1
|
Florido MHC, Ziats NP. Endothelial dysfunction and cardiovascular diseases: The role of human induced pluripotent stem cells and tissue engineering. J Biomed Mater Res A 2024; 112:1286-1304. [PMID: 38230548 DOI: 10.1002/jbm.a.37669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 12/07/2023] [Accepted: 01/02/2024] [Indexed: 01/18/2024]
Abstract
Cardiovascular disease (CVD) remains to be the leading cause of death globally today and therefore the need for the development of novel therapies has become increasingly important in the cardiovascular field. The mechanism(s) behind the pathophysiology of CVD have been laboriously investigated in both stem cell and bioengineering laboratories. Scientific breakthroughs have paved the way to better mimic cell types of interest in recent years, with the ability to generate any cell type from reprogrammed human pluripotent stem cells. Mimicking the native extracellular matrix using both organic and inorganic biomaterials has allowed full organs to be recapitulated in vitro. In this paper, we will review techniques from both stem cell biology and bioengineering which have been fruitfully combined and have fueled advances in the cardiovascular disease field. We will provide a brief introduction to CVD, reviewing some of the recent studies as related to the role of endothelial cells and endothelial cell dysfunction. Recent advances and the techniques widely used in both bioengineering and stem cell biology will be discussed, providing a broad overview of the collaboration between these two fields and their overall impact on tissue engineering in the cardiovascular devices and implications for treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Mary H C Florido
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
- Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, USA
- Harvard Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA
| | - Nicholas P Ziats
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
- Departments of Biomedical Engineering and Anatomy, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
2
|
Lee AS, Kim Y, Hur HJ, Lee SH, Sung MJ. Chrysanthemum coronarium L. Extract Attenuates Homocysteine-Induced Vascular Inflammation in Vascular Smooth Muscle Cells. J Med Food 2023; 26:869-876. [PMID: 38010869 DOI: 10.1089/jmf.2023.k.0058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2023] Open
Abstract
Hyperhomocysteinemia is a main risk factor for phenotypic modulation of vascular smooth muscle cells (VSMCs) and atherosclerosis. Phenotypic switching and proliferation of VSMCs are related to the progression of vascular inflammation. Chrysanthemum coronarium L. is a leafy vegetable with various biological functions, such as antioxidative, anti-inflammatory, and antiproliferative effects. In this study, we aimed to identify the mechanisms underlying the therapeutic and preventive effects of C. coronarium L. extract (CC) in regulating homocysteine (Hcy)-induced vascular inflammation in human aortic VSMCs. CC did not exhibit cytotoxicity and inhibited Hcy-stimulated VSMC proliferation and migration. In addition, CC promoted Hcy-induced expression of VSMC contractile phenotype proteins, including alpha-smooth muscle actin, calponin, and smooth muscle 22α. CC also decreased Hcy-induced accumulation of reactive oxygen species and expression of inflammatory markers nicotinamide adenine dinucleotide phosphate oxidase-4 and soluble epoxide hydrolase. These results showed that CC attenuates Hcy-induced inflammatory responses, highlighting its potential as a therapeutic or preventive target for Hcy-induced vascular inflammation.
