1
|
Qian L, Lu S, Jiang W, Mu Q, Lin Y, Gu Z, Wu Y, Ge X, Miao L. Lactobacillus plantarum Alters Gut Microbiota and Metabolites Composition to Improve High Starch Metabolism in Megalobrama amblycephala. Animals (Basel) 2025; 15:583. [PMID: 40003065 PMCID: PMC11852042 DOI: 10.3390/ani15040583] [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: 01/06/2025] [Revised: 01/20/2025] [Accepted: 01/23/2025] [Indexed: 02/27/2025] Open
Abstract
The aim of this study was to explore the effects of adding Lactobacillus plantarum (LAB) to a high-starch diet on glucose and lipid metabolism, gut microbiota, and the composition of metabolites in Megalobrama amblycephala. This experiment was equipped with three isonitrogenous and isoenergetic feeds as control group (LW), high starch group (HW), and high starch with LAB group (HP). A total of 180 experimental fish (13.5 ± 0.5 g) were randomly divided into three treatments, and three floating cages (1 m × 1 m × 1 m) were set up for each treatment. A total of 20 fish per net were kept in an outdoor pond for 8 weeks. The results showed that both the HW and HP groups had an altered structure and a reduced diversity of gut microbiota. LAB increased the abundance of Cetobacterium and the ratio of Firmicutes/Bacteroidota and decreased PC (16:1/20:5) and taurochenodeoxycholic acid levels. LAB promoted the expression of genes related to the intestinal bile acid cycle (fxr, hmgcr, rxr, shp and hnf4α) and inhibited the expression of pparβ and g6pase (p < 0.05). LAB reduced the expression of genes related to transported cholesterol (lxr and ldlr) (p < 0.05) in the liver. In conclusion, LAB addition could regulate the gut microbiota disorders caused by high starch levels, promote cholesterol metabolism, produce bile acids, and reduce lipid deposition.
Collapse
Affiliation(s)
- Linjie Qian
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China; (L.Q.); (Q.M.)
| | - Siyue Lu
- Key Laboratory for Genetic Breeding of Aquatic Animals and Aquaculture Biology, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (S.L.); (W.J.); (Y.L.); (Z.G.)
| | - Wenqiang Jiang
- Key Laboratory for Genetic Breeding of Aquatic Animals and Aquaculture Biology, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (S.L.); (W.J.); (Y.L.); (Z.G.)
| | - Qiaoqiao Mu
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China; (L.Q.); (Q.M.)
| | - Yan Lin
- Key Laboratory for Genetic Breeding of Aquatic Animals and Aquaculture Biology, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (S.L.); (W.J.); (Y.L.); (Z.G.)
| | - Zhengyan Gu
- Key Laboratory for Genetic Breeding of Aquatic Animals and Aquaculture Biology, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (S.L.); (W.J.); (Y.L.); (Z.G.)
| | - Yeyang Wu
- ANYOU Biotechnology Group Co., Ltd., Taicang 215421, China;
| | - Xianping Ge
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China; (L.Q.); (Q.M.)
- Key Laboratory for Genetic Breeding of Aquatic Animals and Aquaculture Biology, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (S.L.); (W.J.); (Y.L.); (Z.G.)
| | - Linghong Miao
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China; (L.Q.); (Q.M.)
- Key Laboratory for Genetic Breeding of Aquatic Animals and Aquaculture Biology, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (S.L.); (W.J.); (Y.L.); (Z.G.)
| |
Collapse
|
2
|
Vieiros M, Navarro-Tapia E, Ramos-Triguero A, García-Meseguer À, Martínez L, García-Algar Ó, Andreu-Fernández V. Analysis of alcohol-metabolizing enzymes genetic variants and RAR/RXR expression in patients diagnosed with fetal alcohol syndrome: a case-control study. BMC Genomics 2024; 25:610. [PMID: 38886650 PMCID: PMC11184718 DOI: 10.1186/s12864-024-10516-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 06/11/2024] [Indexed: 06/20/2024] Open
Abstract
Understanding the mechanisms underlying alcohol metabolism and its regulation, including the effect of polymorphisms in alcohol-metabolizing enzymes, is crucial for research on Fetal Alcohol Spectrum Disorders. The aim of this study was to identify specific single nucleotide polymorphisms in key alcohol-metabolizing enzymes in a cohort of 71 children, including children with fetal alcohol syndrome, children prenatally exposed to ethanol but without fetal alcohol spectrum disorder, and controls. We hypothesized that certain genetic variants related to alcohol metabolism may be fixed in these populations, giving them a particular alcohol metabolism profile. In addition, the difference in certain isoforms of these enzymes determines their affinity for alcohol, which also affects the metabolism of retinoic acid, which is key to the proper development of the central nervous system. Our results showed that children prenatally exposed to ethanol without fetal alcohol spectrum disorder traits had a higher frequency of the ADH1B*3 and ADH1C*1 alleles, which are associated with increased alcohol metabolism and therefore a protective factor against circulating alcohol in the fetus after maternal drinking, compared to FAS children who had an allele with a lower affinity for alcohol. This study also revealed the presence of an ADH4 variant in the FAS population that binds weakly to the teratogen, allowing increased circulation of the toxic agent and direct induction of developmental abnormalities in the fetus. However, both groups showed dysregulation in the expression of genes related to the retinoic acid pathway, such as retinoic acid receptor and retinoid X receptor, which are involved in the development, regeneration, and maintenance of the nervous system. These findings highlight the importance of understanding the interplay between alcohol metabolism, the retinoic acid pathway and genetic factors in the development of fetal alcohol syndrome.
Collapse
Affiliation(s)
- Melina Vieiros
- Grup de Recerca Infància i Entorn (GRIE), Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- IdiPAZ - Instituto de Investigación Hospital Universitario La Paz, Madrid, Spain
- Department de Cirurgia i Especialitats Mèdico-Quirúrgiques, Universitat de Barcelona, Barcelona, Spain
| | - Elisabet Navarro-Tapia
- IdiPAZ - Instituto de Investigación Hospital Universitario La Paz, Madrid, Spain.
- Faculty of Health Sciences, Valencian International University, Valencia, Spain.
| | - Anna Ramos-Triguero
- Grup de Recerca Infància i Entorn (GRIE), Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Àgueda García-Meseguer
- Grup de Recerca Infància i Entorn (GRIE), Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Leopoldo Martínez
- IdiPAZ - Instituto de Investigación Hospital Universitario La Paz, Madrid, Spain
- Department of Pediatric Surgery, Hospital Universitario La Paz, Madrid, Spain
| | - Óscar García-Algar
- Grup de Recerca Infància i Entorn (GRIE), Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Department of Neonatology, Hospital Clínic-Maternitat, ICGON, BCNatal, Barcelona, Spain
| | - Vicente Andreu-Fernández
- Grup de Recerca Infància i Entorn (GRIE), Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
- Biosanitary Research Institute, Valencian International University, Valencia, Spain.
| |
Collapse
|
3
|
Bao Y, Rong W, Zhu A, Chen Y, Chen H, Hong Y, Le J, Wang Q, Naman CB, Xu Z, Liu L, Cui W, Wu X. Retinoic Acid Receptor Is a Novel Therapeutic Target for Postoperative Cognitive Dysfunction. Pharmaceutics 2023; 15:2311. [PMID: 37765280 PMCID: PMC10538227 DOI: 10.3390/pharmaceutics15092311] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 09/02/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Postoperative cognitive dysfunction (POCD) is a clinical syndrome characterizing by cognitive impairments in the elderly after surgery. There is limited effective treatment available or clear pathological mechanisms known for this syndrome. In this study, a Connectivity Map (CMap) bioinformatics model of POCD was established by using differently expressed landmark genes in the serum samples of POCD and non-POCD patients from the only human transcriptome study. The predictability and reliability of this model were further supported by the positive CMap scores of known POCD inducers and the negative CMap scores of anti-POCD drug candidates. Most retinoic acid receptor (RAR) agonists were negatively associated with POCD in this CMap model, suggesting that RAR might be a novel target for POCD. Most importantly, acitretin, a clinically used RAR agonist, significantly inhibited surgery-induced cognitive impairments and prevented the reduction in RARα and RARα-target genes in the hippocampal regions of aged mice. The study denotes a reliable CMap bioinformatics model of POCD for future use and establishes that RAR is a novel therapeutic target for treating this clinical syndrome.
Collapse
Affiliation(s)
- Yongjie Bao
- Department of Anesthesiology, The First Affiliated Hospital of Ningbo University, Ningbo 315010, China; (Y.B.)
- Translational Medicine Center of Pain, Emotion and Cognition, Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, China
| | - Wenni Rong
- Translational Medicine Center of Pain, Emotion and Cognition, Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, China
| | - An Zhu
- Translational Medicine Center of Pain, Emotion and Cognition, Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, China
| | - Yuan Chen
- Translational Medicine Center of Pain, Emotion and Cognition, Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, China
| | - Huiyue Chen
- Translational Medicine Center of Pain, Emotion and Cognition, Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, China
| | - Yirui Hong
- Translational Medicine Center of Pain, Emotion and Cognition, Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, China
| | - Jingyang Le
- Department of Anesthesiology, The First Affiliated Hospital of Ningbo University, Ningbo 315010, China; (Y.B.)
- Translational Medicine Center of Pain, Emotion and Cognition, Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, China
| | - Qiyao Wang
- Department of Anesthesiology, The First Affiliated Hospital of Ningbo University, Ningbo 315010, China; (Y.B.)
- Translational Medicine Center of Pain, Emotion and Cognition, Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, China
| | - C. Benjamin Naman
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, Ningbo University, Ningbo 315211, China
| | - Zhipeng Xu
- Department of Anesthesiology, The First Affiliated Hospital of Ningbo University, Ningbo 315010, China; (Y.B.)
| | - Lin Liu
- Department of Anesthesiology, The First Affiliated Hospital of Ningbo University, Ningbo 315010, China; (Y.B.)
| | - Wei Cui
- Department of Anesthesiology, The First Affiliated Hospital of Ningbo University, Ningbo 315010, China; (Y.B.)
- Translational Medicine Center of Pain, Emotion and Cognition, Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, China
| | - Xiang Wu
- Department of Anesthesiology, The First Affiliated Hospital of Ningbo University, Ningbo 315010, China; (Y.B.)
| |
Collapse
|
4
|
Han L, Huang D, Wu S, Liu S, Wang C, Sheng Y, Lu X, Broxmeyer HE, Wan J, Yang L. Lipid droplet-associated lncRNA LIPTER preserves cardiac lipid metabolism. Nat Cell Biol 2023; 25:1033-1046. [PMID: 37264180 PMCID: PMC10344779 DOI: 10.1038/s41556-023-01162-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 05/03/2023] [Indexed: 06/03/2023]
Abstract
Lipid droplets (LDs) are cellular organelles critical for lipid homeostasis, with intramyocyte LD accumulation implicated in metabolic disorder-associated heart diseases. Here we identify a human long non-coding RNA, Lipid-Droplet Transporter (LIPTER), essential for LD transport in human cardiomyocytes. LIPTER binds phosphatidic acid and phosphatidylinositol 4-phosphate on LD surface membranes and the MYH10 protein, connecting LDs to the MYH10-ACTIN cytoskeleton and facilitating LD transport. LIPTER and MYH10 deficiencies impair LD trafficking, mitochondrial function and survival of human induced pluripotent stem cell-derived cardiomyocytes. Conditional Myh10 deletion in mouse cardiomyocytes leads to LD accumulation, reduced fatty acid oxidation and compromised cardiac function. We identify NKX2.5 as the primary regulator of cardiomyocyte-specific LIPTER transcription. Notably, LIPTER transgenic expression mitigates cardiac lipotoxicity, preserves cardiac function and alleviates cardiomyopathies in high-fat-diet-fed and Leprdb/db mice. Our findings unveil a molecular connector role of LIPTER in intramyocyte LD transport, crucial for lipid metabolism of the human heart, and hold significant clinical implications for treating metabolic syndrome-associated heart diseases.
