1
|
Amason ME, Li L, Harvest CK, Lacey CA, Miao EA. Validation of the Intermolecular Disulfide Bond in Caspase-2. BIOLOGY 2024; 13:49. [PMID: 38248479 PMCID: PMC10813798 DOI: 10.3390/biology13010049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/05/2024] [Accepted: 01/15/2024] [Indexed: 01/23/2024]
Abstract
Caspases are a family of proteins involved in cell death. Although several caspase members have been well characterized, caspase-2 remains enigmatic. Caspase-2 has been implicated in several phenotypes, but there has been no consensus in the field about its upstream activating signals or its downstream protein targets. In addition, the unique ability of caspase-2 to form a disulfide-bonded dimer has not been studied in depth. Herein, we investigate the disulfide bond in the context of inducible dimerization, showing that disulfide bond formation is dimerization dependent. We also explore and review several stimuli published in the caspase-2 field, test ferroptosis-inducing stimuli, and study in vivo infection models. We hypothesize that the disulfide bond will ultimately prove to be essential for the evolved function of caspase-2. Proving this will require the discovery of cell death phenotypes where caspase-2 is definitively essential.
Collapse
Affiliation(s)
- Megan E. Amason
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Lupeng Li
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Carissa K. Harvest
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Carolyn A. Lacey
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Edward A. Miao
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
2
|
Zeber-Lubecka N, Kulecka M, Suchta K, Dąbrowska M, Ciebiera M, Hennig EE. Association of Mitochondrial Variants with the Joint Occurrence of Polycystic Ovary Syndrome and Hashimoto's Thyroiditis. Antioxidants (Basel) 2023; 12:1983. [PMID: 38001836 PMCID: PMC10669137 DOI: 10.3390/antiox12111983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/01/2023] [Accepted: 11/03/2023] [Indexed: 11/26/2023] Open
Abstract
BACKGROUND The prevalence of Hashimoto's thyroiditis (HT) among women with polycystic ovary syndrome (PCOS) is higher than in the general female population, but the factors predisposing to the coexistence of these disorders remain unclear. This study employed whole genome sequencing of mitochondrial DNA to identify genetic variants potentially associated with the development of PCOS and HT and predisposing to their joint occurrence. RESULTS A total of 84 women participated, including patients with PCOS, HT, coexisting PCOS and HT (PCOS + HT) and healthy women. Both Fisher's exact and Mann-Whitney U statistical analyses were performed to compare the frequency of variants between groups. Ten differentiating variants were common to both analyses in PCOS + HT vs. PCOS, one in PCOS + HT vs. HT, and six in PCOS + HT vs. control. Several variants differentiating the PCOS + HT group from PCOS and controls were identified, located both in the mitochondrial genes (including the MT-CYB, MT-ND1, MT-ND2, MT-ND4, MT-ND6, MT-CO1, MT-CO3) and the D-loop region. Only two variants differentiated PCOS + HT and HT groups. One variant (13237a in MT-ND5) was common for all three comparisons and underrepresented in the PCOS + HT group. Functional enrichment analysis showed 10 pathways that were unique for the comparison of PCOS + HT and PCOS groups, especially related to ATP production and oxidative phosphorylation, and one pathway, the NADH-quinone oxidoreductase, chain M/4, that was unique for the comparison of PCOS + HT and control groups. Notably, nine pathways shared commonality between PCOS + HT vs. PCOS and PCOS + HT vs. control, related to the biogenesis and assembly of Complex I. CONCLUSION This study provides novel insights into the genetic variants associated with oxidative stress in women with coexisting PCOS and HT. Mitochondrial dysfunction and oxidative stress appear to play a role in the pathogenesis of both conditions. However, more mitochondrial variants were found to differentiate women with both PCOS and HT from those with PCOS alone than from those with HT alone.
Collapse
Affiliation(s)
- Natalia Zeber-Lubecka
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, 02-781 Warsaw, Poland; (N.Z.-L.); (M.K.)
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland;
| | - Maria Kulecka
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, 02-781 Warsaw, Poland; (N.Z.-L.); (M.K.)
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland;
| | - Katarzyna Suchta
- Department of Gynaecological Endocrinology, Medical University of Warsaw, 00-315 Warsaw, Poland;
| | - Michalina Dąbrowska
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland;
| | - Michał Ciebiera
- Second Department of Obstetrics and Gynecology, Centre of Postgraduate Medical Education, 00-189 Warsaw, Poland;
- Warsaw Institute of Women’s Health, 00-189 Warsaw, Poland
| | - Ewa E. Hennig
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, 02-781 Warsaw, Poland; (N.Z.-L.); (M.K.)
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland;
| |
Collapse
|
3
|
Zeber-Lubecka N, Ciebiera M, Hennig EE. Polycystic Ovary Syndrome and Oxidative Stress-From Bench to Bedside. Int J Mol Sci 2023; 24:14126. [PMID: 37762427 PMCID: PMC10531631 DOI: 10.3390/ijms241814126] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/06/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Oxidative stress (OS) is a condition that occurs as a result of an imbalance between the production of reactive oxygen species (ROS) and the body's ability to detoxify and neutralize them. It can play a role in a variety of reproductive system conditions, including polycystic ovary syndrome (PCOS), endometriosis, preeclampsia, and infertility. In this review, we briefly discuss the links between oxidative stress and PCOS. Mitochondrial mutations may lead to impaired oxidative phosphorylation (OXPHOS), decreased adenosine triphosphate (ATP) production, and an increased production of ROS. These functional consequences may contribute to the metabolic and hormonal dysregulation observed in PCOS. Studies have shown that OS negatively affects ovarian follicles and disrupts normal follicular development and maturation. Excessive ROS may damage oocytes and granulosa cells within the follicles, impairing their quality and compromising fertility. Impaired OXPHOS and mitochondrial dysfunction may contribute to insulin resistance (IR) by disrupting insulin signaling pathways and impairing glucose metabolism. Due to dysfunctional OXPHOS, reduced ATP production, may hinder insulin-stimulated glucose uptake, leading to IR. Hyperandrogenism promotes inflammation and IR, both of which can increase the production of ROS and lead to OS. A detrimental feedback loop ensues as IR escalates, causing elevated insulin levels that exacerbate OS. Exploring the relations between OS and PCOS is crucial to fully understand the role of OS in the pathophysiology of PCOS and to develop effective treatment strategies to improve the quality of life of women affected by this condition. The role of antioxidants as potential therapies is also discussed.
Collapse
Affiliation(s)
- Natalia Zeber-Lubecka
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland;
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Michał Ciebiera
- Second Department of Obstetrics and Gynecology, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland;
- Warsaw Institute of Women’s Health, 00-189 Warsaw, Poland
| | - Ewa E. Hennig
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland;
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| |
Collapse
|
4
|
Yang J, Wang E, Jiang M, Tan Y, Yao F, Sun C, Pan L, Gao L, Yao J. Integrated fecal microbiota and metabolomics analysis of the orlistat intervention effect on polycystic ovary syndrome rats induced by letrozole combined with a high-fat diet. J Ovarian Res 2023; 16:109. [PMID: 37277785 DOI: 10.1186/s13048-023-01193-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 05/24/2023] [Indexed: 06/07/2023] Open
Abstract
BACKGROUND This study aimed to compare the characteristics of the gut microbiota and their metabolite profiles between polycystic ovary syndrome (PCOS) and orlistat-treated PCOS rats (ORL-PCOS), which could help to better understand the underlying mechanism of the effect of orlistat on PCOS. METHODS PCOS rat models were established using letrozole combined with a high-fat diet. Ten rats were randomly selected as a PCOS control group (PCOS). The other three groups (n = 10/group) were additionally supplemented with different doses of orlistat (low, medium, high). Then, fecal samples of the PCOS and ORL-PCOS groups were analysed by 16S rRNA gene sequencing and untargeted metabolomics. Blood samples were collected to detect serum sex hormones and lipids. RESULTS The results showed that orlistat attenuated the body weight gain, decreased the levels of T, LH, the LH/FSH ratio, TC, TG and LDL-C; increased the level of E2; and improved estrous cycle disorder in PCOS rats. The bacterial richness and diversity of the gut microbiota in the ORL-PCOS group were higher than those in the PCOS group. The ratio of Firmicutes to Bacteroidetes was decreased with orlistat treatment. Moreover, orlistat treatment led to a significant decrease in the relative abundance of Ruminococcaceae and Lactobacillaceae, and increases in the abundances of Muribaculaceae and Bacteroidaceae. Metabolic analysis identified 216 differential fecal metabolites in total and 6 enriched KEGG pathways between the two groups, including steroid hormone biosynthesis, neuroactive ligand-receptor interaction and vitamin digestion and absorption. Steroid hormone biosynthesis was the pathway with the most significant enrichment. The correlations between the gut microbiota and differential metabolites were calculated, which may provide a basis for understanding the composition and function of microbial communities. CONCLUSIONS Our data suggested that orlistat exerts a PCOS treatment effect, which may be mediated by modifying the structure and composition of the gut microbiota, as well as the metabolite profiles of PCOS rats.
Collapse
Affiliation(s)
- Jianmei Yang
- Department of Pediatric Endocrinology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276006, Shandong, China
- Department of Pediatric Endocrinology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Enli Wang
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276006, Shandong, China
| | - Mingmin Jiang
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276006, Shandong, China
| | - Yujun Tan
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276006, Shandong, China
| | - Fangfang Yao
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276006, Shandong, China
| | - Chenghong Sun
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276006, Shandong, China
| | - Lihong Pan
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276006, Shandong, China
| | - Ling Gao
- Scientific Center, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Jingchun Yao
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276006, Shandong, China.
| |
Collapse
|
5
|
Wang Y, Yang J, Wang Y, Chen Y, Wang Y, Kuang H, Feng X. Upregulation of TXNIP contributes to granulosa cell dysfunction in polycystic ovary syndrome via activation of the NLRP3 inflammasome. Mol Cell Endocrinol 2023; 561:111824. [PMID: 36450326 DOI: 10.1016/j.mce.2022.111824] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/22/2022] [Accepted: 11/25/2022] [Indexed: 11/29/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a complex endocrine disease. Thioredoxin-interacting protein (TXNIP) promotes oxidative stress and triggers inflammation. Herein, we investigated the role and potential mechanism of TXNIP in PCOS. In a mouse model of dehydroepiandrosterone (DHEA)-induced PCOS, we found that TXNIP was upregulated in the ovaries, especially in granulosa cells (GCs). TXNIP was also upregulated in testosterone (T)-treated GCs in vitro. Knockdown of TXNIP by lentivirus-constructed shRNA attenuated T-induced GC injury and oxidative stress, as well as inflammation and the NLRP3 inflammasome. The mechanism by which TXNIP promotes inflammation may involve TXNIP dissociation from the TXNIP-TRX complex and binding to NLRP3 to form the inflammasome. Additionally, we verified that knockdown of TXNIP ameliorated ovarian injury and inflammation in mice with DHEA-induced PCOS in vivo. Collectively, we demonstrated that TXNIP is involved in GC inflammation by promoting NLRP3 inflammasome activation in PCOS.
