1
|
Devi S, Negi S, Tandel N, Dalai SK, Tyagi RK. Oleuropein: a viable therapeutic option for malaria and cancer. Drug Discov Today 2025; 30:104254. [PMID: 39608487 DOI: 10.1016/j.drudis.2024.104254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 11/19/2024] [Accepted: 11/22/2024] [Indexed: 11/30/2024]
Abstract
Oleuropein (OLP) holds promise as a therapeutic candidate for both Plasmodium falciparum infection and cancer. It modulates the phosphoinositide 3-kinase (PI3K)-Akt1 signaling pathway to regulate inflammation and restore immune homeostasis. Moreover, it influences the cell death/autophagy axis, along with increasing the antimalarial efficacy of artemisinin. Our findings indicate that the anti-breast-cancer effect of OLP could be mediated by regulating the balance of T helper 17 and regulatory T cells. Additionally, we discuss the use of hematopoietic-stem-cell-transplanted immunodeficient mice with a humanized immune system for validating the antimalarial activity, autophagy and anticancer activity of OLP.
Collapse
Affiliation(s)
- Sonia Devi
- Division of Cell Biology and Immunology, Biomedical Parasitology and Translational-immunology Lab, CSIR-Institute of Microbial Technology (IMTECH), Chandigarh 160036, India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad 201002, India
| | - Sushmita Negi
- Division of Cell Biology and Immunology, Biomedical Parasitology and Translational-immunology Lab, CSIR-Institute of Microbial Technology (IMTECH), Chandigarh 160036, India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad 201002, India
| | - Nikunj Tandel
- CSIR-Centre For Cellular & Molecular Biology (CCMB), Hyderabad, Telangana 500007, India
| | - Sarat K Dalai
- Institute of Science, Nirma University, SG Highway, Gujarat 382481, India
| | - Rajeev K Tyagi
- Division of Cell Biology and Immunology, Biomedical Parasitology and Translational-immunology Lab, CSIR-Institute of Microbial Technology (IMTECH), Chandigarh 160036, India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
2
|
Makhlouf S, Quinn C, Toss M, Alsaleem M, Atallah NM, Ibrahim A, Rutland CS, Mongan NP, Rakha EA. Quantitative expression of oestrogen receptor in breast cancer: Clinical and molecular significance. Eur J Cancer 2024; 197:113473. [PMID: 38103327 DOI: 10.1016/j.ejca.2023.113473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 12/04/2023] [Accepted: 12/04/2023] [Indexed: 12/19/2023]
Abstract
BACKGROUND Oestrogen receptor (ER) positive breast cancer (BC) patients are eligible for endocrine therapy (ET), regardless of ER immunohistochemical expression level. There is a wide spectrum of ER expression and the response to ET is not uniform. This study aimed to assess the clinical and molecular consequences of ER heterogeneity with respect to ET-response. METHODS ER expression, categorised by percentage and staining intensity in a large BC cohort (n = 7559) was correlated with clinicopathological parameters and patient ET response. The Cancer Genome Atlas Data BC cohort (n = 1047) was stratified by ER expression and transcriptomic analysis completed to better understand the molecular basis of ER heterogeneity. RESULTS The quantitative proportional increase in ER expression was positively associated with favourable prognostic parameters. Tumours with 1-9% ER expression were characteristically similar to ER-negative (<1%) tumours. Maximum ET-response was observed in tumours with 100% ER expression, with responses significantly different to tumours exhibiting ER at < 100% and significantly decreased survival rates were observed in tumours with 50% and 10% of ER expression. The Histochemical-score (H-score), which considers both staining intensity and percentage, added significant prognostic value over ER percentage alone with significant outcome differences observed at H-scores of 30, 100 and 200. There was a positive correlation between ER expression and ESR1 mRNA expression and expression of ER-regulated genes. Pathway analysis identified differential expression in key cancer-related pathways in different ER-positive groups. CONCLUSION ET-response is statistically proportionally related to ER expression with significant differences observed at 10%, 50% and 100%. The H-score adds prognostic and predictive information.
Collapse
Affiliation(s)
- Shorouk Makhlouf
- Academic Unit for Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK; Department of Pathology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Cecily Quinn
- Irish National Breast Screening Programme and Department of Histopathology, St. Vincent's University Hospital, Dublin, Ireland
| | - Michael Toss
- Academic Unit for Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK; Department of Histopathology, Sheffield Teaching Hospitals NHS Trust, Sheffield, UK
| | - Mansour Alsaleem
- Academic Unit for Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK; Unit of Scientific Research, Applied College, Qassim University, Saudi Arabia
| | - Nehal M Atallah
- Academic Unit for Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK; Department of Pathology, Faculty of Medicine, Menoufia University, Menoufia, Egypt
| | - Asmaa Ibrahim
- Academic Unit for Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| | - Catrin S Rutland
- School of Veterinary Medicine and Sciences, University of Nottingham, Sutton Bonington, UK
| | - Nigel P Mongan
- Biodiscovery Institute, School of Veterinary Medicine and Sciences, University of Nottingham, Nottingham, UK; Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Emad A Rakha
- Academic Unit for Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK; Department of Histopathology, Nottingham University Hospitals NHS Trust, Nottingham, UK; Department of Pathology, Hamad Medical Corporation, Doha, Qatar.
| |
Collapse
|
3
|
Lin H, Jia Y, Han F, Xia C, Zhao Q, Zhang J, Li E. Toxic effects of waterborne benzylparaben on the growth, antioxidant capacity and lipid metabolism of Nile tilapia (Oreochromis niloticus). AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2022; 248:106197. [PMID: 35623196 DOI: 10.1016/j.aquatox.2022.106197] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/13/2022] [Accepted: 05/14/2022] [Indexed: 06/15/2023]
Abstract
Benzylparaben (BzP) is a potential endocrine disruptor; however, its antioxidant defense, lipotoxicity and underlying mechanism of BzP in aquatic organisms are unknown. This study investigated the impacts of waterborne low-, environmental-related and high-level benzylparaben on the growth, antioxidant capacity, lipid metabolism and lipidomic response of Nile tilapia (Oreochromis niloticus). Juvenile tilapia (0.60 ± 0.11 g) were exposed to 0, 5, 50, 500 and 5000 ng/L benzylparaben for 8 weeks in quadruplicate for each group. Benzylparaben increased the body crude fat content but decreased brain acetylcholinesterase activity in O. niloticus. Benzylparaben caused oxidative stress, leading to hepatic morphology damage and lipid metabolism disorders in fish. Lipidomic analysis identified 13 lipid classes in fish liver. Benzylparaben exposure induced metabolic disorders of glycerol phospholipids, glycerolipids and sphingomyelins in fish liver. These findings indicate that environmentally related benzylparaben levels (5 to 50 ng/L) could induce an antioxidant response, result in triglyceride accumulation, and increase adipocyte formation and fatty acid intake in tilapia. However, high benzylparaben concentrations inhibit lipid deposition, presumably due to the effects of the antioxidant system, and induce tissue inflammation. Therefore, this study provides new insights into the toxic effects and potential mechanism of benzylparaben in fish, especially from the aspect of lipid metabolism.
Collapse
Affiliation(s)
- Hongxing Lin
- Key Laboratory of Tropical Hydrobiology and Biotechnology of Hainan Province, Hainan Aquaculture Breeding Engineering Research Center, College of Marine Sciences, Hainan University, Haikou, Hainan 570228, China
| | - Yongyi Jia
- Key Laboratory of Healthy Freshwater Aquaculture, Ministry of Agriculture and Rural Affairs; Key Laboratory of Fish Health and Nutrition of Zhejiang Province, Zhejiang Institute of Freshwater Fisheries, Huzhou 313001, China
| | - Fenglu Han
- Key Laboratory of Tropical Hydrobiology and Biotechnology of Hainan Province, Hainan Aquaculture Breeding Engineering Research Center, College of Marine Sciences, Hainan University, Haikou, Hainan 570228, China.
| | - Chuyan Xia
- Key Laboratory of Tropical Hydrobiology and Biotechnology of Hainan Province, Hainan Aquaculture Breeding Engineering Research Center, College of Marine Sciences, Hainan University, Haikou, Hainan 570228, China
| | - Qun Zhao
- Key Laboratory of Tropical Hydrobiology and Biotechnology of Hainan Province, Hainan Aquaculture Breeding Engineering Research Center, College of Marine Sciences, Hainan University, Haikou, Hainan 570228, China
| | - Jiliang Zhang
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, Key Laboratory of Tropical Animal and Plant Ecology of Hainan Province, College of Life Sciences, Hainan Normal University, Haikou, Hainan, China
| | - Erchao Li
- Key Laboratory of Tropical Hydrobiology and Biotechnology of Hainan Province, Hainan Aquaculture Breeding Engineering Research Center, College of Marine Sciences, Hainan University, Haikou, Hainan 570228, China.
| |
Collapse
|
4
|
Vail GM, Walley SN, Yasrebi A, Maeng A, Degroat TJ, Conde KM, Roepke TA. Implications of estrogen receptor alpha (ERa) with the intersection of organophosphate flame retardants and diet-induced obesity in adult mice. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2022; 85:397-413. [PMID: 35045790 PMCID: PMC8916992 DOI: 10.1080/15287394.2022.2026849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Previously, organophosphate flame retardants (OPFRs) were found to produce intersecting disruptions of energy homeostasis using an adult mouse model of diet-induced obesity. Using the same mixture consisting of 1 mg/kg/day of each triphenyl phosphate, tricresyl phosphate, and tris(1,3-dichloro-2-propyl)phosphate, the current study aimed to identify the role of estrogen receptor alpha (ERα) in OPFR-induced disruption, utilizing ERα knockout (ERαKO) mice fed either a low-fat diet (LFD) or high-fat diet (HFD). Body weight and composition, food intake patterns, glucose and insulin tolerance, circulating peptide hormones, and expression of hypothalamic genes associated with energy homeostasis were measured. When fed HFD, no marked direct effects of OPFR were observed in mice lacking ERα, suggesting a role for ERα in generating previously reported wildtype (WT) findings. Male ERαKO mice fed LFD experienced decreased feeding efficiency and altered insulin tolerance, whereas their female counterparts displayed less fat mass and circulating ghrelin when exposed to OPFRs. These effects were not noted in the previous WT study, indicating that loss of ERα may sensitize animals fed LFD to alternate pathways of endocrine disruption by OFPRs. Collectively, these data demonstrate both direct and indirect actions of OPFRs on ERα-mediated pathways governing energy homeostasis and support a growing body of evidence urging concern for risk of human exposure.
Collapse
Affiliation(s)
- Gwyndolin M. Vail
- Joint Graduate Program in Toxicology, Rutgers, The State University of New Jersey, Piscataway, NJ. USA
| | - Sabrina N. Walley
- Joint Graduate Program in Toxicology, Rutgers, The State University of New Jersey, Piscataway, NJ. USA
| | - Ali Yasrebi
- Department of Animal Sciences, School of Environmental & Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ. USA
| | - Angela Maeng
- Department of Animal Sciences, School of Environmental & Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ. USA
| | - Thomas J. Degroat
- Graduate Program in Endocrinology and Animal Biosciences, School of Environmental & Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ. USA
| | - Kristie M. Conde
- Graduate Program in Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ. USA
| | - Troy A. Roepke
- Joint Graduate Program in Toxicology, Rutgers, The State University of New Jersey, Piscataway, NJ. USA
- Department of Animal Sciences, School of Environmental & Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ. USA
- Graduate Program in Endocrinology and Animal Biosciences, School of Environmental & Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ. USA
- Graduate Program in Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ. USA
| |
Collapse
|
5
|
The Glitazars Paradox: Cardiotoxicity of the Metabolically Beneficial Dual PPARα and PPARγ Activation. J Cardiovasc Pharmacol 2021; 76:514-526. [PMID: 33165133 DOI: 10.1097/fjc.0000000000000891] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The most common complications in patients with type-2 diabetes are hyperglycemia and hyperlipidemia that can lead to cardiovascular disease. Alleviation of these complications constitutes the major therapeutic approach for the treatment of diabetes mellitus. Agonists of peroxisome proliferator-activated receptor (PPAR) alpha and PPARγ are used for the treatment of hyperlipidemia and hyperglycemia, respectively. PPARs belong to the nuclear receptors superfamily and regulate fatty acid metabolism. PPARα ligands, such as fibrates, reduce circulating triglyceride levels, and PPARγ agonists, such as thiazolidinediones, improve insulin sensitivity. Dual-PPARα/γ agonists (glitazars) were developed to combine the beneficial effects of PPARα and PPARγ agonism. Although they improved metabolic parameters, they paradoxically aggravated congestive heart failure in patients with type-2 diabetes via mechanisms that remain elusive. Many of the glitazars, such as muraglitazar, tesaglitazar, and aleglitazar, were abandoned in phase-III clinical trials. The objective of this review article pertains to the understanding of how combined PPARα and PPARγ activation, which successfully targets the major complications of diabetes, causes cardiac dysfunction. Furthermore, it aims to suggest interventions that will maintain the beneficial effects of dual PPARα/γ agonism and alleviate adverse cardiac outcomes in diabetes.
Collapse
|
6
|
Kim JY, Park EJ, Kim SM, Lee HJ. Optimization of adipogenic differentiation conditions for canine adipose-derived stem cells. J Vet Sci 2021; 22:e53. [PMID: 34170094 PMCID: PMC8318799 DOI: 10.4142/jvs.2021.22.e53] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/24/2021] [Accepted: 06/04/2021] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Canine adipose-derived stem cells (cADSCs) exhibit various differentiation properties and are isolated from the canine subcutaneous fat. Although cADSCs are valuable as tools for research on adipogenic differentiation, studies focusing on adipogenic differentiation methods and the underlying mechanisms are still lacking. OBJECTIVES In this study, we aimed to establish an optimal method for adipogenic differentiation conditions of cADSCs and evaluate the role of peroxisome proliferator-activated receptor gamma (PPARγ) and estrogen receptor (ER) signaling in the adipogenic differentiation. METHODS To induce adipogenic differentiation of cADSCs, 3 different adipogenic medium conditions, MDI, DRI, and MDRI, using 3-isobutyl-1-methylxanthine (M), dexamethasone (D), insulin (I), and rosiglitazone (R) were tested. RESULTS MDRI, addition of PPARγ agonist rosiglitazone to MDI, was the most significantly facilitated cADSC into adipocyte. GW9662, an antagonist of PPARγ, significantly reduced adipogenic differentiation induced by rosiglitazone. Adipogenic differentiation was also stimulated when 17β-estradiol was added to MDI and DRI, and this stimulation was inhibited by the ER antagonist ICI182,780. CONCLUSIONS Taken together, our results suggest that PPARγ and ER signaling are related to the adipogenic differentiation of cADSCs. This study could provide basic information for future research on obesity or anti-obesity mechanisms in dogs.