Collapse
Affiliation(s)
- Lei Han
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Dayang Huang
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Shiyong Wu
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sheng Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Cheng Wang
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yi Sheng
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xiongbin Lu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Hal E Broxmeyer
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jun Wan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Lei Yang
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
5
|
Ma L, Shao M, Cheng W, Jiang J, Chen X, Tan N, Ling G, Yang Y, Wang Q, Yang R, Li C, Wang Y. Neocryptotanshinone ameliorates insufficient energy production in heart failure by targeting retinoid X receptor alpha. Biomed Pharmacother 2023; 163:114868. [PMID: 37201263 DOI: 10.1016/j.biopha.2023.114868] [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: 03/31/2023] [Revised: 05/08/2023] [Accepted: 05/10/2023] [Indexed: 05/20/2023] Open
Abstract
Retinoid X receptor alpha (RXRα) is a nuclear transcription factor that extensively regulates energy metabolism in cardiovascular diseases. Identification of targeted RXRα drugs for heart failure (HF) therapy is urgently needed. Neocryptotanshinone (NCTS) is a component derived from Salvia miltiorrhiza Bunge, the effect and mechanism of which for treating HF have not been reported. The goal of this study was to explore the pharmacological effects of NCTS on energy metabolism to protect against HF post-acute myocardial infarction (AMI) via RXRα. We established a left anterior descending artery ligation-induced HF post-AMI model in mice and an oxygen-glucose deprivation-reperfusion-induced H9c2 cell model to investigate the cardioprotective effect of NCTS. Component-target binding techniques, surface plasmon resonance (SPR), microscale thermophoresis (MST) and small interfering RNA (siRNA) transfection were applied to explore the potential mechanism by which NCTS targets RXRα. The results showed that NCTS protects the heart against ischaemic damage, evidenced by improvement of cardiac dysfunction and attenuation of cellular hypoxic injury. Importantly, the SPR and MST results showed that NCTS has a high binding affinity for RXRα. Meanwhile, the critical downstream target genes of RXRα/PPARα, which are involved in fatty acid metabolism, including Cd36 and Cpt1a, were upregulated under NCTS treatment. Moreover, NCTS enhanced TFAM levels, promoted mitochondrial biogenesis and increased myocardial adenosine triphosphate levels by activating RXRα. In conclusion, we confirmed that NCTS improves myocardial energy metabolism, including fatty acid oxidation and mitochondrial biogenesis, by regulating the RXRα/PPARα pathway in mice with HF post-AMI.
Collapse
Affiliation(s)
- Lin Ma
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Mingyan Shao
- School of Life Science, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Wenkun Cheng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jinchi Jiang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xu Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Nannan Tan
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Guanjing Ling
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Ye Yang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Qian Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Ran Yang
- Guang'anmen Hospital China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Chun Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Yong Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; School of Life Science, Beijing University of Chinese Medicine, Beijing 100029, China; Beijing Key Laboratory of TCM Syndrome and Formula, Beijing 100029, China; Key Laboratory of Beijing University of Chinese Medicine, Ministry of Education, Beijing 100029, China.
| |
Collapse
|
6
|
The RIG-I-NRF2 axis regulates the mesenchymal stromal niche for bone marrow transplantation. Blood 2022; 139:3204-3221. [PMID: 35259210 DOI: 10.1182/blood.2021013048] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 02/18/2022] [Indexed: 11/20/2022] Open
Abstract
Bone marrow-derived mesenchymal stem cells (BMSCs) support bone formation and constitute the stromal niche in regulating hematopoietic stem cells (HSCs). Stromal niche dysfunction affects HSC engraftment during transplantation; however, the underlying mechanisms remain elusive. In the present study, we found that all-trans retinoic acid (ATRA) and inflammation stress upregulated retinoic acid-inducible gene I (RIG-I) in BMSCs. Excess RIG-I expression damaged the clonogenicity, bone-forming ability of BMSCs and, particularly, their stromal niche function that supports HSC expansion in vitro and engraftment in vivo. Mechanistically, RIG-I elevation promoted the degradation of NRF2, a checkpoint for antioxidant cellular response, by altering the RIG-I-Trim25-Keap1-NRF2 complex, leading to reactive oxygen species (ROS) accumulation and BMSC damage. Genetic inhibition of RIG-I sustained NRF2 protein levels and reduced ROS levels in ATRA-treated BMSCs, thus preserving their clonogenicity, bone-forming ability, and stromal niche function in supporting HSC engraftment in mice. More importantly, RIG-I inhibition recovered the ATRA-treated stromal niche function, to enhance HSC engraftment and emergency myelopoiesis for innate immunity against the bacterium Listeria monocytogenes during transplantation. Overall, we identified a non-canonical role of RIG-I in the regulation of the stromal niche for HSC transplantation.
Collapse
|
7
|
Paredes A, Santos-Clemente R, Ricote M. Untangling the Cooperative Role of Nuclear Receptors in Cardiovascular Physiology and Disease. Int J Mol Sci 2021; 22:ijms22157775. [PMID: 34360540 PMCID: PMC8346021 DOI: 10.3390/ijms22157775] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/13/2021] [Accepted: 07/16/2021] [Indexed: 12/12/2022] Open
Abstract
The heart is the first organ to acquire its physiological function during development, enabling it to supply the organism with oxygen and nutrients. Given this early commitment, cardiomyocytes were traditionally considered transcriptionally stable cells fully committed to contractile function. However, growing evidence suggests that the maintenance of cardiac function in health and disease depends on transcriptional and epigenetic regulation. Several studies have revealed that the complex transcriptional alterations underlying cardiovascular disease (CVD) manifestations such as myocardial infarction and hypertrophy is mediated by cardiac retinoid X receptors (RXR) and their partners. RXRs are members of the nuclear receptor (NR) superfamily of ligand-activated transcription factors and drive essential biological processes such as ion handling, mitochondrial biogenesis, and glucose and lipid metabolism. RXRs are thus attractive molecular targets for the development of effective pharmacological strategies for CVD treatment and prevention. In this review, we summarize current knowledge of RXR partnership biology in cardiac homeostasis and disease, providing an up-to-date view of the molecular mechanisms and cellular pathways that sustain cardiomyocyte physiology.
Collapse
|
8
|
Exploring the Antihyperglycemic Chemical Composition and Mechanisms of Tea Using Molecular Docking. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:8871088. [PMID: 33343682 PMCID: PMC7725569 DOI: 10.1155/2020/8871088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 09/11/2020] [Accepted: 10/19/2020] [Indexed: 11/18/2022]
Abstract
Tea, a widely consumed beverage, has long been utilized for promoting human health with a close correlation to hyperglycemia. The Tea Metabolome Database (TMDB), the most complete and comprehensive curated collection of tea compounds data containing 1271 identified small molecule compounds from the tea plant (Camellia sinensis), was established previously by our research team. More recently, our studies have found that various tea types possess an antihyperglycemic effect in mice. However, the bioactive ingredients from tea have potential antihyperglycemic activity and their underlying molecular mechanisms remain unclear. In this study, we used a molecular docking approach to investigate the potential interactions between a selected 747 constituents contained in tea and 11 key protein targets of clinical antihyperglycemic drugs. According to our results, the main antihyperglycemic targets of tea composition were consistent with those of the drug rosiglitazone. The screening results showed that GCG, ECG3'Me, TMDB-01443, and CG had great target binding capacity. The results indicated that these chemicals of tea might affect hyperglycemia by acting on protein targets of rosiglitazone.
Collapse
|
9
|
Chai Q, Miao J, Liu M, Zhang Z, Meng Z, Wu W. Knockdown of SGLT1 prevents the apoptosis of cardiomyocytes induced by glucose fluctuation via relieving oxidative stress and mitochondrial dysfunction. Biochem Cell Biol 2020; 99:356-363. [PMID: 33259229 DOI: 10.1139/bcb-2020-0491] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Fluctuations in the concentration of glucose in the blood is more detrimental than a constantly high level of glucose with respect to the development of cardiovascular complications associated with diabetes mellitus (DM). Sodium glucose cotransporter 2 (SGLT2) inhibitors have been developed as antidiabetic drugs with cardiovascular benefits; however, whether inhibition of SGLT1 protects the diabetic heart remains to be determined. This study investigated the role of SGLT1 in rat H9c2 cardiomyocytes subjected to fluctuating levels of glucose and the underlying mechanisms. The results indicated that knockdown of SGLT1 restored cell proliferation and suppressed the cytotoxicity associated with fluctuating glucose levels. Oxidative stress was induced in H9c2 cells subjected to fluctuating glucose levels, but these changes were effectively reversed by knockdown of SGLT1, as manifested by reductions in the level of intracellular reactive oxygen species and increased antioxidant activity. Further study demonstrated that knockdown of SGLT1 attenuated the mitochondrial dysfunction in H9c2 cells exposed to fluctuating glucose levels, by restoring mitochondrial membrane potential and promoting mitochondrial fusion. In addition, knockdown of SGLT1 downregulated the expression of Bax, upregulated the expression of Bcl-2, and reduced the activation of caspase-3 in H9c2 cells subjected to fluctuating levels of glucose. Collectively, our results show that knockdown of SGLT1 ameliorates the apoptosis of cardiomyocyte caused by fluctuating glucose levels via regulating oxidative stress and combatting mitochondrial dysfunction.
Collapse
Affiliation(s)
- Qian Chai
- Department of Endocrinology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, P.R. China
| | - Jiajing Miao
- Department of Endocrinology, Affiliated Hospital of Jilin Medical College, Jilin 132013, P.R. China
| | - Meili Liu
- Department of Endocrinology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, P.R. China
| | - Ziying Zhang
- Department of Endocrinology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, P.R. China
| | - Ziang Meng
- Department of Urology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin 150001, P.R. China
| | - Weihua Wu
- Department of Endocrinology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, P.R. China
| |
Collapse
|
10
|
Retinoid X receptor agonists attenuates cardiomyopathy in streptozotocin-induced type 1 diabetes through LKB1-dependent anti-fibrosis effects. Clin Sci (Lond) 2020; 134:609-628. [PMID: 32175563 DOI: 10.1042/cs20190985] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 02/27/2020] [Accepted: 03/16/2020] [Indexed: 12/15/2022]
Abstract
Diabetic cardiac fibrosis increases ventricular stiffness and facilitates the occurrence of diastolic dysfunction. Retinoid X receptor (RXR) plays an important role in cardiac development and has been implicated in cardiovascular diseases. In the present study, we investigated the effects of RXR agonist treatment on streptozotocin (STZ)-induced diabetic cardiomyopathy (DCM) and the underlying mechanism. Sprague-Dawley (SD) rats induced by STZ injection were treated with either RXR agonist bexarotene (Bex) or vehicle alone. Echocardiography was performed to determine cardiac structure and function. Cardiac fibroblasts (CFs) were treated with high glucose (HG) with or without the indicated concentration of Bex or the RXR ligand 9-cis-retinoic acid (9-cis-RA). The protein abundance levels were measured along with collagen, body weight (BW), blood biochemical indexes and transforming growth factor-β (TGF-β) levels. The effects of RXRα down-regulation by RXRα small interfering RNA (siRNA) were examined. The results showed that bexarotene treatment resulted in amelioration of left ventricular dysfunction by inhibiting cardiomyocyte apoptosis and myocardial fibrosis. Immunoblot with heart tissue homogenates from diabetic rats revealed that bexarotene activated liver kinase B1 (LKB1) signaling and inhibited p70 ribosomal protein S6 kinase (p70S6K). The increased collagen levels in the heart tissues of DCM rats were reduced by bexarotene treatment. Treatment of CFs with HG resulted in significantly reduced LKB1 activity and increased p70S6K activity. RXRα mediated the antagonism of 9-cis-RA on HG-induced LKB1/p70S6K activation changes in vitro. Our findings suggest that RXR agonist ameliorates STZ-induced DCM by inhibiting myocardial fibrosis via modulation of the LKB1/p70S6K signaling pathway. RXR agonists may serve as novel therapeutic agents for the treatment of DCM.
