1
|
Kusewitt DF, Sharma G, Woods CD, Rosas E, Hathaway HJ, Prossnitz ER. GPER expression prevents estrogen-induced urinary retention in obese mice. J Steroid Biochem Mol Biol 2024; 244:106607. [PMID: 39197539 PMCID: PMC11444091 DOI: 10.1016/j.jsbmb.2024.106607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 09/01/2024]
Abstract
Long-term administration of exogenous estrogen is known to cause urinary retention and marked, often fatal, bladder distention in both male and female mice. Estrogen-treated mice have increased bladder pressure and decreased urine flow, suggesting that urinary retention in estrogen-treated mice is due to infravesicular obstruction to urine outflow. Thus, the condition is commonly referred to as bladder outlet obstruction (BOO). Obesity can also lead to urinary retention. As the effects of estrogen are mediated by multiple receptors, including estrogen receptors ERα and ERβ and the G protein-coupled estrogen receptor (GPER), we sought to determine whether GPER plays a role in estrogen-induced BOO, particularly in the context of obesity. Wild type and GPER knockout (KO) mice fed a high-fat diet were ovariectomized or left ovary-intact (sham surgery) and supplemented with slow-release estrogen or vehicle-only pellets. Supplementing both GPER KO and wild type obese mice with estrogen for 8 weeks resulted in weight loss, splenic enlargement, and thymic atrophy, as expected. However, estrogen-treated obese GPER KO mice developed abdominal distension, debilitation, and ulceration of the skin surrounding the urogenital opening. At necropsy, these mice had prominently distended bladders and hydronephrosis. In contrast, estrogen-treated obese wild type mice only rarely displayed these signs. Our results suggest that, under conditions of obesity, estrogen induces BOO as a result of ERα-driven pathways and that GPER expression is protective against BOO.
Collapse
Affiliation(s)
- Donna F Kusewitt
- Department of Pathology, University of New Mexico Health Science Center, Albuquerque, NM, USA; University of New Mexico Comprehensive Cancer Center, University of New Mexico Health Science Center, Albuquerque, NM, USA.
| | - Geetanjali Sharma
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM, USA
| | - Christine D Woods
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM, USA
| | - Emmanuel Rosas
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM, USA
| | - Helen J Hathaway
- Department of Cell Biology & Physiology, University of New Mexico Health Science Center, Albuquerque, NM, USA; University of New Mexico Comprehensive Cancer Center, University of New Mexico Health Science Center, Albuquerque, NM, USA
| | - Eric R Prossnitz
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM, USA; Center of Biomedical Research Excellence in Autophagy, Inflammation and Metabolism, University of New Mexico Health Science Center, Albuquerque, NM, USA; University of New Mexico Comprehensive Cancer Center, University of New Mexico Health Science Center, Albuquerque, NM, USA.
| |
Collapse
|
2
|
Son SE, Im DS. Activation of G Protein-Coupled Estrogen Receptor 1 (GPER) Attenuates Obesity-Induced Asthma by Switching M1 Macrophages to M2 Macrophages. Int J Mol Sci 2024; 25:9532. [PMID: 39273478 PMCID: PMC11395149 DOI: 10.3390/ijms25179532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/29/2024] [Accepted: 08/31/2024] [Indexed: 09/15/2024] Open
Abstract
The prevalence of obesity-induced asthma increases in women after menopause. We hypothesized that the increase in obese asthma in middle-aged women results from estrogen loss. In particular, we focused on the acute action of estrogen through the G protein-coupled estrogen receptor 1 (GPER), previously known as GPR30. We investigated whether GPER activation ameliorates obesity-induced asthma with a high-fat diet (HFD) using G-1, the GPER agonist, and G-36, the GPER antagonist. Administration of G-1 (0.5 mg/kg) suppressed HFD-induced airway hypersensitivity (AHR), and increased immune cell infiltration, whereas G-36 co-treatment blocked it. Histological analysis showed that G-1 treatment inhibited HFD-induced inflammation, fibrosis, and mucus hypersecretion in a GPER-dependent manner. G-1 inhibited the HFD-induced rise in the mRNA levels of pro-inflammatory cytokines in the gonadal white adipose tissue and lungs, whereas G-36 co-treatment reversed this effect. G-1 increased anti-inflammatory M2 macrophages and inhibited the HFD-induced rise in pro-inflammatory M1 macrophages in the lungs. In addition, G-1 treatment reversed the HFD-induced increase in leptin expression and decrease in adiponectin expression in the lungs and gonadal white adipose tissue. The results suggest that activation of GPER could be a therapeutic option for obesity-induced asthma.
Collapse
Affiliation(s)
- So-Eun Son
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Dong-Soon Im
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Fundamental Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
3
|
Wu Z, Xiao C, Wang J, Zhou M, You F, Li X. 17β-estradiol in colorectal cancer: friend or foe? Cell Commun Signal 2024; 22:367. [PMID: 39030619 PMCID: PMC11264751 DOI: 10.1186/s12964-024-01745-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 07/10/2024] [Indexed: 07/21/2024] Open
Abstract
Colorectal cancer (CRC) is a common gastrointestinal malignancy with higher incidence and mortality rates in men compared to women, potentially due to the effects of estrogen signaling. There is substantial evidence supporting the significant role of 17β-Estradiol (E2) in reducing CRC risk in females, although this perspective remains debated. E2 has been demonstrated to inhibit CRC cell proliferation and migration at the cellular level by enhancing DNA mismatch repair, modulating key gene expression, triggering cell cycle arrest, and reducing activity of migration factors. Furthermore, E2 contributes to promote a tumor microenvironment unfavorable for CRC growth by stimulating ERβ expression, reducing inflammatory responses, reversing immunosuppression, and altering the gut microbiome composition. Conversely, under conditions of high oxidative stress, hypoxia, and nutritional deficiencies, E2 may facilitate CRC development through GPER-mediated non-genomic signaling. E2's influence on CRC involves the genomic and non-genomic signals mediated by ERβ and GPER, respectively, leading to its dual roles in anticancer activity and carcinogenesis. This review aims to summarize the potential mechanisms by which E2 directly or indirectly impacts CRC development, providing insights into the phenomenon of sexual dimorphism in CRC and suggesting potential strategies for prevention and treatment.
Collapse
Affiliation(s)
- Zihong Wu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Chong Xiao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Jiamei Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Min Zhou
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University (Chongqing Health Center for Women and Children), Chongqing, 401147, China
| | - Fengming You
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
- Oncology Teaching and Research Department of Chengdu, University of Traditional Chinese Medicine, Chengdu, 610072, China.
| | - Xueke Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| |
Collapse
|
4
|
Darvishzadeh Mahani F, Raji-Amirhasani A, Khaksari M, Mousavi MS, Bashiri H, Hajializadeh Z, Alavi SS. Caloric and time restriction diets improve acute kidney injury in experimental menopausal rats: role of silent information regulator 2 homolog 1 and transforming growth factor beta 1. Mol Biol Rep 2024; 51:812. [PMID: 39007943 DOI: 10.1007/s11033-024-09716-x] [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: 03/28/2024] [Accepted: 06/10/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND Estrogen has a protective impact on acute kidney injury (AKI); moreover, reducing the daily intake of calories impedes developing diseases. The present study aimed to determine the effects of calorie restriction (CR) and time restriction (TR) diets on the expression of silent information regulator 2 homolog 1 (SIRT1), transforming growth factor beta 1 (TGF-β1), and other indicators in the presence and absence of ovaries in AKI female rats. METHODS The female rats were divided into two groups, ovariectomized (OVX) and sham, and were placed on CR and TR diets for eight weeks; afterward, AKI was induced by injecting glycerol, and kidney injury indicators and biochemical parameters were measured before and after AKI. RESULTS After AKI, the levels of urine albumin excretion rate, urea, and creatinine in serum, and TGF-β1 increased, while creatinine clearance and SIRT1 decreased in kidney tissue. CR improved kidney indicators and caused a reduction in TGF-β1 and an increase in SIRT1 in ovary-intact rats. Moreover, CR prevented total antioxidant capacity (TAC) decrease and malondialdehyde (MDA) increase resulting from AKI. Before AKI, an increase in body weight, fasting blood sugar (FBS), low-density lipoprotein (LDL), triglyceride (TG), and total cholesterol (TC), and a decrease in high-density lipoprotein (HDL) were observed in OVX rats compared to sham rats, but CR prevented these changes. The effects of TR were similar to those of CR in all indicators except for TGF-β1, SIRT1, urea, creatinine, and albumin. CONCLUSION The present study indicated that CR is more effective than TR in preventing AKI, probably by increasing SIRT1 and decreasing TGF-β1 in ovary-intact animals.
Collapse
Affiliation(s)
- Fatemeh Darvishzadeh Mahani
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Alireza Raji-Amirhasani
- Endocrinology and Metabolism Research Center, Kerman University of Medical SciencesKerman, Kerman, Iran
- Department of Physiology and Pharmacology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Khaksari
- Endocrinology and Metabolism Research Center, Kerman University of Medical SciencesKerman, Kerman, Iran.
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
- Physiology Research Center, Department of Physiology and Pharmacology, 22 Bahman Blvd, Kerman, Iran.
| | - Maryam Sadat Mousavi
- Clinical Research Development Unit, Shafa Hospital, Kerman University of Medical Sciences, Kerman, Iran
| | - Hamideh Bashiri
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Zahra Hajializadeh
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Samaneh Sadat Alavi
- Department of Physiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
5
|
Zhang M, Mao C, Dai Y, Xu X, Wang X. Qixian granule inhibits ferroptosis in vascular endothelial cells by modulating TRPML1 in the lysosome to prevent postmenopausal atherosclerosis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 328:118076. [PMID: 38521431 DOI: 10.1016/j.jep.2024.118076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/29/2024] [Accepted: 03/18/2024] [Indexed: 03/25/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE QiXian Granule (QXG) is an integrated traditional Chinese medicine formula used to treat postmenopausal atherosclerotic (AS) cardiovascular diseases. The previous studies have found that QXG inhibited isoproterenol (ISO)-induced myocardial remodeling. And its active ingredient, Icraiin, can inhibit ferroptosis by promoting oxidized low-density lipoprotein (xo-LDL)-induced vascular endothelial cell injury and autophagy in atherosclerotic mice. Another active ingredient, Salvianolic Acid B, can suppress ferroptosis and apoptosis during myocardial ischemia/reperfusion injury by reducing ubiquitin-proteasome degradation of Glutathione Peroxidase 4 (GPX4) and down-regulating the reactive oxygen species (ROS)- c-Jun N-terminal kinases (JNK)/mitogen-activated protein kinase (MAPK) pathway. AIM OF THE STUDY The objective of this research was to assess the possible impact of QXG on atherosclerosis in postmenopausal individuals and investigate its underlying mechanisms. MATERIALS AND METHODS Female ApoE-/- mice underwent ovariectomy and were subjected to a high-fat diet (HFD) to establish a postmenopausal atherosclerosis model. The therapeutic effects of QXG were observed in vivo and in vitro through intraperitoneal injection of erastin, G-protein Coupled Estrogen Receptor (GPER) inhibitor (G15), and silent Mucolipin Transient Receptor Potential Channel 1 (TRPML1) adenovirus injection via tail vein. UPLC-MS and molecular docking techniques identified and evaluated major QXG components, contributing to the investigation of QXG's anti-postmenopausal atherosclerotic effects. RESULTS QXG increased serum Estradiol levels, decreased follicle-stimulating hormone (FSH) and luteinizing hormone (LH) levels, which indicated QXG had estrogen-like effects in Ovx/ApoE-/- mice. Furthermore, QXG demonstrated the potential to impede the progression of AS in Ovx/ApoE-/- mice, as evidenced by reductions in serum triglycerides (TG), total cholesterol (TC), and low-density lipoprotein-cholesterol (LDL-C) levels. Additionally, QXG inhibited ferroptosis in Ovx/ApoE-/- mice. Notably, UPLC-MS analysis identified a total of 106 active components in QXG. The results of molecular docking analysis demonstrated that Epmedin B, Astragaloside II, and Orientin exhibit strong binding affinity towards TRPML1. QXG alleviates the progression of atherosclerosis by activating TRPML1 through the GPER pathway or directly activating TRPML1, thereby inhibiting GPX4 and ferritin heavy chain (FTH1)-mediated iron pendant disease. In vitro, QXG-treated serum suppressed proliferation, migration, and ox-LDL-induced MMP and ROS elevation in HAECs. CONCLUSION QXG inhibited GPX4 and FTH1-mediated ferroptosis in vascular endothelial cells through up-regulating GPER/TRPML1 signaling, providing a potential therapeutic option for postmenopausal females seeking a safe and effective medication to prevent atherosclerosis. The study highlights QXG's estrogenic properties and its promising role in combating postmenopausal atherosclerosis.
Collapse
Affiliation(s)
- Meng Zhang
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Chenhan Mao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China
| | - Yang Dai
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210028, China
| | - Xiaojin Xu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China
| | - Xindong Wang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China.
| |
Collapse
|
6
|
Hanafi D, Onyenwoke RU, Kimbro KS. The G-Protein-Coupled Estrogen Receptor Selective Agonist G-1 Attenuates Cell Viability and Migration in High-Grade Serous Ovarian Cancer Cell Lines. Int J Mol Sci 2024; 25:6499. [PMID: 38928205 PMCID: PMC11203932 DOI: 10.3390/ijms25126499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/07/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
The G-protein-coupled estrogen receptor (GPER; G-protein-coupled estrogen receptor 30, also known as GPR30) is a novel estrogen receptor and has emerged as a promising target for ovarian cancer. GPER, a seven-transmembrane receptor, suppresses cellular viability and migration in studied ovarian cancer cells. However, its impact on the fallopian tube, which is the potential origin of high-grade serous (HGSC) ovarian cancer, has not been addressed. This study was conducted to evaluate the relationship of GPER, ovarian cancer subtypes, i.e., high-grade serous cell lines (OV90 and OVCAR420), as well as the cell type that is the potential origin of HGSC ovarian cancer (i.e., the fallopian tube cell line FT190). The selective ligand assessed here is the agonist G-1, which was utilized in an in vitro study to characterize its effects on cellular viability and migration. As a result, this study has addressed the effect of a specific GPER agonist on cell viability, providing a better understanding of the effects of this compound on our diverse group of studied cell lines. Strikingly, attenuated cell proliferation and migration behaviors were observed in the presence of G-1. Thus, our in vitro study reveals the impact of the origin of HGSC ovarian cancers and highlights the GPER agonist G-1 as a potential therapy for ovarian cancer.