Collapse
Affiliation(s)
- Ying Wang
- The Second Department of Gynecology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jiyu Yang
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yu Wang
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yao Chen
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yiran Wang
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Hongying Kuang
- The Second Department of Gynecology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China.
| | - Xiaoling Feng
- The Second Department of Gynecology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China.
| |
Collapse
|
6
|
Li Y, Adur MK, Lonergan SM, Keating AF, Ross JW. MicroRNA21 inhibition affects porcine oocyte maturation and alters protein expression critical for metabolic pathway function. Mol Reprod Dev 2022; 89:443-458. [PMID: 36001642 PMCID: PMC9804257 DOI: 10.1002/mrd.23641] [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: 02/03/2022] [Revised: 06/08/2022] [Accepted: 08/12/2022] [Indexed: 01/05/2023]
Abstract
MicroRNA21 (MIR21) abundance in porcine oocytes and cumulus cells increases during in vitro maturation. The mechanism by which MIR21 regulates oocyte maturation and the effect on the developmental competence of subsequent embryos remains unclear. The objective of this study was to assess the function of MIR21 during porcine oocyte maturation and its effect on embryonic development. Treatment with peptide nucleic acid MIR21 inhibitor (MIR21-PNA), designed to specifically bind to and prevent MIR21 activity during in vitro oocyte maturation, decreased cumulus cell expansion, and the oocyte ability to achieve metaphase II maturation stage when compared to control groups. Following parthenogenetic activation, the cleavage rate at 48 h in the MIR21-PNA group was decreased (p ≤ 0.03) relative to the control groups. Additionally, liquid chromatography-mass spectrometry (LC-MS/MS) of oocyte and cumulus cell total protein following MIR21-PNA treatment during in vitro maturation identified changes in signaling pathways with primary involvement of glucose metabolism (GM) pathways. Furthermore, there was no difference (p = 0.21) in oocyte maturation of control and MIR21-PNA treated oocytes when cultured in pyruvate lacking medium. Finally, MIR21-PNA treatment decreased (p = 0.04) glutathione and increased (p = 0.07) reactive oxygen species production in the oocyte. These data suggest that MIR21 influences porcine oocyte maturation by regulating GM pathways in the cumulus-oocyte complex.
Collapse
Affiliation(s)
- Yunsheng Li
- Department of Animal ScienceIowa State UniversityAmesIowaUSA,College of Animal Science and TechnologyAnhui Agricultural UniversityHefeiChina
| | | | | | | | - Jason W. Ross
- Department of Animal ScienceIowa State UniversityAmesIowaUSA
| |
Collapse
|
7
|
Zhou J, Yue S, Du J, Xue B, Wang L, Peng Q, Zou H, Hu R, Jiang Y, Wang Z, Xue B. Integration of transcriptomic and metabolomic analysis of the mechanism of dietary N-carbamoylglutamate in promoting follicle development in yaks. Front Vet Sci 2022; 9:946893. [PMID: 36105003 PMCID: PMC9464987 DOI: 10.3389/fvets.2022.946893] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 08/01/2022] [Indexed: 12/04/2022] Open
Abstract
Yak is the main livestock in the highlands of China. The low reproductive rate of yaks is a serious constraint on their production and utility. N-carbamylglutamate (NCG) can increase arginine synthesis in mammals and has been shown to improve reproductive performance. Twelve multiparous and simutaneous anoestrous female yaks were randomly divided into two groups, one of which was fed the basal diet (Control, n = 6), and the other was fed the basal diet supplemented with NCG at 6 g/day/yak (NCG, n = 6). All yaks were slaughtered on the 32nd day (the time predicted for the selection of the last wave of dominant follicles), and their ovarian tissues were collected and follicles were classified. NCG supplementation increased the number of large ovarian follicles (diameter > 10 mm), as well as caused significant changes in the transcriptional and metabolic levels in yak ovaries which due to the differential expression of 889 genes and 94 metabolites. Integrated analysis of the transcriptomics and metabolomics data revealed that the differentially expressed genes and differential metabolites were primarily involved in the process of energy metabolism, amino acid metabolic pathways, carbohydrate metabolic pathways, and lipid metabolic pathways. The highlighted changes were associated with amino acid synthesis and metabolism, ovarian steroid hormone synthesis, the pentose phosphate pathway, and the tricarboxylic acid cycle, suggesting that NCG supplementation may promote estrogen synthesis and help regulate follicular development by altering the pathways associated with glucose catabolism. The results present important clues for understanding the mechanisms by which NCG supplementation promotes follicular development in yaks. The findings of this study provide a basis for the development and application of NCG in optimizing animal reproduction, including yak reproductive performance, which may help optimize livestock management and uplift the pastoral economy.
Collapse
Affiliation(s)
- Jia Zhou
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Shuangming Yue
- Department of Bioengineering, Sichuan Water Conservancy College, Chengdu, China
| | - Jingjing Du
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Benchu Xue
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Lizhi Wang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Quanhui Peng
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Huawei Zou
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Rui Hu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Yahui Jiang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Zhisheng Wang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Bai Xue
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
- *Correspondence: Bai Xue
| |
Collapse
|
8
|
Guler SA, Machahua C, Geiser TK, Kocher G, Marti TM, Tan B, Trappetti V, Ryerson CJ, Funke-Chambour M. Dehydroepiandrosterone in fibrotic interstitial lung disease: a translational study. Respir Res 2022; 23:149. [PMID: 35676709 PMCID: PMC9178848 DOI: 10.1186/s12931-022-02076-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 05/31/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Dehydroepiandrosterone (DHEA) is a precursor sex hormone with antifibrotic properties. The aims of this study were to investigate antifibrotic mechanisms of DHEA, and to determine the relationship between DHEA-sulfate (DHEAS) plasma levels, disease severity and survival in patients with fibrotic interstitial lung diseases (ILDs). METHODS Human precision cut lung slices (PCLS) and normal human lung fibroblasts were treated with DHEA and/or transforming growth factor (TGF)-β1 before analysis of pro-fibrotic genes and signal proteins. Cell proliferation, cytotoxicity, cell cycle and glucose-6-phosphate dehydrogenase (G6PD) activity were assessed. DHEAS plasma levels were correlated with pulmonary function, the composite physiologic index (CPI), and time to death or lung transplantation in a derivation cohort of 31 men with idiopathic pulmonary fibrosis (IPF) and in an independent validation cohort of 238 men and women with fibrotic ILDs. RESULTS DHEA decreased the expression of pro-fibrotic markers in-vitro and ex-vivo. There was no cytotoxic effect for the applied concentrations, but DHEA interfered in proliferation by modulating the cell cycle through reduction of G6PD activity. In men with IPF (derivation cohort) DHEAS plasma levels in the lowest quartile were associated with poor lung function and higher CPI (adjusted OR 1.15 [95% CI 1.03-1.38], p = 0.04), which was confirmed in the fibrotic ILD validation cohort (adjusted OR 1.03 [95% CI 1.00-1.06], p = 0.01). In both cohorts the risk of early mortality was higher in patients with low DHEAS levels, after accounting for potential confounding by age in men with IPF (HR 3.84, 95% CI 1.25-11.7, p = 0.02), and for age, sex, IPF diagnosis and prednisone treatment in men and women with fibrotic ILDs (HR 3.17, 95% CI 1.35-7.44, p = 0.008). CONCLUSIONS DHEA reduces lung fibrosis and cell proliferation by inducing cell cycle arrest and inhibition of G6PD activity. The association between low DHEAS levels and disease severity suggests a potential prognostic and therapeutic role of DHEAS in fibrotic ILD.
Collapse
Affiliation(s)
- Sabina A Guler
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 18, 3010, Bern, Switzerland. .,Department for BioMedical Research DBMR, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| | - Carlos Machahua
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 18, 3010, Bern, Switzerland.,Department for BioMedical Research DBMR, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Thomas K Geiser
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 18, 3010, Bern, Switzerland.,Department for BioMedical Research DBMR, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Gregor Kocher
- Department for BioMedical Research DBMR, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Division of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Thomas M Marti
- Department for BioMedical Research DBMR, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Division of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Benjamin Tan
- Department of Medicine, University of British Columbia, Vancouver, Canada
| | | | - Christopher J Ryerson
- Department of Medicine, University of British Columbia, Vancouver, Canada.,Centre for Heart Lung Innovation, University of British Columbia, Vancouver, Canada
| | - Manuela Funke-Chambour
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 18, 3010, Bern, Switzerland.,Department for BioMedical Research DBMR, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
9
|
Chappell NR, Gibbons WE, Blesson CS. Pathology of hyperandrogenemia in the oocyte of polycystic ovary syndrome. Steroids 2022; 180:108989. [PMID: 35189133 PMCID: PMC8920773 DOI: 10.1016/j.steroids.2022.108989] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 02/07/2022] [Accepted: 02/16/2022] [Indexed: 12/01/2022]
Abstract
Polycystic ovary syndrome (PCOS) is the most common ovulatory disorder in the world and is associated with multiple adverse outcomes. The phenotype is widely varied, with several pathologies contributing to the spectrum of the disease including insulin resistance, obesity and hyperandrogenemia. Of these, the role of hyperandrogenemia and the mechanism by which it causes dysfunction remains poorly understood. Early studies have shown that androgens may affect the metabolic pathways of a cell, and this may pose hazards at the level of the mitochondria. As mitochondria are strictly maternally inherited, this would provide an exciting explanation not only to the pathophysiology of PCOS as a disease, but also to the inheritance pattern. This review seeks to summarize what is known about PCOS and associated adverse outcomes with focus on the role of hyperandrogenemia and specific emphasis on the oocyte.
Collapse
Affiliation(s)
- Neil R Chappell
- Reproductive Endocrinology and Infertility Division, Department of Obstetrics and Gynecology, Baylor College of Medicine. One Baylor Plaza, Houston 77030, TX, USA; Family Fertility Center, Texas Children's Hospital, Houston 77030, TX, USA
| | - William E Gibbons
- Reproductive Endocrinology and Infertility Division, Department of Obstetrics and Gynecology, Baylor College of Medicine. One Baylor Plaza, Houston 77030, TX, USA; Family Fertility Center, Texas Children's Hospital, Houston 77030, TX, USA
| | - Chellakkan S Blesson
- Reproductive Endocrinology and Infertility Division, Department of Obstetrics and Gynecology, Baylor College of Medicine. One Baylor Plaza, Houston 77030, TX, USA; Family Fertility Center, Texas Children's Hospital, Houston 77030, TX, USA.
| |
Collapse
|
10
|
Ullah A, Wang MJ, Yang JP, Adu-Gyamfi EA, Czika A, Sah SK, Feng Q, Wang YX. Ovarian inflammatory mRNA profiles of a dehydroepiandrosterone plus high-fat diet-induced polycystic ovary syndrome mouse model. Reprod Biomed Online 2021; 44:791-802. [DOI: 10.1016/j.rbmo.2021.10.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 10/15/2021] [Accepted: 10/25/2021] [Indexed: 10/19/2022]
|
11
|
Cichon I, Ortmann W, Kolaczkowska E. Metabolic Pathways Involved in Formation of Spontaneous and Lipopolysaccharide-Induced Neutrophil Extracellular Traps (NETs) Differ in Obesity and Systemic Inflammation. Int J Mol Sci 2021; 22:ijms22147718. [PMID: 34299338 PMCID: PMC8303382 DOI: 10.3390/ijms22147718] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 02/07/2023] Open
Abstract
Obesity manifests itself with low-grade chronic inflammation that shapes immune responses during infection. Albeit obese individuals are at risk of higher mortality due to comorbidities, they are better protected from systemic inflammation. Recently, we showed that in the vasculature of obese mice kept on high-fat diet (HFD), neutrophils produce less neutrophil extracellular traps (NETs) than in lean controls (normal diet, ND). NETs are used by neutrophils to counteract severe infection, but they also cause collateral damage. Hardly anything is known about metabolic requirements for their formation, especially in the context of obesity and/or sepsis. Thus, we aimed to study the immunometabolism of NET formation by application of ex vivo neutrophil analyses (Seahorse analyzer, selective inhibitors, confocal imaging) and intravital microscopy. The obtained data show that glycolysis and/or pentose phosphate pathway are involved in NETs release by ND neutrophils in both physiological and inflammatory conditions. In contrast, such cells of septic HFD mice utilize these routes only to spontaneously cast NETs, while after secondary ex vivo activation they exhibit so called "exhausted phenotype", which manifests itself in diminished NET release despite high glycolytic potential and flexibility to oxidize fatty acids. Moreover, impact of ATP synthase inhibition on NET formation is revealed. Overall, the study shows that the neutrophil potential to cast NETs depends on both the metabolic and inflammatory state of the individual.
