1
|
Ahmad I, Kaur M, Tyagi D, Singh TB, Kaur G, Afzal SM, Jauhar M. Exploring novel insights into the molecular mechanisms underlying Bisphenol A-induced toxicity: A persistent threat to human health. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2024; 108:104467. [PMID: 38763439 DOI: 10.1016/j.etap.2024.104467] [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: 02/14/2024] [Revised: 04/09/2024] [Accepted: 05/11/2024] [Indexed: 05/21/2024]
Abstract
Bisphenol A (BPA) is a ubiquitous industrial chemical used in the production of polycarbonate plastics and epoxy resins, found in numerous consumer products. Despite its widespread use, its potential adverse health effects have raised significant concerns. This review explores the molecular mechanisms and evidence-based literature underlying BPA-induced toxicities and its implications for human health. BPA is an endocrine-disrupting chemical (EDC) which exhibits carcinogenic properties by influencing various receptors, such as ER, AhR, PPARs, LXRs, and RARs. It induces oxidative stress and contributes to cellular dysfunction, inflammation, and DNA damage, ultimately leading to various toxicities including but not limited to reproductive, cardiotoxicity, neurotoxicity, and endocrine toxicity. Moreover, BPA can modify DNA methylation patterns, histone modifications, and non-coding RNA expression, leading to epigenetic changes and contribute to carcinogenesis. Overall, understanding molecular mechanisms of BPA-induced toxicity is crucial for developing effective strategies and policies to mitigate its adverse effects on human health.
Collapse
Affiliation(s)
- Israel Ahmad
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T. Road, Phagwara, Punjab, India.
| | - Mandeep Kaur
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T. Road, Phagwara, Punjab, India.
| | - Devansh Tyagi
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T. Road, Phagwara, Punjab, India.
| | - Tejinder Bir Singh
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T. Road, Phagwara, Punjab, India.
| | - Gurpreet Kaur
- School of Business Studies, Punjab Agricultural University, Ludhiana, Punjab, India.
| | - Shaikh Mohammad Afzal
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T. Road, Phagwara, Punjab, India.
| | - Mohsin Jauhar
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T. Road, Phagwara, Punjab, India.
| |
Collapse
|
2
|
Chang KH, Chen CM. The Role of NRF2 in Trinucleotide Repeat Expansion Disorders. Antioxidants (Basel) 2024; 13:649. [PMID: 38929088 PMCID: PMC11200942 DOI: 10.3390/antiox13060649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/20/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024] Open
Abstract
Trinucleotide repeat expansion disorders, a diverse group of neurodegenerative diseases, are caused by abnormal expansions within specific genes. These expansions trigger a cascade of cellular damage, including protein aggregation and abnormal RNA binding. A key contributor to this damage is oxidative stress, an imbalance of reactive oxygen species that harms cellular components. This review explores the interplay between oxidative stress and the NRF2 pathway in these disorders. NRF2 acts as the master regulator of the cellular antioxidant response, orchestrating the expression of enzymes that combat oxidative stress. Trinucleotide repeat expansion disorders often exhibit impaired NRF2 signaling, resulting in inadequate responses to excessive ROS production. NRF2 activation has been shown to upregulate antioxidative gene expression, effectively alleviating oxidative stress damage. NRF2 activators, such as omaveloxolone, vatiquinone, curcumin, sulforaphane, dimethyl fumarate, and resveratrol, demonstrate neuroprotective effects by reducing oxidative stress in experimental cell and animal models of these diseases. However, translating these findings into successful clinical applications requires further research. In this article, we review the literature supporting the role of NRF2 in the pathogenesis of these diseases and the potential therapeutics of NRF2 activators.
Collapse
Affiliation(s)
- Kuo-Hsuan Chang
- Department of Neurology, Chang Gung Memorial Hospital, Linkou Medical Center, Kueishan, Taoyuan 333, Taiwan;
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Chiung-Mei Chen
- Department of Neurology, Chang Gung Memorial Hospital, Linkou Medical Center, Kueishan, Taoyuan 333, Taiwan;
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| |
Collapse
|
3
|
Abstract
Heart failure (HF) is a significant public health problem worldwide. It has long been noted that premenopausal women, compared to postmenopausal women and men, have lower rates for developing this disease, as well as subsequent morbidity and mortality. This difference has been attributed to estrogen playing a cardioprotective role in these women, though exactly how it does so remains unclear. In this review, we examine the presence of estrogen receptors within the cardiovascular system, as well as the role they play behind the cardioprotective effect attributed to estrogen. Furthermore, we highlight the underlying mechanisms behind their alleviation of HF, as well as possible treatment approaches, such as hormone replacement therapy and exercise regimens, to manipulate these mechanisms in treating and preventing HF.
Collapse
Affiliation(s)
- Chenyue Qian
- The Second Clinical Medical College, Jinan University, Shenzhen, 518020, Guangdong, China
| | - Jingjin Liu
- The Second Clinical Medical College, Jinan University, Shenzhen, 518020, Guangdong, China.
- Department of Cardiology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China.
- Department of Cardiology, Shenzhen Cardiovascular Minimally Invasive Medical Engineering Technology Research and Development Center, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China.
- Shenzhen Key Laboratory of Stem Cell Research and Clinical Transformation, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China.
- Department of GeriatricsThe Second Clinical Medical CollegeThe First Affiliated Hospital, Shenzhen People's HospitalJinan UniversitySouthern University of Science and Technology), Shenzhen, 518020, Guangdong, People's Republic of China.
| | - Huadong Liu
- The Second Clinical Medical College, Jinan University, Shenzhen, 518020, Guangdong, China.
- Department of Cardiology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China.
- Department of Cardiology, Shenzhen Cardiovascular Minimally Invasive Medical Engineering Technology Research and Development Center, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China.
- Shenzhen Key Laboratory of Stem Cell Research and Clinical Transformation, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China.
- Department of GeriatricsThe Second Clinical Medical CollegeThe First Affiliated Hospital, Shenzhen People's HospitalJinan UniversitySouthern University of Science and Technology), Shenzhen, 518020, Guangdong, People's Republic of China.
| |
Collapse
|
4
|
Dela Justina V, Miguez JSG, Priviero F, Sullivan JC, Giachini FR, Webb RC. Sex Differences in Molecular Mechanisms of Cardiovascular Aging. FRONTIERS IN AGING 2022; 2:725884. [PMID: 35822017 PMCID: PMC9261391 DOI: 10.3389/fragi.2021.725884] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/25/2021] [Indexed: 12/12/2022]
Abstract
Cardiovascular disease (CVD) is still the leading cause of illness and death in the Western world. Cardiovascular aging is a progressive modification occurring in cardiac and vascular morphology and physiology where increased endothelial dysfunction and arterial stiffness are observed, generally accompanied by increased systolic blood pressure and augmented pulse pressure. The effects of biological sex on cardiovascular pathophysiology have long been known. The incidence of hypertension is higher in men, and it increases in postmenopausal women. Premenopausal women are protected from CVD compared with age-matched men and this protective effect is lost with menopause, suggesting that sex-hormones influence blood pressure regulation. In parallel, the heart progressively remodels over the course of life and the pattern of cardiac remodeling also differs between the sexes. Lower autonomic tone, reduced baroreceptor response, and greater vascular function are observed in premenopausal women than men of similar age. However, postmenopausal women have stiffer arteries than their male counterparts. The biological mechanisms responsible for sex-related differences observed in cardiovascular aging are being unraveled over the last several decades. This review focuses on molecular mechanisms underlying the sex-differences of CVD in aging.