Collapse
Affiliation(s)
- Ae Sin Lee
- Research Group of Natural Materials and Metabolism, Food Functionality Research, Korea Food Research Institute, Wanju-gun, Jeollabuk-do, Republic of Korea
| | - Yiseul Kim
- Research Group of Natural Materials and Metabolism, Food Functionality Research, Korea Food Research Institute, Wanju-gun, Jeollabuk-do, Republic of Korea
| | - Haeng Jeon Hur
- Research Group of Natural Materials and Metabolism, Food Functionality Research, Korea Food Research Institute, Wanju-gun, Jeollabuk-do, Republic of Korea
| | - Sang-Hee Lee
- Research Group of Natural Materials and Metabolism, Food Functionality Research, Korea Food Research Institute, Wanju-gun, Jeollabuk-do, Republic of Korea
| | - Mi Jeong Sung
- Research Group of Natural Materials and Metabolism, Food Functionality Research, Korea Food Research Institute, Wanju-gun, Jeollabuk-do, Republic of Korea
| |
Collapse
|
3
|
Chen W, Zhang Y, Li R, Huang W, Wei X, Zeng D, Liang Y, Zeng Y, Chen M, Zhang L, Gao W, Zhu Y, Li Y, Zhang G. Notch3 Transactivates Glycogen Synthase Kinase-3-Beta and Inhibits Epithelial-to-Mesenchymal Transition in Breast Cancer Cells. Cells 2022; 11:cells11182872. [PMID: 36139447 PMCID: PMC9497076 DOI: 10.3390/cells11182872] [Citation(s) in RCA: 3] [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: 05/26/2022] [Revised: 08/10/2022] [Accepted: 08/19/2022] [Indexed: 02/05/2023] Open
Abstract
As a critical transformational process in the attributes of epithelial cells, epithelial-to-mesenchymal transition (EMT) is involved in tumor invasion, metastasis, and resistance to treatment, which contributes to the ultimate death of some patients with breast cancer. Glycogen synthase kinase-3-beta (GSK3β) is thought to be an EMT suppressor that down-regulates the protein, snail, a zinc finger transcription inhibitor, and regulates E-cadherin expression and the Wnt signaling pathway. Our previous studies have shown that Notch3 also inhibits EMT in breast cancer. In mammary gland cells, GSK3β physically bound and phosphorylated the intracellular domain of two Notch paralogs: N1ICD was positively regulated, but N2ICD was negatively regulated; however, the relationship between Notch3, GSK3β, and EMT in breast cancer is still unclear and crosstalk between Notch3 and GSK3β has not been widely investigated. In this study, we revealed that Notch3 was an essential antagonist of EMT in breast cancer cells by transcriptionally upregulating GSK3β. In breast cancer, MCF-7 and MDA-MB-231 cell lines, the silencing of Notch3 reduced GSK3β expression, which is sufficient to induce EMT. Conversely, ectopic Notch3 expression re-activated GSK3β and E-cadherin. Mechanistically, Notch3 can bind to the GSK3β promoter directly and activate GSK3β transcription. In human breast cancer samples, Notch3 expression is positively associated with GSK3β (r = 0.416, p = 0.001); moreover, high expressions of Notch3 and GSK3β mRNA are correlated to better relapse-free survival in all breast cancer patients via analysis in "the Kaplan-Meier plotter" database. In summary, our preliminary results suggested that Notch3 might inhibit EMT by trans-activating GSK3β in breast cancer cells. The suppression of Notch3 expression may contribute to EMT by transcriptionally downregulating GSK3β in breast cancer.
Collapse
Affiliation(s)
- Weiling Chen
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Key Laboratory for Endocrine-Related Cancer Precision Medicine of Xiamen, No. 2000 Xiang’an East Road, Xiamen 361101, China
- Xiamen Research Center of Clinical Medicine in Breast & Thyroid Cancers, No. 2000 Xiang’an East Road, Xiamen 361101, China
| | - Yongqu Zhang
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Key Laboratory for Endocrine-Related Cancer Precision Medicine of Xiamen, No. 2000 Xiang’an East Road, Xiamen 361101, China
- Xiamen Research Center of Clinical Medicine in Breast & Thyroid Cancers, No. 2000 Xiang’an East Road, Xiamen 361101, China
| | - Ronghui Li
- Department of Medical Oncology, Xiang’an Hospital of Xiamen University, No. 2000 Xiang’an East Road, Xiamen 361101, China
| | - Wenhe Huang
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Key Laboratory for Endocrine-Related Cancer Precision Medicine of Xiamen, No. 2000 Xiang’an East Road, Xiamen 361101, China
- Xiamen Research Center of Clinical Medicine in Breast & Thyroid Cancers, No. 2000 Xiang’an East Road, Xiamen 361101, China
| | - Xiaolong Wei
- Department of Pathology, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou 515041, China
| | - De Zeng
- Department of Medical Oncology, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou 515041, China