Collapse
|
11
|
Chen C, Ke L, Chan H, Chu C, Lee A, Lin K, Lee M, Hsiao P, Chen C, Shin S. Electronegative low-density lipoprotein of patients with metabolic syndrome induces pathogenesis of aorta through disruption of the stimulated by retinoic acid 6 cascade. J Diabetes Investig 2020; 11:535-544. [PMID: 31597015 PMCID: PMC7232312 DOI: 10.1111/jdi.13158] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 10/01/2019] [Accepted: 10/06/2019] [Indexed: 12/26/2022] Open
Abstract
AIMS/INTRODUCTION Electronegative low-density lipoprotein (L5) is the most atherogenic fraction of low-density lipoprotein and is elevated in people with metabolic syndrome (MetS), whereas the retinol-binding protein 4 receptor (stimulated by retinoic acid 6 [STRA6]) cascade is disrupted in various organs of patients with obesity-related diseases. Our objective was to investigate whether L5 from MetS patients capably induces pathogenesis of aorta through disrupting the STRA6 cascade. MATERIAL AND METHODS We examined the in vivo and in vitro effects of L5 on the STRA6 cascade and aortic atherogenic markers. To investigate the role of this cascade on atherosclerotic formation, crbp1 transfection was carried out in vitro. RESULTS This study shows that L5 activates atherogenic markers (p38 mitogen-activated protein kinases, pSmad2 and matrix metallopeptidase 9) and simultaneously suppresses STRA6 signals (STRA6, cellular retinol-binding protein 1, lecithin-retinol acyltransferase, retinoic acid receptor-α and retinoid X receptor-α) in aortas of L5-injected mice and L5-treated human aortic endothelial cell lines and human aortic smooth muscle cell lines. These L5-induced changes of the STRA6 cascade and atherogenic markers were reversed in aortas of LOX1-/- mice and in LOX1 ribonucleic acid-silenced human aortic endothelial cell lines and human aortic smooth muscle cell lines. Furthermore, crbp1 gene transfection reversed the disruption of the STRA6 cascade, the phosphorylation of p38 mitogen-activated protein kinases and Smad2, and the elevation of matrix metallopeptidase 9 in L5-treated human aortic endothelial cell lines. CONCLUSIONS This study shows that L5 from MetS patients induces atherogenic markers by disrupting STRA6 signaling. Suppression of STRA6 might be one novel pathogenesis of aorta in patients with MetS.
Collapse
Affiliation(s)
- Chao‐Hung Chen
- School of MedicineCollege of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
- Divison of Endocrinology and MetabolismDepartment of Internal MedicineKaohsiung Medical University HospitalKaohsiungTaiwan
| | - Liang‐Yin Ke
- Lipid Science and Aging Research CenterKaohsiung Medical UniversityKaohsiungTaiwan
- Department of Medical Laboratory Science and BiotechnologyCollege of Health SciencesKaohsiung Medical UniversityKaohsiungTaiwan
| | - Hua‐Chen Chan
- Department of Medical Laboratory Science and BiotechnologyCollege of Health SciencesKaohsiung Medical UniversityKaohsiungTaiwan
| | - Chih‐Sheng Chu
- Division of CardiologyDepartment of Internal MedicineKaohsiung Medical University HospitalKaohsiungTaiwan
| | - An‐Sheng Lee
- Department of MedicineMackay Medical CollegeNew TaipeiTaiwan
| | - Kun‐Der Lin
- School of MedicineCollege of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
- Divison of Endocrinology and MetabolismDepartment of Internal MedicineKaohsiung Medical University HospitalKaohsiungTaiwan
- Vascular and Medical ResearchTexas Heart InstituteHoustonTexasUSA
| | - Mei‐Yueh Lee
- School of MedicineCollege of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
- Divison of Endocrinology and MetabolismDepartment of Internal MedicineKaohsiung Medical University HospitalKaohsiungTaiwan
| | - Pi‐Jung Hsiao
- School of MedicineCollege of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
- Divison of Endocrinology and MetabolismDepartment of Internal MedicineKaohsiung Medical University HospitalKaohsiungTaiwan
| | - Chu‐Huang Chen
- Lipid Science and Aging Research CenterKaohsiung Medical UniversityKaohsiungTaiwan
- Vascular and Medical ResearchTexas Heart InstituteHoustonTexasUSA
- Department of Internal MedicineKaohsiung Ta‐Tung Municipal HospitalKaohsiung Medical University HospitalKaohsiungTaiwan
| | - Shyi‐Jang Shin
- School of MedicineCollege of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
- Divison of Endocrinology and MetabolismDepartment of Internal MedicineKaohsiung Medical University HospitalKaohsiungTaiwan
- Department of Medical Laboratory Science and BiotechnologyCollege of Health SciencesKaohsiung Medical UniversityKaohsiungTaiwan
| |
Collapse
|
12
|
All-Trans Retinoic Acid Attenuates Fibrotic Processes by Downregulating TGF-β1/Smad3 in Early Diabetic Nephropathy. Biomolecules 2019; 9:biom9100525. [PMID: 31557800 PMCID: PMC6843855 DOI: 10.3390/biom9100525] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 09/04/2019] [Accepted: 09/18/2019] [Indexed: 02/07/2023] Open
Abstract
Diabetic nephropathy (DN) involves damage associated to hyperglycemia and oxidative stress. Renal fibrosis is a major pathologic feature of DN. The aim of this study was to evaluate anti-fibrogenic and renoprotective effects of all-trans retinoic acid (ATRA) in isolated glomeruli and proximal tubules of diabetic rats. Diabetes was induced by single injection of streptozotocin (STZ, 60 mg/Kg). ATRA (1 mg/Kg) was administered daily by gavage, from days 3–21 after STZ injection. ATRA attenuated kidney injury through the reduction of proteinuria, renal hypertrophy, increase in natriuresis, as well as early markers of damage such as β2-microglobulin, kidney injury molecule-1 (KIM-1), and neutrophil gelatinase-associated lipocalin (NGAL). The following parameters increased: macrophage infiltration, localization of alpha-smooth muscle actin (αSMA)-positive cells in renal tissue, and pro-fibrotic proteins such as transforming growth factor-β (TGF-β1), laminin beta 1 (LAM-β1), and collagens IV and I. Remarkably, ATRA treatment ameliorated these alterations and attenuated expression and nuclear translocation of Smad3, with increment of glomerular and tubular Smad7. The diabetic condition decreased expression of retinoic acid receptor alpha (RAR-α) through phosphorylation in serine residues mediated by the activation of c-Jun N-terminal kinase (JNK). ATRA administration restored the expression of RAR-α and inhibited direct interactions of JNK/RAR-α. ATRA prevented fibrogenesis through down-regulation of TGF-β1/Smad3 signaling.
Collapse
|
13
|
Chen CH, Lin KD, Ke LY, Liang CJ, Kuo WC, Lee MY, Lee YL, Hsiao PJ, Hsu CC, Shin SJ. O-GlcNAcylation disrupts STRA6-retinol signals in kidneys of diabetes. Biochim Biophys Acta Gen Subj 2019; 1863:1059-1069. [DOI: 10.1016/j.bbagen.2019.03.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 03/19/2019] [Accepted: 03/20/2019] [Indexed: 12/31/2022]
|
14
|
El-Baz FK, Hussein RA, Saleh DO, Abdel Jaleel GAR. Zeaxanthin Isolated from Dunaliella salina Microalgae Ameliorates Age Associated Cardiac Dysfunction in Rats through Stimulation of Retinoid Receptors. Mar Drugs 2019; 17:md17050290. [PMID: 31091726 PMCID: PMC6562725 DOI: 10.3390/md17050290] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 04/30/2019] [Accepted: 05/04/2019] [Indexed: 12/11/2022] Open
Abstract
Retinoids are essential during early cardiovascular morphogenesis. However, recent studies showed their important role in cardiac remodeling in rats with hypertension and following myocardial infarction. The present study aimed to investigate the effect of zeaxanthin heneicosylate (ZH); a carotenoid ester isolated from Dunaliella salina microalgae, on cardiac dysfunction ensuing d-galactose injection in rats. Rats injected with d-GAL (200 mg/kg; I.P) for 8 weeks were orally treated with ZH (250 μg/kg) for 28 consecutive days. Results showed that d-GAL injection caused dramatic electrocardiographic changes as well as marked elevation in serum levels of homocysteine, creatinine kinase isoenzyme and lactate dehydrogenase. A reduction in the cardiac contents of glucose transporter-4 and superoxide dismutase along with the elevation of inducible nitric oxide synthetase and interleukin-6 was also noticed. Oral administration of ZH significantly improved the above mentioned cardiac aging manifestations; this was further emphasized through histopathological examinations. The effect of ZH is mediated through the interaction with retinoid receptor alpha (RAR-α) as evidenced through a significant elevation of RAR-α expression in cardiac tissue following the lead of an in silico molecular docking study. In conclusion, zeaxanthin heneicosylate isolated from D. salina ameliorated age-associated cardiac dysfunction in rats through the activation of retinoid receptors.
Collapse
Affiliation(s)
- Farouk Kamel El-Baz
- Plant Biochemistry Department, National Research Centre (NRC), 33 El Buhouth St. (Former El Tahrir St.), Dokki, Giza P.O.12622, Egypt.
| | - Rehab Ali Hussein
- Pharmacognosy Department, National Research Centre (NRC), 33 El Buhouth St. (Former El Tahrir St.), Dokki, Giza P.O.12622, Egypt.
| | - Dalia Osama Saleh
- Pharmacology Department, National Research Centre (NRC), 33 El Buhouth St. (Former El Tahrir St.), Dokki, Giza P.O.12622, Egypt.
| | - Gehad Abdel Raheem Abdel Jaleel
- Pharmacology Department, National Research Centre (NRC), 33 El Buhouth St. (Former El Tahrir St.), Dokki, Giza P.O.12622, Egypt.
| |
Collapse
|
15
|
Matsuoka S, Bariuan JV, Nakagiri S, Abd Eldaim MA, Okamatsu-Ogura Y, Kimura K. Linking pathways and processes: Retinoic acid and glucose. MOLECULAR NUTRITION: CARBOHYDRATES 2019:247-264. [DOI: 10.1016/b978-0-12-849886-6.00013-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
|
16
|
Mukai Y, Yamanishi R. Retinol but not retinoic acid can enhance the glutathione level, in a manner similar to β-carotene, in a murine cultured macrophage cell line. Food Sci Nutr 2018; 6:1650-1656. [PMID: 30258608 PMCID: PMC6145222 DOI: 10.1002/fsn3.726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 03/01/2018] [Accepted: 06/08/2018] [Indexed: 11/10/2022] Open
Abstract
SCOPE We evaluated the potential of retinol and retinoic acid (RA) to enhance intracellular glutathione (GSH) levels in a murine cultured macrophage cell line, RAW264, to investigate whether the RA signaling pathway is involved in the β-carotene-induced GSH enhancement. METHODS AND RESULTS We examined GSH levels in RAW264 cells cultured in media supplemented with β-carotene and various inhibitors (ER50891 for RA receptor (RAR)α, CD2665 for RARβ/γ, or HX531 for all subtypes of retinoid X receptor (RXR)), to verify each inhibitor's activity against β-carotene, as well as in media supplemented with various stimulants (AM80 for RARα, CD2314 for RARβ, CD437 for RARγ, or SR11237 for RXR), to compare their activity with that of β-carotene. We also examined the GSH level and glutamate-cysteine-ligase (GCL) expression in RAW264 cells cultured in all-trans RA- or retinol-supplemented media. Enhanced GSH production was not inhibited by any tested antagonist, and, apart from β-carotene, no agonist induced GSH production. Retinol, but not all-trans RA, enhanced GSH synthesis and increased GCL expression, similar to that observed with β-carotene. CONCLUSION The RA signaling pathway may not be involved in the β-carotene-induced enhancement of GSH levels in RAW264 cells, whereas, like β-carotene, retinol can enhance the GSH level and GCL expression.