Collapse
Affiliation(s)
- Vanessa Dela Justina
- Graduate Program in Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | | | - Fernanda Priviero
- Cardiovascular Translational Research Center, University of South Carolina, Columbia, SC, United States
| | - Jennifer C Sullivan
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Fernanda R Giachini
- Graduate Program in Biological Sciences, Federal University of Goiás, Goiânia, Brazil.,Institute of Biological Sciences and Health, Federal University of Mato Grosso, Barra do Garças, Brazil
| | - R Clinton Webb
- Cardiovascular Translational Research Center, University of South Carolina, Columbia, SC, United States
| |
Collapse
|
5
|
Hu X, Li X, Deng P, Zhang Y, Liu R, Cai D, Xu Q, Jiang X, Sun J, Bai W. The consequence and mechanism of dietary flavonoids on androgen profiles and disorders amelioration. Crit Rev Food Sci Nutr 2022; 63:11327-11350. [PMID: 35796699 DOI: 10.1080/10408398.2022.2090893] [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] [Indexed: 11/03/2022]
Abstract
Androgen is a kind of steroid hormone that plays a vital role in reproductive system and homeostasis of the body. Disrupted androgen balance serves as the causal contributor to a series of physiological disorders and even diseases. Flavonoids, as an extremely frequent family of natural polyphenols, exist widely in plants and foods and have received great attention when considering their inevitable consumption and estrogen-like effects. Mounting evidence illustrates that flavonoids have a propensity to interfere with androgen synthesis and metabolism, and also have a designated improvement effect on androgen disorders. Therefore, flavonoids were divided into six subclasses based on the structural feature in this paper, and the literature about their effects on androgens published in the past ten years was summarized. It could be concluded that flavonoids have the potential to regulate androgen levels and biological effects, mainly by interfering with the hypothalamic-pituitary-gonadal axis, androgen synthesis and metabolism, androgen binding with its receptors and membrane receptors, and antioxidant effects. The faced challenges about androgen regulation by flavonoids masterly include target mechanism exploration, individual heterogeneity, food matrixes interaction, and lack of clinical study. This review also provides a scientific basis for nutritional intervention using flavonoids to improve androgen disorder symptoms.
Collapse
Affiliation(s)
- Xiang Hu
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Jinan University, Guangzhou, PR China
| | - Xusheng Li
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Jinan University, Guangzhou, PR China
| | - Pan Deng
- College of Pharmaceutical Sciences, Soochow University, Suzhou, PR China
| | - Yulin Zhang
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Jinan University, Guangzhou, PR China
| | - Ruijing Liu
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Jinan University, Guangzhou, PR China
- Key Laboratory for Bio-Based Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou, PR China
| | - Dongbao Cai
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Jinan University, Guangzhou, PR China
| | - Qingjie Xu
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Jinan University, Guangzhou, PR China
| | - Xinwei Jiang
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Jinan University, Guangzhou, PR China
| | - Jianxia Sun
- School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou, PR China
| | - Weibin Bai
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Jinan University, Guangzhou, PR China
| |
Collapse
|
6
|
Zhang H, Li Y, Zhang X, Chen W, Liang Q, Li C, Knibbs LD, Huang C, Wang Q. Potential occupational exposure of parents to endocrine disrupting chemicals, adverse birth outcomes, and the modification effects of multi-vitamins supplement and infant sex. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 233:113314. [PMID: 35189520 DOI: 10.1016/j.ecoenv.2022.113314] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 02/11/2022] [Accepted: 02/12/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Maternal occupational exposure to endocrine disrupting chemicals (EDCs) may have adverse effect on birth outcomes. However, little is known about paternal EDCs exposure and the combined effect of parental exposure on birth outcomes. OBJECTIVES To assess the effects of both maternal and paternal occupational EDCs exposure on adverse birth outcomes, and further explore if multi-vitamins supplement and infant sex modify the association. METHODS We conducted a prospective cohort study of 5421 mother-father-newborn groups in Guangzhou, China. A questionnaire informed by a job exposure matrix (JEM) was applied to collect parental occupational EDCs exposure based on the type of work performed. We used logistic regression to estimate association between parental EDCs exposure and birth outcomes (including preterm birth (PTB), low birth weight (LBW), birth defects and congenital heart defects (CHD)). Stratified analyses and Cochran Q tests were performed to assess the modifying effect of maternal multi-vitamins supplement use and infant sex. RESULTS Compared with mothers unexposed, we found that mothers those exposed to EDCs were associated with increased odds of birth defects (aOR=1.70, 95% confidence interval (CI): 1.10-2.62), especially for those exposed for > 1.5 years (aOR= 3.00, 95% CIs: 1.78-5.03), or those with directly occupational exposed to EDCs (aOR= 2.94, 95% CIs: 1.72-5.04). Maternal exposure for > 1.5 years and direct exposure increased the risk of CHD, with aORs of 2.47 (1.21-5.02) and 2.79 (1.37-5.69), respectively. Stronger adverse effects were also observed when mothers and fathers were both exposed to EDCs. Paternal occupational EDCs exposure and exposure ≤ 1.5 years was associated with increased odds of LBW, with aORs of 2.14 (1.63-2.79) and 1.54 (1.10-2.15), respectively. When stratified by multi-vitamins supplement and infant sex, we observed slightly stronger effects for maternal exposure on birth defects/CHD as well as paternal EDCs exposure on PTB and LBW, among those without multi-vitamins supplement and among male babies, although the modification effects were not significant. CONCLUSION Maternal exposure to EDCs was associated with greater odds of birth defects and CHD, while paternal exposure was mainly associated with greater odds of LBW. These effects tend to be stronger among mothers without multi-vitamins supplement and among male babies.
Collapse
Affiliation(s)
- Huanhuan Zhang
- School of Public Health, Sun Yat-sen University, Guangzhou, China; School of Public Health, Zhengzhou University, Zhengzhou, China
| | - Yanqiu Li
- Guangzhou Panyu Maternal Child Health Hospital (Guangzhou Panyu District He Xian Memorial Hospital), Guangzhou, China
| | - Xiaoxin Zhang
- School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Weiyi Chen
- Guangzhou Panyu Maternal Child Health Hospital (Guangzhou Panyu District He Xian Memorial Hospital), Guangzhou, China
| | - Qianhong Liang
- Guangzhou Panyu Maternal Child Health Hospital (Guangzhou Panyu District He Xian Memorial Hospital), Guangzhou, China
| | - Changchang Li
- Department of Sexually Transmitted Disease Prevention and Control, Dermatology Hospital of Southern Medical University, Guangzhou, China; Institute for Global Health and Sexually Transmitted Infections, Southern Medical University, Guangzhou, China
| | - Luke D Knibbs
- School of Public Health, The University of Sydney, NSW 2006, Australia
| | - Cunrui Huang
- School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Qiong Wang
- School of Public Health, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
7
|
Zulkifli S, Rahman AA, Kadir SHSA, Nor NSM. Bisphenol A and its effects on the systemic organs of children. Eur J Pediatr 2021; 180:3111-3127. [PMID: 33893858 DOI: 10.1007/s00431-021-04085-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/12/2021] [Accepted: 04/18/2021] [Indexed: 02/06/2023]
Abstract
For the past two decades, growing research has been pointing to multiple repercussions of bisphenol A (BPA) exposure to human health. BPA is a synthetic oestrogen which primarily targets the endocrine system; however, the compound also disturbs other systemic organ functions, in which the magnitude of impacts in those other systems is as comparable to those in the endocrine system. To date, the discoveries on the association between BPA and health outcomes mainly came from animal and in vitro studies, with limited human studies which emphasised on children's health. In this comprehensive review, we summarised studies on human, in vivo and in vitro models to understand the consequences of pre-, post- and perinatal BPA exposure on the perinatal, children and adult health, encompassing cardiovascular, neurodevelopmental, endocrine and reproductive effects.Conclusion: Evidence from in vitro and animal studies may provide further support and better understanding on the correlation between environmental BPA exposure and its detrimental effects in humans and child development, despite the difficulties to draw direct causal relations of BPA effects on the pathophysiology of the diseases/syndromes in children, due to differences in body system complexity between children and adults, as well as between animal and in vitro models and humans. What is known: • Very limited reviews are available on how BPA adversely affects children's health. • Previous papers mainly covered two systems in children. What is new: • Comprehensive review on the detrimental effects of BPA on children health outcomes, including expectations on adult health outcomes following perinatal BPA exposure, as well as covering a small part of BPA alternatives. • Essentially, BPA exposure during pregnancy has huge impacts on the foetus in which it may cause changes in foetal epigenetic programming, resulting in disease onsets during childhood as well as adulthood.