Collapse
|
12
|
Lipinska P, Sell-Kubiak E, Pawlak P, Madeja ZE, Warzych E. Response of Bovine Cumulus-Oocytes Complexes to Energy Pathway Inhibition during In Vitro Maturation. Genes (Basel) 2021; 12:genes12060838. [PMID: 34072406 PMCID: PMC8228821 DOI: 10.3390/genes12060838] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/13/2021] [Accepted: 05/25/2021] [Indexed: 12/25/2022] Open
Abstract
Glucose or fatty acids (FAs) metabolisms may alter the ovarian follicle environment and thus determine oocyte and the nascent embryo quality. The aim of the experiment was to investigate the effect of selective inhibition of glucose (iodoacetate + DHEA) or FA (etomoxir) metabolism on in vitro maturation (IVM) of bovine COCs (cumulus-oocyte complexes) to investigate oocyte's development, quality, and energy metabolism. After in vitro fertilization, embryos were cultured to the blastocyst stage. Lipid droplets, metabolome, and lipidome were analyzed in oocytes and cumulus cells. mRNA expression of the selected genes was measured in the cumulus cells. ATP and glutathione relative levels were measured in oocytes. Changes in FA content in the maturation medium were evaluated by mass spectrometry. Our results indicate that only glucose metabolism is substantial to the oocyte during IVM since only glucose inhibition decreased embryo culture efficiency. The most noteworthy differences in the reaction to the applied inhibition systems were observed in cumulus cells. The upregulation of ketone body metabolism in the cumulus cells of the glucose inhibition group suggest possibly failed attempts of cells to switch into lipid consumption. On the contrary, etomoxir treatment of the oocytes did not affect embryo development, probably due to undisturbed metabolism in cumulus cells. Therefore, we suggest that the energy pathways analyzed in this experiment are not interchangeable alternatives in bovine COCs.
Collapse
|
13
|
Gündoğan Gİ, Kıg C, Karacan M, Doğruman H. Investigation of Physiological Effects Induced by Dehydroepiandrosterone in Human Endothelial Cells and Ovarian Cancer Cell Line. Turk J Pharm Sci 2021; 18:185-191. [PMID: 33902257 DOI: 10.4274/tjps.galenos.2020.58827] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Objectives Dehydroepiandrosterone (DHEA) is an endogenous hormone that acts as a ligand for several cellular receptors. An age-dependent decline in circulating levels of DHEA is linked to changes in various physiological functions. In gynecological clinical practice, DHEA is commonly prescribed to induce ovulation. Some clinical studies report a positive association between high serum concentrations of DHEA and an increased risk of developing ovarian cancer. However, the in vitro physiological effects of DHEA on ovarian cancerous cells have not been explored thus far. In this study, we aimed to investigate the physiological effects of DHEA treatment (0-200 μM, 24-72 hours) on MDAH-2774 human ovarian cancer cell line and primary HuVeC human endothelial cells. Materials and Methods The physiological effects of DHEA treatment (0-200 μM, 24-72 hours) on MDAH-2774 human ovarian cancer cell line and primary HuVeC human endothelial cells were investigated with the (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) test, acridine orange/ethidium bromide staining, and scratch assay. Results DHEA treatment promoted proliferation of the MDAH-2774 cancer cell line in a dose-dependent manner (r=0.6906, p<0.0001, for 24 hours) (r=0.6802, p<0.0001, for 48 hours) (r=0.7969, p<0.0001, for 72 hours). In contrast, DHEA inhibited proliferation of the primary HuVeC cells (r=0.9490, p<0.0001, for 24 hours) (r=0.9533, p<0.0001, for 48 hours) (r=0.9584, p<0.0001, for 72 hours). In agreement with these observations, DHEA treatment resulted in a dose-dependent increase in the number of necrotic cells in the primary HuVeC cells (r=0.97, p<0.0001). However, the number of necrotic or apoptotic cells did not change significantly when the MDAH-2774 cells was exposed to DHEA. Moreover, we found that DHEA treatment reduced the migration rate of HuVeC cells in a dose-dependent manner (r=0.9868, p<0.0001), whereas only a slight increase was observed in the MDAH-2774 ovarian cancer cell line (r=0.8938, p<0.05). Conclusion Our findings suggest that DHEA promotes the proliferation of ovarian cancer cells in a dose-dependent manner in vitro. Moreover, DHEA induced necrosis and inhibited proliferation in endothelial cells. Although mechanistic evidence is required, our preliminary findings imply that exposure to high doses of DHEA may be associated with an increased risk of developing ovarian cancer.
Collapse
Affiliation(s)
- Gül İpek Gündoğan
- Istanbul Yeni Yuzyil University Faculty of Medicine, Department of Histology and Embryology, Istanbul, Turkey
| | - Cenk Kıg
- Istanbul Yeni Yuzyil University Faculty of Medicine, Department of Medical Biology and Genetics, Istanbul, Turkey
| | - Meriç Karacan
- Istanbul Yeni Yuzyil University Faculty of Medicine, Department of Gynecology and Obstetrics, Istanbul, Turkey
| | - Hüsniye Doğruman
- Istanbul Yeni Yuzyil University Faculty of Medicine, Department of Histology and Embryology, Istanbul, Turkey
| |
Collapse
|
14
|
Moiz B, Garcia J, Basehore S, Sun A, Li A, Padmanabhan S, Albus K, Jang C, Sriram G, Clyne AM. 13C Metabolic Flux Analysis Indicates Endothelial Cells Attenuate Metabolic Perturbations by Modulating TCA Activity. Metabolites 2021; 11:metabo11040226. [PMID: 33917224 PMCID: PMC8068087 DOI: 10.3390/metabo11040226] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/30/2021] [Accepted: 04/01/2021] [Indexed: 11/16/2022] Open
Abstract
Disrupted endothelial metabolism is linked to endothelial dysfunction and cardiovascular disease. Targeted metabolic inhibitors are potential therapeutics; however, their systemic impact on endothelial metabolism remains unknown. In this study, we combined stable isotope labeling with 13C metabolic flux analysis (13C MFA) to determine how targeted inhibition of the polyol (fidarestat), pentose phosphate (DHEA), and hexosamine biosynthetic (azaserine) pathways alters endothelial metabolism. Glucose, glutamine, and a four-carbon input to the malate shuttle were important carbon sources in the baseline human umbilical vein endothelial cell (HUVEC) 13C MFA model. We observed two to three times higher glutamine uptake in fidarestat and azaserine-treated cells. Fidarestat and DHEA-treated HUVEC showed decreased 13C enrichment of glycolytic and TCA metabolites and amino acids. Azaserine-treated HUVEC primarily showed 13C enrichment differences in UDP-GlcNAc. 13C MFA estimated decreased pentose phosphate pathway flux and increased TCA activity with reversed malate shuttle direction in fidarestat and DHEA-treated HUVEC. In contrast, 13C MFA estimated increases in both pentose phosphate pathway and TCA activity in azaserine-treated cells. These data show the potential importance of endothelial malate shuttle activity and suggest that inhibiting glycolytic side branch pathways can change the metabolic network, highlighting the need to study systemic metabolic therapeutic effects.
Collapse
Affiliation(s)
- Bilal Moiz
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (B.M.); (A.S.); (A.L.); (S.P.); (K.A.)
| | - Jonathan Garcia
- School of Bioengineering, Science, and Heath Systems, Drexel University, Philadelphia, PA 19104, USA; (J.G.); (S.B.)
| | - Sarah Basehore
- School of Bioengineering, Science, and Heath Systems, Drexel University, Philadelphia, PA 19104, USA; (J.G.); (S.B.)
| | - Angela Sun
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (B.M.); (A.S.); (A.L.); (S.P.); (K.A.)
| | - Andrew Li
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (B.M.); (A.S.); (A.L.); (S.P.); (K.A.)
| | - Surya Padmanabhan
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (B.M.); (A.S.); (A.L.); (S.P.); (K.A.)
| | - Kaitlyn Albus
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (B.M.); (A.S.); (A.L.); (S.P.); (K.A.)
| | - Cholsoon Jang
- Department of Biological Chemistry, Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA 92697, USA;
| | - Ganesh Sriram
- Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, MD 20742, USA;
| | - Alisa Morss Clyne
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (B.M.); (A.S.); (A.L.); (S.P.); (K.A.)
- Correspondence: ; Tel.: +1-301-405-9806
| |
Collapse
|
15
|
Imanaka S, Shigetomi H, Kobayashi H. Reprogramming of glucose metabolism of cumulus cells and oocytes and its therapeutic significance. Reprod Sci 2021; 29:653-667. [PMID: 33675030 DOI: 10.1007/s43032-021-00505-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 02/21/2021] [Indexed: 12/17/2022]
Abstract
The aim of this review is to summarize our current understanding of the molecular mechanism for the glucose metabolism, especially pyruvate dehydrogenase (PDH), during oocyte maturation, as well as future perspectives of therapeutic strategies for aging focusing on metabolic regulation between aerobic glycolysis and the tricarboxylic acid (TCA) cycle/oxidative phosphorylation (OXPHOS). Each keyword alone or in combination was used to search from PubMed. Glucose metabolism is a dynamic process involving "On" and "Off" switches by the pyruvate dehydrogenase kinase (PDK)-PDH axis, which is crucial for energy metabolism and mitochondrial efficiency in cumulus cell differentiation and oocyte maturation. Activation of PDK suppresses the conversion of pyruvate to acetyl-coenzyme A (acetyl-CoA) through the inactivation of PDH, which allows the cumulus cells to supply sufficient amounts of pyruvate, lactate, and nicotinamide adenine dinucleotide phosphate (NADPH) to the oocytes. On the other hand, inactivation of PDK in oocytes can produce adenosine triphosphate (ATP) through a metabolic shift from aerobic glycolysis to the TCA cycle/OXPHOS. The metabolic balance between aerobic glycolysis and TCA cycle/OXPHOS presents us with a number of enzymes, ligands, receptors, and antioxidants that are potential therapeutic targets, some of which have already been successfully pursued to improve fertility outcomes. However, there are also many reports that question their efficacy. In conclusion, understanding the molecular mechanisms involved in the PDK-PDH axis is a crucial step to advance in novel therapeutic strategies to improve oocyte quality.