Collapse
Affiliation(s)
- Yuuka Mukai
- Department of Food Hygiene and FunctionSchool of Nutrition and DieteticsFaculty of Health and Social WorkKanagawa University of Human ServicesKanagawaJapan
| | - Rintaro Yamanishi
- Department of Food Science and NutritionSchool of Nutrition and DieteticsFaculty of Health and Social WorkKanagawa University of Human ServicesKanagawaJapan
| |
Collapse
|
17
|
Zhou H, Sun Y, Zhang L, Kang W, Li N, Li Y. The RhoA/ROCK pathway mediates high glucose-induced cardiomyocyte apoptosis via oxidative stress, JNK, and p38MAPK pathways. Diabetes Metab Res Rev 2018; 34:e3022. [PMID: 29745021 DOI: 10.1002/dmrr.3022] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 04/09/2018] [Accepted: 04/20/2018] [Indexed: 12/21/2022]
Abstract
AIMS To understand the roles of the RhoA/ROCK and mitogen-activated protein kinase (MAPK) pathways in high glucose (HG)-induced apoptosis and oxidative stress in cardiomyocytes. MATERIALS AND METHODS Neonatal rat cardiomyocytes were cultured in Dulbecco's modified Eagle's medium, supplemented with 5.5 or 30 mmol/L D-glucose, in the presence or absence of fasudil (50 or 100 μM), SB203580, SP600125, or PD98059 (10 μM, respectively). The percentage of early apoptotic cardiomyocytes was evaluated using flow cytometry. The superoxide dismutase activity and malondialdehyde contents in the cellular supernatants were measured. The Bax and Bcl-2 mRNA levels were determined by quantitative real-time PCR. Phosphorylation of myosin phosphatase target subunit 1 (MYPT1), p38MAPK, JNK, and ERK as well as the protein levels of Bax, Bcl-2, and cleaved caspase-3 was analysed by Western blot. RESULTS Fasudil, SB203580, and SP600125 effectively inhibited the HG-induced early apoptosis increase and decreased Bax mRNA expression, the Bax/Bcl-2 protein expression ratio, and cleaved caspase-3 protein levels in the cardiomyocytes; this was accompanied by upregulation of the Bcl-2 mRNA. Moreover, fasudil markedly increased the superoxide dismutase activity level and suppressed the elevation in HG-induced malondialdehyde content and the phosphorylation of MYPT1, p38MAPK and JNK. CONCLUSIONS The RhoA/ROCK pathway mediates HG-induced cardiomyocyte apoptosis via oxidative stress and activation of p38MAPK and JNK in neonatal rats in vitro. Fasudil effectively ameliorates HG-induced cardiomyocyte apoptosis by suppressing oxidative stress and the p38MAPK and JNK pathways.
Collapse
Affiliation(s)
- Hong Zhou
- Department of Endocrinology, the Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yonghong Sun
- Nutriology, the Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lihui Zhang
- Department of Endocrinology, the Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Wenyuan Kang
- Department of Endocrinology, the Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Na Li
- Department of Endocrinology, the Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yongjun Li
- Cardiology, the Second Hospital of Hebei Medical University, Shijiazhuang, China
- Hebei Institute of Cardiovascular and Cerebrovascular Diseases, Shijiazhuang, China
| |
Collapse
|
18
|
Dott W, Wright J, Cain K, Mistry P, Herbert KE. Integrated metabolic models for xenobiotic induced mitochondrial toxicity in skeletal muscle. Redox Biol 2018; 14:198-210. [PMID: 28942197 PMCID: PMC5610037 DOI: 10.1016/j.redox.2017.09.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 09/11/2017] [Accepted: 09/13/2017] [Indexed: 11/16/2022] Open
Abstract
There is a need for robust in vitro models to sensitively capture skeletal muscle adverse toxicities early in the research and development of novel xenobiotics. To this end, an in vitro rat skeletal muscle model (L6) was used to study the translation of transcriptomics data generated from an in vivo rat model. Novel sulfonyl isoxazoline herbicides were associated with skeletal muscle toxicity in an in vivo rat model. Gene expression pathway analysis on skeletal muscle tissues taken from in vivo repeat dose studies identified enriched pathways associated with mitochondrial dysfunction, oxidative stress, energy metabolism, protein regulation and cell cycle. Mitochondrial dysfunction and oxidative stress were further explored using in vitro L6 metabolic models. These models demonstrated that the sulfonyl isoxazoline compounds induced mitochondrial dysfunction, mitochondrial superoxide production and apoptosis. These in vitro findings accurately concurred with the in vivo transcriptomics data, thereby confirming the ability of the L6 skeletal muscle models to identify relevant in vivo mechanisms of xenobiotic-induced toxicity. Moreover, these results highlight the sensitivity of the L6 galactose media model to study mitochondrial perturbation associated with skeletal muscle toxicity; this model may be utilised to rank the potency of novel xenobiotics upon further validation.
Collapse
Affiliation(s)
- William Dott
- Department of Cardiovascular Sciences, University of Leicester, UK
| | | | - Kelvin Cain
- MRC Toxicology Unit, University of Leicester, Leicester, UK
| | | | - Karl E Herbert
- Department of Cardiovascular Sciences, University of Leicester, UK.
| |
Collapse
|
19
|
Qiu F, Li Y, Lu X, Xie C, Nong Q, Wu D, Chen J, Yang L, Zhou Y, Lu J. Rare variant ofMAP2K7is associated with increased risk of COPD in southern and eastern Chinese. Respirology 2017; 22:691-698. [DOI: 10.1111/resp.12976] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 09/11/2016] [Accepted: 09/29/2016] [Indexed: 12/25/2022]
Affiliation(s)
- Fuman Qiu
- Biomedicine Research Center of The Third Affiliated Hospital of GMU, The State Key Lab of Respiratory Disease, The Institute for Chemical Carcinogenesis, Collaborative Innovation Center for Environmental Toxicity; Guangzhou Medical University; Guangzhou China
| | - Yinyan Li
- Biomedicine Research Center of The Third Affiliated Hospital of GMU, The State Key Lab of Respiratory Disease, The Institute for Chemical Carcinogenesis, Collaborative Innovation Center for Environmental Toxicity; Guangzhou Medical University; Guangzhou China
| | - Xiaoxiao Lu
- School of Arts and Sciences; Colby-Sawyer College; New London New Hampshire USA
| | - Chenli Xie
- Biomedicine Research Center of The Third Affiliated Hospital of GMU, The State Key Lab of Respiratory Disease, The Institute for Chemical Carcinogenesis, Collaborative Innovation Center for Environmental Toxicity; Guangzhou Medical University; Guangzhou China
| | - Qingqing Nong
- Department of Environmental Health; Guangxi Medical University; Nanning China
| | - Di Wu
- Biomedicine Research Center of The Third Affiliated Hospital of GMU, The State Key Lab of Respiratory Disease, The Institute for Chemical Carcinogenesis, Collaborative Innovation Center for Environmental Toxicity; Guangzhou Medical University; Guangzhou China
| | - Jiansong Chen
- Biomedicine Research Center of The Third Affiliated Hospital of GMU, The State Key Lab of Respiratory Disease, The Institute for Chemical Carcinogenesis, Collaborative Innovation Center for Environmental Toxicity; Guangzhou Medical University; Guangzhou China
| | - Lei Yang
- Biomedicine Research Center of The Third Affiliated Hospital of GMU, The State Key Lab of Respiratory Disease, The Institute for Chemical Carcinogenesis, Collaborative Innovation Center for Environmental Toxicity; Guangzhou Medical University; Guangzhou China
| | - Yifeng Zhou
- Department of Genetics; Medical College of Soochow University; Suzhou China
| | - Jiachun Lu
- Biomedicine Research Center of The Third Affiliated Hospital of GMU, The State Key Lab of Respiratory Disease, The Institute for Chemical Carcinogenesis, Collaborative Innovation Center for Environmental Toxicity; Guangzhou Medical University; Guangzhou China
| |
Collapse
|
20
|
Zhu S, Guleria RS, Thomas CM, Roth A, Gerilechaogetu F, Kumar R, Dostal DE, Baker KM, Pan J. Loss of myocardial retinoic acid receptor α induces diastolic dysfunction by promoting intracellular oxidative stress and calcium mishandling in adult mice. J Mol Cell Cardiol 2016; 99:100-112. [PMID: 27539860 DOI: 10.1016/j.yjmcc.2016.08.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 08/10/2016] [Accepted: 08/12/2016] [Indexed: 01/09/2023]
Abstract
Retinoic acid receptor (RAR) has been implicated in pathological stimuli-induced cardiac remodeling. To determine whether the impairment of RARα signaling directly contributes to the development of heart dysfunction and the involved mechanisms, tamoxifen-induced myocardial specific RARα deletion (RARαKO) mice were utilized. Echocardiographic and cardiac catheterization studies showed significant diastolic dysfunction after 16wks of gene deletion. However, no significant differences were observed in left ventricular ejection fraction (LVEF), between RARαKO and wild type (WT) control mice. DHE staining showed increased intracellular reactive oxygen species (ROS) generation in the hearts of RARαKO mice. Significantly increased NOX2 (NADPH oxidase 2) and NOX4 levels and decreased SOD1 and SOD2 levels were observed in RARαKO mouse hearts, which were rescued by overexpression of RARα in cardiomyocytes. Decreased SERCA2a expression and phosphorylation of phospholamban (PLB), along with decreased phosphorylation of Akt and Ca2+/calmodulin-dependent protein kinase II δ (CaMKII δ) was observed in RARαKO mouse hearts. Ca2+ reuptake and cardiomyocyte relaxation were delayed by RARα deletion. Overexpression of RARα or inhibition of ROS generation or NOX activation prevented RARα deletion-induced decrease in SERCA2a expression/activation and delayed Ca2+ reuptake. Moreover, the gene and protein expression of RARα was significantly decreased in aged or metabolic stressed mouse hearts. RARα deletion accelerated the development of diastolic dysfunction in streptozotocin (STZ)-induced type 1 diabetic mice or in high fat diet fed mice. In conclusion, myocardial RARα deletion promoted diastolic dysfunction, with a relative preserved LVEF. Increased oxidative stress have an important role in the decreased expression/activation of SERCA2a and Ca2+ mishandling in RARαKO mice, which are major contributing factors in the development of diastolic dysfunction. These data suggest that impairment of cardiac RARα signaling may be a novel mechanism that is directly linked to pathological stimuli-induced diastolic dysfunction.
