1
|
Claro-Cala CM, Rivero-Pino F, Torrecillas-López M, Jimenez-Gonzalez V, Montserrat-de la Paz S. Immunonutrition: future perspective in neurodegenerative disorders. Nutr Neurosci 2024:1-12. [PMID: 39561029 DOI: 10.1080/1028415x.2024.2425565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
The relevance of lifestyle, including diet and exercise, has been associated with improved learning and memory capacity, delayed age-related cognitive decline, and a reduced risk of neurodegeneration. Most neurodegenerative diseases are defined as complex multifactorial disorders in which genetic and environmental factors greatly contribute to their onset. Although inflammatory cells produce reactive oxygen species (ROS), oxidative stress itself might exert pro-inflammatory effects and an uncontrolled response could lead to a state of chronic inflammation. Anti-inflammatory dietary approaches unify the disciplines of nutrition, immunity, and neurology. Personalized dietary interventions will be developed based on an individual's genetic makeup, metabolic profile, and gut microbiota composition, thanks to advances in genomics, metabolomics, and microbiome research. The relevance of dietary patterns in decreasing inflammation relies on the role of specific antioxidant nutrients, which might contribute to a decrease in the levels of ROS. This review aims to summarize recent advancements in neuroscience and immunology that have revealed the crucial role that diet and the immune system play in brain function and disease progression. Nutrition influences the immune system, and in turn, the immune system impacts neurological health. This bidirectional relationship suggests that targeted nutritional interventions could modulate immune responses to delay or mitigate the progression of neurodegenerative diseases potentially. This approach focuses on the use of specific nutrients and dietary components that influence the immune system and inflammatory pathway. Key elements of immunonutrition include omega-3 fatty acids, antioxidants, vitamins and various bioactive compounds found in foods.
Collapse
Affiliation(s)
- Carmen María Claro-Cala
- Department of Pharmacology, Pediatrics, and Radiology, School of Medicine, Universidad de Sevilla, Seville, Spain
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocio/CSIC/Universidad de Sevilla, Seville, Spain
| | - Fernando Rivero-Pino
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocio/CSIC/Universidad de Sevilla, Seville, Spain
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, Universidad de Sevilla, Seville, Spain
| | - María Torrecillas-López
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocio/CSIC/Universidad de Sevilla, Seville, Spain
| | - Víctor Jimenez-Gonzalez
- CITIUS (Centre for Research, Technology, and Innovation), University of Seville, Seville, Spain
| | - Sergio Montserrat-de la Paz
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocio/CSIC/Universidad de Sevilla, Seville, Spain
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, Universidad de Sevilla, Seville, Spain
| |
Collapse
|
2
|
Jia K, Luo X, Yi J, Zhang C. Hormonal influence: unraveling the impact of sex hormones on vascular smooth muscle cells. Biol Res 2024; 57:61. [PMID: 39227995 PMCID: PMC11373308 DOI: 10.1186/s40659-024-00542-w] [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: 04/30/2024] [Accepted: 08/26/2024] [Indexed: 09/05/2024] Open
Abstract
Sex hormones play a pivotal role as endocrine hormones that exert profound effects on the biological characteristics and vascular function of vascular smooth muscle cells (VSMCs). By modulating intracellular signaling pathways, activating nuclear receptors, and regulating gene expression, sex hormones intricately influence the morphology, function, and physiological state of VSMCs, thereby impacting the biological properties of vascular contraction, relaxation, and growth. Increasing evidence suggests that abnormal phenotypic changes in VSMCs contribute to the initiation of vascular diseases, including atherosclerosis. Therefore, understanding the factors governing phenotypic alterations in VSMCs and elucidating the underlying mechanisms can provide crucial insights for refining interventions targeted at vascular diseases. Additionally, the varying levels of different types of sex hormones in the human body, influenced by sex and age, may also affect the phenotypic conversion of VSMCs. This review aims to explore the influence of sex hormones on the phenotypic switching of VSMCs and the development of associated vascular diseases in the human body.
Collapse
Affiliation(s)
- Keran Jia
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xin Luo
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, 646000, China
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Jingyan Yi
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, 646000, China.
| | - Chunxiang Zhang
- Department of Cardiology, The Affiliated Hospital, Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, 646000, China.
| |
Collapse
|
3
|
Shi X, Zhang S, Li J, Ke Y, Bai Y. Fibronectin/α5 Integrin Contribute to Hypertension-Associated Arterial Ageing and Calcification through Affecting BMP2/MGP Imbalance and Enhancing Vascular Smooth Muscle Cell Phenotypic Transformation. Gerontology 2024; 70:858-875. [PMID: 38824923 DOI: 10.1159/000539399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 05/14/2024] [Indexed: 06/04/2024] Open
Abstract
INTRODUCTION Hypertension can accelerate and aggravate the process of arterial ageing and calcification. However, the mechanism behind has yet to be well elucidated. METHODS Here, we monitored the dynamic changes of fibronectin (FN)/α5 integrin, bone morphogenetic protein 2/matrix Gla protein (BMP2/MGP), and Runx2 in the aorta of spontaneously hypertensive rats (SHRs) and thoracic aortic vascular smooth muscle cells (VSMCs), also the phenotypic transformation of VSMCs during the process of arterial ageing and calcification. Further, study on arterial ageing and calcification through antagonist experiments at the molecular level was explored. RESULTS We found extracellular FN and its α5 integrin receptor expressions were positively associated with arterial ageing and calcification in SHR during ageing, as well in VSMCs from SHR in vitro. Integrin receptor inhibitor of GRGDSP would delay this arterial ageing and calcification process. Moreover, the elevated FN and α5 integrin receptor expression evoked the disequilibrium of BMP2/MGP, where the expression of BMP2, a potent osteogenic inducer, increased while MGP, a calcification inhibitor, decreased. Furthermore, it was followed by the upregulation of Runx2 and the phenotypic transformation of VSMCs from the contractile phenotype into the osteoblast-like cells. Notably, BMP2 antagonist of rmNoggin was sufficient to ameliorate the ageing and calcification process of VSMCs and exogenous BMP2-adding accelerate and aggregate the process. CONCLUSION Our study revealed that hypertension-associated arterial ageing and calcification might be a consequence that hypertension up-regulated FN and its high binding affinity integrin α5 receptor in the aortic wall, which in turn aggravated the imbalance of BMP2/MGP, promoted the transcription of Runx2, and induced the phenotypic transformation of VSMCs from the contractile phenotype into the osteoblast-like cells. Our study would provide insights into hypertension-associated arterial ageing and calcification and shed new light on the control of arterial calcification, especially for those with hypertension.
Collapse
Affiliation(s)
- Xiaoyun Shi
- Department of Geriatrics, Fujian Key Laboratory of Vascular Aging (Fujian Medical University), Fujian Clinical Research Center for Senile Vascular Aging and Brain Aging, Fujian Medical University Union Hospital, Fuzhou, China
| | - Siduo Zhang
- Department of Geriatrics, Fujian Key Laboratory of Vascular Aging (Fujian Medical University), Fujian Clinical Research Center for Senile Vascular Aging and Brain Aging, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jinghui Li
- Department of Geriatrics, Fujian Key Laboratory of Vascular Aging (Fujian Medical University), Fujian Clinical Research Center for Senile Vascular Aging and Brain Aging, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yilang Ke
- Department of Geriatrics, Fujian Key Laboratory of Vascular Aging (Fujian Medical University), Fujian Clinical Research Center for Senile Vascular Aging and Brain Aging, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yajing Bai
- Department of Geriatrics, Fujian Key Laboratory of Vascular Aging (Fujian Medical University), Fujian Clinical Research Center for Senile Vascular Aging and Brain Aging, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
4
|
Li Y, Li J, Tang X, Xu J, Liu R, Jiang L, Tian J, Zhang Y, Wang D, Sun K, Xu B, Zhao W, Hui R, Gao R, Song L, Yuan J, Zhao X. Association of NPC1L1 and HMGCR gene polymorphisms with coronary artery calcification in patients with premature triple-vessel coronary disease. BMC Med Genomics 2024; 17:22. [PMID: 38233830 PMCID: PMC10795340 DOI: 10.1186/s12920-024-01802-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 01/09/2024] [Indexed: 01/19/2024] Open
Abstract
BACKGROUND Coronary artery calcification (CAC) is a highly specific marker of atherosclerosis. Niemann-Pick C1-like 1 (NPC1L1) and 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR) are the therapeutic targets of ezetimibe and statins, respectively, which are important for the progression of atherosclerosis. However, CAC's genetic susceptibility with above targets is still unknown. We aimed to investigate the association of NPC1L1 and HMGCR gene polymorphisms with CAC in patients with premature triple-vessel disease (PTVD). METHODS Four single nucleotide polymorphisms (SNPs) (rs11763759, rs4720470, rs2072183, rs2073547) of NPC1L1, and three SNPs (rs12916, rs2303151, rs4629571) of HMGCR were genotyped in 872 PTVD patients. According to the coronary angiography results, patients were divided into low-degree CAC group and high-degree CAC group. RESULTS A total of 872 PTVD patients (mean age, 47.71 ± 6.12; male, 72.8%) were finally included for analysis. Multivariate logistic regression analysis showed no significant association between the SNPs of NPC1L1 and HMGCR genes and high-degree CAC in the total population (P > 0.05). Subgroup analysis by gender revealed that the variant genotype (TT/CT) of rs4720470 on NPC1L1 gene was associated with increased risk for high-degree CAC in male patients only (OR = 1.505, 95% CI: 1.008-2.249, P = 0.046) in dominant model, but no significant association was found in female population, other SNPs of NPC1L1 and HMGCR genes (all P > 0.05). CONCLUSIONS We reported for the first time that the rs4720470 on NPC1L1 gene was associated with high-degree CAC in male patients with PTVD. In the future, whether therapies related to this target could reduce CAC and cardiovascular events deserves further investigation.