Collapse
Affiliation(s)
- Shogo Imanaka
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Japan.,Ms.Clinic MayOne, Kashihara, Japan
| | - Hiroshi Shigetomi
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Japan.,Aska Ladies Clinic, Nara, Japan
| | - Hiroshi Kobayashi
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Japan. .,Ms.Clinic MayOne, Kashihara, Japan.
| |
Collapse
|
16
|
Wang F, Xie N, Wu Y, Zhang Q, Zhu Y, Dai M, Zhou J, Pan J, Tang M, Cheng Q, Shi B, Guo Q, Li X, Xie L, Wang B, Yang D, Weng Q, Guo L, Ye J, Pan M, Zhang S, Zhou H, Zhen C, Liu P, Ning K, Brackenridge L, Hardiman PJ, Qu F. Association between circadian rhythm disruption and polycystic ovary syndrome. Fertil Steril 2020; 115:771-781. [PMID: 33358334 DOI: 10.1016/j.fertnstert.2020.08.1425] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 08/27/2020] [Accepted: 08/28/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To explore the association of circadian rhythm disruption with polycystic ovary syndrome (PCOS) and the potential underlying mechanism in ovarian granulosa cells (GCs). DESIGN Multicenter questionnaire-based survey, in vivo and ex vivo studies. SETTING Twelve hospitals in China, animal research center, and research laboratory of a women's hospital. PATIENTS/ANIMALS A total of 436 PCOS case subjects and 715 control subjects were recruited for the survey. In vivo and ex vivo studies were conducted in PCOS-model rats and on ovarian GCs collected from women with PCOS and control subjects. INTERVENTION(S) The PCOS rat model was established with the use of testosterone propionate. MAIN OUTCOME MEASURE(S) Assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-seq), RNA sequencing, rhythmicity analysis, functional enrichment analysis. RESULT(S) There was a significant correlation between night shift work and PCOS. PCOS-model rats presented distinct differences in the circadian variation of corticotropin-releasing hormone, adrenocorticotropic hormone, prolactin, and a 4-h phase delay in thyrotropic hormone levels. The motif enrichment analysis of ATAC-seq revealed the absence of clock-related transcription factors in specific peaks of PCOS group, and RNA sequencing ex vivo at various time points over 24 hours demonstrated the differential rhythmic expression patterns of women with PCOS. Kyoto Encyclopedia of Genes and Genomes analysis further highlighted metabolic dysfunction, including both carbohydrate and amino acid metabolism and the tricarboxylic acid cycle. CONCLUSION(S) There is a significant association of night shift work with PCOS, and genome-wide chronodisruption exists in ovarian GCs.
Collapse
Affiliation(s)
- Fangfang Wang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Institute for Women's Health, University College London, London, United Kingdom
| | - Ningning Xie
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Yan Wu
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Qing Zhang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Yuhang Zhu
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Minchen Dai
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Jue Zhou
- College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China; Institute for Women's Health, University College London, London, United Kingdom
| | - Jiexue Pan
- First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Mengling Tang
- School of Public Health, Zhejiang University, Hangzhou, China
| | - Qi Cheng
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Biwei Shi
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Qinyuan Guo
- Maternal and Child Healthcare Hospital of Liuzhou, Liuzhou, China
| | - Xinling Li
- Maternal and Child Healthcare Hospital of Liuzhou, Liuzhou, China
| | - Lifeng Xie
- Maternal and Child Healthcare Hospital of Liuzhou, Liuzhou, China
| | - Bing Wang
- Second Hospital of Jiaxing, Jiaxing, China
| | - Dongxia Yang
- Second Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qing Weng
- First People's Hospital of Yuhang District of Hangzhou, Hangzhou, China
| | - Lanzhong Guo
- Dongyang Women's and Children's Hospital, Dongyang, China
| | - Jisheng Ye
- Dongyang Women's and Children's Hospital, Dongyang, China
| | - Mingwo Pan
- Guangdong Women and Children Hospital, Guangzhou, China
| | - Shuyi Zhang
- Baiyin City Maternity and Childcare Hospital, Baiyin, China
| | - Hua Zhou
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Cailan Zhen
- People's Hospital of Lucheng, Lucheng, China
| | - Ping Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second Hospital, Chengdu, People's Republic of China
| | - Ke Ning
- Department of Social Science, Institute of Education, University College London, London, United Kingdom
| | - Lisa Brackenridge
- Institute for Women's Health, University College London, London, United Kingdom
| | - Paul J Hardiman
- Institute for Women's Health, University College London, London, United Kingdom
| | - Fan Qu
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Institute for Women's Health, University College London, London, United Kingdom.
| |
Collapse
|
17
|
Chimote BN, Chimote NM. Dehydroepiandrosterone sulphate (DHEAS) concentrations stringently regulate fertilisation, embryo development and IVF outcomes: are we looking at a potentially compelling 'oocyte-related factor' in oocyte activation? J Assist Reprod Genet 2020; 38:193-202. [PMID: 33161515 DOI: 10.1007/s10815-020-02001-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 11/01/2020] [Indexed: 11/30/2022] Open
Affiliation(s)
- Bindu N Chimote
- IVF Embryology Laboratory, Vaunshdhara Fertility Centre, 9, Dr. Munje Marg, Congress Nagar, Nagpur, Maharashtra, India.
| | - Natachandra M Chimote
- Department of Reproductive Endocrinology, Vaunshdhara Fertility Centre, 9, Dr. Munje Marg, Congress Nagar, Nagpur, Maharashtra, India
| |
Collapse
|
18
|
Risk Factors of Overweight and Obesity Related to Diet and Disordered Eating Attitudes in Adolescent Girls with Clinical Features of Polycystic Ovary Syndrome. J Clin Med 2020; 9:jcm9093041. [PMID: 32967289 PMCID: PMC7564079 DOI: 10.3390/jcm9093041] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/10/2020] [Accepted: 09/15/2020] [Indexed: 12/16/2022] Open
Abstract
Background: We aimed to find the difference between girls with clinical features of Polycystic ovary syndrome (PCOS), divided into two groups: Overweight/obesity (Ov/Ob) and normal weight (N), related to diet, disordered eating attitudes (DEA), metabolic and hormonal differences, and to identify the risk factors of being overweight or obese. Methods: Seventy-eight adolescents with PCOS, aged 14-18 years, were divided into Ov/Ob and N groups. Patients underwent blood tests for determination of follicle-stimulating hormone (FSH), luteinizing hormone (LH), total testosterone, DHEA-S, estradiol, of sex hormone-binding globulin (SHBG), fasting glucose, insulin, Homeostatic Model Assessment of Insulin Resistance (HOMA-IR), and lipid profile. Nutrition was evaluated using a 3-day food record. To examine the level of DEA, the Eating Attitudes Test-26 (EAT-26) was used. We defined an EAT-26 score ≥20 as positive for DEA. Logistic regression was carried out to identify the independent predictors of being overweight and obese. Results: An increase of 10 g in plant protein intake decreased the probability of being overweight and of obesity (OR = 0.54; p = 0.036). EAT-26 score ≥20 was correlated with a 7-fold (OR = 6.88; p = 0.02) increased odds of being overweight or of obesity. Conclusion: Being overweight and obesity in adolescents with PCOS may be associated with DEA and the type and amount of protein intake.
Collapse
|
19
|
Urich ML, Henderson WM, MacLeod AH, Yonkos LT, Bringolf RB. Gonad metabolomics and blood biochemical analysis reveal differences associated with testicular oocytes in wild largemouth bass (Micropterus salmoides). Comp Biochem Physiol B Biochem Mol Biol 2020; 250:110491. [PMID: 32827749 DOI: 10.1016/j.cbpb.2020.110491] [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: 05/29/2020] [Revised: 08/05/2020] [Accepted: 08/12/2020] [Indexed: 11/25/2022]
Abstract
Adverse reproductive effects associated with gonadal intersex among freshwater fish could hold considerable implications for population sustainability. Presence of testicular oocytes (TO) is the most common form of intersex and is widespread among centrarchids (sunfishes) of North America and other freshwater teleosts. Placing TO within the toxicological context of adverse outcome pathways (AOPs) to assess ecological risk is a priority for ecotoxicologists due to the association of TO with harmful chemical exposure and adverse reproductive effects in some cases. However, key event relationships between EDC exposure, incidence of TO, and apical outcomes have yet to be fully elucidated - in part due to a lack of knowledge of relationships between intersex gonad physiology and fish health. Understanding the physiological status of intersex fish is critical to assess ecological risk, understand mechanisms of induction, and to establish biomarkers of intersex in fish. In the present study, features of gonad metabolite profiles associated with TO in largemouth bass (LMB, Micropterus salmoides) from an impoundment in Georgia (USA) were determined using GC-MS-based metabolomics. Clinical blood biochemical screens were used to evaluate markers of fish health associated with TO. Results suggest that physiological changes in energy expenditure as well as relatively 'feminized' gonad lipid and protein metabolism may be related to the occurrence of TO in male LMB, and highlight the need to understand relationships between intersex and physical stressors such as elevated temperature and hypoxia. These results provide novel insight to AOPs associated with TO and identify candidate analytes for biomarker discovery.
Collapse
Affiliation(s)
- Matthew L Urich
- University of Georgia, Warnell School of Forestry & Natural Resources, Interdisciplinary Toxicology Program, Athens, GA, USA
| | - W Matthew Henderson
- United State Environmental Protection Agency, Office of Research and Development, Center for Environmental Measurement and Modeling, Athens, GA, United States
| | - Alexander H MacLeod
- University of Maryland, Environmental Sciences Department, College of Agriculture and Natural Resources, College Park, MD, USA
| | - Lance T Yonkos
- University of Maryland, Environmental Sciences Department, College of Agriculture and Natural Resources, College Park, MD, USA
| | - Robert B Bringolf
- University of Georgia, Warnell School of Forestry & Natural Resources, Interdisciplinary Toxicology Program, Athens, GA, USA.
| |
Collapse
|
20
|
Metabolomic biomarkers of polycystic ovary syndrome related-obesity: a review of the literature. REV ROMANA MED LAB 2020. [DOI: 10.2478/rrlm-2020-0017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Abstract
Background and objectives: Polycystic ovary syndrome (PCOS) displays a phenotype-dependent cardio-metabolic risk. By performing a systematic search of the literature, we aimed to summarize metabolomic signatures associated with obesity in PCOS women.
Data sources and study eligibility criteria: We conducted a comprehensive search including: Embase, PubMed, and Web of Science until 31st of May 2019. We used the terms: metabolomics and polycystic ovary syndrome. We excluded the following papers: animal studies, studies that included only lean PCOS women, reviews, meta-analyses, results of interventional studies, those that did not apply metabolomic techniques.
Results: The lipid signature in obese women with PCOS showed increased levels of free fatty acids (carnitine, adipic acid, linoleic acid, oleic acid) and lower levels of lysophosphatidylcholines and glycerolphosphocholine compared with non-obese PCOS women. Regarding carbohydrate metabolism, a decrease in citric and lactic acid levels characterized obese PCOS women. Decreased lactic acid in obese PCOS women suggests augmented insulin stimulated glucose muscle use in lean, but not in obese women. Considering amino acid metabolomic markers, valine, glycine, serine, threonine, isoleucine and lysine were higher in obese PCOS women. Patients with visceral obesity presented a diminished uptake of essential amino acids, BCAA, leucine and serine in the skeletal muscle. α-ketoglutarate was significantly higher in obese women with PCOS in comparison with lean women with PCOS, distinguishing these 2 subgroups of PCOS with high ‘predictive accuracy’.
Limitations: Overall, a small number of studies have focused on the impact of obesity on the metabolic fingerprints of PCOS women. There is need for properly controlled, high-quality studies.