| | - Yuanke Liang
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Shantou University Medical College, No. 57 Changping Road, Shantou 515041, China
| | - Yunzhu Zeng
- Department of Pathology, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou 515041, China
| | - Min Chen
- Key Laboratory for Endocrine-Related Cancer Precision Medicine of Xiamen, No. 2000 Xiang’an East Road, Xiamen 361101, China
| | - Lixin Zhang
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Key Laboratory for Endocrine-Related Cancer Precision Medicine of Xiamen, No. 2000 Xiang’an East Road, Xiamen 361101, China
- Xiamen Research Center of Clinical Medicine in Breast & Thyroid Cancers, No. 2000 Xiang’an East Road, Xiamen 361101, China
| | - Wenliang Gao
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Key Laboratory for Endocrine-Related Cancer Precision Medicine of Xiamen, No. 2000 Xiang’an East Road, Xiamen 361101, China
- Xiamen Research Center of Clinical Medicine in Breast & Thyroid Cancers, No. 2000 Xiang’an East Road, Xiamen 361101, China
| | - Yuanyuan Zhu
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Key Laboratory for Endocrine-Related Cancer Precision Medicine of Xiamen, No. 2000 Xiang’an East Road, Xiamen 361101, China
- Xiamen Research Center of Clinical Medicine in Breast & Thyroid Cancers, No. 2000 Xiang’an East Road, Xiamen 361101, China
| | - Yaochen Li
- Department of Central Lab, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou 515041, China
- Correspondence: (Y.L.); (G.Z.)
| | - Guojun Zhang
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Key Laboratory for Endocrine-Related Cancer Precision Medicine of Xiamen, No. 2000 Xiang’an East Road, Xiamen 361101, China
- Xiamen Research Center of Clinical Medicine in Breast & Thyroid Cancers, No. 2000 Xiang’an East Road, Xiamen 361101, China
- Correspondence: (Y.L.); (G.Z.)
| |
Collapse
|
4
|
Davis CM, Zhang WH, Bah TM, Roese NE, Allen EM, Leung P, Boutros SJ, Marzulla T, Patel E, Nie X, Alkayed FN, Huang JH, Jensen MA, Raber J, Pike MM, Alkayed NJ. Age-dependent cognitive impairment, hydrocephalus and leukocyte infiltration in transgenic mice with endothelial expression of human EPHX2. NPJ AGING 2022; 8:9. [PMID: 35927273 PMCID: PMC9256583 DOI: 10.1038/s41514-022-00090-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 05/31/2022] [Indexed: 12/30/2022]
Abstract
Soluble epoxide hydrolase (sEH) is upregulated in microvascular endothelium of human brain with vascular cognitive impairment (VCI). Transgenic endothelial expression of human sEH in mice (Tie2hsEH) induces endothelial dysfunction (ED), a pathogenetic mechanism of VCI. We sought to determine if endothelial upregulation of sEH is sufficient to cause cognitive impairment, and if cognitive impairment due to chronic hypoperfusion induced by unilateral common carotid artery occlusion (CCAO) is exacerbated in Tie2hsEH mice. Behavioral performance was assessed by the open field, rotarod, novel object, Morris water maze and fear conditioning tests. Cerebral blood flow and brain morphology were evaluated by MRI, and inflammatory changes investigated using immunohistochemistry and flow cytometry. We demonstrate that transgenic endothelial expression of sEH is sufficient to induce cognitive impairment, associated with leukocyte infiltration, brain atrophy and accelerated, age-dependent ventriculomegaly, identifying ED and sEH upregulation as potential underlying mechanisms and therapeutic targets for VCI.
Collapse
Affiliation(s)
- Catherine M Davis
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Wenri H Zhang
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Thierno M Bah
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Natalie E Roese
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Elyse M Allen
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Philberta Leung
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Sydney J Boutros
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Tessa Marzulla
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Esha Patel
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Xiao Nie
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Farah N Alkayed
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Justin H Huang
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Michael A Jensen
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA
- Departments of Neurology and Radiation Medicine, Division of Neuroscience, ONPRC, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Martin M Pike
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Nabil J Alkayed
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, 97239, USA.