Collapse
Affiliation(s)
- Sarah Zulkifli
- Institute of Medical Molecular Biotechnology, Universiti Teknologi MARA (UiTM), Cawangan Selangor, Kampus Sungai Buloh, Jalan Hospital, 47000, Sungai Buloh, Malaysia
| | - Amirah Abdul Rahman
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Universiti Teknologi MARA (UiTM), Cawangan Selangor, Kampus Sungai Buloh, Jalan Hospital, 47000, Sungai Buloh, Malaysia
| | - Siti Hamimah Sheikh Abdul Kadir
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Universiti Teknologi MARA (UiTM), Cawangan Selangor, Kampus Sungai Buloh, Jalan Hospital, 47000, Sungai Buloh, Malaysia.,Institute for Pathology, Laboratory and Forensic Medicine (I-PPerForM), Universiti Teknologi MARA (UiTM), Cawangan Selangor, Kampus Sungai Buloh, Jalan Hospital, 47000, Sungai Buloh, Malaysia
| | - Noor Shafina Mohd Nor
- Institute for Pathology, Laboratory and Forensic Medicine (I-PPerForM), Universiti Teknologi MARA (UiTM), Cawangan Selangor, Kampus Sungai Buloh, Jalan Hospital, 47000, Sungai Buloh, Malaysia. .,Department of Paediatrics, Faculty of Medicine, Universiti Teknologi MARA (UiTM), Cawangan Selangor, Kampus Sungai Buloh, Jalan Hospital, 47000, Sungai Buloh, Malaysia.
| |
Collapse
|
8
|
Campesi I, Montella A, Franconi F. Human monocytes respond to lipopolysaccharide (LPS) stimulation in a sex-dependent manner. J Cell Physiol 2021; 237:580-588. [PMID: 34252202 PMCID: PMC9292909 DOI: 10.1002/jcp.30503] [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: 03/23/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 01/08/2023]
Abstract
Monocytes play a critical role in inflammation and immune response, their activity being sex‐dependent. However, the basis of sex differences is not well understood. Therefore, we investigated the lipopolysaccharide (LPS) effects on tumor necrosis factor‐α (TNF‐α) release, autophagy, and chemotaxis in freshly isolated monocytes from healthy young men and women. In basal conditions, male and female monocytes had similar TNF‐α release, chemotaxis, and estrogen receptors (ER‐α) and ER‐β expression, while the LC3II/I ratio was significantly higher in males. LPS treatment induced qualitative and quantitative sex differences. It reduced autophagy and increased TNF‐α release only in male monocytes, while, chemotaxis was significantly influenced only in female cells. Moreover, it reduced the expression of ER‐α only in female cells, while ER‐β expression was reduced in both sexes, but more markedly in female cells. Finally, the interplay between LPS treatment and 17‐β‐estradiol (E2) was present only in female cells. Globally, these findings expand the concept that sex plays a role in regulating monocytes' functions, being sex differences cell‐ and parameter‐specific.
Collapse
Affiliation(s)
- Ilaria Campesi
- Dipartimento di Scienze Biomediche, Università Degli Studi di Sassari, Sassari, Italy.,Laboratorio Nazionale di Medicina e Farmacologia di Genere, Istituto Nazionale Biostrutture Biosistemi, Sassari, Italy
| | - Andrea Montella
- Dipartimento di Scienze Biomediche, Università Degli Studi di Sassari, Sassari, Italy
| | - Flavia Franconi
- Laboratorio Nazionale di Medicina e Farmacologia di Genere, Istituto Nazionale Biostrutture Biosistemi, Sassari, Italy
| |
Collapse
|
9
|
Association between Phthalate Exposure and Frailty among Community-Dwelling Older Adults: A Repeated Panel Data Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18041985. [PMID: 33670787 PMCID: PMC7922338 DOI: 10.3390/ijerph18041985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/08/2021] [Accepted: 02/15/2021] [Indexed: 11/16/2022]
Abstract
Only a few studies have examined the impacts of environmental exposure on frailty. This study investigated the association between phthalates and frailty among community-dwelling older adults. The Korean Elderly Environmental Panel II (KEEP II) study is a repeated panel data study of 800 community-dwelling older adults in South Korea. Frailty was measured with five items defined by Fried and colleagues. Environmental pollutants in the form of two types of metabolites for Di-ethylhexyl phthalate (DEHPs)-Mono (2-ethyl-5-hydroxyhexyl) phthalate (MEHHP) and Mono (2-ethyl-5-oxohexyl) phthalate (MEOHP)-were obtained from urine specimens. Analyses were performed using repeated linear mixed models. The concentration levels of both MEOHP and MEHHP in urine were significantly higher in the pre-frail or frail group than its counterparts. While adjusting for covariates, MEOHP level was positively associated with the likelihood of being pre-frail or frail in both males and females; the concentration level of MEHHP also had a positive impact on the likelihood of being pre-frail or frail in females. The DEHP metabolite concentrations were significantly lower among adults with daily fruit consumption in both males and females. DEHPs, measured by metabolite concentrations, may increase the risk of frailty among older men and women; further studies are necessary. The preventive effects of nutrition on DEHP risk should also be further investigated.
Collapse
|
10
|
Begum M, Paul P, Das D, Chakraborty K, Bhattacharjee A, Ghosh S. Genes regulating development and behavior exhibited altered expression in Drosophila melanogaster exposed to bisphenol A: use of real-time quantitative PCR (qRT-PCR) and droplet digital PCR (ddPCR) in genotoxicity study. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:7090-7104. [PMID: 33025430 DOI: 10.1007/s11356-020-10805-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Accepted: 09/09/2020] [Indexed: 06/11/2023]
Abstract
Toxicity of bisphenol A on morphological and life-history traits of model insect Drosophila melanogaster was reported in our previous work. In the present study, we have analyzed the adversity of bisphenol A on the reproductive behavior of adult and on the expression of selected genes in the larva and adult stage of fruit fly exposed to bisphenol A (0.007 g/2 ml. or 3.5 mg/ml), in addition to determination of LC50 value of bisphenol A in larva and pupal stage. We employed both the quantitative reverse transcriptase PCR and droplet digital PCR for analyzing the expression profile of seven genes namely, decapentaplegic, vestigial, wingless, foraging, insulin-like receptor, doublesex, and fruitless. We found bisphenol A has more adverse effects on male sexual behavior than females. Moreover, we observed significant downregulation of all the selected genes in treated larvae except, fruitless in male where it showed significant upregulation. On contrary among the treated adult flies, significant downregulation of all target genes in both sexes is evident, except, doublesex and fruitless in males which showed significant upregulation. We did not observe any deviation of male: female sex ratio from 1:1 under bisphenol A exposure. All these results suggest bisphenol A adversely affects the optimum functioning of genes which are involved in the regulation of metabolic pathways, behavioral pattern, stress response, endocrine homeostasis, neural functioning, and the development of the specific organ in Drosophila melanogaster. Our result not only provides a foundation to study further the bisphenol A toxicity on different pivotal genes in Drosophila but also suggests the use of the droplet digital PCR technology in toxicity measurement at the molecular level in eukaryotic model systems.
Collapse
Affiliation(s)
- Morium Begum
- Department of Zoology, Cytogenetics & Genomics Research Unit, University of Calcutta, Taraknath-Palit-Siksha-Prangan (Ballygunge Science College Campus), 35, Ballygunge Circular Road.Kolkata, West Bengal, 700019, India
| | - Pallab Paul
- Department of Zoology, Cytogenetics & Genomics Research Unit, University of Calcutta, Taraknath-Palit-Siksha-Prangan (Ballygunge Science College Campus), 35, Ballygunge Circular Road.Kolkata, West Bengal, 700019, India
| | - Debasmita Das
- Department of Zoology, Cytogenetics & Genomics Research Unit, University of Calcutta, Taraknath-Palit-Siksha-Prangan (Ballygunge Science College Campus), 35, Ballygunge Circular Road.Kolkata, West Bengal, 700019, India
| | - Kaustav Chakraborty
- Amity Institute of Biotechnology, Amity-University Kolkata, Plot no 36, 37, and 38, Major Arterial Road (South-East), Action Area II, Newtown, Kolkata, West Bengal, 700135, India
| | - Ashima Bhattacharjee
- Amity Institute of Biotechnology, Amity-University Kolkata, Plot no 36, 37, and 38, Major Arterial Road (South-East), Action Area II, Newtown, Kolkata, West Bengal, 700135, India
| | - Sujay Ghosh
- Department of Zoology, Cytogenetics & Genomics Research Unit, University of Calcutta, Taraknath-Palit-Siksha-Prangan (Ballygunge Science College Campus), 35, Ballygunge Circular Road.Kolkata, West Bengal, 700019, India.
| |
Collapse
|
11
|
Human Umbilical Cord: Information Mine in Sex-Specific Medicine. Life (Basel) 2021; 11:life11010052. [PMID: 33451112 PMCID: PMC7828611 DOI: 10.3390/life11010052] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/08/2021] [Accepted: 01/12/2021] [Indexed: 12/12/2022] Open
Abstract
Biological differences between sexes should be considered in all stages of research, as sexual dimorphism starts in utero leading to sex-specific fetal programming. In numerous biomedical fields, there is still a lack of stratification by sex despite primary cultured cells retaining memory of the sex and of the donor. The sex of donors in biological research must be known because variations in cells and cellular components can be used as endpoints, biomarkers and/or targets of pharmacological studies. This selective review focuses on the current findings regarding sex differences observed in the umbilical cord, a widely used source of research samples, both in the blood and in the circulating cells, as well as in the different cellular models obtainable from it. Moreover, an overview on sex differences in fetal programming is reported. As it emerges that the sex variable is still often forgotten in experimental models, we suggest that it should be mandatory to adopt sex-oriented research, because only awareness of these issues can lead to innovative research.