Collapse
Affiliation(s)
- Jong Yeon Kim
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam 13120, Korea
| | - Eun Jung Park
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam 13120, Korea.,Institute for Aging and Clinical Nutrition Research, Gachon University, Seongnam 13120, Korea
| | - Sung Min Kim
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam 13120, Korea.,Institute for Aging and Clinical Nutrition Research, Gachon University, Seongnam 13120, Korea
| | - Hae Jeung Lee
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam 13120, Korea.,Institute for Aging and Clinical Nutrition Research, Gachon University, Seongnam 13120, Korea.
| |
Collapse
|
7
|
Estrogen and Glycemic Homeostasis: The Fundamental Role of Nuclear Estrogen Receptors ESR1/ESR2 in Glucose Transporter GLUT4 Regulation. Cells 2021; 10:cells10010099. [PMID: 33430527 PMCID: PMC7827878 DOI: 10.3390/cells10010099] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/03/2021] [Accepted: 01/04/2021] [Indexed: 02/07/2023] Open
Abstract
Impaired circulating estrogen levels have been related to impaired glycemic homeostasis and diabetes mellitus (DM), both in females and males. However, for the last twenty years, the relationship between estrogen, glycemic homeostasis and the mechanisms involved has remained unclear. The characterization of estrogen receptors 1 and 2 (ESR1 and ESR2) and of insulin-sensitive glucose transporter type 4 (GLUT4) finally offered a great opportunity to shed some light on estrogen regulation of glycemic homeostasis. In this manuscript, we review the relationship between estrogen and DM, focusing on glycemic homeostasis, estrogen, ESR1/ESR2 and GLUT4. We review glycemic homeostasis and GLUT4 expression (muscle and adipose tissues) in Esr1−/− and Esr2−/− transgenic mice. We specifically address estradiol-induced and ESR1/ESR2-mediated regulation of the solute carrier family 2 member 4 (Slc2a4) gene, examining ESR1/ESR2-mediated genomic mechanisms that regulate Slc2a4 transcription, especially those occurring in cooperation with other transcription factors. In addition, we address the estradiol-induced translocation of ESR1 and GLUT4 to the plasma membrane. Studies make it clear that ESR1-mediated effects are beneficial, whereas ESR2-mediated effects are detrimental to glycemic homeostasis. Thus, imbalance of the ESR1/ESR2 ratio may have important consequences in metabolism, highlighting that ESR2 hyperactivity assumes a diabetogenic role.
Collapse
|
8
|
Hirao-Suzuki M, Takeda S, Koga T, Takiguchi M, Toda A. Cannabidiolic acid dampens the expression of cyclooxygenase-2 in MDA-MB-231 breast cancer cells: Possible implication of the peroxisome proliferator-activated receptor β/δ abrogation. J Toxicol Sci 2020; 45:227-236. [PMID: 32238697 DOI: 10.2131/jts.45.227] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
A growing body of experimental evidence strongly suggests that cannabidiolic acid (CBDA), a major component of the fiber-type cannabis plant, exerts a variety of biological activities. We have reported that CBDA can abrogate cyclooxygenase-2 (COX-2) expression and its enzymatic activity. It is established that aberrant expression of COX-2 correlates with the degree of malignancy in breast cancer. Although the reduction of COX-2 expression by CBDA offers an attractive medicinal application, the molecular mechanisms underlying these effects have not fully been established. It has been reported that COX-2 expression is positively controlled by peroxisome proliferator-activated receptor β/δ (PPARβ/δ) in some cancerous cells, although there is "no" modulatory element for PPARβ/δ on the COX-2 promoter. No previous studies have examined whether an interaction between PPARβ/δ-mediated signaling and COX-2 expression exists in MDA-MB-231 cells. We confirmed, for the first time, that COX-2 expression is positively modulated by PPARβ/δ-mediated signaling in MDA-MB-231 cells. CBDA inhibits PPARβ/δ-mediated transcriptional activation stimulated by the PPARβ/δ-specific agonist, GW501516. Furthermore, the disappearance of cellular actin stress fibers, a hallmark of PPARβ/δ and COX-2 pathway activation, as evoked by the GW501516, was effectively reversed by CBDA. Activator protein-1 (AP-1)-driven transcriptional activity directly involved in the regulation of COX-2 was abrogated by the PPARβ/δ-specific inverse agonists (GSK0660/ST-247). Thus, it is implicated that there is positive interaction between PPARβ/δ and AP-1 in regulation of COX-2. These data support the concept that CBDA is a functional down-regulator of COX-2 through the abrogation of PPARβ/δ-related signaling, at least in part, in MDA-MB-231 cells.
Collapse
Affiliation(s)
- Masayo Hirao-Suzuki
- Laboratory of Xenobiotic Metabolism and Environmental Toxicology, Faculty of Pharmaceutical Sciences, Hiroshima International University (HIU)
| | - Shuso Takeda
- Laboratory of Xenobiotic Metabolism and Environmental Toxicology, Faculty of Pharmaceutical Sciences, Hiroshima International University (HIU)
| | - Takayuki Koga
- Department of Hygienic Chemistry, Daiichi University of Pharmacy
| | - Masufumi Takiguchi
- Laboratory of Xenobiotic Metabolism and Environmental Toxicology, Faculty of Pharmaceutical Sciences, Hiroshima International University (HIU)
| | - Akihisa Toda
- Department of Hygienic Chemistry, Daiichi University of Pharmacy
| |
Collapse
|
9
|
Augimeri G, Giordano C, Gelsomino L, Plastina P, Barone I, Catalano S, Andò S, Bonofiglio D. The Role of PPARγ Ligands in Breast Cancer: From Basic Research to Clinical Studies. Cancers (Basel) 2020; 12:cancers12092623. [PMID: 32937951 PMCID: PMC7564201 DOI: 10.3390/cancers12092623] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/11/2020] [Accepted: 09/12/2020] [Indexed: 02/06/2023] Open
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ), belonging to the nuclear receptor superfamily, is a ligand-dependent transcription factor involved in a variety of pathophysiological conditions such as inflammation, metabolic disorders, cardiovascular disease, and cancers. In this latter context, PPARγ is expressed in many tumors including breast cancer, and its function upon binding of ligands has been linked to the tumor development, progression, and metastasis. Over the last decade, much research has focused on the potential of natural agonists for PPARγ including fatty acids and prostanoids that act as weak ligands compared to the strong and synthetic PPARγ agonists such as thiazolidinedione drugs. Both natural and synthetic compounds have been implicated in the negative regulation of breast cancer growth and progression. The aim of the present review is to summarize the role of PPARγ activation in breast cancer focusing on the underlying cellular and molecular mechanisms involved in the regulation of cell proliferation, cell cycle, and cell death, in the modulation of motility and invasion as well as in the cross-talk with other different signaling pathways. Besides, we also provide an overview of the in vivo breast cancer models and clinical studies. The therapeutic effects of natural and synthetic PPARγ ligands, as antineoplastic agents, represent a fascinating and clinically a potential translatable area of research with regards to the battle against cancer.
Collapse
Affiliation(s)
- Giuseppina Augimeri
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (C.G.); (L.G.); (P.P.); (I.B.); (S.C.); (S.A.)
| | - Cinzia Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (C.G.); (L.G.); (P.P.); (I.B.); (S.C.); (S.A.)
- Centro Sanitario, University of Calabria, 87036 Arcavacata di Rende (CS), Italy
| | - Luca Gelsomino
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (C.G.); (L.G.); (P.P.); (I.B.); (S.C.); (S.A.)
| | - Pierluigi Plastina
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (C.G.); (L.G.); (P.P.); (I.B.); (S.C.); (S.A.)
| | - Ines Barone
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (C.G.); (L.G.); (P.P.); (I.B.); (S.C.); (S.A.)
- Centro Sanitario, University of Calabria, 87036 Arcavacata di Rende (CS), Italy
| | - Stefania Catalano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (C.G.); (L.G.); (P.P.); (I.B.); (S.C.); (S.A.)
- Centro Sanitario, University of Calabria, 87036 Arcavacata di Rende (CS), Italy
| | - Sebastiano Andò
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (C.G.); (L.G.); (P.P.); (I.B.); (S.C.); (S.A.)
| | - Daniela Bonofiglio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (C.G.); (L.G.); (P.P.); (I.B.); (S.C.); (S.A.)
- Centro Sanitario, University of Calabria, 87036 Arcavacata di Rende (CS), Italy
- Correspondence: ; Tel.: +39-0984-496208
| |
Collapse
|
10
|
Sex Hormone-Dependent Physiology and Diseases of Liver. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17082620. [PMID: 32290381 PMCID: PMC7216036 DOI: 10.3390/ijerph17082620] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/04/2020] [Accepted: 04/07/2020] [Indexed: 12/12/2022]
Abstract
Sexual dimorphism is associated not only with somatic and behavioral differences between men and women, but also with physiological differences reflected in organ metabolism. Genes regulated by sex hormones differ in expression in various tissues, which is especially important in the case of liver metabolism, with the liver being a target organ for sex hormones as its cells express estrogen receptors (ERs: ERα, also known as ESR1 or NR3A; ERβ; GPER (G protein-coupled ER, also known as GPR 30)) and the androgen receptor (AR) in both men and women. Differences in sex hormone levels and sex hormone-specific gene expression are mentioned as some of the main variations in causes of the incidence of hepatic diseases; for example, hepatocellular carcinoma (HCC) is more common in men, while women have an increased risk of autoimmune liver disease and show more acute liver failure symptoms in alcoholic liver disease. In non-alcoholic fatty liver disease (NAFLD), the distinction is less pronounced, but increased incidences are suggested among men and postmenopausal women, probably due to an increased tendency towards visceral fat accumulation.
Collapse
|
11
|
Sun SX, Zhang YN, Lu DL, Wang WL, Limbu SM, Chen LQ, Zhang ML, Du ZY. Concentration-dependent effects of 17β-estradiol and bisphenol A on lipid deposition, inflammation and antioxidant response in male zebrafish (Danio rerio). CHEMOSPHERE 2019; 237:124422. [PMID: 31352104 DOI: 10.1016/j.chemosphere.2019.124422] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 07/18/2019] [Accepted: 07/20/2019] [Indexed: 06/10/2023]
Abstract
Environmental estrogenic compounds are important pollutants, which are widely distributed in natural water bodies. They produce various adverse effects on fish, but their concentration-dependent toxicities in fish metabolism and health are not fully understood. This study investigated the effects of 17β-estradiol (E2) and bisphenol A (BPA) at low and high concentrations on lipid deposition, inflammation and antioxidant response in male zebrafish. We measured fish growth parameters, gonad development, lipid contents and the activities of inflammatory and antioxidant enzymes, as well as their mRNA expressions. All E2 and BPA concentrations used increased body weight, damaged gonad structure and induced feminization in male zebrafish. The exposure of zebrafish to E2 and BPA promoted lipid accumulation by increasing total fat, liver triglycerides and free fatty acid contents, and also upregulated lipogenic genes expression, although they decreased total cholesterol content. Notably, zebrafish exposed to low concentrations of E2 (200 ng/L) and BPA (100 μg/L) had higher lipid synthesis and deposition compared to high concentrations (2000 ng/L and 2000 μg/L, respectively). However, the high concentrations of E2 and BPA increased inflammation and antioxidant response. Furthermore, BPA caused greater damage to fish gonad development and more severe lipid peroxidation compared to E2. Overall, the results suggest that the toxic effects of E2 and BPA on zebrafish are concentration-dependent such that, the relative low concentrations used induced lipid deposition, whereas the high ones caused adverse effects on inflammation and antioxidant response.
Collapse
Affiliation(s)
- Sheng-Xiang Sun
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yun-Ni Zhang
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Dong-Liang Lu
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Wei-Li Wang
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Samwel Mchele Limbu
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, 200241, China; Department of Aquatic Sciences and Fisheries Technology, University of Dar as Salaam, Dar es Salaam, Tanzania
| | - Li-Qiao Chen
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Mei-Ling Zhang
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Zhen-Yu Du
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| |
Collapse
|
12
|
Dong T, Zhang Y, Jia S, Shang H, Fang W, Chen D, Fang M. Human Indoor Exposome of Chemicals in Dust and Risk Prioritization Using EPA's ToxCast Database. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2019; 53:7045-7054. [PMID: 31081622 DOI: 10.1021/acs.est.9b00280] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Humans spend most of their time indoors and thus have long-term exposure to chemicals. Dust is a sink for most indoor chemicals, and its ingestion is an important pathway for chemical uptake. Therefore, the chemical atlas from dust is an ideal environmental sample to investigate the indoor exposome and associated risk. In this study, we aimed to establish an indoor exposome database through comprehensive data mining on the occurrence of identified compounds in dust, and we prioritize chemicals of health concern. Through an extensive literature review (2849 articles), 355 chemicals and their concentrations were documented and analyzed for human exposure. Together with 81 compounds without concentration and 75 volatile organic compounds, we have established an indoor exposome database with 511 chemicals. Sixteen toxicological end points were selected for toxicity prioritization. Toxic equivalency factor (TEF)-based toxicity, calculated from EPA's ToxCast database, revealed a comprehensive atlas of the chemicals that had a primary contribution. Many of the prioritized compounds are currently neglected or are not actively studied. Overall, this investigation provides one of the most comprehensive analyses on chemical occurrence in indoor dust and prioritizes their chemical toxicity. Our findings can be used as a database for future exposome studies of the indoor environment and provide guidance for indoor risk assessments.
