1
|
Liang Q, Liu X, Xu X, Chen Z, Luo T, Su Y, Xie C. Molecular mechanisms and therapeutic perspectives of luteolin on diabetes and its complications. Eur J Pharmacol 2025; 1000:177691. [PMID: 40311831 DOI: 10.1016/j.ejphar.2025.177691] [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: 03/10/2025] [Revised: 04/13/2025] [Accepted: 04/29/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND Extensive preclinical studies have established luteolin, a flavonoid with potent antidiabetic activity, as a therapeutic candidate for preventing and managing various diabetic complications including cardiomyopathy, nephropathy, and osteopathy. This systematic review evaluates current evidence regarding luteolin's antidiabetic potential. AIM OF THE STUDY This study evaluates luteolin's efficacy in diabetes management through evidence synthesis, while critically assessing current research challenges and translational opportunities. METHODS A comprehensive literature search was conducted across Pubmed, Embase, Web of Science, and Google Scholar databases, encompassing articles published between 2000 and 2024. RESULTS Luteolin is a naturally occurring flavonoid that has strong antidiabetic properties. It regulates intestinal microenvironmental homeostasis, lipogenesis and catabolism, and the absorption of carbohydrates. It also modulates nine diabetic complications by reducing inflammation, oxidative stress, apoptosis, and autophagy. Luteolin's potential nutritional and physiological benefits notwithstanding, attention must be directed immediately to its bioavailability, innovative formulations, safety assessment, synergistic effects, and optimal dosage and time for supplementation. In particular, clinical studies are needed to validate efficacy and safety and provide a reliable scientific basis. CONCLUSION Luteolin may act as a pleiotropic molecule targeting multiple signaling cascades to exert antidiabetic bioactivity.
Collapse
Affiliation(s)
- Qingzhi Liang
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610072, China
| | - Xiaoqin Liu
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610072, China
| | - Xin Xu
- Department of Emergency, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610072, China
| | - Zhengtao Chen
- Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, 330006, China
| | - Ting Luo
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610072, China
| | - Yi Su
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610072, China
| | - Chunguang Xie
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, 610072, China; Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610072, China.
| |
Collapse
|
2
|
Li T, Liu Y, Cao J, Lu X, Lu Y, Wang Y, Zhang C, Wu M, Deng S, Li L, Shi M. Triphenyl phosphate induces lipid metabolism disorder and promotes obesity through PI3K/AKT signaling pathway. ENVIRONMENT INTERNATIONAL 2025; 198:109428. [PMID: 40199182 DOI: 10.1016/j.envint.2025.109428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/26/2025] [Accepted: 03/30/2025] [Indexed: 04/10/2025]
Abstract
Triphenyl phosphate (TPHP) is a widely used organic phosphate flame retardant that has been reported as a potential environmental obesogen. However, the potential impact and mechanism of action of TPHP on adipose tissue are still unclear. This study investigates the potential impact of TPHP on lipid metabolism disorders through in vivo and in vitro experiments. Male and female BALB/c mice were exposed to TPHP (0, 1, 10, and 150 mg/kg/day) for 60 days, and 3T3-L1 preadipocytes were treated with concentrations of TPHP (0, 0.1, 1, 10 μM) during differentiation. The results showed that exposure to TPHP could cause gender specific dyslipidemia, with male mice exhibiting dose-dependent increases in inguinal adipose tissue coefficient, adipocyte hypertrophy, and upregulation of adipose differentiation and adipogenesis-related genes. In contrast, female mice did not show significant changes in tissue morphology. This suggested that TPHP might promote the potential occurrence of adiposity by disrupting the lipid metabolism homeostasis of male adipose tissue. During the differentiation and maturation process of 3T3-L1 preadipocytes, exposure to TPHP led to increased lipid accumulation and disrupted lipid homeostasis by simultaneous activation adipogenesis and lipolysis. Multiple omics data showed that the activation of the peroxisome proliferator-activated receptor γ (PPARγ) signaling pathway and fatty acid metabolism was the core mechanism of TPHP induced metabolic dysfunction. Further research showed that TPHP activated the PI3K/AKT pathway, and PI3K inhibitor (LY294002) could rescue TPHP induced lipid droplet formation and normalize the expression of adipogenic markers. These findings confirm that TPHP is a potential environmental obesogen that can disrupt the metabolic homeostasis of white adipose tissue through the PPARγ and PI3K/AKT signaling pathways, with higher susceptibility in males. This study provides compelling evidence for the obesogenic effects of TPHP and information for risk assessment of organophosphorus flame retardants.
Collapse
Affiliation(s)
- Tianlan Li
- Dongguan Key Laboratory of Environmental Medicine, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808 Guangdong Province, China
| | - Yiwa Liu
- Dongguan Key Laboratory of Environmental Medicine, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808 Guangdong Province, China
| | - Jingyi Cao
- Dongguan Key Laboratory of Environmental Medicine, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808 Guangdong Province, China
| | - Xianzhu Lu
- Dongguan Key Laboratory of Environmental Medicine, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808 Guangdong Province, China
| | - Yinghan Lu
- Dongguan Key Laboratory of Environmental Medicine, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808 Guangdong Province, China
| | - Yuhan Wang
- Dongguan Key Laboratory of Environmental Medicine, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808 Guangdong Province, China
| | - Chunmei Zhang
- Dongguan Key Laboratory of Environmental Medicine, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808 Guangdong Province, China
| | - Meifen Wu
- Dongguan Key Laboratory of Environmental Medicine, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808 Guangdong Province, China
| | - Song Deng
- Dongguan Key Laboratory of Environmental Medicine, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808 Guangdong Province, China
| | - Li Li
- Dongguan Key Laboratory of Environmental Medicine, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808 Guangdong Province, China.
| | - Ming Shi
- Dongguan Key Laboratory of Environmental Medicine, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808 Guangdong Province, China.
| |
Collapse
|
3
|
Lara-Hernández F, Melero R, Quiroz-Rodríguez ME, Moya-Valera C, de Jesús Gallardo-Espinoza M, Álvarez L, Valarezo-Torres IL, Briongos-Figuero L, Abadía-Otero J, Mena-Martin FJ, Saez G, Redon J, Martín-Escudero JC, García-García AB, Ayala G, Chaves FJ. Genetic interaction between oxidative stress and body mass index in a Spanish population. Redox Biol 2025; 80:103531. [PMID: 39923398 PMCID: PMC11849672 DOI: 10.1016/j.redox.2025.103531] [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: 12/05/2024] [Revised: 01/21/2025] [Accepted: 02/03/2025] [Indexed: 02/11/2025] Open
Abstract
Oxidative stress may act as a contributing factor in the development of an elevated body mass index (BMI). Oxidative stress has the potential to modulate genetic activity at various levels, including gene transcription and protein function regulation. Nevertheless, the interplay between genetic variants and oxidative stress in relation to BMI remains to be elucidated. Based on this premise, we studied the potential association between 723 single-nucleotide polymorphisms (SNPs) located within a set of 212 genes and both BMI and oxidative stress parameters in 1502 adults from the general Spanish population (Hortega Study). Oxidative stress parameters measured included malondialdehyde (MDA) levels, 8-oxo-2'-deoxyguanosine (8-oxo-dG) levels and oxidised/reduced glutathione ratio (GSSG/GSH). We also examined the potential impact of the interaction between these SNPs and oxidative stress levels on BMI. The genes selected regulate several key biological processes, including obesity, blood pressure, inflammation, lipid metabolism and redox homeostasis. Our findings indicate a robust association between specific genes and both BMI and oxidative stress parameters. Significant BMI-related interactions between genes and oxidative stress parameters were identified, which have a multifactorial impact on oxidative stress modulation and on BMI. SNPs identified in genes such as NPPA, CPT1A, DDIT3, NOX and IL6ST were significantly associated with all oxidative stress parameters analysed, indicating a substantial influence on BMI modulation. The results provide compelling evidence of a significant relationship between oxidative stress levels and genetic background. Our data provide new insights into BMI modulation by oxidative stress levels, highlighting a role for TNF as a key player in the interrelation of oxidative stress and BMI.
Collapse
Affiliation(s)
| | - Rebeca Melero
- Genomics and Diabetes Unit. INCLIVA Biomedical Research Institute, 46010, Valencia, Spain
| | | | - Celeste Moya-Valera
- Genomics and Diabetes Unit. INCLIVA Biomedical Research Institute, 46010, Valencia, Spain
| | | | - Luis Álvarez
- Genomics and Diabetes Unit. INCLIVA Biomedical Research Institute, 46010, Valencia, Spain
| | | | | | - Jessica Abadía-Otero
- Internal Medicine Service. Rio Hortega University Hospital, 47012, Valladolid, Spain
| | | | - Guillermo Saez
- Department of Biochemistry and Molecular Biology, Faculty of Medicine and Odontology. University of Valencia, 46010, Valencia, Spain; Service of Clinical Analysis. University Hospital Dr. Peset-FISABIO, Spain
| | - Josep Redon
- Cardiometabolic Renal Risk Research Group, INCLIVA Biomedical Research Institute, University of Valencia, 46010, Valencia, Spain; CIBEROBN, ISCIII, 28029, Madrid, Spain
| | - Juan-Carlos Martín-Escudero
- Internal Medicine Service. Rio Hortega University Hospital, 47012, Valladolid, Spain; Department of Medicine, Faculty of Medicine, University of Valladolid, 47002, Valladolid, Spain
| | - Ana-Bárbara García-García
- Genomics and Diabetes Unit. INCLIVA Biomedical Research Institute, 46010, Valencia, Spain; CIBERDEM, ISCIII, 28029, Madrid, Spain.
| | - Guillermo Ayala
- Department of Statistics and Operation Research, University of Valencia, Burjassot, 46100, Valencia, Spain
| | - Felipe Javier Chaves
- Genomics and Diabetes Unit. INCLIVA Biomedical Research Institute, 46010, Valencia, Spain; CIBERDEM, ISCIII, 28029, Madrid, Spain
| |
Collapse
|
4
|
Zhang H, Zhou W, Wang X, Men H, Wang J, Xu J, Zhou S, Liu Q, Cai L. Exacerbation by knocking-out metallothionein gene of obesity-induced cardiac remodeling is associated with the activation of CARD9 signaling. Int J Biol Sci 2025; 21:1032-1046. [PMID: 39897024 PMCID: PMC11781176 DOI: 10.7150/ijbs.105513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 12/28/2024] [Indexed: 02/04/2025] Open
Abstract
Obesity increases the risk of metabolic syndrome including insulin resistance, dyslipidemia, and cardiovascular disease. We demonstrated insulin resistance, cardiac hypertrophy, and cardiac inflammation in an obese mouse model induced by a high-fat diet (HFD). Caspase recruitment domain-containing protein 9 (CARD9) and B-cell lymphoma/leukemia 10 (BCL10) were upregulated, and p38 MAPK was activated in these mice. Zinc supplementation prevented these changes with upregulation of metallothionein (MT). Deletion of MT exacerbated palmitate-triggered expression of BCL10 and p38 MAPK activation and eliminated the protective benefits of zinc in palmitate-treated cardiomyocytes. Here we further investigated the mechanisms by which endogenous MT expression affects HFD-induced cardiac remodeling and the CARD9/BCL10/p38 MAPK pathway. Male MT knockout and 129S wild-type mice were assigned to receive either a normal diet or a HFD from 8-week-age for 18 weeks. MT knockout (KO) aggravated HFD-induced obesity and systemic metabolic disorder, reflected by increased body weight, perirenal white adipose tissue, and plasma cholesterol, and cardiac hypertrophy and fibrosis. Obese MT-KO mice had abundant cardiac macrophages, upregulated cardiac proinflammatory cytokines, chemokines, adhesion molecules, CARD9, and BCL10 and activated NF-κB. MT-KO exacerbated HFD-induced trace metal dyshomeostasis and oxidative stress. MT-KO combined with HFD-induced obesity synergistically promotes cardiac remodeling, possibly via trace metal dyshomeostasis-induced oxidative stress to trigger CARD9/BCL10-mediated NF-κB activation.