Collapse
Affiliation(s)
- Donia Hanafi
- Julius L. Chambers Biomedical/Biotechnology Research Institute, North Carolina Central University, Durham, NC 27707, USA;
- Department of Biological and Biomedical Sciences, North Carolina Central University, Durham, NC 27707, USA;
| | - Rob U. Onyenwoke
- Department of Biological and Biomedical Sciences, North Carolina Central University, Durham, NC 27707, USA;
- Biomanufacturing Research Institute and Technology Enterprise (BRITE), North Carolina Central University, Durham, NC 27707, USA
| | - K. Sean Kimbro
- Julius L. Chambers Biomedical/Biotechnology Research Institute, North Carolina Central University, Durham, NC 27707, USA;
- Department of Biological and Biomedical Sciences, North Carolina Central University, Durham, NC 27707, USA;
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine (MSM), Atlanta, GA 30310, USA
| |
Collapse
|
7
|
Hixon JC, Rivas Zarete JI, White J, Hilaire M, Muhammad A, Yusuf AP, Adu-Addai B, Yates CC, Mahavadi S. Epigenetic Modulation of GPER Expression in Gastric and Colonic Smooth Muscle of Male and Female Non-Obese Diabetic (NOD) Mice: Insights into H3K4me3 and H3K27ac Modifications. Int J Mol Sci 2024; 25:5260. [PMID: 38791299 PMCID: PMC11121689 DOI: 10.3390/ijms25105260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/04/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Type 1 diabetes (T1D) affects gastrointestinal (GI) motility, favoring gastroparesis, constipation, and fecal incontinence, which are more prevalent in women. The mechanisms are unknown. Given the G-protein-coupled estrogen receptor's (GPER) role in GI motility, we investigated sex-related diabetes-induced epigenetic changes in GPER. We assessed GPER mRNA and protein expression levels using qPCR and Western blot analyses, and quantified the changes in nuclear DNA methyltransferases and histone modifications (H3K4me3, H3Ac, and H3K27Ac) by ELISA kits. Targeted bisulfite and chromatin immunoprecipitation assays were used to evaluate DNA methylation and histone modifications around the GPER promoter by chromatin immunoprecipitation assays in gastric and colonic smooth muscle tissues of male and female control (CTR) and non-obese diabetic (NOD) mice. GPER expression was downregulated in NOD, with sex-dependent variations. In the gastric smooth muscle, not in colonic smooth muscle, downregulation coincided with differences in methylation ratios between regions 1 and 2 of the GPER promoter of NOD. DNA methylation was higher in NOD male colonic smooth muscle than in NOD females. H3K4me3 and H3ac enrichment decreased in NOD gastric smooth muscle. H3K4me3 levels diminished in the colonic smooth muscle of NOD. H3K27ac levels were unaffected, but enrichment decreased in NOD male gastric smooth muscle; however, it increased in the NOD male colonic smooth muscle and decreased in the female NOD colonic smooth muscle. Male NOD colonic smooth muscle exhibited decreased H3K27ac levels, not female, whereas female NOD colonic smooth muscle demonstrated diminished enrichment of H3ac at the GPER promoter, contrary to male NOD. Sex-specific epigenetic mechanisms contribute to T1D-mediated suppression of GPER expression in the GI tract. These insights advance our understanding of T1D complications and suggest promising avenues for targeted therapeutic interventions.
Collapse
MESH Headings
- Animals
- Female
- Male
- Mice
- Colon/metabolism
- Colon/pathology
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/genetics
- DNA Methylation
- Epigenesis, Genetic
- Histones/metabolism
- Mice, Inbred NOD
- Muscle, Smooth/metabolism
- Promoter Regions, Genetic
- Receptors, Estrogen/metabolism
- Receptors, Estrogen/genetics
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Stomach/pathology
Collapse
Affiliation(s)
- Juanita C. Hixon
- Center for Cancer Research, Department of Biology, Tuskegee University, Tuskegee, AL 36088, USA; (J.C.H.); (J.W.); (A.M.)
| | - Jatna I. Rivas Zarete
- Department of Biomedical Sciences, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL 36088, USA; (J.I.R.Z.); (B.A.-A.)
| | - Jason White
- Center for Cancer Research, Department of Biology, Tuskegee University, Tuskegee, AL 36088, USA; (J.C.H.); (J.W.); (A.M.)
| | - Mariline Hilaire
- Department of Environment & Nutrition Sciences, College of Agriculture, Tuskegee University, Tuskegee, AL 36088, USA;
| | - Aliyu Muhammad
- Center for Cancer Research, Department of Biology, Tuskegee University, Tuskegee, AL 36088, USA; (J.C.H.); (J.W.); (A.M.)
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, P.M.B. 1044, Zaria 810211, Kaduna State, Nigeria
| | - Abdurrahman Pharmacy Yusuf
- Department of Biochemistry, Federal University of Technology, P.M.B. 65, Minna 920101, Niger State, Nigeria;
| | - Benjamin Adu-Addai
- Department of Biomedical Sciences, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL 36088, USA; (J.I.R.Z.); (B.A.-A.)
| | - Clayton C. Yates
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21218, USA;
| | - Sunila Mahavadi
- Center for Cancer Research, Department of Biology, Tuskegee University, Tuskegee, AL 36088, USA; (J.C.H.); (J.W.); (A.M.)
| |
Collapse
|
8
|
Lappano R, Maggiolini M, Mallet C, Jacquot Y. Commentary: harnessing the first peptidic modulator of the estrogen receptor GPER. Front Pharmacol 2024; 15:1413058. [PMID: 38751778 PMCID: PMC11094232 DOI: 10.3389/fphar.2024.1413058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 04/17/2024] [Indexed: 05/18/2024] Open
Affiliation(s)
- Rosamaria Lappano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Italy
| | - Marcello Maggiolini
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Italy
| | - Christophe Mallet
- Institut National de la Santé et de la Recherche Médicale (INSERM), NEURO-DOL Basics and Clinical Pharmacology of Pain, Université Clermont Auvergne, Clermont-Ferrand, France
- Faculty of Medicine, ANALGESIA Institute, Clermont-Ferrand, France
| | - Yves Jacquot
- Faculty of Pharmacy of Paris, Cibles Thérapeutiques et Conception de Médicaments (CiTCoM), Centre National de la Recherche Scientifique (CNRS) UMR 8038, INSERM U1268, Université Paris Cité, Paris, France
| |
Collapse
|
9
|
Ethier R, Krishnamurthy A, Jeffrey M, Tompkins TA. Profiling of Metabolites in a Fermented Soy Dietary Supplement Reinforces its Role in the Management of Intestinal Inflammation. Mol Nutr Food Res 2024; 68:e2300770. [PMID: 38522032 DOI: 10.1002/mnfr.202300770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 02/19/2024] [Indexed: 03/25/2024]
Abstract
SCOPE Gastro-AD (GAD) is a soy flour derived product that undergoes an industrial fermentation with Lactobacillus delbrueckii R0187 and has demonstrated clinical effects in gastroesophageal reflux and peptic ulcer symptom resolution. The aim of this study is to describe and link GAD's metabolomic profile to plausible mechanisms that manifest and explain the documented clinical outcomes. METHODS AND RESULTS 1H NMR spectroscopy with multivariate statistical analysis is used to characterize the prefermented soy flour and GAD products. The acquired spectra are screened using various resources and the molecular assignments are confirmed using total correlation spectroscopy (TOCSY). Peaks corresponding to different metabolites are integrated and compared between the two products for relative changes. HPLC and GC are used to quantify some specific molecules. NMR analyses demonstrate significant changes in the composition of various assigned bioactive moieties. HPLC and GC analysis demonstrate deglycation of isoflavones after fermentation, resulting in estrogenically active secondary metabolites that have been previously shown to help to reduce inflammation. CONCLUSION The identification of bioactive molecules, such as genistein and SCFAs, capable of modulating anti-inflammatory signaling cascades in the stomach's gastric and neuroendocrine tissues can explain the reported biological effects in GAD and is supported by in vivo data.
Collapse
Affiliation(s)
- Richard Ethier
- Richard Ethier Consulting, Montreal, Quebec, H4C 2J9, Canada
| | - Arun Krishnamurthy
- Purity-IQ Inc., Suite# 102, 150 Research Lane, Guelph, Ontario, N1G 4T2, Canada
| | - Michael Jeffrey
- Faculty of Science, Engineering & Information Technology, Durham College, Oshawa, Ontario, L1G 0C5, Canada
| | - Thomas A Tompkins
- Lallemand Bio-Ingredients, 1620 rue Prefontaine, Montreal, Quebec, H1W 2N8, Canada
| |
Collapse
|
10
|
Muhammad A, Hixon JC, Pharmacy Yusuf A, Rivas Zarete JI, Johnson I, Miller J, Adu-Addai B, Yates C, Mahavadi S. Sex-specific epigenetics drive low GPER expression in gastrointestinal smooth muscles in type 2 diabetic mice. Sci Rep 2024; 14:5633. [PMID: 38453938 PMCID: PMC10920797 DOI: 10.1038/s41598-024-54213-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 02/09/2024] [Indexed: 03/09/2024] Open
Abstract
Type 2 diabetes mellitus (T2D) causes gastroparesis, delayed intestinal transit, and constipation, for unknown reasons. Complications are predominant in women than men (particularly pregnant and postmenopausal women), suggesting a female hormone-mediated mechanism. Low G-protein coupled estrogen receptor (GPER) expression from epigenetic modifications may explain it. We explored sexually differentiated GPER expression and gastrointestinal symptoms related to GPER alterations in wild-type (WT) and T2D mice (db/db). We also created smooth muscle-specific GPER knockout (GPER KO) mice to phenotypically explore the effect of GPER deficiency on gastrointestinal motility. GPER mRNA and protein expression, DNA methylation and histone modifications were measured from stomach and colon samples of db/db and WT mice. Changes in gut motility were also evaluated as daily fecal pellet production patterns. We found that WT female tissues have the highest GPER mRNA and protein expressions. The expression is lowest in all db/db. GPER downregulation is associated with promoter hypermethylation and reduced enrichment of H3K4me3 and H3K27ac marks around the GPER promoter. We also observed sex-specific disparities in fecal pellet production patterns of the GPER KO mice compared to WT. We thus, conclude that T2D impairs gut GPER expression, and epigenetic sex-specific mechanisms matter in the downregulation.
Collapse
Affiliation(s)
- Aliyu Muhammad
- Department of Biology, Center for Cancer Research, Tuskegee University, Tuskegee, AL, 36088, USA
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, P.M.B. 1044, Zaria, Kaduna State, Nigeria
| | - Juanita C Hixon
- Department of Biology, Center for Cancer Research, Tuskegee University, Tuskegee, AL, 36088, USA
| | | | - Jatna I Rivas Zarete
- Department of Biomedical Sciences, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL, 36088, USA
| | - India Johnson
- Department of Biology, Center for Cancer Research, Tuskegee University, Tuskegee, AL, 36088, USA
| | - Jamial Miller
- Department of Biology, Center for Cancer Research, Tuskegee University, Tuskegee, AL, 36088, USA
| | - Benjamin Adu-Addai
- Department of Biomedical Sciences, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL, 36088, USA
| | - Clayton Yates
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sunila Mahavadi
- Department of Biology, Center for Cancer Research, Tuskegee University, Tuskegee, AL, 36088, USA.
| |
Collapse
|
11
|
Cappelletto E, Fasiolo LT, Salizzato V, Piccin L, Fabozzi A, Contato A, Bianco PD, Pasello G, Chiarion-Sileni V, Gion M, Fabricio ASC. Cytokine and soluble programmed death-ligand 1 levels in serum and plasma of cancer patients treated with immunotherapy: Preanalytical and analytical considerations. Int J Biol Markers 2024; 39:9-22. [PMID: 38407953 DOI: 10.1177/03936155231226234] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
AIM To evaluate cytokine and soluble programmed death ligand-1 (sPD-L1) levels in the serum and plasma of cancer patients treated with immunotherapy, and to test different assays. METHODS Three Luminex xMAP assays and two ELLA microfluidic cartridges were used to screen 28 immune-related biomarkers in 38 paired serum and citrate-theophylline-adenosine-dipyridamole (CTAD) plasma samples collected from 10 advanced melanoma or non-small cell lung cancer (NSCLC) patients at different time points during immunotherapy. RESULTS Twenty-three of 28 biomarkers were detected both in serum and plasma by at least one of the assays, including IL-1β, IL-2, IL-4, IL-5, IL-6, IL-8, IL-10, IL-12p70, GM-CSF, IFN-γ, TNF-α, VEGF, IP-10, MCP-1, eotaxin, fractalkine, G-CSF, IFN-α, IL-1RA, IL-13, IL-17A, MIP-1β and sPD-L1. Conversely, FGF-2 and IL-1α were not detected in both matrices; GRO-α factor and EGF were detected only in serum and MIP-1α only in plasma. sPD-L1, MCP-1, IFN-γ, IL-8, MIP-1β and VEGF were, respectively, 1.15-, 1.44-, 1.83-, 2.43-, 2.82-, 6.72-fold higher in serum, whereas IL-10, IL-4, IL-2 and IL-5 were 1.05-, 1.19-, 1.92- and 2.17-fold higher, respectively, in plasma. IP-10 levels were higher in plasma but, as well as for VEGF, the bias serum versus plasma varied depending on the assay used (IP-10: -5.7% to -145%; VEGF: 115% to 165%). No significant differences were found for the remaining nine analyzed cytokines. CONCLUSION The cytokine and sPD-L1 levels may differ between serum and plasma samples collected from cancer patients treated with immunotherapy, and the results obtained can be influenced by the different characteristics of the tested assays. The standardization of pre-analytical and analytical procedures is therefore needed for the future implementation of these circulating biomarkers in clinical practice.
Collapse
Affiliation(s)
- Elia Cappelletto
- Regional Center for Biomarkers, Department of Clinical Pathology, AULSS3 Serenissima, Venice, Italy
| | | | | | - Luisa Piccin
- Medical Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Alessio Fabozzi
- Medical Oncology 3, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Anna Contato
- Regional Center for Biomarkers, Department of Clinical Pathology, AULSS3 Serenissima, Venice, Italy
| | - Paola Del Bianco
- Clinical Research Unit, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Giulia Pasello
- Medical Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | | | - Massimo Gion
- Regional Center for Biomarkers, Department of Clinical Pathology, AULSS3 Serenissima, Venice, Italy
| | - Aline S C Fabricio
- Medical Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| |
Collapse
|
12
|
Arisawa K, Matsuoka A, Ozawa N, Ishikawa T, Ichi I, Fujiwara Y. GPER/PKA-Dependent Enhancement of Hormone-Sensitive Lipase Phosphorylation in 3T3-L1 Adipocytes by Piceatannol. Nutrients 2023; 16:38. [PMID: 38201867 PMCID: PMC10781143 DOI: 10.3390/nu16010038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/06/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
We previously reported that piceatannol (PIC) had an anti-obesity effect only in ovariectomized (OVX) postmenopausal obesity mice. PIC was found to induce the phosphorylation of hormone-sensitive lipase (pHSL) in OVX mice. To elucidate the mechanism by which PIC activates HSL, we investigated the effect of PIC using 3T3-L1 adipocytes. PIC induced HSL phosphorylation at Ser563 in 3T3-L1 cells, as in vivo experiments showed. pHSL (Ser563) is believed to be activated through the β-adrenergic receptor (β-AR) and protein kinase A (PKA) pathways; however, the addition of a selective inhibitor of β-AR did not inhibit the effect of PIC. The addition of a PKA inhibitor with PIC blocked pHSL (Ser563), suggesting that the effects are mediated by PKA in a different pathway than β-AR. The addition of G15, a selective inhibitor of the G protein-coupled estrogen receptor (GPER), reduced the activation of HSL by PIC. Furthermore, PIC inhibited insulin signaling and did not induce pHSL (Ser565), which represents its inactive form. These results suggest that PIC acts as a phytoestrogen and phosphorylates HSL through a novel pathway that activates GPER and its downstream PKA, which may be one of the inhibitory actions of PIC on fat accumulation in estrogen deficiency.
