying the reactive mode composition factor (RMCF) analysis, which quantifies the kinetic energy distribution (KED) at the reactive mode (RM) of transition states, we show that reactions following the OH-rebound coordinate concentrate the RM kinetic energy on the motion of the reacting oxygen atom and the nascent substrate radical, whereas reactions following the dissociation channel localize most of their kinetic energy in H-atom motion. These motion signatures serve to predict the post-HAA selectivity, and since KED is affected by the free energy of reaction and asynchronicity (factor η) of HAA, we show that bimolecular HAA reactions in solution that are electron transfer-driven and highly exergonic have the lowest fraction of KED on the transferred H-atom and the highest chance to follow rebound hydroxylation. Finally, the RMCF analysis predicts that the H/D primary kinetic isotope effect can serve as a probe for these mechanisms, as confirmed in virtually all reported examples in the literature.
Collapse
Affiliation(s)
- Mauricio Maldonado-Domínguez
- J. Heyrovský Institute of Physical Chemistry, The Czech Academy of Sciences , Dolejškova 3 , Prague 8 18223 , Czech Republic
| | - Martin Srnec
- J. Heyrovský Institute of Physical Chemistry, The Czech Academy of Sciences , Dolejškova 3 , Prague 8 18223 , Czech Republic
| |
Collapse
|
48
|
Wu K, Song W, Zhang Z, Ge W. Disruption of dmrt1 rescues the all-male phenotype of the cyp19a1a mutant in zebrafish - a novel insight into the roles of aromatase/estrogens in gonadal differentiation and early folliculogenesis. Development 2020; 147:dev.182758. [PMID: 32001440 DOI: 10.1242/dev.182758] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 01/20/2020] [Indexed: 12/27/2022]
Abstract
Sex determination and differentiation are complex processes controlled by many different factors; however, the relationships among these factors are poorly understood. Zebrafish gonadal differentiation exhibits high plasticity involving multiple factors and pathways, which provides an excellent model for investigating the interactions between them. Ovarian aromatase (cyp19a1a) and dmrt1 are key factors in directing vertebrate ovary and testis differentiation, respectively. Knockout of zebrafish cyp19a1a leads to all-male offspring, whereas the loss of dmrt1 results in a female-biased sex ratio. In the present study, we established dmrt1-/- ;cyp19a1a-/- double mutant zebrafish and discovered that the introduction of the dmrt1 mutation into the cyp19a1a mutant could rescue the all-male phenotype of the latter. Interestingly, despite the lack of aromatase/estrogens, the follicles in the ovary of the rescued cyp19a1a mutant could develop normally up to the previtellogenic stage. Further evidence suggested the ovarian aromatase directed ovarian differentiation by suppressing dmrt1 expression via nuclear estrogen receptors (nERs). Our results provide solid evidence for an interaction between cyp19a1a and dmrt1 in zebrafish gonadal differentiation, and for the dispensability of estrogens in controlling early folliculogenesis.
Collapse
Affiliation(s)
- Kun Wu
- Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau 999078, China
| | - Weiyi Song
- Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau 999078, China
| | - Zhiwei Zhang
- Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau 999078, China
| | - Wei Ge
- Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau 999078, China
| |
Collapse
|
49
|
Identification of PARP-1, Histone H1 and SIRT-1 as New Regulators of Breast Cancer-Related Aromatase Promoter I.3/II. Cells 2020; 9:cells9020427. [PMID: 32059481 PMCID: PMC7072628 DOI: 10.3390/cells9020427] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 02/06/2020] [Accepted: 02/10/2020] [Indexed: 12/19/2022] Open
Abstract
Paracrine interactions between malignant estrogen receptor positive (ER+) breast cancer cells and breast adipose fibroblasts (BAFs) stimulate estrogen biosynthesis by aromatase in BAFs. In breast cancer, mainly the cAMP-responsive promoter I.3/II-region mediates excessive aromatase expression. A rare single nucleotide variant (SNV) in this promoter region, which caused 70% reduction in promoter activity, was utilized for the identification of novel regulators of aromatase expression. To this end, normal and mutant promoter activities were measured in luciferase reporter gene assays. DNA-binding proteins were captured by DNA-affinity and identified by mass spectrometry. The DNA binding of proteins was analyzed using electrophoretic mobility shift assays, immunoprecipitation-based in vitro binding assays and by chromatin