1
|
Lolescu BA, Furdui-Lința AV, Ilie CA, Sturza A, Zară F, Muntean DM, Blidișel A, Crețu OM. Adipose tissue as target of environmental toxicants: focus on mitochondrial dysfunction and oxidative inflammation in metabolic dysfunction-associated steatotic liver disease. Mol Cell Biochem 2024:10.1007/s11010-024-05165-z. [PMID: 39704874 DOI: 10.1007/s11010-024-05165-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 11/10/2024] [Indexed: 12/21/2024]
Abstract
Obesity, diabetes, and their cardiovascular and hepatic comorbidities are alarming public health issues of the twenty-first century, which share mitochondrial dysfunction, oxidative stress, and chronic inflammation as common pathophysiological mechanisms. An increasing body of evidence links the combined exposure to multiple environmental toxicants with the occurrence and severity of metabolic diseases. Endocrine disruptors (EDs) are ubiquitous chemicals or mixtures with persistent deleterious effects on the living organisms beyond the endocrine system impairment; in particular, those known as metabolism-disrupting chemicals (MDCs), increase the risk of the metabolic pathologies in adult organism or its progeny. Being largely lipophilic, MDCs mainly target the adipose tissue and elicit mitochondrial dysfunction by interfering with mitochondrial bioenergetics, biogenesis, dynamics and/or other functions. Plastics, when broken down into micro- and nano-plastics (MNPs), have been detected in several human tissues, including the liver. The harmful interplay between inflammatory and redox processes, which mutually interact in a positive feed-back loop, hence the term oxidative inflammation ("OxInflammation"), occurs both at systemic and organ level. In both liver and adipose tissue, oxinflammation contributes to the progression of the metabolic dysfunction-associated steatotic liver disease (MASLD). Moreover, it has been reported that individuals with MASLD may be more susceptible to the harmful effects of toxicants (mainly, those related to mitochondria) and that chronic exposure to EDs/MDCs or MNPs may play a role in the development of the disease. While liver has been systematically investigated as major target organ for ambient chemicals, surprisingly, less information is available in the literature with respect to the adipose tissue. In this narrative review, we delve into the current literature on the most studied environmental toxicants (bisphenols, polychlorinated biphenyls, phthalates, tolylfluanid and tributyltin, per-fluoroalkyl and polyfluoroalkyl substances, heavy metals and MNPs), summarize their deleterious effects on adipose tissue, and address the role of dysregulated mitochondria and oxinflammation, particularly in the setting of MASLD.
Collapse
Affiliation(s)
- Bogdan A Lolescu
- Doctoral School Medicine, Center for Translational Research and Systems Medicine, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
- Center for Translational Research and Systems Medicine, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
| | - Adina V Furdui-Lința
- Doctoral School Medicine, Center for Translational Research and Systems Medicine, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
- Center for Translational Research and Systems Medicine, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
- Department III Functional Sciences-Chair of Pathophysiology, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
| | - Cosmin A Ilie
- Center for Translational Research and Systems Medicine, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
- Department III Functional Sciences-Chair of Public Health & Sanitary Management, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
| | - Adrian Sturza
- Center for Translational Research and Systems Medicine, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
- Department III Functional Sciences-Chair of Pathophysiology, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
| | - Flavia Zară
- Department II Microscopic Morphology-Chair of Histology, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
- Department of Pathology, Timisoara Municipal Emergency Clinical Hospital, Timișoara, Romania
| | - Danina M Muntean
- Center for Translational Research and Systems Medicine, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
- Department III Functional Sciences-Chair of Pathophysiology, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
| | - Alexandru Blidișel
- Department of Surgery I-Clinic of Surgical Semiotics & Thoracic Surgery, Center for Hepato-Biliary and Pancreatic Surgery, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Eftimie Murgu Sq., No.2, 300041, Timișoara, Romania.
| | - Octavian M Crețu
- Department of Surgery I-Clinic of Surgical Semiotics & Thoracic Surgery, Center for Hepato-Biliary and Pancreatic Surgery, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Eftimie Murgu Sq., No.2, 300041, Timișoara, Romania
| |
Collapse
|
2
|
Im S, Kang S, Son WJ, Son M, Oh SJ, Yoon HJ, Pak YK. Dioxin-Induced PAI-1 Expression: A Novel Pathway to Pancreatic β-Cell Failure in Type 2 Diabetes. Int J Mol Sci 2024; 25:11974. [PMID: 39596044 PMCID: PMC11594116 DOI: 10.3390/ijms252211974] [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: 10/14/2024] [Revised: 11/01/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
Exposure to environment-polluting chemicals (EPCs), which are ligands of the aryl hydrocarbon receptor (AhR), is associated with the development of type 2 diabetes (T2D). This study explores the mechanisms by which AhR ligands contribute to β-cell failure in T2D. Incubation of RINm5F rat pancreatic β-cells with low-dose 2,3,7,8-tetrachlorodibenzodioxin (TCDD), the most potent AhR ligand, inhibited glucose-stimulated insulin secretion (GSIS). A single injection of TCDD in wild type mice reduced the size of Langerhans islets, but not in AhR liver knock-out mice (AhR-LKO). RNA-seq database analysis identified Serpine1, encoding for plasminogen activator inhibitor type-1 (PAI-1) as a TCDD-mediated secretory protein that is synthesized in an AhR-dependent manner in the liver. Elevated PAI-1 levels were shown to induce Caspase-3/7-dependent apoptosis in RINm5F cells, suggesting a novel pathway through which EPCs exacerbate T2D. These findings support the hypothesis that chronic exposure to AhR ligands may directly inhibit GSIS in pancreatic β-cells and indirectly induce β-cell apoptosis through increased PAI-1. This study provides new insights into the EPC-PAI-1 axis as a missing link between pancreatic β-cell failure and the progression of T2D and offers a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Suyeol Im
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.I.); (S.K.); (W.J.S.); (S.J.O.)
- Department of Physiology, School of Medicine, Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sora Kang
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.I.); (S.K.); (W.J.S.); (S.J.O.)
- Department of Physiology, School of Medicine, Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Woo Jung Son
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.I.); (S.K.); (W.J.S.); (S.J.O.)
| | - Minuk Son
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (M.S.); (H.J.Y.)
| | - Seung Jun Oh
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.I.); (S.K.); (W.J.S.); (S.J.O.)
| | - Hye Ji Yoon
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (M.S.); (H.J.Y.)
| | - Youngmi Kim Pak
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.I.); (S.K.); (W.J.S.); (S.J.O.)
- Department of Physiology, School of Medicine, Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (M.S.); (H.J.Y.)
| |
Collapse
|
3
|
Pavlíková N, Šrámek J, Němcová V, Bajard L. Effects of novel flame retardants tris(1,3-dichloro-2-propyl) phosphate (TDCIPP) and triphenyl phosphate (TPhP) on function and homeostasis in human and rat pancreatic beta-cell lines. Arch Toxicol 2024; 98:3859-3874. [PMID: 39192017 PMCID: PMC11489283 DOI: 10.1007/s00204-024-03841-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: 06/24/2024] [Accepted: 08/08/2024] [Indexed: 08/29/2024]
Abstract
Despite the fact that environmental pollution has been implicated in the global rise of diabetes, the research on the impact of emerging pollutants such as novel flame retardants remains limited. In line with the shift towards the use of non-animal approaches in toxicological testing, this study aimed to investigate the effects of two novel flame retardants tris(1,3-dichloro-2-propyl) phosphate (TDCIPP) and triphenyl phosphate (TPhP) in rat (INS1E) and human (NES2Y) pancreatic beta-cell lines. One-week exposure to 1 μM and 10 μM TDCIPP and TPhP altered intracellular insulin and proinsulin levels, but not the levels of secreted insulin (despite the presence of a statistically insignificant trend). The exposures also altered the protein expression of several factors involved in beta-cell metabolic pathways and signaling, including ATP citrate lyase, isocitrate dehydrogenase 1, perilipins, glucose transporters, ER stress-related factors, and antioxidant enzymes. This study has brought new and valuable insights into the toxicity of TDCIPP and TPhP on beta-cell function and revealed alterations that might impact insulin secretion after more extended exposure. It also adds to the scarce studies using in vitro pancreatic beta-cells models in toxicological testing, thereby promoting the development of non-animal testing strategy for identifying pro-diabetic effects of chemical pollutants.
Collapse
Affiliation(s)
- Nela Pavlíková
- 3LF UK, Departement of Biochemistry, Cell and Molecular Biology & Center for Research On Nutrition, Metabolism, and Diabetes, Third Faculty of Medicine, Charles University, Ruska 87, 100 00, Prague, Czech Republic.
| | - Jan Šrámek
- 3LF UK, Departement of Biochemistry, Cell and Molecular Biology & Center for Research On Nutrition, Metabolism, and Diabetes, Third Faculty of Medicine, Charles University, Ruska 87, 100 00, Prague, Czech Republic
| | - Vlasta Němcová
- 3LF UK, Departement of Biochemistry, Cell and Molecular Biology & Center for Research On Nutrition, Metabolism, and Diabetes, Third Faculty of Medicine, Charles University, Ruska 87, 100 00, Prague, Czech Republic
| | - Lola Bajard
- Faculty of Science, RECETOX, Masaryk University, Kotlarska 2, 611 37, Brno, Czech Republic
| |
Collapse
|
4
|
Nishitha-Hiresha V, Varsha R, Srinidhi S, Jayasuriya R, Harithpriya K, Chakraborty P, Ramkumar KM. Effect of Bis (methyl glycol) phthalate on endoplasmic reticulum stress in endothelial cells. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2024; 111:104569. [PMID: 39326718 DOI: 10.1016/j.etap.2024.104569] [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: 05/23/2024] [Revised: 09/14/2024] [Accepted: 09/23/2024] [Indexed: 09/28/2024]
Abstract
Phthalate-based polymeric plasticizers are widely used for their durability, transparency, and odorless nature, resulting in human exposure through inhalation, ingestion, or contaminated water. Epidemiological studies have identified bis-phthalate as a potential cardiovascular disease risk factor, though its mechanisms remain unclear. This study investigates the effects of bis-phthalate on endothelial dysfunction (ED), an early event in cardiovascular complications, with a focus on Endoplasmic Reticulum (ER) stress pathways. We observed dose- and time-dependent cytotoxicity in endothelial cells exposed to bis-phthalate, accompanied by elevated expression of ER stress markers (GRP78, IRE-1α, CHOP) and oxidative stress markers (TXNIP, P22phox), as measured by qPCR. Reactive oxygen species (ROS) levels also increased dose-dependently, as determined by H2DCFDA using flow cytometry. These findings suggest that bis-phthalate exposure induces both oxidative and ER stress, leading to the development of ED, providing insights into its potential role in cardiovascular disease progression.
Collapse
Affiliation(s)
- Verma Nishitha-Hiresha
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603 203, India
| | - Raghavan Varsha
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603 203, India
| | - S Srinidhi
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603 203, India
| | - Ravichandran Jayasuriya
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603 203, India
| | - Kannan Harithpriya
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603 203, India
| | - Paromita Chakraborty
- Environmental Science and Technology Research Group, Centre for Research in Environment, Sustainability Advocacy and Climate Change, Directorate of Research, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Kunka Mohanram Ramkumar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603 203, India.
| |
Collapse
|
5
|
Michelangeli F, Mohammed NA, Jones B, Tairu M, Al‐Mousa F. Cytotoxicity by endocrine disruptors through effects on ER Ca 2+ transporters, aberrations in Ca 2+ signalling pathways and ER stress. FEBS Open Bio 2024; 14:1384-1396. [PMID: 39138623 PMCID: PMC11492318 DOI: 10.1002/2211-5463.13880] [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: 04/10/2024] [Revised: 06/24/2024] [Accepted: 08/05/2024] [Indexed: 08/15/2024] Open
Abstract
Concerns regarding man-made organic chemicals pervading our ecosystem and having adverse and detrimental effects upon organisms, including man, have now been studied for several decades. Since the 1970s, some environmental pollutants were identified as having endocrine disrupting affects. These endocrine disrupting chemicals (EDC) were initially shown to have estrogenic or anti-estrogenic properties and some were also shown to bind to a variety of hormone receptors. However, since the 1990s it has also been identified that many of these EDC additionally, have the ability of causing abnormal alterations in Ca2+ signalling pathways (also commonly involved in hormone signalling), leading to exaggerated elevations in cytosolic [Ca2+] levels, that is known to cause activation of a number of cell death pathways. The major emphasis of this review is to present a personal perspective of the evidence for some types of EDC, specifically alkylphenols and brominated flame retardants (BFRs), causing direct effects on Ca2+ transporters (mainly the SERCA Ca2+ ATPases), culminating in acute cytotoxicity and cell death. Evidence is also presented to indicate that this Ca2+ATPase inhibition, which leads to abnormally elevated cytosolic [Ca2+], as well as a decreased luminal ER [Ca2+], which triggers the ER stress response, are both involved in acute cytotoxicity.
Collapse
Affiliation(s)
- Francesco Michelangeli
- Chester Medical SchoolUniversity of ChesterUK
- School of BiosciencesUniversity of BirminghamUK
| | - Noor A. Mohammed
- School of BiosciencesUniversity of BirminghamUK
- Department of BiologyUniversity of DuhokIraq
| | | | | | - Fawaz Al‐Mousa
- General Directorate of Poison Control CentreMinistry of HealthRiyadhSaudi Arabia
| |
Collapse
|
6
|
He K, Chen R, Xu S, Ding Y, Wu Z, Bao M, He B, Li S. Environmental endocrine disruptor-induced mitochondrial dysfunction: a potential mechanism underlying diabetes and its complications. Front Endocrinol (Lausanne) 2024; 15:1422752. [PMID: 39211449 PMCID: PMC11357934 DOI: 10.3389/fendo.2024.1422752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 07/08/2024] [Indexed: 09/04/2024] Open
Abstract
Diabetes and its complications significantly affect individuals' quality of life. The etiology of diabetes mellitus and its associated complications is complex and not yet fully understood. There is an increasing emphasis on investigating the effects of endocrine disruptors on diabetes, as these substances can impact cellular processes, energy production, and utilization, ultimately leading to disturbances in energy homeostasis. Mitochondria play a crucial role in cellular energy generation, and any impairment in these organelles can increase susceptibility to diabetes. This review examines the most recent epidemiological and pathogenic evidence concerning the link between endocrine disruptors and diabetes, including its complications. The analysis suggests that endocrine disruptor-induced mitochondrial dysfunction-characterized by disruptions in the mitochondrial electron transport chain, dysregulation of calcium ions (Ca2+), overproduction of reactive oxygen species (ROS), and initiation of signaling pathways related to mitochondrial apoptosis-may be key mechanisms connecting endocrine disruptors to the development of diabetes and its complications.