Collapse
Affiliation(s)
- Kotoko Arisawa
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8577, Japan;
- Graduate School of Humanities and Sciences, Ochanomizu University, Tokyo 112-8610, Japan; (A.M.); (N.O.)
| | - Ayumi Matsuoka
- Graduate School of Humanities and Sciences, Ochanomizu University, Tokyo 112-8610, Japan; (A.M.); (N.O.)
| | - Natsuki Ozawa
- Graduate School of Humanities and Sciences, Ochanomizu University, Tokyo 112-8610, Japan; (A.M.); (N.O.)
| | - Tomoko Ishikawa
- Institute for Human Life Science, Ochanomizu University, Tokyo 112-8610, Japan; (T.I.); (I.I.)
- Department of Human Nutrition, Seitoku University, Chiba 271-8555, Japan
| | - Ikuyo Ichi
- Institute for Human Life Science, Ochanomizu University, Tokyo 112-8610, Japan; (T.I.); (I.I.)
- Natural Science Division, Faculty of Core Research, Ochanomizu University, Tokyo 112-8610, Japan
| | - Yoko Fujiwara
- Graduate School of Humanities and Sciences, Ochanomizu University, Tokyo 112-8610, Japan; (A.M.); (N.O.)
- Institute for Human Life Science, Ochanomizu University, Tokyo 112-8610, Japan; (T.I.); (I.I.)
| |
Collapse
|
13
|
Sun HY, Lin XY. Analysis of the management and therapeutic performance of diabetes mellitus employing special target. World J Diabetes 2023; 14:1721-1737. [PMID: 38222785 PMCID: PMC10784800 DOI: 10.4239/wjd.v14.i12.1721] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/31/2023] [Accepted: 10/23/2023] [Indexed: 12/14/2023] Open
Abstract
Diabetes mellitus (DM) is a chronic metabolic condition characterized predominantly by hyperglycemia. The most common causes contributing to the pathophysiology of diabetes are insufficient insulin secretion, resistance to insulin's tissue-acting effects, or a combination of both. Over the last 30 years, the global prevalence of diabetes increased from 4% to 6.4%. If no better treatment or cure is found, this amount might climb to 430 million in the coming years. The major factors of the disease's deterioration include age, obesity, and a sedentary lifestyle. Finding new therapies to manage diabetes safely and effectively without jeopardizing patient compliance has always been essential. Among the medications available to manage DM on this journey are glucagon-like peptide-1 agonists, thiazolidinediones, sulphonyl urease, glinides, biguanides, and insulin-targeting receptors discovered more than 10 years ago. Despite the extensive preliminary studies, a few clinical observations suggest this process is still in its early stages. The present review focuses on targets that contribute to insulin regulation and may be employed as targets in treating diabetes since they may be more efficient and secure than current and traditional treatments.
Collapse
Affiliation(s)
- Hong-Yan Sun
- Department of Endocrine and Metabolic Diseases, Yantaishan Hospital, Yantai 264003, Shandong Province, China
| | - Xiao-Yan Lin
- Department of Endocrine and Metabolic Diseases, Yantaishan Hospital, Yantai 264003, Shandong Province, China
| |
Collapse
|
14
|
Jouffre B, Acramel A, Jacquot Y, Daulhac L, Mallet C. GPER involvement in inflammatory pain. Steroids 2023; 200:109311. [PMID: 37734514 DOI: 10.1016/j.steroids.2023.109311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/11/2023] [Accepted: 09/18/2023] [Indexed: 09/23/2023]
Abstract
Chronic pain is a worldwide refractory health disease that causes major financial and emotional burdens and that is devastating for individuals and society. One primary source of pain is inflammation. Current treatments for inflammatory pain are weakly effective, although they usually replace analgesics, such as opioids and non-steroidal anti-inflammatory drugs, which display serious side effects. Emerging evidence indicates that the membrane G protein-coupled estrogen receptor (GPER) may play an important role in the regulation of inflammation and pain. Herein, we focus on the consequences of pharmacological and genetic GPER modulation in different animal models of inflammatory pain. We also provide a brief overview of the putative mechanisms including the direct action of GPER on pain transmission and inflammation.
Collapse
Affiliation(s)
- Baptiste Jouffre
- Université Clermont Auvergne, Inserm U1107 Neuro-Dol, Pharmacologie Fondamentale et Clinique de la Douleur, Clermont-Ferrand, France; ANALGESIA Institute, Faculty of Medicine, 63000 Clermont-Ferrand, France
| | - Alexandre Acramel
- CiTCoM, CNRS - UMR 8038, INSERM U1268, Faculty of Pharmacy of Paris, University Paris Cité, 75270 Paris Cedex 06, France; Department of Pharmacy, Institut Curie, 75248 Paris Cedex 06, France
| | - Yves Jacquot
- CiTCoM, CNRS - UMR 8038, INSERM U1268, Faculty of Pharmacy of Paris, University Paris Cité, 75270 Paris Cedex 06, France
| | - Laurence Daulhac
- Université Clermont Auvergne, Inserm U1107 Neuro-Dol, Pharmacologie Fondamentale et Clinique de la Douleur, Clermont-Ferrand, France; ANALGESIA Institute, Faculty of Medicine, 63000 Clermont-Ferrand, France
| | - Christophe Mallet
- Université Clermont Auvergne, Inserm U1107 Neuro-Dol, Pharmacologie Fondamentale et Clinique de la Douleur, Clermont-Ferrand, France; ANALGESIA Institute, Faculty of Medicine, 63000 Clermont-Ferrand, France.
| |
Collapse
|
15
|
Sommer B, González-Ávila G, Flores-Soto E, Montaño LM, Solís-Chagoyán H, Romero-Martínez BS. Phytoestrogen-Based Hormonal Replacement Therapy Could Benefit Women Suffering Late-Onset Asthma. Int J Mol Sci 2023; 24:15335. [PMID: 37895016 PMCID: PMC10607548 DOI: 10.3390/ijms242015335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/10/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
It has been observed that plasmatic concentrations of estrogens, progesterone, or both correlate with symptoms in asthmatic women. Fluctuations in female sex steroid concentrations during menstrual periods are closely related to asthma symptoms, while menopause induces severe physiological changes that might require hormonal replacement therapy (HRT), that could influence asthma symptoms in these women. Late-onset asthma (LOA) has been categorized as a specific asthmatic phenotype that includes menopausal women and novel research regarding therapeutic alternatives that might provide relief to asthmatic women suffering LOA warrants more thorough and comprehensive analysis. Therefore, the present review proposes phytoestrogens as a promising HRT that might provide these females with relief for both their menopause and asthma symptoms. Besides their well-recognized anti-inflammatory and antioxidant capacities, phytoestrogens activate estrogen receptors and promote mild hormone-like responses that benefit postmenopausal women, particularly asthmatics, constituting therefore a very attractive potential therapy largely due to their low toxicity and scarce side effects.
Collapse
Affiliation(s)
- Bettina Sommer
- Departamento de Investigación en Hiperreactividad Bronquial, Instituto Nacional de Enfermedades Respiratorias ‘Ismael Cosio Villegas’, Calzada de Tlalpan 4502, Colonia Sección XVI, Mexico City CP 14080, Mexico
| | - Georgina González-Ávila
- Laboratorio de Oncología Biomédica, Departamento de Enfermedades Crónico Degenerativas, Instituto Nacional de Enfermedades Respiratorias ‘Ismael Cosio Villegas’, Mexico City CP 14080, Mexico;
| | - Edgar Flores-Soto
- Departmento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City CP 04510, Mexico; (E.F.-S.); (L.M.M.); (B.S.R.-M.)
| | - Luis M. Montaño
- Departmento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City CP 04510, Mexico; (E.F.-S.); (L.M.M.); (B.S.R.-M.)
| | - Héctor Solís-Chagoyán
- Neurociencia Cognitiva Evolutiva, Centro de Investigación en Ciencias Cognitivas, Universidad Autónoma del Estado de Morelos, Cuernavaca CP 62209, Morelos, Mexico;
| | - Bianca S. Romero-Martínez
- Departmento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City CP 04510, Mexico; (E.F.-S.); (L.M.M.); (B.S.R.-M.)
| |
Collapse
|
16
|
He W, Gao Z, Liu S, Tan L, Wu Y, Liu J, Zheng Z, Fan W, Luo Y, Chen Z, Song S. G protein-coupled estrogen receptor activation by bisphenol-A disrupts lipid metabolism and induces ferroptosis in the liver. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 334:122211. [PMID: 37454720 DOI: 10.1016/j.envpol.2023.122211] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 07/12/2023] [Accepted: 07/14/2023] [Indexed: 07/18/2023]
Abstract
As a metabolic disruptor, bisphenol A (BPA) has been widely reported to disrupt lipid balance. Moreover, BPA has gained significant attention due to its estrogenic activity. While both ferroptosis and the G-protein-coupled estrogen receptor (GPER) have been implicated in lipid metabolism, their link to BPA-induced lipid accumulation remains unclear. In this study, chickens were randomly assigned to three groups and housed them for 4 weeks: a control group (0 μg/L BPA), a low dose group (50 μg/L BPA) and a high dose group (5000 μg/L BPA) to investigate the underlying mechanism of BPA-induced hepatotoxicity. Our results showed that BPA exposure significantly increased the contents of TG, TC, and LDL-C while decreasing HDL-C levels. We also found that BPA treatment altered the levels of genes involved in fatty acid β-oxidation (ampkα, cpt-1, and ppaα), synthesis (acc, fas, scd-1, and srebp-1) and absorption (lpl and cd36). Moreover, the results showed that the BPA group had higher levels of IL-1β, IL-18 and TNF-α. These results indicated that BPA exposure disrupted lipid metabolism and induced inflammation in the liver. We also demonstrated that BPA caused hepatic ferroptosis by raising iron content and the expression of genes related to lipid peroxidation (lpcat3, acsl4 and alox15), while reducing the expression of antioxidant system-associated genes (gpx4, slc7a11 and slc3a2). Importantly, BPA remarkably activated GPER expression in the liver. Interestingly, inhibition of GPER remarkably ameliorated BPA-induced lipid metabolism disorder, inflammatory response, and ferroptosis, indicating the crucial role of GPER in BPA-induced liver abnormalities. These findings highlight the link between GPER and ferroptosis in BPA-induced hepatotoxicity, providing new insights into the potential hazard of BPA.
Collapse
Affiliation(s)
- Wanqiu He
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Zhangshan Gao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Shuhui Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Lei Tan
- Shenzhen Institute of Quality & Safety Inspection and Research, Shenzhen, 518000, China
| | - Yuting Wu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Jiwen Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Ziyi Zheng
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Wentao Fan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Yan Luo
- Shenzhen Institute of Quality & Safety Inspection and Research, Shenzhen, 518000, China
| | - Zeguo Chen
- Shenzhen Institute of Quality & Safety Inspection and Research, Shenzhen, 518000, China
| | - Suquan Song
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China.
| |
Collapse
|
17
|
Han S, Mueller C, Wuebbolt C, Kilcullen S, Nayyar V, Calle Gonzalez B, Mahdavi Fard A, Floss JC, Morales MJ, Patel SP. Selective effects of estradiol on human corneal endothelial cells. Sci Rep 2023; 13:15279. [PMID: 37714879 PMCID: PMC10504266 DOI: 10.1038/s41598-023-42290-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 09/07/2023] [Indexed: 09/17/2023] Open
Abstract
In Fuchs endothelial corneal dystrophy (FECD), mitochondrial and oxidative stresses in corneal endothelial cells (HCEnCs) contribute to cell demise and disease progression. FECD is more common in women than men, but the basis for this observation is poorly understood. To understand the sex disparity in FECD prevalence, we studied the effects of the sex hormone 17-β estradiol (E2) on growth, oxidative stress, and metabolism in primary cultures of HCEnCs grown under physiologic ([O2]2.5) and hyperoxic ([O2]A) conditions. We hypothesized that E2 would counter the damage of oxidative stress generated at [O2]A. HCEnCs were treated with or without E2 (10 nM) for 7-10 days under both conditions. Treatment with E2 did not significantly alter HCEnC density, viability, ROS levels, oxidative DNA damage, oxygen consumption rates, or extracellular acidification rates in either condition. E2 disrupted mitochondrial morphology in HCEnCs solely from female donors in the [O2]A condition. ATP levels were significantly higher at [O2]2.5 than at [O2]A in HCEnCs from female donors only, but were not affected by E2. Our findings demonstrate the resilience of HCEnCs against hyperoxic stress. The effects of hyperoxia and E2 on HCEnCs from female donors suggest cell sex-specific mechanisms of toxicity and hormonal influences.
Collapse
Affiliation(s)
- Seoyoung Han
- Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Christian Mueller
- Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Caitlin Wuebbolt
- Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Sean Kilcullen
- Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Varinda Nayyar
- Research Service, Veterans Administration Western New York Healthcare System, Buffalo, NY, USA
- Department of Ophthalmology, Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Brayan Calle Gonzalez
- Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Ali Mahdavi Fard
- Department of Ophthalmology, Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Jamie C Floss
- Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Michael J Morales
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Sangita P Patel
- Research Service, Veterans Administration Western New York Healthcare System, Buffalo, NY, USA.
- Department of Ophthalmology, Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA.
- Ophthalmology Service, Veterans Administration Western New York Healthcare System, Buffalo, NY, USA.
| |
Collapse
|
18
|
Visniauskas B, Kilanowski-Doroh I, Ogola BO, Mcnally AB, Horton AC, Imulinde Sugi A, Lindsey SH. Estrogen-mediated mechanisms in hypertension and other cardiovascular diseases. J Hum Hypertens 2023; 37:609-618. [PMID: 36319856 PMCID: PMC10919324 DOI: 10.1038/s41371-022-00771-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 10/06/2022] [Accepted: 10/18/2022] [Indexed: 06/08/2023]
Abstract
Cardiovascular disease (CVD) is the leading cause of death globally for men and women. Premenopausal women have a lower incidence of hypertension and other cardiovascular events than men of the same age, but diminished sex differences after menopause implicates 17-beta-estradiol (E2) as a protective agent. The cardioprotective effects of E2 are mediated by nuclear estrogen receptors (ERα and ERβ) and a G protein-coupled estrogen receptor (GPER). This review summarizes both established as well as emerging estrogen-mediated mechanisms that underlie sex differences in the vasculature during hypertension and CVD. In addition, remaining knowledge gaps inherent in the association of sex differences and E2 are identified, which may guide future clinical trials and experimental studies in this field.