Collapse
Affiliation(s)
- Sen Zhu
- Department of Medicine, College of Medicine, Texas A&M University Health Science Center, Central Texas Veterans Health Care System, Baylor Scott & White Health, Temple, TX, United States
| | - Rakeshwar S Guleria
- Department of Medicine, College of Medicine, Texas A&M University Health Science Center, Central Texas Veterans Health Care System, Baylor Scott & White Health, Temple, TX, United States; Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Central Texas Veterans Health Care System, Baylor Scott & White Health, Temple, TX, United States.
| | - Candice M Thomas
- Department of Medicine, College of Medicine, Texas A&M University Health Science Center, Central Texas Veterans Health Care System, Baylor Scott & White Health, Temple, TX, United States
| | - Amanda Roth
- Department of Medicine, College of Medicine, Texas A&M University Health Science Center, Central Texas Veterans Health Care System, Baylor Scott & White Health, Temple, TX, United States
| | - Fnu Gerilechaogetu
- Department of Medicine, College of Medicine, Texas A&M University Health Science Center, Central Texas Veterans Health Care System, Baylor Scott & White Health, Temple, TX, United States
| | - Rajesh Kumar
- Department of Medicine, College of Medicine, Texas A&M University Health Science Center, Central Texas Veterans Health Care System, Baylor Scott & White Health, Temple, TX, United States; Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Central Texas Veterans Health Care System, Baylor Scott & White Health, Temple, TX, United States
| | - David E Dostal
- Department of Medicine, College of Medicine, Texas A&M University Health Science Center, Central Texas Veterans Health Care System, Baylor Scott & White Health, Temple, TX, United States; Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Central Texas Veterans Health Care System, Baylor Scott & White Health, Temple, TX, United States
| | - Kenneth M Baker
- Department of Medicine, College of Medicine, Texas A&M University Health Science Center, Central Texas Veterans Health Care System, Baylor Scott & White Health, Temple, TX, United States
| | - Jing Pan
- Department of Medicine, College of Medicine, Texas A&M University Health Science Center, Central Texas Veterans Health Care System, Baylor Scott & White Health, Temple, TX, United States; Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Central Texas Veterans Health Care System, Baylor Scott & White Health, Temple, TX, United States.
| |
Collapse
|
21
|
Chen CH, Ke LY, Chan HC, Lee AS, Lin KD, Chu CS, Lee MY, Hsiao PJ, Hsu C, Chen CH, Shin SJ. Electronegative low density lipoprotein induces renal apoptosis and fibrosis: STRA6 signaling involved. J Lipid Res 2016; 57:1435-46. [PMID: 27256691 PMCID: PMC4959859 DOI: 10.1194/jlr.m067215] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Indexed: 02/07/2023] Open
Abstract
Dyslipidemia has been proven to capably develop and aggravate chronic kidney disease. We also report that electronegative LDL (L5) is the most atherogenic LDL. On the other hand, retinoic acid (RA) and RA receptor (RAR) agonist are reported to be beneficial in some kidney diseases. “Stimulated by retinoic acid 6” (STRA6), one retinol-binding protein 4 receptor, was recently identified to regulate retinoid homeostasis. Here, we observed that L5 suppressed STRA6 cascades [STRA6, cellular retinol-binding protein 1 (CRBP1), RARs, retinoid X receptor α, and retinol, RA], but L5 simultaneously induced apoptosis and fibrosis (TGFβ1, Smad2, collagen 1, hydroxyproline, and trichrome) in kidneys of L5-injected mice and L5-treated renal tubular cells. These L5-induced changes of STRA6 cascades, renal apoptosis, and fibrosis were reversed in kidneys of LOX1−/− mice. LOX1 RNA silencing and inhibitor of c-Jun N-terminal kinase and p38MAPK rescued the suppression of STRA6 cascades and apoptosis and fibrosis in L5-treated renal tubular cells. Furthermore, crbp1 gene transfection reversed downregulation of STRA6 cascades, apoptosis, and fibrosis in L5-treated renal tubular cells. For mimicking STRA6 deficiency, efficient silencing of STRA6 RNA was performed and was found to repress STRA6 cascades and caused apoptosis and fibrosis in L1-treated renal tubular cells. In summary, this study reveals that electronegative L5 can cause kidney apoptosis and fibrosis via the suppression of STRA6 cascades, and implicates that STRA6 signaling may be involved in dyslipidemia-mediated kidney disease.
Collapse
Affiliation(s)
- Chao-Hung Chen
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Liang-Yin Ke
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan Lipid Science and Aging Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hua-Chen Chan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan Lipid Science and Aging Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - An-Sheng Lee
- Department of Medicine, Mackay Medical College, New Taipei, Taiwan
| | - Kun-Der Lin
- Divisions of Endocrinology and Metabolism Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Chih-Sheng Chu
- Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Mei-Yueh Lee
- Divisions of Endocrinology and Metabolism Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Pi-Jung Hsiao
- Divisions of Endocrinology and Metabolism Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan Departments of Internal Medicine College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chin Hsu
- Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chu-Huang Chen
- Lipid Science and Aging Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan Department of Vascular and Medical Research, Texas Heart Institute, Houston, TX
| | - Shyi-Jang Shin
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan Lipid Science and Aging Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan Divisions of Endocrinology and Metabolism Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan Departments of Internal Medicine College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
22
|
Hamilton JP, Koganti L, Muchenditsi A, Pendyala VS, Huso D, Hankin J, Murphy RC, Huster D, Merle U, Mangels C, Yang N, Potter JJ, Mezey E, Lutsenko S. Activation of liver X receptor/retinoid X receptor pathway ameliorates liver disease in Atp7B(-/-) (Wilson disease) mice. Hepatology 2016; 63:1828-41. [PMID: 26679751 PMCID: PMC4874878 DOI: 10.1002/hep.28406] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 10/30/2015] [Accepted: 12/15/2015] [Indexed: 01/07/2023]
Abstract
UNLABELLED Wilson disease (WD) is a hepatoneurological disorder caused by mutations in the copper-transporter, ATP7B. Copper accumulation in the liver is a hallmark of WD. Current therapy is based on copper chelation, which decreases the manifestations of liver disease, but often worsens neurological symptoms. We demonstrate that in Atp7b(-/-) mice, an animal model of WD, liver function can be significantly improved without copper chelation. Analysis of transcriptional and metabolic changes in samples from WD patients and Atp7b(-/-) mice identified dysregulation of nuclear receptors (NRs), especially the liver X receptor (LXR)/retinoid X receptor heterodimer, as an important event in WD pathogenesis. Treating Atp7b(-/-) mice with the LXR agonist, T0901317, ameliorated disease manifestations despite significant copper overload. Genetic markers of liver fibrosis and inflammatory cytokines were significantly decreased, lipid profiles normalized, and liver function and histology were improved. CONCLUSIONS The results demonstrate the major role of an altered NR function in the pathogenesis of WD and suggest that modulation of NR activity should be explored as a supplementary approach to improving liver function in WD. (Hepatology 2016;63:1828-1841).
Collapse
Affiliation(s)
- JP Hamilton
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - L Koganti
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - A Muchenditsi
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - VS Pendyala
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - D Huso
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - J Hankin
- University of Colorado, Denver, CO
| | | | - D Huster
- Deakoness Hospital, Leipzig, Germany
| | - U Merle
- University of Heidelberg, Germany
| | - C Mangels
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - N Yang
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - JJ Potter
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - E Mezey
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - S. Lutsenko
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD,Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
23
|
Lee SE, Koo YD, Lee JS, Kwak SH, Jung HS, Cho YM, Park YJ, Chung SS, Park KS. Retinoid X receptor α overexpression alleviates mitochondrial dysfunction-induced insulin resistance through transcriptional regulation of insulin receptor substrate 1. Mol Cells 2015; 38:356-61. [PMID: 25728751 PMCID: PMC4400311 DOI: 10.14348/molcells.2015.2280] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 12/22/2014] [Accepted: 12/22/2014] [Indexed: 11/27/2022] Open
Abstract
Mitochondrial dysfunction is associated with insulin resistance and diabetes. We previously showed that retinoid X receptor α (RXRα) played an important role in transcriptional regulation of oxidative phosphorylation (OXPHOS) genes in cells with mitochondrial dysfunction caused by mitochondrial DNA mutation. In this study, we investigated whether mitochondrial dysfunction induced by incubation with OXPHOS inhibitors affects insulin receptor substrate 1 (IRS1) mRNA and protein levels and whether RXRα activation or overexpression can restore IRS1 expression. Both IRS1 and RXRα protein levels were significantly reduced when C2C12 myotubes were treated with the OXPHOS complex inhibitors, rotenone and antimycin A. The addition of RXRα agonists, 9-cis retinoic acid (9cRA) and LG1506, increased IRS1 transcription and protein levels and restored mitochondrial function, which ultimately improved insulin signaling. RXRα overexpression also increased IRS1 transcription and mitochondrial function. Because RXRα overexpression, knock-down, or activation by LG1506 regulated IRS1 transcription mostly independently of mitochondrial function, it is likely that RXRα directly regulates IRS1 transcription. Consistent with the hypothesis, we showed that RXRα bound to the IRS1 promoter as a heterodimer with peroxisome proliferator-activated receptor δ (PPARδ). These results suggest that RXRα overexpression or activation alleviates insulin resistance by increasing IRS1 expression.
Collapse
Affiliation(s)
- Seung Eun Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 110-744,
Korea
| | - Young Do Koo
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 110-744,
Korea
| | - Ji Seon Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 110-744,
Korea
| | - Soo Heon Kwak
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 110-744,
Korea
| | - Hye Seung Jung
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 110-744,
Korea
| | - Young Min Cho
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 110-744,
Korea
| | - Young Joo Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 110-744,
Korea
| | - Sung Soo Chung
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 110-744,
Korea
| | - Kyong Soo Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 110-744,
Korea
| |
Collapse
|
24
|
Kovalevich J, Yen W, Ozdemir A, Langford D. Cocaine induces nuclear export and degradation of neuronal retinoid X receptor-γ via a TNF-α/JNK- mediated mechanism. J Neuroimmune Pharmacol 2015; 10:55-73. [PMID: 25586717 PMCID: PMC4336643 DOI: 10.1007/s11481-014-9573-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 11/26/2014] [Indexed: 12/14/2022]
Abstract
Cocaine abuse represents an immense societal health and economic burden for which no effective treatment currently exists. Among the numerous intracellular signaling cascades impacted by exposure to cocaine, increased and aberrant production of pro-inflammatory cytokines in the CNS has been observed. Additionally, we have previously reported a decrease in retinoid-X-receptor-gamma (RXR-γ) in brains of mice chronically exposed to cocaine. Through obligate heterodimerization with a number of nuclear receptors, RXRs serve as master regulatory transcription factors, which can potentiate or suppress expression of a wide spectrum of genes. Little is known about the regulation of RXR levels, but previous studies indicate cellular stressors such as cytokines negatively regulate levels of RXRs in vitro. To evaluate the mechanism underlying the cocaine-induced decreases in RXR-γ levels observed in vivo, we exposed neurons to cocaine in vitro and examined pathways which may contribute to disruption in RXR signaling, including activation of stress pathways by cytokine induction. In these studies, we provide the first evidence that cocaine exposure disrupts neuronal RXR-γ signaling in vitro by promoting its nuclear export and degradation. Furthermore, we demonstrate this effect may be mediated, at least in part, by cocaine-induced production of TNF-α and its downstream effector c-Jun-NH-terminal kinase (JNK). Findings from this study are therefore applicable to both cocaine abuse and to pathological conditions characterized by neuroinflammatory factors, such as neurodegenerative disease.
Collapse
Affiliation(s)
- Jane Kovalevich
- Department of Neuroscience, Temple University School of Medicine, Medical Education Research Building, 3500 North Broad Street, Philadelphia, PA, 19140, USA
| | | | | | | |
Collapse
|
25
|
Zhu J, Ning RB, Lin XY, Chai DJ, Xu CS, Xie H, Zeng JZ, Lin JX. Retinoid X receptor agonists inhibit hypertension-induced myocardial hypertrophy by modulating LKB1/AMPK/p70S6K signaling pathway. Am J Hypertens 2014; 27:1112-24. [PMID: 24603314 DOI: 10.1093/ajh/hpu017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Retinoid X receptor (RXR) has been demonstrated to play an important role in cardiac development and has been implicated in cardiovascular diseases. This study aimed to examine the effects of RXRα agonist bexarotene on pathological left ventricular hypertrophy (LVH) in a spontaneously hypertensive rat (SHR) model and the underlying mechanism. METHODS WKY rats served as controls. SHRs were randomized into 3 groups at the age of 4 weeks and were treated (once daily for 12 weeks) with either bexarotene (30 or 100mg/kg body weight) or vehicle alone. Echocardiography was performed to determine cardiac structure and function. Neonatal cardiomyocytes were treated with AngII (10(-7) mmol/L) with or without the indicated concentration of RXRα ligand 9-cis-RA. The protein abundances of β-actin, RXRα, LKB1, phospho-LKB1, AMPK, phospho-AMPK, P70S6K, phospho-P70S6K, ACE, and AT1 receptor were measured along with blood pressure, body weight and angiotensin II (Ang II) levels. The effects of LKB1 downregulation by LKB1 small, interfering RNA were examined. RESULTS Treatment of SHRs with bexarotene resulted in significant inhibition of LVH without eliminating hypertension. Immunoblot with heart tissue homogenates from SHRs revealed that bexarotene activated the LKB1/AMPK signaling pathway and inhibited p70S6K. However, the increased Ang II levels in SHR serum and heart tissue were not reduced by bexarotene treatment. Treatment of cardiomyocytes with Ang II resulted in significantly reduced LKB1/AMPK activity and increased p70S6K activity. 9-cis-RA antagonized Ang II-induced LKB1/AMPK and p70S6K activation changes in vitro. CONCLUSIONS RXR agonists prevent the inhibition of the LKB1/AMPK/p70S6K pathway and regulate protein synthesis to reduce LVH. This antihypertrophic effect of bexarotene is independent of blood pressure.