Collapse
Affiliation(s)
- Ting Dong
- School of Environment , Jinan University , Guangdong Guangzhou , P. R. China 511443
- School of Civil and Environmental Engineering , Nanyang Technological University , Singapore , Singapore 639798
| | - Yingdan Zhang
- School of Civil and Environmental Engineering , Nanyang Technological University , Singapore , Singapore 639798
| | - Shenglan Jia
- Residues and Resource Reclamation Centre, Nanyang Environment & Water Research Institute , Nanyang Technological University , Singapore , Singapore 637141
| | - Hongtao Shang
- Residues and Resource Reclamation Centre, Nanyang Environment & Water Research Institute , Nanyang Technological University , Singapore , Singapore 637141
| | - Wenjuan Fang
- Residues and Resource Reclamation Centre, Nanyang Environment & Water Research Institute , Nanyang Technological University , Singapore , Singapore 637141
| | - Da Chen
- School of Environment , Jinan University , Guangdong Guangzhou , P. R. China 511443
| | - Mingliang Fang
- School of Civil and Environmental Engineering , Nanyang Technological University , Singapore , Singapore 639798
- Residues and Resource Reclamation Centre, Nanyang Environment & Water Research Institute , Nanyang Technological University , Singapore , Singapore 637141
- Singapore Phenome Center, Lee Kong Chian School of Medicine , Nanyang Technological University , Singapore , Singapore 636921
| |
Collapse
|
13
|
Cannon RE, Trexler AW, Knudsen GA, Evans RA, Birnbaum LS. Tetrabromobisphenol A (TBBPA) Alters ABC Transport at the Blood-Brain Barrier. Toxicol Sci 2019; 169:475-484. [PMID: 30830211 PMCID: PMC6542337 DOI: 10.1093/toxsci/kfz059] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Tetrabromobisphenol A (TBBPA, CAS No. 79-94-7) is a brominated flame retardant used in 90% of epoxy coated circuit boards. Exposures to TBBPA can induce neurotoxicity and disrupt MAPK, estrogen, thyroid, and PPAR-associated signaling pathways. Because these pathways also regulate transporters of the central nervous system barriers, we sought to determine the effect of TBBPA on the expression and activity of 3 ATP binding cassette (ABC) transporters of the blood-brain barrier (BBB). Using a confocal based assay, we measured the ex vivo and in vivo effects of TBBPA on P-glycoprotein (P-gp), breast cancer resistant protein (BCRP), and multidrug resistance-associated protein 2 (MRP2) transport activity in rat brain capillaries. Our rationale for using a rat model was based on tissue availability, ease of handling, and availability of historical TBBPA toxicokinetic data. We found that TBBPA (1-1000 nM) exposure had no significant effect on multidrug resistance-associated protein 2 transport activity in either sex, suggesting TBBPA does not compromise the physical integrity of the BBB. However, low concentrations of TBBPA (1-100 nM) significantly decreased breast cancer resistant protein transport activity in both sexes. Additionally, TBBPA exposures (1-100 nM), elicited a sex-dependent response in P-gp transport: increasing transport activity in males and decreasing transport activity in females. All TBBPA dependent changes in transport activity were dose- and time-dependent. Inhibitors of either transcription or translation abolished the TBBPA dependent increases in male P-gp transport activity. Western blot and immunofluorescent assays confirmed the TBBPA dependent P-gp increases expression in males and decreases in females. Antagonizing PPAR-γ abolished the TBBPA dependent increases in males but not the decreases in females. However, the decreases in female P-gp transport were blocked by an ER-α antagonist. This work indicates that environmentally relevant concentrations of TBBPA (1-100 nM) alter ABC transporter function at the BBB. Moreover, permeability changes in the BBB can alter brain homeostasis, hinder central nervous system drug delivery, and increase the brain's exposure to harmful xenobiotic toxicants.
Collapse
Affiliation(s)
- Ronald E Cannon
- Laboratory of Toxicology and Toxicokinetics, National Cancer Institute, National Institutes of Health, Research Triangle Park, North Carolina 27709
| | - Andrew W Trexler
- Laboratory of Toxicology and Toxicokinetics, National Cancer Institute, National Institutes of Health, Research Triangle Park, North Carolina 27709
| | - Gabriel A Knudsen
- Laboratory of Toxicology and Toxicokinetics, National Cancer Institute, National Institutes of Health, Research Triangle Park, North Carolina 27709
| | - Rebecca A Evans
- University of North Carolina School of Medicine, Chapel Hill, North Carolina 27516
| | - Linda S Birnbaum
- Laboratory of Toxicology and Toxicokinetics, National Cancer Institute, National Institutes of Health, Research Triangle Park, North Carolina 27709
| |
Collapse
|
14
|
Broekema M, Savage D, Monajemi H, Kalkhoven E. Gene-gene and gene-environment interactions in lipodystrophy: Lessons learned from natural PPARγ mutants. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:715-732. [PMID: 30742913 DOI: 10.1016/j.bbalip.2019.02.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 01/13/2019] [Accepted: 02/02/2019] [Indexed: 12/13/2022]
|
15
|
Eldahshan W, Ishrat T, Pillai B, Sayed MA, Alwhaibi A, Fouda AY, Ergul A, Fagan SC. Angiotensin II type 2 receptor stimulation with compound 21 improves neurological function after stroke in female rats: a pilot study. Am J Physiol Heart Circ Physiol 2019; 316:H1192-H1201. [PMID: 30822121 PMCID: PMC6580399 DOI: 10.1152/ajpheart.00446.2018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 01/18/2019] [Accepted: 02/26/2019] [Indexed: 12/22/2022]
Abstract
The angiotensin II type 2 receptor (AT2R) agonist, compound 21 (C21), has been shown to be neurovascularly protective after ischemic stroke in male rats. In the current study, we aim to study the impact of C21 treatment on female rats. Young female Wistar rats were subjected to different durations of middle cerebral artery occlusion (MCAO) (3 h, 2 h, and 1 h) using a silicone-coated monofilament, treated at reperfusion with 0.03 mg/kg ip of C21 and followed up for different times (1, 3, and 14 days) after stroke. Behavioral tests were performed (Bederson, paw grasp, beam walk, and rotarod), and animals were euthanized for infarct size analysis and Western blot analysis. In vitro, primary male and female brain microvascular endothelial cells (ECs) were grown in culture, and the expression of the AT2R was compared between males and females. At 1 day, C21 treatment resulted in an improvement in Bederson scores. However, at 3 days and 14 days, the impact of C21 on stroke outcomes was less robust. In vitro, the expression of the AT2R was significantly higher in female ECs compared with male ECs. In conclusion, C21 improves Bederson scores after stroke in female rats when administered early at reperfusion. The ability of C21 to exert its neuroprotective effects might be affected by fluctuating levels of female hormones. NEW & NOTEWORTHY The present study shows the neuroprotective impact of C21 on ischemic stroke in female rats and how the protective effects of C21 can be influenced by the hormonal status of female rodents.
Collapse
MESH Headings
- Animals
- Behavior, Animal/drug effects
- Brain/blood supply
- Brain/drug effects
- Brain/physiopathology
- Cells, Cultured
- Disease Models, Animal
- Endothelial Cells/drug effects
- Endothelial Cells/metabolism
- Female
- Infarction, Middle Cerebral Artery/diagnosis
- Infarction, Middle Cerebral Artery/drug therapy
- Infarction, Middle Cerebral Artery/physiopathology
- Infarction, Middle Cerebral Artery/psychology
- Male
- Microvessels/drug effects
- Microvessels/metabolism
- Motor Activity/drug effects
- Neuroprotective Agents/pharmacology
- PPAR gamma/agonists
- PPAR gamma/metabolism
- Pilot Projects
- Rats, Wistar
- Receptor, Angiotensin, Type 2/agonists
- Receptor, Angiotensin, Type 2/genetics
- Receptor, Angiotensin, Type 2/metabolism
- Recovery of Function
- Sex Factors
- Signal Transduction
- Sulfonamides/pharmacology
- Thiophenes/pharmacology
- Time Factors
Collapse
Affiliation(s)
- Wael Eldahshan
- Program in Clinical and Experimental Therapeutics, Charlie Norwood Veterans Affairs Medical Center and University of Georgia, College of Pharmacy , Augusta, Georgia
| | - Tauheed Ishrat
- Program in Clinical and Experimental Therapeutics, Charlie Norwood Veterans Affairs Medical Center and University of Georgia, College of Pharmacy , Augusta, Georgia
| | - Bindu Pillai
- Program in Clinical and Experimental Therapeutics, Charlie Norwood Veterans Affairs Medical Center and University of Georgia, College of Pharmacy , Augusta, Georgia
| | - Mohammed A Sayed
- Program in Clinical and Experimental Therapeutics, Charlie Norwood Veterans Affairs Medical Center and University of Georgia, College of Pharmacy , Augusta, Georgia
| | - Abdulrahman Alwhaibi
- Program in Clinical and Experimental Therapeutics, Charlie Norwood Veterans Affairs Medical Center and University of Georgia, College of Pharmacy , Augusta, Georgia
| | - Abdelrahman Y Fouda
- Program in Clinical and Experimental Therapeutics, Charlie Norwood Veterans Affairs Medical Center and University of Georgia, College of Pharmacy , Augusta, Georgia
| | - Adviye Ergul
- Department of Physiology, Augusta University , Augusta, Georgia
| | - Susan C Fagan
- Program in Clinical and Experimental Therapeutics, Charlie Norwood Veterans Affairs Medical Center and University of Georgia, College of Pharmacy , Augusta, Georgia
- Department of Neurology, Augusta University , Augusta, Georgia
| |
Collapse
|
16
|
Crawford KA, Clark BW, Heiger-Bernays WJ, Karchner SI, Claus Henn BG, Griffith KN, Howes BL, Schlezinger DR, Hahn ME, Nacci DE, Schlezinger JJ. Altered lipid homeostasis in a PCB-resistant Atlantic killifish (Fundulus heteroclitus) population from New Bedford Harbor, MA, U.S.A. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2019; 210:30-43. [PMID: 30822701 PMCID: PMC6544361 DOI: 10.1016/j.aquatox.2019.02.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 02/13/2019] [Accepted: 02/16/2019] [Indexed: 05/09/2023]
Abstract
Sentinel species such as the Atlantic killifish (Fundulus heteroclitus) living in urban waterways can be used as toxicological models to understand impacts of environmental metabolism disrupting compound (MDC) exposure on both wildlife and humans. Exposure to MDCs is associated with increased risk of metabolic syndrome, including impaired lipid and glucose homeostasis, adipogenesis, appetite control, and basal metabolism. MDCs are ubiquitous in the environment, including in aquatic environments. New Bedford Harbor (NBH), Massachusetts is polluted with polychlorinated biphenyls (PCBs), and, as we show for the first time, tin (Sn). PCBs and organotins are ligands for two receptor systems known to regulate lipid homeostasis, the aryl hydrocarbon receptor (AHR) and the peroxisome proliferator-activated receptors (PPARs), respectively. In the current study, we compared lipid homeostasis in laboratory-reared killifish from NBH (F2) and a reference location (Scorton Creek, Massachusetts; F1 and F2) to evaluate how adaptation to local conditions may influence responses to MDCs. Adult killifish from each population were exposed to 3,3',4,4',5-pentachlorobiphenyl (PCB126, dioxin-like), 2,2',4,4',5,5'-hexachlorobiphenyl (PCB153, non-dioxin-like), or tributyltin (TBT, a PPARγ ligand) by a single intraperitoneal injection and analyzed after 3 days. AHR activation was assessed by measuring cyp1a mRNA expression. Lipid homeostasis was evaluated phenotypically by measuring liver triglycerides and organosomatic indices, and at the molecular level by measuring the mRNA expression of pparg and ppara and a target gene for each receptor. Acute MDC exposure did not affect phenotypic outcomes. However, overall NBH killifish had higher liver triglycerides and adiposomatic indices than SC killifish. Both season and population were significant predictors of the lipid phenotype. Acute MDC exposure altered hepatic gene expression only in male killifish from SC. PCB126 exposure induced cyp1a and pparg, whereas PCB153 exposure induced ppara. TBT exposure did not induce ppar-dependent pathways. Comparison of lipid homeostasis in two killifish populations extends our understanding of how MDCs act on fish and provides a basis to infer adaptive benefits of these differences in the wild.
Collapse
Affiliation(s)
- Kathryn A Crawford
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA; Boston University Superfund Research Program, Boston, MA, USA.
| | - Bryan W Clark
- Oak Ridge Institute for Science and Education at the Office of Research and Development, US Environmental Protection Agency, Narragansett, RI, USA
| | - Wendy J Heiger-Bernays
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA; Boston University Superfund Research Program, Boston, MA, USA
| | - Sibel I Karchner
- Boston University Superfund Research Program, Boston, MA, USA; Biology Department, Woods Hole Oceanographic Institution, Woods Hole, MA, USA
| | - Birgit G Claus Henn
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA
| | - Kevin N Griffith
- Department of Health Law, Policy and Management, Boston University School of Public Health, Boston, MA, USA
| | - Brian L Howes
- School for Marine Science and Technology, University of Massachusetts, Dartmouth, New Bedford, MA, USA
| | - David R Schlezinger
- School for Marine Science and Technology, University of Massachusetts, Dartmouth, New Bedford, MA, USA
| | - Mark E Hahn
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA; Biology Department, Woods Hole Oceanographic Institution, Woods Hole, MA, USA
| | - Diane E Nacci
- Atlantic Ecology Division, Office of Research and Development, US Environmental Protection Agency, Narragansett, RI, USA
| | - Jennifer J Schlezinger
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA; Boston University Superfund Research Program, Boston, MA, USA
| |
Collapse
|
17
|
Hirao-Suzuki M, Takeda S, Watanabe K, Takiguchi M, Aramaki H. Δ 9-Tetrahydrocannabinol upregulates fatty acid 2-hydroxylase (FA2H) via PPARα induction: A possible evidence for the cancellation of PPARβ/δ-mediated inhibition of PPARα in MDA-MB-231 cells. Arch Biochem Biophys 2018; 662:219-225. [PMID: 30553767 DOI: 10.1016/j.abb.2018.12.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 12/04/2018] [Accepted: 12/08/2018] [Indexed: 01/30/2023]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are a family of ligand-activated nuclear transcription factors, with three characterized subtypes: PPARα, PPARβ/δ, and PPARγ. The biological correlation between the two PPAR subtypes PPARα and γ and carcinogenesis is well-characterized; however, substantially less is known about the biological functions of PPARβ/δ. PPARβ/δ has been reported to repress transcription when PPARβ/δ and PPARα or PPARγ are simultaneously expressed in some cells, and MDA-MB-231 cells express functional levels of PPARβ/δ. We have previously reported that Δ9-tetrahydrocannabinol (Δ9-THC), a major cannabinoid component of the drug-type cannabis plant, can stimulate the expression of fatty acid 2-hydroxylase (FA2H) via upregulation of PPARα expression in human breast cancer MDA-MB-231 cells. Although the possibility of an inhibitory interaction between PPARα and PPARβ/δ has not been demonstrated in MDA-MB-231 cells, we reasoned if this interaction were to exist, Δ9-THC should make PPARα free to achieve FA2H induction. Here, we show that a PPARβ/δ-mediated suppression of PPARα function, but not of PPARγ, exists in MDA-MB-231 cells and Δ9-THC causes FA2H induction via mechanisms underlying the cancellation of PPARβ/δ-mediated inhibition of PPARα, in addition to the upregulation of PPARα.