Collapse
Affiliation(s)
- Haina Zhang
- Pediatric Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY, 40202, USA
- Department of Cardiology, The Second Hospital of Jilin University, Jilin University, Changchun, 130041, China
| | - Wenqian Zhou
- Pediatric Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY, 40202, USA
- Department of Cardiovascular Diseases, The First Hospital of Jilin University, Jilin University, Changchun, 130021, China
| | - Xiang Wang
- Pediatric Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY, 40202, USA
- Department of Cardiovascular Diseases, The First Hospital of Jilin University, Jilin University, Changchun, 130021, China
| | - Hongbo Men
- Pediatric Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY, 40202, USA
- Department of Cardiovascular Diseases, The First Hospital of Jilin University, Jilin University, Changchun, 130021, China
| | - Jiqun Wang
- Pediatric Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY, 40202, USA
- Department of Cardiovascular Diseases, The First Hospital of Jilin University, Jilin University, Changchun, 130021, China
| | - Jianxiang Xu
- Pediatric Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY, 40202, USA
| | - Shanshan Zhou
- Department of Cardiovascular Diseases, The First Hospital of Jilin University, Jilin University, Changchun, 130021, China
| | - Quan Liu
- Department of Cardiovascular Diseases, The First Hospital of Jilin University, Jilin University, Changchun, 130021, China
| | - Lu Cai
- Pediatric Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY, 40202, USA
- Departments of Radiation Oncology, Pharmacology and Toxicology, University of Louisville, Louisville, KY, 40202, USA
| |
Collapse
|
5
|
Zhu S, Ruan F, Ye L, Jiang S, Yang C, Zuo Z, He C. Black phosphorus quantum dots induce lipid accumulation through PPARγ activation and mitochondrial dysfunction in adipocytes. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 958:177972. [PMID: 39662394 DOI: 10.1016/j.scitotenv.2024.177972] [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: 09/13/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 12/13/2024]
Abstract
Black phosphorus quantum dots (BPQDs) are believed to have broad prospects for application. Obesity has garnered significant attention, but the association between BPQDs and lipid metabolism has not been thoroughly investigated. Mice were orally exposed to BPQDs at doses of 0.1 and 1 mg/kg for 28 d. The exposed mice exhibited reduced insulin sensitivity, hypertrophy of white adipose tissues, and reduced thermogenic function of brown adipose tissues. In white adipocyte line (3T3-L1), exposure to 5-20 μg/mL BPQDs induced lipid accumulation, oxidative stress, and upregulated the expression of PPARγ and genes involved in de novo lipogenesis. Moreover, both a reactive oxygen species (ROS) scavenger and a PPARγ inhibitor were able to attenuate lipid accumulation and downregulate the expression of lipid-associated genes in white adipocytes. In mouse brown adipocytes, BPQDs exposure caused oxidative stress, mitochondrial dysfunction, and downregulation of thermogenic genes such as UCP1. The ROS scavenger attenuated the oxidative stress and improved the mitochondrial thermogenic function in brown adipocytes. In summary, this work demonstrates that oxidative stress induced by BPQDs mediates the lipid accumulation possibly through PPARγ activation and mitochondrial dysfunction of adipose tissues, highlighting the potential obesogenic effect of BPQDs. Our findings provide novel insights into the biosafety of BPQDs and their potential health risks to humans, offering important considerations for the sustainable application of BP materials. ENVIRONMENTAL IMPLICATION: BPQDs are a novel type of nanomaterials with unique physicochemical properties, and have broad applications in various fields, particularly in biomedicine. However, during the production and use of BPQDs as medical materials, they inevitably contact with the human body for long periods of time. Therefore, it is necessary to investigate the effects of BPQDs on organisms under long-term exposure, especially lipid metabolism. This study would be helpful decreasing the environmental health risk of BP materials and promoting their sustainable development of nanotechnology in biomedicine.
Collapse
Affiliation(s)
- Sihao Zhu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Endocrinology, Xiang'an Hospital of Xiamen University, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China
| | - Fengkai Ruan
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Endocrinology, Xiang'an Hospital of Xiamen University, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China
| | - Lingxiao Ye
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Endocrinology, Xiang'an Hospital of Xiamen University, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China
| | - Suhua Jiang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Endocrinology, Xiang'an Hospital of Xiamen University, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China
| | - Chunyan Yang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Endocrinology, Xiang'an Hospital of Xiamen University, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China
| | - Zhenghong Zuo
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Endocrinology, Xiang'an Hospital of Xiamen University, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China
| | - Chengyong He
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Endocrinology, Xiang'an Hospital of Xiamen University, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China..
| |
Collapse
|
6
|
Ju HY, Song SE, Shin SK, Jeong GS, Cho HC, Im SS, Song DK. Fulvic acid inhibits the differentiation of 3T3-L1 adipocytes by activating the Ca 2+/CaMKⅡ/AMPK pathway. Biochem Biophys Res Commun 2025; 743:151173. [PMID: 39673972 DOI: 10.1016/j.bbrc.2024.151173] [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: 11/30/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 12/16/2024]
Abstract
Type 2 diabetes increases the risk of developing obesity. Although fulvic acid alleviates back fat thickness in pigs, the mechanism underlying its anti-obesity effect remains unclear. Therefore, we investigated the anti-obesity mechanism of fulvic acid using 3T3-L1 adipocytes. We examined the effects of fulvic acid on adipocyte differentiation, cell viability, and lipid accumulation using molecular techniques. Fulvic acid treatment significantly decreased intracellular lipid accumulation in 3T3-L1 cells during the differentiation compared with that in the control group. Western blotting revealed fulvic acid-induced downregulated expression of the adipocyte differentiation-related markers peroxisome proliferator-activated receptor gamma, CCAAT/enhancer-binding protein alpha, and sterol regulatory element-binding protein 1. The fulvic acid treatment decreased the expression of the lipid uptake-related markers fatty acid-binding protein 4 and the cluster of differentiation 36 in 3T3-L1 cells. Moreover, fulvic acid significantly increased cytosolic Ca2+ concentration via Ca2+ sequestration from the endoplasmic reticulum, enhanced Ca2+/calmodulin-dependent protein kinase II (CaMKII) activity, and upregulated AMP-activated protein kinase (AMPK), thereby reducing adipocyte differentiation. Conclusively, fulvic acid attenuates adipocyte differentiation by activating the Ca2+/CaMKⅡ/AMPK pathway, suggesting its anti-obesity potential.
Collapse
Affiliation(s)
- Hyeon Yeong Ju
- Department of Physiology, Obesity-mediated Disease Research Center, Keimyung University School of Medicine, 1095 Dalgubeoldae-ro, Dalseo-gu, Daegu, 42601, Republic of Korea
| | - Seung-Eun Song
- Department of Physiology, Obesity-mediated Disease Research Center, Keimyung University School of Medicine, 1095 Dalgubeoldae-ro, Dalseo-gu, Daegu, 42601, Republic of Korea
| | - Su-Kyung Shin
- Department of Food Science and Nutrition, Kyungpook National University, 80 Daehak-ro, Pook-gu, Daegu, 41566, Republic of Korea
| | - Gil-Saeng Jeong
- College of Pharmacy, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Republic of Korea
| | - Ho-Chan Cho
- Department of Endocrinology, Internal Medicine, Keimyung University School of Medicine, 1095 Dalgubeoldae-ro, Dalseo-gu, Daegu, 42601, Republic of Korea
| | - Seung-Soon Im
- Department of Physiology, Obesity-mediated Disease Research Center, Keimyung University School of Medicine, 1095 Dalgubeoldae-ro, Dalseo-gu, Daegu, 42601, Republic of Korea
| | - Dae-Kyu Song
- Department of Physiology, Obesity-mediated Disease Research Center, Keimyung University School of Medicine, 1095 Dalgubeoldae-ro, Dalseo-gu, Daegu, 42601, Republic of Korea.
| |
Collapse
|
7
|
Lupu A, Fotea S, Jechel E, Starcea IM, Ioniuc I, Knieling A, Salaru DL, Sasaran MO, Cirstea O, Revenco N, Mihai CM, Lupu VV, Nedelcu AH. Is oxidative stress - antioxidants imbalance the physiopathogenic core in pediatric obesity? Front Immunol 2024; 15:1394869. [PMID: 39176098 PMCID: PMC11338799 DOI: 10.3389/fimmu.2024.1394869] [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: 03/02/2024] [Accepted: 07/23/2024] [Indexed: 08/24/2024] Open
Abstract
Despite the early recognition of obesity as an epidemic with global implications, research on its pathogenesis and therapeutic approach is still on the rise. The literature of the 21st century records an excess weight found in up to 1/3 of children. Both the determining factors and its systemic effects are multiple and variable. Regarding its involvement in the potentiation of cardio-vascular, pulmonary, digestive, metabolic, neuro-psychic or even dermatological diseases, the information is already broadly outlined. The connection between the underlying disease and the associated comorbidities seems to be partially attributable to oxidative stress. In addition to these, and in the light of the recent COVID-19 pandemic, the role played by oxidative stress in the induction, maintenance and potentiation of chronic inflammation among overweight children and adolescents becomes a topic of interest again. Thus, this review's purpose is to update general data on obesity, with an emphasis on the physiopathological mechanisms that underlie it and involve oxidative stress. At the same time, we briefly present the latest principles of pathology diagnosis and management. Among these, we will mainly emphasize the impact played by endogenous and exogenous antioxidants in the evolutionary course of pediatric obesity. In order to achieve our objectives, we will refer to the most recent studies published in the specialized literature.
Collapse
Affiliation(s)
- Ancuta Lupu
- Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| | - Silvia Fotea
- Clinical Medical Department, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, Galati, Romania
| | - Elena Jechel
- Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| | | | - Ileana Ioniuc
- Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| | - Anton Knieling
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| | - Delia Lidia Salaru
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| | - Maria Oana Sasaran
- Pediatrics, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology, Targu Mures, Romania
| | - Olga Cirstea
- Pediatrics, Nicolae Testemitanu State University of Medicine and Pharmacy, Chisinau, Moldova
| | - Ninel Revenco
- Pediatrics, Nicolae Testemitanu State University of Medicine and Pharmacy, Chisinau, Moldova
| | | | - Vasile Valeriu Lupu
- Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| | - Alin Horatiu Nedelcu
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| |
Collapse
|
8
|
Hong C, Li X, Zhang K, Huang Q, Li B, Xin H, Hu B, Meng F, Zhu X, Tang D, Hu C, Tao C, Li J, Cao Y, Wang H, Deng B, Wang S. Novel perspectives on autophagy-oxidative stress-inflammation axis in the orchestration of adipogenesis. Front Endocrinol (Lausanne) 2024; 15:1404697. [PMID: 38982993 PMCID: PMC11232368 DOI: 10.3389/fendo.2024.1404697] [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: 03/21/2024] [Accepted: 05/30/2024] [Indexed: 07/11/2024] Open
Abstract
Adipose tissue, an indispensable organ, fulfils the pivotal role of energy storage and metabolism and is instrumental in maintaining the dynamic equilibrium of energy and health of the organism. Adipocyte hypertrophy and adipocyte hyperplasia (adipogenesis) are the two primary mechanisms of fat deposition. Mature adipocytes are obtained by differentiating mesenchymal stem cells into preadipocytes and redifferentiation. However, the mechanisms orchestrating adipogenesis remain unclear. Autophagy, an alternative cell death pathway that sustains intracellular energy homeostasis through the degradation of cellular components, is implicated in regulating adipogenesis. Furthermore, adipose tissue functions as an endocrine organ, producing various cytokines, and certain inflammatory factors, in turn, modulate autophagy and adipogenesis. Additionally, autophagy influences intracellular redox homeostasis by regulating reactive oxygen species, which play pivotal roles in adipogenesis. There is a growing interest in exploring the involvement of autophagy, inflammation, and oxidative stress in adipogenesis. The present manuscript reviews the impact of autophagy, oxidative stress, and inflammation on the regulation of adipogenesis and, for the first time, discusses their interactions during adipogenesis. An integrated analysis of the role of autophagy, inflammation and oxidative stress will contribute to elucidating the mechanisms of adipogenesis and expediting the exploration of molecular targets for treating obesity-related metabolic disorders.
Collapse
Affiliation(s)
- Chun Hong
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Xinming Li
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Kunli Zhang
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangdong Provincial Key Laboratory of Livestock Disease Prevention Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Guangzhou, China
| | - Qiuyan Huang
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Baohong Li
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Haiyun Xin
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Bin Hu
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Fanming Meng
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Xiangxing Zhu
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Sciences and Engineering, Foshan University, Foshan, China
| | - Dongsheng Tang
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Sciences and Engineering, Foshan University, Foshan, China
| | - Chuanhuo Hu
- College of Animal Science and Technology, Guangxi University, Nanning, China
- Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, Guangxi University, Nanning, China
| | - Chenyu Tao
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, Hebei, China
| | - Jianhao Li
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Yang Cao
- Branch of Animal Husbandry, Jilin Academy of Agricultural Science, Gongzhuling, China
| | - Hai Wang
- Chinese Academy of Sciences (CAS) Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health- Hong Kong University (GIBH-HKU) Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Bo Deng
- Division of Nephrology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sutian Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, China
| |
Collapse
|
9
|
Zhou M, Ma J, Kang M, Tang W, Xia S, Yin J, Yin Y. Flavonoids, gut microbiota, and host lipid metabolism. Eng Life Sci 2024; 24:2300065. [PMID: 38708419 PMCID: PMC11065335 DOI: 10.1002/elsc.202300065] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 08/19/2023] [Accepted: 08/30/2023] [Indexed: 05/07/2024] Open
Abstract
Flavonoids are widely distributed in nature and have a variety of beneficial biological effects, including antioxidant, anti-inflammatory, and anti-obesity effects. All of these are related to gut microbiota, and flavonoids also serve as a bridge between the host and gut microbiota. Flavonoids are commonly used to modify the composition of the gut microbiota by promoting or inhibiting specific microbial species within the gut, as well as modifying their metabolites. In turn, the gut microbiota extensively metabolizes flavonoids. Hence, this reciprocal relationship between flavonoids and the gut microbiota may play a crucial role in maintaining the balance and functionality of the metabolism system. In this review, we mainly highlighted the biological effects of antioxidant, anti-inflammatory and antiobesity, and discussed the interaction between flavonoids, gut microbiota and lipid metabolism, and elaborated the potential mechanisms on host lipid metabolism.