Collapse
Affiliation(s)
- Jiang Zhu
- First Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China
| | - Ruo-Bing Ning
- First Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China
| | - Xiao-Yan Lin
- Echocardiological Department, First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Da-Jun Chai
- Cardiovascular Department, First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China;
| | - Chang-Sheng Xu
- Cardiovascular Department, First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Hong Xie
- Cardiovascular Department, First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Jin-Zhang Zeng
- School of Pharmaceutical Sciences and Institute for Biomedical Research, Xiamen University, Xiamen, China
| | - Jin-Xiu Lin
- Cardiovascular Department, First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China;
| |
Collapse
|
26
|
Zálešák M, Blažíček P, Pancza D, Ledvényiová V, Barteková M, Nemčeková M, Čarnická S, Ziegelhöffer A, Ravingerová T. Severity of lethal ischemia/reperfusion injury in rat hearts subjected to ischemic preconditioning is increased under conditions of simulated hyperglycemia. Physiol Res 2014; 63:577-85. [PMID: 24908083 DOI: 10.33549/physiolres.932652] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The aim of our study was to characterize resistance to ischemia/reperfusion (I/R) injury in Langendorff-perfused rat hearts and effectivity of ischemic preconditioning (PC) under condition of simulated acute hyperglycemia (SAHG) by perfusion of the hearts with Krebs-Henseleit (KH) solution with elevated glucose concentration (22 mmol/l). I/R injury was induced by 30-min coronary occlusion followed by 120-min reperfusion and PC by two cycles of 5-min occlusion/5-min reperfusion, prior to I/R. The severity of I/R injury was characterized by determination of the size of infarction (IS, expressed in % of area at risk size) and the amount of heart-type fatty acid binding protein (h-FABP, a marker of cell injury) released from the hearts to the effluent. Significantly smaller IS (8.8+/-1 %) and lower total amount of released h-FABP (1808+/-660 pmol) in PC group compared with IS 17.1+/-1.2 % (p<0.01) and amount of h-FABP (8803+/-2415 pmol, p<0.05) in the non-PC control hearts perfused with standard KH solution (glucose 11 mmol/l) confirmed protective effects of PC. In contrast, in SAHG groups, PC enhanced IS (21.4+/-2.2 vs. 14.3+/-1.3 %, p<0.05) and increased total amount of h-FABP (5541+/-229 vs. 3458+/-283 pmol, p<0.05) compared with respective non-PC controls. Results suggest that PC has negative effect on resistance of the hearts to I/R injury under conditions of elevated glucose in vitro.
Collapse
Affiliation(s)
- M Zálešák
- Institute for Heart Research, Slovak Academy of Science, Bratislava, Slovak Republic.
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Pan J, Guleria RS, Zhu S, Baker KM. Molecular Mechanisms of Retinoid Receptors in Diabetes-Induced Cardiac Remodeling. J Clin Med 2014; 3:566-94. [PMID: 26237391 PMCID: PMC4449696 DOI: 10.3390/jcm3020566] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 03/17/2014] [Accepted: 03/25/2014] [Indexed: 02/07/2023] Open
Abstract
Diabetic cardiomyopathy (DCM), a significant contributor to morbidity and mortality in diabetic patients, is characterized by ventricular dysfunction, in the absence of coronary atherosclerosis and hypertension. There is no specific therapeutic strategy to effectively treat patients with DCM, due to a lack of a mechanistic understanding of the disease process. Retinoic acid, the active metabolite of vitamin A, is involved in a wide range of biological processes, through binding and activation of nuclear receptors: retinoic acid receptors (RAR) and retinoid X receptors (RXR). RAR/RXR-mediated signaling has been implicated in the regulation of glucose and lipid metabolism. Recently, it has been reported that activation of RAR/RXR has an important role in preventing the development of diabetic cardiomyopathy, through improving cardiac insulin resistance, inhibition of intracellular oxidative stress, NF-κB-mediated inflammatory responses and the renin-angiotensin system. Moreover, downregulated RAR/RXR signaling has been demonstrated in diabetic myocardium, suggesting that impaired RAR/RXR signaling may be a trigger to accelerate diabetes-induced development of DCM. Understanding the molecular mechanisms of retinoid receptors in the regulation of cardiac metabolism and remodeling under diabetic conditions is important in providing the impetus for generating novel therapeutic approaches for the prevention and treatment of diabetes-induced cardiac complications and heart failure.
Collapse
Affiliation(s)
- Jing Pan
- Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A & M Health Science Center, Baylor Scott & White Health, Central Texas Veterans Health Care System, Temple, TX, 76504, USA.
| | - Rakeshwar S Guleria
- Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A & M Health Science Center, Baylor Scott & White Health, Central Texas Veterans Health Care System, Temple, TX, 76504, USA.
| | - Sen Zhu
- Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A & M Health Science Center, Baylor Scott & White Health, Central Texas Veterans Health Care System, Temple, TX, 76504, USA.
| | - Kenneth M Baker
- Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A & M Health Science Center, Baylor Scott & White Health, Central Texas Veterans Health Care System, Temple, TX, 76504, USA.
| |
Collapse
|
28
|
Richards EM, Wood CE, Rabaglino MB, Antolic A, Keller-Wood M. Mechanisms for the adverse effects of late gestational increases in maternal cortisol on the heart revealed by transcriptomic analyses of the fetal septum. Physiol Genomics 2014; 46:547-59. [PMID: 24867915 DOI: 10.1152/physiolgenomics.00009.2014] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
We have previously shown in sheep that 10 days of modest chronic increase in maternal cortisol resulting from maternal infusion of cortisol (1 mg/kg/day) caused fetal heart enlargement and Purkinje cell apoptosis. In subsequent studies we extended the cortisol infusion to term, finding a dramatic incidence of stillbirth in the pregnancies with chronically increased cortisol. To investigate effects of maternal cortisol on the heart, we performed transcriptomic analyses on the septa using ovine microarrays and Webgestalt and Cytoscape programs for pathway inference. Analyses of the transcriptomic effects of maternal cortisol infusion for 10 days (130 day cortisol vs 130 day control), or ∼25 days (140 day cortisol vs 140 day control) and of normal maturation (140 day control vs 130 day control) were performed. Gene ontology terms related to immune function and cytokine actions were significantly overrepresented as genes altered by both cortisol and maturation in the septa. After 10 days of cortisol, growth factor and muscle cell apoptosis pathways were significantly overrepresented, consistent with our previous histologic findings. In the term fetuses (∼25 days of cortisol) nutrient pathways were significantly overrepresented, consistent with altered metabolism and reduced mitochondria. Analysis of mitochondrial number by mitochondrial DNA expression confirmed a significant decrease in mitochondria. The metabolic pathways modeled as altered by cortisol treatment to term were different from those modeled during maturation of the heart to term, and thus changes in gene expression in these metabolic pathways may be indicative of the fetal heart pathophysiologies seen in pregnancies complicated by stillbirth, including gestational diabetes, Cushing's disease and chronic stress.
Collapse
Affiliation(s)
- Elaine M Richards
- Department of Pharmacodynamics, University of Florida, Gainesville, Florida; and
| | - Charles E Wood
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
| | - Maria Belen Rabaglino
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
| | - Andrew Antolic
- Department of Pharmacodynamics, University of Florida, Gainesville, Florida; and
| | - Maureen Keller-Wood
- Department of Pharmacodynamics, University of Florida, Gainesville, Florida; and
| |
Collapse
|
29
|
Bao W, Pan F, Chen L, Su G, Gao X, Li Y, Sun Q, Sun J, He K, Song H. The PI3K/AKT Pathway and FOXO3a Transcription Factor Mediate High Glucose-Induced Apoptosis in Neonatal Rat Ventricular Myocytes. IRANIAN RED CRESCENT MEDICAL JOURNAL 2014; 16:e14914. [PMID: 24910802 PMCID: PMC4028775 DOI: 10.5812/ircmj.14914] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 12/18/2013] [Accepted: 01/28/2014] [Indexed: 01/09/2023]
Abstract
Background: PI3K/AKT pathway plays major roles in regulating cardiomyocyte metabolism. The roles of PI3K/AKT pathway and FOXO3a in mediating high glucose-induced apoptosis in cardiomyocytes remain unclear. Objectives: In this experimental study, we investigated the mechanisms of the PI3K/AKT pathway and FOXO3a in mediating hyperglycemia-induced apoptosis in neonatal rat ventricular myocytes (NRVMs). Materials and Methods: NRVMs were adopted as the cell model to investigate the roles of PI3K/AKT and FOXO3a in mediating hyperglycemia-induced apoptosis in cardiomyocytes. Annexin-V-FITC staining and PI staining were used to evaluate the apoptosis in NRVMs under indicated conditions of serum starvation, high glucose exposure, and pharmacological or genetic manipulations on the expressions of PI3K/AKT and FOXO3a. Western blotting was conducted to evaluate the cytoplasmic/nuclear localization of FOXO3a in NRVMs exposed to high glucose. FOXO3a transcriptional activity was measured by luciferase reporter assay. Results: High glucose (30 mM) induced significant apoptosis in serum-starved NRVMs as compared with normal glucose (5 mM) control (12.01 ± 0.76% vs. 2.86 ± 0.55%; P < 0.001). Treatment with IGF1 attenuated hyperglycemia-induced apoptosis by 68% (3.23 ± 0.76% vs. 9.97 ± 1.29%; P < 0.001; n = 3) in comparison with the non-treated control. Treatment with PI3K inhibitor LY294002 enhanced hyperglycemia-induced apoptosis by 109% (20.83 ± 1.87% vs. 9.97 ± 1.29%; P < 0.001; n = 3) in comparison with the non-treated control. Over-expression of AKT by transduction with CA-AKT attenuated hyperglycemia-induced apoptosis by 47% (5.48 ± 0.35% vs.10.31 ± 0.94%; P < 0.001; n = 3) in comparison with the empty-vector control. Transduction with DN-AKT enhanced high glucose-induced apoptosis by 105% (21.13 ± 1.11% vs. 10.31 ± 0.94%; P < 0.001; n = 3) in comparison with the empty-vector control. Western blotting showed that high glucose induced a significant increase in FOXO3a nuclear localization. Luciferase reporter assay showed that high glucose induced a significant increase of 310% (P < 0.001; n = 3) in FOXO3a transcriptional activity against Fas ligand when NRVMs were transducted with TM-FOXO3a in comparison with the empty-vector control. Conclusions: The PI3K/AKT pathway mediated hyperglycemia-induced apoptosis of NRVMs through the translocation of FOXO3a to nuclei and the resultant enhanced transcriptional activity of FOXO3.