immunoprecipitation in BAFs in vivo. Protein expression and parylation were analyzed by western blotting. Aromatase activities and RNA-expression were measured in BAFs. Functional consequences of poly (ADP-ribose) polymerase-1 (PARP-1) knock-out, rescue or overexpression, respectively, were analyzed in murine embryonic fibroblasts (MEFs) and the 3T3-L1 cell model. In summary, PARP-1 and histone H1 (H1) were identified as critical regulators of aromatase expression. PARP-1-binding to the SNV-region was crucial for aromatase promoter activation. PARP-1 parylated H1 and competed with H1 for DNA-binding, thereby inhibiting its gene silencing action. In MEFs (PARP-1 knock-out and wild-type) and BAFs, PARP-1-mediated induction of the aromatase promoter showed bi-phasic dose responses in overexpression and inhibitor experiments, respectively. The HDAC-inhibitors butyrate, panobinostat and selisistat enhanced promoter I.3/II-mediated gene expression dependent on PARP-1-activity. Forskolin stimulation of BAFs increased promoter I.3/II-occupancy by PARP-1, whereas SIRT-1 competed with PARP-1 for DNA binding but independently activated the promoter I.3/II. Consistently, the inhibition of both PARP-1 and SIRT-1 increased the NAD+/NADH-ratio in BAFs. This suggests that cellular NAD+/NADH ratios control the complex interactions of PARP-1, H1 and SIRT-1 and regulate the interplay of parylation and acetylation/de-acetylation events with low NAD+/NADH ratios (reverse Warburg effect), promoting PARP-1 activation and estrogen synthesis in BAFs. Therefore, PARP-1 inhibitors could be useful in the treatment of estrogen-dependent breast cancers.
Collapse
|
50
|
Barrientos G, Llanos P, Basualto-Alarcón C, Estrada M. Androgen-Regulated Cardiac Metabolism in Aging Men. Front Endocrinol (Lausanne) 2020; 11:316. [PMID: 32499759 PMCID: PMC7243157 DOI: 10.3389/fendo.2020.00316] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 04/24/2020] [Indexed: 12/21/2022] Open
Abstract
The prevalence of cardiovascular mortality is higher in men than in age-matched premenopausal women. Gender differences are linked to circulating sex-related steroid hormone levels and their cardio-specific actions, which are critical factors involved in the prevalence and features of age-associated cardiovascular disease. In women, estrogens have been described as cardioprotective agents, while in men, testosterone is the main sex steroid hormone. The effects of testosterone as a metabolic regulator and cardioprotective agent in aging men are poorly understood. With advancing age, testosterone levels gradually decrease in men, an effect associated with increasing fat mass, decrease in lean body mass, dyslipidemia, insulin resistance and adjustment in energy substrate metabolism. Aging is associated with a decline in metabolism, characterized by modifications in cardiac function, excitation-contraction coupling, and lower efficacy to generate energy. Testosterone deficiency -as found in elderly men- rapidly becomes an epidemic condition, associated with prominent cardiometabolic disorders. Therefore, it is highly probable that senior men showing low testosterone levels will display symptoms of androgen deficiency, presenting an unfavorable metabolic profile and increased cardiovascular risk. Moreover, recent reports establish that testosterone replacement improves cardiomyocyte bioenergetics, increases glucose metabolism and reduces insulin resistance in elderly men. Thus, testosterone-related metabolic signaling and gene expression may constitute relevant therapeutic target for preventing, or treating, age- and gender-related cardiometabolic diseases in men. Here, we will discuss the impact of current evidence showing how cardiac metabolism is regulated by androgen levels in aging men.
Collapse
Affiliation(s)
- Genaro Barrientos
- Programa de Fisiología y Biofísica, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
- Centro de Estudios en Ejercicio, Metabolismo y Cáncer (CEMC), Universidad de Chile, Santiago, Chile
| | - Paola Llanos
- Centro de Estudios en Ejercicio, Metabolismo y Cáncer (CEMC), Universidad de Chile, Santiago, Chile
- Facultad de Odontología, Instituto de Investigación en Ciencias Odontológicas (ICOD), Universidad de Chile, Santiago, Chile
| | - Carla Basualto-Alarcón
- Departamento de Ciencias de la Salud, Universidad de Aysén, Coyhaique, Chile
- Departamento de Anatomía y Medicina Legal, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Manuel Estrada
- Programa de Fisiología y Biofísica, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
- *Correspondence: Manuel Estrada
| |
Collapse
|