Collapse
Affiliation(s)
- Bruna Visniauskas
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| | | | - Benard O Ogola
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Alexandra B Mcnally
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Alec C Horton
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Ariane Imulinde Sugi
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Sarah H Lindsey
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA.
- Tulane Center of Excellence in Sex-Based Biology and Medicine, New Orleans, LA, USA.
- Tulane Brain Institute, New Orleans, LA, USA.
| |
Collapse
|
19
|
Yuan J, Yang J, Xu X, Wang Z, Jiang Z, Ye Z, Ren Y, Wang Q, Wang T. Bisphenol A (BPA) Directly Activates the G Protein-Coupled Estrogen Receptor 1 and Triggers the Metabolic Disruption in the Gonadal Tissue of Apostichopus japonicus. BIOLOGY 2023; 12:798. [PMID: 37372083 DOI: 10.3390/biology12060798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 05/26/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023]
Abstract
The sea cucumber, Apostichopus japonicus, is a marine benthic organism that feeds on small benthic particulate matter and is easily affected by pollutants. Bisphenol A (BPA, 4,4'-isopropylidenediphenol) has been identified as an endocrine disruptor. It is ubiquitously detectable in oceans and affects a variety of marine animals. It functions as an estrogen analog and typically causes reproductive toxicity by interfering with the endocrine system. To comparatively analyze the reproductive effects of estradiol (E2) and BPA on sea cucumbers, we identified a G protein-coupled estrogen receptor 1 (GPER1) in A. japonicus and investigated its effects on reproduction. The results showed that BPA and E2 exposure activated A. japonicus AjGPER1, thereby mediating the mitogen-activated protein kinase signaling pathways. High-level expression of AjGPER1 in the ovarian tissue was confirmed by qPCR. Furthermore, metabolic changes were induced by 100 nM (22.83 μg/L) BPA exposure in the ovarian tissue, leading to a notable increase in the activities of trehalase and phosphofructokinase. Overall, our findings suggest that AjGPER1 is directly activated by BPA and affects sea cucumber reproduction by disrupting ovarian tissue metabolism, suggesting that marine pollutants pose a threat to the conservation of sea cucumber resources.
Collapse
Affiliation(s)
- Jieyi Yuan
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science and Technology College, Zhejiang Ocean University, Zhoushan 316022, China
| | - Jingwen Yang
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science and Technology College, Zhejiang Ocean University, Zhoushan 316022, China
| | - Xiuwen Xu
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science and Technology College, Zhejiang Ocean University, Zhoushan 316022, China
| | - Zexianghua Wang
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science and Technology College, Zhejiang Ocean University, Zhoushan 316022, China
| | - Zhijing Jiang
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science and Technology College, Zhejiang Ocean University, Zhoushan 316022, China
| | - Zhiqing Ye
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science and Technology College, Zhejiang Ocean University, Zhoushan 316022, China
| | - Yucheng Ren
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science and Technology College, Zhejiang Ocean University, Zhoushan 316022, China
| | - Qing Wang
- Research and Development Center for Efficient Utilization of Coastal Bioresources, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China
| | - Tianming Wang
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science and Technology College, Zhejiang Ocean University, Zhoushan 316022, China
| |
Collapse
|
20
|
Han S, Mueller C, Wuebbolt C, Kilcullen S, Nayyar V, Gonzalez BC, Fard AM, Floss JC, Morales MJ, Patel SP. Selective effects of estradiol on human corneal endothelial cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.27.538629. [PMID: 37162976 PMCID: PMC10168450 DOI: 10.1101/2023.04.27.538629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Fuchs endothelial corneal dystrophy (FECD) results from genetic and environmental factors triggering mitochondrial and oxidative stress in corneal endothelial cells (CEnCs) leading to CEnC death and corneal opacification. FECD is more common in women than men, but the basis for this observation is unknown. Because FECD is commonly diagnosed around the time of the menopausal transition in women when estrogen levels decrease precipitously, we studied the effects of the potent estrogen,17-β estradiol (E2) on growth, oxidative stress, and metabolism in primary cultures of human CEnCs (HCEnCs) under conditions of physiologic 2.5% O 2 ([O 2 ] 2.5 ) and under hyperoxic stress ([O 2 ] A : room air + 5% CO 2 ). We hypothesized that E2 would counter the stresses of the hyperoxic environment in HCEnCs. HCEnCs were treated ± 10 nM E2 for 7-10 days at [O 2 ] 2.5 and [O 2 ] A followed by measurements of cell density, viability, reactive oxygen species (ROS), mitochondrial morphology, oxidative DNA damage, ATP levels, mitochondrial respiration (O 2 consumption rate [OCR]), and glycolysis (extracellular acidification rate [ECAR]). There were no significant changes in HCEnC density, viability, ROS levels, oxidative DNA damage, OCR, and ECAR in response to E2 under either O 2 condition. We found that E2 disrupted mitochondrial morphology in HCEnCs from female donors but not male donors at the [O 2 ] A condition. ATP levels were significantly higher at [O 2 ] 2.5 compared to [O 2 ] A in HCEnCs from female donors only, but were not affected by E2. Our findings demonstrate the overall resilience of primary HCEnCs against hyperoxic stress. The selective detrimental effects of hyperoxia and estradiol on HCEnCs from female but not male donors suggests mechanisms of toxicity based upon cell-sex in addition to hormonal environment.
Collapse
Affiliation(s)
- Seoyoung Han
- Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, USA
| | - Christian Mueller
- Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, USA
| | - Caitlin Wuebbolt
- Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, USA
| | - Sean Kilcullen
- Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, USA
| | - Varinda Nayyar
- Research Service, Veterans Administration of Western New York Healthcare System, Buffalo, New York, USA
- Ross Eye Institute, Department of Ophthalmology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, USA
| | - Brayan Calle Gonzalez
- Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, USA
| | - Ali Mahdavi Fard
- Ross Eye Institute, Department of Ophthalmology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, USA
| | - Jamie C. Floss
- Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, USA
| | - Michael J. Morales
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, USA
| | - Sangita P. Patel
- Research Service, Veterans Administration of Western New York Healthcare System, Buffalo, New York, USA
- Ross Eye Institute, Department of Ophthalmology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, USA
- Ophthalmology Service, Veterans Administration of Western New York Healthcare System, Buffalo, New York, USA
| |
Collapse
|
21
|
Quan J, Zhang T, Gu Y, Meng G, Zhang Q, Liu L, Wu H, Zhang S, Wang X, Zhang J, Sun S, Wang X, Zhou M, Jia Q, Song K, Niu K. Green tea intake and the risk of hypertension in premenopausal women: the TCLSIH cohort study. Food Funct 2023; 14:4406-4413. [PMID: 37097224 DOI: 10.1039/d2fo03342a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2023]
Abstract
Background and aims: Tea polyphenols, such as green tea polyphenols, have been extensively studied as agents that ameliorate cardiovascular disease and blood pressure in vitro and in animal studies. However, epidemiological evidence for the association of green tea consumption with hypertension (HTN) is inconsistent. In addition, such an association has not been prospectively examined in the general adult population, particularly among young women. Therefore, we designed a cohort study to examine whether green tea consumption increases the risk of HTN in premenopausal women. Methods and results: This prospective cohort study investigated 6633 premenopausal female participants without hypertension, cardiovascular disease, and cancer at the baseline. Green tea consumption was measured at the baseline using a validated food frequency questionnaire. Hypertension was confirmed with the SBP ≥140 mm Hg-1 or with the DBP ≥90 mm Hg-1. Cox proportional hazards regression models were used to examine the association of green tea consumption with incident hypertension. A total of 488 first incident cases of hypertension occurred during 24 957 person-years of follow-up (median follow-up of 4.0 years). After adjustment for potential confounding variables, the multivariable hazard ratios (95% confidence intervals) for incident hypertension in premenopausal female participants with different green tea consumption frequencies were 1.00 (reference) for almost never, 0.84 (0.67, 1.07) for 1 cup per week, 1.02 (0.77, 1.35) for 2-6 cups per week, and 0.65 (0.44, 0.96) for ≥1 cup per day. Conclusions: The results from our prospective study indicate that the consumption of green tea is associated with a reduced risk of HTN in premenopausal women.
Collapse
Affiliation(s)
- Jing Quan
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.
- School of Public Health of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China.
| | - Tingjing Zhang
- School of Public Health of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- School of Public Health, Wannan Medical College, 22 West Wenchang Road, Wuhu, Anhui Province, 241002, People's Republic of China
| | - Yeqing Gu
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.
| | - Ge Meng
- School of Public Health of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Qing Zhang
- Health Management Centre, Tianjin Medical University General Hospital, Tianjin, China
| | - Li Liu
- Health Management Centre, Tianjin Medical University General Hospital, Tianjin, China
| | - Hongmei Wu
- School of Public Health of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China.
| | - Shunming Zhang
- School of Public Health of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China.
| | - Xuena Wang
- School of Public Health of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China.
| | - Juanjuan Zhang
- School of Public Health of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China.
| | - Shaomei Sun
- Health Management Centre, Tianjin Medical University General Hospital, Tianjin, China
| | - Xing Wang
- Health Management Centre, Tianjin Medical University General Hospital, Tianjin, China
| | - Ming Zhou
- Health Management Centre, Tianjin Medical University General Hospital, Tianjin, China
| | - Qiyu Jia
- Health Management Centre, Tianjin Medical University General Hospital, Tianjin, China
| | - Kun Song
- Health Management Centre, Tianjin Medical University General Hospital, Tianjin, China
| | - Kaijun Niu
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.
- School of Public Health of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China.
- Health Management Centre, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
- Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin, China
| |
Collapse
|
22
|
Le G, Baumann CW, Warren GL, Lowe DA. In vivo potentiation of muscle torque is enhanced in female mice through estradiol-estrogen receptor signaling. J Appl Physiol (1985) 2023; 134:722-730. [PMID: 36735234 PMCID: PMC10027088 DOI: 10.1152/japplphysiol.00731.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/13/2023] [Accepted: 02/02/2023] [Indexed: 02/04/2023] Open
Abstract
Estradiol affects several properties of skeletal muscle in females including strength. Here, we developed an approach to measure in vivo posttetanic twitch potentiation (PTP) of the anterior crural muscles of anesthetized mice and tested the hypothesis that 17β-estradiol (E2) enhances PTP through estrogen receptor (ER) signaling. Peak torques of potentiated twitches were ∼40%-60% greater than those of unpotentiated twitches and such PTP was greater in ovary-intact mice, or ovariectomized (Ovx) mice treated with E2, compared with Ovx mice (P ≤ 0.047). PTP did not differ between mice with and without ERα ablated in skeletal muscle fibers (P = 0.347). Treatment of ovary-intact and Ovx mice with ERβ antagonist and agonist (PHTPP and DPN, respectively) did not affect PTP (P ≥ 0.258). Treatment with G1, an agonist of the G protein-coupled estrogen receptor (GPER), significantly increased PTP in Ovx mice from 41 ± 10% to 66 ± 21% (means ± SD; P = 0.034). Collectively, these data indicate that E2 signals through GPER, and not ERα or ERβ, in skeletal muscles of female mice to augment an in vivo parameter of strength, namely, PTP.NEW & NOTEWORTHY A novel in vivo approach was developed to measure potentiation of skeletal muscle torque in female mice and highlight another parameter of strength that is impacted by estradiol. The enhancement of PTP by estradiol is mediated distinctively through the G-protein estrogen receptor, GPER.
Collapse
Affiliation(s)
- Gengyun Le
- Division of Rehabilitation Science and Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, United States
| | - Cory W Baumann
- Division of Rehabilitation Science and Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, United States
| | - Gordon L Warren
- Department of Physical Therapy, Georgia State University, Atlanta, Georgia, United States
| | - Dawn A Lowe
- Division of Rehabilitation Science and Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, United States
| |
Collapse
|
23
|
Roles of Estrogen, Estrogen Receptors, and Estrogen-Related Receptors in Skeletal Muscle: Regulation of Mitochondrial Function. Int J Mol Sci 2023; 24:ijms24031853. [PMID: 36768177 PMCID: PMC9916347 DOI: 10.3390/ijms24031853] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/13/2023] [Accepted: 01/14/2023] [Indexed: 01/19/2023] Open
Abstract
Estrogen is an essential sex steroid hormone that functions primarily in female reproductive system, as well as in a variety of tissues and organs with pleiotropic effects, such as in cardiovascular, nervous, immune, and musculoskeletal systems. Women with low estrogen, as exemplified by those in postmenopause, are therefore prone to suffer from various disorders, i.e., cardiovascular disease, dementia, metabolic syndrome, osteoporosis, sarcopenia, frailty, and so on. Estrogen regulates the expression of its target genes by binding to its cognate receptors, estrogen receptors (ERs) α and β. Notably, the estrogen-related receptors (ERRs) α, β, and γ are originally identified as orphan receptors that share substantial structural homology and common transcriptional targets with ERs. Accumulating evidence suggests that ERs and ERRs play crucial roles in skeletal muscles, such as muscle mass maintenance, muscle exercise physiology, and muscle regeneration. In this article, we review potential regulatory roles of ERs and ERRs in muscle physiology, particularly with regard to mitochondrial function and metabolism.