Collapse
Affiliation(s)
- Weiguo Bao
- Department of Cardiac Surgery, Second Hospital of Shandong University, Jinan, PR China
| | - Feng Pan
- Department of Cardiology, Jinan Central Hospital of Shandong University, Jinan, PR China
| | - Ling Chen
- Department of Cardiac Surgery, Second Hospital of Shandong University, Jinan, PR China
| | - Guohai Su
- Department of Cardiology, Jinan Central Hospital of Shandong University, Jinan, PR China
| | - Xiaoyuan Gao
- Department of Cardiology, Jinan Central Hospital of Shandong University, Jinan, PR China
| | - Ying Li
- Department of Cardiology, Jinan Central Hospital of Shandong University, Jinan, PR China
| | - Qiang Sun
- Department of Cardiac Surgery, Second Hospital of Shandong University, Jinan, PR China
| | - Jinhui Sun
- Department of Cardiac Surgery, Second Hospital of Shandong University, Jinan, PR China
| | - Kun He
- Department of Cardiology, Jinan Central Hospital of Shandong University, Jinan, PR China
| | - Hui Song
- Department of Cardiology, Jinan Central Hospital of Shandong University, Jinan, PR China
- School of Public Health, Shandong University, Jinan, PR China
- Corresponding Author: Hui Song, Department of Cardiology, Jinan Central Hospital of Shandong University, Jinan, PR China, Tel: +86-13370582767, E-mail:
| |
Collapse
|
30
|
Chen JY, Chou HC, Chen YH, Chan HL. High glucose-induced proteome alterations in hepatocytes and its possible relevance to diabetic liver disease. J Nutr Biochem 2013; 24:1889-910. [DOI: 10.1016/j.jnutbio.2013.05.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 05/16/2013] [Accepted: 05/24/2013] [Indexed: 12/11/2022]
|
31
|
Soubeyrand S, Naing T, Martinuk A, McPherson R. ERK1/2 regulates hepatocyte Trib1 in response to mitochondrial dysfunction. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:3405-3414. [PMID: 24161842 DOI: 10.1016/j.bbamcr.2013.10.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Revised: 09/13/2013] [Accepted: 10/02/2013] [Indexed: 01/10/2023]
Abstract
The TRIB1 locus (8q24.13) is a novel locus identified and replicated by several genome-wide association studies for associations with plasma triglycerides, apolipoprotein B and coronary artery disease. The TRIB1 protein product, tribbles-like protein 1 (Trib1), regulates MAPK activity. MAP kinases transduce a large variety of external signals, leading to a wide range of cellular responses, including growth, differentiation, inflammation and apoptosis. Importantly, Trib1 has been shown to regulate hepatic lipogenesis and very low density lipoprotein production. Despite the relevance of hepatocyte Trib1 to lipid metabolism and atherosclerosis, little is known about the mechanisms regulating Trib1 itself. Here, we identify the mitochondria axis as a regulator of Trib1. Treatment of HepG2 cells with a short pulse of a low oligomycin concentration led to a potent and prolonged increase in the Trib1 mRNA, an effect that was shared with other mitochondria stressors. HuH7 cells as well murine hepatocytes were also responsive albeit to a weaker extent. The upregulation appeared largely independent of reactive oxygen species generation or metabolic stress and was mainly under transcriptional control, with ERK1/2 playing an important regulating role in the process. While the presence of the Trib1 protein could be inferred, attempts to correlate the increased mRNA to changes in protein level were unsuccessful due to the lack of recognizable Trib1 signal. Our data enrich the current paradigm of Trib1 as an activator of the MAPK pathway by uncovering a role for MAPK in regulating Trib1.
Collapse
Affiliation(s)
| | - Thet Naing
- Atherogenomics Laboratory, University of Ottawa Heart Institute, Ottawa, Canada
| | - Amy Martinuk
- Atherogenomics Laboratory, University of Ottawa Heart Institute, Ottawa, Canada
| | - Ruth McPherson
- Atherogenomics Laboratory, University of Ottawa Heart Institute, Ottawa, Canada; Division of Cardiology, University of Ottawa Heart Institute, Ottawa, Canada.
| |
Collapse
|
32
|
Song SE, Kim YW, Kim JY, Lee DH, Kim JR, Park SY. IGFBP5 mediates high glucose-induced cardiac fibroblast activation. J Mol Endocrinol 2013; 50:291-303. [PMID: 23417767 DOI: 10.1530/jme-12-0194] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
This study examined whether IGF-binding protein 5 (IGFBP5) is involved in the high glucose-induced deteriorating effects in cardiac cells. Cardiac fibroblasts and cardiomyocytes were isolated from the hearts of 1- to 3-day-old Sprague Dawley rats. Treatment of fibroblasts with 25 mM glucose increased the number of cells and the mRNA levels of collagen III, matrix metalloproteinase 2 (MMP2), and MMP9. High glucose increased ERK1/2 activity, and the ERK1/2 inhibitor PD98059 suppressed high glucose-mediated fibroblast proliferation and increased collagen III mRNA levels. Whereas high glucose increased both mRNA and protein levels of IGFBP5 in fibroblasts, high glucose did not affect IGFBP5 protein levels in cardiomyocytes. The high glucose-induced increase in IGFBP5 protein levels was inhibited by PD98059 in fibroblasts. While recombinant IGFBP5 increased ERK phosphorylation, cell proliferation, and the mRNA levels of collagen III, MMP2, and MMP9 in fibroblasts, IGFBP5 increased c-Jun N-terminal kinase phosphorylation and induced apoptosis in cardiomyocytes. The knockdown of IGFBP5 inhibited high glucose-induced cell proliferation and collagen III mRNA levels in fibroblasts. Although high glucose increased IGF1 levels, IGF1 did not increase IGFBP5 levels in fibroblasts. The hearts of Otsuka Long-Evans Tokushima Fatty rats and the cardiac fibroblasts of streptozotocin-induced diabetic rats showed increased IGFBP5 expression. These results suggest that IGFBP5 mediates high glucose-induced profibrotic effects in cardiac fibroblasts.
Collapse
Affiliation(s)
- Seung Eun Song
- Department of Physiology, College of Medicine, Yeungnam University, Daegu 705-717, South Korea
| | | | | | | | | | | |
Collapse
|
33
|
Ding F, Yu L, Wang M, Xu S, Xia Q, Fu G. O-GlcNAcylation involvement in high glucose-induced cardiac hypertrophy via ERK1/2 and cyclin D2. Amino Acids 2013; 45:339-49. [PMID: 23665912 DOI: 10.1007/s00726-013-1504-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2013] [Accepted: 04/19/2013] [Indexed: 01/26/2023]
Abstract
Continuous hyperglycemia is considered to be the most significant pathogenesis of diabetic cardiomyopathy, which manifests as cardiac hypertrophy and subsequent heart failure. O-GlcNAcylation has attracted attention as a post-translational protein modification in the past decade. The role of O-GlcNAcylation in high glucose-induced cardiomyocyte hypertrophy remains unclear. We studied the effect of O-GlcNAcylation on neonatal rat cardiomyocytes that were exposed to high glucose and myocardium in diabetic rats induced by streptozocin. High glucose (30 mM) incubation induced a greater than twofold increase in cell size and increased hypertrophy marker gene expression accompanied by elevated O-GlcNAcylation protein levels. High glucose increased ERK1/2 but not p38 MAPK or JNK activity, and cyclin D2 expression was also increased. PUGNAc, an inhibitor of β-N-acetylglucosaminidase, enhanced O-GlcNAcylation and imitated the effects of high glucose. OGT siRNA and ERK1/2 inhibition with PD98059 treatment blunted the hypertrophic response and cyclin D2 upregulation. OGT inhibition also prevented ERK1/2 activation. We also observed concentric hypertrophy and similar changes of O-GlcNAcylation level, ERK1/2 activation and cyclin D2 expression in myocardium of diabetic rats induced by streptozocin. In conclusion, O-GlcNAcylation plays a role in high glucose-induced cardiac hypertrophy via ERK1/2 and cyclin D2.
Collapse
Affiliation(s)
- Fang Ding
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, China
| | | | | | | | | | | |
Collapse
|
34
|
Ahmadian M, Suh JM, Hah N, Liddle C, Atkins AR, Downes M, Evans RM. PPARγ signaling and metabolism: the good, the bad and the future. Nat Med 2013; 19:557-66. [PMID: 23652116 PMCID: PMC3870016 DOI: 10.1038/nm.3159] [Citation(s) in RCA: 1654] [Impact Index Per Article: 137.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 03/06/2013] [Indexed: 11/09/2022]
Abstract
Thiazolidinediones (TZDs) are potent insulin sensitizers that act through the nuclear receptor peroxisome proliferator-activated receptor-γ (PPARγ) and are highly effective oral medications for type 2 diabetes. However, their unique benefits are shadowed by the risk for fluid retention, weight gain, bone loss and congestive heart failure. This raises the question as to whether it is possible to build a safer generation of PPARγ-specific drugs that evoke fewer side effects while preserving insulin-sensitizing potential. Recent studies that have supported the continuing physiologic and therapeutic relevance of the PPARγ pathway also provide opportunities to develop newer classes of molecules that reduce or eliminate adverse effects. This review highlights key advances in understanding PPARγ signaling in energy homeostasis and metabolic disease and also provides new explanations for adverse events linked to TZD-based therapy.
Collapse
Affiliation(s)
- Maryam Ahmadian
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, California, USA
| | | | | | | | | | | | | |
Collapse
|
35
|
Rassaf T, Kelm M. Protection from diabetic cardiomyopathy - putative role of the retinoid receptor-mediated signaling. J Mol Cell Cardiol 2013; 59:179-80. [PMID: 23542309 DOI: 10.1016/j.yjmcc.2013.03.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Revised: 03/09/2013] [Accepted: 03/19/2013] [Indexed: 02/05/2023]
|
36
|
Miao X, Cui W, Sun W, Xin Y, Wang B, Tan Y, Cai L, Miao L, Fu Y, Su G, Wang Y. Therapeutic effect of MG132 on the aortic oxidative damage and inflammatory response in OVE26 type 1 diabetic mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:879516. [PMID: 23589759 PMCID: PMC3622385 DOI: 10.1155/2013/879516] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 02/28/2013] [Indexed: 01/01/2023]
Abstract
The present study tested whether MG132 increases vascular nuclear factor E2-related factor-2 (Nrf2) expression and transcription to provide a therapeutic effect on diabetes-induced pathogenic changes in the aorta. To this end, three-month-old OVE26 diabetic and age-matched control mice were intraperitoneally injected with MG-132, 10 μ g/kg daily for 3 months. OVE26 transgenic type 1 diabetic mice develop hyperglycemia at 2-3 weeks of age and exhibit albuminuria at 3 months of age with mild increases in TNF- α expression and 3-NT accumulation in the aorta. Diabetes-induced significant increases in the wall thickness and structural derangement of aorta were found in OVE26 mice with significant increases in aortic oxidative and nitrosative damage, inflammation, and remodeling at 6 months of diabetes, but not at 3 months of diabetes. However, these pathological changes seen at the 6 months of diabetes were abolished in OVE26 mice treated with MG-132 for 3 months that were also associated with a significant increase in Nrf2 expression in the aorta as well as transcription of downstream genes. These results suggest that chronic treatment with low-dose MG132 can afford an effective therapy for diabetes-induced pathogenic changes in the aorta, which is associated with the increased Nrf2 expression and transcription.