Collapse
Affiliation(s)
- Yulong Li
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Jiawen Li
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Xiaofang Tang
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Jingjing Xu
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Ru Liu
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Lin Jiang
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Jian Tian
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Yin Zhang
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Dong Wang
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Kai Sun
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Bo Xu
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Wei Zhao
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Rutai Hui
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Runlin Gao
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Lei Song
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Beilishi Road, Xicheng District, Beijing, 100037, China.
| | - Jinqing Yuan
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Beilishi Road, Xicheng District, Beijing, 100037, China.
| | - Xueyan Zhao
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Beilishi Road, Xicheng District, Beijing, 100037, China.
| |
Collapse
|
5
|
Li P, Hong J, Liang C, Li Y, Gao L, Wu L, Yao R, Zhang Y. Endothelial cell-released extracellular vesicles trigger pyroptosis and vascular inflammation to induce atherosclerosis through the delivery of HIF1A-AS2. FASEB J 2023; 37:e22942. [PMID: 37178006 DOI: 10.1096/fj.202201399rrr] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 04/04/2023] [Accepted: 04/17/2023] [Indexed: 05/15/2023]
Abstract
Extracellular vesicles (EVs) possess great potential in the modulation of cardiovascular diseases. Our current work intended to assay the clinical significance of endothelial cell (EC)-derived EVs in atherosclerosis (AS). Expression of HIF1A-AS2, miR-455-5p, and ESRRG in plasma from AS patients and mice and EVs from ox-LDL-treated ECs was measured. Interactions among HIF1A-AS2, miR-455-5p, ESRRG, and NLRP3 were analyzed. Next, EVs were co-cultured with ECs, and ectopic expression and depletion experimentations of HIF1A-AS2, miR-455-5p, ESRRG, and/or NLRP3 were carried out to assay their roles in pyroptosis and inflammation of ECs in AS. At last, the effects of HIF1A-AS2 shuttled by EC-derived EVs on EC pyroptosis and vascular inflammation in AS were verified in vivo. HIF1A-AS2 and ESRRG were highly expressed, while miR-455-5p was poorly expressed in AS. HIF1A-AS2 could sponge miR-455-5p to elevate the expression of ESRRG and NLRP3. Both in vitro and in vivo experiments revealed that ECs-derived EVs carrying HIF1A-AS2 induced the pyroptosis and vascular inflammation of ECs to promote the progression of AS by sponging miR-455-5p via ESRRG/NLRP3. HIF1A-AS2 shuttled by ECs-derived EVs can accelerate the progression of AS by downregulating miR-455-5p and upregulating ESRRG and NLRP3.
Collapse
Affiliation(s)
- Pengcheng Li
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, P. R. China
| | - Jin Hong
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, P. R. China
| | - Cui Liang
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, P. R. China
| | - Yapeng Li
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, P. R. China
| | - Lu Gao
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, P. R. China
| | - Leiming Wu
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, P. R. China
| | - Rui Yao
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, P. R. China
| | - Yanzhou Zhang
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, P. R. China
| |
Collapse
|
6
|
Cerutti C, Shi JR, Vanacker JM. Multifaceted Transcriptional Network of Estrogen-Related Receptor Alpha in Health and Disease. Int J Mol Sci 2023; 24:ijms24054265. [PMID: 36901694 PMCID: PMC10002233 DOI: 10.3390/ijms24054265] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/15/2023] [Accepted: 02/18/2023] [Indexed: 02/23/2023] Open
Abstract
Estrogen-related receptors (ERRα, β and γ in mammals) are orphan members of the nuclear receptor superfamily acting as transcription factors. ERRs are expressed in several cell types and they display various functions in normal and pathological contexts. Amongst others, they are notably involved in bone homeostasis, energy metabolism and cancer progression. In contrast to other nuclear receptors, the activities of the ERRs are apparently not controlled by a natural ligand but they rely on other means such as the availability of transcriptional co-regulators. Here we focus on ERRα and review the variety of co-regulators that have been identified by various means for this receptor and their reported target genes. ERRα cooperates with distinct co-regulators to control the expression of distinct sets of target genes. This exemplifies the combinatorial specificity of transcriptional regulation that induces discrete cellular phenotypes depending on the selected coregulator. We finally propose an integrated view of the ERRα transcriptional network.
Collapse
|
7
|
An Inverse Agonist of Estrogen-Related Receptor Gamma, GSK5182, Enhances Na +/I - Symporter Function in Radioiodine-Refractory Papillary Thyroid Cancer Cells. Cells 2023; 12:cells12030470. [PMID: 36766812 PMCID: PMC9914548 DOI: 10.3390/cells12030470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/02/2023] [Accepted: 01/30/2023] [Indexed: 02/04/2023] Open
Abstract
Previously, we reported that an inverse agonist of estrogen-related receptor gamma (ERRγ), GSK5182, enhances sodium iodide (Na+/I-) symporter (NIS) function through mitogen-activated protein (MAP) kinase signaling in anaplastic thyroid cancer cells. This finding helped us to further investigate the effects of GSK5182 on NIS function in papillary thyroid cancer (PTC) refractory to radioactive iodine (RAI) therapy. Herein, we report the effects of ERRγ on the regulation of NIS function in RAI-resistant PTC cells using GSK5182. RAI-refractory BCPAP cells were treated with GK5182 for 24 h at various concentrations, and radioiodine avidity was determined with or without potassium perchlorate (KClO4) as an NIS inhibitor. We explored the effects of GSK5182 on ERRγ, the mitogen-activated protein (MAP) kinase pathway, and iodide metabolism-related genes. We examined whether the MAP pathway affected GSK5182-mediated NIS function using U0126, a selective MEK inhibitor. A clonogenic assay was performed to evaluate the cytotoxic effects of I-131. GSK5182 induced an increase in radioiodine avidity in a dose-dependent manner, and the enhanced uptake was completely inhibited by KClO4 in BCPAP cells. We found that ERRγ was downregulated and phosphorylated extracellular signal-regulated kinase (ERK)1/2 was upregulated in BCPAP cells, with an increase in total and membranous NIS and iodide metabolism-related genes. MEK inhibitors reversed the increase in radioiodine avidity induced by GSK5182. Clonogenic examination revealed the lowest survival in cells treated with a combination of GSK5182 and I-131 compared to those treated with either GSK518 or I-131 alone. We demonstrate that an inverse agonist of ERRγ, GSK5182, enhances the function of NIS protein via the modulation of ERRγ and MAP kinase signaling, thereby leading to increased responsiveness to radioiodine in RAI-refractory papillary thyroid cancer cells.
Collapse
|
8
|
Yamamoto H, Tanaka Y, Sawada M, Kihara S. ERRα Attenuates Vascular Inflammation via Enhanced NFκB Degradation Pathway. Endocrinology 2023; 164:6936569. [PMID: 36534970 DOI: 10.1210/endocr/bqac212] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022]
Abstract
We have previously reported that β-aminoisobutyric acid (BAIBA), a muscle-derived exercise mimetic, had anti-inflammatory and reactive oxygen species (ROS) scavenging effects in vascular endothelial cells through the enhanced expression of peroxisome proliferator-activated receptor gamma coactivator-1β (PGC-1β). Although BAIBA also increased the expression of estrogen-related receptor α (ERRα), the roles of ERRα in vascular endothelial cells have yet to be fully elucidated. Here, we found that human aortic endothelial cells (HAECs) infected with ERRα-expressing adenovirus had significantly decreased mRNA levels of tumor necrosis factor α-stimulated proinflammatory molecules. However, ERRα overexpression had little effect on the mRNA levels of PGC-1β, peroxisome proliferator-activated receptors, and almost all ROS scavenging molecules, except for superoxide dismutase 2. ERRα expression significantly decreased NFκB reporter activities in a dose-dependent manner with unaltered IκBα phosphorylation levels but with a significant increase in the mRNA levels of PDZ and LIM domain protein 2 (PDLIM2) and copper metabolism gene MURR1 domain-containing protein (COMMD1), which enhance the ubiquitination and degradation of NFκB. Also, PDLIM2 and COMMD1 mRNA levels were upregulated in BAIBA-treated HAECs. Finally, we identified the ERRα-response element in the COMMD1 promoter region (-283 to -29 bp). These results indicated that ERRα exerted anti-inflammatory effects in vascular endothelial cells through COMMD1-mediated attenuation of NFκB activity, which could be an atheroprotective mechanism of physical exercise.
Collapse
Affiliation(s)
- Hiroyasu Yamamoto
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, 1-7 Yamada-oka, Suita City, Osaka 565-0871, Japan
| | - Yuya Tanaka
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, 1-7 Yamada-oka, Suita City, Osaka 565-0871, Japan
| | - Miho Sawada
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, 1-7 Yamada-oka, Suita City, Osaka 565-0871, Japan
| | - Shinji Kihara
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, 1-7 Yamada-oka, Suita City, Osaka 565-0871, Japan
| |
Collapse
|
9
|
Zhao X, Li X, Liu P, Li P, Xu X, Chen Y, Cheng Y, Zhu D, Fu X. 17β-estradiol promotes angiogenesis through non-genomic activation of Smad1 signaling in endometriosis. Vascul Pharmacol 2021; 142:106932. [PMID: 34763099 DOI: 10.1016/j.vph.2021.106932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 09/24/2021] [Accepted: 11/04/2021] [Indexed: 12/19/2022]
Abstract
17β-estradiol (E2) plays a key role in endometriosis through regulation of angiogenesis. Smad1 has been reported to be up-regulated in patients with endometriosis. However, the role of Smad1 in E2-mediated angiogenesis during the development of endometriosis remains to be determined. This study aimed to explore the role of Smad1 in E2-mediated angiogenesis during endometriosis and its underlying mechanisms. Immunofluorescence staining and Western blotting were performed to examine the expression of p-Smad1 in ectopic and control endometrium. Western blotting was used to examine activation of Smad1 signaling in NMECs, EMECs and HUVECs. Tube formation assay was performed to examine the effect of E2 on angiogenesis. Cell proliferation and migration was determined using in real-time by xCELLigence RTCA DP instrument. We found that the expression of p-Smad1 was significantly up-regulated in ectopic endometrium and ectopic intima microvascular endothelial cells. E2 non-genomically stimulated phosphorylation of Smad1 in HUVECs. c-Src and p44/42 MAPK(ERK1/2) signaling pathways are required for E2's induction on Smad1 phosphorylation. Moreover, caveolae is involved in E2-induced Smad1 phosphorylation in vascular endothelial cells. E2 promoted tube formation of vascular endothelial cells through c-Src/ERK1/2/Smad1 signaling pathway. Knockdown of Smad1 expression attenuated E2-induced proliferation and migration of HUVECs. In conclusion, E2 promotes proliferation, migration and tube formation of HUVECs through c-Src/ERK1/2/Smad1 signaling pathway. Our data shed new lights on the mechanisms through which E2 contributes to endometriosis, and may provide novel strategies to treat endometriosis.