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, 97239, USA.
| |
Collapse
|
5
|
Kim SA, Lee AS, Lee HB, Hur HJ, Lee SH, Sung MJ. Soluble epoxide hydrolase inhibitor, TPPU, attenuates progression of atherosclerotic lesions and vascular smooth muscle cell phenotypic switching. Vascul Pharmacol 2022; 145:107086. [PMID: 35752378 DOI: 10.1016/j.vph.2022.107086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/22/2022] [Accepted: 06/20/2022] [Indexed: 11/29/2022]
Abstract
Atherosclerosis manifests as a chronic inflammation resulting from multiple interactions between circulating factors and various cell types in blood vessel walls. Growing evidence shows that phenotypic switching and proliferation of vascular smooth muscle cells (VSMCs) plays an important role in the progression of atherosclerosis. Soluble epoxide hydrolase (sEH)/epoxyeicosatrienoic acids are mediated by vascular inflammation. N-[1-(1-oxopropyl)-4-piperidinyl]-N'-[4-(trifluoromethoxy)phenyl]-urea (TPPU) is an sEH inhibitor. This study investigated the therapeutic effect of TPPU on atherosclerosis in vivo and homocysteine-induced vascular inflammation in vitro and explored their molecular mechanisms. We found that TPPU decreased WD-induced atherosclerotic plaque lesions, inflammation, expression of sEH, and nicotinamide adenine dinucleotide phosphate oxidase-4 (Nox4), and increased the expression of contractile phenotype marker of aortas in ApoE (-/-) mice. TPPU also inhibited homocysteine-stimulated VSMC proliferation, migration, and phenotypic switching, and reduced Nox4 in human-aorta-VSMC regulation. We conclude that TPPU has anti-atherosclerotic effects, potentially because of the suppression of VSMC phenotype switching. Thus, TPPU could be a potential therapeutic target for phenotypic switching attenuation in atherosclerosis.
Collapse
Affiliation(s)
- So Ah Kim
- Research Group of Natural Materials and Metabolism, Food Functionality Research, Korea Food Research Institute, Jeollabuk-Do, Republic of Korea; Department of Food Biotechnology, Chonbuk National University, Jeollabuk-Do, Republic of Korea
| | - Ae Sin Lee
- Research Group of Natural Materials and Metabolism, Food Functionality Research, Korea Food Research Institute, Jeollabuk-Do, Republic of Korea
| | - Han Bit Lee
- Research Group of Natural Materials and Metabolism, Food Functionality Research, Korea Food Research Institute, Jeollabuk-Do, Republic of Korea
| | - Haeng Jeon Hur
- Research Group of Natural Materials and Metabolism, Food Functionality Research, Korea Food Research Institute, Jeollabuk-Do, Republic of Korea
| | - Sang Hee Lee
- Research Group of Natural Materials and Metabolism, Food Functionality Research, Korea Food Research Institute, Jeollabuk-Do, Republic of Korea
| | - Mi Jeong Sung
- Research Group of Natural Materials and Metabolism, Food Functionality Research, Korea Food Research Institute, Jeollabuk-Do, Republic of Korea.
| |
Collapse
|
6
|
Tagi VM, Mainieri F, Chiarelli F. Hypertension in Patients with Insulin Resistance: Etiopathogenesis and Management in Children. Int J Mol Sci 2022; 23:ijms23105814. [PMID: 35628624 PMCID: PMC9144705 DOI: 10.3390/ijms23105814] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/19/2022] [Accepted: 05/19/2022] [Indexed: 11/16/2022] Open
Abstract
Insulin resistance (IR) is a key component in the etiopathogenesis of hypertension (HS) in patients with diabetes mellitus (DM). Several pathways have been found to be involved in this mechanism in recent literature. For the above-mentioned reasons, treatment of HS should be specifically addressed in patients affected by DM. Two relevant recently published guidelines have stressed this concept, giving specific advice in the treatment of HS in children belonging to this group: the European Society of HS guidelines for the management of high blood pressure in children and adolescents and the American Academy of Pediatrics Clinical Practice Guideline for Screening and Management of High Blood Pressure in Children and Adolescents. Our aim is to summarize the main pathophysiological mechanisms through which IR causes HS and to highlight the specific principles of treatment of HS for children with DM.