Collapse
Affiliation(s)
- Xiao Miao
- The Second Hospital of Jilin University, Changchun 130041, China
- KCHRI, Department of Pediatrics, University of Louisville, Louisville, KY 40202, USA
- Department of Ophthalmology, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun 130041, China
| | - Wenpeng Cui
- The Second Hospital of Jilin University, Changchun 130041, China
- KCHRI, Department of Pediatrics, University of Louisville, Louisville, KY 40202, USA
| | - Weixia Sun
- KCHRI, Department of Pediatrics, University of Louisville, Louisville, KY 40202, USA
- The First Hospital of Jilin University, Changchun 130021, China
| | - Ying Xin
- KCHRI, Department of Pediatrics, University of Louisville, Louisville, KY 40202, USA
- Normal Bethune Medical College of Jilin University, Changchun 130021, China
| | - Bo Wang
- KCHRI, Department of Pediatrics, University of Louisville, Louisville, KY 40202, USA
- Department of Pathology, Inner Mongolia Forestry General Hospital, Yakeshi, Inner Mongolia 022150, China
| | - Yi Tan
- KCHRI, Department of Pediatrics, University of Louisville, Louisville, KY 40202, USA
- Chinese American Research Institute for Diabetic Complications, Wenzhou Medical College, Wenzhou 325000, China
| | - Lu Cai
- KCHRI, Department of Pediatrics, University of Louisville, Louisville, KY 40202, USA
- Chinese American Research Institute for Diabetic Complications, Wenzhou Medical College, Wenzhou 325000, China
- Departments of Radiation Oncology and Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA
| | - Lining Miao
- The Second Hospital of Jilin University, Changchun 130041, China
| | - Yaowen Fu
- The First Hospital of Jilin University, Changchun 130021, China
| | - Guanfang Su
- The Second Hospital of Jilin University, Changchun 130041, China
- Department of Ophthalmology, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun 130041, China
| | - Yuehui Wang
- The Second Hospital of Jilin University, Changchun 130041, China
- Department of Cardiology, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun 130041, China
| |
Collapse
|
37
|
Cui W, Bai Y, Luo P, Miao L, Cai L. Preventive and therapeutic effects of MG132 by activating Nrf2-ARE signaling pathway on oxidative stress-induced cardiovascular and renal injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:306073. [PMID: 23533688 PMCID: PMC3606804 DOI: 10.1155/2013/306073] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Accepted: 02/14/2013] [Indexed: 12/25/2022]
Abstract
So far, cardiovascular and renal diseases have brought us not only huge economic burden but also serious society problems. Since effective therapeutic strategies are still limited, to find new methods for the prevention or therapy of these diseases is important. Oxidative stress has been found to play a critical role in the initiation and progression of cardiovascular and renal diseases. In addition, activation of nuclear-factor-E2-related-factor-2- (Nrf2-) antioxidant-responsive element (ARE) signaling pathway protects cells and tissues from oxidative damage. As a proteasomal inhibitor, MG132 was reported to activate Nrf2 expression and function, which was accompanied with significant preventive and/or therapeutic effect on cardiovascular and renal diseases under most conditions; therefore, MG132 seems to be a potentially effective drug to be used in the prevention of oxidative damage. In this paper, we will summarize the information available regarding the effect of MG132 on oxidative stress-induced cardiovascular and renal damage, especially through Nrf2-ARE signaling pathway.
Collapse
Affiliation(s)
- Wenpeng Cui
- Department of Nephrology, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun, Jilin Province 130041, China
- KCHRI at the Department of Pediatrics, University of Louisville, 570 South Preston Street, Baxter I, Suite 304F, Louisville, KY 40202, USA
| | - Yang Bai
- KCHRI at the Department of Pediatrics, University of Louisville, 570 South Preston Street, Baxter I, Suite 304F, Louisville, KY 40202, USA
- Department of Cardiology, The People's Hospital of Jilin Province, Changchun 130021, China
| | - Ping Luo
- Department of Nephrology, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun, Jilin Province 130041, China
| | - Lining Miao
- Department of Nephrology, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun, Jilin Province 130041, China
| | - Lu Cai
- KCHRI at the Department of Pediatrics, University of Louisville, 570 South Preston Street, Baxter I, Suite 304F, Louisville, KY 40202, USA
| |
Collapse
|
38
|
Chae S, Ahn BY, Byun K, Cho YM, Yu MH, Lee B, Hwang D, Park KS. A Systems Approach for Decoding Mitochondrial Retrograde Signaling Pathways. Sci Signal 2013; 6:rs4. [DOI: 10.1126/scisignal.2003266] [Citation(s) in RCA: 140] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
39
|
Guleria RS, Singh AB, Nizamutdinova IT, Souslova T, Mohammad AA, Kendall JA, Baker KM, Pan J. Activation of retinoid receptor-mediated signaling ameliorates diabetes-induced cardiac dysfunction in Zucker diabetic rats. J Mol Cell Cardiol 2013; 57:106-18. [PMID: 23395853 DOI: 10.1016/j.yjmcc.2013.01.017] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Revised: 01/07/2013] [Accepted: 01/29/2013] [Indexed: 01/04/2023]
Abstract
Diabetic cardiomyopathy (DCM) is a significant contributor to the morbidity and mortality associated with diabetes and metabolic syndrome. Retinoids, through activation of retinoic acid receptor (RAR) and retinoid x receptor (RXR), have been linked to control glucose and lipid homeostasis, with effects on obesity and diabetes. However, the functional role of RAR and RXR in the development of DCM remains unclear. Zucker diabetic fatty (ZDF) and lean rats were treated with Am580 (RARα agonist) or LGD1069 (RXR agonist) for 16 weeks, and cardiac function and metabolic alterations were determined. Hyperglycemia, hyperlipidemia and insulin resistance were observed in ZDF rats. Diabetic cardiomyopathy was characterized in ZDF rats by increased oxidative stress, apoptosis, fibrosis, inflammation, activation of MAP kinases and NF-κB signaling and diminished Akt phosphorylation, along with decreased glucose transport and increased cardiac lipid accumulation, and ultimately diastolic dysfunction. Am580 and LGD1069 attenuated diabetes-induced cardiac dysfunction and the pathological alterations, by improving glucose tolerance and insulin resistance; facilitating Akt activation and glucose utilization, and attenuating oxidative stress and interrelated MAP kinase and NF-κB signaling pathways. Am580 inhibited body weight gain, attenuated the increased cardiac fatty acid uptake, β-oxidation and lipid accumulation in the hearts of ZDF rats. However, LGD1069 promoted body weight gain, hyperlipidemia and cardiac lipid accumulation. In conclusion, our data suggest that activation of RAR and RXR may have therapeutic potential in the treatment of diabetic cardiomyopathy. However, further studies are necessary to clarify the role of RAR and RXR in the regulation of lipid metabolism and homeostasis.
Collapse
Affiliation(s)
- Rakeshwar S Guleria
- Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M Health Science Center, Central Texas Veterans Health Care System, Temple, TX, USA
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Nizamutdinova IT, Guleria RS, Singh AB, Kendall JA, Baker KM, Pan J. Retinoic acid protects cardiomyocytes from high glucose-induced apoptosis through inhibition of NF-κB signaling pathway. J Cell Physiol 2013; 228:380-92. [PMID: 22718360 DOI: 10.1002/jcp.24142] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
We have previously shown that retinoic acid (RA) has protective effects on high glucose (HG)-induced cardiomyocyte apoptosis. To further elucidate the molecular mechanisms of RA effects, we determined the interaction between nuclear factor (NF)-κB and RA signaling. HG induced a sustained phosphorylation of IKK/IκBα and transcriptional activation of NF-κB in cardiomyocytes. Activated NF-κB signaling has an important role in HG-induced cardiomyocyte apoptosis and gene expression of interleukin-6 (IL-6), tumor necrosis factor (TNF)-α, and monocyte chemoattractant protein-1 (MCP-1). All-trans RA (ATRA) and LGD1069, through activation of RAR/RXR-mediated signaling, inhibited the HG-mediated effects in cardiomyocytes. The inhibitory effect of RA on NF-κB activation was mediated through inhibition of IKK/IκBα phosphorylation. ATRA and LGD1069 treatment promoted protein phosphatase 2A (PP2A) activity, which was significantly suppressed by HG stimulation. The RA effects on IKK and IκBα were blocked by okadaic acid or silencing the expression of PP2Ac-subunit, indicating that the inhibitory effect of RA on NF-κB is regulated through activation of PP2A and subsequent dephosphorylation of IKK/IκBα. Moreover, ATRA and LGD1069 reversed the decreased PP2A activity and inhibited the activation of IKK/IκBα and gene expression of MCP-1, IL-6, and TNF-α in the hearts of Zucker diabetic fatty rats. In summary, our findings suggest that the suppressed activation of PP2A contributed to sustained activation of NF-κB in HG-stimulated cardiomyocytes; and that the protective effect of RA on hyperglycemia-induced cardiomyocyte apoptosis and inflammatory responses is partially regulated through activation of PP2A and suppression of NF-κB-mediated signaling and downstream targets.
Collapse
Affiliation(s)
- Irina T Nizamutdinova
- Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M Health Science Center, Temple, Texas 76504, USA
| | | | | | | | | | | |
Collapse
|
41
|
Chen YH, Chen JY, Chen YW, Lin ST, Chan HL. High glucose-induced proteome alterations in retinal pigmented epithelium cells and its possible relevance to diabetic retinopathy. MOLECULAR BIOSYSTEMS 2012; 8:3107-24. [PMID: 23051786 DOI: 10.1039/c2mb25331c] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2023]
Abstract
Diabetic retinopathy can cause poor vision and blindness. Previous research has shown that high blood glucose weakens retinal capillaries and induces glycoxidation. However, the detailed molecular mechanisms underlying the effects of high blood glucose on development of diabetic retinopathy have yet to be elucidated. In this study, we cultured a retinal pigmented epithelium cell line (ARPE-19) in mannitol-balanced 5.5 mM, 25 mM, and 100 mM d-glucose media, and evaluated protein expression and redox-regulation. We identified 56 proteins that showed significant changes in protein expression, and 33 proteins showing significant changes in thiol reactivity, in response to high glucose concentration. Several proteins that are involved in signal transduction, gene regulation, and transport showed significant changes in expression, whereas proteins involved in metabolism, transport, and cell survival displayed changes in thiol reactivity. Further analyses of clinical plasma specimens confirmed that the proteins lamin B2, PUMA, WTAP, ASGR1, and prohibitin 2 showed type 2 diabetic retinopathy-dependent alterations. In summary, in this study, we used a comprehensive retinal cell-based proteomic approach for the identification of changes in protein expression and redox-associated retinal markers induced by high glucose concentration. Some of the identified proteins have been validated with clinical samples and provide potential targets for the prognosis and diagnosis of diabetic retinopathy.
Collapse
Affiliation(s)
- You-Hsuan Chen
- Institute of Bioinformatics and Structural Biology & Department of Medical Sciences, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | | | | | | | | |
Collapse
|
42
|
Chen CH, Hsieh TJ, Lin KD, Lin HY, Lee MY, Hung WW, Hsiao PJ, Shin SJ. Increased unbound retinol-binding protein 4 concentration induces apoptosis through receptor-mediated signaling. J Biol Chem 2012; 287:9694-9707. [PMID: 22308028 DOI: 10.1074/jbc.m111.301721] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The increase of apo-/holo-retinol-binding protein 4 (RBP4) concentrations has been found in subjects with renal dysfunction and even in diabetic patients with microalbuminuria. Holo-RBP4 is recognized to possess cytoprotective function. Therefore, we supposed that the relative increase in apo-RBP4 might induce cell damage. In this study, we investigated the signal transduction that activated apoptosis in response to the increase of apo-/holo-RBP4 concentration. We found that increase of apo-/holo-RBP4 concentration ratio delayed the displacement of RBP4 with "stimulated by retinoic acid 6" (STRA6), enhanced Janus kinase 2 (JAK2)/STAT5 cascade, up-regulated adenylate cyclase 6 (AC6), increased cAMP, enhanced JNK1/p38 cascade, suppressed CRBP-I/RARα (cellular retinol-binding protein/retinoic acid receptor α) expression, and led to apoptosis in HK-2 and human umbilical vein endothelial cells. Furthermore, STRA6, JAK2, STAT5, JNK1, or p38 siRNA and cAMP-PKA inhibitor reversed the repression of CRBP-I/RARα and apoptosis in apo-RBP4 stimulation. In conclusion, this study indicates that the increase of apo-/holo-RBP4 concentration may influence STRA6 signaling, finally causing apoptosis.
Collapse
Affiliation(s)
- Chao-Hung Chen
- Graduate Institute of Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Tusty-Jiuan Hsieh
- School of Medicine, College of Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Kun-Der Lin
- Division of Endocrinology and Metabolism, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Hsing-Yi Lin
- Division of Endocrinology and Metabolism, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Mei-Yueh Lee
- Division of Endocrinology and Metabolism, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Wei-Wen Hung
- Division of Endocrinology and Metabolism, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Pi-Jung Hsiao
- School of Medicine, College of Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Division of Endocrinology and Metabolism, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Shyi-Jang Shin
- School of Medicine, College of Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Division of Endocrinology and Metabolism, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| |
Collapse
|