Collapse
|
24
|
Zhang S, Ma J, Wang X, Zhao D, Zhang J, Jiang L, Duan W, Wang X, Hong Z, Li Z, Liu J. GPR30 Alleviates Pressure Overload-Induced Myocardial Hypertrophy in Ovariectomized Mice by Regulating Autophagy. Int J Mol Sci 2023; 24:ijms24020904. [PMID: 36674423 PMCID: PMC9867279 DOI: 10.3390/ijms24020904] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/26/2022] [Accepted: 12/28/2022] [Indexed: 01/06/2023] Open
Abstract
The incidence of heart failure mainly resulting from cardiac hypertrophy and fibrosis increases sharply in post-menopausal women compared with men at the same age, which indicates a cardioprotective role of estrogen. Previous studies in our group have shown that the novel estrogen receptor G Protein Coupled Receptor 30 (GPR30) could attenuate myocardial fibrosis caused by ischemic heart disease. However, the role of GPR30 in myocardial hypertrophy in ovariectomized mice has not been investigated yet. In this study, female mice with bilateral ovariectomy or sham surgery underwent transverse aortic constriction (TAC) surgery. After 8 weeks, mice in the OVX + TAC group exhibited more severe myocardial hypertrophy and fibrosis than mice in the TAC group. G1, the specific agonist of GPR30, could attenuate myocardial hypertrophy and fibrosis of mice in the OVX + TAC group. Furthermore, the expression of LC3II was significantly higher in the OVX + TAC group than in the OVX + TAC + G1 group, which indicates that autophagy might play an important role in this process. An in vitro study showed that G1 alleviated AngiotensionII (AngII)-induced hypertrophy and reduced the autophagy level of H9c2 cells, as revealed by LC3II expression and tandem mRFP-GFP-LC3 fluorescence analysis. Additionally, Western blot results showed that the AKT/mTOR pathway was inhibited in the AngII group, whereas it was restored in the AngII + G1 group. To further verify the mechanism, PI3K inhibitor LY294002 or autophagy activator rapamycin was added in the AngII + G1 group, and the antihypertrophy effect of G1 on H9c2 cells was blocked by LY294002 or rapamycin. In summary, our results demonstrate that G1 can attenuate cardiac hypertrophy and fibrosis and improve the cardiac function of mice in the OVX + TAC group through AKT/mTOR mediated inhibition of autophagy. Thus, this study demonstrates a potential option for the drug treatment of pressure overload-induced cardiac hypertrophy in postmenopausal women.
Collapse
|
25
|
Muhammad A, Forcados GE, Yusuf AP, Abubakar MB, Sadiq IZ, Elhussin I, Siddique MAT, Aminu S, Suleiman RB, Abubakar YS, Katsayal BS, Yates CC, Mahavadi S. Comparative G-Protein-Coupled Estrogen Receptor (GPER) Systems in Diabetic and Cancer Conditions: A Review. Molecules 2022; 27:molecules27248943. [PMID: 36558071 PMCID: PMC9786783 DOI: 10.3390/molecules27248943] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/05/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022] Open
Abstract
For many patients, diabetes Mellitus and Malignancy are frequently encountered comorbidities. Diabetes affects approximately 10.5% of the global population, while malignancy accounts for 29.4 million cases each year. These troubling statistics indicate that current treatment approaches for these diseases are insufficient. Alternative therapeutic strategies that consider unique signaling pathways in diabetic and malignancy patients could provide improved therapeutic outcomes. The G-protein-coupled estrogen receptor (GPER) is receiving attention for its role in disease pathogenesis and treatment outcomes. This review aims to critically examine GPER' s comparative role in diabetes mellitus and malignancy, identify research gaps that need to be filled, and highlight GPER's potential as a therapeutic target for diabetes and malignancy management. There is a scarcity of data on GPER expression patterns in diabetic models; however, for diabetes mellitus, altered expression of transport and signaling proteins has been linked to GPER signaling. In contrast, GPER expression in various malignancy types appears to be complex and debatable at the moment. Current data show inconclusive patterns of GPER expression in various malignancies, with some indicating upregulation and others demonstrating downregulation. Further research should be conducted to investigate GPER expression patterns and their relationship with signaling pathways in diabetes mellitus and various malignancies. We conclude that GPER has therapeutic potential for chronic diseases such as diabetes mellitus and malignancy.
Collapse
Affiliation(s)
- Aliyu Muhammad
- Center for Cancer Research, Department of Biology, Tuskegee University, Tuskegee, AL 36088, USA
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria P.M.B. 1044, Nigeria
| | | | - Abdurrahman Pharmacy Yusuf
- Department of Biochemistry, School of Life Sciences, Federal University of Technology, Minna P.M.B. 65, Nigeria
| | - Murtala Bello Abubakar
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Usmanu Danfodiyo University, Sokoto P.M.B. 2254, Nigeria
- Centre for Advanced Medical Research & Training (CAMRET), Usmanu Danfodiyo University, Sokoto P.M.B. 2254, Nigeria
| | - Idris Zubairu Sadiq
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria P.M.B. 1044, Nigeria
| | - Isra Elhussin
- Center for Cancer Research, Department of Biology, Tuskegee University, Tuskegee, AL 36088, USA
| | - Md Abu Talha Siddique
- Center for Cancer Research, Department of Biology, Tuskegee University, Tuskegee, AL 36088, USA
| | - Suleiman Aminu
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria P.M.B. 1044, Nigeria
| | - Rabiatu Bako Suleiman
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria P.M.B. 1044, Nigeria
| | - Yakubu Saddeeq Abubakar
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria P.M.B. 1044, Nigeria
| | - Babangida Sanusi Katsayal
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria P.M.B. 1044, Nigeria
| | - Clayton C Yates
- Center for Cancer Research, Department of Biology, Tuskegee University, Tuskegee, AL 36088, USA
| | - Sunila Mahavadi
- Center for Cancer Research, Department of Biology, Tuskegee University, Tuskegee, AL 36088, USA
| |
Collapse
|
26
|
Wang D, Li X, Zhang P, Cao Y, Zhang K, Qin P, Guo Y, Li Z, Tian Y, Kang X, Liu X, Li H. ELOVL gene family plays a virtual role in response to breeding selection and lipid deposition in different tissues in chicken (Gallus gallus). BMC Genomics 2022; 23:705. [PMID: 36253734 PMCID: PMC9575239 DOI: 10.1186/s12864-022-08932-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 10/10/2022] [Indexed: 11/11/2022] Open
Abstract
Background Elongases of very long chain fatty acids (ELOVLs), a family of first rate-limiting enzymes in the synthesis of long-chain fatty acids, play an essential role in the biosynthesis of complex lipids. Disrupting any of ELOVLs affects normal growth and development in mammals. Genetic variations in ELOVLs are associated with backfat or intramuscular fatty acid composition in livestock. However, the effects of ELOVL gene family on breeding selection and lipid deposition in different tissues are still unknown in chickens. Results Genetic variation patterns and genetic associations analysis showed that the genetic variations of ELOVL genes were contributed to breeding selection of commercial varieties in chicken, and 14 SNPs in ELOVL2-6 were associated with body weight, carcass or fat deposition traits. Especially, one SNP rs17631638T > C in the promoter of ELOVL3 was associated with intramuscular fat content (IMF), and its allele frequency was significantly higher in native and layer breeds compared to that in commercial broiler breeds. Quantitative real-time PCR (qRT-PCR) determined that the ELOVL3 expressions in pectoralis were affected by the genotypes of rs17631638T > C. In addition, the transcription levels of ELOVL genes except ELOVL5 were regulated by estrogen in chicken liver and hypothalamus with different regulatory pathways. The expression levels of ELOVL1-6 in hypothalamus, liver, abdominal fat and pectoralis were correlated with abdominal fat weight, abdominal fat percentage, liver lipid content and IMF. Noteworthily, expression of ELOVL3 in pectoralis was highly positively correlated with IMF and glycerophospholipid molecules, including phosphatidyl choline, phosphatidyl ethanolamine, phosphatidyl glycerol and phospholipids inositol, rich in ω-3 and ω-6 long-chain unsaturated fatty acids, suggesting ELOVL3 could contribute to intramuscular fat deposition by increasing the proportion of long-chain unsaturated glycerophospholipid molecules in pectoralis. Conclusions In summary, we demonstrated the genetic contribution of ELOVL gene family to breeding selection for specialized varieties, and revealed the expression regulation of ELOVL genes and their potential roles in regulating lipid deposition in different tissues. This study provides new insights into understanding the functions of ELOVL family on avian growth and lipid deposition in different tissues and the genetic variation in ELOVL3 may aid the marker-assisted selection of meat quality in chicken. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-022-08932-8.
Collapse
Affiliation(s)
- Dandan Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China
| | - Xinyan Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China
| | - Panpan Zhang
- Henan Institute of Veterinary Drug and Feed Control, Zhengzhou, 450002, China
| | - Yuzhu Cao
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China
| | - Ke Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China
| | - Panpan Qin
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China
| | - Yulong Guo
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China
| | - Zhuanjian Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China.,Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou, 450046, China.,International Joint Research Laboratory for Poultry Breeding of Henan, Zhengzhou, 450046, China
| | - Yadong Tian
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China.,Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou, 450046, China.,International Joint Research Laboratory for Poultry Breeding of Henan, Zhengzhou, 450046, China
| | - Xiangtao Kang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China.,Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou, 450046, China.,International Joint Research Laboratory for Poultry Breeding of Henan, Zhengzhou, 450046, China
| | - Xiaojun Liu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China. .,Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou, 450046, China. .,International Joint Research Laboratory for Poultry Breeding of Henan, Zhengzhou, 450046, China.
| | - Hong Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China. .,Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou, 450046, China. .,International Joint Research Laboratory for Poultry Breeding of Henan, Zhengzhou, 450046, China.
| |
Collapse
|
27
|
Brettle H, Tran V, Drummond GR, Franks AE, Petrovski S, Vinh A, Jelinic M. Sex hormones, intestinal inflammation, and the gut microbiome: Major influencers of the sexual dimorphisms in obesity. Front Immunol 2022; 13:971048. [PMID: 36248832 PMCID: PMC9554749 DOI: 10.3389/fimmu.2022.971048] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 08/16/2022] [Indexed: 11/13/2022] Open
Abstract
Obesity is defined as the excessive accumulation of body fat and is associated with an increased risk of developing major health problems such as cardiovascular disease, diabetes and stroke. There are clear sexual dimorphisms in the epidemiology, pathophysiology and sequelae of obesity and its accompanying metabolic disorders, with females often better protected compared to males. This protection has predominantly been attributed to the female sex hormone estrogen and differences in fat distribution. More recently, the sexual dimorphisms of obesity have also been attributed to the differences in the composition and function of the gut microbiota, and the intestinal immune system. This review will comprehensively summarize the pre-clinical and clinical evidence for these sexual dimorphisms and discuss the interplay between sex hormones, intestinal inflammation and the gut microbiome in obesity. Major gaps and limitations of this rapidly growing area of research will also be highlighted in this review.
Collapse
Affiliation(s)
- Holly Brettle
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
| | - Vivian Tran
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
| | - Grant R. Drummond
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
| | - Ashley E. Franks
- Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
| | - Steve Petrovski
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
| | - Antony Vinh
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
| | - Maria Jelinic
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
- *Correspondence: Maria Jelinic,
| |
Collapse
|
28
|
Wang D, Teng M, Wang Y, Cao Y, Tian W, Wang Z, Guo Y, Li H, Li Z, Jiang R, Li G, Tian Y, Liu X. GPNMB promotes abdominal fat deposition in chickens: genetic variation, expressional profile, biological function, and transcriptional regulation. Poult Sci 2022; 101:102216. [PMID: 36279606 PMCID: PMC9597125 DOI: 10.1016/j.psj.2022.102216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 09/23/2022] [Accepted: 09/26/2022] [Indexed: 11/30/2022] Open
Abstract
Glycoprotein nonmetastatic melanoma protein B (GPNMB) is a vital secreted factor that promotes the occurrence of obesity in mammals. However, the effects of GPNMB on abdominal fat deposition is still unknown in chickens. In this study, we looked into the genetic and expression association of GPNMB gene with abdominal fat traits in chicken, and found that a genetic variation rs31126482 in GPNMB promoter was significantly associated with abdominal fat weight (AFW, P < 0.05) and abdominal fat percentage (AFP, P < 0.01). Express profile analysis of the GPNMB indicated that the gene was mainly expressed in abdominal fat tissue, and its expression level was strongly positively correlated with AFW (R2 = 0.6356, P = 4.10E−05) and AFP (R2 = 0.6450, P = 2.90E−05). We then investigated biological function of GPNMB on adipogenesis in chicken, and found that GPNMB could inhibit abdominal preadipocyte proliferation, but promote abdominal preadipocyte differentiation and lipid deposition. Furthermore, we explored regulatory mechanism of GPNMB gene in chicken, and detected one nonclassical estrogen regulatory element (AP1) and one peroxisome proliferator-activated receptor α (PPARα) responsive element in the 2 kb promoter region of GPNMB gene, and demonstrated that estrogen could up-regulate GPNMB mRNA expression in adipose tissue and primary abdominal preadipocytes, while PPARα could down-regulate GPNMB expression in primary preadipocytes. Taken together, this study brings new insights into understanding the function and transcriptional control of GPNMB gene, and provides genetic markers for breeding selection to improve abdominal fat traits in chicken.
Collapse
|
29
|
PCSK9 deficiency results in a specific shedding of excess LDLR in female mice only: Role of hepatic cholesterol. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159217. [PMID: 35985474 DOI: 10.1016/j.bbalip.2022.159217] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/03/2022] [Accepted: 08/04/2022] [Indexed: 11/24/2022]
Abstract
PCSK9 promotes the lysosomal degradation of cell surface LDL receptor (LDLR). We analyzed how excess LDLR generated by PCSK9 deficiency is differently handled in male and female mice to possibly unveil the mechanism leading to the lower efficacy of PCSK9 mAb on LDL-cholesterol levels in women. Analysis of intact or ovariectomized PCSK9 knockout (KO) mice supplemented with placebo or 17β-estradiol (E2) demonstrated that female, but not male mice massively shed the soluble ectodomain of the LDLR in the plasma. Liver-specific PCSK9 KO or alirocumab-treated WT mice exhibit the same pattern. This shedding is distinct from the basal one and is inhibited by ZLDI-8, a metalloprotease inhibitor pointing at ADAM10/ADAM17. In PCSK9 KO female mice, ZLDI-8 raises by 80 % the LDLR liver content in a few hours. This specific shedding is likely cholesterol-dependent: it is prevented in PCSK9 KO male mice that exhibit low intra-hepatic cholesterol levels without activating SREBP-2, and enhanced by mevalonate or high cholesterol feeding, or by E2 known to stimulate cholesterol synthesis via the estrogen receptor-α. Liver transcriptomics demonstrates that critically low liver cholesterol in ovariectomized female or knockout male mice also hampers the cholesterol-dependent G2/M transition of the cell cycle. Finally, higher levels of shed LDLR were measured in the plasma of women treated with PCSK9 mAb. PCSK9 knockout female mice hormonally sustain cholesterol synthesis and shed excess LDLR, seemingly like women. In contrast, male mice rely on high surface LDLR to replenish their stocks, despite 80 % lower circulating LDL.