Collapse
Affiliation(s)
- Masayo Hirao-Suzuki
- Laboratory of Xenobiotic Metabolism and Environmental Toxicology, Faculty of Pharmaceutical Sciences, Hiroshima International University (HIU), 5-1-1 Hiro-koshingai, Kure, Hiroshima, 737-0112, Japan
| | - Shuso Takeda
- Laboratory of Xenobiotic Metabolism and Environmental Toxicology, Faculty of Pharmaceutical Sciences, Hiroshima International University (HIU), 5-1-1 Hiro-koshingai, Kure, Hiroshima, 737-0112, Japan.
| | - Kazuhito Watanabe
- Center for Supporting Pharmaceutical Education, Daiichi University of Pharmacy, 22-1 Tamagawa-cho, Minami-ku, Fukuoka, 815-8511, Japan
| | - Masufumi Takiguchi
- Laboratory of Xenobiotic Metabolism and Environmental Toxicology, Faculty of Pharmaceutical Sciences, Hiroshima International University (HIU), 5-1-1 Hiro-koshingai, Kure, Hiroshima, 737-0112, Japan
| | - Hironori Aramaki
- Department of Molecular Biology, Daiichi University of Pharmacy, 22-1 Tamagawa-cho, Minami-ku, Fukuoka, 815-8511, Japan
| |
Collapse
|
18
|
De Almeida LKS, Pletschke BI, Frost CL. Moderate levels of glyphosate and its formulations vary in their cytotoxicity and genotoxicity in a whole blood model and in human cell lines with different estrogen receptor status. 3 Biotech 2018; 8:438. [PMID: 30306007 DOI: 10.1007/s13205-018-1464-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 09/26/2018] [Indexed: 12/19/2022] Open
Abstract
In vitro studies were conducted to determine the short-term cytotoxic and genotoxic effects of pure glyphosate and two glyphosate formulations (Roundup® and Wipeout®) at concentrations relevant to human exposure using whole blood (cytotoxicity) and various cancer cell lines (cytotoxicity and genotoxicity). Pure glyphosate (pure glyph) and Roundup® (Ro) showed similar non-monotonic toxicological profiles at low dose exposure (from 10 µg/ml), whereas Wipeout® (Wo) demonstrated a monotonic reduction in cell viability from a threshold concentration of 50 µg/ml, when tested in whole blood. We evaluated whether using various cancer cells (the estrogen-E2-responsive HEC1A, MCF7 and the estrogen-insensitive MDA-MB-231) exposed to moderate doses (75-500 µg/ml) would indicate varied toxicity and results indicated significant effects in the HEC1A cancer cells. A non-monotonic reduction in cell viability was observed in HEC1A exposed to pure glyph (75-500 µg/ml) and proliferative effects were observed after exposure to Wo (75, 125 and 250 µg/ml). Genotoxicity assessment (test concentration 500 µg/ml) demonstrated DNA damage in the HEC1A and MDA-MB-231 cells. Adjuvants and/or glyphosate impurities were potential contributing factors of toxicity based on the differential toxicities displayed by Ro and Wo in human whole blood and the HEC1A cells. This study contributes to the existing knowledge about in vitro exposure to moderate concentrations of glyphosate or glyphosate formulations at cytotoxic and genotoxic levels. In addition, a suggestion on the relevance of the estrogen receptor status of the cell lines used is provided, leading to the need to further investigate a potential endocrine disruptive role.
Collapse
Affiliation(s)
- L K S De Almeida
- 1Department of Biochemistry and Microbiology, Rhodes University, P.O. Box 94, Grahamstown, 6140 South Africa
| | - B I Pletschke
- 1Department of Biochemistry and Microbiology, Rhodes University, P.O. Box 94, Grahamstown, 6140 South Africa
| | - C L Frost
- 2Department of Biochemistry and Microbiology, Nelson Mandela University, P.O. Box 77000, Port Elizabeth, 6031 South Africa
| |
Collapse
|
19
|
Liu S, Sun Q. Sex differences, endogenous sex-hormone hormones, sex-hormone binding globulin, and exogenous disruptors in diabetes and related metabolic outcomes. J Diabetes 2018; 10:428-441. [PMID: 27990781 DOI: 10.1111/1753-0407.12517] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 12/13/2016] [Indexed: 12/26/2022] Open
Abstract
In assessing clinical and pathophysiological development of type 2 diabetes (T2D), the critical role of the sex steroids axis is underappreciated, particularly concerning the sex-specific relationships with many relevant cardiometabolic outcomes. In this issue of the Journal of Diabetes, we provide a comprehensive overview of these significant associations of germline variants in the genes governing the sex steroid pathways, plasma levels of steroid hormones, and sex hormone-binding globulin (SHBG) with T2D risk that have been observed in many clinical and high-quality large prospective cohorts of men and women across ethnic populations. Together, this body of evidence indicates that sex steroids and SHBG should be routinely incorporated into clinical characterization of T2D patients, particularly in screening prediabetic patients, such as those with metabolic syndrome, using plasma levels of SHBG. Given that several germline mutations in the SHBG gene have also been directly related to both plasma concentrations of SHBG and clinical manifestation of T2D, targeting signals in the sex steroid axis, particularly SHBG, may have significant utility in the prediction and treatment of T2D. Further, many of the environmental endocrine disrupting chemicals may exert their potential adverse effects on cardiometabolic outcomes via either estrogenic or androgenic signaling pathways, highlighting the importance of using the sex steroids and SHBG as important biochemical markers in both clinical and population studies in studying sex-specific mechanisms in the pathogenesis of T2D and its complications, as well as the need to equitably allocate resources in studying both men and women.
Collapse
Affiliation(s)
- Simin Liu
- Department of Endocrinology, Guangdong General Hospital/Guangdong Academy of Medical Sciences, Guangzhou, China
- Departments of Epidemiology, Brown University, Providence, Rhode Island, USA
- Departments of Medicine, Brown University, Providence, Rhode Island, USA
- Center for Global Cardiometabolic Health, Brown University, Providence, Rhode Island, USA
- Departments of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Qi Sun
- Departments of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
20
|
Bisphenol A and its derivatives decrease expression of chemerin, which reverses its stimulatory action in ovarian cancer cells. Toxicol Lett 2018; 291:61-69. [PMID: 29653259 DOI: 10.1016/j.toxlet.2018.04.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 03/09/2018] [Accepted: 04/09/2018] [Indexed: 12/14/2022]
Abstract
Chemerin is an adipocyte-secreted protein that associates with obesity, inflammation, metabolic dysfunction, and carcinogenesis. Previous studies have shown human granulosa cells to produce bioactive chemerin and its receptor CMKLR1. In the present study, we demonstrated that the mRNA level of chemerin receptor is higher in a granulosa cell tumor cell line than in epithelial cancer cells, whereas chemerin expression and secretion were lower. Various exogenous factors, such as bisphenol A and its halogenated derivatives tetrabromobisphenol A and tetrachlorobisphenol A, can affect adipokine expression. For this reason, we investigated the effects of bisphenol A and its derivatives on the expression of chemerin and its receptor. At low nanomolar concentrations, BPA, TBBPA, and TCBPA decreased chemerin expression and secretion only in granulosa cell tumor COV434 cells by both peroxisome proliferator-activated receptor γ and estrogen receptor signaling pathways. Chemerin treatment had no effect on proliferation of ovarian non-cancer and cancer cell lines. However, we also found evidence to support the inhibition of BPA- and TBBPA-induced cell proliferation by chemerin. Taken together, our results indicate for the first time that BPA and its derivatives down-regulate chemerin expression, which can suppress the ability of BPA to induce proliferation. Moreover, both PPARγ and ERs were involved in the BPA-induced decrease in chemerin expression, and its ratio was crucial to exert these effects.
Collapse
|
21
|
A high throughput metabolomics method and its application in female serum samples in a normal menstrual cycle based on liquid chromatography-mass spectrometry. Talanta 2018; 185:483-490. [PMID: 29759231 DOI: 10.1016/j.talanta.2018.03.087] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 03/20/2018] [Accepted: 03/25/2018] [Indexed: 12/21/2022]
Abstract
Periodical changes of steroid hormones have a great impact on the homeostasis of the female. However, there are few studies concerning the metabolome changes during the cycle. To study the periodic metabolic changes, a female cohort was enrolled with time-series serum samples collected during a menstrual cycle. To meet the requirement of the large-scale sample analysis, a high throughput metabolomics method was established by using an efficient sample preparation on a 96 well filter plate and a rapid LC condition in 12 min, which reduces about 70% of the samples preprocessing time and 60% analysis time. Evaluation of metabolite coverage and separation performances reflected that the method was robust for the large-scale metabolomics study. Using this method, we found that 12.6% of total detected ions including lipids, amino acids, citric acid, and so on were significantly changed during a menstrual cycle. Some metabolites were found periodically changed, which is similar to hormones (estrone and progesterone) during the cycle. These results show the novel high throughput method can be applied in large-scale metabolomics studies.
Collapse
|
22
|
Jang YJ, Son HJ, Ahn J, Jung CH, Ha T. Coumestrol modulates Akt and Wnt/β-catenin signaling during the attenuation of adipogenesis. Food Funct 2018; 7:4984-4991. [PMID: 27868125 DOI: 10.1039/c6fo01127f] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Coumestrol is a natural phytochemical present in plants such as red clover and soy, and has been reported to stimulate the estrogen receptor as a major phytoestrogen. While the molecular mechanisms responsible for the anti-adipogenic effects of phytoestrogens such as genistein and daidzein have been previously investigated, the effects of coumestrol on adipogenesis remain to be elucidated. We observed that coumestrol dose-dependently attenuates MDI (mixture of 3-isobutyl-1-methylxanthine, dexamethasone, and insulin)-induced lipid accumulation, consistent with an earlier study, while significantly inhibiting MDI-induced adipogenesis in the first 48 hours of differentiation, a critical time window for anti-adipogenic effects. Coumestrol treatment suppressed MDI-induced protein expression of PPARγ and C/EBPα in adipocytes, leading to the subsequent downregulation of FAS and aP2 expression. Akt and GSK3β were phosphorylated shortly after MDI stimulation, and these responses were inhibited by coumestrol treatment. Coumestrol also increased LRP6 protein expression, resulting in the recovery of β-catenin downregulation by MDI, while attenuating MDI-induced downregulation of Wnt10b. In addition, mRNA and protein expression of c-Myc and cyclin D1, target genes of β-catenin, were both recovered by coumestrol treatment. These results suggest that coumestrol inhibits adipocyte differentiation via regulation of Akt and Wnt/β-catenin signaling and may have potential for development as an agent to prevent adipogenesis.
Collapse
Affiliation(s)
- Young Jin Jang
- Metabolic Mechanism Research Group, Korea Food Research Institute, Seongnam, Republic of Korea.
| | - Hyo Jeong Son
- Metabolic Mechanism Research Group, Korea Food Research Institute, Seongnam, Republic of Korea.
| | - Jiyun Ahn
- Metabolic Mechanism Research Group, Korea Food Research Institute, Seongnam, Republic of Korea. and Division of Food Biotechnology, University of Science and Technology, Daejeon, Republic of Korea
| | - Chang Hwa Jung
- Metabolic Mechanism Research Group, Korea Food Research Institute, Seongnam, Republic of Korea. and Division of Food Biotechnology, University of Science and Technology, Daejeon, Republic of Korea
| | - Taeyoul Ha
- Metabolic Mechanism Research Group, Korea Food Research Institute, Seongnam, Republic of Korea. and Division of Food Biotechnology, University of Science and Technology, Daejeon, Republic of Korea
| |
Collapse
|
23
|
Shen M, Cao J, Shi H. Effects of Estrogen and Estrogen Receptors on Transcriptomes of HepG2 Cells: A Preliminary Study Using RNA Sequencing. Int J Endocrinol 2018; 2018:5789127. [PMID: 30510575 PMCID: PMC6230429 DOI: 10.1155/2018/5789127] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 08/12/2018] [Indexed: 12/23/2022] Open
Abstract
Men have a much higher incidence of hepatocellular carcinoma (HCC), the predominant form of liver cancer, than women, suggesting that estrogens play a protective role in liver cancer development and progression. To begin to understand the potential mechanisms of estrogens' inhibitory effects on HCC development, RNA sequencing was used to generate comprehensive global transcriptome profiles of the human HCC-derived HepG2 cell line following treatment of vehicle (control), estradiol (E2), estrogen receptor alpha- (ERα-) specific agonist 1,3,5-tris(4-hydroxyphenyl)-4-propyl-1H-pyrazole (PPT), or ERβ-specific agonist 2,3-bis(4-hydroxyphenyl)-propionitrile (DPN) using a small set of cells. Gene ontology (GO) analysis identified increased expression of genes involved in the biological process (BP) of response to different stimuli and metabolic processes by E2 and ER agonists, which enhanced molecular function (MF) in various enzyme activities and chemical bindings. Kyoto Encyclopedia of Genes and Genomes (KEGG) functional pathway analysis indicated enhanced pathways associated with carbohydrate metabolism, complement and coagulation cascades, and HIF-1 signaling pathway by E2 and ER agonists. GO analysis also identified decreased expression of genes by E2, PPT, and DPN involved in BP related to the cell cycle and cell division, which reduced MF in activity of multiple enzymes and microtubule activity. KEGG analysis indicated that E2, PPT, and DPN suppressed pathways associated with the cell cycle; E2 and PPT suppressed pathways associated with chemical carcinogenesis and drug metabolism, and DPN suppressed DNA replication, recombination, and repair. Collectively, these differentially expressed genes across HepG2 cell transcriptome involving cellular and metabolic processes by E2 and ER agonists provided mechanistic insight into protective effects of estrogens in HCC development.
Collapse
Affiliation(s)
- Minqian Shen
- Department of Biology, Miami University, 700 E. High St., Oxford, OH, USA
| | - Jingyi Cao
- Department of Biology, Miami University, 700 E. High St., Oxford, OH, USA
| | - Haifei Shi
- Department of Biology, Miami University, 700 E. High St., Oxford, OH, USA
| |
Collapse
|
24
|
Robinson GA, Waddington KE, Pineda-Torra I, Jury EC. Transcriptional Regulation of T-Cell Lipid Metabolism: Implications for Plasma Membrane Lipid Rafts and T-Cell Function. Front Immunol 2017; 8:1636. [PMID: 29225604 PMCID: PMC5705553 DOI: 10.3389/fimmu.2017.01636] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 11/09/2017] [Indexed: 01/10/2023] Open
Abstract
It is well established that cholesterol and glycosphingolipids are enriched in the plasma membrane (PM) and form signaling platforms called lipid rafts, essential for T-cell activation and function. Moreover, changes in PM lipid composition affect the biophysical properties of lipid rafts and have a role in defining functional T-cell phenotypes. Here, we review the role of transcriptional regulators of lipid metabolism including liver X receptors α/β, peroxisome proliferator-activated receptor γ, estrogen receptors α/β (ERα/β), and sterol regulatory element-binding proteins in T-cells. These receptors lie at the interface between lipid metabolism and immune cell function and are endogenously activated by lipids and/or hormones. Importantly, they regulate cellular cholesterol, fatty acid, glycosphingolipid, and phospholipid levels but are also known to modulate a broad spectrum of immune responses. The current evidence supporting a role for lipid metabolism pathways in controlling immune cell activation by influencing PM lipid raft composition in health and disease, and the potential for targeting lipid biosynthesis pathways to control unwanted T-cell activation in autoimmunity is reviewed.