Collapse
Affiliation(s)
- Kunhui He
- The 1 Affiliate Hospital of Changsha Medical University, Changsha Medical University, Changsha, China
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, School of Pharmaceutical Science, Changsha Medical University, Changsha, China
| | - Rumeng Chen
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Shuling Xu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yining Ding
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Zhu Wu
- The Hunan Provincial Key Laboratory of the TCM Agricultural Biogenomics, Changsha Medical University, Changsha, China
| | - Meihua Bao
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, School of Pharmaceutical Science, Changsha Medical University, Changsha, China
- The Hunan Provincial Key Laboratory of the TCM Agricultural Biogenomics, Changsha Medical University, Changsha, China
| | - Binsheng He
- The Hunan Provincial Key Laboratory of the TCM Agricultural Biogenomics, Changsha Medical University, Changsha, China
| | - Sen Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
7
|
Lința AV, Lolescu BM, Ilie CA, Vlad M, Blidișel A, Sturza A, Borza C, Muntean DM, Crețu OM. Liver and Pancreatic Toxicity of Endocrine-Disruptive Chemicals: Focus on Mitochondrial Dysfunction and Oxidative Stress. Int J Mol Sci 2024; 25:7420. [PMID: 39000526 PMCID: PMC11242905 DOI: 10.3390/ijms25137420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 06/29/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
In recent years, the worldwide epidemic of metabolic diseases, namely obesity, metabolic syndrome, diabetes and metabolic-associated fatty liver disease (MAFLD) has been strongly associated with constant exposure to endocrine-disruptive chemicals (EDCs), in particular, the ones able to disrupt various metabolic pathways. EDCs have a negative impact on several human tissues/systems, including metabolically active organs, such as the liver and pancreas. Among their deleterious effects, EDCs induce mitochondrial dysfunction and oxidative stress, which are also the major pathophysiological mechanisms underlying metabolic diseases. In this narrative review, we delve into the current literature on EDC toxicity effects on the liver and pancreatic tissues in terms of impaired mitochondrial function and redox homeostasis.
Collapse
Affiliation(s)
- Adina V. Lința
- Department of Functional Sciences—Chair of Pathophysiology, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timișoara, Romania; (A.V.L.); (A.S.); (C.B.)
- Centre for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timișoara, Romania; (B.M.L.); (C.A.I.)
- Doctoral School Medicine-Pharmacy, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq., No. 2, 300041 Timișoara, Romania
| | - Bogdan M. Lolescu
- Centre for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timișoara, Romania; (B.M.L.); (C.A.I.)
- Doctoral School Medicine-Pharmacy, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq., No. 2, 300041 Timișoara, Romania
| | - Cosmin A. Ilie
- Centre for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timișoara, Romania; (B.M.L.); (C.A.I.)
- Department of Functional Sciences—Chair of Public Health & Sanitary Management, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timișoara, Romania
| | - Mihaela Vlad
- Department of Internal Medicine II—Chair of Endocrinology, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq., No. 2, 300041 Timișoara, Romania;
| | - Alexandru Blidișel
- Department of Surgery I—Chair of Surgical Semiotics & Thoracic Surgery, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timişoara, Romania; (A.B.); (O.M.C.)
- Centre for Hepato-Biliary and Pancreatic Surgery, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timişoara, Romania
| | - Adrian Sturza
- Department of Functional Sciences—Chair of Pathophysiology, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timișoara, Romania; (A.V.L.); (A.S.); (C.B.)
- Centre for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timișoara, Romania; (B.M.L.); (C.A.I.)
| | - Claudia Borza
- Department of Functional Sciences—Chair of Pathophysiology, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timișoara, Romania; (A.V.L.); (A.S.); (C.B.)
- Centre for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timișoara, Romania; (B.M.L.); (C.A.I.)
| | - Danina M. Muntean
- Department of Functional Sciences—Chair of Pathophysiology, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timișoara, Romania; (A.V.L.); (A.S.); (C.B.)
- Centre for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timișoara, Romania; (B.M.L.); (C.A.I.)
| | - Octavian M. Crețu
- Department of Surgery I—Chair of Surgical Semiotics & Thoracic Surgery, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timişoara, Romania; (A.B.); (O.M.C.)
- Centre for Hepato-Biliary and Pancreatic Surgery, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timişoara, Romania
| |
Collapse
|
8
|
Alymbaeva D, Szabo C, Jocsak G, Bartha T, Zsarnovszky A, Kovago C, Ondrasovicova S, Kiss DS. Analysis of arsenic-modulated expression of hypothalamic estrogen receptor, thyroid receptor, and peroxisome proliferator-activated receptor gamma mRNA and simultaneous mitochondrial morphology and respiration rates in the mouse. PLoS One 2024; 19:e0303528. [PMID: 38753618 PMCID: PMC11098319 DOI: 10.1371/journal.pone.0303528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 04/26/2024] [Indexed: 05/18/2024] Open
Abstract
Arsenic has been identified as an environmental toxicant acting through various mechanisms, including the disruption of endocrine pathways. The present study assessed the ability of a single intraperitoneal injection of arsenic, to modify the mRNA expression levels of estrogen- and thyroid hormone receptors (ERα,β; TRα,β) and peroxisome proliferator-activated receptor gamma (PPARγ) in hypothalamic tissue homogenates of prepubertal mice in vivo. Mitochondrial respiration (MRR) was also measured, and the corresponding mitochondrial ultrastructure was analyzed. Results show that ERα,β, and TRα expression was significantly increased by arsenic, in all concentrations examined. In contrast, TRβ and PPARγ remained unaffected after arsenic injection. Arsenic-induced dose-dependent changes in state 4 mitochondrial respiration (St4). Mitochondrial morphology was affected by arsenic in that the 5 mg dose increased the size but decreased the number of mitochondria in agouti-related protein- (AgRP), while increasing the size without affecting the number of mitochondria in pro-opiomelanocortin (POMC) neurons. Arsenic also increased the size of the mitochondrial matrix per host mitochondrion. Complex analysis of dose-dependent response patterns between receptor mRNA, mitochondrial morphology, and mitochondrial respiration in the neuroendocrine hypothalamus suggests that instant arsenic effects on receptor mRNAs may not be directly reflected in St3-4 values, however, mitochondrial dynamics is affected, which predicts more pronounced effects in hypothalamus-regulated homeostatic processes after long-term arsenic exposure.
Collapse
Affiliation(s)
- Daiana Alymbaeva
- Department of Physiology and Biochemistry, University of Veterinary Medicine, Budapest, Hungary
| | - Csaba Szabo
- Department of Animal Physiology and Health, Hungarian University of Agricultural and Life Sciences, Godollo, Hungary
| | - Gergely Jocsak
- Department of Physiology and Biochemistry, University of Veterinary Medicine, Budapest, Hungary
| | - Tibor Bartha
- Department of Physiology and Biochemistry, University of Veterinary Medicine, Budapest, Hungary
| | - Attila Zsarnovszky
- Department of Physiology and Biochemistry, University of Veterinary Medicine, Budapest, Hungary
- Department of Animal Physiology and Health, Hungarian University of Agricultural and Life Sciences, Godollo, Hungary
- Agribiotechnology and Precision Breeding for Food Security National Laboratory, Department of Animal Physiology and Health, Institute of Physiology and Nutrition, Hungarian University of Agricultural and Life Sciences, Kaposvar, Hungary
| | - Csaba Kovago
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, Budapest, Hungary
| | - Silvia Ondrasovicova
- Department of Biology and Physiology, University of Veterinary Medicine and Pharmacy in Košice, Košice, Slovakia
| | - David Sandor Kiss
- Department of Physiology and Biochemistry, University of Veterinary Medicine, Budapest, Hungary
| |
Collapse
|
9
|
Ferreira T, Rodriguez S. Mitochondrial DNA: Inherent Complexities Relevant to Genetic Analyses. Genes (Basel) 2024; 15:617. [PMID: 38790246 PMCID: PMC11121663 DOI: 10.3390/genes15050617] [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: 04/17/2024] [Revised: 05/09/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Mitochondrial DNA (mtDNA) exhibits distinct characteristics distinguishing it from the nuclear genome, necessitating specific analytical methods in genetic studies. This comprehensive review explores the complex role of mtDNA in a variety of genetic studies, including genome-wide, epigenome-wide, and phenome-wide association studies, with a focus on its implications for human traits and diseases. Here, we discuss the structure and gene-encoding properties of mtDNA, along with the influence of environmental factors and epigenetic modifications on its function and variability. Particularly significant are the challenges posed by mtDNA's high mutation rate, heteroplasmy, and copy number variations, and their impact on disease susceptibility and population genetic analyses. The review also highlights recent advances in methodological approaches that enhance our understanding of mtDNA associations, advocating for refined genetic research techniques that accommodate its complexities. By providing a comprehensive overview of the intricacies of mtDNA, this paper underscores the need for an integrated approach to genetic studies that considers the unique properties of mitochondrial genetics. Our findings aim to inform future research and encourage the development of innovative methodologies to better interpret the broad implications of mtDNA in human health and disease.
Collapse
Affiliation(s)
- Tomas Ferreira
- Bristol Medical School, University of Bristol, Bristol BS8 1UD, UK
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SL, UK
| | - Santiago Rodriguez
- Bristol Medical School, University of Bristol, Bristol BS8 1UD, UK
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 1QU, UK
| |
Collapse
|
10
|
Kabekkodu SP, Gladwell LR, Choudhury M. The mitochondrial link: Phthalate exposure and cardiovascular disease. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119708. [PMID: 38508420 DOI: 10.1016/j.bbamcr.2024.119708] [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: 11/14/2023] [Revised: 02/17/2024] [Accepted: 03/09/2024] [Indexed: 03/22/2024]
Abstract
Phthalates' pervasive presence in everyday life poses concern as they have been revealed to induce perturbing health defects. Utilized as a plasticizer, phthalates are riddled throughout many common consumer products including personal care products, food packaging, home furnishings, and medical supplies. Phthalates permeate into the environment by leaching out of these products which can subsequently be taken up by the human body. It is previously established that a connection exists between phthalate exposure and cardiovascular disease (CVD) development; however, the specific mitochondrial link in this scenario has not yet been described. Prior studies have indicated that one possible mechanism for how phthalates exert their effects is through mitochondrial dysfunction. By disturbing mitochondrial structure, function, and signaling, phthalates can contribute to the development of the foremost cause of death worldwide, CVD. This review will examine the potential link among phthalates and their effects on the mitochondria, permissive of CVD development.
Collapse
Affiliation(s)
- Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Lauren Rae Gladwell
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M Health Science Center, College Station, TX, USA
| | - Mahua Choudhury
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M Health Science Center, College Station, TX, USA.
| |
Collapse
|
11
|
Heindel JJ, Lustig RH, Howard S, Corkey BE. Obesogens: a unifying theory for the global rise in obesity. Int J Obes (Lond) 2024; 48:449-460. [PMID: 38212644 PMCID: PMC10978495 DOI: 10.1038/s41366-024-01460-3] [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: 08/31/2023] [Revised: 12/14/2023] [Accepted: 01/02/2024] [Indexed: 01/13/2024]
Abstract
Despite varied treatment, mitigation, and prevention efforts, the global prevalence and severity of obesity continue to worsen. Here we propose a combined model of obesity, a unifying paradigm that links four general models: the energy balance model (EBM), based on calories as the driver of weight gain; the carbohydrate-insulin model (CIM), based on insulin as a driver of energy storage; the oxidation-reduction model (REDOX), based on reactive oxygen species (ROS) as a driver of altered metabolic signaling; and the obesogens model (OBS), which proposes that environmental chemicals interfere with hormonal signaling leading to adiposity. We propose a combined OBS/REDOX model in which environmental chemicals (in air, food, food packaging, and household products) generate false autocrine and endocrine metabolic signals, including ROS, that subvert standard regulatory energy mechanisms, increase basal and stimulated insulin secretion, disrupt energy efficiency, and influence appetite and energy expenditure leading to weight gain. This combined model incorporates the data supporting the EBM and CIM models, thus creating one integrated model that covers significant aspects of all the mechanisms potentially contributing to the obesity pandemic. Importantly, the OBS/REDOX model provides a rationale and approach for future preventative efforts based on environmental chemical exposure reduction.