Collapse
Affiliation(s)
- Miao Zhou
- College of Animal Science and TechnologyHunan Agricultural UniversityChangshaChina
| | - Jie Ma
- College of Animal Science and TechnologyHunan Agricultural UniversityChangshaChina
| | - Meng Kang
- College of Animal Science and TechnologyHunan Agricultural UniversityChangshaChina
| | - Wenjie Tang
- Sichuan Animal Science AcademyLivestock and Poultry Biological Products Key Laboratory of Sichuan ProvinceSichuan Animtech Feed Co., LtdChengduSichuanChina
| | - Siting Xia
- College of Animal Science and TechnologyHunan Agricultural UniversityChangshaChina
| | - Jie Yin
- College of Animal Science and TechnologyHunan Agricultural UniversityChangshaChina
| | - Yulong Yin
- College of Animal Science and TechnologyHunan Agricultural UniversityChangshaChina
| |
Collapse
|
10
|
Ramos-León J, Valencia C, Gutiérrez-Mariscal M, Rivera-Miranda DA, García-Meléndrez C, Covarrubias L. The loss of antioxidant activities impairs intestinal epithelium homeostasis by altering lipid metabolism. Exp Cell Res 2024; 437:113965. [PMID: 38378126 DOI: 10.1016/j.yexcr.2024.113965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 02/02/2024] [Accepted: 02/08/2024] [Indexed: 02/22/2024]
Abstract
Reactive oxygens species (ROS) are common byproducts of metabolic reactions and could be at the origin of many diseases of the elderly. Here we investigated the role of ROS in the renewal of the intestinal epithelium in mice lacking catalase (CAT) and/or nicotinamide nucleotide transhydrogenase (NNT) activities. Cat-/- mice have delayed intestinal epithelium renewal and were prone to develop necrotizing enterocolitis upon starvation. Interestingly, crypts lacking CAT showed fewer intestinal stem cells (ISC) and lower stem cell activity than wild-type. In contrast, crypts lacking NNT showed a similar number of ISCs as wild-type but increased stem cell activity, which was also impaired by the loss of CAT. No alteration in the number of Paneth cells (PCs) was observed in crypts of either Cat-/- or Nnt-/- mice, but they showed an evident decline in the amount of lysozyme. Cat deficiency caused fat accumulation in crypts, and a fall in the remarkable high amount of adipose triglyceride lipase (ATGL) in PCs. Notably, the low levels of ATGL in the intestine of Cat -/- mice increased after a treatment with the antioxidant N-acetyl-L-cysteine. Supporting a role of ATGL in the regulation of ISC activity, its inhibition halt intestinal organoid development. These data suggest that the reduction in the renewal capacity of intestine originates from fatty acid metabolic alterations caused by peroxisomal ROS.
Collapse
Affiliation(s)
- Javier Ramos-León
- Departamento de Genética Del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mor., Mexico
| | - Concepción Valencia
- Departamento de Genética Del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mor., Mexico
| | - Mariana Gutiérrez-Mariscal
- Departamento de Genética Del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mor., Mexico
| | - David-Alejandro Rivera-Miranda
- Departamento de Genética Del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mor., Mexico
| | - Celina García-Meléndrez
- Departamento de Genética Del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mor., Mexico
| | - Luis Covarrubias
- Departamento de Genética Del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mor., Mexico.
| |
Collapse
|
11
|
Pradel-Mora JJ, Marín G, Castillo-Rangel C, Hernández-Contreras KA, Vichi-Ramírez MM, Zarate-Calderon C, Herran Motta FS. Oxidative Stress in Postbariatric Patients: A Systematic Literature Review Exploring the Long-term Effects of Bariatric Surgery. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2024; 12:e5646. [PMID: 38515558 PMCID: PMC10956951 DOI: 10.1097/gox.0000000000005646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 01/16/2024] [Indexed: 03/23/2024]
Abstract
Background The present study investigates the impact of oxidative stress after bariatric surgery in patients with obesity. This field of study has gained great interest in recent years due to the role that oxidative stress plays in metabolic diseases. Obesity, by itself, can generate an increase in reactive oxygen and nitrogen species, intensifying cellular damage and promoting the progression of adverse metabolic conditions. In this context, bariatric surgery emerges as a candidate capable of modifying oxidative stress biomarkers, facilitating the patient's metabolic recovery. Methods A systematic review was carried out, identifying 30 studies found in databases such as PubMed, Scopus, Web of Science, and Google Scholar. It looked at the link between oxidative stress and recovery after bariatric surgery in patients. The selection of studies was based on the measurement of oxidative stress biomarkers before and after surgical intervention. Results The results reveal a significant decrease in oxidative stress biomarkers after bariatric surgery. However, a notable variability in antioxidant activity is observed between different patients, as well as a significant influence of comorbidities. Conclusions Bariatric surgery is postulated as an effective intervention in reducing oxidative stress in patients with obesity, enhancing antioxidant activity and improving patient recovery. This finding highlights the importance of considering oxidative stress management as an integral part of postoperative care, suggesting the need to implement complementary treatment strategies to optimize health outcomes.
Collapse
Affiliation(s)
- Jessica Juliana Pradel-Mora
- From the Plastic and Reconstructive Surgery, “UMAE Hospital de Especialidades Dr. Bernardo Sepúlveda Gutiérrez, Centro Médico Nacional Siglo XXI, Mexican Social Security Institute (IMSS), Mexico City, Mexico
| | - Gerardo Marín
- Neural Dynamics and Modulation Lab, Cleveland Clinic, Cleveland, Ohio
| | - Carlos Castillo-Rangel
- Department of Neurosurgery, “Hospital Regional 1º de Octubre,” Institute of Social Security and Services for State Workers (ISSSTE), Mexico City, Mexico
| | | | | | | | - Fanny Stella Herran Motta
- Plastic and Reconstructive Surgery, “Centro Médico Nacional 20 de noviembre,” Institute of Social Security and Services for State Workers (ISSSTE), Mexico City, Mexico
| |
Collapse
|
12
|
Liu Y, Zhao H, Yang Y, Liu Y, Ao CY, Zeng JM, Ban JQ, Li J. Mechanism by which HDAC3 regulates manganese induced H3K27ac in SH-SY5Y cells and intervention by curcumin. Arch Biochem Biophys 2024; 752:109878. [PMID: 38151197 DOI: 10.1016/j.abb.2023.109878] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 12/29/2023]
Abstract
Long-term excessive exposure to manganese can impair neuronal function in the brain, but the underlying pathological mechanism remains unclear. Oxidative stress plays a central role in manganese-induced neurotoxicity. Numerous studies have established a strong link between abnormal histone acetylation levels and the onset of various diseases. Histone deacetylase inhibitors and activators, such as TSA and ITSA-1, are often used to investigate the intricate mechanisms of histone acetylation in disease. In addition, recent experiments have provided substantial evidence demonstrating that curcumin (Cur) can act as an epigenetic regulator. Given these findings, this study aims to investigate the mechanisms underlying oxidative damage in SH-SY5Y cells exposed to MnCl2·4H2O, with a particular focus on histone acetylation, and to assess the potential therapeutic efficacy of Cur. In this study, SH-SY5Y cells were exposed to manganese for 24 h, were treated with TSA or ITSA-1, and were treated with or without Cur. The results suggested that manganese exposure, which leads to increased expression of HDAC3, induced H3K27 hypoacetylation, inhibited the transcription of antioxidant genes, decreased antioxidant enzyme activities, and induced oxidative damage in cells. Pretreatment with an HDAC3 inhibitor (TSA) increased the acetylation of H3K27 and the transcription of antioxidant genes and thus slowed manganese exposure-induced cellular oxidative damage. In contrast, an HDAC3 activator (ITSA-1) partially increased manganese-induced cellular oxidative damage, while Cur prevented manganese-induced oxidative damage. In summary, these findings suggest that inhibiting H3K27ac is a possible mechanism for ameliorating manganese-induced damage to dopaminergic neurons and that Cur exerts a certain protective effect against manganese-induced damage to dopaminergic neurons.
Collapse
Affiliation(s)
- Ying Liu
- School of Public Heath, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - Hua Zhao
- School of Public Heath, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - Yue Yang
- Guiyang Stomatological Hospital, Guiyang, Guizhou, 550002, China
| | - Yan Liu
- School of Public Heath, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - Chun-Yan Ao
- School of Public Heath, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - Jia-Min Zeng
- School of Public Heath, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - Jia-Qi Ban
- School of Public Heath, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, 550025, China.
| | - Jun Li
- School of Public Heath, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, 550025, China.
| |
Collapse
|
13
|
Balan AI, Halațiu VB, Scridon A. Oxidative Stress, Inflammation, and Mitochondrial Dysfunction: A Link between Obesity and Atrial Fibrillation. Antioxidants (Basel) 2024; 13:117. [PMID: 38247541 PMCID: PMC10812976 DOI: 10.3390/antiox13010117] [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: 12/18/2023] [Revised: 01/11/2024] [Accepted: 01/16/2024] [Indexed: 01/23/2024] Open
Abstract
The adipose tissue has long been thought to represent a passive source of triglycerides and fatty acids. However, extensive data have demonstrated that the adipose tissue is also a major endocrine organ that directly or indirectly affects the physiological functions of almost all cell types. Obesity is recognized as a risk factor for multiple systemic conditions, including metabolic syndrome, type 2 diabetes mellitus, sleep apnea, cardiovascular disorders, and many others. Obesity-related changes in the adipose tissue induce functional and structural changes in cardiac myocytes, promoting a wide range of cardiovascular disorders, including atrial fibrillation (AF). Due to the wealth of epidemiologic data linking AF to obesity, the mechanisms underlying AF occurrence in obese patients are an area of rich ongoing investigation. However, progress has been somewhat slowed by the complex phenotypes of both obesity and AF. The triad inflammation, oxidative stress, and mitochondrial dysfunction are critical for AF pathogenesis in the setting of obesity via multiple structural and functional proarrhythmic changes at the level of the atria. The aim of this paper is to provide a comprehensive view of the close relationship between obesity-induced oxidative stress, inflammation, and mitochondrial dysfunction and the pathogenesis of AF. The clinical implications of these mechanistic insights are also discussed.
Collapse
Affiliation(s)
- Alkora Ioana Balan
- Center for Advanced Medical and Pharmaceutical Research, University of Medicine, Pharmacy, Science and Technology “George Emil Palade” of Târgu Mureș, 540142 Târgu Mureș, Romania;
| | - Vasile Bogdan Halațiu
- Physiology Department, University of Medicine, Pharmacy, Science and Technology “George Emil Palade” of Târgu Mureș, 540142 Târgu Mureș, Romania;
| | - Alina Scridon
- Center for Advanced Medical and Pharmaceutical Research, University of Medicine, Pharmacy, Science and Technology “George Emil Palade” of Târgu Mureș, 540142 Târgu Mureș, Romania;
- Physiology Department, University of Medicine, Pharmacy, Science and Technology “George Emil Palade” of Târgu Mureș, 540142 Târgu Mureș, Romania;
| |
Collapse
|
14
|
Niveta JPS, John CM, Arockiasamy S. Monoamine oxidase mediated oxidative stress: a potential molecular and biochemical crux in the pathogenesis of obesity. Mol Biol Rep 2023; 51:29. [PMID: 38142252 DOI: 10.1007/s11033-023-08938-9] [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: 08/31/2023] [Accepted: 11/14/2023] [Indexed: 12/25/2023]
Abstract
Obesity has become a global health concern with an increasing prevalence as years pass by but the researchers have not come to a consensus on the exact pathophysiological mechanism underlying this disease. In the past three decades, Monoamine Oxidases (MAO), has come into limelight for a possible involvement in orchestrating the genesis of obesity but the exact mechanism is not well elucidated. MAO is essentially an enzyme involved in the catabolism of neurotransmitters and other biogenic amines to form a corresponding aldehyde, hydrogen peroxide (H2O2) and ammonia. This review aims to highlight the repercussions of MAO's catabolic activity on the redox balance, carbohydrate metabolism and lipid metabolism of adipocytes which ultimately leads to obesity. The H2O2 produced by these enzymes seems to be the culprit causing oxidative stress in pre-adipocytes and goes on to mimic insulin's activity independent of its presence via the Protein Kinase B Pathway facilitating glucose influx. The H2O2 activates Sterol regulatory-element binding protein-1c and peroxisome proliferator activated receptor gamma crucial for encoding enzymes like fatty acid synthase, acetyl CoA carboxylase 1, Adenosine triphosphate-citrate lyase, phosphoenol pyruvate carboxykinase etc., which helps promoting lipogenesis at the same time inhibits lipolysis. More reactive oxygen species production occurs via NADPH Oxidases enzymes and is also able activate Nuclear Factor kappa B leading to inflammation in the adipocyte microenvironment. This chronic inflammation is the seed for insulin resistance.