Collapse
|
30
|
Khaksari M, Raji-Amirhasani A, Bashiri H, Ebrahimi MN, Azizian H. Protective effects of combining SERMs with estrogen on metabolic parameters in postmenopausal diabetic cardiovascular dysfunction: The role of cytokines and angiotensin II. Steroids 2022; 183:109023. [PMID: 35358567 DOI: 10.1016/j.steroids.2022.109023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 03/23/2022] [Indexed: 12/20/2022]
Abstract
INTRODUCTION The beneficial effects of the administration of selective estrogen receptor modulators (SERMs) and estrogen (E2), alone or in combination with each other, have been reported in postmenopausal diabetic cardiovascular dysfunction. In the present study, we determined the mechanism of action of SERMs and E2 on inflammatory balance, angiotensin II (Ang II) serum levels, and glycemic profile in a postmenopausal diabetic rat model. METHODS Ovariectomized rats with type 2 diabetes received daily SERMs (tamoxifen and raloxifene) and E2 for one month. After treatment, cardiovascular risk indices, glycemic profile, and serum Ang II, TNF-α and IL-10 levels were measured. RESULTS Type 2 diabetes caused an abnormal glycemic profile, which was exacerbated by ovariectomy. All treatments inhibited the effects of diabetes and ovariectomy on the glycemic profile, with combined treatments (SERMs + E2) showing stronger effects. Cardiovascular risk indices that became abnormal by diabetes and worsened by ovariectomy were improved in all treatment modalities. Also, combined treatment reduced serum Ang II, TNF-α, and the ratio of TNF-α to IL-10, indicating an improvement in inflammatory balance. CONCLUSION Our study showed the administration of SERMs and E2, alone or in combination, could be an effective alternative in the treatment of menopausal diabetes, and generally, the beneficial effects of combined treatments were more effective than the effects of E2 or SERMs alone. It appears that E2 or SERMs benefit the cardiovascular system by improving inflammatory balance and reducing Ang II levels.
Collapse
Affiliation(s)
- Mohammad Khaksari
- Endocrinology and Metabolism Research Center, Kerman University of Medical Sciences, Kerman, Iran.
| | - Alireza Raji-Amirhasani
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Hamideh Bashiri
- Cardiovascular Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Hossein Azizian
- Neurobiomedical Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
31
|
Comparative effects of estrogen and silibinin on cardiovascular risk biomarkers in ovariectomized rats. Gene 2022; 823:146365. [PMID: 35257789 DOI: 10.1016/j.gene.2022.146365] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 02/14/2022] [Accepted: 02/18/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND Silibinin is a polyphenolic compound that could modulate estrogen receptor activation. Vascular dysfunction is considered a key initiator in atherosclerosis and may occur in the postmenopausal period. This manuscript compares estrogen and silibinin's impacts on factors that change endothelial function in ovariectomized (OVX) rats. METHODS 32 female Wistar rats were subdivided into control; OVX; OVX + estrogen (1 mg/kg/day); and OVX + silibinin (50 mg/kg/day) groups. After the experimental period, lipid profile, atherogenic indices, and histopathology of endothelium were monitored. The vascular oxidative stress, adhesion molecules, inflammatory cytokine levels, nitric oxide (NO), angiotensin-II (Ang-II), and endothelin-1 (ET-1) were also analyzed. RESULTS Silibinin treatment, similar to estrogen, significantly normalized the adverse changes of OVX on vascular function, including improved lipid profile and oxidative stress, increased endothelial nitric oxide synthase (eNOS) expression, diminished inflammatory status, and reduced adhesion molecule levels, ET-1 and Ang-II substances. Our findings also revealed that the administration with estrogen or silibinin resulted in a normal endothelium layer in the aorta tissues of OVX rats. CONCLUSION Estrogen and silibinin have similar effects in improving vascular function. These treatments' protective impacts on vasculature indicate their potential benefits on the cardiovascular system in the postmenopausal period.
Collapse
|
32
|
Bubb M, Beyer ASL, Dasgupta P, Kaemmerer D, Sänger J, Evert K, Wirtz RM, Schulz S, Lupp A. Assessment of G Protein-Coupled Oestrogen Receptor Expression in Normal and Neoplastic Human Tissues Using a Novel Rabbit Monoclonal Antibody. Int J Mol Sci 2022; 23:ijms23095191. [PMID: 35563581 PMCID: PMC9099907 DOI: 10.3390/ijms23095191] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/03/2022] [Accepted: 05/04/2022] [Indexed: 12/23/2022] Open
Abstract
In addition to the classical oestrogen receptors, ERα and ERβ, a G protein-coupled oestrogen receptor (GPER) has been identified that primarily mediates the rapid, non-genomic signalling of oestrogens. Data on GPER expression at the protein level are contradictory; therefore, the present study was conducted to re-evaluate GPER expression by immunohistochemistry to obtain broad GPER expression profiles in human non-neoplastic and neoplastic tissues, especially those not investigated in this respect so far. We developed and thoroughly characterised a novel rabbit monoclonal anti-human GPER antibody, 20H15L21, using Western blot analyses and immunocytochemistry. The antibody was then applied to a large series of formalin-fixed, paraffin-embedded human tissue samples. In normal tissue, GPER was identified in distinct cell populations of the cortex and the anterior pituitary; islets and pancreatic ducts; fundic glands of the stomach; the epithelium of the duodenum and gallbladder; hepatocytes; proximal tubules of the kidney; the adrenal medulla; and syncytiotrophoblasts and decidua cells of the placenta. GPER was also expressed in hepatocellular, pancreatic, renal, and endometrial cancers, pancreatic neuroendocrine tumours, and pheochromocytomas. The novel antibody 20H15L21 will serve as a valuable tool for basic research and the identification of GPER-expressing tumours during histopathological examinations.
Collapse
Affiliation(s)
- Maria Bubb
- Institute of Pharmacology and Toxicology, Jena University Hospital, 07747 Jena, Germany; (M.B.); (A.-S.L.B.); (P.D.); (S.S.)
| | - Anna-Sophia Lieselott Beyer
- Institute of Pharmacology and Toxicology, Jena University Hospital, 07747 Jena, Germany; (M.B.); (A.-S.L.B.); (P.D.); (S.S.)
| | - Pooja Dasgupta
- Institute of Pharmacology and Toxicology, Jena University Hospital, 07747 Jena, Germany; (M.B.); (A.-S.L.B.); (P.D.); (S.S.)
| | - Daniel Kaemmerer
- Department of General and Visceral Surgery, Zentralklinik Bad Berka, 99438 Bad Berka, Germany;
| | - Jörg Sänger
- Laboratory of Pathology and Cytology Bad Berka, 99438 Bad Berka, Germany;
| | - Katja Evert
- Department of Pathology, University of Regensburg, 93053 Regensburg, Germany;
- Institute of Pathology, University Medicine of Greifswald, 17475 Greifswald, Germany
| | - Ralph M. Wirtz
- STRATIFYER Molecular Pathology GmbH, 50935 Cologne, Germany;
| | - Stefan Schulz
- Institute of Pharmacology and Toxicology, Jena University Hospital, 07747 Jena, Germany; (M.B.); (A.-S.L.B.); (P.D.); (S.S.)
| | - Amelie Lupp
- Institute of Pharmacology and Toxicology, Jena University Hospital, 07747 Jena, Germany; (M.B.); (A.-S.L.B.); (P.D.); (S.S.)
- Correspondence: ; Tel.: +49-3641-9325678; Fax: +49-3641-9325652
| |
Collapse
|
33
|
Shi D, Li H, Zhang Z, He Y, Chen M, Sun L, Zhao P. Cryptotanshinone inhibits proliferation and induces apoptosis of breast cancer MCF-7 cells via GPER mediated PI3K/AKT signaling pathway. PLoS One 2022; 17:e0262389. [PMID: 35061800 PMCID: PMC8782479 DOI: 10.1371/journal.pone.0262389] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 12/23/2021] [Indexed: 12/15/2022] Open
Abstract
G protein-coupled estrogen receptor (GPER) was reported to be a potential target in the breast cancer therapy. This study aimed to illuminate the function of GPER and its mediated PI3K/AKT pathway in cryptotanshinone (CPT) inducing cell apoptosis and antiproliferation effect on GPER positive breast cancer MCF-7 cells. Cell proliferation was tested by MTT assay. Apoptosis rates were tested by Annexin V-FITC/PI double staining and the cell cycle was researched by flow cytometry. Autodock vina was applied to make molecular docking between CPT or estradiol and GPER. siRNA technique and GPER specific agonist G-1 or antagonist G-15 were applied to verify the mediated function of GPER. Apoptosis and cell cycle related proteins, as well as the key proteins on PI3K/AKT signaling pathway were detected by western blot. The results indicated that CPT could exert antiproliferation effects by arresting cell cycle in G2/M phase and downregulating the expression of cyclin D, cyclin B and cyclin A. Besides, apoptosis induced by CPT was observed. CPT might be a novel GPER binding compounds. Significantly, suppression of PI3K/AKT signal transduction by CPT was further increased by G-1 and decreased by G-15. The study revealed that the effect of antiproliferation and apoptosis treating with CPT on MCF-7 cells might be through the downregulation of PI3K/AKT pathway mediated by activated GPER.
Collapse
Affiliation(s)
- Danning Shi
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Hongbo Li
- Department of Gynecology, Affiliated Hospital of Shaanxi University of Chinese Medicine, Shaanxi, 712000, China
| | - Zeye Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yueshuang He
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Meng Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Liping Sun
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Piwen Zhao
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
- * E-mail:
| |
Collapse
|
34
|
A high carbohydrate and soda diet influences metabolic variables in Wistar rats. Life Sci 2022; 291:120295. [PMID: 34998837 DOI: 10.1016/j.lfs.2021.120295] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/22/2021] [Accepted: 12/30/2021] [Indexed: 11/21/2022]
Abstract
AIMS High carbohydrate diet and carbonated soda consumption have individually been associated with metabolic dysfunction, with links to glucose and insulin homeostasis, affecting metabolic variables associated with feeding, satiety and adiposity. Our objective is to determine the combined effect of a high carbohydrate and carbonated soda diet on metabolic variables in male and female Wistar rats. MATERIALS AND METHODS Thirty-two female and male weanlings were equally divided into four dietary groups; Control, Soda, High Carbohydrate diet (HCD), and High Carbohydrate diet/Soda (HCD/Soda), and fed ad libitum for fourteen weeks. Bodyweight, thoracic circumference, abdominal circumference and glucose was determined; Insulin, leptin, adiponectin, Tumor Necrotic Factor (TNF)-α, Interleukin (IL)-6 and lipid profile were assayed and used to determine the metabolic effects. KEY FINDINGS Soda and HCD/Soda had increased body weight in male rats, while HCD-fed rats were reduced compared to respective controls. Abdominal circumference, total cholesterol and reduced HDL of Soda were elevated in both sexes. Although HCD/Soda groups had elevated abdominal circumference in both sexes, total cholesterol and reduced high-density lipoprotein (HDL) were both reduced in females. Insulin and malondialdehyde (MDA) concentrations in Soda-fed rats was significantly reduced, however, MDA was elevated in both sexes in HCD and HCD/Soda fed rats. Female HCD and HCD/Soda groups had a significant increase in glutathione (GSH) concentration and a significant reduction in catalase. TNF-α was increased in both Soda and HCD/Soda groups. SIGNIFICANCE The results of this study suggest that HCD and Soda consumption results in alteration in phenotype and variables impacting metabolism.
Collapse
|
35
|
Sharma G, Prossnitz ER. Assessment of Metabolic Regulation by Estrogen Receptors. Methods Mol Biol 2022; 2418:383-404. [PMID: 35119676 DOI: 10.1007/978-1-0716-1920-9_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Estrogens, predominantly 17β-estradiol (E2), are a class of steroid hormones critical for diverse functions in the body both during normal physiology and disease. Primary actions of E2 include reproduction and development of secondary sexual characteristics. In addition, E2 action is involved in the nervous, immune, vascular, muscular, skeletal, and endocrine systems, all of which contribute to multiple aspects of metabolism. The actions of E2 have traditionally been attributed to the classical nuclear estrogen receptors (ERα and ERβ) that largely mediate transcriptional/genomic activities. However, over the last decade, the G protein-coupled estrogen receptor (GPER/GPR30) has become recognized as a mediator of rapid as well as transcriptional actions of E2, employing both in vitro and in vivo approaches. Recent evidence strongly supports the role of GPER in metabolic regulation. Murine genetic knockout (KO) models and pharmacological tools (agonists and antagonists) represent important approaches to understand the mechanisms of E2 action in physiology and disease via GPER. Studies in cells and GPER KO mice have revealed functions for GPER in the regulation of body weight and metabolism. This chapter focuses on methods relevant for the evaluation of metabolic parameters in vivo, ex vivo, and in vitro. We have emphasized glucose homeostasis through the determination of glucose and insulin tolerance, pancreatic islet function, and glucose uptake. In addition, we describe methods of adipocyte isolation, differentiation of preadipocytes, and evaluation of mitochondrial function.
Collapse
Affiliation(s)
- Geetanjali Sharma
- Department of Internal Medicine, Division of Molecular Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, USA.
- Center of Biomedical Research Excellence in Autophagy, Inflammation and Metabolism, University of New Mexico Health Sciences Center, Albuquerque, NM, USA.
| | - Eric R Prossnitz
- Department of Internal Medicine, Division of Molecular Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, USA.
- Center of Biomedical Research Excellence in Autophagy, Inflammation and Metabolism, University of New Mexico Health Sciences Center, Albuquerque, NM, USA.
- University of New Mexico Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM, USA.
| |
Collapse
|
36
|
Britz J, Ojo E, Dhukhwa A, Saito T, Saido TC, Hascup ER, Hascup KN, Tischkau SA. Assessing Sex-Specific Circadian, Metabolic, and Cognitive Phenotypes in the AβPP/PS1 and APPNL-F/NL-F Models of Alzheimer's Disease. J Alzheimers Dis 2022; 85:1077-1093. [PMID: 34897085 PMCID: PMC8900657 DOI: 10.3233/jad-210629] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Circadian disruption has long been recognized as a symptom of Alzheimer's disease (AD); however, emerging data suggests that circadian dysfunction occurs early on in disease development, potentially preceding any noticeable cognitive deficits. OBJECTIVE This study compares the onset of AD in male and female wild type (C57BL6/J), transgenic (AβPP/PS1), and knock-in (APPNL-F/NL-F) AD mouse models from the period of plaque initiation (6 months) through 12 months. METHODS Rhythmic daily activity patterns, glucose sensitivity, cognitive function (Morris water maze, MWM), and AD pathology (plaques formation) were assessed. A comparison was made across sexes. RESULTS Sex-dependent hyperactivity in AβPP/PS1 mice was observed. In comparison to C57BL/6J animals, 6-month-old male AβPP/PS1 demonstrated nighttime hyperactivity, as did 12-month-old females. Female AβPP/PS1 animals performed significantly worse on a MWM task than AβPP/PS1 males at 12 months and trended toward increased plaque pathology. APPNL-F/NL-F 12-month-old males performed significantly worse on the MWM task compared to 12-month-old females. Significantly greater plaque pathology occurred in AβPP/PS1 animals as compared to APPNL-F/NL-F animals. Female AβPP/PS1 animals performed significantly worse than APPNL-F/NL-F animals in spatial learning and memory tasks, though this was reversed in males. CONCLUSION Taken together, this study provides novel insights into baseline sex differences, as well as characterizes baseline diurnal activity variations, in the AβPP/PS1 and APPNL-F/NL-F AD mouse models.