Collapse
Affiliation(s)
- Jerrold J Heindel
- Healthy Environment and Endocrine Disruptor Strategies (HEEDS), Bozeman, MT, 59715, USA.
| | - Robert H Lustig
- Department of Pediatrics and Institute for Health Policy Studies, University of California, San Francisco, CA, 94143, USA
| | - Sarah Howard
- Healthy Environment and Endocrine Disruptor Strategies (HEEDS), Bozeman, MT, 59715, USA
| | - Barbara E Corkey
- Department of Medicine, Boston University, Chobanian and Avedisian School of Medicine, Boston, MA, 02118, USA
| |
Collapse
|
12
|
Sheikh AM, Yano S, Tabassum S, Nagai A. The Role of the Vascular System in Degenerative Diseases: Mechanisms and Implications. Int J Mol Sci 2024; 25:2169. [PMID: 38396849 PMCID: PMC10889477 DOI: 10.3390/ijms25042169] [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: 01/12/2024] [Revised: 02/03/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
Degenerative diseases, encompassing a wide range of conditions affecting various organ systems, pose significant challenges to global healthcare systems. This comprehensive review explores the intricate interplay between the vascular system and degenerative diseases, shedding light on the underlying mechanisms and profound implications for disease progression and management. The pivotal role of the vascular system in maintaining tissue homeostasis is highlighted, as it serves as the conduit for oxygen, nutrients, and immune cells to vital organs and tissues. Due to the vital role of the vascular system in maintaining homeostasis, its dysfunction, characterized by impaired blood flow, endothelial dysfunction, and vascular inflammation, emerges as a common denominator of degenerative diseases across multiple systems. In the nervous system, we explored the influence of vascular factors on neurodegenerative diseases such as Alzheimer's and Parkinson's, emphasizing the critical role of cerebral blood flow regulation and the blood-brain barrier. Within the kidney system, the intricate relationship between vascular health and chronic kidney disease is scrutinized, unraveling the mechanisms by which hypertension and other vascular factors contribute to renal dysfunction. Throughout this review, we emphasize the clinical significance of understanding vascular involvement in degenerative diseases and potential therapeutic interventions targeting vascular health, highlighting emerging treatments and prevention strategies. In conclusion, a profound appreciation of the role of the vascular system in degenerative diseases is essential for advancing our understanding of degenerative disease pathogenesis and developing innovative approaches for prevention and treatment. This review provides a comprehensive foundation for researchers, clinicians, and policymakers seeking to address the intricate relationship between vascular health and degenerative diseases in pursuit of improved patient outcomes and enhanced public health.
Collapse
Affiliation(s)
- Abdullah Md. Sheikh
- Department of Laboratory Medicine, Faculty of Medicine, Shimane University, 89-1 Enya Cho, Izumo 693-8501, Japan; (S.Y.); (S.T.); (A.N.)
| | - Shozo Yano
- Department of Laboratory Medicine, Faculty of Medicine, Shimane University, 89-1 Enya Cho, Izumo 693-8501, Japan; (S.Y.); (S.T.); (A.N.)
| | - Shatera Tabassum
- Department of Laboratory Medicine, Faculty of Medicine, Shimane University, 89-1 Enya Cho, Izumo 693-8501, Japan; (S.Y.); (S.T.); (A.N.)
| | - Atsushi Nagai
- Department of Laboratory Medicine, Faculty of Medicine, Shimane University, 89-1 Enya Cho, Izumo 693-8501, Japan; (S.Y.); (S.T.); (A.N.)
- Department of Neurology, Faculty of Medicine, Shimane University, 89-1 Enya Cho, Izumo 693-8501, Japan
| |
Collapse
|
13
|
Saikia UK, Kumar A. Endocrine disruptors in the pathogenesis of metabolic syndrome. METABOLIC SYNDROME 2024:235-248. [DOI: 10.1016/b978-0-323-85732-1.00018-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
14
|
Wang H, Akbari-Alavijeh S, Parhar RS, Gaugler R, Hashmi S. Partners in diabetes epidemic: A global perspective. World J Diabetes 2023; 14:1463-1477. [PMID: 37970124 PMCID: PMC10642420 DOI: 10.4239/wjd.v14.i10.1463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/01/2023] [Accepted: 09/01/2023] [Indexed: 10/09/2023] Open
Abstract
There is a recent increase in the worldwide prevalence of both obesity and diabetes. In this review we assessed insulin signaling, genetics, environment, lipid metabolism dysfunction and mitochondria as the major determinants in diabetes and to identify the potential mechanism of gut microbiota in diabetes diseases. We searched relevant articles, which have key information from laboratory experiments, epidemiological evidence, clinical trials, experimental models, meta-analysis and review articles, in PubMed, MEDLINE, EMBASE, Google scholars and Cochrane Controlled Trial Database. We selected 144 full-length articles that met our inclusion and exclusion criteria for complete assessment. We have briefly discussed these associations, challenges, and the need for further research to manage and treat diabetes more efficiently. Diabetes involves the complex network of physiological dysfunction that can be attributed to insulin signaling, genetics, environment, obesity, mitochondria and stress. In recent years, there are intriguing findings regarding gut microbiome as the important regulator of diabetes. Valid approaches are necessary for speeding medical advances but we should find a solution sooner given the burden of the metabolic disorder - What we need is a collaborative venture that may involve laboratories both in academia and industries for the scientific progress and its application for the diabetes control.
Collapse
Affiliation(s)
- Huan Wang
- College of Bioscience and Biotechnology, Shenyang Agricultural University, Shenyang 110866, Liaoning Province, China
- Rutgers Center for Vector Biology, Rutgers University, New Brunswick, NJ 08901, United States
| | - Safoura Akbari-Alavijeh
- Rutgers Center for Vector Biology, Rutgers University, New Brunswick, NJ 08901, United States
- Department of Food Science and Technology, College of Agriculture, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - Ranjit S Parhar
- Department of Biological and Medical Research, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Randy Gaugler
- Rutgers Center for Vector Biology, Rutgers University, New Brunswick, NJ 08901, United States
| | - Sarwar Hashmi
- Rutgers Center for Vector Biology, Rutgers University, New Brunswick, NJ 08901, United States
- Research and Diagnostics, Ghazala and Sanya Hashmi Foundation, Holmdel, NJ 07733, United States
| |
Collapse
|
15
|
Corkey BE. Reactive oxygen species: role in obesity and mitochondrial energy efficiency. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220210. [PMID: 37482778 PMCID: PMC10363708 DOI: 10.1098/rstb.2022.0210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 05/29/2023] [Indexed: 07/25/2023] Open
Abstract
Changes correlating with increasing obesity include insulin resistance, hyperlipidaemia, hyperinsulinaemia, highly processed food and environmental toxins including plastics and air pollution. The relationship between the appearance of each of these potential causes and the onset of obesity is unknown. The cause(s) must precede obesity, the consequence, and temporally relate to its rising incidence. Macronutrients such as carbohydrates or fats are unlikely to cause obesity since these have long been constituents of human diets. Furthermore, food consumption and body weight have been well-regulated in most humans and other species until recent times. Thus, attention must focus on changes that have occurred in the last half-century and the relationship between such changes and specific populations that are impacted. The hypothesis presented here is that substances that have entered our bodies recently cause obesity by generating false and misleading information about energy status. We propose that this misinformation is caused by changes in the oxidation-reduction (redox) potential of metabolites that circulate and communicate to organs throughout the body. Examples are provided of food additives that generate reactive oxygen species and impact redox state, thereby, eliciting inappropriate tissue-specific functional changes, including insulin secretion. Reversal requires identification, neutralization, or removal of these compounds. This article is part of a discussion meeting issue 'Causes of obesity: theories, conjectures and evidence (Part I)'.
Collapse
Affiliation(s)
- Barbara E. Corkey
- Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
16
|
Panda C, Komarnytsky S, Fleming MN, Marsh C, Barron K, Le Brun-Blashka S, Metzger B. Guided Metabolic Detoxification Program Supports Phase II Detoxification Enzymes and Antioxidant Balance in Healthy Participants. Nutrients 2023; 15:2209. [PMID: 37432335 DOI: 10.3390/nu15092209] [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: 04/12/2023] [Revised: 05/03/2023] [Accepted: 05/04/2023] [Indexed: 07/12/2023] Open
Abstract
Adequate antioxidant supply is essential for maintaining metabolic homeostasis and reducing oxidative stress during detoxification. The emerging evidence suggests that certain classes of phytonutrients can help support the detoxification process by stimulating the liver to produce detoxification enzymes or acting as antioxidants that neutralize the harmful effects of free radicals. This study was designed to examine the effects of a guided 28-day metabolic detoxification program in healthy adults. The participants were randomly assigned to consume a whole food, multi-ingredient supplement (n = 14, education and intervention) or control (n = 18, education and healthy meal) daily for the duration of the trial. The whole food supplement contained 37 g/serving of a proprietary, multicomponent nutritional blend in the form of a rehydratable shake. Program readiness was ensured at baseline using a validated self-perceived wellness score and a blood metabolic panel, indicating stable emotional and physical well-being in both groups. No significant changes or adverse effects were found on physical or emotional health, cellular glutathione (GSH) and the GSH:GSSG ratio, porphyrin, and hepatic detoxification biomarkers in urine. The intervention was positively associated with a 23% increase in superoxide dismutase (p = 0.06) and a 13% increase in glutathione S-transferase (p = 0.003) activities in the blood. This resulted in a 40% increase in the total cellular antioxidant capacity (p = 0.001) and a 13% decrease in reactive oxygen species (p = 0.002) in isolated PBMCs from participants in the detoxification group. Our findings indicate that consuming a whole food nutritional intervention as a part of the guided detoxification program supported phase II detoxification, in part, by promoting enhanced free radical scavenging and maintaining redox homeostasis under the body's natural glutathione recycling capacity.
Collapse
Affiliation(s)
- Chinmayee Panda
- Nutrition Innovation Center, Standard Process Inc., 150 N Research Campus Dr, Kannapolis, NC 28081, USA
| | - Slavko Komarnytsky
- Plants for Human Health Institute, NC State University, 600 Laureate Way, Kannapolis, NC 28081, USA
- Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, 400 Dan Allen Drive, Raleigh, NC 27695, USA
| | - Michelle Norton Fleming
- College of Chiropractic, Northwestern Health Sciences University, 2501 W 84th Street, Bloomington, MN 55431, USA
| | - Carissa Marsh
- College of Chiropractic, Northwestern Health Sciences University, 2501 W 84th Street, Bloomington, MN 55431, USA
| | - Keri Barron
- Nutrition Innovation Center, Standard Process Inc., 150 N Research Campus Dr, Kannapolis, NC 28081, USA
| | - Sara Le Brun-Blashka
- Nutrition Innovation Center, Standard Process Inc., 150 N Research Campus Dr, Kannapolis, NC 28081, USA
| | - Brandon Metzger
- Nutrition Innovation Center, Standard Process Inc., 150 N Research Campus Dr, Kannapolis, NC 28081, USA
| |
Collapse
|
17
|
The potential role of environmental factors in modulating mitochondrial DNA epigenetic marks. VITAMINS AND HORMONES 2023; 122:107-145. [PMID: 36863791 DOI: 10.1016/bs.vh.2023.01.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Many studies implicate mitochondrial dysfunction in the development and progression of numerous chronic diseases. Mitochondria are responsible for most cellular energy production, and unlike other cytoplasmic organelles, mitochondria contain their own genome. Most research to date, through investigating mitochondrial DNA copy number, has focused on larger structural changes or alterations to the entire mitochondrial genome and their role in human disease. Using these methods, mitochondrial dysfunction has been linked to cancers, cardiovascular disease, and metabolic health. However, like the nuclear genome, the mitochondrial genome may experience epigenetic alterations, including DNA methylation that may partially explain some of the health effects of various exposures. Recently, there has been a movement to understand human health and disease within the context of the exposome, which aims to describe and quantify the entirety of all exposures people encounter throughout their lives. These include, among others, environmental pollutants, occupational exposures, heavy metals, and lifestyle and behavioral factors. In this chapter, we summarize the current research on mitochondria and human health, provide an overview of the current knowledge on mitochondrial epigenetics, and describe the experimental and epidemiologic studies that have investigated particular exposures and their relationships with mitochondrial epigenetic modifications. We conclude the chapter with suggestions for future directions in epidemiologic and experimental research that is needed to advance the growing field of mitochondrial epigenetics.
Collapse
|
18
|
Giamaki D, Tsiotsiou M, Oglou SC, Touraki M. Interactions of Bisphenol A with Artemia franciscana and the ameliorative effect of probiotics. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2023; 98:104064. [PMID: 36640920 DOI: 10.1016/j.etap.2023.104064] [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: 09/15/2022] [Revised: 11/25/2022] [Accepted: 01/09/2023] [Indexed: 06/17/2023]
Abstract
In the present study, the bidirectional interactions of Artemia franciscana with BPA, administered either alone or following treatment with the probiotics Bacillus subtilis, Lactococcus lactis or Lactobacillus plantarum, were evaluated. A 24 h exposure to BPA below LC50 induced oxidative stress to Artemia, indicated by diminished activity of superoxide dismutase, glutathione reductase, glutathione transferase and phenoloxidase, increased lipid peroxidation and decreased survival. Probiotic treatment prior to BPA exposure, led to increased survival, reduced lipid peroxidation and increased enzyme activities. BPA quantification in Artemia and its culture medium, showed a time dependent reduction in its levels, more evident in probiotic series, indicating its biotransformation. ESI-MS analysis confirmed the presence of the tentative BPA metabolites hydroquinone and BPA-sulfate, while BPA-disulfate formation was confirmed in only in the probiotic series. Our results provide evidence that probiotics alleviate the oxidative stress response induced by BPA, by enhancing the BPA biotransformation ability of Artemia.