Collapse
Affiliation(s)
- J P Shirley Niveta
- Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | - Cordelia Mano John
- Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | | |
Collapse
|
15
|
Joung HY, Oh JM, Song MS, Kwon YB, Chun S. Selegiline Modulates Lipid Metabolism by Activating AMPK Pathways of Epididymal White Adipose Tissues in HFD-Fed Obese Mice. Pharmaceutics 2023; 15:2539. [PMID: 38004519 PMCID: PMC10675427 DOI: 10.3390/pharmaceutics15112539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/24/2023] [Accepted: 10/25/2023] [Indexed: 11/26/2023] Open
Abstract
Obesity, as a major cause of many chronic diseases such as diabetes, cardiovascular disease, and cancer, is among the most serious health problems. Increased monoamine oxidase (MAO) activity has been observed in the adipose tissue of obese humans and animals. Although previous studies have already demonstrated the potential of MAO-B inhibitors as a treatment for this condition, the mechanism of their effect has been insufficiently elucidated. In this study, we investigated the anti-obesity effect of selegiline, a selective MAO-B inhibitor, using in vivo animal models. The effect was evaluated through an assessment of body energy homeostasis, glucose tolerance tests, and biochemical analysis. Pharmacological inhibition of MAO-B by selegiline was observed to reduce body weight and fat accumulation, and improved glucose metabolism without a corresponding change in food intake, in HFD-fed obese mice. We also observed that both the expression of adipogenenic markers, including C/EBPα and FABP4, and lipogenic markers such as pACC were significantly reduced in epididymal white adipose tissues (eWATs). Conversely, increased expression of lipolytic markers such as ATGL and pHSL and AMPK phosphorylation were noted. Treating obese mice with selegiline significantly increased expression levels of UCP1 and promoted eWAT browning, indicating increased energy expenditure. These results suggest that selegiline, by inhibiting MAO-B activity, is a potential anti-obesity treatment.
Collapse
Affiliation(s)
- Hye-Young Joung
- Department of Physiology, Jeonbuk National University Medical School, Jeonju 54907, Republic of Korea; (H.-Y.J.); (J.-M.O.)
| | - Jung-Mi Oh
- Department of Physiology, Jeonbuk National University Medical School, Jeonju 54907, Republic of Korea; (H.-Y.J.); (J.-M.O.)
- Research Institute for Endocrine Sciences, Jeonbuk National University Medical School, Jeonju 54907, Republic of Korea
| | - Min-Suk Song
- Department of Microbiology, Chungbuk National University College of Medicine and Medical Research Institute, Cheongju 28644, Republic of Korea;
| | - Young-Bae Kwon
- Department of Pharmacology, Jeonbuk National University Medical School, Jeonju 54907, Republic of Korea;
| | - Sungkun Chun
- Department of Physiology, Jeonbuk National University Medical School, Jeonju 54907, Republic of Korea; (H.-Y.J.); (J.-M.O.)
- Research Institute for Endocrine Sciences, Jeonbuk National University Medical School, Jeonju 54907, Republic of Korea
| |
Collapse
|
16
|
Man AWC, Zhou Y, Xia N, Li H. Perivascular Adipose Tissue Oxidative Stress in Obesity. Antioxidants (Basel) 2023; 12:1595. [PMID: 37627590 PMCID: PMC10451984 DOI: 10.3390/antiox12081595] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/31/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
Perivascular adipose tissue (PVAT) adheres to most systemic blood vessels in the body. Healthy PVAT exerts anticontractile effects on blood vessels and further protects against cardiovascular and metabolic diseases. Healthy PVAT regulates vascular homeostasis via secreting an array of adipokine, hormones, and growth factors. Normally, homeostatic reactive oxygen species (ROS) in PVAT act as secondary messengers in various signalling pathways and contribute to vascular tone regulation. Excessive ROS are eliminated by the antioxidant defence system in PVAT. Oxidative stress occurs when the production of ROS exceeds the endogenous antioxidant defence, leading to a redox imbalance. Oxidative stress is a pivotal pathophysiological process in cardiovascular and metabolic complications. In obesity, PVAT becomes dysfunctional and exerts detrimental effects on the blood vessels. Therefore, redox balance in PVAT emerges as a potential pathophysiological mechanism underlying obesity-induced cardiovascular diseases. In this review, we summarise new findings describing different ROS, the major sources of ROS and antioxidant defence in PVAT, as well as potential pharmacological intervention of PVAT oxidative stress in obesity.
Collapse
Affiliation(s)
| | | | | | - Huige Li
- Department of Pharmacology, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany; (A.W.C.M.); (Y.Z.); (N.X.)
| |
Collapse
|
17
|
Georgieva MK, Anastassova N, Stefanova D, Yancheva D. Radical Scavenging Mechanisms of 1-Arylhydrazone Benzimidazole Hybrids with Neuroprotective Activity. J Phys Chem B 2023; 127:4364-4373. [PMID: 37163390 DOI: 10.1021/acs.jpcb.2c05784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Benzimidazole-arylhydrazone hybrids showed promising potential as multifunctional drugs for the treatment of neurodegenerative disorders. The neuroprotection studies conducted using an in vitro model of H2O2-induced oxidative stress on the SH-SY5Y cell line revealed a remarkable activity of the compound possessing a vanilloid structural fragment. The cell viability was preserved up to 84% and this effect was significantly higher than the one exerted by the reference compounds melatonin and rasagiline. Another compound with a catecholic moiety demonstrated the second-best neuroprotective activity. Computational studies were further conducted to characterize in depth the antioxidant properties of both compounds. The possible radical scavenging mechanisms were estimated as well as the most reactive sites through which the compounds may deactivate a variety of free radicals. Both of the compounds are able to deactivate not only the highly reactive hydroxyl radicals but also alkoxyl and hydroperoxyl radicals, following hydrogen atom transfer or radical adduct formation mechanism. In nonpolar medium, 3e is predicted to react slightly faster than 3a with alkoxyl radicals and around two orders of magnitude faster than 3a with hydroperoxyl radicals. The most reactive sites for formal hydrogen atom transfer in 3a are the meta-hydroxy group in the phenyl ring in water and the amide N-H group in benzene; in 3e, the amide N-H group is more reactive in both solvents. The radical adduct formation can occur at several positions in 3a and 3e, the most active being C4, C6, and C14. The stability of the formed radicals was estimated by NBO calculations. The NBO calculations indicated that the spin density in the radicals formed by the abstraction of a hydrogen atom from the amide groups of both compounds is delocalized over the phenyl ring and the hydrazone chain. The obtained theoretical data for the better radical scavenging ability of the vanilloid hybrid corroborate its experimentally established better neuroprotective activity.
Collapse
Affiliation(s)
- Miglena K Georgieva
- LAQV-REQUIMTE, Department of Chemistry, NOVA School of Science and Technology, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal
| | - Neda Anastassova
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Building 9, 1113 Sofia, Bulgaria
| | - Denitsa Stefanova
- Laboratory of Drug Metabolism and Drug Toxicity, Department of Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University-Sofia, 2 Dunav Str., 1000 Sofia, Bulgaria
| | - Denitsa Yancheva
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Building 9, 1113 Sofia, Bulgaria
| |
Collapse
|
18
|
Li J, Sun Y, Xue C, Yang X, Duan Y, Zhao D, Han J. Nogo-B deficiency suppresses white adipogenesis by regulating β-catenin signaling. Life Sci 2023; 321:121571. [PMID: 36931495 DOI: 10.1016/j.lfs.2023.121571] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/21/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023]
Abstract
AIMS Obesity is a global epidemic around the world. Reticulon-4B (Nogo-B) is an endoplasmic reticulum-resident protein. Our previous work demonstrated that Nogo-B deficiency inhibited obesity and decreased the size of white adipocytes. However, the underlying molecular mechanism of Nogo-B in white adipogenesis remains poorly understood. This study aims to explore the effect of Nogo-B in white adipogenesis, as well as its underlying molecular mechanisms. MAIN METHODS AND FINDINGS The study adopted mouse embryonic fibroblasts (MEFs) and 3T3-L1 preadipocytes to induce white adipogenesis and investigate the effect of Nogo-B on adipogenesis using qRT-PCR, Western blotting, immunofluorescence, lipid quantification, and Oil Red O staining. During white adipogenesis, Nogo-B expression was increased accompanied by upregulation of adipogenic markers. In contrast, Nogo-B deficiency inhibited white adipocyte markers expression and lipid accumulation. Furthermore, the mechanism study showed that Nogo-B deficiency decreased the destruction complex [AXIN1-APC-glycogen synthase kinase 3β (GSK3β)] levels through activating protein kinase B 2 (AKT2), resulting in β-catenin translocating into the nucleus and inhibiting the expression of adipogenic markers. Moreover, Nogo-B deficiency promoted the expression of brown/beige adipocytes markers while improving mitochondrial thermogenesis by activating β-catenin pathway. In addition, Nogo-B deficiency reduced the levels of inflammatory molecules during white adipogenic differentiation. SIGNIFICANCE This study revealed that Nogo-B deficiency inhibited white adipogenesis through AKT2/GSK3β/β-catenin pathway. Meanwhile, Nogo-B deficiency increased the expression of brown/beige adipocyte markers and promoted mitochondrial thermogenesis. In addition, Nogo-B deficiency reduced inflammatory cytokine levels caused by adipogenesis. Collectively, blocking Nogo-B expression may be a potential strategy to suppress white adipogenesis.
Collapse
Affiliation(s)
- Jiaqi Li
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Yuyao Sun
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Chao Xue
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Xiaoxiao Yang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Yajun Duan
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Dan Zhao
- Joint National Laboratory for Antibody Drug Engineering, the First Affiliated Hospital of Henan University, Kaifeng, China.
| | - Jihong Han
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China; Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China.
| |
Collapse
|
19
|
Basu T, Selman A, Reddy AP, Reddy PH. Current Status of Obesity: Protective Role of Catechins. Antioxidants (Basel) 2023; 12:antiox12020474. [PMID: 36830032 PMCID: PMC9952428 DOI: 10.3390/antiox12020474] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/31/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023] Open
Abstract
Obesity is a growing health concern in today's society. Current estimates indicate that obesity occurs in both adults and young people. Recent research also found that the Hispanic population in the U.S. is 1.9 times more likely to be overweight as compared to their non-Hispanic population. Obesity is a multifactorial disease that has a variety of causes. All current treatment options incorporate dietary changes aimed at establishing a negative energy balance. According to current scientific research, multiple factors are involved with the development of obesity, including genetic, biochemical, psychological, environmental, behavioral, and socio-demographic factors. The people who suffer from obesity are far more likely to suffer serious health problems, such as stroke, diabetes, lung disease, bone and joint disease, cancer, heart disease, neurological disorders, and poor mental health. Studies indicate that multiple cellular changes are implicated in the progression of obesity, mitochondrial dysfunction, deregulated microRNAs, inflammatory changes, hormonal deregulation, and others. This article highlights the role that oxidative stress plays in obesity and current obesity-prevention techniques with an emphasis on the impact of catechins to prevent and treat obesity.
Collapse
Affiliation(s)
- Tanisha Basu
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Ashley Selman
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Arubala P. Reddy
- Nutritional Sciences Department, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA
| | - P. Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Nutritional Sciences Department, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA
- Neurology, Departments of School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Correspondence: ; Tel.: +1-806-743-3194; Fax: +1-806-743-2334
| |
Collapse
|
20
|
Zhao L, Zheng M, Cai H, Chen J, Lin Y, Wang F, Wang L, Zhang X, Liu J. The activity comparison of six dietary flavonoids identifies that luteolin inhibits 3T3-L1 adipocyte differentiation through reducing ROS generation. J Nutr Biochem 2023; 112:109208. [PMID: 36370929 DOI: 10.1016/j.jnutbio.2022.109208] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 07/22/2022] [Accepted: 09/22/2022] [Indexed: 11/10/2022]
Abstract
Mitochondrial reactive oxygen species (ROS)generation plays an essential role in the process of adipocyte differentiation and is involved in the development of obesity and associated metabolic diseases. Various dietary flavonoids possess the substantial anti-adipogenic activity. However, it is unclear whether these flavonoids inhibit adipocyte differentiation by reducing ROS generation. In this study, the effects of six common dietary flavonoids on adipocyte differentiation were assessed in 3T3-L1 cells. The flavonoids with the same backbone of 5,7-dihydroxylflavone, including flavones apigenin, chrysin, luteolin and flavonols kaempferol, myricetin, quercetin, dose-dependently inhibited 3T3-L1 adipocyte differentiation, suggesting an associated hierarchy of inhibitory capability: luteolin > quercetin > myricetin > apigenin/kaempferol > chrysin. Meanwhile, six flavonoids were found to inhibit adipogenic gene expression and the early stage of adipocyte differentiation. Among the tested flavonoids, luteolin significantly reduced both intracellular and mitochondrial ROS generation during adipocyte differentiation. Further, luteolin treatment depressed the elevation of H2O2 concentration in the early stage of 3T3-L1 differentiation and reversed the facilitated effects of exogenous H2O2 on 3T3-L1 adipocyte differentiation and ROS generation. Altogether, the activity comparison of six dietary flavonoids identifies that luteolin inhibits 3T3-L1 adipocyte differentiation through reducing ROS generation, elucidating a new mechanism underlying the anti-adipogenic actions of flavonoids.