Collapse
Affiliation(s)
- George A. Robinson
- Centre of Rheumatology, Division of Medicine, University College London, London, United Kingdom
| | - Kirsty E. Waddington
- Centre of Rheumatology, Division of Medicine, University College London, London, United Kingdom
- Clinical Pharmacology, Division of Medicine, University College London, London, United Kingdom
| | - Ines Pineda-Torra
- Clinical Pharmacology, Division of Medicine, University College London, London, United Kingdom
| | - Elizabeth C. Jury
- Centre of Rheumatology, Division of Medicine, University College London, London, United Kingdom
| |
Collapse
|
25
|
Kim DH, Suh J, Surh YJ, Na HK. Regulation of the tumor suppressor PTEN by natural anticancer compounds. Ann N Y Acad Sci 2017; 1401:136-149. [PMID: 28891094 DOI: 10.1111/nyas.13422] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 05/31/2017] [Accepted: 06/05/2017] [Indexed: 12/20/2022]
Abstract
The tumor suppressor phosphatase and tensin homologue (PTEN) has phosphatase activity, with phosphatidylinositol (3,4,5)-trisphosphate (PIP3), a product of phosphatidylinositol 3-kinase (PI3K), as one of the principal substrates. PTEN is a negative regulator of the Akt pathway, which plays a fundamental role in controlling cell growth, survival, and proliferation. Loss of PTEN function has been observed in many different types of cancer. Functional inactivation of PTEN as a consequence of germ-line mutations or promoter hypermethylation predisposes individuals to malignancies. PTEN undergoes posttranslational modifications, such as oxidation, acetylation, phosphorylation, SUMOylation, and ubiquitination, which influence its catalytic activity, interactions with other proteins, and subcellular localization. Cellular redox status is crucial for posttranslational modification of PTEN and its functional consequences. Oxidative stress and inflammation are major causes of loss of PTEN function. Pharmacologic or nutritional restoration of PTEN function is considered a reliable strategy in the management of PTEN-defective cancer. In this review, we highlight natural compounds, such as curcumin, indol-3 carbinol, and omega-3 fatty acids, that have the potential to restore or potentiate PTEN expression/activity, thereby suppressing cancer cell proliferation, survival, and resistance to chemotherapeutic agents.
Collapse
Affiliation(s)
- Do-Hee Kim
- Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Jinyoung Suh
- Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Young-Joon Surh
- Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, South Korea.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science, Seoul National University, Seoul, South Korea.,Cancer Research Institute, Seoul National University, Seoul, South Korea
| | - Hye-Kyung Na
- Department of Food Science and Biotechnology, College of Knowledge-Based Services Engineering, Sungshin Women's University, Seoul, South Korea
| |
Collapse
|
26
|
|
27
|
Mediating Roles of PPARs in the Effects of Environmental Chemicals on Sex Steroids. PPAR Res 2017; 2017:3203161. [PMID: 28819354 PMCID: PMC5551527 DOI: 10.1155/2017/3203161] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/23/2017] [Accepted: 06/21/2017] [Indexed: 12/18/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are ligand-activated nuclear receptors that are widely involved in various physiological functions. They are widely expressed through the reproductive system. Their roles in the metabolism and function of sex steroids and thus the etiology of reproductive disorders receive great concern. Various kinds of exogenous chemicals, especially environmental pollutants, exert their adverse impact on the reproductive system through disturbing the PPAR signaling pathway. Chemicals could bind to PPARs and modulate the transcription of downstream genes containing PPRE (peroxisome proliferator response element). This will lead to altered expression of genes related to metabolism of sex steroids and thus the abnormal physiological function of sex steroids. In this review, various kinds of environmental ligands are summarized and discussed. Their interactions with three types of PPARs are classified by various data from transcript profiles, PPRE reporter in cell line, in silico docking, and gene silencing. The review will contribute to the understanding of the roles of PPARs in the reproductive toxicology of environmental chemicals.
Collapse
|
28
|
Kalo D, Roth Z. Low level of mono(2-ethylhexyl) phthalate reduces oocyte developmental competence in association with impaired gene expression. Toxicology 2016; 377:38-48. [PMID: 27989758 DOI: 10.1016/j.tox.2016.12.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 12/12/2016] [Accepted: 12/15/2016] [Indexed: 12/24/2022]
Abstract
Di-(2-ethylhexyl) phthalate (DEHP) and its metabolite, mono-(2-ethylhexyl) phthalate (MEHP), are reproductive toxicants. However, disruptive effects of MEHP at low concentrations on the oocyte and developing blastocyst are unknown. Previously, we detected low levels of MEHP in follicular fluid aspirated from DEHP-treated cows associated with reduced estradiol levels. Moreover, the MEHP concentrations found were similar to those reported for follicular fluid aspirated from women who have undergone IVF cycles. In the current study, we used an in vitro embryo production model to examine the effect of MEHP at low levels on oocyte developmental competence. We set up several experiments to mimic the follicular fluid content, i.e., low MEHP level and low estradiol. For all experiments, cumulus oocyte complexes (COCs) were aspirated from bovine ovaries, then matured in vitro in standard oocyte maturation medium (OMM) supplemented with: MEHP at a range levels (20-1000nM) or with estradiol at a range levels (0-2000ng/ml). Then, oocytes were fertilized and cultured for an additional 7days to allow blastocyst development. Findings revealed that MEHP at low levels impairs oocyte developmental competence in a dose-dependent manner (P<0.05) and that estradiol by itself does not impair it. Accordingly, in another set of experiments, COCs were matured in vitro with MEHP at two choosen concentrations (20 or 1000nM) with or without estradiol, fertilized and cultured for 7days. Samples of mature oocytes and their derived blastocysts were subjected to quantitative real-time PCR to examine the profiles of selected genes (CYC1, MT-CO1, ATP5B, POU5F1, SOX2 and DNMT3b). Maturation of COCs with MEHP (20 or 1000nM) affected gene expression in the mature oocyte. Maturation of COCs with MEHP (20 or 1000nM) in the absence of estradiol reduced oocyte developmental competence (P<0.05). A differential carryover effect on transcript abundance was recorded in blastocysts developed from MEHP-treated oocytes. In the presence of estradiol, increased expression was recorded for CYC1, ATP5B, SOX2 and DNMT3b. In the absence of estradiol, decreased expression was recorded, with a significant effect for 1000nM MEHP (P<0.05). Taken together, the findings suggest that low levels of phthalate must be taken into consideration in risk assessments.
Collapse
Affiliation(s)
- D Kalo
- Department of Animal Sciences, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University, Rehovot 76100, Israel; Center of Excellence in Agriculture and Environmental Health, The Hebrew University, Rehovot 76100, Israel
| | - Z Roth
- Department of Animal Sciences, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University, Rehovot 76100, Israel; Center of Excellence in Agriculture and Environmental Health, The Hebrew University, Rehovot 76100, Israel.
| |
Collapse
|
29
|
Song Z, Shi Q. The Interaction of PPARα and CYP7B1 with ERα, β Impacted the Occurrence and Development of Intrahepatic Cholestasis in Pregnant Rats. Reprod Sci 2016; 24:627-634. [PMID: 27628953 DOI: 10.1177/1933719116667223] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Intrahepatic cholestasis of pregnancy (ICP) is a disorder of bile acid (BA) synthesis, excretion, and metabolism, with systemic accumulation of BAs, which can lead to prematurity, fetal distress, and intrauterine death. Here, we investigate the expression of peroxisome proliferator-activated receptor alpha and cytochrome P450 oxysterol 7alpha-hydroxylase by exposing to 17α-ethynylestradiol with or without the estrogen receptor signaling pathway in pregnant rats with intrahepatic cholestasis. In vivo and in vitro evidences showed that estrogen receptor alpha (ERα) may be the key point of occurrence and development of intrahepatic cholestasis in pregnant rats. Besides, the abnormalities in genes could be reversed by ERα small interfering RNA. Our findings provide the ERα-centered hypothesis on the mechanisms of ICP. New perspectives are emerging for the treatment of estrogen-induced hepatic complication.
Collapse
Affiliation(s)
- Zhaoyi Song
- 1 Department of Obstetrics and Gynecology, Ninth School of Clinical Medicine, Peking University, Beijing, China
| | - Qingyun Shi
- 2 Department of Obstetrics and Gynecology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
30
|
Lopes C, Madureira TV, Ferreira N, Pinheiro I, Castro LFC, Rocha E. Peroxisome proliferator-activated receptor gamma (PPARγ) in brown trout: Interference of estrogenic and androgenic inputs in primary hepatocytes. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2016; 46:328-336. [PMID: 27541269 DOI: 10.1016/j.etap.2016.08.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 08/05/2016] [Accepted: 08/08/2016] [Indexed: 06/06/2023]
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) is a pivotal regulator of lipid and glucose metabolism in vertebrates. Here, we isolated and characterized for the first time the PPARγ gene from brown trout (Salmo trutta f. fario). Hormones have been reported to interfere with the regulatory function of PPARγ in various organisms, albeit with little focus on fish. Thus, primary hepatocytes isolated from juveniles of brown trout were exposed to 1, 10 and 50μM of ethinylestradiol (EE2) or testosterone (T). A significant (3 fold) decrease was obtained in response to 50μM of EE2 and to 10 and 50μM of T (13 and 14 folds), while a 3 fold increase was observed at 1μM of EE2. Therefore, trout PPARγ seems a target for natural/synthetic compounds with estrogenic or androgenic properties and so, we advocate considering PPARγ as another alert sensor gene when assessing the effects of sex-steroid endocrine disruptors.
Collapse
Affiliation(s)
- Célia Lopes
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), U.Porto - University of Porto, Rua dos Bragas 289, P 4050-123 Porto, Portugal; Institute of Biomedical Sciences Abel Salazar (ICBAS), U.Porto - University of Porto, Laboratory of Histology and Embryology, Department of Microscopy, Rua Jorge Viterbo Ferreira 228, P 4050-313 Porto, Portugal
| | - Tânia Vieira Madureira
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), U.Porto - University of Porto, Rua dos Bragas 289, P 4050-123 Porto, Portugal; Institute of Biomedical Sciences Abel Salazar (ICBAS), U.Porto - University of Porto, Laboratory of Histology and Embryology, Department of Microscopy, Rua Jorge Viterbo Ferreira 228, P 4050-313 Porto, Portugal.
| | - Nádia Ferreira
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), U.Porto - University of Porto, Rua dos Bragas 289, P 4050-123 Porto, Portugal; Institute of Biomedical Sciences Abel Salazar (ICBAS), U.Porto - University of Porto, Laboratory of Histology and Embryology, Department of Microscopy, Rua Jorge Viterbo Ferreira 228, P 4050-313 Porto, Portugal
| | - Ivone Pinheiro
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), U.Porto - University of Porto, Rua dos Bragas 289, P 4050-123 Porto, Portugal; Institute of Biomedical Sciences Abel Salazar (ICBAS), U.Porto - University of Porto, Laboratory of Histology and Embryology, Department of Microscopy, Rua Jorge Viterbo Ferreira 228, P 4050-313 Porto, Portugal
| | - L Filipe C Castro
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), U.Porto - University of Porto, Rua dos Bragas 289, P 4050-123 Porto, Portugal; Faculty of Sciences (FCUP), U.Porto - University of Porto, Department of Biology, Rua do Campo Alegre, P 4169-007 Porto, Portugal
| | - Eduardo Rocha
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), U.Porto - University of Porto, Rua dos Bragas 289, P 4050-123 Porto, Portugal; Institute of Biomedical Sciences Abel Salazar (ICBAS), U.Porto - University of Porto, Laboratory of Histology and Embryology, Department of Microscopy, Rua Jorge Viterbo Ferreira 228, P 4050-313 Porto, Portugal
| |
Collapse
|
31
|
Zhang W, Sun Q, Zhong W, Sun X, Zhou Z. Hepatic Peroxisome Proliferator-Activated Receptor Gamma Signaling Contributes to Alcohol-Induced Hepatic Steatosis and Inflammation in Mice. Alcohol Clin Exp Res 2016; 40:988-99. [PMID: 27062444 DOI: 10.1111/acer.13049] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 02/18/2016] [Indexed: 12/25/2022]
Abstract
BACKGROUND Peroxisome proliferator-activated receptor gamma (PPARγ) signaling has been shown to regulate lipogenesis and lipid accumulation. Previous studies have shown that hepatic PPARγ is up-regulated in steatotic liver of both animal and human. However, the effects of hepatic PPARγ signaling on alcoholic liver disease (ALD) remain elusive. METHODS To determine the role of hepatic PPARγ signaling on ALD, wild-type (WT) and hepatocyte-specific PPARγ knockdown (PPARγ∆Hep) mice were fed a modified Lieber-DeCarli alcohol or isocaloric maltose dextrin control liquid diet for 8 weeks to induce ALD. Blood parameters, hepatic steatosis, and inflammation were measured after 8-week alcohol feeding. RESULTS Alcohol feeding to WT mice resulted in liver damage (alanine aminotransferase [ALT], 94.68 ± 17.05 U/L; aspartate aminotransferase [AST], 55.87 ± 11.29 U/L), which was significantly alleviated by hepatic PPARγ knockdown (ALT, 57.36 ± 14.98 U/L; AST, 38.06 ± 3.35 U/L). Alcohol feeding led to marked lipid accumulation and up-regulation of lipogenic genes including fatty acid transport protein 1 (FATP1), acetyl-CoA carboxylase (ACC), fatty acid synthase (FASN), lipin1 (LIPIN1), diacylglycerol acyltransferase 1 (DGAT1), and diacylglycerol acyltransferase 2 (DGAT2) in the livers of WT mice. Knockdown of hepatic PPARγ significantly alleviated alcohol-induced lipid accumulation and abolished the up-regulation of FASN, DGAT1, and DGAT2. Silencing of PPARγ in FL83B cells significantly decreased ethanol (EtOH)-, linoleic acid-, and EtOH plus linoleic acid-induced lipid accumulation. Knockdown of hepatic PPARγ also significantly reduced alcohol-induced inflammatory chemokine (monocyte chemotactic protein 1 [MCP1], keratinocyte-derived chemokine [KC], interferon gamma-induced protein 10 [IP-10]) and inflammatory infiltration (lymphocyte antigen 6 complex, locus G [Ly6G], and F4/80). CONCLUSIONS The results suggest that hepatic PPARγ signaling contributes to alcohol-induced liver injury by promoting hepatic steatosis and inflammation.