Collapse
Affiliation(s)
- Xinran Zhao
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510260, PR China; Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou City, Guangdong Province, 510630, China
| | - Xiaosa Li
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510260, PR China; Department of Gynecology and Obstetrics, Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511518, PR China
| | - Pei Liu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510260, PR China; Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou City, Guangdong Province, 510630, China
| | - Ping Li
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510260, PR China
| | - Xingyan Xu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510260, PR China
| | - Yiwen Chen
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510260, PR China
| | - Yang Cheng
- Department of Gynecology and Obstetrics, Municipal First People's Hospital of Guangzhou, Guangzhou 510180, PR China.
| | - Dongxing Zhu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510260, PR China.
| | - Xiaodong Fu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510260, PR China.
| |
Collapse
|
10
|
Chinetti G, Neels JG. Roles of Nuclear Receptors in Vascular Calcification. Int J Mol Sci 2021; 22:6491. [PMID: 34204304 PMCID: PMC8235358 DOI: 10.3390/ijms22126491] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/10/2021] [Accepted: 06/14/2021] [Indexed: 12/17/2022] Open
Abstract
Vascular calcification is defined as an inappropriate accumulation of calcium depots occurring in soft tissues, including the vascular wall. Growing evidence suggests that vascular calcification is an actively regulated process, sharing similar mechanisms with bone formation, implicating both inhibitory and inducible factors, mediated by osteoclast-like and osteoblast-like cells, respectively. This process, which occurs in nearly all the arterial beds and in both the medial and intimal layers, mainly involves vascular smooth muscle cells. In the vascular wall, calcification can have different clinical consequences, depending on the pattern, localization and nature of calcium deposition. Nuclear receptors are transcription factors widely expressed, activated by specific ligands that control the expression of target genes involved in a multitude of pathophysiological processes, including metabolism, cancer, inflammation and cell differentiation. Some of them act as drug targets. In this review we describe and discuss the role of different nuclear receptors in the control of vascular calcification.
Collapse
Affiliation(s)
- Giulia Chinetti
- Université Côte d’Azur, CHU, INSERM, C3M, 06204 Nice, France;
| | - Jaap G. Neels
- Université Côte d’Azur, INSERM, C3M, 06204 Nice, France
| |
Collapse
|
11
|
Kim HJ, Yoon HJ, Lee DK, Jin X, Che X, Choi JY. The estrogen-related receptor γ modulator, GSK5182, inhibits osteoclast differentiation and accelerates osteoclast apoptosis. BMB Rep 2021. [PMID: 33612148 PMCID: PMC8167243 DOI: 10.5483/bmbrep.2021.54.5.243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Estrogen-related receptor γ (ERRγ), a member of the orphan nuclear receptor family, is a key mediator in cellular metabolic processes and energy homeostasis. Therefore, ERRγ has become an attractive target for treating diverse metabolic disorders. We recently reported that ERRγ acts as a negative regulator of osteoclastogenesis induced by receptor activator of nuclear factor-κB ligand (RANKL). In the present study, we explored the effects of an ERRγ-specific modulator, GSK5182, on ERRγ-regulated osteoclast differentiation and survival. Interestingly, GSK5182 increased ERRγ protein levels much as does GSK4716, which is an ERRγ agonist. GSK5182 inhibited osteoclast generation from bone-marrow-derived macrophages without affecting cytotoxicity. GSK5182 also attenuated RANKL-mediated expression of c-Fos and nuclear factor of activated T-cells cytoplasmic 1 (NFATc1), pivotal transcription factors for osteoclastogenesis. Arrested osteoclast differentiation was associated with reduced RANK expression, but not with the M-CSF receptor, c-Fms. GSK5182 strongly blocked the phosphorylation of IκBα, c-Jun N-terminal kinase, and extracellular signal-regulated kinase in response to RANKL. GSK5182 also suppressed NF-κB promoter activity in a dose-dependent manner. In addition to osteoclastogenesis, GSK5182 accelerated osteoclast apoptosis by caspase-3 activation. Together, these results suggest that GSK5182, a synthetic ERRγ modulator, may have potential in treating disorders related to bone resorption.
Collapse
Affiliation(s)
- Hyun-Ju Kim
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, Korea Mouse Phenotyping Center, KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Hye-Jin Yoon
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, Korea Mouse Phenotyping Center, KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Dong-Kyo Lee
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, Korea Mouse Phenotyping Center, KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Xian Jin
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, Korea Mouse Phenotyping Center, KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Xiangguo Che
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, Korea Mouse Phenotyping Center, KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Je-Yong Choi
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, Korea Mouse Phenotyping Center, KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| |
Collapse
|
12
|
Pyruvate dehydrogenase kinases (PDKs): an overview toward clinical applications. Biosci Rep 2021; 41:228121. [PMID: 33739396 PMCID: PMC8026821 DOI: 10.1042/bsr20204402] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/12/2021] [Accepted: 03/17/2021] [Indexed: 01/01/2023] Open
Abstract
Pyruvate dehydrogenase kinase (PDK) can regulate the catalytic activity of pyruvate decarboxylation oxidation via the mitochondrial pyruvate dehydrogenase complex, and it further links glycolysis with the tricarboxylic acid cycle and ATP generation. This review seeks to elucidate the regulation of PDK activity in different species, mainly mammals, and the role of PDK inhibitors in preventing increased blood glucose, reducing injury caused by myocardial ischemia, and inducing apoptosis of tumor cells. Regulations of PDKs expression or activity represent a very promising approach for treatment of metabolic diseases including diabetes, heart failure, and cancer. The future research and development could be more focused on the biochemical understanding of the diseases, which would help understand the cellular energy metabolism and its regulation by pharmacological effectors of PDKs.
Collapse
|
13
|
Choe N, Kwon DH, Ryu J, Shin S, Cho HJ, Joung H, Eom GH, Ahn Y, Park WJ, Nam KI, Kim YK, Kook H. miR-27a-3p Targets ATF3 to Reduce Calcium Deposition in Vascular Smooth Muscle Cells. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 22:627-639. [PMID: 33230462 PMCID: PMC7578555 DOI: 10.1016/j.omtn.2020.09.030] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 09/23/2020] [Indexed: 01/16/2023]
Abstract
Vascular calcification, the ectopic deposition of calcium in blood vessels, develops in association with various metabolic diseases and atherosclerosis and is an independent predictor of morbidity and mortality associated with these diseases. Herein, we report that reduction of microRNA-27a-3p (miR-27a-3p) causes an increase in activating transcription factor 3 (ATF3), a novel osteogenic transcription factor, in vascular smooth muscle cells. Both microRNA (miRNA) and mRNA microarrays were performed with rat vascular smooth muscle cells, and reciprocally regulated pairs of miRNA and mRNA were selected after bioinformatics analysis. Inorganic phosphate significantly reduced the expression of miR-27a-3p in A10 cells. The transcript level was also reduced in vitamin D3-administered mouse aortas. miR-27a-3p mimic reduced calcium deposition, whereas miR-27a-3p inhibitor increased it. The Atf3 mRNA level was upregulated in a cellular vascular calcification model, and miR-27a-3p reduced the Atf3 mRNA and protein levels. Transfection with Atf3 could recover the miR-27a-3p-induced reduction of calcium deposition. Our results suggest that reduction of miR-27a-3p may contribute to the development of vascular calcification by de-repression of ATF3.
Collapse
Affiliation(s)
- Nakwon Choe
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, Jeollanamdo, Republic of Korea
| | - Duk-Hwa Kwon
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, Jeollanamdo, Republic of Korea
| | - Juhee Ryu
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, Jeollanamdo, Republic of Korea.,Department of Biochemistry, Chonnam National University Medical School, Hwasun, Jeollanamdo, Republic of Korea
| | - Sera Shin
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, Jeollanamdo, Republic of Korea
| | - Hye Jung Cho
- Department of Anatomy, Chonnam National University Medical School, Hwasun, Jeollanamdo, Republic of Korea
| | - Hosouk Joung
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, Jeollanamdo, Republic of Korea
| | - Gwang Hyeon Eom
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, Jeollanamdo, Republic of Korea
| | - Youngkeun Ahn
- Department of Cardiology, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Woo Jin Park
- College of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Kwang-Il Nam
- Department of Anatomy, Chonnam National University Medical School, Hwasun, Jeollanamdo, Republic of Korea
| | - Young-Kook Kim
- Department of Biochemistry, Chonnam National University Medical School, Hwasun, Jeollanamdo, Republic of Korea
| | - Hyun Kook
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, Jeollanamdo, Republic of Korea
| |
Collapse
|
14
|
Min Y, Kim D, Suminda GGD, Zhao X, Kim M, Zhao Y, Son YO. GSK5182, 4-Hydroxytamoxifen Analog, a New Potential Therapeutic Drug for Osteoarthritis. Pharmaceuticals (Basel) 2020; 13:ph13120429. [PMID: 33261216 PMCID: PMC7761342 DOI: 10.3390/ph13120429] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/23/2020] [Accepted: 11/26/2020] [Indexed: 11/16/2022] Open
Abstract
Estrogen-related receptors (ERRs) are the first identified orphan nuclear receptors. The ERR family consists of ERRα, ERRβ, and ERRγ, regulating diverse isoform-specific functions. We have reported the importance of ERRγ in osteoarthritis (OA) pathogenesis. However, therapeutic approaches with ERRγ against OA associated with inflammatory mechanisms remain limited. Herein, we examined the therapeutic potential of a small-molecule ERRγ inverse agonist, GSK5182 (4-hydroxytamoxifen analog), in OA, to assess the relationship between ERRγ expression and pro-inflammatory cytokines in mouse articular chondrocyte cultures. ERRγ expression increased following chondrocyte exposure to various pro-inflammatory cytokines, including interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α. Pro-inflammatory cytokines dose-dependently increased ERRγ protein levels. In mouse articular chondrocytes, adenovirus-mediated ERRγ overexpression upregulated matrix metalloproteinase (MMP)-3 and MMP-13, which participate in cartilage destruction during OA. Adenovirus-mediated ERRγ overexpression in mouse knee joints or ERRγ transgenic mice resulted in OA. In mouse joint tissues, genetic ablation of Esrrg obscured experimental OA. These results indicate that ERRγ is involved in OA pathogenesis. In mouse articular chondrocytes, GSK5182 inhibited pro-inflammatory cytokine-induced catabolic factors. Consistent with the in vitro results, GSK5182 significantly reduced cartilage degeneration in ERRγ-overexpressing mice administered intra-articular Ad-Esrrg. Overall, the ERRγ inverse agonist GSK5182 represents a promising therapeutic small molecule for OA.
Collapse
Affiliation(s)
- Yunhui Min
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju City 63243, Korea; (Y.M.); (G.G.D.S.); (X.Z.)
| | - Dahye Kim
- Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju City 63243, Korea; (D.K.); (M.K.)
| | - Godagama Gamaarachchige Dinesh Suminda
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju City 63243, Korea; (Y.M.); (G.G.D.S.); (X.Z.)
| | - Xiangyu Zhao
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju City 63243, Korea; (Y.M.); (G.G.D.S.); (X.Z.)
| | - Mangeun Kim
- Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju City 63243, Korea; (D.K.); (M.K.)
| | - Yaping Zhao
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, Shanghai 200240, China;
| | - Young-Ok Son
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju City 63243, Korea; (Y.M.); (G.G.D.S.); (X.Z.)
- Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju City 63243, Korea; (D.K.); (M.K.)
- Bio-Health Materials Core-Facility Center, Jeju National University, Jeju City 63243, Korea
- Practical Translational Research Center, Jeju National University, Jeju City 63243, Korea
- Correspondence: ; Tel.: +82-(64)-754-3331
| |
Collapse
|
15
|
Zhang X, Chen J, Meng Q, Li D, Hu FZ, Zhu YQ, Huang YY, Liu YN, Sun L, Liang QH. The protective effects of long non-coding RNA-ANCR on arterial calcification. J Bone Miner Metab 2020; 38:421-431. [PMID: 31974677 DOI: 10.1007/s00774-019-01076-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 12/17/2019] [Indexed: 01/17/2023]
Abstract
INTRODUCTION Arterial calcification is a major factor for cardiovascular events and is characterized by vascular smooth muscle cells (VSMCs) transformed into osteoblast-like cells. Long non-coding RNAs (lncRNA) were recognized as important regulators of diverse biological processes. Previous studies have demonstrated that lncRNAs could regulate the proliferation and apoptosis of VSMCs. LncRNA-ANCR (Anti-differentiation ncRNA) is an essential mediator governing the differentiation of human osteoblast. However, it is unclear whether ANCR could regulate the osteoblastic differentiation of VSMCs. In this study, we determined the effect of ANCR on VSMCs differentiation and arterial calcification. MATERIALS AND METHODS Both cellular and mouse model of arterial calcification were, respectively, established to investigate the role of ANCR in the mechanism of arterial calcification. ANCR overexpressing lentivirus were used to investigate the effects of ANCR on the expression of bone proteins and autophagy-related molecules. RESULTS ANCR could inhibit β-glycerophosphate (β-GP)-induced VSMCs osteoblastic differentiation and mineralization due to decreased expressions of Runt-related transcription factor 2, bone morphogenetic protein-2, and formation of mineralized nodule, and attenuate high calcitriol-induced mice model of arterial calcification. Furthermore, ANCR could significantly increase LC3 and autophagy protein 5 expression in β-GP-stimulated VSMCs, and the effect could be inhibited by 3-methyladenine, a pharmacological inhibitor of autophagy. CONCLUSION ANCR may inhibit the osteoblastic differentiation of VSMCs and attenuate mice arterial calcification through activating autophagy.
Collapse
Affiliation(s)
- Xue Zhang
- Department of Endocrinology, Affiliated Hospital of Jining Medical University, Jining, Shandong, People's Republic of China
| | - Jing Chen
- Department of Endocrinology, Affiliated Hospital of Jining Medical University, Jining, Shandong, People's Republic of China
| | - Qiang Meng
- Department of Endocrinology, Affiliated Hospital of Jining Medical University, Jining, Shandong, People's Republic of China
| | - Dong Li
- Department of Endocrinology, Affiliated Hospital of Jining Medical University, Jining, Shandong, People's Republic of China
| | - Fang-Zhi Hu
- Department of Endocrinology, Affiliated Hospital of Jining Medical University, Jining, Shandong, People's Republic of China
| | - Yu-Qing Zhu
- Department of Endocrinology, Affiliated Hospital of Jining Medical University, Jining, Shandong, People's Republic of China
| | - Yuan-Yuan Huang
- Department of Endocrinology, Affiliated Hospital of Jining Medical University, Jining, Shandong, People's Republic of China
| | - Ya-Nan Liu
- Department of Endocrinology, Affiliated Hospital of Jining Medical University, Jining, Shandong, People's Republic of China
| | - Lin Sun
- Department of Endocrinology, Affiliated Hospital of Jining Medical University, Jining, Shandong, People's Republic of China.
| | - Qiu-Hua Liang
- Department of Endocrinology, Affiliated Hospital of Jining Medical University, Jining, Shandong, People's Republic of China.
| |
Collapse
|
16
|
Tripathi M, Yen PM, Singh BK. Estrogen-Related Receptor Alpha: An Under-Appreciated Potential Target for the Treatment of Metabolic Diseases. Int J Mol Sci 2020; 21:E1645. [PMID: 32121253 PMCID: PMC7084735 DOI: 10.3390/ijms21051645] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 02/24/2020] [Accepted: 02/24/2020] [Indexed: 12/14/2022] Open
Abstract
The estrogen-related receptor alpha (ESRRA) is an orphan nuclear receptor (NR) that significantly influences cellular metabolism. ESRRA is predominantly expressed in metabolically-active tissues and regulates the transcription of metabolic genes, including those involved in mitochondrial turnover and autophagy. Although ESRRA activity is well-characterized in several types of cancer, recent reports suggest that it also has an important role in metabolic diseases. This minireview focuses on the regulation of cellular metabolism and function by ESRRA and its potential as a target for the treatment of metabolic disorders.
Collapse
Affiliation(s)
| | | | - Brijesh Kumar Singh
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore 169857, Singapore; (M.T.); (P.M.Y.)
| |
Collapse
|
17
|
Liu D, Chen L, Dong S, Yang H, Li L, Liu J, Zhou H, Zhou R. Low bone mass is associated with carotid calcification plaque in Chinese postmenopausal women: the Chongqing osteoporosis study. Climacteric 2019; 23:237-244. [PMID: 31612731 DOI: 10.1080/13697137.2019.1671818] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background: The aim of this study was to examine the relationship between low bone mass and the risk of carotid calcification plaques in Chinese postmenopausal women.Methods: We conducted a 5 years prospective study. Bone mineral density (BMD) was measured by dual-energy X-ray absorptiometry (DXA) scanning. Carotid computed tomography angiography (CTA) was conducted using a 64-multidetector row scanner to assess carotid arterial plaque at baseline and during follow-up. Cox proportional hazards analysis was used to evaluate the association of BMD and risk of carotid calcification plaques.Results: Four hundred and eighty-eight women sustained prospective carotid plaques during the follow-up. Women with carotid calcification plaques had low BMD than those with carotid non-calcification plaques. After adjustment for potential confounders, BMD, age, years since menopause, levels of plasma osteoprotegerin and adiponectin, hypertension, diabetes mellitus and hyperlipidemia were independently associated with increased risk of carotid calcification plaques. For carotid calcification plaques, a significant inverse correlation was indicated between BMD and the plaques, and a significant positive correlation was indicated between bone loss and plaques.Conclusions: This study suggested that lower BMD and increased loss rate of BMD were associated with a higher risk of carotid calcification plaques in Chinese postmenopausal women.
Collapse
Affiliation(s)
- D Liu
- Trauma Center, Daping Hospital, Army Medical University, Chongqing, China
| | - L Chen
- Postgraduate School, Bengbu Medical College, Anhui, China
| | - S Dong
- Postgraduate School, Bengbu Medical College, Anhui, China
| | - H Yang
- Department of Neurology, Daping hospital, Army Medical University, Chongqing, China
| | - L Li
- Department of Neurology, Daping hospital, Army Medical University, Chongqing, China
| | - J Liu
- Department of Neurology, Daping hospital, Army Medical University, Chongqing, China
| | - H Zhou
- Department of Neurology, Daping hospital, Army Medical University, Chongqing, China
| | - R Zhou
- Department of Orthopedics, The Orthopedic Surgery Center of Chinese PLA, Southwest Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW This review addresses recent developments in studies of lipid regulation of calcific disease of arteries and cardiac valves, including the role of nuclear receptors. The role of lipid-soluble signals and their receptors is timely given the recent evidence and concerns that lipid-lowering treatment may increase the rate of progression of coronary artery calcification, which has been long associated with increased cardiovascular risk. Understanding the mechanisms will be important for interpreting such clinical information. RECENT FINDINGS New findings support regulation of calcific vascular and valvular disease by nuclear receptors, including the vitamin D receptor, glucocorticoid receptor, nutrient-sensing nuclear receptors (liver X receptor, farnesoid X receptor, and peroxisome proliferator-activated receptors), and sex hormone (estrogen and androgen) receptors. There were two major unexpected findings: first, vitamin D supplementation, which was previously believed to prevent or reduce vascular calcification, showed no cardiovascular benefit in large randomized, controlled trials. Second, both epidemiological studies and coronary intravascular ultrasound studies suggest that treatment with HMG-CoA reductase inhibitors increases progression of coronary artery calcification, raising a question of whether there are mechanically stable and unstable forms of coronary calcification. SUMMARY For clinical practice and research, these new findings offer new fundamental mechanisms for vascular calcification and provide new cautionary insights for therapeutic avenues.
Collapse
Affiliation(s)
- Tamer Sallam
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1679
- Department of Physiology, University of California, Los Angeles, Los Angeles, CA 90095-1679
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095-1679
| | - Yin Tintut
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1679
- Department of Physiology, University of California, Los Angeles, Los Angeles, CA 90095-1679
- Department of Orthopaedic Surgery, University of California, Los Angeles, Los Angeles, CA 90095-1679
| | - Linda L. Demer
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1679
- Department of Physiology, University of California, Los Angeles, Los Angeles, CA 90095-1679
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095-1679
| |
Collapse
|
19
|
Jiménez-González V, Ogalla-García E, García-Quintanilla M, García-Quintanilla A. Deciphering GRINA/Lifeguard1: Nuclear Location, Ca 2+ Homeostasis and Vesicle Transport. Int J Mol Sci 2019; 20:ijms20164005. [PMID: 31426446 PMCID: PMC6719933 DOI: 10.3390/ijms20164005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 07/31/2019] [Accepted: 08/12/2019] [Indexed: 01/31/2023] Open
Abstract
The Glutamate Receptor Ionotropic NMDA-Associated Protein 1 (GRINA) belongs to the Lifeguard family and is involved in calcium homeostasis, which governs key processes, such as cell survival or the release of neurotransmitters. GRINA is mainly associated with membranes of the endoplasmic reticulum, Golgi, endosome, and the cell surface, but its presence in the nucleus has not been explained yet. Here we dissect, with the help of different software tools, the potential roles of GRINA in the cell and how they may be altered in diseases, such as schizophrenia or celiac disease. We describe for the first time that the cytoplasmic N-terminal half of GRINA (which spans a Proline-rich domain) contains a potential DNA-binding sequence, in addition to cleavage target sites and probable PY-nuclear localization sequences, that may enable it to be released from the rest of the protein and enter the nucleus under suitable conditions, where it could participate in the transcription, alternative splicing, and mRNA export of a subset of genes likely involved in lipid and sterol synthesis, ribosome biogenesis, or cell cycle progression. To support these findings, we include additional evidence based on an exhaustive review of the literature and our preliminary data of the protein–protein interaction network of GRINA.