Collapse
Affiliation(s)
- Lingli Zhao
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, P R China
| | - Mengfei Zheng
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, P R China
| | - Hao Cai
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, P R China
| | - Juan Chen
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, P R China
| | - Yan Lin
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, P R China.
| | - Fangbin Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, P R China
| | - Lu Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, P R China
| | - Xian Zhang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, P R China
| | - Jian Liu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, P R China; Engineering Research Center of Bioprocess, Ministry of Education, Hefei University of Technology, Hefei, Anhui, P R China.
| |
Collapse
|
21
|
Wang CJ, Noble PB, Elliot JG, James AL, Wang KCW. From Beneath the Skin to the Airway Wall: Understanding the Pathological Role of Adipose Tissue in Comorbid Asthma-Obesity. Compr Physiol 2023; 13:4321-4353. [PMID: 36715283 DOI: 10.1002/cphy.c220011] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
This article provides a contemporary report on the role of adipose tissue in respiratory dysfunction. Adipose tissue is distributed throughout the body, accumulating beneath the skin (subcutaneous), around organs (visceral), and importantly in the context of respiratory disease, has recently been shown to accumulate within the airway wall: "airway-associated adipose tissue." Excessive adipose tissue deposition compromises respiratory function and increases the severity of diseases such as asthma. The mechanisms of respiratory impairment are inflammatory, structural, and mechanical in nature, vary depending on the anatomical site of deposition and adipose tissue subtype, and likely contribute to different phenotypes of comorbid asthma-obesity. An understanding of adipose tissue-driven pathophysiology provides an opportunity for diagnostic advancement and patient-specific treatment. As an exemplar, the potential impact of airway-associated adipose tissue is highlighted, and how this may change the management of a patient with asthma who is also obese. © 2023 American Physiological Society. Compr Physiol 13:4321-4353, 2023.
Collapse
Affiliation(s)
- Carolyn J Wang
- School of Human Sciences, The University of Western Australia, Crawley, Western Australia, Australia
| | - Peter B Noble
- School of Human Sciences, The University of Western Australia, Crawley, Western Australia, Australia
| | - John G Elliot
- School of Human Sciences, The University of Western Australia, Crawley, Western Australia, Australia.,Department of Pulmonary Physiology and Sleep Medicine, West Australian Sleep Disorders Research Institute, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | - Alan L James
- Department of Pulmonary Physiology and Sleep Medicine, West Australian Sleep Disorders Research Institute, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia.,Medical School, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Kimberley C W Wang
- School of Human Sciences, The University of Western Australia, Crawley, Western Australia, Australia.,Telethon Kids Institute, The University of Western Australia, Nedlands, Western Australia, Australia
| |
Collapse
|
22
|
Exploring the Role of Obesity in Dilated Cardiomyopathy Based on Bio-informatics Analysis. J Cardiovasc Dev Dis 2022; 9:jcdd9120462. [PMID: 36547458 PMCID: PMC9783214 DOI: 10.3390/jcdd9120462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/08/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
(1) Background: Obesity is a major risk factor for cardiovascular disease (CVD), contributing to increasing global disease burdens. Apart from heart failure, coronary artery disease, and arrhythmia, recent research has found that obesity also elevates the risk of dilated cardiomyopathy (DCM). The main purpose of this study was to investigate the underlying biological role of obesity in increasing the risk of DCM. (2) Methods: The datasets GSE120895, GSE19303, and GSE2508 were downloaded from the Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) were analyzed using GSE120895 for DCM and GSE2508 for obesity, and the findings were compiled to discover the common genes. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were conducted for the common genes in RStudio. In addition, CIBERSORT was used to obtain the immune cellular composition from DEGs. The key genes were identified in the set of common genes by the least absolute shrinkage and selection operator (LASSO) algorithm, the prognostic risk models of which were verified by receiver operator characteristic (ROC) curves in GSE19303. Finally, Spearman's correlation was used to explore the connections between key genes and immune cells. (3) Results: GO and KEGG pathway enrichment analyses showed that the main enriched terms of the common genes were transforming growth factor-beta (TGF-β), fibrillar collagen, NADPH oxidase activity, and multiple hormone-related signaling pathways. Both obesity and DCM had a disordered immune environment, especially obesity. The key genes NOX4, CCDC80, COL1A2, HTRA1, and KLHL29 may be primarily responsible for the changes. Spearman's correlation analysis performed for key genes and immune cells indicated that KLHL29 closely correlated to T cells and M2 macrophages, and HTRA1 very tightly correlated to plasma cells. (4) Conclusions: Bio-informatics analyses performed for DCM and obesity in our study suggested that obesity disturbed the immune micro-environment, promoted oxidative stress, and increased myocardial fibrosis, resulting in ventricular remodeling and an increased risk of DCM. The key genes KLHL29 and HTRA1 may play critical roles in obesity-related DCM.
Collapse
|
23
|
Su L, Zeng Y, Li G, Chen J, Chen X. Quercetin improves high-fat diet-induced obesity by modulating gut microbiota and metabolites in C57BL/6J mice. Phytother Res 2022; 36:4558-4572. [PMID: 35906097 DOI: 10.1002/ptr.7575] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 07/09/2022] [Accepted: 07/12/2022] [Indexed: 12/13/2022]
Abstract
High-fat diet-induced obesity is characterized by low-grade inflammation, which has been linked to gut microbiota dysbiosis. We hypothesized that quercetin supplementation would alter gut microbiota and reduce inflammation in obese mice. Male C57BL/6J mice, 4 weeks of age, were divided into 3 groups, including a low-fat diet group, a high-fat diet (HFD) group, and a high-fat diet plus quercetin (HFD+Q) group. The mice in HFD+Q group were given 50 mg per kg BW quercetin by gavage for 20 weeks. The body weight, fat accumulation, gut barrier function, glucose tolerance, and adipose tissue inflammation were determined in mice. 16 s rRNA amplicon sequence and non-targeted metabolomics analysis were used to explore the alteration of gut microbiota and metabolites. We found that quercetin significantly alleviated HFD-induced obesity, improved glucose tolerance, recovered gut barrier function, and reduced adipose tissue inflammation. Moreover, quercetin ameliorated HFD-induced gut microbiota disorder by regulating the abundance of gut microbiota, such as Adlercreutzia, Allobaculum, Coprococcus_1, Lactococcus, and Akkermansia. Quercetin influenced the production of metabolites that were linked to alterations in obesity-related inflammation and oxidative stress, such as Glycerophospho-N-palmitoyl ethanolamine, sanguisorbic acid dilactone, O-Phospho-L-serine, and P-benzoquinone. Our results demonstrate that the anti-obesity effects of quercetin may be mediated through regulation in gut microbiota and metabolites.
Collapse
Affiliation(s)
- Lijie Su
- Department of Nutrition and Food Hygiene, School of Public Health, Guangzhou Medical University, Guangzhou, China
| | - Yupeng Zeng
- Department of Nutrition and Food Hygiene, School of Public Health, Guangzhou Medical University, Guangzhou, China
| | - Guokun Li
- Department of Nutrition and Food Hygiene, School of Public Health, Guangzhou Medical University, Guangzhou, China
| | - Jing Chen
- Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Xiaoyi Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
24
|
Yuan Q, Zeng ZL, Yang S, Li A, Zu X, Liu J. Mitochondrial Stress in Metabolic Inflammation: Modest Benefits and Full Losses. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8803404. [PMID: 36457729 PMCID: PMC9708372 DOI: 10.1155/2022/8803404] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/24/2022] [Accepted: 10/26/2022] [Indexed: 09/02/2023]
Abstract
Energy intake and metabolic balance are the pillars of health preservation. Overnutrition causes nonspecific, persistently low inflammatory state known as metabolic inflammation. This condition contributes to the pathophysiology of various metabolic disorders, such as atherosclerosis, obesity, diabetes mellitus, and metabolic syndrome. The mitochondria maintain the balance of energy metabolism. Excessive energy stress can lead to mitochondrial dysfunction, which promotes metabolic inflammation. The inflammatory environment further impairs mitochondrial function. Accordingly, excellent organism design keeps the body metabolically healthy in the context of mitochondrial dysfunction, and moderate mitochondrial stress can have a beneficial effect. This review summarises the research progress on the multifaceted characterisation of mitochondrial dysfunction and its role in metabolic inflammation.
Collapse
Affiliation(s)
- Qing Yuan
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Diabetes Clinical Medical Research Center of Hunan Province, Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Z. L. Zeng
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Diabetes Clinical Medical Research Center of Hunan Province, Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Shiqi Yang
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Diabetes Clinical Medical Research Center of Hunan Province, Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Anqi Li
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Diabetes Clinical Medical Research Center of Hunan Province, Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Xuyu Zu
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Jianghua Liu
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Diabetes Clinical Medical Research Center of Hunan Province, Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
25
|
Hameed M, Geerling E, Pinto AK, Miraj I, Weger-Lucarelli J. Immune response to arbovirus infection in obesity. Front Immunol 2022; 13:968582. [PMID: 36466818 PMCID: PMC9716109 DOI: 10.3389/fimmu.2022.968582] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 11/04/2022] [Indexed: 12/26/2023] Open
Abstract
Obesity is a global health problem that affects 650 million people worldwide and leads to diverse changes in host immunity. Individuals with obesity experience an increase in the size and the number of adipocytes, which function as an endocrine organ and release various adipocytokines such as leptin and adiponectin that exert wide ranging effects on other cells. In individuals with obesity, macrophages account for up to 40% of adipose tissue (AT) cells, three times more than in adipose tissue (10%) of healthy weight individuals and secrete several cytokines and chemokines such as interleukin (IL)-1β, chemokine C-C ligand (CCL)-2, IL-6, CCL5, and tumor necrosis factor (TNF)-α, leading to the development of inflammation. Overall, obesity-derived cytokines strongly affect immune responses and make patients with obesity more prone to severe symptoms than patients with a healthy weight. Several epidemiological studies reported a strong association between obesity and severe arthropod-borne virus (arbovirus) infections such as dengue virus (DENV), chikungunya virus (CHIKV), West Nile virus (WNV), and Sindbis virus (SINV). Recently, experimental investigations found that DENV, WNV, CHIKV and Mayaro virus (MAYV) infections cause worsened disease outcomes in infected diet induced obese (DIO) mice groups compared to infected healthy-weight animals. The mechanisms leading to higher susceptibility to severe infections in individuals with obesity remain unknown, though a better understanding of the causes will help scientists and clinicians develop host directed therapies to treat severe disease. In this review article, we summarize the effects of obesity on the host immune response in the context of arboviral infections. We have outlined that obesity makes the host more susceptible to infectious agents, likely by disrupting the functions of innate and adaptive immune cells. We have also discussed the immune response of DIO mouse models against some important arboviruses such as CHIKV, MAYV, DENV, and WNV. We can speculate that obesity-induced disruption of innate and adaptive immune cell function in arboviral infections ultimately affects the course of arboviral disease. Therefore, further studies are needed to explore the cellular and molecular aspects of immunity that are compromised in obesity during arboviral infections or vaccination, which will be helpful in developing specific therapeutic/prophylactic interventions to prevent immunopathology and disease progression in individuals with obesity.