Collapse
|
12
|
Zhang YF, Shan C, Wang Y, Qian LL, Jia DD, Zhang YF, Hao XD, Xu HM. Cardiovascular toxicity and mechanism of bisphenol A and emerging risk of bisphenol S. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 723:137952. [PMID: 32213405 DOI: 10.1016/j.scitotenv.2020.137952] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 02/24/2020] [Accepted: 03/13/2020] [Indexed: 05/20/2023]
Abstract
Epidemiological and animal studies indicate that increased exposure to bisphenol A (BPA) induces various human cardiovascular diseases (CVDs), including myocardial infarction, arrhythmias, dilated cardiomyopathy, atherosclerosis, and hypertension. Bisphenol S (BPS), an alternative to BPA, is increasingly present in various consumer products and human bodies worldwide. Recently, emerging evidence has shown that BPS might be related to cardiovascular disorders. In this review, we present striking evidence of the correlation between BPA exposure and various CVDs, and show that a nonmonotonic dose-response curve (NMDRC) was common in studies of the CV effects of BPA in vivo. The CV impairment induced by low doses of BPA should be highlighted, especially during developmental exposure or during coexposure with other risk factors. Furthermore, we explored the possible underlying mechanisms of these effects-particularly nuclear receptor signaling, ion channels, and epigenetic mechanisms-and the possible participation of lipid metabolism, oxidative stress and cell signaling. As the potential risks of BPA exposure in humans are still noteworthy, studies of BPA in CVDs should be strengthened, especially with respect to the mechanisms, prevention and treatment. Moreover, the potential CV risk of BPS reported by in vivo studies calls for immediate epidemiological investigations and animal studies to reveal the relationships of BPS and other BPA alternatives with human CVDs.
Collapse
Affiliation(s)
- Yin-Feng Zhang
- Institute for Translational Medicine, College of Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China.
| | - Chan Shan
- Institute for Translational Medicine, College of Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| | - Yu Wang
- Institute for Translational Medicine, College of Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| | - Li-Li Qian
- Institute for Translational Medicine, College of Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| | - Dong-Dong Jia
- Institute for Translational Medicine, College of Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| | - Yi-Fei Zhang
- Institute for Translational Medicine, College of Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| | - Xiao-Dan Hao
- Institute for Translational Medicine, College of Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| | - Hai-Ming Xu
- Department of Occupational and Environmental Medicine, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China.
| |
Collapse
|
13
|
La Rosa P, Petrillo S, Bertini ES, Piemonte F. Oxidative Stress in DNA Repeat Expansion Disorders: A Focus on NRF2 Signaling Involvement. Biomolecules 2020; 10:biom10050702. [PMID: 32369911 PMCID: PMC7277112 DOI: 10.3390/biom10050702] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 04/24/2020] [Accepted: 04/27/2020] [Indexed: 12/13/2022] Open
Abstract
DNA repeat expansion disorders are a group of neuromuscular and neurodegenerative diseases that arise from the inheritance of long tracts of nucleotide repetitions, located in the regulatory region, introns, or inside the coding sequence of a gene. Although loss of protein expression and/or the gain of function of its transcribed mRNA or translated product represent the major pathogenic effect of these pathologies, mitochondrial dysfunction and imbalance in redox homeostasis are reported as common features in these disorders, deeply affecting their severity and progression. In this review, we examine the role that the redox imbalance plays in the pathological mechanisms of DNA expansion disorders and the recent advances on antioxidant treatments, particularly focusing on the expression and the activity of the transcription factor NRF2, the main cellular regulator of the antioxidant response.
Collapse
|
14
|
Sickinghe AA, Korporaal SJA, den Ruijter HM, Kessler EL. Estrogen Contributions to Microvascular Dysfunction Evolving to Heart Failure With Preserved Ejection Fraction. Front Endocrinol (Lausanne) 2019; 10:442. [PMID: 31333587 PMCID: PMC6616854 DOI: 10.3389/fendo.2019.00442] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 06/19/2019] [Indexed: 12/14/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a syndrome involving microvascular dysfunction. No treatment is available yet and as the HFpEF patient group is expanding due to the aging population, more knowledge on dysfunction of the cardiac microvasculature is required. Endothelial dysfunction, impaired angiogenesis, (perivascular) fibrosis and the pruning of capillaries (rarefaction) may all contribute to microvascular dysfunction in the heart and other organs, e.g., the kidneys. The HFpEF patient group consists mainly of post-menopausal women and female sex itself is a risk factor for this syndrome. This may point toward a role of estrogen depletion after menopause in the development of HFpEF. Estrogens favor the ratio of vasodilating over vasoconstricting factors, which results in an overall lower blood pressure in women than in men. Furthermore, estrogens improve angiogenic capacity and attenuate (perivascular) fibrosis formation. Therefore, we hypothesize that the drop of estrogen levels after menopause contributes to myocardial microvascular dysfunction and renders post-menopausal women more vulnerable for heart diseases that involve the microvasculature. This review provides a detailed summary of molecular targets of estrogen, which might guide future research and treatment options.
Collapse
Affiliation(s)
| | | | | | - Elise L. Kessler
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
15
|
Gao F, Huang Y, Zhang L, Liu W. Involvement of estrogen receptor and GPER in bisphenol A induced proliferation of vascular smooth muscle cells. Toxicol In Vitro 2019; 56:156-162. [DOI: 10.1016/j.tiv.2019.01.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 01/02/2019] [Accepted: 01/21/2019] [Indexed: 01/01/2023]
|
16
|
Abstract
PURPOSE OF REVIEW To give an overview of recently published articles addressing the mechanisms underlying sex bias in autoimmune disease. RECENT FINDINGS Recent studies investigating the origins of sex bias in autoimmune disease have revealed an extensive and interconnected network of genetic, hormonal, microbial, and environmental influences. Investigation of sex hormones has moved beyond profiling the effects of hormones on activity and prevalence of immune cell types to defining the specific immunity-related genes driving these changes. Deeper examination of the genetic content of the X and Y chromosomes and genetic escapees of X chromosome inactivation has revealed some key drivers of female-biased autoimmunity. Animal studies are offering further insights into the connections among microbiota, particularly that of the gut, and the immune system. SUMMARY Sex bias in autoimmune disease is the manifestation of a complex interplay of the sex chromosomes, sex hormones, the microbiota, and additional environmental and sociological factors.
Collapse
|
17
|
Dai R, Edwards MR, Heid B, Ahmed SA. 17β-Estradiol and 17α-Ethinyl Estradiol Exhibit Immunologic and Epigenetic Regulatory Effects in NZB/WF1 Female Mice. Endocrinology 2019; 160:101-118. [PMID: 30418530 PMCID: PMC6305969 DOI: 10.1210/en.2018-00824] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Accepted: 11/06/2018] [Indexed: 02/07/2023]
Abstract
17α-Ethinyl estradiol (EE), a synthetic analog of natural estrogen 17β-estradiol (E2), is extensively used in hormonal contraceptives and estrogen replacement therapy, and it has also been found in sewage effluents. Given that E2 is a well-known immunomodulator, surprisingly there has been only limited information on the cellular and molecular immunologic consequences of exposure to EE. To address this fundamental gap, we directly compared the effects of EE with E2 on splenic leukocytes of New Zealand Black × New Zealand White F1 progeny (NZB/WF1) mice during the preautoimmune period. We found that EE and E2 have common, as well as distinctive, immunologic effects, with EE exposure resulting in more profound effects. Both EE and E2 increased numbers of splenic neutrophils, enhanced neutrophil serine proteases and myeloperoxidase expression, promoted the production of nitric oxide and monocyte chemoattractant protein-1, and altered adaptive immune T cell subsets. However, activation of splenic leukocytes through the T cell receptor or Toll-like receptor (TLR)4 revealed not only common (IL-10), but also hormone-specific alterations of cytokines (IFNγ, IL-1β, ΤΝFα, IL-2). Furthermore, in EE-exposed mice, TLR9 stimulation suppressed IFNα, in contrast to increased IFNα from E2-exposed mice. EE and E2 regulated common and hormone-specific expression of immune-related genes. Furthermore, EE exposure resulted in more marked alterations in miRNA expression levels than for E2. Only EE was able to reduce global DNA methylation significantly in splenic leukocytes. Taken together, our novel data revealed that EE and E2 exposure confers more similar effects in innate immune system-related cell development and responses, but has more differential regulatory effects in adaptive immune-related cell development and responses.