Conclusions: There is compelling evidence of significant alterations in carbohydrate, lipid, and amino acid metabolism in women with PCOS and obesity. Metabolomics may identify new metabolic pathways involved in PCOS and improve our understanding of the complex relation between PCOS and obesity in order to personalize PCOS therapy.
Collapse
|
21
|
Lowe JL, Bathgate R, Grupen CG. Effect of carbohydrates on lipid metabolism during porcine oocyte IVM. Reprod Fertil Dev 2019; 31:557-569. [PMID: 31039975 DOI: 10.1071/rd18043] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 09/12/2018] [Indexed: 01/06/2023] Open
Abstract
Porcine oocytes contain a large amount of endogenous lipid, which is thought to function as an intracellular source of energy. The aim of this study was to determine the effects of stimulating or inhibiting lipid metabolism using l-carnitine or etomoxir respectively on the IVM of porcine oocytes cultured in media of varying carbohydrate composition. In the presence of pyruvate and lactate, exclusion of glucose inhibited oocyte nuclear and cytoplasmic maturation compared with oocytes matured in media containing low (1.5mM) and high (4.0mM) concentrations of glucose. In the absence of pyruvate and lactate in low-glucose medium only, a greater proportion of l-carnitine-treated oocytes progressed to the MII stage compared with untreated oocytes. The inclusion of pyruvate and lactate significantly altered the distribution of cytoplasmic lipid droplets and elevated the ATP content of oocytes, whereas the l-carnitine treatment did not. Further, the inhibitory effect of etomoxir on nuclear maturation was decreased in high- compared with low-glucose medium. The results indicate that carbohydrate substrates are absolutely necessary for effective porcine oocyte maturation, and that l-carnitine supplementation can only partially compensate for deficiencies in carbohydrate provision.
Collapse
Affiliation(s)
- Jenna L Lowe
- Sydney School of Veterinary Science, Faculty of Science, The University of Sydney, 425 Werombi Road, Camden, NSW 2570, Australia
| | - Roslyn Bathgate
- Sydney School of Veterinary Science, Faculty of Science, The University of Sydney, 425 Werombi Road, Camden, NSW 2570, Australia
| | - Christopher G Grupen
- Sydney School of Veterinary Science, Faculty of Science, The University of Sydney, 425 Werombi Road, Camden, NSW 2570, Australia
| |
Collapse
|
22
|
Luderer U, Eskenazi B, Hauser R, Korach KS, McHale CM, Moran F, Rieswijk L, Solomon G, Udagawa O, Zhang L, Zlatnik M, Zeise L, Smith MT. Proposed Key Characteristics of Female Reproductive Toxicants as an Approach for Organizing and Evaluating Mechanistic Data in Hazard Assessment. ENVIRONMENTAL HEALTH PERSPECTIVES 2019; 127:75001. [PMID: 31322437 PMCID: PMC6791466 DOI: 10.1289/ehp4971] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 06/17/2019] [Accepted: 06/24/2019] [Indexed: 05/13/2023]
Abstract
BACKGROUND Identification of female reproductive toxicants is currently based largely on integrated epidemiological and in vivo toxicology data and, to a lesser degree, on mechanistic data. A uniform approach to systematically search, organize, integrate, and evaluate mechanistic evidence of female reproductive toxicity from various data types is lacking. OBJECTIVE We sought to apply a key characteristics approach similar to that pioneered for carcinogen hazard identification to female reproductive toxicant hazard identification. METHODS A working group of international experts was convened to discuss mechanisms associated with chemical-induced female reproductive toxicity and identified 10 key characteristics of chemicals that cause female reproductive toxicity: 1) alters hormone receptor signaling; alters reproductive hormone production, secretion, or metabolism; 2) chemical or metabolite is genotoxic; 3) induces epigenetic alterations; 4) causes mitochondrial dysfunction; 5) induces oxidative stress; 6) alters immune function; 7) alters cell signal transduction; 8) alters direct cell–cell interactions; 9) alters survival, proliferation, cell death, or metabolic pathways; and 10) alters microtubules and associated structures. As proof of principle, cyclophosphamide and diethylstilbestrol (DES), for which both human and animal studies have demonstrated female reproductive toxicity, display at least 5 and 3 key characteristics, respectively. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD), for which the epidemiological evidence is mixed, exhibits 5 key characteristics. DISCUSSION Future efforts should focus on evaluating the proposed key characteristics against additional known and suspected female reproductive toxicants. Chemicals that exhibit one or more of the key characteristics could be prioritized for additional evaluation and testing. A key characteristics approach has the potential to integrate with pathway-based toxicity testing to improve prediction of female reproductive toxicity in chemicals and potentially prevent some toxicants from entering common use. https://doi.org/10.1289/EHP4971.
Collapse
Affiliation(s)
- Ulrike Luderer
- Center for Occupational and Environmental Health, University of California, Irvine, Irvine, California, USA
| | - Brenda Eskenazi
- School of Public Health, University of California, Berkeley, Berkeley, California, USA
| | - Russ Hauser
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Kenneth S. Korach
- National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina, USA
| | - Cliona M. McHale
- School of Public Health, University of California, Berkeley, Berkeley, California, USA
| | - Francisco Moran
- Office of Environmental Health Hazard Assessment, California Environmental Protection Agency, Sacramento, California, USA
| | - Linda Rieswijk
- School of Public Health, University of California, Berkeley, Berkeley, California, USA
- Institute of Data Science, Maastricht University, Maastricht, Netherlands
| | - Gina Solomon
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Osamu Udagawa
- Center for Health and Environmental Risk Research, National Institute of Environmental Studies, Tsukuba-City, Ibaraki, Japan
| | - Luoping Zhang
- School of Public Health, University of California, Berkeley, Berkeley, California, USA
| | - Marya Zlatnik
- Department of Obstetrics and Gynecology, University of California, San Francisco, San Francisco, California, USA
| | - Lauren Zeise
- Office of Environmental Health Hazard Assessment, California Environmental Protection Agency, Sacramento, California, USA
| | - Martyn T. Smith
- School of Public Health, University of California, Berkeley, Berkeley, California, USA
| |
Collapse
|
23
|
Troisi J, Cinque C, Giugliano L, Symes S, Richards S, Adair D, Cavallo P, Sarno L, Scala G, Caiazza M, Guida M. Metabolomic change due to combined treatment with myo-inositol, D-chiro-inositol and glucomannan in polycystic ovarian syndrome patients: a pilot study. J Ovarian Res 2019; 12:25. [PMID: 30904021 PMCID: PMC6431025 DOI: 10.1186/s13048-019-0500-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 03/07/2019] [Indexed: 02/08/2023] Open
Abstract
Background Polycystic ovarian syndrome (PCOS) is a highly variable syndrome and one of the most common female endocrine disorders. Although the association inositols-glucomannan may represent a good therapeutic strategy in the treatment of PCOS women with insulin resistance, the effect of inositols on the metabolomic profile of these women has not been described yet. Results Fifteen PCOS-patients and 15 controls were enrolled. Patients were treated with myo-inositol (1.75 g/day), D-chiro-inositol (0.25 g/day) and glucomannan (4 g/day) for 3 months. Blood concentrations of glucose, insulin, triglycerides and cholesterol, and ovary volumes and antral follicles count, as well as metabolomic profiles, were evaluated for control subjects and for cases before and after treatment. PCOS-patients had higher BMI compared with Controls, BMI decreased significantly after 3 months of treatment although it remained significantly higher compared to controls. 3-methyl-1-hydroxybutyl-thiamine-diphosphate, valine, phenylalanine, ketoisocapric, linoleic, lactic, glyceric, citric and palmitic acid, glucose, glutamine, creatinine, arginine, choline and tocopherol emerged as the most relevant metabolites for distinguishing cases from controls. Conclusion Our pilot study has identified a complex network of serum molecules that appear to be correlated with PCOS, and with a combined treatment with inositols and glucomannan. Trial registration ClinicalTial.gov, NCT03608813. Registered 1st August 2018 - Retrospectively registered, . Electronic supplementary material The online version of this article (10.1186/s13048-019-0500-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jacopo Troisi
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana", University of Salerno, Baronissi, SA, Italy. .,Theoreo srl, Via degli Ulivi 3, 84090, Montecorvino Pugliano, SA, Italy. .,European Biomedical Research Institute of Salerno (EBRIS), Via S. de Renzi, 3, 84125, Salerno, SA, Italy.
| | - Claudia Cinque
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana", University of Salerno, Baronissi, SA, Italy
| | - Luigi Giugliano
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana", University of Salerno, Baronissi, SA, Italy
| | - Steven Symes
- Department of Chemistry and Physics, University of Tennessee at Chattanooga, 615 McCallie Ave., Chattanooga, TN, 37403, USA.,Department of Obstetrics and Gynecology, University of Tennessee College of Medicine, Chattanooga, TN, USA
| | - Sean Richards
- Department of Obstetrics and Gynecology, University of Tennessee College of Medicine, Chattanooga, TN, USA.,Department of Biology, Geology and Environmental Sciences, University of Tennessee at Chattanooga, 615 McCallie Ave., Chattanooga, TN, 37403, USA
| | - David Adair
- Department of Obstetrics and Gynecology, University of Tennessee College of Medicine, Chattanooga, TN, USA
| | - Pierpaolo Cavallo
- Department of Physics, University of Salerno, Fisciano, SA, Italy.,Istituto Sistemi Complessi - Consiglio Nazionale delle Ricerche, Rome, Italy
| | - Laura Sarno
- Department of Neurosciences and Reproductive and Dentistry Sciences, University of Naples Federico II, Naples, Italy
| | - Giovanni Scala
- Theoreo srl, Via degli Ulivi 3, 84090, Montecorvino Pugliano, SA, Italy.,Hosmotic srl, Via Raffale Bosco 78, 80069, Vico Equense, NA, Italy
| | - Maria Caiazza
- Azienda Sanitaria Locale, distretto sanitario 66, via Vernieri, 14, 84124, Salerno, SA, Italy
| | - Maurizio Guida
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana", University of Salerno, Baronissi, SA, Italy.,Theoreo srl, Via degli Ulivi 3, 84090, Montecorvino Pugliano, SA, Italy
| |
Collapse
|
24
|
Buszewska-Forajta M, Rachoń D, Stefaniak A, Wawrzyniak R, Konieczna A, Kowalewska A, Markuszewski MJ. Identification of the metabolic fingerprints in women with polycystic ovary syndrome using the multiplatform metabolomics technique. J Steroid Biochem Mol Biol 2019; 186:176-184. [PMID: 30670174 DOI: 10.1016/j.jsbmb.2018.10.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 10/16/2018] [Accepted: 10/24/2018] [Indexed: 12/19/2022]
Abstract
In addition to chronic anovulation and clinical signs of hyperandrogenism women with polycystic ovary syndrome (PCOS) are insulin resistant and therefore, develop central obesity with its long term consequences such as dyslipidaemia, hypertension, atherosclerosis and type 2 diabetes mellitus (T2DM), which all lead to the development of cardiovascular disease (CVD). Due to the polysymptomatic nature of this syndrome and lack of consensus on its diagnostic criteria there is a strong need of finding a reliable biochemical or molecular marker, which would facilitate making the accurate diagnosis of PCOS. Therefore, the aim of our study was to perform a metabolomics analysis with the use of two complementary techniques: gas chromatography and liquid chromatography coupled with mass spectrometry, of the serum samples from women with PCOS (n = 30) and to compare them with healthy age and BMI matched controls (n = 30). Obtained results were subjected to one-dimensional statistical analysis (student's t-test or its non-parametric equivalent U Mann-Whitney test) and multivariate statistical analysis (the principal component analysis [PCA], variable importance into projection [VIP] and selectivity ratio [SR]). The results of our study showed that women with PCOS are characterised by metabolic disorders of the amino acids, carbohydrates, steroid hormones, lipids and purines. Compared to control subjects, women with PCOS had increased serum levels of phospholipids, aromatic amino acids, organic acids, hormones and sphinganine and decreased total cholesterol. Among the identified compounds, total cholesterol, phenylalanine and dehydroepiandrosterone sulfate, uric and lactic acid were the compounds with the strongest discriminating power.