Collapse
Affiliation(s)
- Despoina Giamaki
- Laboratory of General Biology, Division of Genetics, Development and Molecular Biology, Department of Biology, School of Sciences, Aristotle University of Thessaloniki (A.U.TH.), 54 124 Thessaloniki, Greece.
| | - Malamati Tsiotsiou
- Laboratory of General Biology, Division of Genetics, Development and Molecular Biology, Department of Biology, School of Sciences, Aristotle University of Thessaloniki (A.U.TH.), 54 124 Thessaloniki, Greece.
| | - Sevnta Chousein Oglou
- Laboratory of General Biology, Division of Genetics, Development and Molecular Biology, Department of Biology, School of Sciences, Aristotle University of Thessaloniki (A.U.TH.), 54 124 Thessaloniki, Greece.
| | - Maria Touraki
- Laboratory of General Biology, Division of Genetics, Development and Molecular Biology, Department of Biology, School of Sciences, Aristotle University of Thessaloniki (A.U.TH.), 54 124 Thessaloniki, Greece.
| |
Collapse
|
19
|
Tris(1,3-dichloro-2-propyl) phosphate is a metabolism-disrupting chemical in male mice. Toxicol Lett 2023; 374:31-39. [PMID: 36493961 PMCID: PMC9869283 DOI: 10.1016/j.toxlet.2022.11.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 10/11/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022]
Abstract
Tris(1,3-dichloro-2-propyl) phosphate (TDCPP) is an organophosphate flame retardant. The primary TDCPP metabolite, bis(1,3-dichloro-2-propyl) phosphate (BDCPP), is detectable in the urine of over 90 % of Americans. Epidemiological studies show sex-specific associations between urinary BDCPP levels and metabolic syndrome, which is an established risk factor for type 2 diabetes, heart disease, and stroke. We used a mouse model to determine whether TDCPP exposure disrupts glucose homeostasis. Six-week old male and female C57BL/6J mice were given ad libitum access to diets containing vehicle (0.1 % DMSO) and TDCPP resulting in the following treatment groups: 0 mg/kg/day, 0.02 mg/kg/day, 1 mg/kg/day, or 100 mg/kg/day. After being on the experimental diet for five weeks without interruption, body composition was analyzed, glucose and insulin tolerance tests were performed, and fasting glucose and insulin levels were quantified. TDCPP at 100 mg/kg/day caused male sex-specific adiposity, fasting hyperglycemia, and insulin resistance. TDCPP-induced modulation of nuclear receptor activation was investigated using an in vitro screen to identify potential mechanisms of metabolic disruption. TDCPP activated farnesoid X receptor (FXR) and pregnane X receptor (PXR), and inhibited the androgen receptor (AR). PXR target genes, but not FXR target genes, were upregulated in livers from mice exposed to 100 mg TDCPP/kg/day. Interestingly, PXR target genes were differentially expressed in livers from both males and females. It remains to be determined whether TDCPP-induced metabolic disruption occurs via modulation of nuclear receptor activity. Taken together, these studies build upon the association of TDCPP exposure and metabolic syndrome in humans by identifying sex-specific effects of TDCPP on glucose homeostasis in mice.
Collapse
|
20
|
Kuretu A, Arineitwe C, Mothibe M, Ngubane P, Khathi A, Sibiya N. Drug-induced mitochondrial toxicity: Risks of developing glucose handling impairments. Front Endocrinol (Lausanne) 2023; 14:1123928. [PMID: 36860368 PMCID: PMC9969099 DOI: 10.3389/fendo.2023.1123928] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/31/2023] [Indexed: 02/15/2023] Open
Abstract
Mitochondrial impairment has been associated with the development of insulin resistance, the hallmark of type 2 diabetes mellitus (T2DM). However, the relationship between mitochondrial impairment and insulin resistance is not fully elucidated due to insufficient evidence to support the hypothesis. Insulin resistance and insulin deficiency are both characterised by excessive production of reactive oxygen species and mitochondrial coupling. Compelling evidence states that improving the function of the mitochondria may provide a positive therapeutic tool for improving insulin sensitivity. There has been a rapid increase in reports of the toxic effects of drugs and pollutants on the mitochondria in recent decades, interestingly correlating with an increase in insulin resistance prevalence. A variety of drug classes have been reported to potentially induce toxicity in the mitochondria leading to skeletal muscle, liver, central nervous system, and kidney injury. With the increase in diabetes prevalence and mitochondrial toxicity, it is therefore imperative to understand how mitochondrial toxicological agents can potentially compromise insulin sensitivity. This review article aims to explore and summarise the correlation between potential mitochondrial dysfunction caused by selected pharmacological agents and its effect on insulin signalling and glucose handling. Additionally, this review highlights the necessity for further studies aimed to understand drug-induced mitochondrial toxicity and the development of insulin resistance.
Collapse
Affiliation(s)
- Auxiliare Kuretu
- Pharmacology Division, Faculty of Pharmacy, Rhodes University, Makhanda, South Africa
| | - Charles Arineitwe
- Pharmacology Division, Faculty of Pharmacy, Rhodes University, Makhanda, South Africa
| | - Mamosheledi Mothibe
- Pharmacology Division, Faculty of Pharmacy, Rhodes University, Makhanda, South Africa
| | - Phikelelani Ngubane
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Andile Khathi
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Ntethelelo Sibiya
- Pharmacology Division, Faculty of Pharmacy, Rhodes University, Makhanda, South Africa
- *Correspondence: Ntethelelo Sibiya,
| |
Collapse
|
21
|
Liang R, Yu L, Liu W, Dong C, Tan Q, Wang M, Ye Z, Zhang Y, Li M, Wang B, Feng X, Zhou M, Chen W. Associations of bifenthrin exposure with glucose homeostasis and type 2 diabetes mellitus in a general Chinese population: Roles of protein carbonylation. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 315:120352. [PMID: 36216181 DOI: 10.1016/j.envpol.2022.120352] [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: 04/11/2022] [Revised: 09/18/2022] [Accepted: 10/01/2022] [Indexed: 06/16/2023]
Abstract
The adverse health effects of pyrethroids exposure have attracted wide concern. We aimed to assess the associations of bifenthrin, a widely used pyrethroid, with glucose homeostasis and risk of type 2 diabetes mellitus (T2DM) and to explore the underlying mechanism. Serum bifenthrin, fasting plasma glucose (FPG), fasting plasma insulin (FPI), and plasma protein carbonyl (PCO) were determined among 3822 participants from the Wuhan-Zhuhai cohort. Glucose homeostasis was evaluated by FPG, FPI, homeostasis model assessment of insulin resistance (HOMA-IR), impaired fasting glucose (IFG), and abnormal glucose regulation (AGR). The associations of serum bifenthrin with glucose homeostasis and risk of T2DM were assessed by generalized linear models and logistic regression models. The role of PCO in the above associations was evaluated by mediation analyses. After adjusting for covariates, each 2-fold increase in serum bifenthrin was associated with a 0.21 mmol/L increase in FPG and a 5.19%, 10.49%, and 12.18% increase in FPI, HOMA-IR, and PCO levels, respectively. Monotonically elevated ORs of IFG and AGR (all P and P for trend <0.05), but not T2DM (P > 0.05) were detected to be associated with increased bifenthrin. Compared with the participants with low bifenthrin and low PCO, participants with high bifenthrin exposure and high PCO showed a 0.40 mmol/L, 11.07%, and 22.50% increase in FPG, FPI, and HOMA-IR, as well as a 119.97% and 48.88% increase in risks of IFG and AGR, respectively (P for trend <0.05). Moreover, PCO mediated 13.61%-24.98% of the serum bifenthrin-associated glucose dyshomeostasis. The study suggested that bifenthrin exposure was dose-dependently associated with glucose dyshomeostasis in the general Chinese urban adults, and these associations were exacerbated and partly mediated by PCO. Given that other pollutants were not included in this study, the effect of co-exposure of pyrethroids with multiple pollutants is necessary to be considered in future studies.
Collapse
Affiliation(s)
- Ruyi Liang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Linling Yu
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Wei Liu
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Chaoqian Dong
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Qiyou Tan
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Mengyi Wang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Zi Ye
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Yongfang Zhang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Minjing Li
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Bin Wang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Xiaobing Feng
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Min Zhou
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| | - Weihong Chen
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| |
Collapse
|
22
|
Kasongo AA, Leroux M, Amrouche-Mekkioui I, Belhadji-Domecq M, Aguer C. BPA exposure in L6 myotubes increased basal glucose metabolism in an estrogen receptor-dependent manner but induced insulin resistance. Food Chem Toxicol 2022; 170:113505. [DOI: 10.1016/j.fct.2022.113505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/24/2022] [Accepted: 10/27/2022] [Indexed: 11/08/2022]
|
23
|
Nayak D, Adiga D, Khan NG, Rai PS, Dsouza HS, Chakrabarty S, Gassman NR, Kabekkodu SP. Impact of Bisphenol A on Structure and Function of Mitochondria: A Critical Review. REVIEWS OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2022; 260:10. [DOI: 10.1007/s44169-022-00011-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 10/26/2022] [Indexed: 04/02/2024]
Abstract
AbstractBisphenol A (BPA) is an industrial chemical used extensively to manufacture polycarbonate plastics and epoxy resins. Because of its estrogen-mimicking properties, BPA acts as an endocrine-disrupting chemical. It has gained attention due to its high chances of daily and constant human exposure, bioaccumulation, and the ability to cause cellular toxicities and diseases at extremely low doses. Several elegant studies have shown that BPA can exert cellular toxicities by interfering with the structure and function of mitochondria, leading to mitochondrial dysfunction. Exposure to BPA results in oxidative stress and alterations in mitochondrial DNA (mtDNA), mitochondrial biogenesis, bioenergetics, mitochondrial membrane potential (MMP) decline, mitophagy, and apoptosis. Accumulation of reactive oxygen species (ROS) in conjunction with oxidative damage may be responsible for causing BPA-mediated cellular toxicity. Thus, several reports have suggested using antioxidant treatment to mitigate the toxicological effects of BPA. The present literature review emphasizes the adverse effects of BPA on mitochondria, with a comprehensive note on the molecular aspects of the structural and functional alterations in mitochondria in response to BPA exposure. The review also confers the possible approaches to alleviate BPA-mediated oxidative damage and the existing knowledge gaps in this emerging area of research.
Collapse
|
24
|
Environmental Chemical Exposures and Mitochondrial Dysfunction: a Review of Recent Literature. Curr Environ Health Rep 2022; 9:631-649. [PMID: 35902457 PMCID: PMC9729331 DOI: 10.1007/s40572-022-00371-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/20/2022] [Indexed: 01/31/2023]
Abstract
PURPOSE OF REVIEW Mitochondria play various roles that are important for cell function and survival; therefore, significant mitochondrial dysfunction may have chronic consequences that extend beyond the cell. Mitochondria are already susceptible to damage, which may be exacerbated by environmental exposures. Therefore, the aim of this review is to summarize the recent literature (2012-2022) looking at the effects of six ubiquitous classes of compounds on mitochondrial dysfunction in human populations. RECENT FINDINGS The literature suggests that there are a number of biomarkers that are commonly used to identify mitochondrial dysfunction, each with certain advantages and limitations. Classes of environmental toxicants such as polycyclic aromatic hydrocarbons, air pollutants, heavy metals, endocrine-disrupting compounds, pesticides, and nanomaterials can damage the mitochondria in varied ways, with changes in mtDNA copy number and measures of oxidative damage the most commonly measured in human populations. Other significant biomarkers include changes in mitochondrial membrane potential, calcium levels, and ATP levels. This review identifies the biomarkers that are commonly used to characterize mitochondrial dysfunction but suggests that emerging mitochondrial biomarkers, such as cell-free mitochondria and blood cardiolipin levels, may provide greater insight into the impacts of exposures on mitochondrial function. This review identifies that the mtDNA copy number and measures of oxidative damage are commonly used to characterize mitochondrial dysfunction, but suggests using novel approaches in addition to well-characterized ones to create standardized protocols. We identified a dearth of studies on mitochondrial dysfunction in human populations exposed to metals, endocrine-disrupting chemicals, pesticides, and nanoparticles as a gap in knowledge that needs attention.
Collapse
|
25
|
Prabhu NB, Adiga D, Kabekkodu SP, Bhat SK, Satyamoorthy K, Rai PS. Bisphenol A exposure modulates reproductive and endocrine system, mitochondrial function and cellular senescence in female adult rats: A hallmarks of polycystic ovarian syndrome phenotype. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2022; 96:104010. [PMID: 36334871 DOI: 10.1016/j.etap.2022.104010] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 10/29/2022] [Accepted: 11/01/2022] [Indexed: 06/16/2023]
Abstract
Bisphenol A (BPA) mimics estrogen and consequently suspected to be detrimental to female reproductive system. Biomonitoring confirms the BPA burden in body leading to a complex condition called polycystic ovarian syndrome (PCOS) which is frequently attributed to female infertility. Due to unclear precise molecular pathomechanisms of BPA in PCOS, we intend to examine the molecular mechanisms of the reproductive, endocrine, mitochondrial features, and cellular senescence in BPA-treated rats. We analyzed vaginal smears and ovarian follicles using microscope, assessed sex hormones by ELISA, analyzed BPA target gene expression by semi-quantitative RT-PCR, assessed senescence induction by β-galactosidase staining and immunofluorescence in BPA-treated rats. Our data showed hormonal imbalance, impaired folliculogenesis, abnormal expression patterns of target genes, CDKN2A overexpression and enhanced ROS levels in BPA-treated rats. This study provides insights on the effects of BPA exposure on ovulatory, hormonal, mitochondrial dysfunction, and senescence that benefit in better understanding of PCOS induced by BPA.