Collapse
Affiliation(s)
- Rujuan Dai
- Department of Biomedical Sciences and Pathobiology, Infectious Disease Research Facility (IDRF), Virginia-Maryland College of Veterinary Medicine, Virginia Tech/Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | - Michael R Edwards
- Department of Biomedical Sciences and Pathobiology, Infectious Disease Research Facility (IDRF), Virginia-Maryland College of Veterinary Medicine, Virginia Tech/Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | - Bettina Heid
- Department of Biomedical Sciences and Pathobiology, Infectious Disease Research Facility (IDRF), Virginia-Maryland College of Veterinary Medicine, Virginia Tech/Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | - S Ansar Ahmed
- Department of Biomedical Sciences and Pathobiology, Infectious Disease Research Facility (IDRF), Virginia-Maryland College of Veterinary Medicine, Virginia Tech/Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| |
Collapse
|
18
|
Put "gender glasses" on the effects of phenolic compounds on cardiovascular function and diseases. Eur J Nutr 2018; 57:2677-2691. [PMID: 29696400 DOI: 10.1007/s00394-018-1695-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 04/19/2018] [Indexed: 12/15/2022]
Abstract
INTRODUCTION The influence of sex and gender is particularly relevant in cardiovascular diseases (CVD) as well as in several aspects of drug pharmacodynamics and pharmacokinetics. Anatomical and physiological differences between the sexes may influence the activity of many drugs, including the possibility of their interaction with other drugs, bioactive compounds, foods and beverages. Phenolic compounds could interact with our organism at organ, cellular, and molecular levels triggering a preventive action against chronic diseases, including CVD. RESULTS This article will review the role of sex on the activity of these bioactive molecules, considering the existence of sex differences in oxidative stress. It describes the pharmacokinetics of phenolic compounds, their effects on vessels, on cardiovascular system, and during development, including the role of nuclear receptors and microbiota. CONCLUSIONS Although there is a large gap between the knowledge of the sex differences in the phenolic compounds' activity and safety, and the urgent need for more research, available data underlie the possibility that plant-derived phenolic compounds could differently influence the health of male and female subjects.
Collapse
|
19
|
Edwards M, Dai R, Ahmed SA. Our Environment Shapes Us: The Importance of Environment and Sex Differences in Regulation of Autoantibody Production. Front Immunol 2018; 9:478. [PMID: 29662485 PMCID: PMC5890161 DOI: 10.3389/fimmu.2018.00478] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 02/22/2018] [Indexed: 01/17/2023] Open
Abstract
Consequential differences exist between the male and female immune systems’ ability to respond to pathogens, environmental insults or self-antigens, and subsequent effects on immunoregulation. In general, females when compared with their male counterparts, respond to pathogenic stimuli and vaccines more robustly, with heightened production of antibodies, pro-inflammatory cytokines, and chemokines. While the precise reasons for sex differences in immune response to different stimuli are not yet well understood, females are more resistant to infectious diseases and much more likely to develop autoimmune diseases. Intrinsic (i.e., sex hormones, sex chromosomes, etc.) and extrinsic (microbiome composition, external triggers, and immune modulators) factors appear to impact the overall outcome of immune responses between sexes. Evidence suggests that interactions between environmental contaminants [e.g., endocrine disrupting chemicals (EDCs)] and host leukocytes affect the ability of the immune system to mount a response to exogenous and endogenous insults, and/or return to normal activity following clearance of the threat. Inherently, males and females have differential immune response to external triggers. In this review, we describe how environmental chemicals, including EDCs, may have sex differential influence on the outcome of immune responses through alterations in epigenetic status (such as modulation of microRNA expression, gene methylation, or histone modification status), direct and indirect activation of the estrogen receptors to drive hormonal effects, and differential modulation of microbial sensing and composition of host microbiota. Taken together, an intriguing question develops as to how an individual’s environment directly and indirectly contributes to an altered immune response, dysregulation of autoantibody production, and influence autoimmune disease development. Few studies exist utilizing well-controlled cohorts of both sexes to explore the sex differences in response to EDC exposure and the effects on autoimmune disease development. Translational studies incorporating multiple environmental factors in animal models of autoimmune disease are necessary to determine the interrelationships that occur between potential etiopathological factors. The presence or absence of autoantibodies is not a reliable predictor of disease. Therefore, future studies should incorporate all the susceptibility/influencing factors, coupled with individual genomics, epigenomics, and proteomics, to develop a model that better predicts, diagnoses, and treats autoimmune diseases in a personalized-medicine fashion.
Collapse
Affiliation(s)
- Michael Edwards
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Rujuan Dai
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - S Ansar Ahmed
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
20
|
Ventura-Clapier R, Dworatzek E, Seeland U, Kararigas G, Arnal JF, Brunelleschi S, Carpenter TC, Erdmann J, Franconi F, Giannetta E, Glezerman M, Hofmann SM, Junien C, Katai M, Kublickiene K, König IR, Majdic G, Malorni W, Mieth C, Miller VM, Reynolds RM, Shimokawa H, Tannenbaum C, D'Ursi AM, Regitz-Zagrosek V. Sex in basic research: concepts in the cardiovascular field. Cardiovasc Res 2018; 113:711-724. [PMID: 28472454 DOI: 10.1093/cvr/cvx066] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 05/01/2017] [Indexed: 01/01/2023] Open
Abstract
Women and men, female and male animals and cells are biologically different, and acknowledgement of this fact is critical to advancing medicine. However, incorporating concepts of sex-specific analysis in basic research is largely neglected, introducing bias into translational findings, clinical concepts and drug development. Research funding agencies recently approached these issues but implementation of policy changes in the scientific community is still limited, probably due to deficits in concepts, knowledge and proper methodology. This expert review is based on the EUGenMed project (www.eugenmed.eu) developing a roadmap for implementing sex and gender in biomedical and health research. For sake of clarity and conciseness, examples are mainly taken from the cardiovascular field that may serve as a paradigm for others, since a significant amount of knowledge how sex and oestrogen determine the manifestation of many cardiovascular diseases (CVD) has been accumulated. As main concepts for implementation of sex in basic research, the study of primary cell and animals of both sexes, the study of the influence of genetic vs. hormonal factors and the analysis of sex chromosomes and sex specific statistics in genome wide association studies (GWAS) are discussed. The review also discusses methodological issues, and analyses strength, weaknesses, opportunities and threats in implementing sex-sensitive aspects into basic research.