Collapse
Affiliation(s)
- Magdalena Buszewska-Forajta
- Department of Biopharmaceutics and Pharmacodynamics, Medical University of Gdańsk, Hallera 107, 80-416 Gdańsk, Poland
| | - Dominik Rachoń
- Department of Clinical and Experimental Endocrinology, Medical University of Gdańsk, Dębinki 7, 80-210 Gdańsk, Poland
| | - Anna Stefaniak
- Department of Biopharmaceutics and Pharmacodynamics, Medical University of Gdańsk, Hallera 107, 80-416 Gdańsk, Poland
| | - Renata Wawrzyniak
- Department of Biopharmaceutics and Pharmacodynamics, Medical University of Gdańsk, Hallera 107, 80-416 Gdańsk, Poland
| | - Aleksandra Konieczna
- Department of Clinical and Experimental Endocrinology, Medical University of Gdańsk, Dębinki 7, 80-210 Gdańsk, Poland
| | | | - Michał Jan Markuszewski
- Department of Biopharmaceutics and Pharmacodynamics, Medical University of Gdańsk, Hallera 107, 80-416 Gdańsk, Poland.
| |
Collapse
|
25
|
Tamadon A, Hu W, Cui P, Ma T, Tong X, Zhang F, Li X, Shao LR, Feng Y. How to choose the suitable animal model of polycystic ovary syndrome? TRADITIONAL MEDICINE AND MODERN MEDICINE 2018. [DOI: 10.1142/s2575900018300047] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is a gynecological metabolic and endocrine disorder with uncertain etiology. To understand the etiology of PCOS or the evaluation of various therapeutic agents, different animal models have been introduced. Considering this fact that is difficult to develop an animal model that mimics all aspects of this syndrome, but, similarity of biological, anatomical, and/or biochemical features of animal model to the human PCOS phenotypes can increase its application. This review paper evaluates the recently researched animal models and introduced the best models for different research purposes in PCOS studies. During January 2013 to January 2017, 162 studies were identified which applied various kinds of animal models of PCOS including rodent, primate, ruminant and fish. Between these models, prenatal and pre-pubertal androgen rat models and then prenatal androgen mouse model have been studied in detail than others. The comparison of main features of these models with women PCOS demonstrates higher similarity of these three models to human conditions. Thereafter, letrozole models can be recommended for the investigation of various aspects of PCOS. Interestingly, similarity of PCOS features of post-pubertal insulin and human chorionic gonadotropin rat models with women PCOS were considerable which can make it as a good choice for future investigations.
Collapse
Affiliation(s)
- Amin Tamadon
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P. R. China
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Fudan University, Shanghai 200032, P. R. China
- Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai 200032, P. R. China
| | - Wei Hu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P. R. China
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Fudan University, Shanghai 200032, P. R. China
- Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai 200032, P. R. China
| | - Peng Cui
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P. R. China
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Fudan University, Shanghai 200032, P. R. China
- Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai 200032, P. R. China
| | - Tong Ma
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P. R. China
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Fudan University, Shanghai 200032, P. R. China
- Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai 200032, P. R. China
| | - Xiaoyu Tong
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P. R. China
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Fudan University, Shanghai 200032, P. R. China
- Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai 200032, P. R. China
| | - Feifei Zhang
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, P. R. China
| | - Xin Li
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, P. R. China
| | - Linus R. Shao
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 40530, Sweden
| | - Yi Feng
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P. R. China
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Fudan University, Shanghai 200032, P. R. China
- Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai 200032, P. R. China
| |
Collapse
|
26
|
Mahmoud YI, Mahmoud AA, Abo-Zeid FS, Fares NH. Effects of dehydroepiandrosterone on the ovarian reserve and pregnancy outcomes in perimenopausal rats (DHEA and fertility in perimenopausal rats). Life Sci 2018. [DOI: 10.1016/j.lfs.2018.03.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
27
|
Kiersztan A, Trojan N, Tempes A, Nalepa P, Sitek J, Winiarska K, Usarek M. DHEA supplementation to dexamethasone-treated rabbits alleviates oxidative stress in kidney-cortex and attenuates albuminuria. J Steroid Biochem Mol Biol 2017; 174:17-26. [PMID: 28782595 DOI: 10.1016/j.jsbmb.2017.07.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 07/01/2017] [Accepted: 07/18/2017] [Indexed: 01/13/2023]
Abstract
Our recent study has shown that dehydroepiandrosterone (DHEA) administered to rabbits partially ameliorated several dexamethasone (dexP) effects on hepatic and renal gluconeogenesis, insulin resistance and plasma lipid disorders. In the current investigation, we present the data on DHEA protective action against dexP-induced oxidative stress and albuminuria in rabbits. Four groups of adult male rabbits were used in the in vivo experiment: (1) control, (2) dexP-treated, (3) DHEA-treated and (4) both dexP- and DHEA-treated. Administration of dexP resulted in accelerated generation of renal hydroxyl free radicals (HFR) and malondialdehyde (MDA), accompanied by diminished superoxide dismutase (SOD) and catalase activities and a dramatic rise in urinary albumin/creatinine ratio. Treatment with DHEA markedly reduced dexP-induced oxidative stress in kidney-cortex due to a decline in NADPH oxidase activity and enhancement of catalase activity. Moreover, DHEA effectively attenuated dexP-evoked albuminuria. Surprisingly, dexP-treated rabbits exhibited elevation of GSH/GSSG ratio, accompanied by a decrease in glutathione peroxidase (GPx) and glutathione-S-transferase (GST) activities as well as an increase in glucose-6-phosphate dehydrogenase (G6PDH) activity. Treatment with DHEA resulted in a decline in GSH/GSSG ratio and glutathione reductase (GR) activity, accompanied by an elevation of GPx activity. Interestingly, rabbits treated with both dexP and DHEA remained the control values of GSH/GSSG ratio. As the co-administration of DHEA with dexP resulted in (i) reduction of oxidative stress in kidney-cortex, (ii) attenuation of albuminuria and (iii) normalization of glutathione redox state, DHEA might limit several undesirable renal side effects during chronic GC treatment of patients suffering from allergies, asthma, rheumatoid arthritis and lupus. Moreover, its supplementation might be particularly beneficial for the therapy of patients with glucocorticoid-induced diabetes.
Collapse
Affiliation(s)
- Anna Kiersztan
- Department of Metabolic Regulation, Institute of Biochemistry, Faculty of Biology, University of Warsaw, I. Miecznikowa 1, 02-096 Warsaw, Poland.
| | - Nina Trojan
- Department of Metabolic Regulation, Institute of Biochemistry, Faculty of Biology, University of Warsaw, I. Miecznikowa 1, 02-096 Warsaw, Poland.
| | - Aleksandra Tempes
- Department of Metabolic Regulation, Institute of Biochemistry, Faculty of Biology, University of Warsaw, I. Miecznikowa 1, 02-096 Warsaw, Poland.
| | - Paweł Nalepa
- Department of Metabolic Regulation, Institute of Biochemistry, Faculty of Biology, University of Warsaw, I. Miecznikowa 1, 02-096 Warsaw, Poland.
| | - Joanna Sitek
- Department of Metabolic Regulation, Institute of Biochemistry, Faculty of Biology, University of Warsaw, I. Miecznikowa 1, 02-096 Warsaw, Poland.
| | - Katarzyna Winiarska
- Department of Metabolic Regulation, Institute of Biochemistry, Faculty of Biology, University of Warsaw, I. Miecznikowa 1, 02-096 Warsaw, Poland.
| | - Michał Usarek
- Department of Metabolic Regulation, Institute of Biochemistry, Faculty of Biology, University of Warsaw, I. Miecznikowa 1, 02-096 Warsaw, Poland.
| |
Collapse
|
28
|
|
29
|
DAS DEBABRATA, ARUR SWATHI. Conserved insulin signaling in the regulation of oocyte growth, development, and maturation. Mol Reprod Dev 2017; 84:444-459. [PMID: 28379636 PMCID: PMC5477485 DOI: 10.1002/mrd.22806] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 03/31/2017] [Indexed: 01/01/2023]
Abstract
Insulin signaling regulates various aspects of physiology, such as glucose homeostasis and aging, and is a key determinant of female reproduction in metazoans. That insulin signaling is crucial for female reproductive health is clear from clinical data linking hyperinsulinemic and hypoinsulinemic condition with certain types of ovarian dysfunction, such as altered steroidogenesis, polycystic ovary syndrome, and infertility. Thus, understanding the signaling mechanisms that underlie the control of insulin-mediated ovarian development is important for the accurate diagnosis of and intervention for female infertility. Studies of invertebrate and vertebrate model systems have revealed the molecular determinants that transduce insulin signaling as well as which biological processes are regulated by the insulin-signaling pathway. The molecular determinants of the insulin-signaling pathway, from the insulin receptor to its downstream signaling components, are structurally and functionally conserved across evolution, from worms to mammals-yet, physiological differences in signaling still exist. Insulin signaling acts cooperatively with gonadotropins in mammals and lower vertebrates to mediate various aspects of ovarian development, mainly owing to evolution of the endocrine system in vertebrates. In contrast, insulin signaling in Drosophila and Caenorhabditis elegans directly regulates oocyte growth and maturation. In this review, we compare and contrast insulin-mediated regulation of ovarian functions in mammals, lower vertebrates, C. elegans, and Drosophila, and highlight conserved signaling pathways and regulatory mechanisms in general while illustrating insulin's unique role in specific reproductive processes.
Collapse
Affiliation(s)
- DEBABRATA DAS
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - SWATHI ARUR
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
30
|
Reynolds KA, Boudoures AL, Chi MMY, Wang Q, Moley KH. Adverse effects of obesity and/or high-fat diet on oocyte quality and metabolism are not reversible with resumption of regular diet in mice. Reprod Fertil Dev 2017; 27:716-24. [PMID: 25775080 DOI: 10.1071/rd14251] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 02/04/2015] [Indexed: 01/29/2023] Open
Abstract
Obesity adversely affects reproduction and results in oocyte defects in both mice and humans. In the present study we used a mouse model to examine whether the adverse effects of an obesogenic diet on oocyte metabolism and morphology can be reversed by return to a control diet. The intervention group consisted of C57BL6/J mice placed on a high-fat diet (HFD; 35.8% fat and 20.2% protein by nutritional content) for 6 weeks and then switched to an isocaloric control diet (CD; 13% fat and 25% protein) for 8 weeks (HFD/CD mice). The control group consisted of age-matched C57BL6/J mice maintained on CD for 14 weeks (CD/CD mice). Although metabolic parameters (weight, glucose tolerance and cholesterol levels) of HFD/CD mice returned to normal after this 'diet reversal' period, several oocyte defects were not reversible. These HFD/CD oocytes demonstrated significantly higher percentages of abnormal meiotic spindles, lower mitochondrial membrane potential and lower ATP and citrate levels, and higher percentages of abnormal lipid accumulation and mitochondrial distribution compared with CD/CD mice. These results suggest that the negative effects of an obesogenic diet on oocyte quality are not reversible, despite reversal of metabolic parameters. These data may provide better insight when counselling obese women regarding reproductive options and success.