Collapse
Affiliation(s)
- Muddassar Hameed
- Department of Biomedical Sciences and Pathobiology, VA-MD Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Elizabeth Geerling
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, MO, United States
| | - Amelia K. Pinto
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, MO, United States
| | - Iqra Miraj
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, China
| | - James Weger-Lucarelli
- Department of Biomedical Sciences and Pathobiology, VA-MD Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
26
|
Chao Y, Gao L, Wang X, Cai Y, Shu Y, Zou X, Qin Y, Hu C, Dai Y, Zhu M, Shen Z, Zou C. Dysregulated adipose tissue expansion and impaired adipogenesis in Prader-Willi syndrome children before obesity-onset. Metabolism 2022; 136:155295. [PMID: 36007622 DOI: 10.1016/j.metabol.2022.155295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 08/10/2022] [Accepted: 08/18/2022] [Indexed: 10/31/2022]
Abstract
OBJECTIVE Prader-Willi syndrome (PWS) is a rare genetic imprinting disorder resulting from the expression loss of genes on the paternally inherited chromosome 15q11-13. Early-onset life-thriving obesity and hyperphagia represent the clinical hallmarks of PWS. The noncoding RNA gene SNORD116 within the minimal PWS genetic lesion plays a critical role in the pathogenesis of the syndrome. Despite advancements in understanding the genetic basis for PWS, the pathophysiology of obesity development in PWS remains largely uncharacterized. Here, we aimed to investigate the signatures of adipose tissue development and expansion pathways and associated adipose biology in PWS children without obesity-onset at an early stage, mainly from the perspective of the adipogenesis process, and further elucidate the underlying molecular mechanisms. METHODS We collected inguinal (subcutaneous) white adipose tissues (ingWATs) from phase 1 PWS and healthy children with normal weight aged from 6 M to 2 Y. Adipose morphology and histological characteristics were assessed. Primary adipose stromal vascular fractions (SVFs) were isolated, cultured in vitro, and used to determine the capacity and function of white and beige adipogenic differentiation. High-throughput RNA-sequencing (RNA-seq) was performed in adipose-derived mesenchymal stem cells (AdMSCs) to analyze transcriptome signatures in PWS subjects. Transient repression of SNORD116 was conducted to evaluate its functional relevance in adipogenesis. The changes in alternative pre-mRNA splicing were investigated in PWS and SNORD116 deficient cells. RESULTS In phase 1 PWS children, impaired white adipose tissue (WAT) development and unusual fat expansion occurred long before obesity onset, which was characterized by the massive enlargement of adipocytes accompanied by increased apoptosis. White and beige adipogenesis programs were impaired and differentiated adipocyte functions were disturbed in PWS-derived SVFs, despite increased proliferation capacity, which were consistent with the results of RNA-seq analysis of PWS AdMSCs. We also experimentally validated disrupted beige adipogenesis in adipocytes with transient SNORD116 downregulation. The transcript and protein levels of PPARγ, the adipogenesis master regulator, were significantly lower in PWS than in control AdMSCs as well as in SNORD116 deficient AdMSCs/adipocytes than in scramble (Scr) cells, resulting in the inhibited adipogenic program. Additionally, through RNA-seq, we observed aberrant transcriptome-wide alterations in alternative RNA splicing patterns in PWS cells mediated by SNORD116 loss and specifically identified a changed PRDM16 gene splicing profile in vitro. CONCLUSIONS Imbalance in the WAT expansion pathway and developmental disruption are primary defects in PWS displaying aberrant adipocyte hypertrophy and impaired adipogenesis process, in which SNORD116 deficiency plays a part. Our findings suggest that dysregulated adiposity specificity existing at an early phase is a potential pathological mechanism exacerbating hyperphagic obesity onset in PWS. This mechanistic evidence on adipose biology in young PWS patients expands knowledge regarding the pathogenesis of PWS obesity and may aid in developing a new therapeutic strategy targeting disturbed adipogenesis and driving AT plasticity to combat abnormal adiposity and associated metabolic disorders for PWS patients.
Collapse
Affiliation(s)
- Yunqi Chao
- Department of Endocrinology, The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou 310052, Zhejiang, China
| | - Lei Gao
- Department of Urology, The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou 310052, Zhejiang, China
| | - Xiangzhi Wang
- Department of Endocrinology, The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou 310052, Zhejiang, China
| | - Yuqing Cai
- Department of Endocrinology, The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou 310052, Zhejiang, China
| | - Yingying Shu
- Department of Endocrinology, The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou 310052, Zhejiang, China
| | - Xinyi Zou
- Zhejiang University City College, Hangzhou 310015, Zhejiang, China
| | - Yifang Qin
- Department of Endocrinology, The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou 310052, Zhejiang, China
| | - Chenxi Hu
- Department of Endocrinology, The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou 310052, Zhejiang, China
| | - Yangli Dai
- Department of Endocrinology, The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou 310052, Zhejiang, China
| | - Mingqiang Zhu
- Department of Endocrinology, The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou 310052, Zhejiang, China
| | - Zheng Shen
- Lab Center, The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou 310052, Zhejiang, China
| | - Chaochun Zou
- Department of Endocrinology, The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou 310052, Zhejiang, China.
| |
Collapse
|
27
|
Kleiboeker B, Lodhi IJ. Peroxisomal regulation of energy homeostasis: Effect on obesity and related metabolic disorders. Mol Metab 2022; 65:101577. [PMID: 35988716 PMCID: PMC9442330 DOI: 10.1016/j.molmet.2022.101577] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/01/2022] [Accepted: 08/16/2022] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Peroxisomes are single membrane-bound organelles named for their role in hydrogen peroxide production and catabolism. However, their cellular functions extend well beyond reactive oxygen species (ROS) metabolism and include fatty acid oxidation of unique substrates that cannot be catabolized in mitochondria, and synthesis of ether lipids and bile acids. Metabolic functions of peroxisomes involve crosstalk with other organelles, including mitochondria, endoplasmic reticulum, lipid droplets and lysosomes. Emerging studies suggest that peroxisomes are important regulators of energy homeostasis and that disruption of peroxisomal functions influences the risk for obesity and the associated metabolic disorders, including type 2 diabetes and hepatic steatosis. SCOPE OF REVIEW Here, we focus on the role of peroxisomes in ether lipid synthesis, β-oxidation and ROS metabolism, given that these functions have been most widely studied and have physiologically relevant implications in systemic metabolism and obesity. Efforts are made to mechanistically link these cellular and systemic processes. MAJOR CONCLUSIONS Circulating plasmalogens, a form of ether lipids, have been identified as inversely correlated biomarkers of obesity. Ether lipids influence metabolic homeostasis through multiple mechanisms, including regulation of mitochondrial morphology and respiration affecting brown fat-mediated thermogenesis, and through regulation of adipose tissue development. Peroxisomal β-oxidation also affects metabolic homeostasis through generation of signaling molecules, such as acetyl-CoA and ROS that inhibit hydrolysis of stored lipids, contributing to development of hepatic steatosis. Oxidative stress resulting from increased peroxisomal β-oxidation-generated ROS in the context of obesity mediates β-cell lipotoxicity. A better understanding of the roles peroxisomes play in regulating and responding to obesity and its complications will provide new opportunities for their treatment.
Collapse
Affiliation(s)
- Brian Kleiboeker
- Division of Endocrinology, Metabolism & Lipid Research, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Irfan J Lodhi
- Division of Endocrinology, Metabolism & Lipid Research, Washington University School of Medicine, St. Louis, MO 63110 USA.
| |
Collapse
|
28
|
Long S, Wang Y, Chen Y, Fang T, Yao Y, Fu K. Pan-cancer analysis of cuproptosis regulation patterns and identification of mTOR-target responder in clear cell renal cell carcinoma. Biol Direct 2022; 17:28. [PMID: 36209249 PMCID: PMC9548146 DOI: 10.1186/s13062-022-00340-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 09/19/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The mechanism of cuproptosis, a novel copper-induced cell death by regulating tricarboxylic acid cycle (TCA)-related genes, has been reported to regulate oxidative phosphorylation system (OXPHOS) in cancers and can be regarded as potential therapeutic strategies in cancer; however, the characteristics of cuproptosis in pan-cancer have not been elucidated. METHODS The multi-omics data of The Cancer Genome Atlas were used to evaluate the cuproptosis-associated characteristics across 32 tumor types. A cuproptosis enrichment score (CEScore) was established using a single sample gene enrichment analysis (ssGSEA) in pan-cancer. Spearman correlation analysis was used to identify pathway most associated with CEScore. Lasso-Cox regression was used to screen prognostic genes associated with OXPHOS and further construct a cuproptosis-related prognostic model in clear cell renal cell carcinoma (ccRCC). RESULTS We revealed that most cuproptosis-related genes (CRGs) were differentially expressed between tumors and normal tissues, and somatic copy number alterations contributed to their aberrant expression. We established a CEScore index to indicate cuproptosis status which was associated with prognosis in most cancers. The CEScore was negatively correlated with OXPHOS and significantly featured prognosis in ccRCC. The ccRCC patients with high-risk scores show worse survival outcomes and bad clinical benefits of Everolimus (mTOR inhibitor). CONCLUSIONS Our findings indicate the importance of abnormal CRGs expression in cancers. In addition, identified several prognostic CRGs as potential markers for prognostic distinction and drug response in the specific tumor. These results accelerate the understanding of copper-induced death in tumor progression and provide cuproptosis-associated novel therapeutic strategies.
Collapse
Affiliation(s)
- Shichao Long
- grid.452223.00000 0004 1757 7615Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan China
| | - Ya Wang
- grid.452223.00000 0004 1757 7615Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan China
| | - Yuqiao Chen
- grid.452223.00000 0004 1757 7615Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan China
| | - Tianshu Fang
- grid.452223.00000 0004 1757 7615Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan China
| | - Yuanbing Yao
- grid.452223.00000 0004 1757 7615Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan China
| | - Kai Fu
- Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China. .,Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China. .,Hunan Key Laboratory of Animal Models for Human Diseases, Central South University, Changsha, Hunan, China. .,National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, China. .,Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
29
|
CORM-3 Attenuates Oxidative Stress-Induced Bone Loss via the Nrf2/HO-1 Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5098358. [PMID: 36035220 PMCID: PMC9402314 DOI: 10.1155/2022/5098358] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 08/05/2022] [Indexed: 12/01/2022]
Abstract
Bone metabolism occurs in the entire life of an individual and is required for maintaining skeletal homeostasis. The imbalance between osteogenesis and osteoclastogenesis eventually leads to osteoporosis. Oxidative stress is considered a major cause of bone homeostasis disorder, and relieving excessive oxidative stress in bone mesenchymal stem cells (BMSCs) is a potential treatment strategy for osteoporosis. Carbon monoxide releasing molecule-3 (CORM-3), the classical donor of carbon monoxide (CO), possesses antioxidation, antiapoptosis, and anti-inflammatory properties. In our study, we found that CORM-3 could reduce reactive oxygen species (ROS) accumulation and prevent mitochondrial dysfunction thereby restoring the osteogenic potential of the BMSCs disrupted by hydrogen peroxide (H2O2) exposure. The action of CORM-3 was preliminarily considered the consequence of Nrf2/HO-1 axis activation. In addition, CORM-3 inhibited osteoclast formation in mouse primary bone marrow monocytes (BMMs) by inhibiting H2O2-induced polarization of M1 macrophages and endowing macrophages with M2 polarizating ability. Rat models further demonstrated that CORM-3 treatment could restore bone mass and enhance the expression of Nrf2 and osteogenic markers in the distal femurs. In summary, CORM-3 is a potential therapeutic agent for the treatment of osteoporosis.
Collapse
|
30
|
Bacil GP, Cogliati B, Cardoso DR, Barbisan LF, Romualdo GR. Are isothiocyanates and polyphenols from Brassicaceae vegetables emerging as preventive/therapeutic strategies for NAFLD? The landscape of recent preclinical findings. Food Funct 2022; 13:8348-8362. [PMID: 35899794 DOI: 10.1039/d2fo01488b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a lipid impairment-related chronic metabolic disease that affects almost 25% of the worldwide population and has become the leading cause of liver transplantation in the United States of America (USA). NAFLD may progress from simple hepatic steatosis (HS) to nonalcoholic steatohepatitis (NASH), which occurs simultaneously in an inflammatory and fibrotic microenvironment and affects approximately 5% of the global population. Recently, NASH has been suggested to be a relevant driver in progressive liver cirrhosis and a population-attributable factor in hepatocellular carcinoma patients. Moreover, predictions show that NAFLD-related annual health costs in the USA have reached ∼$100 bi., but effective therapies are still scarce. Thus, new preventative strategies for this hepatic disease urgently need to be developed. The Brassicaceae vegetable family includes almost 350 genera and 3500 species and these are one of the main types of vegetables harvested and produced worldwide. These vegetables are well-known sources of glucobrassicin-derivative molecules, such as isothiocyanates and phenolic compounds, which have shown antioxidant and antilipogenic effects in preclinical NAFLD data. In this review, we gathered prominent evidence of the in vivo and in vitro effects of these vegetable-derived nutraceutical compounds on the gut-liver-adipose axis, which is a well-known regulator of NAFLD and may represent a new strategy for disease control.
Collapse
Affiliation(s)
- Gabriel P Bacil
- São Paulo State University (UNESP), Botucatu Medical School, Department of Pathology, Botucatu, SP, Brazil.
| | - Bruno Cogliati
- University of São Paulo (USP), School of Veterinary and Animal Science, Department of Pathology, São Paulo, SP, Brazil
| | - Daniel R Cardoso
- University of São Paulo (USP), São Carlos Institute of Chemistry (IQSC), São Carlos, SP, Brazil
| | - Luís Fernando Barbisan
- São Paulo State University (UNESP), Department of Structural and Functional Biology, SP, Brazil
| | - Guilherme R Romualdo
- São Paulo State University (UNESP), Botucatu Medical School, Department of Pathology, Botucatu, SP, Brazil. .,São Paulo State University (UNESP), Department of Structural and Functional Biology, SP, Brazil
| |
Collapse
|
31
|
Yang Y, Liu Y, Zhang AL, Tang SF, Ming Q, Ao CY, Liu Y, Li CZ, Yu C, Zhao H, Chen L, Li J. Curcumin protects against manganese-induced neurotoxicity in rat by regulating oxidative stress-related gene expression via H3K27 acetylation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 236:113469. [PMID: 35367881 DOI: 10.1016/j.ecoenv.2022.113469] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/22/2022] [Accepted: 03/26/2022] [Indexed: 06/14/2023]
Abstract
Long-term manganese exposure causes a neurodegenerative disorder referred to as manganese poisoning, but the mechanism remains unclear and no specific treatment is available. Oxidative stress is widely recognised as one of the main causes of manganese-induced neurotoxicity. In recent years, the role of histone acetylation in neurodegenerative diseases has been widely concerned. curcumin is a natural polyphenol compound extracted from the rhizome of turmeric and exhibits both antioxidant and neuroprotective properties. Therefore, we aimed to investigate whether and how curcumin protects against manganese-induced neurotoxicity from the perspective of histone acetylation, based on the reversibility of histone acetylation modification. In this study, rats were treated with or without curcumin and subjected to long-term manganese exposure. Results that treatment of manganese decreased the protein expression of H3K18 acetylation and H3K27 acetylation at the promoters of oxidative stress-related genes and inhibited the expression of these genes. Nevertheless, curcumin increased the H3K27 acetylation level at the manganese superoxide dismutase (SOD2) gene promoter and promoted the expression of SOD2 gene. Oxidative damage in the rat striatum as well as learning and memory dysfunction were ameliorated after curcumin treatment. Taken together, our results suggest that the regulation of oxidative stress by histone acetylation may be a key mechanism of manganese-induced neurotoxicity. In addition, curcumin ameliorates Mn-induced neurotoxicity may be due to alleviation of oxidative damage mediated by increased activation of H3K27 acetylation at the SOD2 gene promoter.