Collapse
Affiliation(s)
- Renée Ventura-Clapier
- Signalisation et Physiopathologie Cardiovasculaire UMR-S 1180, Inserm, Univ. Paris-Sud, Université Paris-Saclay, 92296 Châtenay-Malabry, France
| | - Elke Dworatzek
- Institute of Gender in Medicine and Center for Cardiovascular Research, Charité Universitaetsmedizin Berlin, 10115 Berlin, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Germany
| | - Ute Seeland
- Institute of Gender in Medicine and Center for Cardiovascular Research, Charité Universitaetsmedizin Berlin, 10115 Berlin, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Germany
| | - Georgios Kararigas
- Institute of Gender in Medicine and Center for Cardiovascular Research, Charité Universitaetsmedizin Berlin, 10115 Berlin, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Germany
| | - Jean-Francois Arnal
- Faculté Médecine Toulouse-Rangueil, Université de Toulouse, Toulouse, France
| | - Sandra Brunelleschi
- Department of Health Sciences, School of Medicine, University of Eastern Piedmont, Novara, Italy
| | - Thomas C Carpenter
- College of Medicine and Veterinary Medicine, University of Edinburgh, EH16 4TJ Edinburgh, UK
| | - Jeanette Erdmann
- Institut für Kardiogenetik, Universität zu Lübeck, 23562 Lübeck, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Germany
| | - Flavia Franconi
- Department of Biomedical Science, University of Sassari,Sassari, Italy
| | - Elisa Giannetta
- Ricercatore TD in Endocrinologia, Dipartimento di Medicina Sperimentale, Sezione di Fisiopatologia Medica, Sapienza University of Rome, Roma, Italy
| | - Marek Glezerman
- International Society for Gender Medicine, Research Center for Medicine, Rabin Medical Center, and Tel Aviv University, Israel
| | - Susanna M Hofmann
- Medizinische Klinik und Poliklinik IV, Klinikum der LMU München, Munich 80336, Germany; Institute for Diabetes and Regeneration, Helmholtz Center Munich, Germany; German Center for Diabetes Research (DZD) München-Neuherberg, Germany
| | - Claudine Junien
- BDR Biologie du Développement et Reproduction Developmental Biology and Reproduction UMR, INRA, France
| | - Miyuki Katai
- Section of Gender Medicine, Department of General Medicine, Tokyo Women's Medical University, 162-8666 Tokyo, Japan
| | - Karolina Kublickiene
- Centre for Gender Medicine and Departments of Obstetrics and Gynecology and Renal Medicine, Karolinska Institutet, 14186 Stockholm, Sweden
| | - Inke R König
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Germany.,Institut für Medizinische Biometrie und Statistik, Universität zu Lübeck, 235620 Lübeck, Germany
| | - Gregor Majdic
- Institute for Preclinical Sciences, Veterinary Faculty, University of Ljubljana & Institute of Physiology, Medical Faculty, University of Maribor, Maribor, Slovenia
| | - Walter Malorni
- National Center for Gender-Specific Medicine, Istituto Superiore di Sanità, 00161 Roma, Italy
| | - Christin Mieth
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Germany.,Max-Delbrück-Centrum für Molekulare Medizin in der Helmholtz-Gemeinschaft (MDC), Berlin, Germany
| | | | - Rebecca M Reynolds
- Center for Cardiovascular Science, Queen's Medical Research Institute, EH16 4TJ Edinburgh, UK
| | - Hiroaki Shimokawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Japan
| | - Cara Tannenbaum
- Institute of Gender and Health, Canadian Institutes of Health Research (CIHR), Canada
| | - Anna Maria D'Ursi
- Medicinal Chemistry DIFARMA, Università di Salerno, 84084 Fisciano, Italy
| | - Vera Regitz-Zagrosek
- Institute of Gender in Medicine and Center for Cardiovascular Research, Charité Universitaetsmedizin Berlin, 10115 Berlin, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Germany
| |
Collapse
|
21
|
Donoghue LJ, Neufeld TI, Li Y, Arao Y, Coons LA, Korach KS. Differential Activation of a Mouse Estrogen Receptor β Isoform (mERβ2) with Endocrine-Disrupting Chemicals (EDCs). ENVIRONMENTAL HEALTH PERSPECTIVES 2017; 125:634-642. [PMID: 27634370 PMCID: PMC5381991 DOI: 10.1289/ehp396] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 08/03/2016] [Accepted: 08/17/2016] [Indexed: 05/09/2023]
Abstract
BACKGROUND Endocrine-disrupting chemicals (EDCs) are suspected of altering estrogenic signaling through estrogen receptor (ER) α or β (mERβ1 in mice). Several EDC effects have been reported in animal studies and extrapolated to human studies. Unlike humans, rodents express a novel isoform of ERβ (mERβ2) with a modified ligand-binding domain sequence. EDC activity through this isoform remains uncharacterized. OBJECTIVES We identified the expression pattern of mERβ2 in mouse tissues and assessed the estrogenic activity of EDCs through mERβ2. METHODS mERβ2 mRNA expression was measured in mouse tissues. HepG2 cells were used to assess the transactivation activity of mERβ isoforms with EDCs and ER co-activators. 293A cells transiently transfected with mER isoforms were used to detect EDC-mediated changes in endogenous ER target gene expression. RESULTS Expression of mERβ2 mRNA was detected in mouse reproductive tissues (ovary, testis, and prostate) and lung and colon tissues from both female and male mice. Five (E2, DES, DPN, BPAF, Coum, 1-BP) of 16 compounds tested by reporter assay had estrogenic activity through mERβ2. mERβ2 had a compound-specific negative effect on ERβ/ligand-mediated activity and ER target genes when co-expressed with mERβ1. mERβ2 recruited coactivators SRC2 or SRC3 in the presence of EDCs, but showed less recruitment than mERβ1. CONCLUSION mERβ2 showed weaker estrogenic activity than mERβ1 in our in vitro system, and can dampen mERβ1 activity. In vivo models of EDC activity and ER-mediated toxicity should consider the role of mERβ2, as rodent tissue responses involving mERβ2 may not be reproduced in human biology.
Collapse
Affiliation(s)
| | | | | | | | | | - Kenneth S. Korach
- Address correspondence to K.S. Korach, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, 111 T.W. Alexander Dr., P.O. Box 12233, Research Triangle Park, NC 27709 USA. Telephone: (919) 541-3512. E-mail:
| |
Collapse
|
22
|
Marcoccia D, Pellegrini M, Fiocchetti M, Lorenzetti S, Marino M. Food components and contaminants as (anti)androgenic molecules. GENES AND NUTRITION 2017; 12:6. [PMID: 28239427 PMCID: PMC5312591 DOI: 10.1186/s12263-017-0555-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 01/23/2017] [Indexed: 01/14/2023]
Abstract
Androgens, the main male sex steroids, are the critical factors responsible for the development of the male phenotype during embryogenesis and for the achievement of sexual maturation and puberty. In adulthood, androgens remain essential for the maintenance of male reproductive function and behavior. Androgens, acting through the androgen receptor (AR), regulate male sexual differentiation during development, sperm production beginning from puberty, and maintenance of prostate homeostasis. Several substances present in the environment, now classified as endocrine disruptors (EDCs), strongly interfere with androgen actions in reproductive and non-reproductive tissues. EDCs are a heterogeneous group of xenobiotics which include synthetic chemicals used as industrial solvents/lubricants, plasticizers, additives, agrochemicals, pharmaceutical agents, and polyphenols of plant origin. These compounds are even present in the food as components (polyphenols) or food/water contaminants (pesticides, plasticizers used as food packaging) rendering the diet as the main route of exposure to EDCs for humans. Although huge amount of literature reports the (anti)estrogenic effects of different EDCs, relatively scarce information is available on the (anti)androgenic effects of EDCs. Here, the effects and mechanism of action of phytochemicals and pesticides and plasticizers as possible modulators of AR activities will be reviewed taking into account that insight derived from principles of endocrinology are required to estimate EDC consequences on endocrine deregulation and disease.
Collapse
Affiliation(s)
- Daniele Marcoccia
- Dpt. of Food Safety and Veterinary Public Health, Food and Veterinary Toxicology Unit, Istituto Superiore di Sanità - ISS, Viale Regina Elena 299, I-00161 Rome, Italy.,Present address: Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia-Romagna, via A. Bianchi 9, 25124 Brescia, Italy
| | - Marco Pellegrini
- Department of Science, University Roma Tre, Viale G. Marconi 446, I-00146 Rome, Italy.,Present address: Department of Molecular Medicine, University of Padova, Via Ugo Bassi, 58/b, 35131 Padova, Italy
| | - Marco Fiocchetti
- Department of Science, University Roma Tre, Viale G. Marconi 446, I-00146 Rome, Italy
| | - Stefano Lorenzetti
- Dpt. of Food Safety and Veterinary Public Health, Food and Veterinary Toxicology Unit, Istituto Superiore di Sanità - ISS, Viale Regina Elena 299, I-00161 Rome, Italy
| | - Maria Marino
- Department of Science, University Roma Tre, Viale G. Marconi 446, I-00146 Rome, Italy
| |
Collapse
|
23
|
Franconi F, Rosano G, Basili S, Montella A, Campesi I. Human cells involved in atherosclerosis have a sex. Int J Cardiol 2016; 228:983-1001. [PMID: 27915217 DOI: 10.1016/j.ijcard.2016.11.118] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 11/06/2016] [Indexed: 12/30/2022]
Abstract
The influence of sex has been largely described in cardiovascular diseases. Atherosclerosis is a complex process that involves many cell types such as vessel cells, immune cells and endothelial progenitor cells; however, many, if not all, studies do not report the sex of the cells. This review focuses on sex differences in human cells involved in the atherosclerotic process, emphasizing the role of sex hormones. Furthermore, we report sex differences and issues related to the processes that determine the fate of the cells such as apoptotic and autophagic mechanisms. The analysis of the data reveals that there are still many gaps in our knowledge regarding sex influences in atherosclerosis, largely for the cell types that have not been well studied, stressing the urgent need for a clear definition of experimental conditions and the inclusion of both sexes in preclinical studies.