Collapse
Affiliation(s)
- Jesse Britz
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Emmanuel Ojo
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Asmita Dhukhwa
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Takaomi C. Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Saitama, Japan
| | - Erin R. Hascup
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA,Department of Neurology, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Kevin N. Hascup
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA,Department of Neurology, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Southern Illinois University School of Medicine, Springfield, IL, USA,Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Shelley A. Tischkau
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA,Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA,Correspondence to: Shelley A. Tischkau, PhD, 801 N. Rutledge, Room 3289, Springfield, IL 62794-9629, USA. Tel.: +1 217 840 6724;
| |
Collapse
|
37
|
Chen G, Liu W, Yan B. Breast Cancer MCF-7 Cell Spheroid Culture for Drug Discovery and Development. JOURNAL OF CANCER THERAPY 2022; 13:117-130. [PMID: 36311820 PMCID: PMC9611733 DOI: 10.4236/jct.2022.133009] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In vitro 3D cancer spheroids (tumoroids) exhibit a drug resistance profile similar to that found in solid tumors. 3D spheroid culture methods recreate more physiologically relevant microenvironments for cells. Therefore, these models are more appropriate for cancer drug screening. We have recently developed a protocol for MCF-7 cell spheroid culture, and used this method to test the effects of different types of drugs on this estrogen-dependent breast cancer cell spheroid. Our results demonstrated that MCF-7 cells can grow spheroid in medium using a low attachment plate. We managed to grow one spheroid in each well, and the spheroid can grow over a month, the size of the spheroid can grow over a hundred times in volume. Our targeted drug experimental results suggest that estrogen sulfotransferase, steroid sulfatase, and G protein-coupled estrogen receptor may play critical roles in MCF-7 cell spheroid growth, while estrogen receptors α and β may not play an essential role in MCF-7 spheroid growth. Organoids are the miniatures of in vivo tissues and reiterate the in vivo microenvironment of a specific organ, best fit for the in vitro studies of diseases and drug development. Tumoroid, developed from cancer cell lines or patients’ tumor tissue, is the best in vitro model of in vivo tumors. 3D spheroid technology will be the best future method for drug development of cancers and other diseases. Our reported method can be developed clinically to develop personalized drugs when the patient’s tumor tissues are used to develop a spheroid culture for drug screening.
Collapse
Affiliation(s)
- Guangping Chen
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, USA
- ,
| | - William Liu
- College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | - Bingfang Yan
- College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
- ,
| |
Collapse
|
38
|
Yuasa T, Takata Y, Aki N, Kunimi K, Satoh M, Nii M, Izumi Y, Otoda T, Hashida S, Osawa H, Aihara KI. Insulin receptor cleavage induced by estrogen impairs insulin signaling. BMJ Open Diabetes Res Care 2021; 9:9/2/e002467. [PMID: 34969688 PMCID: PMC8719150 DOI: 10.1136/bmjdrc-2021-002467] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 12/06/2021] [Indexed: 11/21/2022] Open
Abstract
INTRODUCTION Soluble insulin receptor (sIR), which is the ectodomain of insulin receptor (IR), is present in human plasma. Plasma sIR levels are positively correlated with blood glucose levels and negatively correlated with insulin sensitivity. An in vitro model of IR cleavage shows that extracellular calpain 2 directly cleaves IR, which generates sIR, and sequential cleavage of the IRβ subunit by γ-secretase impairs insulin signaling in a glucose concentration-dependent manner. Nevertheless, sIR levels vary among subjects with normal glucose levels. RESEARCH DESIGN AND METHODS We examined sIR levels of pregnant women throughout gestation. Using an in vitro model, we also investigated the molecular mechanisms of IR cleavage induced by estradiol. RESULTS In pregnant women, sIR levels were positively correlated with estrogen levels and significantly increased at late pregnancy independent of glucose levels. Using an in vitro model, estrogen elicited IR cleavage and impaired cellular insulin signaling. Estradiol-induced IR cleavage was inhibited by targeting of calpain 2 and γ-secretase. Estrogen exerted these biological effects via G protein-coupled estrogen receptor, and its selective ligand upregulated calpain 2 expression and promoted exosome secretion, which significantly increased extracellular calpain 2. Simultaneous stimulation of estrogen and high glucose levels had a synergic effect on IR cleavage. Metformin prevented calpain 2 release in exosomes and restored insulin signaling impaired by estrogen. CONCLUSIONS Estradiol-induced IR cleavage causes cellular insulin resistance, and its molecular mechanisms are shared with those by high glucose levels. sIR levels at late pregnancy are significantly elevated along with estrogen levels. Therefore, estradiol-induced IR cleavage is preserved in pregnant women and could be part of the etiology of insulin resistance in gestational diabetes mellitus and overt diabetes during pregnancy.
Collapse
Affiliation(s)
- Tomoyuki Yuasa
- Community Medicine and Medical Science, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Yasunori Takata
- Diabetes and Molecular Genetics, Ehime University Graduate School of Medicine, Toon, Japan
| | - Nanako Aki
- Internal Medicine, Anan Kyoei Hospital, Anan, Japan
| | - Kotaro Kunimi
- Obstetrics and Gynecology, Anan Kyoei Hospital, Anan, Japan
| | - Miki Satoh
- Obstetrics and Gynecology, Anan Kyoei Hospital, Anan, Japan
| | - Mari Nii
- Obstetrics and Gynecology, Anan Kyoei Hospital, Anan, Japan
| | | | - Toshiki Otoda
- Community Medicine and Medical Science, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Seiichi Hashida
- Diabetes and Molecular Genetics, Ehime University Graduate School of Medicine, Toon, Japan
| | - Haruhiko Osawa
- Diabetes and Molecular Genetics, Ehime University Graduate School of Medicine, Toon, Japan
| | - Ken-Ichi Aihara
- Community Medicine and Medical Science, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| |
Collapse
|
39
|
Tong Y, Xu S, Huang L, Chen C. Obesity and insulin resistance: Pathophysiology and treatment. Drug Discov Today 2021; 27:822-830. [PMID: 34767960 DOI: 10.1016/j.drudis.2021.11.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 09/27/2021] [Accepted: 11/01/2021] [Indexed: 12/15/2022]
Abstract
The prevalence of obesity is a major cause of many chronic metabolic disorders, including type 2 diabetes mellitus (T2DM), cardiovascular disease (CVD), and cancer. Insulin resistance is often associated with metabolic unhealthy obesity (MUO). Therapeutic approaches aiming to improve insulin sensitivity are believed to be central for the prevention and treatment of MUO. However, current antiobesity drugs are reported as multitargeted and their insulin-sensitizing effects remain unclear. In this review, we discuss current understanding of the mechanisms of insulin resistance from the aspects of endocrine disturbance, inflammation, oxidative, and endoplasmic reticulum stress (ERS). We then summarize the antiobesity drugs, focusing on their effects on insulin sensitivity. Finally, we discuss strategies for obesity treatment.
Collapse
Affiliation(s)
- Yue Tong
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Sai Xu
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Lili Huang
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD 4072, Australia.
| | - Chen Chen
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
40
|
Chen G, Zeng H, Li X, Liu J, Li Z, Xu R, Ma Y, Liu C, Xue B. Activation of G protein coupled estrogen receptor prevents chemotherapy-induced intestinal mucositis by inhibiting the DNA damage in crypt cell in an extracellular signal-regulated kinase 1- and 2- dependent manner. Cell Death Dis 2021; 12:1034. [PMID: 34718327 PMCID: PMC8557214 DOI: 10.1038/s41419-021-04325-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 10/12/2021] [Accepted: 10/14/2021] [Indexed: 12/13/2022]
Abstract
Chemotherapy-induced intestinal mucositis (CIM) is a common adverse reaction to antineoplastic treatment with few appropriate, specific interventions. We aimed to identify the role of the G protein coupled estrogen receptor (GPER) in CIM and its mechanism. Adult male C57BL/6 mice were intraperitoneally injected with 5-fluorouracil to establish the CIM model. The selective GPER agonist G-1 significantly inhibited weight loss and histological damage in CIM mice and restored mucosal barrier dysfunction, including improving the expression of ZO-1, increasing the number of goblet cells, and decreasing mucosal permeability. Moreover, G-1 treatment did not alter the antitumor effect of 5-fluorouracil. In the CIM model, G-1 therapy reduced the expression of proapoptotic protein and cyclin D1 and cyclin B1, reversed the changes in the number of TUNEL+ cells, Ki67+ and bromodeoxyuridine+ cells in crypts. The selective GPER antagonist G15 eliminated all of the above effects caused by G-1 on CIM, and application of G15 alone increased the severity of CIM. GPER was predominantly expressed in ileal crypts, and G-1 inhibited the DNA damage induced by 5-fluorouracil in vivo and vitro, as confirmed by the decrease in the number of γH2AX+ cells in the crypts and the comet assay results. Referring to the data from GEO dataset we verified GPER activation restored ERK1/2 activity in CIM and 5-fluorouracil-treated IEC-6 cells. Once the effects of G-1 on ERK1/2 activity were abolished with the ERK1/2 inhibitor PD0325901, the effects of G-1 on DNA damage both in vivo and in vitro were eliminated. Correspondingly, all of the manifestations of G-1 protection against CIM were inhibited by PD0325901, such as body weight and histological changes, the mucosal barrier, the apoptosis and proliferation of crypt cells. In conclusion, GPER activation prevents CIM by inhibiting crypt cell DNA damage in an ERK1/2-dependent manner, suggesting GPER might be a target preventing CIM.
Collapse
Affiliation(s)
- Guanyu Chen
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Honghui Zeng
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xinyun Li
- The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University, Jinan, China
| | - Jianbo Liu
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhao Li
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Runze Xu
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yuntao Ma
- Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Chuanyong Liu
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bing Xue
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
41
|
Alemany M. Estrogens and the regulation of glucose metabolism. World J Diabetes 2021; 12:1622-1654. [PMID: 34754368 PMCID: PMC8554369 DOI: 10.4239/wjd.v12.i10.1622] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/10/2021] [Accepted: 04/14/2021] [Indexed: 02/06/2023] Open
Abstract
The main estrogens: estradiol, estrone, and their acyl-esters have been studied essentially related to their classical estrogenic and pharmacologic functions. However, their main effect in the body is probably the sustained control of core energy metabolism. Estrogen nuclear and membrane receptors show an extraordinary flexibility in the modulation of metabolic responses, and largely explain gender and age differences in energy metabolism: part of these mechanisms is already sufficiently known to justify both. With regard to energy, the estrogen molecular species act essentially through four key functions: (1) Facilitation of insulin secretion and control of glucose availability; (2) Modulation of energy partition, favoring the use of lipid as the main energy substrate when more available than carbohydrates; (3) Functional protection through antioxidant mechanisms; and (4) Central effects (largely through neural modulation) on whole body energy management. Analyzing the different actions of estrone, estradiol and their acyl esters, a tentative classification based on structure/effects has been postulated. Either separately or as a group, estrogens provide a comprehensive explanation that not all their quite diverse actions are related solely to specific molecules. As a group, they constitute a powerful synergic action complex. In consequence, estrogens may be considered wardens of energy homeostasis.
Collapse
Affiliation(s)
- Marià Alemany
- Faculty of Biology, University of Barcelona, Barcelona 08028, Catalonia, Spain
| |
Collapse
|
42
|
Xu L, Xu Y, Zhu Z, Gu H, Chen C, Chen J. Tanshinone IIA attenuates renal injury during hypothermic preservation via the MEK/ERK1/2/GSK-3β pathway. BMC Complement Med Ther 2021; 21:257. [PMID: 34625061 PMCID: PMC8501657 DOI: 10.1186/s12906-021-03427-7] [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: 01/28/2021] [Accepted: 09/29/2021] [Indexed: 12/25/2022] Open
Abstract
Background Oxidative stress-induced injury during hypothermic preservation is a universal problem that delays graft function and decrease the success of organ transplantation. Tanshinone IIA (Tan IIA) was reported to exhibit a variety of biochemical activities, including protection against oxidative stress. Therefore, the specific molecular pathway by which Tan IIA protects renal tissues during preservation was investigated in this study. Methods In vivo study, Sprague-Dawley (SD) rats were divided into twelve groups and the kidneys were isolated and preserved in different solutions for 0, 24 or 48 h, respectively: control group (Celsior solution) and Tan II groups (Celsior solution containing 10, 50,100 μM). In vitro study, primary renal cell from SD rats was cultured which was treated H2O2 (800 μM) for 6 h to mimic oxidative stress injury. Four groups were finally divided: control group; H2O2 group; H2O2 + Tan IIA group; H2O2 + Tan IIA + G15 group. Results In present study, we demonstrate data indicating that a significant increase in the superoxide dismutase (SOD) activity and a decrease in the reactive oxygen species (ROS) content were observed in the kidneys and renal cells preserved with Tan IIA compared with those preserved with the Celsior solution alone after 24 h and 48 h of hypothermic preservation (P < 0.01). The expression of phosphorylated mitogen-activated protein kinase kinase (MEK), phosphorylated extracellular signal-regulated protein kinases 1/2 (ERK1/2), phosphorylated glycogen synthase kinase-3β (GSK-3β) and cleaved caspase-3 was lower in the kidneys and renal cells preserved with Tan IIA than in those preserved with the Celsior solution alone after 24 h and 48 h of hypothermic preservation (P < 0.01). The mitochondrial morphology was rescued and adenosine triphophate (ATP) production and mitochondrial membrane potential were increased in the Tan IIA groups. Finally, Tan IIA also decreased cell apoptosis. Conclusion It suggests that the supplementation of the standard Celsior solution with Tan IIA may significantly improve long-term kidney preservation. Tan IIA attenuated oxidative stress injury and decreased apoptosis levels via activation of the MEK/ERK1/2/GSK-3β signaling pathway during kidney hypothermic preservation. Supplementary Information The online version contains supplementary material available at 10.1186/s12906-021-03427-7.