Collapse
Affiliation(s)
| | - Elena Ogalla-García
- Department of Pharmacology, School of Pharmacy, University of Seville, 41012 Seville, Spain
| | - Meritxell García-Quintanilla
- Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville, 41013 Seville, Spain
| | - Albert García-Quintanilla
- Department of Biochemistry and Molecular Biology, School of Pharmacy, University of Seville, 41012 Seville, Spain.
| |
Collapse
|
20
|
Hyaluronan negatively regulates vascular calcification involving BMP2 signaling. J Transl Med 2018; 98:1320-1332. [PMID: 29785051 DOI: 10.1038/s41374-018-0076-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 03/21/2018] [Accepted: 04/16/2018] [Indexed: 01/10/2023] Open
Abstract
Vascular calcification is a highly regulated biological process similar to bone formation involving osteogenic differentiation of vascular smooth muscle cells (VSMCs). Hyaluronan (HA), a major structural component of the extracellular matrix in cartilage, has been shown to inhibit osteoblast differentiation. However, whether HA affects osteogenic differentiation and calcification of VSMCs remains unclear. In the present study, we used in vitro and ex vivo models of vascular calcification to investigate the role of HA in vascular calcification. Both high and low molecular weight HA treatment significantly reduced calcification of rat VSMCs in a dose-dependent manner, as detected by alizarin red staining and calcium content assay. Ex vivo study further confirmed the inhibitory effect of HA on vascular calcification. Similarly, HA treatment decreased ALP activity and expression of bone-related molecules including Runx2, BMP2 and Msx2. By contrast, inhibition of HA synthesis by 4-methylumbelliferone (4MU) promoted calcification of rat VSMCs. In addition, adenovirus-mediated overexpression of HA synthase 2 (HAS2), a major HA synthase in VSMCs, also inhibited calcification of VSMCs, whereas CRISPR/Cas9-mediated HAS2 knockout promoted calcification of rat A10 cells. Furthermore, we found that BMP2 signaling was inhibited in VSMCs after HA treatment. Recombinant BMP2 enhanced high calcium and phosphate-induced VSMC calcification, which can be blocked by HA treatment. Taken together, these findings suggest that HA inhibits vascular calcification involving BMP2 signaling.
Collapse
|
21
|
Zhang JJ, Xi GS. Morphological changes in different caste adult ant specificities of Polyrhachis vicina Roger (Hymenoptera, Formicidae) caused in estrogen-related receptor. Gen Comp Endocrinol 2018; 266:29-37. [PMID: 29746854 DOI: 10.1016/j.ygcen.2018.04.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 03/11/2018] [Accepted: 04/02/2018] [Indexed: 11/29/2022]
Abstract
The estrogen-related receptor (ERR) gene is a member of the nuclear receptor subfamily. Previous studies have indicated that ERR plays important roles in regulating insect growth and development. How ERR is associated with ant caste specificities remains unclear. In this study, we attempted to identify the role of ERR in the regulation of different adult caste specificities of Polyrhachis vicina Roger. Significant variations were detected in the ants including PvERR expressions, some physiological indexes and morphological traits including survival rate, body weight, body length, head width and abdominal appearance by different techniques. The results revealed that when PvERR expressions is up-regulated, boundaries of the abdominal segments were indistinct on the ventral side of the abdomen in males. Down-regulation of PvERR expressions caused abdominal swelling in males and a distended ventral abdomen in females and workers. Variation in PvERR expressions led to a remarkable decline in ant survival rates, particularly for males. These results indicated that different caste adults appeared to have different degrees of sensitivity in physiological response and morphological changes caused by variation in PvERR expressions. Thus, our data demonstrate that PvERR plays an important role in regulating the different adult caste specificities of P. vicina.
Collapse
Affiliation(s)
- Juan-Juan Zhang
- Institute of Zoology, Department of Life Science, Shaanxi Normal University, Xi'an, Shaanxi Province 710119, PR China
| | - Geng-Si Xi
- Institute of Zoology, Department of Life Science, Shaanxi Normal University, Xi'an, Shaanxi Province 710119, PR China.
| |
Collapse
|
22
|
Single extracorporeal shock-wave lithotripsy for proximal ureter stones: Can CT texture analysis technique help predict the therapeutic effect? Eur J Radiol 2018; 107:84-89. [PMID: 30292278 DOI: 10.1016/j.ejrad.2018.08.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 08/13/2018] [Accepted: 08/21/2018] [Indexed: 01/08/2023]
Abstract
PURPOSE To explore whether the computed tomography texture analysis (CTTA) technique can help predict the curative effects of a single extracorporeal shock-wave lithotripsy (ESWL) for proximal ureteral stones. MATERIALS AND METHODS In all, 100 patients with proximal ureteral stone underwent non-enhanced multi-detector computed tomography (MDCT) before ESWL. The patients were divided into success and failure groups. Success of ESWL was defined as the patients being stone-free or having residual stone fragments of ≤2 mm. Traditional characteristics, such as stone size, body mass index (BMI), and skin-to-stone distance (SSD), and CTTA metrics, such as the mean Hounsfield unit (HU) density, entropy, kurtosis, and skewness, were analyzed and compared between two groups by univariate and multivariate logistic regression analyses. Receiver operating characteristic (ROC) curves were generated to determine Youden index-based cutoff values. RESULT Failure of stone removal was observed in 36 patients (36%). Stone height, stone cross-sectional diameter, largest cross-sectional area, stone volume, stone density (mean HU), and CTTA metrics (kurtosis and entropy) were the significant independent predictors of ESWL success on univariate analysis (p < 0.05). On multivariate analysis, mean HU, skewness, and kurtosis were shown to be significant predictors of ESWL success (p < 0.05). In subgroup analysis based on the cutoff value of mean stone density (HU = 857), the only significant independent factor associated with both subgroups was kurtosis (p < 0.05). CONCLUSIONS As a quantitative analysis method, CTTA may be helpful in selecting appropriate ESWL patients. High kurtosis and low mean HU values simultaneously indicate a relatively higher ESWL success rate.
Collapse
|
23
|
Yaguchi T. The endocrine disruptor bisphenol A promotes nuclear ERRγ translocation, facilitating cell proliferation of Grade I endometrial cancer cells via EGF-dependent and EGF-independent pathways. Mol Cell Biochem 2018; 452:41-50. [DOI: 10.1007/s11010-018-3410-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 07/13/2018] [Indexed: 12/16/2022]
|
24
|
Zhu H, Huang L, He Z, Zou Z, Luo Y. Estrogen-related receptor γ regulates expression of 17β-hydroxysteroid dehydrogenase type 1 in fetal growth restriction. Placenta 2018; 67:38-44. [PMID: 29941172 DOI: 10.1016/j.placenta.2018.05.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 05/24/2018] [Accepted: 05/28/2018] [Indexed: 02/01/2023]
Abstract
INTRODUCTION Estrogen-related receptor γ (ERRγ) and 17β-hydroxysteroid dehydrogenase type 1 (HSD17B1) have important roles in cell invasion and in the proliferation of many types of cancer cells. However, it remains unknown whether ERRγ and HSD17B1 contribute to abnormal placental structure and dysfunction which characterize fetal growth restriction (FGR). Therefore, the aim of this study was to investigate the expression profiles of ERRγ and HSD17B1 in placenta tissues affected by FGR and to examine a possible molecular mechanism by which ERRγ is able to regulate HSD17B1 during development of FGR. METHODS Placenta tissues were collected from women affected by FGR (n = 28) and from women with appropriately gestational age (AGA) (n = 30). Relative mRNA and protein levels of ERRγ and HSD17B1 in both groups were assessed by quantitative real-time PCR, immunohistochemistry, and Western blot analyses. The effect of ERRγ on trophoblast function and its associated mechanistic details were studied in the trophoblast cell line, HTR-8/SVneo, which was transfected with small interfering RNA (siRNA) targeting ERRγ. RESULTS Both mRNA and protein levels of ERRγ and HSD17B1 were significantly lower in FGR placentae (P < 0.05). When ERRγ expression was knocked down in HTR-8/SVneo cells with siRNA, invasion and proliferation were inhibited. In addition, HSD17B1 expression was significantly decreased. In dual luciferase reporter assays, ERRγ stimulated transcription of HSD17B1 by targeting the ERRγ response element within its 5'-flanking promoter region. DISCUSSION Aberrant ERRγ expression may contribute to the pathogenesis of FGR by regulating the transcriptional activity of HSD17B1.
Collapse
Affiliation(s)
- Hui Zhu
- Department of Obstetrics & Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Linhuan Huang
- Department of Obstetrics & Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhiming He
- Department of Obstetrics & Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhiyong Zou
- Department of Obstetrics & Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yanmin Luo
- Department of Obstetrics & Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
25
|
Zhang X, Li R, Qin X, Wang L, Xiao J, Song Y, Sheng X, Guo M, Ji X. Sp1 Plays an Important Role in Vascular Calcification Both In Vivo and In Vitro. J Am Heart Assoc 2018; 7:e007555. [PMID: 29572322 PMCID: PMC5907546 DOI: 10.1161/jaha.117.007555] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 02/20/2018] [Indexed: 01/01/2023]
Abstract
BACKGROUND Vascular calcification and increased cardiovascular morbidity and mortality are closely related in patients with end-stage renal disease and diabetes mellitus. Specific protein 1 (Sp1) is a transactivation molecule that plays a crucial role in the regulation of apoptosis, fibrosis, angiogenesis, and other pathological disorders. There is evidence that specific protein 1 (Sp1) directly stimulates the transcription of bone morphogenetic protein 2 (BMP2) and that BMP2 plays a key role in the calcification process in the BMP2-expressing F9 cell model system. Here, we investigated whether Sp1 plays an important role in vascular calcification and its potential regulatory mechanism in vascular calcification. METHODS AND RESULTS In this study, vascular calcification was induced in male Wistar rats by administration of nicotine (25 mg/kg) and vitamin D3 (300 000 IU/kg). These rats were randomly selected for treatment with adenovirus harboring Sp1 knockdown gene or empty virus. The mechanism of Sp1 in vascular smooth muscle cells cultured in high phosphate medium was studied. Based on our findings, the Sp1 gene silencing or inhibition improved calcium deposition, which was partly achieved by inhibiting phenotype switch, apoptosis, and matrix vesicle release of vascular smooth muscle cells. Moreover, Sp1 can activate BMP2 transcription by binding to the Sp1-binding element of the BMP2 promoter. CONCLUSIONS Overall, elevated Sp1 exerts a pro-apoptotic effect, promoting BMP2 transcription and further accumulating vascular calcification. Proper and timely regulation of Sp1 expression may be a potential strategy for treatment of aging, end-stage renal disease, and diabetic-related macrovascular disease treatment.