Collapse
Affiliation(s)
- Flavia Franconi
- Assessorato alle Politiche per la Persona of Basilicata Region, Potenza, Italy; Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Giuseppe Rosano
- Cardiovascular and Cell Sciences Research Institute, St. George's University of London, United Kingdom
| | - Stefania Basili
- Department of Internal Medicine and Medical Specialties - Research Center on Gender and Evaluation and Promotion of Quality in Medicine (CEQUAM), Sapienza University of Rome, Italy
| | - Andrea Montella
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Ilaria Campesi
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy; Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Osilo, Italy.
| |
Collapse
|
24
|
Campesi I, Marino M, Montella A, Pais S, Franconi F. Sex Differences in Estrogen Receptor α and β Levels and Activation Status in LPS-Stimulated Human Macrophages. J Cell Physiol 2016; 232:340-345. [PMID: 27171902 DOI: 10.1002/jcp.25425] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 05/09/2016] [Indexed: 12/11/2022]
Abstract
Immune function, inflammation, and atherosclerosis display sex differences and are influenced by 17β-estradiol through estrogen receptors subtypes ERα and ERβ. Male tissues express active ERs, but their possible involvement in inflammation in males has never been assessed. Macrophages express both ERα and ERβ and offer the opportunity to evaluate the role of ER levels and activation in inflammation. We assessed the ability of lipopolysaccharide (LPS) to modulate, in a sex-specific way, the expression and the activation status of ERα and ERβ in blood monocytes-derived macrophages (MDMs) from men and women. MDMs were incubated with 100 ng/ml LPS for 24 h and used to evaluate ERα, ERβ, P-ERα, p38, and P-p38 expression by Western Blotting. In basal conditions, ERα and ERβ were significantly higher in female MDMs than in male MDMs. LPS up-regulated ERα and ERα phosphorylation in both sexes, with a significantly higher effect observed in male MDMs, and down-regulated ERβ level only in female MDMs. p38 and P-p38 proteins, indicative of ERβ activity, did not show sex differences both in basal conditions and after LPS treatment. Finally, ERα/ERβ and P-ERα/ERα ratios were significantly higher in male MDMs than in female ones. Our data indicate, for the first time, that LPS affects ERα but not ERβ activation status. We identify a significant role of ERα in LPS-mediated inflammatory responses in MDMs, which represents an initial step in understanding the influence of sex in the relationship between LPS and ERα. J. Cell. Physiol. 232: 340-345, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ilaria Campesi
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy. .,Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Osilo, Italy.
| | - Maria Marino
- Cell Physiology Lab, Department of Science, University Roma Tre, Rome, Italy.
| | - Andrea Montella
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Sara Pais
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Flavia Franconi
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy.,Assessorato alle Politiche per la Persona of Basilicata Region, Potenza, Italy
| |
Collapse
|
25
|
Hong H, Shen J, Ng HW, Sakkiah S, Ye H, Ge W, Gong P, Xiao W, Tong W. A Rat α-Fetoprotein Binding Activity Prediction Model to Facilitate Assessment of the Endocrine Disruption Potential of Environmental Chemicals. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2016; 13:372. [PMID: 27023588 PMCID: PMC4847034 DOI: 10.3390/ijerph13040372] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 03/10/2016] [Accepted: 03/22/2016] [Indexed: 11/21/2022]
Abstract
Endocrine disruptors such as polychlorinated biphenyls (PCBs), diethylstilbestrol (DES) and dichlorodiphenyltrichloroethane (DDT) are agents that interfere with the endocrine system and cause adverse health effects. Huge public health concern about endocrine disruptors has arisen. One of the mechanisms of endocrine disruption is through binding of endocrine disruptors with the hormone receptors in the target cells. Entrance of endocrine disruptors into target cells is the precondition of endocrine disruption. The binding capability of a chemical with proteins in the blood affects its entrance into the target cells and, thus, is very informative for the assessment of potential endocrine disruption of chemicals. α-fetoprotein is one of the major serum proteins that binds to a variety of chemicals such as estrogens. To better facilitate assessment of endocrine disruption of environmental chemicals, we developed a model for α-fetoprotein binding activity prediction using the novel pattern recognition method (Decision Forest) and the molecular descriptors calculated from two-dimensional structures by Mold² software. The predictive capability of the model has been evaluated through internal validation using 125 training chemicals (average balanced accuracy of 69%) and external validations using 22 chemicals (balanced accuracy of 71%). Prediction confidence analysis revealed the model performed much better at high prediction confidence. Our results indicate that the model is useful (when predictions are in high confidence) in endocrine disruption risk assessment of environmental chemicals though improvement by increasing number of training chemicals is needed.
Collapse
Affiliation(s)
- Huixiao Hong
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA.
| | - Jie Shen
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA.
| | - Hui Wen Ng
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA.
| | - Sugunadevi Sakkiah
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA.
| | - Hao Ye
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA.
| | - Weigong Ge
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA.
| | - Ping Gong
- Environmental Laboratory, U.S. Army Engineer Research and Development Center, 3909 Halls Ferry Road, Vicksburg, MS 39180, USA.
| | - Wenming Xiao
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA.
| | - Weida Tong
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA.
| |
Collapse
|
26
|
Campesi I, Capobianco G, Dessole S, Occhioni S, Montella A, Franconi F. Estrogenic Compounds Have Divergent Effects on Human Endothelial Progenitor Cell Migration according to Sex of the Donor. J Vasc Res 2016; 52:273-8. [DOI: 10.1159/000443403] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 12/12/2015] [Indexed: 11/19/2022] Open
|
27
|
Muka T, Vargas KG, Jaspers L, Wen KX, Dhana K, Vitezova A, Nano J, Brahimaj A, Colpani V, Bano A, Kraja B, Zaciragic A, Bramer WM, van Dijk GM, Kavousi M, Franco OH. Estrogen receptor β actions in the female cardiovascular system: A systematic review of animal and human studies. Maturitas 2016; 86:28-43. [PMID: 26921926 DOI: 10.1016/j.maturitas.2016.01.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Accepted: 01/14/2016] [Indexed: 12/27/2022]
Abstract
Five medical databases were searched for studies that assessed the role of ERβ in the female cardiovascular system and the influence of age and menopause on ERβ functioning. Of 9472 references, 88 studies met our inclusion criteria (71 animal model experimental studies, 15 human model experimental studies and 2 population based studies). ERβ signaling was shown to possess vasodilator and antiangiogenic properties by regulating the activity of nitric oxide, altering membrane ionic permeability in vascular smooth muscle cells, inhibiting vascular smooth muscle cell migration and proliferation and by regulating adrenergic control of the arteries. Also, a possible protective effect of ERβ signaling against left ventricular hypertrophy and ischemia/reperfusion injury via genomic and non-genomic pathways was suggested in 27 studies. Moreover, 5 studies reported that the vascular effects of ERβ may be vessel specific and may differ by age and menopause status. ERβ seems to possess multiple functions in the female cardiovascular system. Further studies are needed to evaluate whether isoform-selective ERβ-ligands might contribute to cardiovascular disease prevention.
Collapse
Affiliation(s)
- Taulant Muka
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands.
| | - Kris G Vargas
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - Loes Jaspers
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - Ke-xin Wen
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - Klodian Dhana
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - Anna Vitezova
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - Jana Nano
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - Adela Brahimaj
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - Veronica Colpani
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - Arjola Bano
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - Bledar Kraja
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands; Department of Biomedical Sciences, Faculty of Medicine, University of Medicine, Tirana, Albania; University Clinic of Gastrohepatology, University Hospital Center Mother Teresa, Tirana, Albania
| | - Asija Zaciragic
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | | | - Gaby M van Dijk
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - Maryam Kavousi
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - Oscar H Franco
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
28
|
Abstract
Bisphenol A (BPA) exposure has been associated with serious endocrine-disrupting effects in humans and wildlife. Toxicological and epidemiological studies evidenced that BPA increases body mass index and disrupts normal cardiovascular physiology by interfering with endogenous hormones in rodents, nonhuman primates, and cell culture test systems. The BPA concentration derived from these experiments were used by government regulatory agencies to determine the safe exposure levels of BPA in humans. However, accumulating literature in vivo and in vitro indicate that at concentrations lower than that reported in toxicological studies, BPA could elicit a different endocrine-disrupting capacity. To further complicate this picture, BPA effects rely on several and diverse mechanisms that converge upon endocrine and reproductive systems. If all or just few of these mechanisms concur to the endocrine-disrupting potential of low doses of BPA is at present still unclear. Thus, taking into account that the incidence and/or prevalence of health problems associated with endocrine disruption have increased worldwide, the goal of the present review is to give an overview of the many mechanisms of BPA action in order to decipher whether different mechanisms are at the root of the effect of low dose of BPA on endocrine system.