Collapse
Affiliation(s)
- Kasey A Reynolds
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St Louis, MO 63130, USA
| | - Anna L Boudoures
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St Louis, MO 63130, USA
| | - Maggie M-Y Chi
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St Louis, MO 63130, USA
| | - Qiang Wang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Kelle H Moley
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St Louis, MO 63130, USA
| |
Collapse
|
31
|
Yuan B, Liang S, Kwon JW, Jin YX, Park SH, Wang HY, Sun TY, Zhang JB, Kim NH. The Role of Glucose Metabolism on Porcine Oocyte Cytoplasmic Maturation and Its Possible Mechanisms. PLoS One 2016; 11:e0168329. [PMID: 27997591 PMCID: PMC5173360 DOI: 10.1371/journal.pone.0168329] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 11/30/2016] [Indexed: 12/30/2022] Open
Abstract
In the present study, we investigated the potential role of glucose and pyruvate in the cytoplasmic maturation of porcine oocytes by investigating the effect of glucose and/or pyruvate supplementation, in the presence or absence of 10% porcine follicular fluid (PFF), on meiotic maturation and subsequent embryo development. In the absence of 10% PFF, without exogenous addition of glucose and pyruvate, the medium seemed unable to support maturation. In the presence of 10% PFF, the addition of 5.6 mM glucose and/or 2 mM pyruvate during in vitro maturation of cumulus enclosed oocytes increased MII oocyte and blastocyst rates. In contrast, oocytes denuded of cumulus cells were not able to take full advantage of the glucose in the medium, as only pyruvate was able to increase the MII rate and the subsequent early embryo developmental ability. Treatment of cumulus enclosed oocytes undergoing maturation with 200 μM dehydroepiandrosterone (DHEA), a pentose phosphate pathway inhibitor, or 2 μM iodoacetate (IA), a glycolysis inhibitor, significantly reduced GHS, intra-oocyte ATP, maternal gene expression, and MPF activity levels. DHEA was also able to increase ROS and reduce the levels of NADPH. Moreover, blastocysts of the DHEA- or IA-treated groups presented higher apoptosis rates and markedly lower cell proliferation cell rates than those of the non-treated group. In conclusion, our results suggest that oocytes maturing in the presence of 10% PFF can make full use of energy sources through glucose metabolism only when they are accompanied by cumulus cells, and that pentose phosphate pathway (PPP) and glycolysis promote porcine oocyte cytoplasmic maturation by supplying energy, regulating maternal gene expression, and controlling MPF activity.
Collapse
Affiliation(s)
- Bao Yuan
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, Jilin, China
- Molecular Embryology Laboratory, Department of Animal Sciences, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Shuang Liang
- Molecular Embryology Laboratory, Department of Animal Sciences, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Jeong-Woo Kwon
- Molecular Embryology Laboratory, Department of Animal Sciences, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Yong-Xun Jin
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, Jilin, China
- Molecular Embryology Laboratory, Department of Animal Sciences, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Shun-Ha Park
- Molecular Embryology Laboratory, Department of Animal Sciences, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Hai-Yang Wang
- Molecular Embryology Laboratory, Department of Animal Sciences, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Tian-Yi Sun
- Molecular Embryology Laboratory, Department of Animal Sciences, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Jia-Bao Zhang
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, Jilin, China
- * E-mail: (NHK); (JBZ)
| | - Nam-Hyung Kim
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, Jilin, China
- Molecular Embryology Laboratory, Department of Animal Sciences, Chungbuk National University, Cheongju, Chungbuk, South Korea
- * E-mail: (NHK); (JBZ)
| |
Collapse
|
32
|
Li SY, Song Z, Song MJ, Qin JW, Zhao ML, Yang ZM. Impaired receptivity and decidualization in DHEA-induced PCOS mice. Sci Rep 2016; 6:38134. [PMID: 27924832 PMCID: PMC5141439 DOI: 10.1038/srep38134] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 11/07/2016] [Indexed: 12/27/2022] Open
Abstract
Polycystic ovary syndrome (PCOS), a complex endocrine disorder, is a leading cause of female infertility. An obvious reason for infertility in PCOS women is anovulation. However, success rate with high quality embryos selected by assisted reproduction techniques in PCOS patients still remain low with a high rate of early clinical pregnancy loss, suggesting a problem in uterine receptivity. Using a dehydroepiandrosterone-induced mouse model of PCOS, some potential causes of decreased fertility in PCOS patients were explored. In our study, ovulation problem also causes sterility in PCOS mice. After blastocysts from normal mice are transferred into uterine lumen of pseudopregnant PCOS mice, the rate of embryo implantation was reduced. In PCOS mouse uteri, the implantation-related genes are also dysregulated. Additionally, artificial decidualization is severely impaired in PCOS mice. The serum estrogen level is significantly higher in PCOS mice than vehicle control. The high level of estrogen and potentially impaired LIF-STAT3 pathway may lead to embryo implantation failure in PCOS mice. Although there are many studies about effects of PCOS on endometrium, both embryo transfer and artificial decidualization are applied to exclude the effects from ovulation and embryos in our study.
Collapse
Affiliation(s)
- Shu-Yun Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Zhuo Song
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Min-Jie Song
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Jia-Wen Qin
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Meng-Long Zhao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Zeng-Ming Yang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| |
Collapse
|
33
|
Cavnar SP, Xiao A, Gibbons AE, Rickelmann AD, Neely T, Luker KE, Takayama S, Luker GD. Imaging Sensitivity of Quiescent Cancer Cells to Metabolic Perturbations in Bone Marrow Spheroids. Tomography 2016; 2:146-157. [PMID: 27478871 PMCID: PMC4963031 DOI: 10.18383/j.tom.2016.00157] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Malignant cells from breast cancer and other common cancers such as prostate and melanoma may persist in bone marrow as quiescent, non-dividing cells that remain viable for years or even decades before resuming proliferation to cause recurrent disease. This phenomenon, referred to clinically as tumor dormancy, poses tremendous challenges to curing patients with breast cancer. Quiescent tumor cells resist chemotherapy drugs that predominantly target proliferating cells, limiting success of neo-adjuvant and adjuvant therapies. We recently developed a 3D spheroid model of quiescent breast cancer cells in bone marrow for mechanistic and drug testing studies. We combined this model with optical imaging methods for label-free detection of cells preferentially utilizing glycolysis versus oxidative metabolism to investigate the metabolic state of co-culture spheroids with different bone marrow stromal and breast cancer cells. Through imaging and biochemical assays, we identified different metabolic states of bone marrow stromal cells that control metabolic status and flexibilities of co-cultured breast cancer cells. We tested metabolic stresses and targeted inhibition of specific metabolic pathways to identify approaches to preferentially eliminate quiescent breast cancer cells from bone marrow environments. These studies establish an integrated imaging approach to analyze metabolism in complex tissue environments to identify new metabolically-targeted cancer therapies.
Collapse
Affiliation(s)
- Stephen P. Cavnar
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, Michigan
- Biointerfaces Institute, University of Michigan College of Engineering, Ann Arbor, Michigan
| | - Annie Xiao
- Department of Radiology, Center for Molecular Imaging, University of Michigan Medical School, Ann Arbor, Michigan
| | - Anne E. Gibbons
- Department of Radiology, Center for Molecular Imaging, University of Michigan Medical School, Ann Arbor, Michigan
| | - Andrew D. Rickelmann
- Department of Radiology, Center for Molecular Imaging, University of Michigan Medical School, Ann Arbor, Michigan
| | - Taylor Neely
- Department of Radiology, Center for Molecular Imaging, University of Michigan Medical School, Ann Arbor, Michigan
| | - Kathryn E. Luker
- Department of Radiology, Center for Molecular Imaging, University of Michigan Medical School, Ann Arbor, Michigan
| | - Shuichi Takayama
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, Michigan
- Biointerfaces Institute, University of Michigan College of Engineering, Ann Arbor, Michigan
- Department of Macromolecular Science and Engineering, University of Michigan College of Engineering, Ann Arbor, Michigan; and
| | - Gary D. Luker
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, Michigan
- Department of Radiology, Center for Molecular Imaging, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Macromolecular Science and Engineering, University of Michigan College of Engineering, Ann Arbor, Michigan; and
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
34
|
Zhao H, Zhao Y, Li T, Li M, Li J, Li R, Liu P, Yu Y, Qiao J. Metabolism alteration in follicular niche: The nexus among intermediary metabolism, mitochondrial function, and classic polycystic ovary syndrome. Free Radic Biol Med 2015; 86:295-307. [PMID: 26057937 DOI: 10.1016/j.freeradbiomed.2015.05.013] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 04/21/2015] [Accepted: 05/11/2015] [Indexed: 02/07/2023]
Abstract
Classic polycystic ovary syndrome (PCOS) is a high-risk phenotype accompanied by increased risks of reproductive and metabolic abnormalities; however, the local metabolism characteristics of the ovaries and their effects on germ cell development are unclear. The present study used targeted metabolomics to detect alterations in the intermediate metabolites of follicular fluid from classic PCOS patients, and the results indicated that hyperandrogenism but not obesity induced the changed intermediate metabolites in classic PCOS patients. Regarding the direct contact, we identified mitochondrial function, redox potential, and oxidative stress in cumulus cells which were necessary to support oocyte growth before fertilization, and suggested dysfunction of mitochondria, imbalanced redox potential, and increased oxidative stress in cumulus cells of classic PCOS patients. Follicular fluid intermediary metabolic profiles provide signatures of classic PCOS ovary local metabolism and establish a close link with mitochondria dysfunction of cumulus cells, highlighting the role of metabolic signal and mitochondrial cross talk involved in the pathogenesis of classic PCOS.