Collapse
Affiliation(s)
- Yue Yang
- School of Public Heath, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 550025, China
| | - Ying Liu
- School of Public Heath, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 550025, China
| | - An-Liu Zhang
- Guiyang Center for Disease Control and Prevention, Guiyang, Guizhou 550003, China
| | - Shun-Fang Tang
- School of Public Heath, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 550025, China
| | - Qian Ming
- School of Public Heath, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 550025, China
| | - Chun-Yan Ao
- School of Public Heath, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 550025, China
| | - Yan Liu
- School of Public Heath, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 550025, China
| | - Chang-Zhe Li
- School of Public Heath, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 550025, China
| | - Chun Yu
- School of Public Heath, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 550025, China
| | - Hua Zhao
- School of Public Heath, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 550025, China
| | - Li Chen
- School of Public Heath, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 550025, China
| | - Jun Li
- School of Public Heath, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 550025, China.
| |
Collapse
|
32
|
Wang X, Li P, He S, Xing S, Cao Z, Cao X, Liu B, Li ZH. Effects of tralopyril on histological, biochemical and molecular impacts in Pacific oyster, Crassostrea gigas. CHEMOSPHERE 2022; 289:133157. [PMID: 34871613 DOI: 10.1016/j.chemosphere.2021.133157] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 06/13/2023]
Abstract
Recently, the toxic effects of tralopyril, as a new antifouling biocide, on aquatic organisms have aroused widespread attention about the potential toxicity. However, the mechanism of tralopyril on marine mollusks has not been elaborated clearly. In this study, the histological, biochemical and molecular impacts of tralopyril on adult Crassostrea gigas were investigated. The results indicated that the 96 h LC50 of tralopyril to adult Crassostrea gigas was 911 μg/L. After exposure to tralopyril (0, 40, 80 and 160 μg/L) for 6 days, the mantle mucus secretion coverage ratio of Crassostrea gigas was increased with a dose-dependent pattern. Catalase (CAT) activity was significantly increased, amylase (AMS) activity, acid phosphatase (ACP) activity and calcium ion (Ca2+) concentration significantly decreased. Meanwhile, integrated biomarker responses (IBR) index suggested that higher concentrations of tralopyril caused severer damage to Crassostrea gigas. In addition, the mRNA expression levels of biomineralization related genes in the mantle were significantly upregulated. Collectively, this study firstly revealed the histological, biochemical and molecular impacts of tralopyril exposure on adult Crassostrea gigas, which provided new insights for understanding the toxicity of tralopyril in marine mollusks.
Collapse
Affiliation(s)
- Xu Wang
- Marine College, Shandong University, Weihai, Shandong, 264209, China
| | - Ping Li
- Marine College, Shandong University, Weihai, Shandong, 264209, China
| | - Shuwen He
- Marine College, Shandong University, Weihai, Shandong, 264209, China
| | - Shaoying Xing
- Marine College, Shandong University, Weihai, Shandong, 264209, China
| | - Zhihan Cao
- Marine College, Shandong University, Weihai, Shandong, 264209, China
| | - Xuqian Cao
- Marine College, Shandong University, Weihai, Shandong, 264209, China
| | - Bin Liu
- Marine College, Shandong University, Weihai, Shandong, 264209, China
| | - Zhi-Hua Li
- Marine College, Shandong University, Weihai, Shandong, 264209, China.
| |
Collapse
|
33
|
Chen H, Li J, Zhang Y, Zhang W, Li X, Tang H, Liu Y, Li T, He H, Du B, Li L, Shi M. Bisphenol F suppresses insulin-stimulated glucose metabolism in adipocytes by inhibiting IRS-1/PI3K/AKT pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 231:113201. [PMID: 35051757 DOI: 10.1016/j.ecoenv.2022.113201] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 01/10/2022] [Accepted: 01/12/2022] [Indexed: 06/14/2023]
Abstract
Obesity is one of the risk factors of metabolic diseases. Decreased sensitivity to insulin or impairment of the insulin signaling pathway may affect the metabolism of adipose tissue. Bisphenol F (BPF) has been widely used in various products as a substitute for bisphenol A (BPA). BPA has been defined as "obesogen". However, knowledge about the correlation between BPF and obesity is very limited. This study was aimed to explore the effects of BPF on glucose metabolism and insulin sensitivity in mammalian tissues, using a mouse 3T3-L1 adipocyte line as the model. Differentiated 3T3-L1 adipocytes were treated with BPF at various concentrations for 24 h or 48 h, followed by the measurement of cell viability, lipid accumulation, expression levels of adipocytokines, glucose consumption, and impairment of the insulin signaling pathway. The results indicated that BPF had no effect on the size of 3T3-L1 adipocytes, but the expression of leptin, adiponectin and apelin was decreased, while that of chemerin and resistin was increased after 48 h of BPF treatment. Moreover, BPF inhibited the glucose consumption, the expression of GLUT4, and its translocation to the plasma membranes in 3T3-L1 adipocytes. Western blot analysis indicated that the activation of IRS-1/PI3K/AKT signaling pathway was inhibited by BPF, which resulted in reduced GLUT4 translocation. In conclusion, our data suggest that exposure of adipocytes to BPF may alter the expression of calorie metabolism-related adipokines and suppress insulin-stimulated glucose metabolism by impairing the insulin signaling (IRS-1/PI3K/AKT) pathway.
Collapse
Affiliation(s)
- Huiling Chen
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, Guangdong Province, China
| | - Jiangbin Li
- School of Medical Technology, Guangdong Medical University, Dongguan 523808, Guangdong Province, China
| | - Yanchao Zhang
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, Guangdong Province, China
| | - Wei Zhang
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, Guangdong Province, China
| | - Xing Li
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, Guangdong Province, China
| | - Huanwen Tang
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, Guangdong Province, China
| | - Yungang Liu
- Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Tianlan Li
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, Guangdong Province, China
| | - Haoqi He
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, Guangdong Province, China
| | - Bohai Du
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, Guangdong Province, China
| | - Li Li
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, Guangdong Province, China.
| | - Ming Shi
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, Guangdong Province, China; Dongguan Liaobu Hospital, Dongguan 523808, Guangdong Province, China.
| |
Collapse
|
34
|
Lu XH, Zhang J, Xiong Q. Suppressive effect erythropoietin on oxidative stress by targeting AMPK/Nox4/ROS pathway in renal ischemia reperfusion injury. Transpl Immunol 2022; 72:101537. [PMID: 35031454 DOI: 10.1016/j.trim.2022.101537] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/09/2022] [Accepted: 01/09/2022] [Indexed: 11/27/2022]
Abstract
OBJECTIVE To explore the effect of erythropoietin (EPO) on the AMP-activated protein kinase (AMPK)/nicotinamide adenine dinucleotide phosphatase oxidase 4 (NOX4) signaling pathway during renal ischemia reperfusion injury (RIRI) in rats. METHODS A rat model of RIRI was established by clamping the left renal pedicle and removing the right kidney. The rats in the sham group did not have their left renal pedicle clamped. Rats with a model of RIRI were randomly divided into RIRI alone (control), erythropoietin treatment (EPO/RIRI), and Compound C treatment (CPC/RIRI) groups. Hematoxylin-eosin (H&E) staining was used to examine pathological kidney damage. Serum creatinine and urea nitrogen levels were measured to evaluate renal function. Western blotting was performed to detect the expression levels of phosphorylated p-AMPK and total AMPK protein in the kidneys. RT-PCR was used to evaluate the mRNA levels of Nox4 and p22 in the kidneys. Oxidative stress-related indices (ROS, CAT, GSH, SOD, and MDA) were also measured. RESULTS EPO treatment improved kidney function by preventing kidney damage induced by the RIRI model. Preventing ischemia/reperfusion injury in the RIRI model was correlated with an increased p-AMPK/AMPK ratio and elevated activity of CAT, GSH, and SOD, which ameliorated the expression of NOX4, p22, ROS, and MDA. Moreover, treatment with CPC (an AMPK inhibitor) reduced the effects of EPO in the RIRI model. CONCLUSION EPO treatment protected rats against RIRI in the RIRI model by alleviating oxidative stress by triggering the AMPK/NOX4/ROS pathway.
Collapse
Affiliation(s)
- Xiang-Heng Lu
- Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Jiong Zhang
- Department of Nephrology, University of Electronic Science and Technology, Sichuan Academy of Sciences & Sichuan Provincial People's Hospital, Sichuan Clinical Research Center for Kidney Disease, Chengdu 610072, China
| | - Qin Xiong
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Nanchang University, Nanchang 330006, China.
| |
Collapse
|
35
|
Jakubiak GK, Osadnik K, Lejawa M, Osadnik T, Goławski M, Lewandowski P, Pawlas N. "Obesity and Insulin Resistance" Is the Component of the Metabolic Syndrome Most Strongly Associated with Oxidative Stress. Antioxidants (Basel) 2021; 11:79. [PMID: 35052583 PMCID: PMC8773170 DOI: 10.3390/antiox11010079] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/22/2021] [Accepted: 12/24/2021] [Indexed: 02/06/2023] Open
Abstract
Metabolic syndrome (MS) is not a homogeneous entity, but this term refers to the coexistence of factors that increase the risk for the development of type 2 diabetes and cardiovascular disease. There are different versions of the criteria for the diagnosis of MS, which makes the population of patients diagnosed with MS heterogeneous. Research to date shows that MS is associated with oxidative stress (OS), but it is unclear which MS component is most strongly associated with OS. The purpose of the study was to investigate the relationship between the parameters of OS and the presence of individual elements of MS in young adults, as well as to identify the components of MS by means of principal components analysis (PCA) and to investigate how the parameters of OS correlate with the presence of individual components. The study included 724 young adults with or without a family history of coronary heart disease (population of the MAGNETIC study). Blood samples were taken from the participants of the study to determine peripheral blood counts, biochemical parameters, and selected parameters of OS. In addition, blood pressure and anthropometric parameters were measured. In subjects with MS, significantly lower activity of superoxide dismutase (SOD), copper- and zinc-containing SOD (CuZnSOD), and manganese-containing SOD (MnSOD) were found, along with significantly higher total antioxidant capacity (TAC) and significantly lower concentration of thiol groups per gram of protein (PSH). We identified three components of MS by means of PCA: "Obesity and insulin resistance", "Dyslipidemia", and "Blood pressure", and showed the component "Obesity and insulin resistance" to have the strongest relationship with OS. In conclusion, we documented significant differences in some parameters of OS between young adults with and without MS. We showed that "Obesity and insulin resistance" is the most important component of MS in terms of relationship with OS.
Collapse
Affiliation(s)
- Grzegorz K. Jakubiak
- Department of Pharmacology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Jordana 38 Street, 41-808 Zabrze, Poland; (K.O.); (M.L.); (T.O.); (N.P.)