Collapse
Affiliation(s)
| | | | - Maria Marino
- Department of Science, Roma Tre University, Roma, Italy; INBB-National Laboratory of Gender and Endocrine Disruptors, Roma, Italy
| |
Collapse
|
29
|
Addis R, Campesi I, Fois M, Capobianco G, Dessole S, Fenu G, Montella A, Cattaneo MG, Vicentini LM, Franconi F. Human umbilical endothelial cells (HUVECs) have a sex: characterisation of the phenotype of male and female cells. Biol Sex Differ 2014; 5:18. [PMID: 25535548 PMCID: PMC4273493 DOI: 10.1186/s13293-014-0018-2] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 11/20/2014] [Indexed: 01/04/2023] Open
Abstract
Background Human umbilical endothelial cells (HUVECs) are widely used to study the endothelial physiology and pathology that might be involved in sex and gender differences detected at the cardiovascular level. This study evaluated whether HUVECs are sexually dimorphic in their morphological, proliferative and migratory properties and in the gene and protein expression of oestrogen and androgen receptors and nitric oxide synthase 3 (NOS3). Moreover, because autophagy is influenced by sex, its degree was analysed in male and female HUVECs (MHUVECs and FHUVECs). Methods Umbilical cords from healthy, normal weight male and female neonates born to healthy non-obese and non-smoking women were studied. HUVEC morphology was analysed by electron microscopy, and their function was investigated by proliferation, viability, wound healing and chemotaxis assays. Gene and protein expression for oestrogen and androgen receptors and for NOS3 were evaluated by real-time PCR and Western blotting, respectively, and the expression of the primary molecules involved in autophagy regulation [protein kinase B (Akt), mammalian target of rapamycin (mTOR), beclin-1 and microtubule-associated protein 1 light chain 3 (LC3)] were detected by Western blotting. Results Cell proliferation, migration NOS3 mRNA and protein expression were significantly higher in FHUVECs than in MHUVECs. Conversely, beclin-1 and the LC3-II/LC3-I ratio were higher in MHUVECs than in FHUVECs, indicating that male cells are more autophagic than female cells. The expression of oestrogen and androgen receptor genes and proteins, the protein expression of Akt and mTOR and cellular size and shape were not influenced by sex. Body weights of male and female neonates were not significantly different, but the weight of male babies positively correlated with the weight of the mother, suggesting that the mother’s weight may exert a different influence on male and female babies. Conclusions The results indicate that sex differences exist in prenatal life and are parameter-specific, suggesting that HUVECs of both sexes should be used as an in vitro model to increase the quality and the translational value of research. The sex differences observed in HUVECs could be relevant in explaining the diseases of adulthood because endothelial dysfunction has a crucial role in the pathogenesis of cardiovascular diseases, diabetes mellitus, neurodegeneration and immune disease.
Collapse
Affiliation(s)
- Roberta Addis
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Ilaria Campesi
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy.,National Laboratory of Gender Medicine of the National Institute of Biostructures and Biosystems, Osilo, Sassari Italy
| | - Marco Fois
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Giampiero Capobianco
- Department of Surgical, Microsurgical and Medical Sciences, Gynaecologic and Obstetric Clinic, University of Sassari, Sassari, Italy
| | - Salvatore Dessole
- Department of Surgical, Microsurgical and Medical Sciences, Gynaecologic and Obstetric Clinic, University of Sassari, Sassari, Italy
| | - Grazia Fenu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Andrea Montella
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Maria Grazia Cattaneo
- Department of Medical Biotechnology and Translational Medicine, University of Milano, Milano, Italy
| | - Lucia M Vicentini
- Department of Medical Biotechnology and Translational Medicine, University of Milano, Milano, Italy
| | - Flavia Franconi
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy.,National Laboratory of Gender Medicine of the National Institute of Biostructures and Biosystems, Osilo, Sassari Italy.,Assessorato alle Politiche per la Persona, Region Basilicata, Italy
| |
Collapse
|
30
|
Franconi F, Rosano G, Campesi I. Need for gender-specific pre-analytical testing: the dark side of the moon in laboratory testing. Int J Cardiol 2014; 179:514-35. [PMID: 25465806 DOI: 10.1016/j.ijcard.2014.11.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 10/27/2014] [Accepted: 11/03/2014] [Indexed: 01/16/2023]
Abstract
Many international organisations encourage studies in a sex-gender perspective. However, research with a gender perspective presents a high degree of complexity, and the inclusion of sex-gender variable in experiments presents many methodological questions, the majority of which are still neglected. Overcoming these issues is fundamental to avoid erroneous results. Here, pre-analytical aspects of the research, such as study design, choice of utilised specimens, sample collection and processing, animal models of diseases, and the observer's role, are discussed. Artefacts in this stage of research could affect the predictive value of all analyses. Furthermore, the standardisation of research subjects according to their lifestyles and, if female, to their life phase and menses or oestrous cycle, is urgent to harmonise research worldwide. A sex-gender-specific attention to pre-analytical aspects could produce a decrease in the time for translation from the bench to bedside. Furthermore, sex-gender-specific pre-clinical pharmacological testing will enable adequate assessment of pharmacokinetic and pharmacodynamic actions of drugs and will enable, where appropriate, an adequate gender-specific clinical development plan. Therefore, sex-gender-specific pre-clinical research will increase the gender equity of care and will produce more evidence-based medicine.
Collapse
Affiliation(s)
- Flavia Franconi
- Department of Biomedical Sciences, University of Sassari, National Laboratory of Gender Medicine of the National Institute of Biostructures and Biosystems, Osilo, Sassari, Italy; Vicepresident of Basilicata Region.
| | - Giuseppe Rosano
- Cardiovascular and Cell Sciences Research Institute, St George's University of London, United Kingdom
| | - Ilaria Campesi
- Department of Biomedical Sciences, University of Sassari, National Laboratory of Gender Medicine of the National Institute of Biostructures and Biosystems, Osilo, Sassari, Italy
| |
Collapse
|
31
|
Pesiri V, Totta P, Marino M, Acconcia F. Ubiquitin-activating enzyme is necessary for 17β-estradiol-induced breast cancer cell proliferation and migration. IUBMB Life 2014; 66:578-85. [DOI: 10.1002/iub.1296] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 07/25/2014] [Indexed: 11/12/2022]
Affiliation(s)
- Valeria Pesiri
- Department of Science; Section Biomedical Sciences and Technology, University Roma Tre; Rome Italy
| | - Pierangela Totta
- Department of Science; Section Biomedical Sciences and Technology, University Roma Tre; Rome Italy
| | - Maria Marino
- Department of Science; Section Biomedical Sciences and Technology, University Roma Tre; Rome Italy
| | - Filippo Acconcia
- Department of Science; Section Biomedical Sciences and Technology, University Roma Tre; Rome Italy
| |
Collapse
|
32
|
Impact of bisphenol a on the cardiovascular system - epidemiological and experimental evidence and molecular mechanisms. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2014; 11:8399-413. [PMID: 25153468 PMCID: PMC4143868 DOI: 10.3390/ijerph110808399] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 07/28/2014] [Accepted: 08/07/2014] [Indexed: 12/30/2022]
Abstract
Bisphenol A (BPA) is a ubiquitous plasticizing agent used in the manufacturing of polycarbonate plastics and epoxy resins. There is well-documented and broad human exposure to BPA. The potential risk that BPA poses to the human health has attracted much attention from regulatory agencies and the general public, and has been extensively studied. An emerging and rapidly growing area in the study of BPA's toxicity is its impact on the cardiovascular (CV) system. Recent epidemiological studies have shown that higher urinary BPA concentration in humans is associated with various types of CV diseases, including angina, hypertension, heart attack and coronary and peripheral arterial disease. Experimental studies have demonstrated that acute BPA exposure promotes the development of arrhythmias in female rodent hearts. Chronic exposure to BPA has been shown to result in cardiac remodeling, atherosclerosis, and altered blood pressure in rodents. The underlying mechanisms may involve alteration of cardiac Ca2+ handling, ion channel inhibition/activation, oxidative stress, and genome/transcriptome modifications. In this review, we discuss these recent findings that point to the potential CV toxicity of BPA, and highlight the knowledge gaps in this growing research area.
Collapse
|