Collapse
Affiliation(s)
- Xinyu Zhang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, the State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Rui Li
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, the State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xiaoteng Qin
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, the State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Lei Wang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, the State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Jie Xiao
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, the State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yu Song
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, the State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xi Sheng
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, the State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Mengqi Guo
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, the State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xiaoping Ji
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, the State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| |
Collapse
|
26
|
Li J, Carrillo García C, Riedt T, Brandes M, Szczepanski S, Brossart P, Wagner W, Janzen V. Murine hematopoietic stem cell reconstitution potential is maintained by osteopontin during aging. Sci Rep 2018; 8:2833. [PMID: 29434282 PMCID: PMC5809550 DOI: 10.1038/s41598-018-21324-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 01/29/2018] [Indexed: 12/24/2022] Open
Abstract
In adult mammals, hematopoietic stem cells (HSCs) reside in the bone marrow and are in part regulated by the bone marrow microenvironment, called the stem cell niche. We have previously identified the bone marrow morphogen osteopontin (OPN), which is abundantly present in the bone marrow extracellular matrix, as a negative regulator of the size of the HSC pool under physiological conditions. Here, we study the impact of OPN on HSC function during aging using an OPN-knockout mouse model. We show that during aging OPN deficiency is associated with an increase in lymphocytes and a decline in erythrocytes in peripheral blood. In a bone marrow transplantation setting, aged OPN-deficient stem cells show reduced reconstitution ability likely due to insufficient differentiation of HSCs into more mature cells. In serial bone marrow transplantation, aged OPN−/− bone marrow cells fail to adequately reconstitute red blood cells and platelets, resulting in severe anemia and thrombocytopenia as well as premature deaths of recipient mice. Thus, OPN has different effects on HSCs in aged and young animals and is particularly important to maintain stem cell function in aging mice.
Collapse
Affiliation(s)
- Jin Li
- Department of Internal Medicine III, Division of Hematology/Oncology, University of Bonn, Bonn, Germany
| | - Carmen Carrillo García
- Department of Internal Medicine III, Division of Hematology/Oncology, University of Bonn, Bonn, Germany
| | - Tamara Riedt
- Department of Internal Medicine III, Division of Hematology/Oncology, University of Bonn, Bonn, Germany
| | - Maria Brandes
- Department of Internal Medicine III, Division of Hematology/Oncology, University of Bonn, Bonn, Germany
| | - Sabrina Szczepanski
- Department of Internal Medicine III, Division of Hematology/Oncology, University of Bonn, Bonn, Germany
| | - Peter Brossart
- Department of Internal Medicine III, Division of Hematology/Oncology, University of Bonn, Bonn, Germany
| | - Wolfgang Wagner
- Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University Medical School, Aachen, Germany
| | - Viktor Janzen
- Department of Internal Medicine III, Division of Hematology/Oncology, University of Bonn, Bonn, Germany.
| |
Collapse
|
27
|
Zhang K, Zhang Y, Feng W, Chen R, Chen J, Touyz RM, Wang J, Huang H. Interleukin-18 Enhances Vascular Calcification and Osteogenic Differentiation of Vascular Smooth Muscle Cells Through TRPM7 Activation. Arterioscler Thromb Vasc Biol 2017; 37:1933-1943. [DOI: 10.1161/atvbaha.117.309161] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 08/09/2017] [Indexed: 12/31/2022]
Affiliation(s)
- Kun Zhang
- From the Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology (K.Z., Y.Z., W.F., R.C., J.W., H.H.) and Department of Radiation Oncology (J.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China (K.Z., Y.Z., W.F., R.C., J.C., J.W., H.H.); and Institute of Cardiovascular and Medical Sciences, British Heart Foundation (BHF) Glasgow
| | - Yinyin Zhang
- From the Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology (K.Z., Y.Z., W.F., R.C., J.W., H.H.) and Department of Radiation Oncology (J.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China (K.Z., Y.Z., W.F., R.C., J.C., J.W., H.H.); and Institute of Cardiovascular and Medical Sciences, British Heart Foundation (BHF) Glasgow
| | - Weijing Feng
- From the Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology (K.Z., Y.Z., W.F., R.C., J.W., H.H.) and Department of Radiation Oncology (J.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China (K.Z., Y.Z., W.F., R.C., J.C., J.W., H.H.); and Institute of Cardiovascular and Medical Sciences, British Heart Foundation (BHF) Glasgow
| | - Renhua Chen
- From the Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology (K.Z., Y.Z., W.F., R.C., J.W., H.H.) and Department of Radiation Oncology (J.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China (K.Z., Y.Z., W.F., R.C., J.C., J.W., H.H.); and Institute of Cardiovascular and Medical Sciences, British Heart Foundation (BHF) Glasgow
| | - Jie Chen
- From the Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology (K.Z., Y.Z., W.F., R.C., J.W., H.H.) and Department of Radiation Oncology (J.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China (K.Z., Y.Z., W.F., R.C., J.C., J.W., H.H.); and Institute of Cardiovascular and Medical Sciences, British Heart Foundation (BHF) Glasgow
| | - Rhian M. Touyz
- From the Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology (K.Z., Y.Z., W.F., R.C., J.W., H.H.) and Department of Radiation Oncology (J.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China (K.Z., Y.Z., W.F., R.C., J.C., J.W., H.H.); and Institute of Cardiovascular and Medical Sciences, British Heart Foundation (BHF) Glasgow
| | - Jingfeng Wang
- From the Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology (K.Z., Y.Z., W.F., R.C., J.W., H.H.) and Department of Radiation Oncology (J.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China (K.Z., Y.Z., W.F., R.C., J.C., J.W., H.H.); and Institute of Cardiovascular and Medical Sciences, British Heart Foundation (BHF) Glasgow
| | - Hui Huang
- From the Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology (K.Z., Y.Z., W.F., R.C., J.W., H.H.) and Department of Radiation Oncology (J.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China (K.Z., Y.Z., W.F., R.C., J.C., J.W., H.H.); and Institute of Cardiovascular and Medical Sciences, British Heart Foundation (BHF) Glasgow
| |
Collapse
|
28
|
Estrogen-related receptor γ gene ( ESRRG ) rs1890552 A>G polymorphism in a Korean population: Association with urinary prostaglandin F2α concentration and impaired fasting glucose or newly diagnosed type 2 diabetes. DIABETES & METABOLISM 2017; 43:385-388. [DOI: 10.1016/j.diabet.2016.11.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 10/27/2016] [Accepted: 11/06/2016] [Indexed: 02/08/2023]
|
29
|
Kim M, Yoo HJ, Kim M, Seo H, Chae JS, Lee SH, Lee JH. Influence of estrogen-related receptor γ (ESRRG) rs1890552 A > G polymorphism on changes in fasting glucose and arterial stiffness. Sci Rep 2017; 7:9787. [PMID: 28852080 PMCID: PMC5575041 DOI: 10.1038/s41598-017-10192-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 08/04/2017] [Indexed: 02/01/2023] Open
Abstract
To determine the effects of the estrogen-related receptor γ (ESRRG) rs1890552 A > G polymorphism on dietary advice-mediated changes in fasting glucose and arterial stiffness, 374 subjects with normal fasting glucose (NFG; control group, no treatment) and 142 subjects with impaired fasting glucose (IFG group, dietary advice) were followed for 3.5 years. At follow-up, the GG subjects in the IFG group showed a significant reduction in fasting glucose, which was greater than in the AA subjects. A significant association was observed between ESRRG rs1890552 A > G polymorphism and changes in fasting glucose, brachial-ankle pulse wave velocity (ba-PWV), and 8-epi-prostaglandin F2α in the IFG subjects. At baseline, the GG subjects showed a higher ba-PWV than the AA subjects in the IFG group. At the 3.5-year follow-up, subjects with AA or AG showed significant increases in ba-PWV, whereas subjects with GG showed a decrease from baseline. This study suggests that the ESRRG rs1890552 A > G polymorphism may modulate interindividual differences in atrial stiffness, with a reduction in fasting glucose in response to dietary advice in subjects with IFG after a 3.5-year follow-up.
Collapse
Affiliation(s)
- Minjoo Kim
- Research Center for Silver Science, Institute of Symbiotic Life-TECH, Yonsei University, Seoul, 03722, Korea
| | - Hye Jin Yoo
- National Leading Research Laboratory of Clinical Nutrigenetics/Nutrigenomics, Department of Food and Nutrition, College of Human Ecology, Yonsei University, Seoul, 03722, Korea
- Department of Food & Nutrition, Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University, Seoul, 03722, Korea
| | - Minkyung Kim
- Research Center for Silver Science, Institute of Symbiotic Life-TECH, Yonsei University, Seoul, 03722, Korea
| | - Haengok Seo
- National Leading Research Laboratory of Clinical Nutrigenetics/Nutrigenomics, Department of Food and Nutrition, College of Human Ecology, Yonsei University, Seoul, 03722, Korea
- Department of Food & Nutrition, Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University, Seoul, 03722, Korea
| | - Jey Sook Chae
- Research Center for Silver Science, Institute of Symbiotic Life-TECH, Yonsei University, Seoul, 03722, Korea
| | - Sang-Hyun Lee
- Department of Family Practice, National Health Insurance Corporation, Ilsan Hospital, Goyang, 10444, Korea
| | - Jong Ho Lee
- Research Center for Silver Science, Institute of Symbiotic Life-TECH, Yonsei University, Seoul, 03722, Korea.
- National Leading Research Laboratory of Clinical Nutrigenetics/Nutrigenomics, Department of Food and Nutrition, College of Human Ecology, Yonsei University, Seoul, 03722, Korea.
- Department of Food & Nutrition, Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University, Seoul, 03722, Korea.
| |
Collapse
|
30
|
McRobb LS, McGrath KCY, Tsatralis T, Liong EC, Tan JTM, Hughes G, Handelsman DJ, Heather AK. Estrogen Receptor Control of Atherosclerotic Calcification and Smooth Muscle Cell Osteogenic Differentiation. Arterioscler Thromb Vasc Biol 2017; 37:1127-1137. [PMID: 28473445 DOI: 10.1161/atvbaha.117.309054] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 04/19/2017] [Indexed: 12/28/2022]
Abstract
OBJECTIVE Vascular calcification is associated with increased risk of myocardial infarction and stroke. The objective of this work was to examine the ability of 17β-estradiol (E2) to stimulate calcification of vascular smooth muscle cells (VSMC) in vivo, using aged apolipoprotein E-null mice with advanced atherosclerotic lesions, and subsequently to explore underlying mechanisms in vitro. APPROACH AND RESULTS Silastic E2 capsules were implanted into male and female apolipoprotein E-null mice aged 34 weeks. Plaque and calcified area were measured in the aortic sinus and innominate artery after 8 weeks. Immunohistochemical analysis examined expression of the estrogen receptors (estrogen receptor alpha and estrogen receptor beta [ERβ]). VSMC expression of osteogenic markers was examined using digital polymerase chain reaction. Advanced atherosclerotic lesions were present in all mice at the end of 8 weeks. In both male and female mice, E2 increased calcified area in a site-specific manner in the aortic sinus independently of plaque growth or lipid levels and occurred in association with a site-specific decrease in the proportion of ERβ-positive intimal cells. Calcified lesions expressed collagen I and bone sialoprotein, with decreased matrix Gla protein. In vitro, E2 suppressed ERβ expression and increased VSMC mineralization, demonstrating increased collagen I and II, osteocalcin and bone sialoprotein, and reduced matrix Gla protein and osteopontin. Antagonism or RNA silencing of estrogen receptor alpha, ERβ, or both further increased VSMC mineralization. CONCLUSIONS We have demonstrated that E2 can drive calcification in advanced atherosclerotic lesions by promoting the differentiation of VSMC to osteoblast-like cells, a process which is augmented by inhibition of estrogen receptor alpha or ERβ activity.