- Department and Clinic of Internal Medicine, Angiology, and Physical Medicine, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Batorego 15 Street, 41-902 Bytom, Poland
| | - Kamila Osadnik
- Department of Pharmacology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Jordana 38 Street, 41-808 Zabrze, Poland; (K.O.); (M.L.); (T.O.); (N.P.)
| | - Mateusz Lejawa
- Department of Pharmacology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Jordana 38 Street, 41-808 Zabrze, Poland; (K.O.); (M.L.); (T.O.); (N.P.)
| | - Tadeusz Osadnik
- Department of Pharmacology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Jordana 38 Street, 41-808 Zabrze, Poland; (K.O.); (M.L.); (T.O.); (N.P.)
| | - Marcin Goławski
- Student Research Group, Department of Pharmacology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Jordana 38 Street, 41-808 Zabrze, Poland; (M.G.); (P.L.)
| | - Piotr Lewandowski
- Student Research Group, Department of Pharmacology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Jordana 38 Street, 41-808 Zabrze, Poland; (M.G.); (P.L.)
| | - Natalia Pawlas
- Department of Pharmacology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Jordana 38 Street, 41-808 Zabrze, Poland; (K.O.); (M.L.); (T.O.); (N.P.)
| |
Collapse
|
36
|
Silva-Boghossian CM, Dezonne RS. What Are the Clinical and Systemic Results of Periodontitis Treatment in Obese Individuals? ACTA ACUST UNITED AC 2021; 8:48-65. [PMID: 34367878 PMCID: PMC8327900 DOI: 10.1007/s40496-021-00295-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2021] [Indexed: 12/24/2022]
Abstract
Purpose of Review Periodontitis and obesity are characterized by a dysregulated inflammatory state. Obese individuals have a higher chance of presenting periodontitis. Clinical studies in different populations demonstrate that individuals with obesity have worse periodontal conditions. This current review aims to explore recent literature to understand what the impacts of obesity on periodontal treatment outcomes are and to learn whether periodontal treatment can improve systemic biomarkers in obese individuals. Recent Findings Short- and long-term evaluations demonstrated that non-surgical periodontal treatment could improve clinical parameters in obese individuals, represented as the reduction in mean probing depth, sites with probing depth ≥ 4 mm, and extension of bleeding on probing. However, obese individuals may have less clinical improvement when compared to normal-weight individuals with a similar periodontal profile. Additionally, periodontal treatment may contribute to a reduction in systemic levels of retinol-binding protein 4 and leptin, while promoting an increase in systemic levels of adiponectin. Summary Overall, obese individuals with periodontitis can significantly benefit from non-surgical periodontal treatment. However, clinical improvements seem to be less prominent in obese individuals with periodontitis compared to non-obese individuals with similar periodontal status. Nevertheless, periodontal treatment may impact significantly on the reduction of several biochemical biomarkers of obesity with or without weight reduction. Further investigations are needed to improve our comprehension of the mechanisms underlying those findings.
Collapse
Affiliation(s)
- Carina M. Silva-Boghossian
- Periodontics, School of Dentistry, Federal University of Rio de Janeiro, Rua Professor Rodolpho Paulo Rocco, 325, Cidade Universitaria, Rio de Janeiro, RJ CEP 21941-617 Brazil
| | - Romulo S. Dezonne
- Postgraduate Program in Translational Biomedicine, University of Grande Rio, Duque de Caxias, RJ Brazil
| |
Collapse
|
37
|
Redox Regulation of Lipid Mobilization in Adipose Tissues. Antioxidants (Basel) 2021; 10:antiox10071090. [PMID: 34356323 PMCID: PMC8301038 DOI: 10.3390/antiox10071090] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/30/2021] [Accepted: 07/01/2021] [Indexed: 12/14/2022] Open
Abstract
Lipid mobilization in adipose tissues, which includes lipogenesis and lipolysis, is a paramount process in regulating systemic energy metabolism. Reactive oxygen and nitrogen species (ROS and RNS) are byproducts of cellular metabolism that exert signaling functions in several cellular processes, including lipolysis and lipogenesis. During lipolysis, the adipose tissue generates ROS and RNS and thus requires a robust antioxidant response to maintain tight regulation of redox signaling. This review will discuss the production of ROS and RNS within the adipose tissue, their role in regulating lipolysis and lipogenesis, and the implications of antioxidants on lipid mobilization.
Collapse
|
38
|
Effects of Dufulin on Oxidative Stress and Metabolomic Profile of Tubifex. Metabolites 2021; 11:metabo11060381. [PMID: 34208357 PMCID: PMC8231163 DOI: 10.3390/metabo11060381] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 06/03/2021] [Accepted: 06/03/2021] [Indexed: 12/19/2022] Open
Abstract
Dufulin is a highly effective antiviral pesticide used in plants. In this study, a seven-day experiment was conducted to evaluate the effects of Dufulin at five different concentrations (1 × 10−4, 1 × 10−3, 1 × 10−2, 0.1, and 1 mg/L) on Tubifex. LC-MS-based metabolome analysis detected a total of 5356 features in positive and 9110 features in negative, of which 41 showed significant changes and were identified as differential metabolites. Four metabolic pathways were selected for further study. Detailed analysis revealed that Dufulin exposure affected the urea cycle of Tubifex, probably via argininosuccinate lyase (ASL) inhibition. It also affected the fatty acid metabolism, leading to changes in the concentration of free fatty acids in Tubifex. Furthermore, the changes in metabolites after exposure to Dufulin at 1 × 10−2 mg/L were different from those at the other concentrations.
Collapse
|
39
|
Lee H, Jose PA. Coordinated Contribution of NADPH Oxidase- and Mitochondria-Derived Reactive Oxygen Species in Metabolic Syndrome and Its Implication in Renal Dysfunction. Front Pharmacol 2021; 12:670076. [PMID: 34017260 PMCID: PMC8129499 DOI: 10.3389/fphar.2021.670076] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 04/14/2021] [Indexed: 12/16/2022] Open
Abstract
Metabolic syndrome (MetS), a complex of interrelated risk factors for cardiovascular disease and diabetes, is comprised of central obesity (increased waist circumference), hyperglycemia, dyslipidemia (high triglyceride blood levels, low high-density lipoprotein blood levels), and increased blood pressure. Oxidative stress, caused by the imbalance between pro-oxidant and endogenous antioxidant systems, is the primary pathological basis of MetS. The major sources of reactive oxygen species (ROS) associated with MetS are nicotinamide-adenine dinucleotide phosphate (NADPH) oxidases and mitochondria. In this review, we summarize the current knowledge regarding the generation of ROS from NADPH oxidases and mitochondria, discuss the NADPH oxidase- and mitochondria-derived ROS signaling and pathophysiological effects, and the interplay between these two major sources of ROS, which leads to chronic inflammation, adipocyte proliferation, insulin resistance, and other metabolic abnormalities. The mechanisms linking MetS and chronic kidney disease are not well known. The role of NADPH oxidases and mitochondria in renal injury in the setting of MetS, particularly the influence of the pyruvate dehydrogenase complex in oxidative stress, inflammation, and subsequent renal injury, is highlighted. Understanding the molecular mechanism(s) underlying MetS may lead to novel therapeutic approaches by targeting the pyruvate dehydrogenase complex in MetS and prevent its sequelae of chronic cardiovascular and renal diseases.
Collapse
Affiliation(s)
- Hewang Lee
- Department of Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Pedro A Jose
- Department of Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, United States.,Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| |
Collapse
|
40
|
Contribution of Adipose Tissue Oxidative Stress to Obesity-Associated Diabetes Risk and Ethnic Differences: Focus on Women of African Ancestry. Antioxidants (Basel) 2021; 10:antiox10040622. [PMID: 33921645 PMCID: PMC8073769 DOI: 10.3390/antiox10040622] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/15/2021] [Accepted: 03/26/2021] [Indexed: 12/14/2022] Open
Abstract
Adipose tissue (AT) storage capacity is central in the maintenance of whole-body homeostasis, especially in obesity states. However, sustained nutrients overflow may dysregulate this function resulting in adipocytes hypertrophy, AT hypoxia, inflammation and oxidative stress. Systemic inflammation may also contribute to the disruption of AT redox equilibrium. AT and systemic oxidative stress have been involved in the development of obesity-associated insulin resistance (IR) and type 2 diabetes (T2D) through several mechanisms. Interestingly, fat accumulation, body fat distribution and the degree of how adiposity translates into cardio-metabolic diseases differ between ethnicities. Populations of African ancestry have a higher prevalence of obesity and higher T2D risk than populations of European ancestry, mainly driven by higher rates among African women. Considering the reported ethnic-specific differences in AT distribution and function and higher levels of systemic oxidative stress markers, oxidative stress is a potential contributor to the higher susceptibility for metabolic diseases in African women. This review summarizes existing evidence supporting this hypothesis while acknowledging a lack of data on AT oxidative stress in relation to IR in Africans, and the potential influence of other ethnicity-related modulators (e.g., genetic-environment interplay, socioeconomic factors) for consideration in future studies with different ethnicities.
Collapse
|
41
|
Zhou Y, Li H, Xia N. The Interplay Between Adipose Tissue and Vasculature: Role of Oxidative Stress in Obesity. Front Cardiovasc Med 2021; 8:650214. [PMID: 33748199 PMCID: PMC7969519 DOI: 10.3389/fcvm.2021.650214] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 02/10/2021] [Indexed: 12/15/2022] Open
Abstract
Cardiovascular diseases (CVDs) rank the leading cause of morbidity and mortality globally. Obesity and its related metabolic syndrome are well-established risk factors for CVDs. Therefore, understanding the pathophysiological role of adipose tissues is of great importance in maintaining cardiovascular health. Oxidative stress, characterized by excessive formation of reactive oxygen species, is a common cellular stress shared by obesity and CVDs. While plenty of literatures have illustrated the vascular oxidative stress, very few have discussed the impact of oxidative stress in adipose tissues. Adipose tissues can communicate with vascular systems, in an endocrine and paracrine manner, through secreting several adipocytokines, which is largely dysregulated in obesity. The aim of this review is to summarize current understanding of the relationship between oxidative stress in obesity and vascular endothelial dysfunction. In this review, we briefly describe the possible causes of oxidative stress in obesity, and the impact of obesity-induced oxidative stress on adipose tissue function. We also summarize the crosstalk between adipose tissue and vasculature mediated by adipocytokines in vascular oxidative stress. In addition, we highlight the potential target mediating adipose tissue oxidative stress.
Collapse
Affiliation(s)
- Yawen Zhou
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Huige Li
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Ning Xia
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany
| |
Collapse
|
42
|
Randi EB, Casili G, Jacquemai S, Szabo C. Selenium-Binding Protein 1 (SELENBP1) Supports Hydrogen Sulfide Biosynthesis and Adipogenesis. Antioxidants (Basel) 2021; 10:antiox10030361. [PMID: 33673622 PMCID: PMC7997437 DOI: 10.3390/antiox10030361] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 02/19/2021] [Accepted: 02/24/2021] [Indexed: 12/12/2022] Open
Abstract
Hydrogen sulfide (H2S), a mammalian gasotransmitter, is involved in the regulation of a variety of fundamental processes including intracellular signaling, cellular bioenergetics, cell proliferation, and cell differentiation. Cystathionine γ-lyase (CSE), cystathionine β-synthase (CBS), and 3-mercaptopyruvate sulfurtransferase (3-MST) are currently considered the three principal mammalian H2S-generating enzymes. However, recently, a fourth H2S-producing enzyme, selenium-binding-protein 1 (SELENBP1), has also been identified. The cellular regulatory role(s) of SELENBP1 are incompletely understood. The current study investigated whether SELENBP1 plays a role in the regulation of adipocyte differentiation in vitro. 3T3-L1 preadipocytes with or without SELENBP1 knock-down were subjected to differentiation-inducing conditions, and H2S production, cellular lipid accumulation, cell proliferation, and mitochondrial activity were quantified. Adipocyte differentiation was associated with an upregulation of H2S biosynthesis. SELENBP1 silencing decreased cellular H2S levels, suppressed the expression of the three “classical” H2S-producing enzymes (CBS, CSE, and 3-MST) and significantly suppressed adipocyte differentiation. Treatment of SELENBP1 knock-down cells with the H2S donor GYY4137 partially restored lipid accumulation, increased cellular H2S levels, and exerted a bell-shaped effect on cellular bioenergetics (enhancement at 1 and 3 mM, and inhibition at 6 mM). We conclude that SELENBP1 in adipocytes (1) contributes to H2S biosynthesis and (2) acts as an endogenous stimulator of adipocyte differentiation.
Collapse
|
43
|
Ransy C, Vaz C, Lombès A, Bouillaud F. Use of H 2O 2 to Cause Oxidative Stress, the Catalase Issue. Int J Mol Sci 2020; 21:E9149. [PMID: 33266350 PMCID: PMC7731207 DOI: 10.3390/ijms21239149] [Citation(s) in RCA: 164] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 11/27/2020] [Accepted: 11/28/2020] [Indexed: 01/08/2023] Open
Abstract
Addition of hydrogen peroxide (H2O2) is a method commonly used to trigger cellular oxidative stress. However, the doses used (often hundreds of micromolar) are disproportionally high with regard to physiological oxygen concentration (low micromolar). In this study using polarographic measurement of oxygen concentration in cellular suspensions we show that H2O2 addition results in O2 release as expected from catalase reaction. This reaction is fast enough to, within seconds, decrease drastically H2O2 concentration and to annihilate it within a few minutes. Firstly, this is likely to explain why recording of oxidative damage requires the high concentrations found in the literature. Secondly, it illustrates the potency of intracellular antioxidant (H2O2) defense. Thirdly, it complicates the interpretation of experiments as subsequent observations might result from high/transient H2O2 exposure and/or from the diverse possible consequences of the O2 release.
Collapse
Affiliation(s)
| | | | | | - Frédéric Bouillaud
- Institut Cochin, Université de Paris, INSERM, CNRS, F-75014 Paris, France; (C.R.); (C.V.); (A.L.)
| |
Collapse
|