Collapse
Affiliation(s)
- Wenliang Zhang
- Center for Translational Biomedical Research, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, North Carolina
| | - Qian Sun
- Center for Translational Biomedical Research, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, North Carolina.,Department of Nutrition, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, North Carolina
| | - Wei Zhong
- Center for Translational Biomedical Research, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, North Carolina
| | - Xinguo Sun
- Center for Translational Biomedical Research, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, North Carolina
| | - Zhanxiang Zhou
- Center for Translational Biomedical Research, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, North Carolina.,Department of Nutrition, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, North Carolina
| |
Collapse
|
32
|
PPARs: Protectors or Opponents of Myocardial Function? PPAR Res 2015; 2015:835985. [PMID: 26713088 PMCID: PMC4680114 DOI: 10.1155/2015/835985] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 11/05/2015] [Accepted: 11/08/2015] [Indexed: 12/15/2022] Open
Abstract
Over 5 million people in the United States suffer from the complications of heart failure (HF), which is a rapidly expanding health complication. Disorders that contribute to HF include ischemic cardiac disease, cardiomyopathies, and hypertension. Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear receptor family. There are three PPAR isoforms: PPARα, PPARγ, and PPARδ. They can be activated by endogenous ligands, such as fatty acids, as well as by pharmacologic agents. Activators of PPARs are used for treating several metabolic complications, such as diabetes and hyperlipidemia that are directly or indirectly associated with HF. However, some of these drugs have adverse effects that compromise cardiac function. This review article aims to summarize the current basic and clinical research findings of the beneficial or detrimental effects of PPAR biology on myocardial function.
Collapse
|
33
|
Madureira TV, Malhão F, Pinheiro I, Lopes C, Ferreira N, Urbatzka R, Castro LFC, Rocha E. Estrogenic and anti-estrogenic influences in cultured brown trout hepatocytes: Focus on the expression of some estrogen and peroxisomal related genes and linked phenotypic anchors. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2015; 169:133-142. [PMID: 26539803 DOI: 10.1016/j.aquatox.2015.10.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 09/29/2015] [Accepted: 10/16/2015] [Indexed: 06/05/2023]
Abstract
Estrogens, estrogenic mimics and anti-estrogenic compounds are known to target estrogen receptors (ER) that can modulate other nuclear receptor signaling pathways, such as those controlled by the peroxisome proliferator-activated receptor (PPAR), and alter organelle (inc. peroxisome) morphodynamics. By using primary isolated brown trout (Salmo trutta f. fario) hepatocytes after 72 and 96h of exposure we evaluated some effects in selected molecular targets and in peroxisomal morphological features caused by: (1) an ER agonist (ethinylestradiol-EE2) at 1, 10 and 50μM; (2) an ER antagonist (ICI 182,780) at 10 and 50μM; and (3) mixtures of both (Mix I-10μM EE2 and 50μM ICI; Mix II-1μM EE2 and 10μM ICI and Mix III-1μM EE2 and 50μM ICI). The mRNA levels of the estrogenic targets (ERα, ERβ-1 and vitellogenin A-VtgA) and the peroxisome structure/function related genes (catalase, urate oxidase-Uox, 17β-hydroxysteroid dehydrogenase 4-17β-HSD4, peroxin 11α-Pex11α and PPARα) were analyzed by real-time polymerase chain reaction (RT-PCR). Stereology combined with catalase immunofluorescence revealed a significant reduction in peroxisome volume densities at 50μM of EE2 exposure. Concomitantly, at the same concentration, electron microscopy showed smaller peroxisome profiles, exacerbated proliferation of rough endoplasmic reticulum, and a generalized cytoplasmic vacuolization of hepatocytes. Catalase and Uox mRNA levels decreased in all estrogenic stimuli conditions. VtgA and ERα mRNA increased after all EE2 treatments, while ERβ-1 had an inverse pattern. The EE2 action was reversed by ICI 182,780 in a concentration-dependent manner, for VtgA, ERα and Uox. Overall, our data show the great value of primary brown trout hepatocytes to study the effects of estrogenic/anti-estrogenic inputs in peroxisome kinetics and in ER and PPARα signaling, backing the still open hypothesis of crosstalk interactions between these pathways and calling for more mechanistic experiments.
Collapse
Affiliation(s)
- Tânia Vieira Madureira
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), U.Porto-University of Porto, Rua dos Bragas 289, P 4050-123 Porto, Portugal; Institute of Biomedical Sciences Abel Salazar, U.Porto (ICBAS)-University of Porto, Laboratory of Histology and Embryology, Department of Microscopy, Rua Jorge Viterbo Ferreira 228, P 4050-313 Porto, Portugal.
| | - Fernanda Malhão
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), U.Porto-University of Porto, Rua dos Bragas 289, P 4050-123 Porto, Portugal; Institute of Biomedical Sciences Abel Salazar, U.Porto (ICBAS)-University of Porto, Laboratory of Histology and Embryology, Department of Microscopy, Rua Jorge Viterbo Ferreira 228, P 4050-313 Porto, Portugal
| | - Ivone Pinheiro
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), U.Porto-University of Porto, Rua dos Bragas 289, P 4050-123 Porto, Portugal; Institute of Biomedical Sciences Abel Salazar, U.Porto (ICBAS)-University of Porto, Laboratory of Histology and Embryology, Department of Microscopy, Rua Jorge Viterbo Ferreira 228, P 4050-313 Porto, Portugal
| | - Célia Lopes
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), U.Porto-University of Porto, Rua dos Bragas 289, P 4050-123 Porto, Portugal; Institute of Biomedical Sciences Abel Salazar, U.Porto (ICBAS)-University of Porto, Laboratory of Histology and Embryology, Department of Microscopy, Rua Jorge Viterbo Ferreira 228, P 4050-313 Porto, Portugal
| | - Nádia Ferreira
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), U.Porto-University of Porto, Rua dos Bragas 289, P 4050-123 Porto, Portugal; Institute of Biomedical Sciences Abel Salazar, U.Porto (ICBAS)-University of Porto, Laboratory of Histology and Embryology, Department of Microscopy, Rua Jorge Viterbo Ferreira 228, P 4050-313 Porto, Portugal
| | - Ralph Urbatzka
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), U.Porto-University of Porto, Rua dos Bragas 289, P 4050-123 Porto, Portugal
| | - L Filipe C Castro
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), U.Porto-University of Porto, Rua dos Bragas 289, P 4050-123 Porto, Portugal; Faculty of Sciences (FCUP), U.Porto-University of Porto, Department of Biology, Rua do Campo Alegre, P 4169-007 Porto, Portugal
| | - Eduardo Rocha
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), U.Porto-University of Porto, Rua dos Bragas 289, P 4050-123 Porto, Portugal; Institute of Biomedical Sciences Abel Salazar, U.Porto (ICBAS)-University of Porto, Laboratory of Histology and Embryology, Department of Microscopy, Rua Jorge Viterbo Ferreira 228, P 4050-313 Porto, Portugal
| |
Collapse
|
34
|
Hultman MT, Song Y, Tollefsen KE. 17α-Ethinylestradiol (EE2) effect on global gene expression in primary rainbow trout (Oncorhynchus mykiss) hepatocytes. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2015; 169:90-104. [PMID: 26519835 DOI: 10.1016/j.aquatox.2015.10.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 10/06/2015] [Accepted: 10/06/2015] [Indexed: 06/05/2023]
Abstract
The potential impact of endocrine disrupting chemicals (EDCs) in the aquatic environment has driven the development of screening assays to evaluate the estrogenic properties of chemicals and their effects on aquatic organisms such as fish. However, obtaining full concentration-response relationships in animal (in vivo) exposure studies are laborious, costly and unethical, hence a need for developing feasible alternative (non-animal) methods. Use of in vitro bioassays such as primary fish hepatocytes, which retain many of the native properties of the liver, has been proposed for in vitro screening of estrogen receptor (ER) agonists and antagonists. The aim of present study was to characterize the molecular mode of action (MoA) of the ER agonist 17α-ethinylestradiol (EE2) in primary rainbow trout (Oncorhynchus mykiss) hepatocytes. A custom designed salmonid 60,000-feature (60k) oligonucleotide microarray was used to characterize the potential MoAs after 48h exposure to EE2. The microarray analysis revealed several concentration-dependent gene expression alterations including classical estrogen sensitive biomarker gene expression (e.g. estrogen receptor α, vitellogenin, zona radiata). Gene Ontology (GO) analysis displayed transcriptional changes suggesting interference of cellular growth, fatty acid and lipid metabolism potentially mediated through the estrogen receptor (ER), which were proposed to be associated with modulation of genes involved in endocrine function and reproduction. Pathway analysis supported the identified GOs and revealed modulation of additional genes associated with apoptosis and cholesterol biosynthesis. Differentially expressed genes (DEGs) related to impaired lipid metabolism (e.g. peroxisome proliferator-activated receptor α and γ), growth (e.g. insulin growth factor protein 1), phase I and II biotransformation (e.g. cytochrome P450 1A, sulfotransferase, UDP-glucuronosyltransferase and glutathione S-transferase) provided additional insight into the MoA of EE2 in primary fish hepatocytes. Results from the present study suggest that biotransformation, estrogen receptor-mediated responses, lipid homeostasis, growth and cancer/apoptosis in primary fish hepatocytes may be altered after short-term exposure to ER-agonists such as EE2. In many cases the observed changes were similar to those reported for estrogen-exposed fish in vivo. In conclusion, global transcriptional analysis demonstrated that EE2 affected a number of toxicologically relevant pathways associated with an estrogenic MoA in the rainbow trout hepatocytes.
Collapse
Affiliation(s)
- Maria T Hultman
- Norwegian Institute for Water Research (NIVA), Section of Ecotoxicology and Risk Assessment, Gaustadalléen 21, N-0349 Oslo, Norway; Faculty of Environmental Science & Technology, Department for Environmental Sciences, Norwegian University of Life Sciences (NMBU), Post box 5003, N-1432 Ås, Norway.
| | - You Song
- Norwegian Institute for Water Research (NIVA), Section of Ecotoxicology and Risk Assessment, Gaustadalléen 21, N-0349 Oslo, Norway
| | - Knut Erik Tollefsen
- Norwegian Institute for Water Research (NIVA), Section of Ecotoxicology and Risk Assessment, Gaustadalléen 21, N-0349 Oslo, Norway; Faculty of Environmental Science & Technology, Department for Environmental Sciences, Norwegian University of Life Sciences (NMBU), Post box 5003, N-1432 Ås, Norway
| |
Collapse
|
35
|
Dammann K, Khare V, Lang M, Claudel T, Harpain F, Granofszky N, Evstatiev R, Williams JM, Pritchard DM, Watson A, Gasche C. PAK1 modulates a PPARγ/NF-κB cascade in intestinal inflammation. BIOCHIMICA ET BIOPHYSICA ACTA 2015; 1853:2349-60. [PMID: 26036343 PMCID: PMC4576212 DOI: 10.1016/j.bbamcr.2015.05.031] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 05/13/2015] [Accepted: 05/28/2015] [Indexed: 02/07/2023]
Abstract
P21-activated kinases (PAKs) are multifunctional effectors of Rho GTPases with both kinase and scaffolding activity. Here, we investigated the effects of inflammation on PAK1 signaling and its role in colitis-driven carcinogenesis. PAK1 and p-PAK1 (Thr423) were assessed by immunohistochemistry, immunofluorescence, and Western blot. C57BL6/J wildtype mice were treated with a single intraperitoneal TNFα injection. Small intestinal organoids from these mice and from PAK1-KO mice were cultured with TNFα. NF-κB and PPARγ were analyzed upon PAK1 overexpression and silencing for transcriptional/translational regulation. PAK1 expression and activation was increased on the luminal intestinal epithelial surface in inflammatory bowel disease and colitis-associated cancer. PAK1 was phosphorylated upon treatment with IFNγ, IL-1β, and TNFα. In vivo, mice administered with TNFα showed increased p-PAK1 in intestinal villi, which was associated with nuclear p65 and NF-κB activation. p65 nuclear translocation downstream of TNFα was strongly inhibited in PAK1-KO small intestinal organoids. PAK1 overexpression induced a PAK1-p65 interaction as visualized by co-immunoprecipitation, nuclear translocation, and increased NF-κB transactivation, all of which were impeded by kinase-dead PAK1. Moreover, PAK1 overexpression downregulated PPARγ and mesalamine recovered PPARγ through PAK1 inhibition. On the other hand PAK1 silencing inhibited NF-κB, which was recovered using BADGE, a PPARγ antagonist. Altogether these data demonstrate that PAK1 overexpression and activation in inflammation and colitis-associated cancer promote NF-κB activity via suppression of PPARγ in intestinal epithelial cells.
Collapse
Affiliation(s)
- Kyle Dammann
- Medical University of Vienna, Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Christian Doppler Laboratory for Molecular Cancer Chemoprevention, Vienna, Austria
| | - Vineeta Khare
- Medical University of Vienna, Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Christian Doppler Laboratory for Molecular Cancer Chemoprevention, Vienna, Austria
| | - Michaela Lang
- Medical University of Vienna, Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Christian Doppler Laboratory for Molecular Cancer Chemoprevention, Vienna, Austria
| | - Thierry Claudel
- Medical University of Vienna, Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Hans Popper Laboratory for Molecular Hepatology, Vienna, Austria
| | - Felix Harpain
- Medical University of Vienna, Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Christian Doppler Laboratory for Molecular Cancer Chemoprevention, Vienna, Austria
| | - Nicolas Granofszky
- Medical University of Vienna, Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Christian Doppler Laboratory for Molecular Cancer Chemoprevention, Vienna, Austria
| | - Rayko Evstatiev
- Medical University of Vienna, Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Christian Doppler Laboratory for Molecular Cancer Chemoprevention, Vienna, Austria
| | - Jonathan M Williams
- Department of Gastroenterology, University of Liverpool, Liverpool, United Kingdom
| | - D Mark Pritchard
- Department of Gastroenterology, University of Liverpool, Liverpool, United Kingdom
| | - Alastair Watson
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, United Kingdom
| | - Christoph Gasche
- Medical University of Vienna, Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Christian Doppler Laboratory for Molecular Cancer Chemoprevention, Vienna, Austria.
| |
Collapse
|
36
|
Zanella I, Marrazzo E, Biasiotto G, Penza M, Romani A, Vignolini P, Caimi L, Di Lorenzo D. Soy and the soy isoflavone genistein promote adipose tissue development in male mice on a low-fat diet. Eur J Nutr 2015; 54:1095-107. [PMID: 25341395 DOI: 10.1007/s00394-014-0786-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 10/13/2014] [Indexed: 01/11/2023]
Abstract
PURPOSE Several nutrients act as phytoestrogens, being anti-adipogenic when consumed with a fat-rich diet. Their effect on a low-fat diet (LFD) background is unknown. We tested soy and genistein effects on adipose tissue in LFD-fed mice and genistein activity in the 3T3-L1 adipogenesis model. METHODS C57BL/6 J male mice were fed an 8.5% soy-supplemented LFD (SS-LFD) or a soy-free LFD (SF-LFD) for 147 days. Groups of 3-week-old (pubertal) and 6-week-old (adult) mice on the SF-LFD were also treated with 17ß-estradiol (E2, 5 µg/kg/day) ip or pure genistein (5 mg/kg/day) by gavage for 15 days. Body fat deposition and gene expression profiles were evaluated. E2 and genistein effects on ERα, ERβ and PPARγ transcriptional activities were characterized in ERα- or ERβ-transfected 3T3L1 cells during differentiation, by the use of reporter plasmids. RESULTS The SS-LFD group increased fat mass compared with the SF-LFD group. Genistein alone increased while E2 decreased fat pads in the 15-day-treated mice. In visceral fat, genistein differentially regulated 13 metabolic pathways compared to E2. PPARγ-controlled genes were downregulated by E2, while they were upregulated by genistein. In 3T3-L1 cells, genistein activated ERβ-driven transcription, differentiation and lipid accumulation, while inhibited ERα-driven transcription, without effects on lipid accumulation. E2 activated both ERs only in preadipocytes. In differentiated untransfected cells, genistein inhibited PPARγ, while activated PPARγ in the presence of ERβ. CONCLUSIONS Soy and genistein at nutritional doses induce fat development in LFD-fed mice and adipogenesis in 3T3-L1 cells, with a mechanism that involves, at least in vitro, ERβ and is dependent on cell differentiation stage.