Collapse
Affiliation(s)
- Linhao Xu
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.,Translational Medicine Research Center, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.,School of Basic Medical Sciences and Forensic Medicine, Hangzhou medical college, No. 481 Binwen Road, Binjiang District, Hangzhou, 310053, Zhejiang, China
| | - Yizhou Xu
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Zhoujing Zhu
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, 310051, China
| | - Huiquan Gu
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou medical college, No. 481 Binwen Road, Binjiang District, Hangzhou, 310053, Zhejiang, China
| | - Chaofeng Chen
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Jian Chen
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou medical college, No. 481 Binwen Road, Binjiang District, Hangzhou, 310053, Zhejiang, China.
| |
Collapse
|
43
|
Yang S, Yin Z, Zhu G. A review of the functions of G protein-coupled estrogen receptor 1 in vascular and neurological aging. Eur J Pharmacol 2021; 908:174363. [PMID: 34297966 DOI: 10.1016/j.ejphar.2021.174363] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 07/11/2021] [Accepted: 07/19/2021] [Indexed: 02/07/2023]
Abstract
Aging-related diseases, especially vascular and neurological disorders cause huge economic burden. How to delay vascular and neurological aging is one of the insurmountable questions. G protein-coupled estrogen receptor 1 (GPER) has been extensively investigated in recent years due to its multiple biological responses. In this review, the function of GPER in aging-related diseases represented by vascular diseases, and neurological disorders were discussed. Apart from that, activation of GPER was also found to renovate the aging brain characterized by memory decline, but in a manner different from another two nuclear estrogen receptors estrogen receptor (ER)α and ERβ. This salutary effect would be better clarified from the aspects of synaptic inputs and transmission. Furthermore, we carefully described molecular mechanisms underpinning GPER-mediated effects. This review would update our understanding of GPER in the aging process. Targeting GPER may represent a promising strategy in the aging-related disorders.
Collapse
Affiliation(s)
- Shaojie Yang
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui, 230038, China
| | - Zhe Yin
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui, 230038, China
| | - Guoqi Zhu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui, 230038, China.
| |
Collapse
|
44
|
Azizian H, Khaksari M, Asadikaram G, Esmailidehaj M, Shahrokhi N. Progesterone eliminates 17β-estradiol-Mediated cardioprotection against diabetic cardiovascular dysfunction in ovariectomized rats. Biomed J 2021; 44:461-470. [PMID: 34507919 PMCID: PMC8514797 DOI: 10.1016/j.bj.2020.03.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/23/2020] [Accepted: 03/10/2020] [Indexed: 12/12/2022] Open
Abstract
Background Type2 Diabetes (T2D) remains one of the most important causes of cardiovascular diseases (CVD). Menopause leads to an increase in CVD and metabolic syndrome, which indicates the role of sex steroids as a protective factor. In the present study, we surveyed the effects of 17β-estradiol (E2) alone and in combination with progesterone (P4) on cardiovascular dysfunction in T2D. Methods Female ovariectomized (OVX) diabetic rats were divided into eight groups: Sham-Control, Diabetes (Dia), OVX + Dia, OVX + Dia + Vehicle, OVX + Dia + E2, OVX + Dia + P4, OVX + Dia + E2+P4, and OVX + Dia + E2+Vehicle. T2D was induced by a high-fat diet and streptozotocin. E2 and P4 were administrated every four days for four weeks. The heart cytokines and angiotensin II, lipid profile, insulin, water, and food intake and cardiovascular indices were measured. Results Results showed that single treatment with E2 decreased fasting blood glucose, water, and food intake, atherogenic and cardiac risk indices, and blood pressure. Also, P4 led to a decrease in atherogenic and cardiac risk indices. TNFα and IL-6 levels were increased and IL-10 was decreased in the Dia group, while E2 alone was able to inhibit these changes. The combined use of E2 and P4 eliminated the beneficial effects of E2 on these indices. Although diabetes results in an increment of cholesterol, LDL and triglyceride, hormone therapy with E2 was associated with improved dyslipidemia. Conclusion The use of E2 alone, and not the individual use of P4, and its combination with E2 improved cardiovascular function in OVX diabetic animals, possibly by reducing the amount of inflammatory cytokines and improving metabolic parameters.
Collapse
Affiliation(s)
- Hossein Azizian
- Neurobiomedical Research Center, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Department of Physiology, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Khaksari
- Endocrinology and Metabolism Research, and Physiology Research Centers, Kerman University of Medical Sciences, Kerman, Iran.
| | - Gholamreza Asadikaram
- Department of Biochemistry, and Metabolism & Endocrinology Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Mansour Esmailidehaj
- Neurobiomedical Research Center, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Nader Shahrokhi
- Department of Physiology, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
45
|
Robeva R, Mladenović D, Vesković M, Hrnčić D, Bjekić-Macut J, Stanojlović O, Livadas S, Yildiz BO, Macut D. The interplay between metabolic dysregulations and non-alcoholic fatty liver disease in women after menopause. Maturitas 2021; 151:22-30. [PMID: 34446275 DOI: 10.1016/j.maturitas.2021.06.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/18/2021] [Accepted: 06/26/2021] [Indexed: 12/12/2022]
Abstract
The hypoestrogenic period after menopause and associated metabolic imbalance might facilitate the onset of non-alcoholic fatty liver disease (NAFLD) and its progression. The prevalence of NAFLD increases in patients experiencing premature ovarian insufficiency, as well as surgical or natural menopause. The postmenopausal period is characterized by dyslipidemia and insulin resistance associated with an increased influx of free fatty acids to the liver with consequent steatosis and further progression of NAFLD. More than half of postmenopausal women with diabetes mellitus type 2 suffer from NAFLD. It is suggested that estrogens slow the progression of chronic liver diseases by suppression of inflammation, improvement of mitochondrial function, alleviation of oxidative stress, insulin resistance, and fibrogenesis. The hyperandrogenic state of polycystic ovary syndrome (PCOS) is associated with the development of NAFLD in women of reproductive age, but it is difficult to extend these findings to menopause due to inappropriate diagnosis of PCOS after menopause. Lifestyle intervention, including physical activity and dietary regimens, remains the first-line preventive and therapeutic option for NAFLD. There are contradictory reports on the use of menopausal hormonal therapy (MHT) and NAFLD. It is necessary to investigate the potential effects of estradiol dose, progesterone type, selective estrogen receptor modulators and tissue-selective estrogen complex compounds on NAFLD development and progression in postmenopausal women. The present review aims to explore the pathophysiological and clinical aspects of liver metabolic disturbances in women after menopause, focusing on the possible preventive and therapeutic strategies in NAFLD, including the potential role of MHT.
Collapse
Affiliation(s)
- Ralitsa Robeva
- Department of Endocrinology, USHATE "Acad. Iv. Penchev", Faculty of Medicine, Medical University-Sofia, Sofia, Bulgaria
| | - Dušan Mladenović
- Institute of Pathophysiology "Ljubodrag Buba Mihailović", Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Milena Vesković
- Institute of Pathophysiology "Ljubodrag Buba Mihailović", Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Dragan Hrnčić
- Institute of Medicinal Physiology "Richard Burian", Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Jelica Bjekić-Macut
- Department of Endocrinology, CHC Bežanijska kosa, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Olivera Stanojlović
- Institute of Medicinal Physiology "Richard Burian", Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | | | - Bulent O Yildiz
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Hacettepe University School of Medicine, Ankara, Turkey
| | - Djuro Macut
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Centre of Serbia, Faculty of Medicine, University of Belgrade, Dr Subotića 13, 11000 Belgrade, Serbia.
| |
Collapse
|
46
|
Initial observations describing a lack of endometrial estrogenicity in response to low-dose estrogen vaginal implants. Menopause 2021; 28:969-970. [PMID: 34313614 DOI: 10.1097/gme.0000000000001843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
47
|
The G-Protein-Coupled Membrane Estrogen Receptor Is Present in Horse Cryptorchid Testes and Mediates Downstream Pathways. Int J Mol Sci 2021; 22:ijms22137131. [PMID: 34281183 PMCID: PMC8269005 DOI: 10.3390/ijms22137131] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/22/2021] [Accepted: 06/29/2021] [Indexed: 12/18/2022] Open
Abstract
Cryptorchidism in horses is a commonly occurring malformation. The molecular basis of this pathology is not fully known. In addition, the origins of high intratesticular estrogen levels in horses remain obscure. In order to investigate the role of the G-protein-coupled membrane estrogen receptor (GPER) and establish histological and biochemical cryptorchid testis status, healthy and cryptorchid horse testes were subjected to scanning electron microscopy analysis, histochemical staining for total protein (with naphthol blue black; NBB), acid content (with toluidine blue O; TBO), and polysaccharide content (with periodic acid-Schiff; PAS). The expression of GPER was analyzed by immunohistochemistry and Western blot. GPER-mediated intracellular cAMP and calcium (Ca2+) signaling were measured immunoenzymatically or colorimetrically. Our data revealed changes in the distribution of polysaccharide content but not the protein and acid content in the cryptorchid testis. Polysaccharides seemed to be partially translocated from the interstitial compartment to the seminiferous tubule compartment. Moreover, the markedly decreased expression of GPER and GPER downstream molecules, cAMP and Ca2+, suggests their potential role in testis pathology. Increased estrogen levels in cryptorchid conditions may be linked to disturbed GPER signaling. We postulate that GPER is a prominent key player in testis development and function and may be used as a new biomarker of horse testis in health and disease.
Collapse
|
48
|
Carcinogenesis of Triple-Negative Breast Cancer and Sex Steroid Hormones. Cancers (Basel) 2021; 13:cancers13112588. [PMID: 34070471 PMCID: PMC8197527 DOI: 10.3390/cancers13112588] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 05/19/2021] [Accepted: 05/23/2021] [Indexed: 11/21/2022] Open
Abstract
Simple Summary Triple-negative breast cancer (TNBC) lacks all of three treatment targets (estrogen receptor-α, ER-α; progesterone receptor, PgR; and human epidermal growth factor receptor 2, HER2) and is usually associated with a poor clinical outcome; however, several sex steroid receptors, such as androgen receptor (AR), ER-β, and G-protein-coupled estrogen receptor, are frequently expressed and their biological and clinical importance has been suggested. Despite the structural similarity between sex steroid hormones (androgens and estrogens) or receptors (AR and ER-β), similar signaling mechanisms of these hormones, and the coexistence of these hormones and their receptors in TNBC in a clinical setting, most studies or reviews focused on only one of these receptors, and rarely reviewed them in a comprehensive way. In this review, the carcinogenic or pathobiological role of sex steroid hormones in TNBC is considered, focusing on common and differing features of hormone actions. Abstract Triple-negative breast cancer (TNBC) lacks an effective treatment target and is usually associated with a poor clinical outcome; however, hormone unresponsiveness, which is the most important biological characteristic of TNBC, only means the lack of nuclear estrogenic signaling through the classical estrogen receptor (ER), ER-α. Several sex steroid receptors other than ER-α: androgen receptor (AR), second ER, ER-β, and non-nuclear receptors represented by G-protein-coupled estrogen receptor (GPER), are frequently expressed in TNBC and their biological and clinical importance has been suggested by a large number of studies. Despite the structural similarity between each sex steroid hormone (androgens and estrogens) or each receptor (AR and ER-β), and similarity in the signaling mechanisms of these hormones, most studies or reviews focused on one of these receptors, and rarely reviewed them in a comprehensive way. Considering the coexistence of these hormones and their receptors in TNBC in a clinical setting, a comprehensive viewpoint would be important to correctly understand the association between the carcinogenic mechanism or pathobiology of TNBC and sex steroid hormones. In this review, the carcinogenic or pathobiological role of sex steroid hormones in TNBC is considered, focusing on the common and divergent features of the action of these hormones.
Collapse
|
49
|
Pepermans RA, Sharma G, Prossnitz ER. G Protein-Coupled Estrogen Receptor in Cancer and Stromal Cells: Functions and Novel Therapeutic Perspectives. Cells 2021; 10:cells10030672. [PMID: 33802978 PMCID: PMC8002620 DOI: 10.3390/cells10030672] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/08/2021] [Accepted: 03/10/2021] [Indexed: 02/06/2023] Open
Abstract
Estrogen is involved in numerous physiological and pathophysiological systems. Its role in driving estrogen receptor-expressing breast cancers is well established, but it also has important roles in a number of other cancers, acting both on tumor cells directly as well as in the function of multiple cells of the tumor microenvironment, including fibroblasts, immune cells, and adipocytes, which can greatly impact carcinogenesis. One of its receptors, the G protein-coupled estrogen receptor (GPER), has gained much interest over the last decade in both health and disease. Increasing evidence shows that GPER contributes to clinically observed endocrine therapy resistance in breast cancer while also playing a complex role in a number of other cancers. Recent discoveries regarding the targeting of GPER in combination with immune checkpoint inhibition, particularly in melanoma, have led to the initiation of the first Phase I clinical trial for the GPER-selective agonist G-1. Furthermore, its functions in metabolism and corresponding pathophysiological states, such as obesity and diabetes, are becoming more evident and suggest additional therapeutic value in targeting GPER for both cancer and other diseases. Here, we highlight the roles of GPER in several cancers, as well as in metabolism and immune regulation, and discuss the therapeutic value of targeting this estrogen receptor as a potential treatment for cancer as well as contributing metabolic and inflammatory diseases and conditions.
Collapse
Affiliation(s)
- Richard A. Pepermans
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (R.A.P.); (G.S.)
| | - Geetanjali Sharma
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (R.A.P.); (G.S.)
- Center of Biomedical Research Excellence in Autophagy, Inflammation and Metabolism, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - Eric R. Prossnitz
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (R.A.P.); (G.S.)
- Center of Biomedical Research Excellence in Autophagy, Inflammation and Metabolism, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
- University of New Mexico Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
- Correspondence: ; Tel.: +1-505-272-5647
| |
Collapse
|
50
|
Model JFA, Lima MV, Ohlweiler R, Lopes Vogt É, Rocha DS, Souza SKD, Türck P, Araújo ASDR, Vinagre AS. Liraglutide improves lipid and carbohydrate metabolism of ovariectomized rats. Mol Cell Endocrinol 2021; 524:111158. [PMID: 33444670 DOI: 10.1016/j.mce.2021.111158] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 12/30/2020] [Accepted: 12/31/2020] [Indexed: 12/16/2022]
Abstract
Considering that post-menopausal women and ovariectomized rodents develop obesity associated with increased visceral fat, this study was developed to investigate if liraglutide, a glucagon-like peptide 1 (GLP1) analogue, could improve the metabolism of estrogen (E2) deficient females. Wistar rats were ovariectomized (OVX), and subdivided in four groups: sham saline, sham liraglutide, OVX saline, and OVX liraglutide. After sixty days, metabolic parameters of blood, heart, liver, brown (BAT) and white adipose tissue (WAT) visceral depots, and, heart oxidative homeostasis, were evaluated. Castration increased the animals' body weight, the relative weight of the WAT depots, hepatic triglycerides and cardiac glycogen content. Liraglutide treatment reversed these effects, decreased WAT depots weight and increased glucose oxidation and lipogenesis in BAT and WAT. In addition, liraglutide enhanced adrenalin (A) lipolytic effect. These results indicate that liraglutide may be a promising treatment to restore lipid homeostasis and prevent weight gain associated with E2 deficiency.
Collapse
Affiliation(s)
| | - Matheus Vieira Lima
- Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Renata Ohlweiler
- Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Éverton Lopes Vogt
- Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Débora Santos Rocha
- Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Samir Khal de Souza
- Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Patrick Türck
- Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | | | - Anapaula Sommer Vinagre
- Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.
| |
Collapse
|