Collapse
Affiliation(s)
- Navya B Prabhu
- Department of Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Divya Adiga
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Shashikala K Bhat
- Department of Obstetrics and Gynaecology, Dr. T.M.A Pai Hospital, Melaka Manipal Medical College, Manipal Academy of Higher Education, Manipal 576101, India
| | - Kapaettu Satyamoorthy
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Padmalatha S Rai
- Department of Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, India.
| |
Collapse
|
26
|
Baumert BO, Goodrich JA, Hu X, Walker DI, Alderete TL, Chen Z, Valvi D, Rock S, Berhane K, Gilliland FD, Goran MI, Jones DP, Conti DV, Chatzi L. Plasma concentrations of lipophilic persistent organic pollutants and glucose homeostasis in youth populations. ENVIRONMENTAL RESEARCH 2022; 212:113296. [PMID: 35447156 PMCID: PMC9831292 DOI: 10.1016/j.envres.2022.113296] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 03/15/2022] [Accepted: 04/09/2022] [Indexed: 05/16/2023]
Abstract
BACKGROUND Exposure to lipophilic persistent organic pollutants (POPs) is ubiquitous. POPs are metabolic disrupting chemicals and are potentially diabetogenic. METHODS Using a multi-cohort study including overweight adolescents from the Study of Latino Adolescents at Risk (SOLAR, N = 301, 2001-2012) and young adults from the Southern California Children's Health Study (CHS, N = 135, 2014-2018), we examined associations of POPs and risk factors for type 2 diabetes. SOLAR participants underwent annual visits for a median of 2.2 years and CHS participants performed a single visit, during which a 2-h oral glucose tolerance test was performed. Linear mixed models were used to examine associations between plasma concentrations of POPs [4,4'-dichlorodiphenyldichloroethylene (4,4'-DDE), hexachlorobenzene (HCB), PCBs-153, 138, 118, 180 and PBDEs-154, 153, 100, 85, 47] and changes in glucose homeostasis across age and pubertal stage. RESULTS In SOLAR, exposure to HCB, PCB-118, and PBDE-153 was associated with dysregulated glucose metabolism. For example, each two-fold increase in HCB was associated with approximately 2 mg/dL higher glucose concentrations at 30 min (p = 0.001), 45 min (p = 0.0006), and 60 min (p = 0.03) post glucose challenge. Compared to individuals with low levels of PCB-118, individuals with high levels exhibited a 4.7 mg/dL (p = 0.02) higher glucose concentration at 15 min and a 3.6 mg/dL (p = 0.01) higher glucose concentration at 30 min. The effects observed with exposure to organochlorine compounds were independent of pubertal stages. PBDE-153 was associated with the development of dysregulated glucose metabolism beginning in late puberty. At Tanner stage 4, exposure to PBDE-153 was associated with a 12.7 mg/dL higher 60-min glucose concentration (p = 0.009) and a 16.1 mg*dl-1*hr-1 higher glucose AUC (p = 0.01). These associations persisted at Tanner 5. In CHS, PBDE-153 and total PBDE were associated with similar increases in glucose concentrations. CONCLUSION Our results suggest that childhood exposure to lipophilic POPs is associated with dysregulated glucose metabolism.
Collapse
Affiliation(s)
- Brittney O Baumert
- Department of Preventative Medicine, University of Southern California, Los Angeles, CA, United States.
| | - Jesse A Goodrich
- Department of Preventative Medicine, University of Southern California, Los Angeles, CA, United States
| | - Xin Hu
- Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, GA, United States
| | - Douglas I Walker
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Tanya L Alderete
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
| | - Zhanghua Chen
- Department of Preventative Medicine, University of Southern California, Los Angeles, CA, United States
| | - Damaskini Valvi
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Sarah Rock
- Department of Preventative Medicine, University of Southern California, Los Angeles, CA, United States
| | - Kiros Berhane
- Department of Biostatistics, Columbia University, New York, NY, United States
| | - Frank D Gilliland
- Department of Preventative Medicine, University of Southern California, Los Angeles, CA, United States
| | - Michael I Goran
- Department of Pediatrics, Children's Hospital of Los Angeles, The Saban Research Institute, United States
| | - Dean P Jones
- Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, GA, United States
| | - David V Conti
- Department of Preventative Medicine, University of Southern California, Los Angeles, CA, United States
| | - Leda Chatzi
- Department of Preventative Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
27
|
Motto AE, Lawson-Evi P, Eklu-Gadegbeku K. Antidiabetic and antioxidant potential of total extract and supernatant fraction of the roots of Anogeissus leiocarpus in HFD-fed and Streptozocin -induced diabetic rats. Biomed Pharmacother 2022; 154:113578. [PMID: 36027612 DOI: 10.1016/j.biopha.2022.113578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 08/09/2022] [Accepted: 08/17/2022] [Indexed: 11/02/2022] Open
Abstract
The aim of this study was to evaluate the antidiabetic properties of hydro alcoholic extract and supernatant fraction of the roots of Anogeissus leiocarpus, a plant used by traditional healers to treat Diabetes mellitus. Diabetes mellitus was induced by a single intraperitoneal administration of Streptozocin to Sprague Dawley rats under a fructose-enriched fat diet. Diabetic rats were treated with 500 mg/kg of total extract and 100 mg/kg of supernatant. The antidiabetic activity was assessed by measuring blood glucose level, lipid profile, insulin and biochemical parameters together with the antioxidant potential. The administration of total extract and supernatant exhibited significant decrease (p < 0.01) of the blood glucose level in the diabetic rats after 7 days of treatment compared to the diabetic rats. A significant reduction in the serum concentrations of cholesterol (19.7 %) and triglycerides (56.7 %) was observed in the treated diabetic rats. The levels of insulin did not differ across all the groups. However, compared to diabetic rats, HOMA-IR (Homeostasis Model Assessment for Insulin-resistance) and HOMA-β (Homeostasis Model Assessment for β cell function) showed a statistical decrease in insulin resistance and an increase in pancreatic β cell function in the treated diabetic rats. Moreover, total extract and supernatant significantly increased GSH level and decreased lipid peroxidation because of their antioxidant properties. In comparison, the supernatant fraction exerted stronger antidiabetic and antioxidant effects than the total extract. Hence, the roots of Anogeissus leiocarpus are a potent antidiabetic agent that can be developed as an alternative medicine for diabetes and its complications.
Collapse
Affiliation(s)
- Aku Enam Motto
- Laboratory of Physiology/Pharmacology, Unit of Pathophysiology, Bioactive Substances and Safety, Faculty of Sciences, University of Lomé, BP 1515, Togo.
| | - Povi Lawson-Evi
- Laboratory of Physiology/Pharmacology, Unit of Pathophysiology, Bioactive Substances and Safety, Faculty of Sciences, University of Lomé, BP 1515, Togo
| | - Kwashie Eklu-Gadegbeku
- Laboratory of Physiology/Pharmacology, Unit of Pathophysiology, Bioactive Substances and Safety, Faculty of Sciences, University of Lomé, BP 1515, Togo
| |
Collapse
|
28
|
Anastasiou IA, Eleftheriadou I, Tentolouris A, Sarantis P, Angelopoulou A, Katsaouni A, Mourouzis I, Karamouzis MV, Gorgoulis V, Pantos C, Tentolouris N. Low concentrations of bisphenol A promote the activation of the mitochondrial apoptotic pathway on Beta-TC-6 cells via the generation of intracellular reactive oxygen species and mitochondrial superoxide. J Biochem Mol Toxicol 2022; 36:e23099. [PMID: 35593412 DOI: 10.1002/jbt.23099] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 04/03/2022] [Accepted: 05/11/2022] [Indexed: 11/10/2022]
Abstract
Τhe natural history of type 2 diabetes mellitus is characterized by a progressive loss of pancreatic beta cell function and insulin resistance. Bisphenol A (BPA) is an endocrine-disrupting chemical that is used widely in industry; people are exposed to BPA and its products daily. Studies have delineated that BPA alters the function of pancreatic beta cells. Herein, we examined the effect of low doses of BPA on pancreatic beta cell viability and apoptosis and we tried to elucidate the mechanisms involved in these processes. Beta-TC-6 (ATCC® CRL-11506™) cells were cultured with a medium containing the following dilutions of BPA: 0.002, 0.02, 0.1, 0.2, 2 μΜ up to 72 h. We examined the viability and adenosine triphosphate (ATP) levels of cells. Then, we measured apoptosis, cell cycle, and insulin levels. We quantified the levels of proteins implicated in the mitochondrial pathway of apoptosis; and finally, we quantified the intracellular reactive oxygen species and mitochondrial superoxide. We found that the exposure of Beta-TC-6 cells to BPA results in a decrease in cell viability, ATP levels, and an increase in insulin levels. We found an increase in apoptosis levels and a decrease in cell cycle levels. In addition, we provide evidence of the levels of apoptotic proteins. Finally, we found an increase in the cellular reactive oxygen species and mitochondrial superoxide production. Exposure to low concentrations of BPA triggers the mitochondrial pathway of apoptosis via the generation of intracellular reactive oxygen species and mitochondrial superoxide on Beta-TC-6 cells in a dose-dependent way.
Collapse
Affiliation(s)
- Ioanna A Anastasiou
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Ioanna Eleftheriadou
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Anastasios Tentolouris
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Panagiotis Sarantis
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Andriani Angelopoulou
- Laboratory of Histology-Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Athanasia Katsaouni
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Iordanis Mourouzis
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Michalis V Karamouzis
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Vasilios Gorgoulis
- Laboratory of Histology-Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Constantinos Pantos
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Tentolouris
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| |
Collapse
|
29
|
Qin W, Ren X, Zhao L, Guo L. Exposure to perfluorooctane sulfonate reduced cell viability and insulin release capacity of β cells. J Environ Sci (China) 2022; 115:162-172. [PMID: 34969446 DOI: 10.1016/j.jes.2021.07.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 07/05/2021] [Accepted: 07/05/2021] [Indexed: 05/20/2023]
Abstract
Per- and polyfluoroalkyl substances (PFAS) are found to have multiple adverse outcomes on human health. Recently, epidemiological and toxicological studies showed that exposure to PFAS had adverse impacts on pancreas and showed association with insulin abnormalities. To explore how PFAS may contribute to diabetes, we studied impacts of perfluorooctane sulfonate (PFOS) on cell viability and insulin release capacity of pancreatic β cells by using in vivo and in vitro methods. We found that 28-day administration with PFOS (10 mg/(kg body weight•day)) caused reductions of pancreas weight and islet size in male mice. PFOS administration also led to lower serum insulin level both in fasting state and after glucose infusion among male mice. For cell-based in vitro bioassay, we used mouse β-TC-6 cancer cells and found 48-hr exposure to PFOS decreased the cell viability at 50 μmol/L. By measuring insulin content in supernatant, 48-hr pretreatment of PFOS (100 μmol/L) decreased the insulin release capacity of β-TC-6 cells after glucose stimulation. Although these concentrations were higher than the environmental concentration of PFOS, it might be reasonable for high concentration of PFOS to exert observable toxic effects in mice considering mice had a faster removal efficiency of PFOS than human. PFOS exposure (50 μmol/L) to β-TC-6 cells induced intracellular accumulation of reactive oxidative specie (ROS). Excessive ROS induced the reactive toxicity of cells, which eventually invoke apoptosis and necrosis. Results in this study provide evidence for the possible causal link of exposure to PFOS and diabetes risk.
Collapse
Affiliation(s)
- Weiping Qin
- State Key Laboratory of Environmental Chemistry and Eco-toxicology, Research Center for Eco-environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Xiaomin Ren
- State Key Laboratory of Environmental Chemistry and Eco-toxicology, Research Center for Eco-environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Lixia Zhao
- State Key Laboratory of Environmental Chemistry and Eco-toxicology, Research Center for Eco-environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Lianghong Guo
- Institute of Environmental and Health Sciences, China Jiliang University, Hangzhou 310018, China.
| |
Collapse
|
30
|
Heindel JJ, Howard S, Agay-Shay K, Arrebola JP, Audouze K, Babin PJ, Barouki R, Bansal A, Blanc E, Cave MC, Chatterjee S, Chevalier N, Choudhury M, Collier D, Connolly L, Coumoul X, Garruti G, Gilbertson M, Hoepner LA, Holloway AC, Howell G, Kassotis CD, Kay MK, Kim MJ, Lagadic-Gossmann D, Langouet S, Legrand A, Li Z, Le Mentec H, Lind L, Monica Lind P, Lustig RH, Martin-Chouly C, Munic Kos V, Podechard N, Roepke TA, Sargis RM, Starling A, Tomlinson CR, Touma C, Vondracek J, Vom Saal F, Blumberg B. Obesity II: Establishing causal links between chemical exposures and obesity. Biochem Pharmacol 2022; 199:115015. [PMID: 35395240 PMCID: PMC9124454 DOI: 10.1016/j.bcp.2022.115015] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 03/12/2022] [Accepted: 03/15/2022] [Indexed: 02/06/2023]
Abstract
Obesity is a multifactorial disease with both genetic and environmental components. The prevailing view is that obesity results from an imbalance between energy intake and expenditure caused by overeating and insufficient exercise. We describe another environmental element that can alter the balance between energy intake and energy expenditure: obesogens. Obesogens are a subset of environmental chemicals that act as endocrine disruptors affecting metabolic endpoints. The obesogen hypothesis posits that exposure to endocrine disruptors and other chemicals can alter the development and function of the adipose tissue, liver, pancreas, gastrointestinal tract, and brain, thus changing the set point for control of metabolism. Obesogens can determine how much food is needed to maintain homeostasis and thereby increase the susceptibility to obesity. The most sensitive time for obesogen action is in utero and early childhood, in part via epigenetic programming that can be transmitted to future generations. This review explores the evidence supporting the obesogen hypothesis and highlights knowledge gaps that have prevented widespread acceptance as a contributor to the obesity pandemic. Critically, the obesogen hypothesis changes the narrative from curing obesity to preventing obesity.