Collapse
MESH Headings
- Animals
- Apolipoproteins E/deficiency
- Apolipoproteins E/genetics
- Atherosclerosis/chemically induced
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Calcium-Binding Proteins/metabolism
- Cattle
- Cell Differentiation/drug effects
- Cells, Cultured
- Collagen/metabolism
- Disease Models, Animal
- Drug Implants
- Estradiol/administration & dosage
- Estradiol/toxicity
- Estrogen Receptor Antagonists/pharmacology
- Estrogen Receptor alpha/agonists
- Estrogen Receptor alpha/genetics
- Estrogen Receptor alpha/metabolism
- Estrogen Receptor beta/agonists
- Estrogen Receptor beta/genetics
- Estrogen Receptor beta/metabolism
- Extracellular Matrix Proteins/metabolism
- Female
- Genetic Predisposition to Disease
- Humans
- Integrin-Binding Sialoprotein/metabolism
- Male
- Mice, Knockout
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Neointima
- Osteocalcin/metabolism
- Osteogenesis/drug effects
- Osteopontin/metabolism
- Phenotype
- Plaque, Atherosclerotic
- RNA Interference
- Signal Transduction/drug effects
- Transfection
- Vascular Calcification/chemically induced
- Vascular Calcification/genetics
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
- Matrix Gla Protein
Collapse
Affiliation(s)
- Lucinda S McRobb
- From the Heart Research Institute, Sydney, New South Wales, Australia (L.S.M., K.C.Y.M., T.T., E.C.L., J.T.M.T.); Department of Clinical Medicine, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales, Australia (L.S.M.); School of Life Sciences, Faculty of Science, University of Technology Sydney, New South Wales, Australia (K.C.Y.M.); Sydney Medical School (J.T.M.T.) and ANZAC Research Institute (D.J.H.), University of Sydney, New South Wales, Australia; and Department of Physiology, Otago School of Medical Sciences (G.H., A.K.H.) and HeartOtago (A.K.H.), University of Otago, Dunedin, New Zealand
| | - Kristine C Y McGrath
- From the Heart Research Institute, Sydney, New South Wales, Australia (L.S.M., K.C.Y.M., T.T., E.C.L., J.T.M.T.); Department of Clinical Medicine, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales, Australia (L.S.M.); School of Life Sciences, Faculty of Science, University of Technology Sydney, New South Wales, Australia (K.C.Y.M.); Sydney Medical School (J.T.M.T.) and ANZAC Research Institute (D.J.H.), University of Sydney, New South Wales, Australia; and Department of Physiology, Otago School of Medical Sciences (G.H., A.K.H.) and HeartOtago (A.K.H.), University of Otago, Dunedin, New Zealand
| | - Tania Tsatralis
- From the Heart Research Institute, Sydney, New South Wales, Australia (L.S.M., K.C.Y.M., T.T., E.C.L., J.T.M.T.); Department of Clinical Medicine, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales, Australia (L.S.M.); School of Life Sciences, Faculty of Science, University of Technology Sydney, New South Wales, Australia (K.C.Y.M.); Sydney Medical School (J.T.M.T.) and ANZAC Research Institute (D.J.H.), University of Sydney, New South Wales, Australia; and Department of Physiology, Otago School of Medical Sciences (G.H., A.K.H.) and HeartOtago (A.K.H.), University of Otago, Dunedin, New Zealand
| | - Eleanore C Liong
- From the Heart Research Institute, Sydney, New South Wales, Australia (L.S.M., K.C.Y.M., T.T., E.C.L., J.T.M.T.); Department of Clinical Medicine, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales, Australia (L.S.M.); School of Life Sciences, Faculty of Science, University of Technology Sydney, New South Wales, Australia (K.C.Y.M.); Sydney Medical School (J.T.M.T.) and ANZAC Research Institute (D.J.H.), University of Sydney, New South Wales, Australia; and Department of Physiology, Otago School of Medical Sciences (G.H., A.K.H.) and HeartOtago (A.K.H.), University of Otago, Dunedin, New Zealand
| | - Joanne T M Tan
- From the Heart Research Institute, Sydney, New South Wales, Australia (L.S.M., K.C.Y.M., T.T., E.C.L., J.T.M.T.); Department of Clinical Medicine, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales, Australia (L.S.M.); School of Life Sciences, Faculty of Science, University of Technology Sydney, New South Wales, Australia (K.C.Y.M.); Sydney Medical School (J.T.M.T.) and ANZAC Research Institute (D.J.H.), University of Sydney, New South Wales, Australia; and Department of Physiology, Otago School of Medical Sciences (G.H., A.K.H.) and HeartOtago (A.K.H.), University of Otago, Dunedin, New Zealand
| | - Gillian Hughes
- From the Heart Research Institute, Sydney, New South Wales, Australia (L.S.M., K.C.Y.M., T.T., E.C.L., J.T.M.T.); Department of Clinical Medicine, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales, Australia (L.S.M.); School of Life Sciences, Faculty of Science, University of Technology Sydney, New South Wales, Australia (K.C.Y.M.); Sydney Medical School (J.T.M.T.) and ANZAC Research Institute (D.J.H.), University of Sydney, New South Wales, Australia; and Department of Physiology, Otago School of Medical Sciences (G.H., A.K.H.) and HeartOtago (A.K.H.), University of Otago, Dunedin, New Zealand
| | - David J Handelsman
- From the Heart Research Institute, Sydney, New South Wales, Australia (L.S.M., K.C.Y.M., T.T., E.C.L., J.T.M.T.); Department of Clinical Medicine, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales, Australia (L.S.M.); School of Life Sciences, Faculty of Science, University of Technology Sydney, New South Wales, Australia (K.C.Y.M.); Sydney Medical School (J.T.M.T.) and ANZAC Research Institute (D.J.H.), University of Sydney, New South Wales, Australia; and Department of Physiology, Otago School of Medical Sciences (G.H., A.K.H.) and HeartOtago (A.K.H.), University of Otago, Dunedin, New Zealand
| | - Alison K Heather
- From the Heart Research Institute, Sydney, New South Wales, Australia (L.S.M., K.C.Y.M., T.T., E.C.L., J.T.M.T.); Department of Clinical Medicine, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales, Australia (L.S.M.); School of Life Sciences, Faculty of Science, University of Technology Sydney, New South Wales, Australia (K.C.Y.M.); Sydney Medical School (J.T.M.T.) and ANZAC Research Institute (D.J.H.), University of Sydney, New South Wales, Australia; and Department of Physiology, Otago School of Medical Sciences (G.H., A.K.H.) and HeartOtago (A.K.H.), University of Otago, Dunedin, New Zealand.
| |
Collapse
|
31
|
Misra J, Kim DK, Choi HS. ERRγ: a Junior Orphan with a Senior Role in Metabolism. Trends Endocrinol Metab 2017; 28:261-272. [PMID: 28209382 DOI: 10.1016/j.tem.2016.12.005] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 12/18/2016] [Accepted: 12/22/2016] [Indexed: 01/01/2023]
Abstract
Estrogen-related receptor (ERR)γ is an orphan nuclear hormone receptor that belongs to the ERR subfamily of transcription factors. No endogenous ligand has been identified to date. ERRγ possesses ligand-independent transcriptional activity that is regulated by co-regulator interactions, and post-translational modifications (PTMs). Recent data from animal models have established ERRγ as a crucial mediator of multiple endocrine and metabolic signals. ERRγ plays important roles in pathological conditions such as insulin resistance, alcoholic liver injury, and cardiac hypertrophy, and controls energy metabolism in the heart, skeletal muscle, and pancreatic β cells. These findings corroborate the importance of ERRγ in metabolic homeostasis, and suggest that ERRγ is a good target for the treatment of metabolic diseases.
Collapse
Affiliation(s)
- Jagannath Misra
- National Creative Research Initiatives Center for Nuclear Receptor Signals and Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Don-Kyu Kim
- Department of Molecular Biotechnology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Hueng-Sik Choi
- National Creative Research Initiatives Center for Nuclear Receptor Signals and Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea.
| |
Collapse
|
32
|
Sun M, Chang Q, Xin M, Wang Q, Li H, Qian J. Endogenous bone morphogenetic protein 2 plays a role in vascular smooth muscle cell calcification induced by interleukin 6 in vitro. Int J Immunopathol Pharmacol 2017; 30:227-237. [PMID: 28134597 PMCID: PMC5815263 DOI: 10.1177/0394632016689571] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Systemic inflammation is involved in vascular calcification and cardiovascular disease which is the leading cause of mortality in rheumatoid arthritis (RA). A high level of serum interleukin (IL)-6 plays a key role in local and systemic inflammation in RA. However, the underlying mechanisms remain unclear. We established a human umbilical artery smooth muscle cell (HUASMC) culturing method to investigate the possible role of IL-6 on vascular calcification. HUASMCs were obtained from umbilical arteries of healthy neonates. To detect calcification effects, HUASMCs were treated with (experimental group) or without (control group) recombinant human (rh) IL-6. The calcium deposition stain and calcium concentrations were measured, as well as the mRNA and protein levels of the regulating factor of osteogenic differentiation-bone morphogenetic protein (BMP) 2 and those calcifying related molecules including bone-specific alkaline phosphatase (BAP), osteoprotegerin (OPG), and osteopontin (OPN). Our study showed that rhIL-6 induced calcification of HUASMCs in a time- and dose-dependent manner, and upregulated expressions of BMP2, BAP, OPG, and OPN of HUASMCs. We then used the anti-BMP2 siRNA to knockdown the expression of endogenous BMP2 to confirm its role. HUASMCs were transfected with negative siRNA (control group) or the valid anti-BMP2 siRNA (experimental group) before they were treated with rhIL-6. Cells transfected with negative siRNA without IL-6 stimulating served as the blank group. The results showed that anti-BMP2 siRNA markedly decreased expressions of BMP2, BAP, OPG, and OPN, and also partly reduced the calcification of HUASMCs induced by rhIL-6. Collectively, according to our study, rhIL-6 could induce the extracellular calcification and osteogenic differentiation of human artery smooth muscle cells through upregulating endogenous BMP2 in vitro. This may be one of the underlying mechanisms of the overwhelming vascular calcification in RA.