Collapse
Affiliation(s)
- Hongcui Zhao
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Yue Zhao
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Tianjie Li
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
| | - Min Li
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Junsheng Li
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Rong Li
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Ping Liu
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Yang Yu
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China.
| | - Jie Qiao
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China.
| |
Collapse
|
35
|
Louden ED, Luzzo KM, Jimenez PT, Chi T, Chi M, Moley KH. TallyHO obese female mice experience poor reproductive outcomes and abnormal blastocyst metabolism that is reversed by metformin. Reprod Fertil Dev 2015; 27:31-9. [PMID: 25472042 DOI: 10.1071/rd14339] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Obese women experience worse reproductive outcomes than normal weight women, specifically infertility, pregnancy loss, fetal malformations and developmental delay of offspring. The aim of the present study was to use a genetic mouse model of obesity to recapitulate the human reproductive phenotype and further examine potential mechanisms and therapies. New inbred, polygenic Type 2 diabetic TallyHO mice and age-matched control C57BL/6 mice were superovulated to obtain morula or blastocyst stage embryos that were cultured in human tubal fluid (HTF) medium. Deoxyglucose uptake was determined for individual insulin-stimulated blastocysts. Apoptosis was detected by confocal microscopy using the terminal deoxyribonucleotidyl transferase-mediated dUTP-digoxigenin nick end-labelling (TUNEL) assay and Topro-3 nuclear dye. Embryos were scored for TUNEL-positive as a percentage of total nuclei. AMP-activated protein kinase (AMPK) activation, tumour necrosis factor (TNF)-α expression and adiponectin expression were analysed by western immunoblot and confocal immunofluorescent microscopy. Lipid accumulation was assayed by BODIPY. Comparisons were made between TallyHO morulae cultured to blastocyst embryos in either HTF medium or HTF medium with 25 μg mL(-1) metformin. TallyHO mice developed whole body abnormal insulin tolerance, had decreased litter sizes and increased non-esterified fatty acid levels. Blastocysts from TallyHO mice exhibited increased apoptosis, decreased insulin sensitivity and decreased AMPK. A possible cause for the insulin resistance and abnormal AMPK phosphorylation was the increased TNF-α expression and lipid accumulation, as detected by BODIPY, in TallyHO blastocysts and decreased adiponectin. Culturing TallyHO morulae with the AMPK activator metformin led to a reversal of all the abnormal findings, including increased AMPK phosphorylation, improved insulin-stimulated glucose uptake and normalisation of lipid accumulation. Women with obesity and insulin resistance experience poor pregnancy outcomes. Previously we have shown in mouse models of insulin resistance that AMPK activity is decreased and that activators of AMPK reverse poor embryo outcomes. Here, we show for the first time using a genetically altered obese model, not a diet-induced model, that metformin reverses many of the adverse effects of obesity at the level of the blastocyst. Expanding on this we determine that activation of AMPK via metformin reduces lipid droplet accumulation, presumably by eliminating the inhibitory effects of TNF-α, resulting in normalisation of fatty acid oxidation and HADH2 (hydroxyacyl-CoA dehydrogenase/3-ketoacyl-CoA thiolase/enoyl-CoA hydratase (trifunctional protein), alpha subunit) activity. Metformin exposure in vitro was able to partially reverse these effects, at the level of the blastocyst, and may thus be effective in preventing the adverse effects of obesity on pregnancy and reproductive outcomes.
Collapse
Affiliation(s)
- Erica D Louden
- Department of Obstetrics and Gynecology, Washington University School of Medicine, 425 S. Euclid Ave, Campus Box 8064, St Louis, MO 63110, USA
| | - Kerri M Luzzo
- Department of Obstetrics and Gynecology, Washington University School of Medicine, 425 S. Euclid Ave, Campus Box 8064, St Louis, MO 63110, USA
| | - Patricia T Jimenez
- Department of Obstetrics and Gynecology, Washington University School of Medicine, 425 S. Euclid Ave, Campus Box 8064, St Louis, MO 63110, USA
| | - Tiffany Chi
- Department of Obstetrics and Gynecology, Washington University School of Medicine, 425 S. Euclid Ave, Campus Box 8064, St Louis, MO 63110, USA
| | - Maggie Chi
- Department of Obstetrics and Gynecology, Washington University School of Medicine, 425 S. Euclid Ave, Campus Box 8064, St Louis, MO 63110, USA
| | - Kelle H Moley
- Department of Obstetrics and Gynecology, Washington University School of Medicine, 425 S. Euclid Ave, Campus Box 8064, St Louis, MO 63110, USA
| |
Collapse
|
36
|
Pastò A, Bellio C, Pilotto G, Ciminale V, Silic-Benussi M, Guzzo G, Rasola A, Frasson C, Nardo G, Zulato E, Nicoletto MO, Manicone M, Indraccolo S, Amadori A. Cancer stem cells from epithelial ovarian cancer patients privilege oxidative phosphorylation, and resist glucose deprivation. Oncotarget 2015; 5:4305-19. [PMID: 24946808 PMCID: PMC4147325 DOI: 10.18632/oncotarget.2010] [Citation(s) in RCA: 229] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
We investigated the metabolic profile of cancer stem cells (CSC) isolated from patients with epithelial ovarian cancer. CSC overexpressed genes associated with glucose uptake, oxidative phosphorylation (OXPHOS), and fatty acid β-oxidation, indicating higher ability to direct pyruvate towards the Krebs cycle. Consistent with a metabolic profile dominated by OXPHOS, the CSC showed higher mitochondrial reactive oxygen species (ROS) production and elevated membrane potential, and underwent apoptosis upon inhibition of the mitochondrial respiratory chain. The CSC also had a high rate of pentose phosphate pathway (PPP) activity, which is not typical of cells privileging OXPHOS over glycolysis, and may rather reflect the PPP role in recharging scavenging enzymes. Furthermore, CSC resisted in vitro and in vivo glucose deprivation, while maintaining their CSC phenotype and OXPHOS profile. These observations may explain the CSC resistance to anti-angiogenic therapies, and indicate this peculiar metabolic profile as a possible target of novel treatment strategies.
Collapse
Affiliation(s)
- Anna Pastò
- Department of Surgery, Oncology, and Gastroenterology, Oncology Section, University of Padova, Padova, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Huang Y, Yu Y, Gao J, Li R, Zhang C, Zhao H, Zhao Y, Qiao J. Impaired oocyte quality induced by dehydroepiandrosterone is partially rescued by metformin treatment. PLoS One 2015; 10:e0122370. [PMID: 25811995 PMCID: PMC4374838 DOI: 10.1371/journal.pone.0122370] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 02/15/2015] [Indexed: 12/25/2022] Open
Abstract
The present study evaluated the influence of hyperandrogenism on oocyte quality using a murine PCOS model induced by dehydroepiandrosterone (DHEA) and further explored the effect of metformin treatment. Female BALB/c mice were treated with a vehicle control or DHEA (6 mg /100 g body weight) or DHEA plus metformin (50 mg /100 g body weight) for 20 consecutive days. DHEA-induced mice resembled some characters of human PCOS, such as irregular sexual cycles and polycystic ovaries. After the model validation was completed, metaphase II (MII) oocytes were retrieved and subsequent analyses of oocyte quality were performed. DHEA-treated mice yielded fewer MII oocytes, which displayed decreased mtDNA copy number, ATP content, inner mitochondrial membrane potential, excessive oxidative stress and impaired embryo development competence compared with those in control mice. Metformin treatment partially attenuated those damages, as evidenced by the increased fertilization and blastocyst rate, ATP content, GSH concentration and GSH/GSSG ratio, and decreased reactive oxygen species levels. No significant difference in normal spindle assembly was observed among the three groups. During in vitro maturation (IVM), the periods of germinal vesicle breakdown (GVBD) and the first polar body (PB1) extrusion were extended and the maturation rate of GVBD oocytes was decreased in DHEA mice compared with controls. Metformin treatment decreased the time elapsed of GVBD while had no effect on PB1 extrusion. These results indicated that excessive androgen is detrimental to oocyte quality while metformin treatment is, directly or indirectly, beneficial for oocyte quality improvement.
Collapse
Affiliation(s)
- Ying Huang
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Yang Yu
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
| | - Jiangman Gao
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Rong Li
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Chunmei Zhang
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
| | - Hongcui Zhao
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Yue Zhao
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
- * E-mail: (JQ); (YZ)
| | - Jie Qiao
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- * E-mail: (JQ); (YZ)
| |
Collapse
|
38
|
Oocyte environment: follicular fluid and cumulus cells are critical for oocyte health. Fertil Steril 2014; 103:303-16. [PMID: 25497448 DOI: 10.1016/j.fertnstert.2014.11.015] [Citation(s) in RCA: 410] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 11/08/2014] [Accepted: 11/10/2014] [Indexed: 02/06/2023]
Abstract
Bidirectional somatic cell-oocyte signaling is essential to create a changing intrafollicular microenvironment that controls primordial follicle growth into a cohort of growing follicles, from which one antral follicle is selected to ovulate a healthy oocyte. Such intercellular communications allow the oocyte to determine its own fate by influencing the intrafollicular microenvironment, which in turn provides the necessary cellular functions for oocyte developmental competence, which is defined as the ability of the oocyte to complete meiosis and undergo fertilization, embryogenesis, and term development. These coordinated somatic cell-oocyte interactions attempt to balance cellular metabolism with energy requirements during folliculogenesis, including changing energy utilization during meiotic resumption. If these cellular mechanisms are perturbed by metabolic disease and/or maternal aging, molecular damage of the oocyte can alter macromolecules, induce mitochondrial mutations, and reduce adenosine triphosphate production, all of which can harm the oocyte. Recent technologies are now exploring transcriptional, translational, and post-translational events within the human follicle with the goal of identifying biomarkers that reliably predict oocyte quality in the clinical setting.
Collapse
|
39
|
Gu L, Liu H, Gu X, Boots C, Moley KH, Wang Q. Metabolic control of oocyte development: linking maternal nutrition and reproductive outcomes. Cell Mol Life Sci 2014; 72:251-71. [PMID: 25280482 DOI: 10.1007/s00018-014-1739-4] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2014] [Revised: 09/12/2014] [Accepted: 09/22/2014] [Indexed: 02/01/2023]
Abstract
Obesity, diabetes, and related metabolic disorders are major health issues worldwide. As the epidemic of metabolic disorders continues, the associated medical co-morbidities, including the detrimental impact on reproduction, increase as well. Emerging evidence suggests that the effects of maternal nutrition on reproductive outcomes are likely to be mediated, at least in part, by oocyte metabolism. Well-balanced and timed energy metabolism is critical for optimal development of oocytes. To date, much of our understanding of oocyte metabolism comes from the effects of extrinsic nutrients on oocyte maturation. In contrast, intrinsic regulation of oocyte development by metabolic enzymes, intracellular mediators, and transport systems is less characterized. Specifically, decreased acid transport proteins levels, increased glucose/lipid content and elevated reactive oxygen species in oocytes have been implicated in meiotic defects, organelle dysfunction and epigenetic alteration. Therefore, metabolic disturbances in oocytes may contribute to the diminished reproductive potential experienced by women with metabolic disorders. In-depth research is needed to further explore the underlying mechanisms. This review also discusses several approaches for metabolic analysis. Metabolomic profiling of oocytes, the surrounding granulosa cells, and follicular fluid will uncover the metabolic networks regulating oocyte development, potentially leading to the identification of oocyte quality markers and prevention of reproductive disease and poor outcomes in offspring.
Collapse
Affiliation(s)
- Ling Gu
- College of Animal Science and Technology, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, Jiangsu, China,
| | | | | | | | | | | |
Collapse
|
40
|
Frank LA, Sutton-McDowall ML, Gilchrist RB, Thompson JG. The effect of peri-conception hyperglycaemia and the involvement of the hexosamine biosynthesis pathway in mediating oocyte and embryo developmental competence. Mol Reprod Dev 2014; 81:391-408. [DOI: 10.1002/mrd.22299] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 12/31/2013] [Indexed: 12/12/2022]
Affiliation(s)
- Laura A. Frank
- The Robinson Institute, The Research Centre for Reproductive Health, School of Paediatrics and Reproductive Health, The University of Adelaide; Adelaide South Australia Australia
| | - Melanie L. Sutton-McDowall
- The Robinson Institute, The Research Centre for Reproductive Health, School of Paediatrics and Reproductive Health, The University of Adelaide; Adelaide South Australia Australia
| | - Robert B. Gilchrist
- The Robinson Institute, The Research Centre for Reproductive Health, School of Paediatrics and Reproductive Health, The University of Adelaide; Adelaide South Australia Australia
| | - Jeremy G. Thompson
- The Robinson Institute, The Research Centre for Reproductive Health, School of Paediatrics and Reproductive Health, The University of Adelaide; Adelaide South Australia Australia
| |
Collapse
|