Collapse
Affiliation(s)
- Jerrold J Heindel
- Healthy Environment and Endocrine Disruptor Strategies, Commonweal, Bolinas, CA 92924, USA.
| | - Sarah Howard
- Healthy Environment and Endocrine Disruptor Strategies, Commonweal, Bolinas, CA 92924, USA
| | - Keren Agay-Shay
- Health and Environment Research (HER) Lab, The Azrieli Faculty of Medicine, Bar Ilan University, Israel
| | - Juan P Arrebola
- Department of Preventive Medicine and Public Health University of Granada, Granada, Spain
| | - Karine Audouze
- Department of Systems Biology and Bioinformatics, University of Paris, INSERM, T3S, Paris France
| | - Patrick J Babin
- Department of Life and Health Sciences, University of Bordeaux, INSERM, Pessac France
| | - Robert Barouki
- Department of Biochemistry, University of Paris, INSERM, T3S, 75006 Paris, France
| | - Amita Bansal
- College of Health & Medicine, Australian National University, Canberra, Australia
| | - Etienne Blanc
- Department of Biochemistry, University of Paris, INSERM, T3S, 75006 Paris, France
| | - Matthew C Cave
- Division of Gastroenterology, Hepatology and Nutrition, University of Louisville, Louisville, KY 40402, USA
| | - Saurabh Chatterjee
- Environmental Health and Disease Laboratory, University of South Carolina, Columbia, SC 29208, USA
| | - Nicolas Chevalier
- Obstetrics and Gynecology, University of Cote d'Azur, Cote d'Azur, France
| | - Mahua Choudhury
- College of Pharmacy, Texas A&M University, College Station, TX 77843, USA
| | - David Collier
- Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Lisa Connolly
- The Institute for Global Food Security, School of Biological Sciences, Queen's University, Belfast, Northern Ireland, UK
| | - Xavier Coumoul
- Department of Biochemistry, University of Paris, INSERM, T3S, 75006 Paris, France
| | - Gabriella Garruti
- Department of Endocrinology, University of Bari "Aldo Moro," Bari, Italy
| | - Michael Gilbertson
- Occupational and Environmental Health Research Group, University of Stirling, Stirling, Scotland
| | - Lori A Hoepner
- Department of Environmental and Occupational Health Sciences, School of Public Health, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Alison C Holloway
- McMaster University, Department of Obstetrics and Gynecology, Hamilton, Ontario, CA, USA
| | - George Howell
- Center for Environmental Health Sciences, Mississippi State University, Mississippi State, MS 39762, USA
| | - Christopher D Kassotis
- Institute of Environmental Health Sciences and Department of Pharmacology, Wayne State University, Detroit, MI 48202, USA
| | - Mathew K Kay
- College of Pharmacy, Texas A&M University, College Station, TX 77843, USA
| | - Min Ji Kim
- Sorbonne Paris Nord University, Bobigny, INSERM U1124 (T3S), Paris, France
| | | | - Sophie Langouet
- Univ Rennes, INSERM EHESP, IRSET UMR_5S 1085, 35000 Rennes, France
| | - Antoine Legrand
- Sorbonne Paris Nord University, Bobigny, INSERM U1124 (T3S), Paris, France
| | - Zhuorui Li
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Helene Le Mentec
- Sorbonne Paris Nord University, Bobigny, INSERM U1124 (T3S), Paris, France
| | - Lars Lind
- Clinical Epidemiology, Department of Medical Sciences, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - P Monica Lind
- Occupational and Environmental Medicine, Department of Medical Sciences, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Robert H Lustig
- Division of Endocrinology, Department of Pediatrics, University of California San Francisco, CA 94143, USA
| | | | - Vesna Munic Kos
- Department of Physiology and Pharmacology, Karolinska Institute, Solna, Sweden
| | - Normand Podechard
- Sorbonne Paris Nord University, Bobigny, INSERM U1124 (T3S), Paris, France
| | - Troy A Roepke
- Department of Animal Science, School of Environmental and Biological Science, Rutgers University, New Brunswick, NJ 08901, USA
| | - Robert M Sargis
- Division of Endocrinology, Diabetes and Metabolism, The University of Illinois at Chicago, Chicago, Il 60612, USA
| | - Anne Starling
- Department of Epidemiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Craig R Tomlinson
- Norris Cotton Cancer Center, Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Charbel Touma
- Sorbonne Paris Nord University, Bobigny, INSERM U1124 (T3S), Paris, France
| | - Jan Vondracek
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | - Frederick Vom Saal
- Division of Biological Sciences, The University of Missouri, Columbia, MO 65211, USA
| | - Bruce Blumberg
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| |
Collapse
|
31
|
Sellami A, Réau M, Montes M, Lagarde N. Review of in silico studies dedicated to the nuclear receptor family: Therapeutic prospects and toxicological concerns. Front Endocrinol (Lausanne) 2022; 13:986016. [PMID: 36176461 PMCID: PMC9513233 DOI: 10.3389/fendo.2022.986016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Being in the center of both therapeutic and toxicological concerns, NRs are widely studied for drug discovery application but also to unravel the potential toxicity of environmental compounds such as pesticides, cosmetics or additives. High throughput screening campaigns (HTS) are largely used to detect compounds able to interact with this protein family for both therapeutic and toxicological purposes. These methods lead to a large amount of data requiring the use of computational approaches for a robust and correct analysis and interpretation. The output data can be used to build predictive models to forecast the behavior of new chemicals based on their in vitro activities. This atrticle is a review of the studies published in the last decade and dedicated to NR ligands in silico prediction for both therapeutic and toxicological purposes. Over 100 articles concerning 14 NR subfamilies were carefully read and analyzed in order to retrieve the most commonly used computational methods to develop predictive models, to retrieve the databases deployed in the model building process and to pinpoint some of the limitations they faced.
Collapse
|
32
|
Duarte-Hospital C, Tête A, Brial F, Benoit L, Koual M, Tomkiewicz C, Kim MJ, Blanc EB, Coumoul X, Bortoli S. Mitochondrial Dysfunction as a Hallmark of Environmental Injury. Cells 2021; 11:cells11010110. [PMID: 35011671 PMCID: PMC8750015 DOI: 10.3390/cells11010110] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/24/2021] [Accepted: 12/27/2021] [Indexed: 02/07/2023] Open
Abstract
Environmental factors including diet, sedentary lifestyle and exposure to pollutants largely influence human health throughout life. Cellular and molecular events triggered by an exposure to environmental pollutants are extremely variable and depend on the age, the chronicity and the doses of exposure. Only a fraction of all relevant mechanisms involved in the onset and progression of pathologies in response to toxicants has probably been identified. Mitochondria are central hubs of metabolic and cell signaling responsible for a large variety of biochemical processes, including oxidative stress, metabolite production, energy transduction, hormone synthesis, and apoptosis. Growing evidence highlights mitochondrial dysfunction as a major hallmark of environmental insults. Here, we present mitochondria as crucial organelles for healthy metabolic homeostasis and whose dysfunction induces critical adverse effects. Then, we review the multiple mechanisms of action of pollutants causing mitochondrial toxicity in link with chronic diseases. We propose the Aryl hydrocarbon Receptor (AhR) as a model of “exposome receptor”, whose activation by environmental pollutants leads to various toxic events through mitochondrial dysfunction. Finally, we provide some remarks related to mitotoxicity and risk assessment.
Collapse
Affiliation(s)
- Carolina Duarte-Hospital
- Environmental Toxicity, Therapeutic Targets, Cellular Signaling and Biomarkers, T3S, INSERM UMR-S 1124, F-75006 Paris, France; (C.D.-H.); (A.T.); (F.B.); (L.B.); (M.K.); (C.T.); (M.J.K.); (E.B.B.)
- Faculty of Sciences, Université de Paris, F-75006 Paris, France
| | - Arnaud Tête
- Environmental Toxicity, Therapeutic Targets, Cellular Signaling and Biomarkers, T3S, INSERM UMR-S 1124, F-75006 Paris, France; (C.D.-H.); (A.T.); (F.B.); (L.B.); (M.K.); (C.T.); (M.J.K.); (E.B.B.)
- Faculty of Sciences, Université de Paris, F-75006 Paris, France
| | - François Brial
- Environmental Toxicity, Therapeutic Targets, Cellular Signaling and Biomarkers, T3S, INSERM UMR-S 1124, F-75006 Paris, France; (C.D.-H.); (A.T.); (F.B.); (L.B.); (M.K.); (C.T.); (M.J.K.); (E.B.B.)
| | - Louise Benoit
- Environmental Toxicity, Therapeutic Targets, Cellular Signaling and Biomarkers, T3S, INSERM UMR-S 1124, F-75006 Paris, France; (C.D.-H.); (A.T.); (F.B.); (L.B.); (M.K.); (C.T.); (M.J.K.); (E.B.B.)
- Faculty of Sciences, Université de Paris, F-75006 Paris, France
| | - Meriem Koual
- Environmental Toxicity, Therapeutic Targets, Cellular Signaling and Biomarkers, T3S, INSERM UMR-S 1124, F-75006 Paris, France; (C.D.-H.); (A.T.); (F.B.); (L.B.); (M.K.); (C.T.); (M.J.K.); (E.B.B.)
- Faculty of Sciences, Université de Paris, F-75006 Paris, France
| | - Céline Tomkiewicz
- Environmental Toxicity, Therapeutic Targets, Cellular Signaling and Biomarkers, T3S, INSERM UMR-S 1124, F-75006 Paris, France; (C.D.-H.); (A.T.); (F.B.); (L.B.); (M.K.); (C.T.); (M.J.K.); (E.B.B.)
- Faculty of Sciences, Université de Paris, F-75006 Paris, France
| | - Min Ji Kim
- Environmental Toxicity, Therapeutic Targets, Cellular Signaling and Biomarkers, T3S, INSERM UMR-S 1124, F-75006 Paris, France; (C.D.-H.); (A.T.); (F.B.); (L.B.); (M.K.); (C.T.); (M.J.K.); (E.B.B.)
- Université Sorbonne Paris Nord, F-93000 Bobigny, France
| | - Etienne B. Blanc
- Environmental Toxicity, Therapeutic Targets, Cellular Signaling and Biomarkers, T3S, INSERM UMR-S 1124, F-75006 Paris, France; (C.D.-H.); (A.T.); (F.B.); (L.B.); (M.K.); (C.T.); (M.J.K.); (E.B.B.)
- Faculty of Sciences, Université de Paris, F-75006 Paris, France
| | - Xavier Coumoul
- Environmental Toxicity, Therapeutic Targets, Cellular Signaling and Biomarkers, T3S, INSERM UMR-S 1124, F-75006 Paris, France; (C.D.-H.); (A.T.); (F.B.); (L.B.); (M.K.); (C.T.); (M.J.K.); (E.B.B.)
- Faculty of Sciences, Université de Paris, F-75006 Paris, France
- Correspondence: (X.C.); (S.B.); Tel.: +33-1-76-53-43-70 (S.B.)
| | - Sylvie Bortoli
- Environmental Toxicity, Therapeutic Targets, Cellular Signaling and Biomarkers, T3S, INSERM UMR-S 1124, F-75006 Paris, France; (C.D.-H.); (A.T.); (F.B.); (L.B.); (M.K.); (C.T.); (M.J.K.); (E.B.B.)
- Faculty of Sciences, Université de Paris, F-75006 Paris, France
- Correspondence: (X.C.); (S.B.); Tel.: +33-1-76-53-43-70 (S.B.)
| |
Collapse
|
33
|
Zhou Z, Goodrich JM, Strakovsky RS. Mitochondrial Epigenetics and Environmental Health: Making a Case for Endocrine Disrupting Chemicals. Toxicol Sci 2021; 178:16-25. [PMID: 32777053 DOI: 10.1093/toxsci/kfaa129] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Recent studies implicate mitochondrial dysfunction in the development and progression of numerous chronic diseases, which may be partially due to modifications in mitochondrial DNA (mtDNA). There is also mounting evidence that epigenetic modifications to mtDNA may be an additional layer of regulation that controls mitochondrial biogenesis and function. Several environmental factors (eg, smoking, air pollution) have been associated with altered mtDNA methylation in a handful of mechanistic studies and in observational human studies. However, little is understood about other environmental contaminants that induce mtDNA epigenetic changes. Numerous environmental toxicants are classified as endocrine disrupting chemicals (EDCs). Beyond their actions on hormonal pathways, EDC exposure is associated with elevated oxidative stress, which may occur through or result in mitochondrial dysfunction. Although only a few studies have assessed the impacts of EDCs on mtDNA methylation, the current review provides reasons to consider mtDNA epigenetic disruption as a mechanism of action of EDCs and reviews potential limitations related to currently available evidence. First, there is sufficient evidence that EDCs (including bisphenols and phthalates) directly target mitochondrial function, and more direct evidence is needed to connect this to mtDNA methylation. Second, these and other EDCs are potent modulators of nuclear DNA epigenetics, including DNA methylation and histone modifications. Finally, EDCs have been shown to disrupt several modulators of mtDNA methylation, including DNA methyltransferases and the mitochondrial transcription factor A/nuclear respiratory factor 1 pathway. Taken together, these studies highlight the need for future research evaluating mtDNA epigenetic disruption by EDCs and to detail specific mechanisms responsible for such disruptions.
Collapse
Affiliation(s)
- Zheng Zhou
- Department of Animal Sciences, Michigan State University, East Lansing, Michigan 48824
| | - Jaclyn M Goodrich
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, Michigan 48109
| | - Rita S Strakovsky
- Department of Food Science and Human Nutrition.,Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824
| |
Collapse
|
34
|
El-Sikaily A, Helal M. Environmental pollution and diabetes mellitus. World J Meta-Anal 2021; 9:234-256. [DOI: 10.13105/wjma.v9.i3.234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/17/2021] [Accepted: 06/03/2021] [Indexed: 02/06/2023] Open
Abstract
Diabetes mellitus (DM) is a chromic metabolic disease that affects a large segment of the population worldwide. Physical inactivity, poor nutrition, and genetic predisposition are main risk factors for disease development. In the last decade, it was clear to the scientific community that DM development is linked to a novel disease inducer that was later defined as diabetogenic factors of pollution and endocrine disrupting agents. Environmental pollution is exponentially increasing in uncontrolled manner in several countries. Environmental pollutants are of diverse nature and toxicities, including polyaromatic hydrocarbons (PAHs), pesticides, and heavy metals. In the current review, we shed light on the impact of each class of these pollutants and the underlined molecular mechanism of diabetes induction and biological toxicities. Finally, a brief overview about the connection between coronavirus disease 2019 and diabetes pandemics is presented.