Collapse
Affiliation(s)
- Isabella Zanella
- Biotechnology/3rd Laboratory and Department of Diagnostics, Civic Hospital of Brescia, 25123, Brescia, Italy
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Eleonora Marrazzo
- Biotechnology/3rd Laboratory and Department of Diagnostics, Civic Hospital of Brescia, 25123, Brescia, Italy
| | - Giorgio Biasiotto
- Biotechnology/3rd Laboratory and Department of Diagnostics, Civic Hospital of Brescia, 25123, Brescia, Italy
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Marialetizia Penza
- Biotechnology/3rd Laboratory and Department of Diagnostics, Civic Hospital of Brescia, 25123, Brescia, Italy
| | - Annalisa Romani
- Pharmaceutical Sciences Department, Università di Firenze, Florence, Italy
| | - Pamela Vignolini
- Pharmaceutical Sciences Department, Università di Firenze, Florence, Italy
| | - Luigi Caimi
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Diego Di Lorenzo
- Biotechnology/3rd Laboratory and Department of Diagnostics, Civic Hospital of Brescia, 25123, Brescia, Italy.
| |
Collapse
|
37
|
Hu Y, Song Y, Franke AA, Hu FB, van Dam RM, Sun Q. A Prospective Investigation of the Association Between Urinary Excretion of Dietary Lignan Metabolites and Weight Change in US Women. Am J Epidemiol 2015; 182:503-11. [PMID: 26290574 DOI: 10.1093/aje/kwv091] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Accepted: 03/31/2015] [Indexed: 11/13/2022] Open
Abstract
Results from animal studies have consistently suggested that lignans play a role in the regulation of in body weight, but evidence from human studies has been limited. We examined the associations between urinary excretion of enterolactone and enterodiol, the major intestinal microbial metabolites of dietary lignans, and 10-year prospective weight change using data from 2 well-characterized cohort studies of US women: the Nurses' Health Study (2000-2010) and Nurses' Health Study II (1997-2007). Urinary excretion levels of enterolactone and enterodiol were measured at baseline. Associations with prospective weight change were analyzed using a multivariable-adjusted linear mixed-effects model. We observed that women in the highest quartile of urinary excretion of total lignans had significantly lower baseline body mass indices (weight in kilograms divided by height in meters squared) (mean, 24.6, 95% confidence interval (CI): 23.9, 25.2) than did those in the lowest quartile (mean, 27.7, 95% CI: 27.0, 28.4; P for trend < 0.01). Compared with women in the lowest quartile of enterodiol excretion, those in the highest quartile gained 0.27 kg/year less weight (95% CI: 0.12, 0.41; P for trend < 0.01) during the 10-year follow-up. The association was borderline significant for enterolactone (for the fourth vs. first quartile, least square mean of weight change rate = -0.14 kg/year, 95% CI: -0.29, 0.00). Our data suggest that higher urinary excretion of lignan metabolites, especially enterodiol, is associated with modestly slower weight gain.
Collapse
|
38
|
Combination of Antiestrogens and Omega-3 Fatty Acids for Breast Cancer Prevention. BIOMED RESEARCH INTERNATIONAL 2015; 2015:638645. [PMID: 26339626 PMCID: PMC4538406 DOI: 10.1155/2015/638645] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 12/23/2014] [Indexed: 01/21/2023]
Abstract
The molecular and biological heterogeneity of human breast cancer emphasizes the importance of a multitargeted approach for effective chemoprevention. Targeting the estrogen receptor pathway alone with the antiestrogens, Tamoxifen and Raloxifene reduces the incidence of estrogen receptor positive tumors but is ineffective against the development of hormone independent cancers. Our preclinical data indicate that the administration of omega-3 fatty acids potentiates the antitumor effects of Tamoxifen by inhibiting multiple proliferative and antiapoptotic pathways, several of which interact with estrogen receptor signaling. The complementarity in the mechanism of antitumor action of Tamoxifen and omega-3 fatty acids is well supported by our signaling, genomic, and proteomic studies. Furthermore, administration of omega-3 fatty acids allows the use of lower and, hence, likely less toxic doses of Tamoxifen. If these findings are supported in the clinical setting, the combination of omega-3 fatty acids and anteistrogens may emerge as a promising, effective, and safe chemopreventive strategy to be tested in a large multi-institutional trial using breast cancer incidence as the primary endpoint.
Collapse
|
39
|
Li SY, Wang YX, Wang L, Qian ZB, Ji ML. Cytoplasm estrogen receptor β5 as an improved prognostic factor in thymoma and thymic carcinoma progression. Oncol Lett 2015; 10:2341-2346. [PMID: 26622848 DOI: 10.3892/ol.2015.3555] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2014] [Accepted: 07/23/2015] [Indexed: 02/02/2023] Open
Abstract
A number of previous studies have reported that sex steroid hormones, including estrogens, are involved in the regulation of the thymic function. The aim of the present study was to investigate the expression of estrogen receptor β5 (ERβ5) in thymic tumors and the correlation between ERβ5 expression and thymoma biological characteristics. The expression levels of ERβ5 in thymic epithelial tumors was evaluated in 103 patents using immunohistochemical staining and reverse transcription-quantitative polymerase chain reaction. In addition, an indirect immunofluorescence assay was performed to evaluate the ERβ5 expression levels in the TC1889 and T1682 cell lines. The survival outcome was estimated using Kaplan-Meier plots. The results indicated that ERβ5 expression was mainly located in the thymic tumor cell cytoplasm (87.37%; 90/103 cases) and overexpression was observed in thymic tumors compared with normal thymic tissues (P=0.001). Using the Kruskal-Wallis test, a statistically significant association was observed between cytoplasmic ERβ5 (cERβ5) expression and thymic tumor subtypes (P=0.024) and stages (P=0.003 and R=-0.376). The Kaplan-Meier plots revealed that cERβ5 expression was significantly associated with improved overall and progression-free survival (P=0.008 and P=0.004, respectively). The present study suggested that overexpression of cERβ5 may indicate an improved prognosis and may be involved in the underlying mechanism through which estrogen inhibits thymoma and thymic carcinoma development.
Collapse
Affiliation(s)
- Sheng-Ying Li
- Department of Physiology, Basic Medical College of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Yu-Xia Wang
- Department of Physiology, Basic Medical College of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Lei Wang
- Department of Physiology, Basic Medical College of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Zhi-Bing Qian
- Department of Physiology, Basic Medical College of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Ming-Li Ji
- Department of Physiology, Basic Medical College of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| |
Collapse
|
40
|
Zheng N, Zhang P, Huang H, Liu W, Hayashi T, Zang L, Zhang Y, Liu L, Xia M, Tashiro SI, Onodera S, Ikejima T. ERα down-regulation plays a key role in silibinin-induced autophagy and apoptosis in human breast cancer MCF-7 cells. J Pharmacol Sci 2015; 128:97-107. [PMID: 26117209 DOI: 10.1016/j.jphs.2015.05.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 03/10/2015] [Accepted: 05/01/2015] [Indexed: 02/09/2023] Open
Abstract
The estrogen receptor alpha (ERα) has been proven to be one of the most important therapeutic targets in breast cancer over the last 30 years. Previous studies pointed out that a natural flavonoid, silibinin, induced apoptosis in human breast cancer MCF-7 cells. In the present study we report that exposure of MCF-7 cells to silibinin led to cell death through the down-regulation of ERα expression. Silibinin-induced apoptosis of MCF-7 cells through up-regulation of caspase 6 due to ERα signalling repression was further boosted by ERα antagonist. Moreover, up-regulation of autophagy induced by silibinin accounted for apoptotic exacerbation, being further enhanced by ERα inhibition. Upon ERα activation, series of downstream signalling pathways can be activated. We found that silibinin reduced the expressions of Akt/mTOR and extracellular-signal-related kinase (ERK), which respectively accounted for the induction of autophagy and apoptosis. These effects were further augmented by co-treatment with ERα inhibitor. We conclude that the treatment with silibinin of ERα-positive MCF-7 cells down-regulates the expression of ERα, and subsequently mTOR and ERK signaling pathways, ERα downstream, finally resulting in induction of autophagy and apoptosis.
Collapse
Affiliation(s)
- Nan Zheng
- China-Japan Research Institute of Medical and Pharmaceutical Sciences, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Ping Zhang
- China-Japan Research Institute of Medical and Pharmaceutical Sciences, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Huai Huang
- China-Japan Research Institute of Medical and Pharmaceutical Sciences, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Weiwei Liu
- China-Japan Research Institute of Medical and Pharmaceutical Sciences, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Toshihiko Hayashi
- China-Japan Research Institute of Medical and Pharmaceutical Sciences, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Linghe Zang
- China-Japan Research Institute of Medical and Pharmaceutical Sciences, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Ye Zhang
- China-Japan Research Institute of Medical and Pharmaceutical Sciences, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Lu Liu
- China-Japan Research Institute of Medical and Pharmaceutical Sciences, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Mingyu Xia
- China-Japan Research Institute of Medical and Pharmaceutical Sciences, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Shin-ichi Tashiro
- Institute for Clinical and Biomedical Sciences, Kyoto 603-8072, Japan
| | - Satoshi Onodera
- Department of Clinical and Biomedical Sciences, Showa Pharmaceutical University, Tokyo 194-8543, Japan
| | - Takashi Ikejima
- China-Japan Research Institute of Medical and Pharmaceutical Sciences, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
41
|
Zanella I, Di Lorenzo D. Intracellular receptor regulation of adipose metabolism by the isoflavone genistein. Eur J Nutr 2015; 54:493-5. [PMID: 25662823 DOI: 10.1007/s00394-015-0849-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Isabella Zanella
- Biotechnology/3rd Laboratory and Department of Diagnostics, Civic Hospital of Brescia, 25123, Brescia, Italy
| | | |
Collapse
|
42
|
Bertuloso BD, Podratz PL, Merlo E, de Araújo JFP, Lima LCF, de Miguel EC, de Souza LN, Gava AL, de Oliveira M, Miranda-Alves L, Carneiro MTWD, Nogueira CR, Graceli JB. Tributyltin chloride leads to adiposity and impairs metabolic functions in the rat liver and pancreas. Toxicol Lett 2015; 235:45-59. [PMID: 25819109 DOI: 10.1016/j.toxlet.2015.03.009] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 02/23/2015] [Accepted: 03/21/2015] [Indexed: 12/13/2022]
Abstract
Tributyltin chloride (TBT) is an environmental contaminant used in antifouling paints of boats. Endocrine disruptor effects of TBT are well established in animal models. However, the adverse effects on metabolism are less well understood. The toxicity of TBT in the white adipose tissue (WAT), liver and pancreas of female rats were assessed. Animals were divided into control and TBT (0.1 μg/kg/day) groups. TBT induced an increase in the body weight of the rats by the 15th day of oral exposure. The weight gain was associated with high parametrial (PR) and retroperitoneal (RP) WAT weights. TBT-treatment increased the adiposity, inflammation and expression of ERα and PPARγ proteins in both RP and PR WAT. In 3T3-L1 cells, estrogen treatment reduced lipid droplets accumulation, however increased the ERα protein expression. In contrast, TBT-treatment increased the lipid accumulation and reduced the ERα expression. WAT metabolic changes led to hepatic inflammation, lipid accumulation, increase of PPARγ and reduction of ERα protein expression. Accordingly, there were increases in the glucose tolerance and insulin sensitivity tests with increases in the number of pancreatic islets and insulin levels. These findings suggest that TBT leads to adiposity in WAT specifically, impairing the metabolic functions of the liver and pancreas.
Collapse
Affiliation(s)
- Bruno D Bertuloso
- Department of Morphology, Federal University of Espírito Santo, Brazil
| | | | - Eduardo Merlo
- Department of Morphology, Federal University of Espírito Santo, Brazil
| | | | - Leandro C F Lima
- Institute of Biological Sciences, Federal University of Minas Gerais, Brazil
| | - Emilio C de Miguel
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Brazil
| | | | - Agata L Gava
- Department of Physiology, Federal University of Espírito Santo, Brazil
| | - Miriane de Oliveira
- Department of Internal Medicine, Botucatu School of Medicine, University of São Paulo State, Brazil
| | - Leandro Miranda-Alves
- Experimental Endocrinology Research Group, Institute of Biomedical Sciences, RJ, Brazil
| | | | - Celia R Nogueira
- Department of Internal Medicine, Botucatu School of Medicine, University of São Paulo State, Brazil
| | - Jones B Graceli
- Department of Morphology, Federal University of Espírito Santo, Brazil.
| |
Collapse
|
43
|
Zhang G, Hou X, Gao S. Stimulation of peroxisome proliferator-activated receptor γ inhibits estrogen receptor α transcriptional activity in endometrial carcinoma cells. Oncol Rep 2015; 33:1227-34. [PMID: 25592200 DOI: 10.3892/or.2015.3729] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 12/19/2014] [Indexed: 11/05/2022] Open
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ) and estrogen receptor (ER) belong to a family of nuclear hormone receptors that have been demonstrated to affect each other's transcriptional activity. At present, little is known regarding the effect of PPARγ on ER-mediated transcriptional activity in endometrial carcinoma. In the present study, we aimed to demonstrate the correlation between PPARγ and ER in endometrial carcinoma and to elucidate the biological effects of abnormal expression of PPARγ on endometrial carcinoma cell lines. Immunohistochemical and western blotting methods were used to detect the expression of PPARγ, ERα and ERβ in normal and malignant endometrium. Next, we performed transient transfection to assess the interaction between PPARγ and ER in vitro. Furthermore, we examined cell migration, invasion and proliferation as a biological counterpart. PPARγ and ERα expression levels were significantly associated with pathological grade and clinical stage in endometrial carcinoma (P<0.05). Pearson correlation analysis revealed that PPARγ expression was positively correlated with ERα expression (P<0.05). Using KLE and ERα-positive cells (ECC-1), we demonstrated that the PPARγ regulation of ER expression occurred predominantly through ERα. Moreover, our findings suggest that PPARγ activation inhibited the migration, invasion and proliferation of endometrial carcinoma cells; ECC-1 cells were more sensitive to this inhibition. The present study demonstrated that PPARγ activation inhibited ERα expression in ERα-positive endometrial carcinoma cell lines. This crosstalk may facilitate the development of novel therapeutic methods targeting PPARγ in endometrial carcinoma treatment, particularly ERα-positive carcinomas.