Collapse
Affiliation(s)
- Mingshu Sun
- 1 Department of Rheumatology and Clinical Immunology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qing Chang
- 2 Department of Cardiac surgery, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Miaomiao Xin
- 1 Department of Rheumatology and Clinical Immunology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qian Wang
- 1 Department of Rheumatology and Clinical Immunology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hua Li
- 1 Department of Rheumatology and Clinical Immunology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jiaqi Qian
- 3 Department of Nephrology, Renji Hospital, Shanghai Jiaotong University Medical College, Shanghai, China
| |
Collapse
|
33
|
Kim J, Woo SY, Im CY, Yoo EK, Lee S, Kim HJ, Hwang HJ, Cho JH, Lee WS, Yoon H, Kim S, Kwon OB, Hwang H, Kim KH, Jeon JH, Singh TD, Kim SW, Hwang SY, Choi HS, Lee IK, Kim SH, Jeon YH, Chin J, Cho SJ. Insights of a Lead Optimization Study and Biological Evaluation of Novel 4-Hydroxytamoxifen Analogs as Estrogen-Related Receptor γ (ERRγ) Inverse Agonists. J Med Chem 2016; 59:10209-10227. [DOI: 10.1021/acs.jmedchem.6b01204] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Jina Kim
- New
Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Korea
| | - Seo Yeon Woo
- New
Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Korea
| | - Chun Young Im
- New
Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Korea
| | - Eun Kyung Yoo
- Leading-Edge
Research Center for Drug Discovery and Development for Diabetes and
Metabolic Disease, Kyungpook National University Hospital, Daegu 41404, Korea
| | - Seungmi Lee
- Leading-Edge
Research Center for Drug Discovery and Development for Diabetes and
Metabolic Disease, Kyungpook National University Hospital, Daegu 41404, Korea
| | - Hyo-Ji Kim
- New
Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Korea
| | - Hee-Jong Hwang
- New
Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Korea
| | - Joong-heui Cho
- New
Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Korea
| | - Won Seok Lee
- New
Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Korea
| | - Heeseok Yoon
- New
Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Korea
| | - Shinae Kim
- New
Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Korea
| | - Oh-bin Kwon
- New
Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Korea
| | - Hayoung Hwang
- New
Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Korea
| | - Kyung-Hee Kim
- New
Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Korea
| | - Jae-Han Jeon
- Leading-Edge
Research Center for Drug Discovery and Development for Diabetes and
Metabolic Disease, Kyungpook National University Hospital, Daegu 41404, Korea
- Department
of Internal Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Thoudam Debraj Singh
- Department
of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Sang Wook Kim
- Korea Bio-Medical Science Institute, Seoul 06106, Republic of Korea
| | - Sung Yeoun Hwang
- Korea Bio-Medical Science Institute, Seoul 06106, Republic of Korea
| | - Hueng-Sik Choi
- National
Creative Research Initiatives Center for Nuclear Receptor Signals
and Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - In-Kyu Lee
- Leading-Edge
Research Center for Drug Discovery and Development for Diabetes and
Metabolic Disease, Kyungpook National University Hospital, Daegu 41404, Korea
- Department
of Internal Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Seong Heon Kim
- New
Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Korea
| | - Yong Hyun Jeon
- Leading-Edge
Research Center for Drug Discovery and Development for Diabetes and
Metabolic Disease, Kyungpook National University Hospital, Daegu 41404, Korea
- Department
of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Jungwook Chin
- New
Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Korea
| | - Sung Jin Cho
- New
Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Korea
- Leading-Edge
Research Center for Drug Discovery and Development for Diabetes and
Metabolic Disease, Kyungpook National University Hospital, Daegu 41404, Korea
| |
Collapse
|
34
|
Nanao-Hamai M, Son BK, Hashizume T, Ogawa S, Akishita M. Protective effects of estrogen against vascular calcification via estrogen receptor α-dependent growth arrest-specific gene 6 transactivation. Biochem Biophys Res Commun 2016; 480:429-435. [DOI: 10.1016/j.bbrc.2016.10.066] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 10/19/2016] [Indexed: 11/24/2022]
|
35
|
Zhang L, Wong J, Vanacker JM. The estrogen-related receptors (ERRs): potential targets against bone loss. Cell Mol Life Sci 2016; 73:3781-7. [PMID: 27514376 PMCID: PMC11108346 DOI: 10.1007/s00018-016-2328-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 08/04/2016] [Indexed: 01/20/2023]
Abstract
Bone loss and the resulting skeletal fragility is induced by several pathological or natural conditions, the most prominent of which being aging as well as the decreased levels of circulating estrogens in post-menopause females. To date, most treatments against bone loss aim at preventing excess bone resorption. We here summarize data indicating that the estrogen-related receptors (ERRs) α and γ prevent bone formation. Inhibiting these receptors may thus constitute an anabolic approach by increasing bone formation.
Collapse
Affiliation(s)
- Ling Zhang
- Institut de Génomique Fonctionnelle de Lyon, CNRS UMR5242, Université de Lyon, Université Lyon I, Ecole Normale Supérieure de Lyon, Lyon, France
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Jiemin Wong
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Jean-Marc Vanacker
- Institut de Génomique Fonctionnelle de Lyon, CNRS UMR5242, Université de Lyon, Université Lyon I, Ecole Normale Supérieure de Lyon, Lyon, France.
| |
Collapse
|
36
|
St Hilaire C, Liberman M, Miller JD. Bidirectional Translation in Cardiovascular Calcification. Arterioscler Thromb Vasc Biol 2016; 36:e19-24. [PMID: 26912744 DOI: 10.1161/atvbaha.115.307056] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Cynthia St Hilaire
- From the Department of Medicine, Division of Cardiology & Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA (C.S.H.); Departments of Critical Care Medicine and Cardiology, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil (M.L.); and Departments of Surgery and Physiology & BME, Mayo Clinic, Rochester, MN (J.D.M)
| | - Marcel Liberman
- From the Department of Medicine, Division of Cardiology & Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA (C.S.H.); Departments of Critical Care Medicine and Cardiology, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil (M.L.); and Departments of Surgery and Physiology & BME, Mayo Clinic, Rochester, MN (J.D.M)
| | - Jordan D Miller
- From the Department of Medicine, Division of Cardiology & Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA (C.S.H.); Departments of Critical Care Medicine and Cardiology, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil (M.L.); and Departments of Surgery and Physiology & BME, Mayo Clinic, Rochester, MN (J.D.M)
| | | |
Collapse
|
37
|
Genetic Variants in the Bone Morphogenic Protein Gene Family Modify the Association between Residential Exposure to Traffic and Peripheral Arterial Disease. PLoS One 2016; 11:e0152670. [PMID: 27082954 PMCID: PMC4833382 DOI: 10.1371/journal.pone.0152670] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 03/17/2016] [Indexed: 02/06/2023] Open
Abstract
There is a growing literature indicating that genetic variants modify many of the associations between environmental exposures and clinical outcomes, potentially by increasing susceptibility to these exposures. However, genome-scale investigations of these interactions have been rarely performed particularly in the case of air pollution exposures. We performed race-stratified genome-wide gene-environment interaction association studies on European-American (EA, N = 1623) and African-American (AA, N = 554) cohorts to investigate the joint influence of common single nucleotide polymorphisms (SNPs) and residential exposure to traffic (“traffic exposure”)—a recognized vascular disease risk factor—on peripheral arterial disease (PAD). Traffic exposure was estimated via the distance from the primary residence to the nearest major roadway, defined as the nearest limited access highways or major arterial. The rs755249-traffic exposure interaction was associated with PAD at a genome-wide significant level (P = 2.29x10-8) in European-Americans. Rs755249 is located in the 3’ untranslated region of BMP8A, a member of the bone morphogenic protein (BMP) gene family. Further investigation revealed several variants in BMP genes associated with PAD via an interaction with traffic exposure in both the EA and AA cohorts; this included interactions with non-synonymous variants in BMP2, which is regulated by air pollution exposure. The BMP family of genes is linked to vascular growth and calcification and is a novel gene family for the study of PAD pathophysiology. Further investigation of BMP8A using the Genotype Tissue Expression Database revealed multiple variants with nominally significant (P < 0.05) interaction P-values in our EA cohort were significant BMP8A eQTLs in tissue types highlight relevant for PAD such as rs755249 (tibial nerve, eQTL P = 3.6x10-6) and rs1180341 (tibial artery, eQTL P = 5.3x10-6). Together these results reveal a novel gene, and possibly gene family, associated with PAD via an interaction with traffic air pollution exposure. These results also highlight the potential for interactions studies, particularly at the genome scale, to reveal novel biology linking environmental exposures to clinical outcomes.
Collapse
|
38
|
Leem J, Lee IK. Mechanisms of Vascular Calcification: The Pivotal Role of Pyruvate Dehydrogenase Kinase 4. Endocrinol Metab (Seoul) 2016; 31:52-61. [PMID: 26996423 PMCID: PMC4803561 DOI: 10.3803/enm.2016.31.1.52] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 02/19/2016] [Accepted: 02/23/2016] [Indexed: 01/08/2023] Open
Abstract
Vascular calcification, abnormal mineralization of the vessel wall, is frequently associated with aging, atherosclerosis, diabetes mellitus, and chronic kidney disease. Vascular calcification is a key risk factor for many adverse clinical outcomes, including ischemic cardiac events and subsequent cardiovascular mortality. Vascular calcification was long considered to be a passive degenerative process, but it is now recognized as an active and highly regulated process similar to bone formation. However, despite numerous studies on the pathogenesis of vascular calcification, the mechanisms driving this process remain poorly understood. Pyruvate dehydrogenase kinases (PDKs) play an important role in the regulation of cellular metabolism and mitochondrial function. Recent studies show that PDK4 is an attractive therapeutic target for the treatment of various metabolic diseases. In this review, we summarize our current knowledge regarding the mechanisms of vascular calcification and describe the role of PDK4 in the osteogenic differentiation of vascular smooth muscle cells and development of vascular calcification. Further studies aimed at understanding the molecular mechanisms of vascular calcification will be critical for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Jaechan Leem
- Department of Immunology, Catholic University of Daegu School of Medicine, Daegu, Korea
| | - In Kyu Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, Korea
- BK21 PLUS KNU Biomedical Convergence Program, Kyungpook National University, Daegu, Korea.
| |
Collapse
|