Collapse
Affiliation(s)
- Amany El-Sikaily
- National Institute of Oceanography and Fisheries (NIOF), Cairo 21513, Egypt
| | - Mohamed Helal
- National Institute of Oceanography and Fisheries (NIOF), Cairo 21513, Egypt
| |
Collapse
|
35
|
Cano R, Pérez JL, Dávila LA, Ortega Á, Gómez Y, Valero-Cedeño NJ, Parra H, Manzano A, Véliz Castro TI, Albornoz MPD, Cano G, Rojas-Quintero J, Chacín M, Bermúdez V. Role of Endocrine-Disrupting Chemicals in the Pathogenesis of Non-Alcoholic Fatty Liver Disease: A Comprehensive Review. Int J Mol Sci 2021; 22:4807. [PMID: 34062716 PMCID: PMC8125512 DOI: 10.3390/ijms22094807] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/21/2021] [Accepted: 03/22/2021] [Indexed: 12/15/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is considered the most common liver disorder, affecting around 25% of the population worldwide. It is a complex disease spectrum, closely linked with other conditions such as obesity, insulin resistance, type 2 diabetes mellitus, and metabolic syndrome, which may increase liver-related mortality. In light of this, numerous efforts have been carried out in recent years in order to clarify its pathogenesis and create new prevention strategies. Currently, the essential role of environmental pollutants in NAFLD development is recognized. Particularly, endocrine-disrupting chemicals (EDCs) have a notable influence. EDCs can be classified as natural (phytoestrogens, genistein, and coumestrol) or synthetic, and the latter ones can be further subdivided into industrial (dioxins, polychlorinated biphenyls, and alkylphenols), agricultural (pesticides, insecticides, herbicides, and fungicides), residential (phthalates, polybrominated biphenyls, and bisphenol A), and pharmaceutical (parabens). Several experimental models have proposed a mechanism involving this group of substances with the disruption of hepatic metabolism, which promotes NAFLD. These include an imbalance between lipid influx/efflux in the liver, mitochondrial dysfunction, liver inflammation, and epigenetic reprogramming. It can be concluded that exposure to EDCs might play a crucial role in NAFLD initiation and evolution. However, further investigations supporting these effects in humans are required.
Collapse
Affiliation(s)
- Raquel Cano
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (R.C.); (J.L.P.); (Á.O.); (Y.G.); (H.P.); (A.M.); (M.P.D.A.)
| | - José L. Pérez
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (R.C.); (J.L.P.); (Á.O.); (Y.G.); (H.P.); (A.M.); (M.P.D.A.)
| | - Lissé Angarita Dávila
- Escuela de Nutrición y Dietética, Facultad de Medicina, Universidad Andres Bello, Sede Concepción 4260000, Chile;
| | - Ángel Ortega
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (R.C.); (J.L.P.); (Á.O.); (Y.G.); (H.P.); (A.M.); (M.P.D.A.)
| | - Yosselin Gómez
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (R.C.); (J.L.P.); (Á.O.); (Y.G.); (H.P.); (A.M.); (M.P.D.A.)
| | - Nereida Josefina Valero-Cedeño
- Carrera de Laboratorio Clínico, Facultad de Ciencias de la Salud, Universidad Estatal del Sur de Manabí, Jipijapa E482, Ecuador; (N.J.V.-C.); (T.I.V.C.)
| | - Heliana Parra
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (R.C.); (J.L.P.); (Á.O.); (Y.G.); (H.P.); (A.M.); (M.P.D.A.)
| | - Alexander Manzano
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (R.C.); (J.L.P.); (Á.O.); (Y.G.); (H.P.); (A.M.); (M.P.D.A.)
| | - Teresa Isabel Véliz Castro
- Carrera de Laboratorio Clínico, Facultad de Ciencias de la Salud, Universidad Estatal del Sur de Manabí, Jipijapa E482, Ecuador; (N.J.V.-C.); (T.I.V.C.)
| | - María P. Díaz Albornoz
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (R.C.); (J.L.P.); (Á.O.); (Y.G.); (H.P.); (A.M.); (M.P.D.A.)
| | - Gabriel Cano
- Insitute für Pharmazie, Freie Universitänt Berlin, Königin-Louise-Strabe 2-4, 14195 Berlin, Germany;
| | - Joselyn Rojas-Quintero
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
| | - Maricarmen Chacín
- Facultad de Ciencias de la Salud. Barranquilla, Universidad Simón Bolívar, Barranquilla 55-132, Colombia;
| | - Valmore Bermúdez
- Facultad de Ciencias de la Salud. Barranquilla, Universidad Simón Bolívar, Barranquilla 55-132, Colombia;
| |
Collapse
|
36
|
Endocrine-Disrupting Chemicals and Infectious Diseases: From Endocrine Disruption to Immunosuppression. Int J Mol Sci 2021; 22:ijms22083939. [PMID: 33920428 PMCID: PMC8069594 DOI: 10.3390/ijms22083939] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/07/2021] [Accepted: 04/08/2021] [Indexed: 01/08/2023] Open
Abstract
Endocrine-disrupting chemicals (EDCs) are hormonally active compounds in the environment that interfere with the body's endocrine system and consequently produce adverse health effects. Despite persistent public health concerns, EDCs remain important components of common consumer products, thus representing ubiquitous contaminants to humans. While scientific evidence confirmed their contribution to the severity of Influenza A virus (H1N1) in the animal model, their roles in susceptibility and clinical outcome of the coronavirus disease (COVID-19) cannot be underestimated. Since its emergence in late 2019, clinical reports on COVID-19 have confirmed that severe disease and death occur in persons aged ≥65 years and those with underlying comorbidities. Major comorbidities of COVID-19 include diabetes, obesity, cardiovascular disease, hypertension, cancer, and kidney and liver diseases. Meanwhile, long-term exposure to EDCs contributes significantly to the onset and progression of these comorbid diseases. Besides, EDCs play vital roles in the disruption of the body's immune system. Here, we review the recent literature on the roles of EDCs in comorbidities contributing to COVID-19 mortality, impacts of EDCs on the immune system, and recent articles linking EDCs to COVID-19 risks. We also recommend methodologies that could be adopted to comprehensively study the role of EDCs in COVID-19 risk.
Collapse
|
37
|
Xu P, Liu A, Li F, Tinkov AA, Liu L, Zhou JC. Associations between metabolic syndrome and four heavy metals: A systematic review and meta-analysis. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 273:116480. [PMID: 33486246 DOI: 10.1016/j.envpol.2021.116480] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 12/19/2020] [Accepted: 01/07/2021] [Indexed: 06/12/2023]
Abstract
Four most concerned heavy metal pollutants, arsenic, cadmium, lead, and mercury may share common mechanisms to induce metabolic syndrome (MetS). However, recent studies exploring the relationships between MetS and metal exposure presented inconsistent findings. We aimed to clarify the relationship between heavy metal exposure biomarkers and MetS using a meta-analysis and systematic review approach. Literature search was conducted in international and the Chinese national databases up to June 2020. Of selected studies, we extracted the relevant data and evaluated the quality of each study's methodology. We then calculated the pooled effect sizes (ESs), standardized mean differences (SMDs), and their 95% confidence intervals (CIs) using a random-effect meta-analysis approach followed by stratification analyses for control of potential confounders. Involving 55,536 participants, the included 22 articles covered 52 observational studies reporting ESs and/or metal concentrations on specific metal and gender. Our results show that participants with MetS had significantly higher levels of heavy metal exposure [pooled ES = 1.16, 95% CI: 1.09, 1.23; n = 42, heterogeneity I2 = 75.6%; and SMD = 0.22, 95% CI: 0.15, 0.29; n = 32, I2 = 94.2%] than those without MetS. Pooled ESs in the subgroups stratified by arsenic, cadmium, lead, and mercury were 1.04 (95% CI: 0.97, 1.10; n = 8, I2 = 61.0%), 1.10 (0.95, 1.27; 11, 45.0%), 1.21 (1.00, 1.48; 12, 82.9%), and 1.26 (1.06, 1.48; 11, 67.7%), respectively. Pooled ESs in the subgroups stratified by blood, urine, and the other specimen were 1.22 (95% CI: 1.08, 1.38; n = 26, I2 = 75.8%), 1.06 (1.00, 1.13; 14, 58.1%), and 2.41 (1.30, 4.43; 2, 0.0%), respectively. In conclusion, heavy metal exposure was positively associated with MetS. Further studies are warranted to examine the effects of individual metals and their interaction on the relationship between MetS and heavy metals.
Collapse
Affiliation(s)
- Ping Xu
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, 518100, China
| | - Aiping Liu
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, 518100, China
| | - Fengna Li
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, 518100, China
| | - Alexey A Tinkov
- Yaroslavl State University, 150003, Yaroslavl, Russia; IM Sechenov First Moscow State Medical University (Sechenov University), 119146, Moscow, Russia
| | - Longjian Liu
- Department of Epidemiology and Biostatistics, Dornsife School of Public Health, Drexel University, Philadelphia, PA, 19104, USA
| | - Ji-Chang Zhou
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, 518100, China; Guangdong Province Engineering Laboratory for Nutrition Translation, Guangzhou, 510080, China.
| |
Collapse
|
38
|
Rives C, Fougerat A, Ellero-Simatos S, Loiseau N, Guillou H, Gamet-Payrastre L, Wahli W. Oxidative Stress in NAFLD: Role of Nutrients and Food Contaminants. Biomolecules 2020; 10:E1702. [PMID: 33371482 PMCID: PMC7767499 DOI: 10.3390/biom10121702] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 12/14/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is often the hepatic expression of metabolic syndrome and its comorbidities that comprise, among others, obesity and insulin-resistance. NAFLD involves a large spectrum of clinical conditions. These range from steatosis, a benign liver disorder characterized by the accumulation of fat in hepatocytes, to non-alcoholic steatohepatitis (NASH), which is characterized by inflammation, hepatocyte damage, and liver fibrosis. NASH can further progress to cirrhosis and hepatocellular carcinoma. The etiology of NAFLD involves both genetic and environmental factors, including an unhealthy lifestyle. Of note, unhealthy eating is clearly associated with NAFLD development and progression to NASH. Both macronutrients (sugars, lipids, proteins) and micronutrients (vitamins, phytoingredients, antioxidants) affect NAFLD pathogenesis. Furthermore, some evidence indicates disruption of metabolic homeostasis by food contaminants, some of which are risk factor candidates in NAFLD. At the molecular level, several models have been proposed for the pathogenesis of NAFLD. Most importantly, oxidative stress and mitochondrial damage have been reported to be causative in NAFLD initiation and progression. The aim of this review is to provide an overview of the contribution of nutrients and food contaminants, especially pesticides, to oxidative stress and how they may influence NAFLD pathogenesis.
Collapse
Affiliation(s)
- Clémence Rives
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, EVT, INP-Purpan, UPS, 31300 Toulouse, France; (C.R.); (A.F.); (S.E.-S.); (N.L.); (H.G.)
| | - Anne Fougerat
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, EVT, INP-Purpan, UPS, 31300 Toulouse, France; (C.R.); (A.F.); (S.E.-S.); (N.L.); (H.G.)
| | - Sandrine Ellero-Simatos
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, EVT, INP-Purpan, UPS, 31300 Toulouse, France; (C.R.); (A.F.); (S.E.-S.); (N.L.); (H.G.)
| | - Nicolas Loiseau
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, EVT, INP-Purpan, UPS, 31300 Toulouse, France; (C.R.); (A.F.); (S.E.-S.); (N.L.); (H.G.)
| | - Hervé Guillou
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, EVT, INP-Purpan, UPS, 31300 Toulouse, France; (C.R.); (A.F.); (S.E.-S.); (N.L.); (H.G.)
| | - Laurence Gamet-Payrastre
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, EVT, INP-Purpan, UPS, 31300 Toulouse, France; (C.R.); (A.F.); (S.E.-S.); (N.L.); (H.G.)
| | - Walter Wahli
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, EVT, INP-Purpan, UPS, 31300 Toulouse, France; (C.R.); (A.F.); (S.E.-S.); (N.L.); (H.G.)
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Clinical Sciences Building, 11 Mandalay Road, Singapore 308232, Singapore
- Center for Integrative Genomics, Université de Lausanne, Le Génopode, CH-1015 Lausanne, Switzerland
| |
Collapse
|
39
|
Le Magueresse-Battistoni B. Adipose Tissue and Endocrine-Disrupting Chemicals: Does Sex Matter? INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17249403. [PMID: 33333918 PMCID: PMC7765367 DOI: 10.3390/ijerph17249403] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/10/2020] [Accepted: 12/14/2020] [Indexed: 12/21/2022]
Abstract
Obesity and metabolic-related diseases, among which diabetes, are prominent public health challenges of the 21st century. It is now well acknowledged that pollutants are a part of the equation, especially endocrine-disrupting chemicals (EDCs) that interfere with the hormonal aspect. The aim of the review is to focus on adipose tissue, a central regulator of energy balance and metabolic homeostasis, and to highlight the significant differences in the endocrine and metabolic aspects of adipose tissue between males and females which likely underlie the differences of the response to exposure to EDCs between the sexes. Moreover, the study also presents an overview of several mechanisms of action by which pollutants could cause adipose tissue dysfunction. Indeed, a better understanding of the mechanism by which environmental chemicals target adipose tissue and cause metabolic disturbances, and how these mechanisms interact and sex specificities are essential for developing mitigating and sex-specific strategies against metabolic diseases of chemical origin. In particular, considering that a scenario without pollutant exposure is not a realistic option in our current societies, attenuating the deleterious effects of exposure to pollutants by acting on the gut-adipose tissue axis may constitute a new direction of research.