Collapse
Affiliation(s)
- Guiyu Zhang
- Department of Gynecology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Xinxin Hou
- Department of Reproductive Immunology, Hospital and Institute of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai 200011, P.R. China
| | - Shuhong Gao
- Department of Gynecology and Obstetrics, Dongying Honggang Hospital, Dongying, Shandong 257000, P.R. China
| |
Collapse
|
44
|
Yaacob NS, Nasir R, Norazmi MN. Influence of 17β-estradiol on 15-deoxy-δ12,14 prostaglandin J2 -induced apoptosis in MCF-7 and MDA-MB-231 cells. Asian Pac J Cancer Prev 2014; 14:6761-7. [PMID: 24377602 DOI: 10.7314/apjcp.2013.14.11.6761] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
The nuclear receptor, peroxisome proliferator-activated receptor gamma (PPARγ), is expressed in various cancer cells including breast, prostate, colorectal and cervical examples. An endogenous ligand of PPARγ, 15-deoxy-Δ12,14 prostaglandin J2 (PGJ2), is emerging as a potent anticancer agent but the exact mechanism has not been fully elucidated, especially in breast cancer. The present study compared the anticancer effects of PGJ2 on estrogen receptor alpha (ERα)-positive (MCF-7) and ERα-negative (MDA-MB-231) human breast cancer cells. Based on the reported signalling cross-talk between PPARγ and ERα, the effect of the ERα ligand, 17β-estradiol (E2) on the anticancer activities of PGJ2 in both types of cells was also explored. Here we report that PGJ2 inhibited proliferation of both MCF-7 and MDA-MB-231 cells by inducing apoptotic cell death with active involvement of mitochondria. The presence of E2 potentiated PGJ2-induced apoptosis in MCF-7, but not in MDA-MB-231 cells. The PPARγ antagonist, GW9662, failed to block PGJ2-induced activities but potentiated its effects in MCF-7 cells, instead. Interestingly, GW9662 also proved capable of inducing apoptotic cell death. It can be concluded that E2 enhances PPARγ-independent anticancer effects of PGJ2 in the presence of its receptor.
Collapse
Affiliation(s)
- Nik Soriani Yaacob
- Department of Chemical Pathology, Universiti Sains Malaysia, Health Campus, Kelantan, Malaysia E-mail :
| | | | | |
Collapse
|
45
|
Marlatt VL, Sun J, Curran CA, Bailey HC, Kennedy CK, Elphick JR, Martyniuk CJ. Molecular responses to 17β-estradiol in early life stage salmonids. Gen Comp Endocrinol 2014; 203:203-14. [PMID: 24698784 DOI: 10.1016/j.ygcen.2014.03.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 03/11/2014] [Accepted: 03/13/2014] [Indexed: 01/19/2023]
Abstract
Environmental estrogens (EE) are ubiquitous in many aquatic environments and biological responses to EEs in early developmental stages of salmonids are poorly understood compared to juvenile and adult stages. Using 17β-estradiol (E2) as a model estrogen, waterborne exposures were conducted on early life stage rainbow trout (Oncorhynchus mykiss; egg, alevin, swim-up fry) and both molecular and physiological endpoints were measured to quantify the effects of E2. To investigate developmental stage-specific effects, laboratory exposures of 1 μg/L E2 were initiated pre-hatching as eyed embryos or post-hatching upon entering the alevin stage. High mortality (∼90%) was observed when E2 exposures were initiated at the eyed embryo stage compared to the alevin stage (∼35% mortality), demonstrating stage-specific sensitivity. Gene expression analyses revealed that vitellogenin was detectable in the liver of swim-up fry, and was highly inducible by 1 μg/L E2 (>200-fold higher levels compared to control animals). Experiments also confirmed the induction of vitellogenin protein levels in protein extracts isolated from head and tail regions of swim-up fry after E2 exposure. These findings suggest that induction of vitellogenin, a well-characterized biomarker for estrogenic exposure, can be informative measured at this early life stage. Several other genes of the reproductive endocrine axis (e.g. estrogen receptors and androgen receptors) exhibited decreased expression levels compared to control animals. In addition, chronic exposure to E2 during the eyed embryo and alevin stages resulted in suppressive effects on growth related genes (growth hormone receptors, insulin-like growth factor 1) as well as premature hatching, suggesting that the somatotropic axis is a key target for E2-mediated developmental and growth disruptions. Combining molecular biomarkers with morphological and physiological changes in early life stage salmonids holds considerable promise for further defining estrogen action during development, and for assessing the impacts of endocrine disrupting chemicals in vivo in teleosts.
Collapse
Affiliation(s)
- Vicki L Marlatt
- Nautilus Environmental, 8664 Commerce Court, Burnaby, British Columbia V5A 4N71, Canada.
| | - Jinying Sun
- Canadian Rivers Institute and Department of Biology, University of New Brunswick, Saint John, New Brunswick, Canada
| | - Cat A Curran
- Nautilus Environmental, 8664 Commerce Court, Burnaby, British Columbia V5A 4N71, Canada
| | - Howard C Bailey
- Nautilus Environmental, 8664 Commerce Court, Burnaby, British Columbia V5A 4N71, Canada
| | - Chris K Kennedy
- Department of Biological Sciences, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia, Canada
| | - James R Elphick
- Nautilus Environmental, 8664 Commerce Court, Burnaby, British Columbia V5A 4N71, Canada
| | - Christopher J Martyniuk
- Canadian Rivers Institute and Department of Biology, University of New Brunswick, Saint John, New Brunswick, Canada
| |
Collapse
|
46
|
Balogun KA, Randunu RS, Cheema SK. The effect of dietary omega-3 polyunsaturated fatty acids on plasma lipids and lipoproteins of C57BL/6 mice is age and sex specific. Prostaglandins Leukot Essent Fatty Acids 2014; 91:39-47. [PMID: 24935799 DOI: 10.1016/j.plefa.2014.05.002] [Citation(s) in RCA: 12] [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: 10/07/2013] [Revised: 05/15/2014] [Accepted: 05/20/2014] [Indexed: 12/31/2022]
Abstract
There is clear evidence of the effects of sex and age on the prevalence of cardiovascular disease. We investigated the interactions of dietary omega (n)-3 polyunsaturated fatty acids (PUFA), sex, and age on plasma lipids and lipoproteins in the offspring of C57BL/6 mice exposed to high, medium, or low n-3 PUFA at weaning and 16 weeks postweaning. There was an increase in plasma triglycerides from weaning to 16 weeks in male and female offspring; however, the high n-3 PUFA group showed a reduction in triglycerides in both sexes at 16 weeks. High n-3 PUFA caused an increase in plasma LDL-cholesterol from weaning to 16 weeks in male offspring; however, the LDL particle size was significantly larger in the high n-3 PUFA group. Plasma from male mice showed higher cholesterol efflux compared to females; high n-3 PUFA increased cholesterol efflux. Thus the effects of n-3 PUFA are age and sex dependent.
Collapse
Affiliation(s)
- K A Balogun
- Department of Biochemistry, Memorial University, St. John׳s, NL, Canada
| | - R S Randunu
- Department of Biochemistry, Memorial University, St. John׳s, NL, Canada
| | - S K Cheema
- Department of Biochemistry, Memorial University, St. John׳s, NL, Canada.
| |
Collapse
|
47
|
Franconi F, Campesi I. Sex and gender influences on pharmacological response: an overview. Expert Rev Clin Pharmacol 2014; 7:469-85. [DOI: 10.1586/17512433.2014.922866] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
48
|
Koyama N, Nishida Y, Ishii T, Yoshida T, Furukawa Y, Narahara H. Telmisartan induces growth inhibition, DNA double-strand breaks and apoptosis in human endometrial cancer cells. PLoS One 2014; 9:e93050. [PMID: 24667764 PMCID: PMC3965508 DOI: 10.1371/journal.pone.0093050] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Accepted: 02/28/2014] [Indexed: 12/17/2022] Open
Abstract
Telmisartan, an angiotensin II receptor type 1 blocker, is often used as an antihypertension drug, and it has also been characterized as a peroxisome proliferator-activated receptor-gamma (PPARγ) ligand. The purpose of this study was to elucidate the antitumor effects of telmisartan on endometrial cancer cells. We treated three endometrial cancer cell lines with various concentrations of telmisartan, and we investigated the effects of the telmisartan on the cell proliferation, apoptosis, and their related measurements in vitro. We also administered telmisartan to nude mice with experimental tumors to determine its in vivo effects and toxicity. All three endometrial cancer cell lines were sensitive to the growth-inhibitory effect of telmisartan. The induction of apoptosis was confirmed in concert with the altered expression of genes and proteins related to the apoptosis. We also observed that DNA double-strand breaks (DSBs) were induced in HHUA (human endometrial cancer) cells by telmisartan treatment. In addition, experiments in nude mice showed that telmisartan significantly inhibited human endometrial tumor growth, without toxic side effects. Our results suggest that telmisartan might be a new therapeutic option for the treatment of endometrial cancers.
Collapse
Affiliation(s)
- Naoko Koyama
- Department of Obstetrics and Gynecology, Oita University Faculty of Medicine, Oita, Japan
| | - Yoshihiro Nishida
- Department of Obstetrics and Gynecology, Oita University Faculty of Medicine, Oita, Japan
- * E-mail:
| | - Terukazu Ishii
- Department of Obstetrics and Gynecology, Oita University Faculty of Medicine, Oita, Japan
| | - Toshie Yoshida
- Department of Obstetrics and Gynecology, Oita University Faculty of Medicine, Oita, Japan
| | - Yuichi Furukawa
- Department of Obstetrics and Gynecology, Oita University Faculty of Medicine, Oita, Japan
| | - Hisashi Narahara
- Department of Obstetrics and Gynecology, Oita University Faculty of Medicine, Oita, Japan
| |
Collapse
|
49
|
Lin YM, Velmurugan BK, Yeh YL, Tu CC, Ho TJ, Lai TY, Tsai CH, Tsai FJ, Tsai CH, Huang CY. Activation of estrogen receptors with E2 downregulates peroxisome proliferator-activated receptor γ in hepatocellular carcinoma. Oncol Rep 2013; 30:3027-31. [PMID: 24126791 DOI: 10.3892/or.2013.2793] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 09/18/2013] [Indexed: 12/23/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer-related mortality and occurs more often in men than in women; however, little is known about its underlying molecular mechanisms. The present study investigated the effect of estrogen receptor (ER)α and ERβ on peroxisome proliferator-activated receptor γ (PPARγ) expression in Hep3B cells. We examined PPARγ, ERα and ERβ mRNA and protein expression by RT-PCR and western blotting. In order to determine whether PPARγ plays a central role in HCC, we screened for PPARγ expression in liver cancer patient tissues and differentially differentiated HCC cell lines (HA22T, Huh-7, Hep3B and HepG2). We found that PPARγ expression was highly expressed in liver cancer tissues and in Hep3B cells. Furthermore, overexpression of ERα and ERβ was found to decrease PPARγ expression at the transcriptional as well as at the translational level in a ligand-dependent manner. In summary, the present study demonstrated that both ERα and β were sufficient to inhibit PPARγ and provide a valuable therapeutic option for the treatment of HCC patients.
Collapse
Affiliation(s)
- Yueh-Min Lin
- Department of Pathology, Changhua Christian Hospital, Changhua, Taiwan, R.O.C
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Chu R, van Hasselt A, Vlantis AC, Ng EKW, Liu SYW, Fan MD, Ng SK, Chan ABW, Liu Z, Li XY, Chen GG. The cross-talk between estrogen receptor and peroxisome proliferator-activated receptor gamma in thyroid cancer. Cancer 2013; 120:142-53. [PMID: 24114184 DOI: 10.1002/cncr.28383] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Revised: 08/26/2013] [Accepted: 08/26/2013] [Indexed: 11/08/2022]
Abstract
BACKGROUND Estrogen receptor (ER) and peroxisome proliferator-activated receptor gamma (PPARγ) are associated with thyroid tumorigenesis and treatment. However, the interaction between them has not been studied. METHODS The impact of ER over-expression or down-expression by DNA/small interfering RNA (siRNA) transfection, ERα agonists, and the ERβ agonist diarylpropiolnitrile (DPN) on PPARγ expression/activity was examined in papillary thyroid carcinoma (PTC) and anaplastic thyroid carcinoma (ATC) cells. The effects of PPARγ modulation by rosiglitazone (RTZ), a PPARγ ligand, and of PPARγ siRNA on ER expression were determined. Cellular functions reflected by cell proliferation and migration were assayed. Apoptosis was analyzed by terminal deoxynucleotidyl transferase dUTP nick-end labeling, and apoptotic-related proteins were evaluated by Western blot analysis. RESULTS PPARγ protein and activity were reduced by the over-expression of either ERα or ERβ, whereas repression of ERα or ERβ increased PPARγ expression. The administration of RTZ counteracted the effects of ER and also reduced their expression, particularly in PTC cells. Moreover, knockdown of PPARγ increased ER expression and activity. Functionally, ERα activation offset the inhibitory effect of PPARγ on cellular functions, but ERβ activation aggregated it and induced apoptosis, particularly in PTC cells. Finally, the interaction between ERβ and PPARγ enhanced the expression of proapoptotic molecules, such as caspase-3 and apoptosis-inducing factor. CONCLUSIONS This study provides evidence supporting a cross-talk between ER and PPARγ. The reciprocal interaction between PPARγ and ERβ significantly inhibits the proliferation and migration of thyroid cancer cells, providing a new therapeutic strategy against thyroid cancer.
Collapse
Affiliation(s)
- Ryan Chu
- Department of Otorhinolaryngology, Head and Neck Surgery, The Chinese University of Hong Kong, Hong Kong, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|