Collapse
Affiliation(s)
- Brigitte Le Magueresse-Battistoni
- Univ-Lyon, CarMeN Laboratory, INSERM U1060, INRAé U1397, Université Claude Bernard Lyon1, F-69310 Pierre-Bénite, France; ; Tel.: +33-(0)-426235919; Fax: +33-(0)-426235916
- CarMeN Laboratory, INSERM U1060, Hopital Lyon-Sud, Bâtiment CENS ELI-2D, 165 Chemin du Grand Revoyet, 69310 Pierre-Bénite, France
| |
Collapse
|
40
|
The antiandrogenic vinclozolin induces differentiation delay of germ cells and changes in energy metabolism in 3D cultures of fetal ovaries. Sci Rep 2020; 10:18036. [PMID: 33093579 PMCID: PMC7582921 DOI: 10.1038/s41598-020-75116-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 10/12/2020] [Indexed: 12/14/2022] Open
Abstract
Vinclozolin is a pesticide with antiandrogenic activity as an endocrine disruptor compound. Its effects upon the progression of primordial follicles were assessed in cultures of mouse fetal ovaries from the onset of meiotic differentiation of germ cells (13.5 days post coitum) and from both in vivo exposed mice and in vitro exposed ovaries. Exposure of ovaries to vinclozolin—at in vitro dosages ranging from 10 to 200 μM and in 3D ex vivo culture following in vivo exposure to 50 mg/kg bw/day—showed delays in meiocyte differentiation and in follicle growth, even at the lowest in vitro dose exposure. Immunofluorescent analysis showed the presence of the proteins MSY2 and NOBOX in the primary follicles but no difference in the level of protein signals or in the number of follicles in relation to treatment. However, assessing the cytological differentiation of germ cells by detecting the synaptonemal complex protein SYCP3, the exposure to vinclozolin delayed meiotic differentiation from both in vitro- and in vivo-exposed ovaries. These effects were concomitant with changes in the energy metabolism, detected as a relative increase of glycolytic metabolism in live-cell metabolic assays in exposed ovaries.
Collapse
|
41
|
The EDCMET Project: Metabolic Effects of Endocrine Disruptors. Int J Mol Sci 2020; 21:ijms21083021. [PMID: 32344727 PMCID: PMC7215524 DOI: 10.3390/ijms21083021] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/21/2020] [Accepted: 04/22/2020] [Indexed: 02/08/2023] Open
Abstract
Endocrine disruptors (EDs) are defined as chemicals that mimic, block, or interfere with hormones in the body's endocrine systems and have been associated with a diverse array of health issues. The concept of endocrine disruption has recently been extended to metabolic alterations that may result in diseases, such as obesity, diabetes, and fatty liver disease, and constitute an increasing health concern worldwide. However, while epidemiological and experimental data on the close association of EDs and adverse metabolic effects are mounting, predictive methods and models to evaluate the detailed mechanisms and pathways behind these observed effects are lacking, thus restricting the regulatory risk assessment of EDs. The EDCMET (Metabolic effects of Endocrine Disrupting Chemicals: novel testing METhods and adverse outcome pathways) project brings together systems toxicologists; experimental biologists with a thorough understanding of the molecular mechanisms of metabolic disease and comprehensive in vitro and in vivo methodological skills; and, ultimately, epidemiologists linking environmental exposure to adverse metabolic outcomes. During its 5-year journey, EDCMET aims to identify novel ED mechanisms of action, to generate (pre)validated test methods to assess the metabolic effects of Eds, and to predict emergent adverse biological phenotypes by following the adverse outcome pathway (AOP) paradigm.
Collapse
|
42
|
Klinge CM. Estrogenic control of mitochondrial function. Redox Biol 2020; 31:101435. [PMID: 32001259 PMCID: PMC7212490 DOI: 10.1016/j.redox.2020.101435] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 01/14/2020] [Accepted: 01/16/2020] [Indexed: 12/15/2022] Open
Abstract
Sex-based differences in human disease are caused in part by the levels of endogenous sex steroid hormones which regulate mitochondrial metabolism. This review updates a previous review on how estrogens regulate metabolism and mitochondrial function that was published in 2017. Estrogens are produced by ovaries and adrenals, and in lesser amounts by adipose, breast stromal, and brain tissues. At the cellular level, the mechanisms by which estrogens regulate diverse cellular functions including reproduction and behavior is by binding to estrogen receptors α, β (ERα and ERβ) and G-protein coupled ER (GPER1). ERα and ERβ are transcription factors that bind genomic and mitochondrial DNA to regulate gene transcription. A small proportion of ERα and ERβ interact with plasma membrane-associated signaling proteins to activate intracellular signaling cascades that ultimately alter transcriptional responses, including mitochondrial morphology and function. Although the mechanisms and targets by which estrogens act directly and indirectly to regulate mitochondrial function are not fully elucidated, it is clear that estradiol regulates mitochondrial metabolism and morphology via nuclear and mitochondrial-mediated events, including stimulation of nuclear respiratory factor-1 (NRF-1) transcription that will be reviewed here. NRF-1 is a transcription factor that interacts with coactivators including peroxisome proliferator-activated receptor gamma, coactivator 1 alpha (PGC-1α) to regulate nuclear-encoded mitochondrial genes. One NRF-1 target is TFAM that binds mtDNA to regulate its transcription. Nuclear-encoded miRNA and lncRNA regulate mtDNA-encoded and nuclear-encoded transcripts that regulate mitochondrial function, thus acting as anterograde signals. Other estrogen-regulated mitochondrial activities including bioenergetics, oxygen consumption rate (OCR), and extracellular acidification (ECAR), are reviewed.
Collapse
Affiliation(s)
- Carolyn M Klinge
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, 40292, KY, USA.
| |
Collapse
|
43
|
Azevedo LF, Hornos Carneiro MF, Dechandt CRP, Cassoli JS, Alberici LC, Barbosa F. Global liver proteomic analysis of Wistar rats chronically exposed to low-levels of bisphenol A and S. ENVIRONMENTAL RESEARCH 2020; 182:109080. [PMID: 31901629 DOI: 10.1016/j.envres.2019.109080] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 12/21/2019] [Accepted: 12/21/2019] [Indexed: 05/26/2023]
Abstract
Exposure to bisphenol A (BPA) and bisphenol S (BPS) has been associated with the development of metabolic disorders, such as obesity, dyslipidemias, and nonalcoholic fatty liver disease. Nonetheless, the associated mechanisms are still not fully understood. BPS is being used with no restrictions to replace BPA, which increases the concern regarding its safety and claims for further investigation on its potential mechanisms of toxicity. The present study aims to access liver molecular disturbances which could be associated with systemic metabolic disorders following exposure to BPA or BPS. Therefore, body weight gain and serum biochemical parameters were measured in male Wistar rats chronically exposed to 50 or 500 µg/kg/day of BPA or BPS, while an extensive evaluation of liver protein expression changes was conducted after exposure to 50 µg/kg/day of both compounds. Exposure to the lowest dose of BPA led to the development of hyperglycemia and hypercholesterolemia, while the BPS lowest dose led to the development of hypertriglyceridemia. Besides, exposure to 500 µg/kg/day of BPS significantly increased body weight gain and LDL-cholesterol levels. Hepatic proteins differentially expressed in BPA and BPS-exposed groups compared to the control group were mostly related to lipid metabolism and synthesis, with upregulation of glucokinase activity-related sequence 1 (1.8-fold in BPA and 2.4-fold in BPS), which is involved in glycerol triglycerides synthesis, and hydroxymethylglutaryl-CoA synthase cytoplasmic (2-fold in BPS), an enzyme involved in mevalonate biosynthesis. Essential mitochondrial proteins of the electron transport chain were upregulated after exposure to both contaminants. Also, BPA and BPS dysregulated expression of liver antioxidant enzymes, which are involved in cellular reactive oxygen species detoxification. Altogether, the results of the present study contribute to expand the scientific understanding of how BPA and BPS lead to the development of metabolic disorders and reinforce the risks associated with exposure to these contaminants.
Collapse
Affiliation(s)
- Lara Ferreira Azevedo
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo-USP, Ribeirão Preto, Brazil
| | - Maria Fernanda Hornos Carneiro
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo-USP, Ribeirão Preto, Brazil; Departamento de Farmacia, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
| | - Carlos Roberto Porto Dechandt
- Department of Physics and Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo-USP, Ribeirão Preto, Brazil
| | | | - Luciane Carla Alberici
- Department of Physics and Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo-USP, Ribeirão Preto, Brazil
| | - Fernando Barbosa
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo-USP, Ribeirão Preto, Brazil.
| |
Collapse
|
44
|
Yilmaz B, Terekeci H, Sandal S, Kelestimur F. Endocrine disrupting chemicals: exposure, effects on human health, mechanism of action, models for testing and strategies for prevention. Rev Endocr Metab Disord 2020; 21:127-147. [PMID: 31792807 DOI: 10.1007/s11154-019-09521-z] [Citation(s) in RCA: 307] [Impact Index Per Article: 61.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Endocrine Disrupting Chemicals (EDCs) are a global problem for environmental and human health. They are defined as "an exogenous chemical, or mixture of chemicals, that can interfere with any aspect of hormone action". It is estimated that there are about 1000 chemicals with endocrine-acting properties. EDCs comprise pesticides, fungicides, industrial chemicals, plasticizers, nonylphenols, metals, pharmaceutical agents and phytoestrogens. Human exposure to EDCs mainly occurs by ingestion and to some extent by inhalation and dermal uptake. Most EDCs are lipophilic and bioaccumulate in the adipose tissue, thus they have a very long half-life in the body. It is difficult to assess the full impact of human exposure to EDCs because adverse effects develop latently and manifest at later ages, and in some people do not present. Timing of exposure is of importance. Developing fetus and neonates are the most vulnerable to endocrine disruption. EDCs may interfere with synthesis, action and metabolism of sex steroid hormones that in turn cause developmental and fertility problems, infertility and hormone-sensitive cancers in women and men. Some EDCs exert obesogenic effects that result in disturbance in energy homeostasis. Interference with hypothalamo-pituitary-thyroid and adrenal axes has also been reported. In this review, potential EDCs, their effects and mechanisms of action, epidemiological studies to analyze their effects on human health, bio-detection and chemical identification methods, difficulties in extrapolating experimental findings and studying endocrine disruptors in humans and recommendations for endocrinologists, individuals and policy makers will be discussed in view of the relevant literature.
Collapse
Affiliation(s)
- Bayram Yilmaz
- Department of Physiology, Faculty of Medicine, Yeditepe University, Istanbul, Turkey
| | - Hakan Terekeci
- Department of Internal Medicine, Faculty of Medicine, Yeditepe University, Istanbul, Turkey
| | - Suleyman Sandal
- Department of Physiology, Faculty of Medicine, Inonu University, Malatya, Turkey
| | - Fahrettin Kelestimur
- Department of Endocrinology, Faculty of Medicine, Yeditepe University, Istanbul, Turkey.
| |
Collapse
|
45
|
Relationship Between Oxidative Stress, ER Stress, and Inflammation in Type 2 Diabetes: The Battle Continues. J Clin Med 2019; 8:jcm8091385. [PMID: 31487953 PMCID: PMC6780404 DOI: 10.3390/jcm8091385] [Citation(s) in RCA: 326] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/29/2019] [Accepted: 09/02/2019] [Indexed: 12/15/2022] Open
Abstract
Type 2 diabetes (T2D) is a metabolic disorder characterized by hyperglycemia and insulin resistance in which oxidative stress is thought to be a primary cause. Considering that mitochondria are the main source of ROS, we have set out to provide a general overview on how oxidative stress is generated and related to T2D. Enhanced generation of reactive oxygen species (ROS) and oxidative stress occurs in mitochondria as a consequence of an overload of glucose and oxidative phosphorylation. Endoplasmic reticulum (ER) stress plays an important role in oxidative stress, as it is also a source of ROS. The tight interconnection between both organelles through mitochondrial-associated membranes (MAMs) means that the ROS generated in mitochondria promote ER stress. Therefore, a state of stress and mitochondrial dysfunction are consequences of this vicious cycle. The implication of mitochondria in insulin release and the exposure of pancreatic β-cells to hyperglycemia make them especially susceptible to oxidative stress and mitochondrial dysfunction. In fact, crosstalk between both mechanisms is related with alterations in glucose homeostasis and can lead to the diabetes-associated insulin-resistance status. In the present review, we discuss the current knowledge of the relationship between oxidative stress, mitochondria, ER stress, inflammation, and lipotoxicity in T2D.
Collapse
|
46
|
Azevedo LF, Porto Dechandt CR, Cristina de Souza Rocha C, Hornos Carneiro MF, Alberici LC, Barbosa F. Long-term exposure to bisphenol A or S promotes glucose intolerance and changes hepatic mitochondrial metabolism in male Wistar rats. Food Chem Toxicol 2019; 132:110694. [PMID: 31344369 DOI: 10.1016/j.fct.2019.110694] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 07/20/2019] [Accepted: 07/22/2019] [Indexed: 02/07/2023]
Abstract
The present study evaluates the effects of low-level long-term exposure to bisphenol A (BPA) and bisphenol S (BPS) on serum biochemical markers, glucose homeostasis, mitochondrial energy metabolism, biogenesis and dynamics, and redox status in livers of Wistar rats. While only the exposure to BPS induces a significant body mass gain after 21 weeks, both compounds alter serum lipid levels and lead to the development of glucose intolerance. Regarding mitochondrial metabolism, both bisphenols augment the electron entry by complex II relative to complex I in the mitochondrial respiratory chain (MRC), and reduce mitochondrial content; BPA reduces OXPHOS capacity and uncouples respiration (relative to maximal capacity of MRC) but promotes a significant increase in fatty acid oxidation. Either exposure to BPA or BPS leads to an increase in mitochondrial-derived reactive oxygen species, mainly at complex I. Additionally, BPA and BPS significantly upregulate the expression levels of dynamin-related protein 1 related to mitochondrial fission, while BPA downregulates the expression of proliferator-activated receptor gamma coactivator 1 alpha, a master regulator of mitochondrial biogenesis. In summary, our data shows that exposure to both compounds alters metabolic homeostasis and mitochondrial energy metabolism, providing new mechanisms by which BPA and BPS impair the mitochondrial metabolism.
Collapse
Affiliation(s)
- Lara Ferreira Azevedo
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo-USP, Ribeirão Preto, Brazil
| | - Carlos Roberto Porto Dechandt
- Department of Physics and Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo-USP, Ribeirão Preto, Brazil
| | - Cecília Cristina de Souza Rocha
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo-USP, Ribeirão Preto, Brazil
| | - Maria Fernanda Hornos Carneiro
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo-USP, Ribeirão Preto, Brazil
| | - Luciane Carla Alberici
- Department of Physics and Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo-USP, Ribeirão Preto, Brazil.
| | - Fernando Barbosa
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo-USP, Ribeirão Preto, Brazil.
| |
Collapse
|