Collapse
|
7
|
Al-Shabrawey M, Elmarakby A, Samra Y, Moustafa M, Looney SW, Maddipati KR, Tawfik A. Hyperhomocysteinemia dysregulates plasma levels of polyunsaturated fatty acids-derived eicosanoids. LIFE RESEARCH 2022; 5:14. [PMID: 36341141 PMCID: PMC9632953 DOI: 10.53388/2022-0106-103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Hyperhomocysteinemia (HHcy) contributes to the incidence of many cardiovascular diseases (CVD). Our group have previously established crucial roles of eicosanoids and homocysteine in the incidence of vascular injury in diabetic retinopathy and renal injury. Using cystathionine-β-synthase heterozygous mice (cβs+/-) as a model of HHcy, the current study was designed to determine the impact of homocysteine on circulating levels of lipid mediators derived from polyunsaturated fatty acids (PUFA). Plasma samples were isolated from wild-type (WT) and cβs+/- mice for the assessment of eicosanoids levels using LC/MS. Plasma 12/15-lipoxygenase (12/15-LOX) activity significantly decreased in cβs+/- vs. WT control mice. LOX-derived metabolites from both omega-3 and omega-6 PUFA were also reduced in cβs+/- mice compared to WT control (P < 0.05). Contrary to LOX metabolites, cytochrome P450 (CYP) metabolites from omega-3 and omega-6 PUFA were significantly elevated in cβs+/- mice compared to WT control. Epoxyeicosatrienoic acids (EETs) are epoxides derived from arachidonic acid (AA) metabolism by CYP with anti-inflammatory properties and are known to limit vascular injury, however their physiological role is limited by their rapid degradation by soluble epoxide hydrolase (sEH) to their corresponding diols (DiHETrEs). In cβs+/- mice, a significant decrease in the plasma EETs bioavailability was obvious as evident by the decrease in EETs/ DiHETrEs ratio relative to WT control mice. Cyclooxygenase (COX) metabolites were also significantly decreased in cβs+/- vs. WT control mice. These data suggest that HHcy impacts eicosanoids metabolism through decreasing LOX and COX metabolic activities while increasing CYP metabolic activity. The increase in AA metabolism by CYP was also associated with increase in sEH activity and decrease in EETs bioavailability. Dysregulation of eicosanoids metabolism could be a contributing factor to the incidence and progression of HHcy-induced CVD.
Collapse
Affiliation(s)
- Mohamed Al-Shabrawey
- Department of Foundational Medical Studies and Eye Research Center, Oakland University William Beaumont School of Medicine, Rochester, Michigan, USA
- Eye Research Institute, Oakland University, Rochester, Michigan, USA
| | - Ahmed Elmarakby
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, Georgia, USA
- Departments of Pharmacology & Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Yara Samra
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, Georgia, USA
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Egypt
| | - Mohamed Moustafa
- Department of Foundational Medical Studies and Eye Research Center, Oakland University William Beaumont School of Medicine, Rochester, Michigan, USA
- Eye Research Institute, Oakland University, Rochester, Michigan, USA
| | - Stephen W. Looney
- Department of Population Health Sciences, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Krishna Rao Maddipati
- Bioactive Lipids Research Program, Department of Pathology, Wayne State University, Michigan, USA
| | - Amany Tawfik
- Department of Foundational Medical Studies and Eye Research Center, Oakland University William Beaumont School of Medicine, Rochester, Michigan, USA
- Eye Research Institute, Oakland University, Rochester, Michigan, USA
| |
Collapse
|
8
|
Li H, Bradbury JA, Edin ML, Graves JP, Gruzdev A, Cheng J, Hoopes SL, DeGraff LM, Fessler MB, Garantziotis S, Schurman SH, Zeldin DC. sEH promotes macrophage phagocytosis and lung clearance of Streptococcus pneumoniae. J Clin Invest 2021; 131:129679. [PMID: 34591792 DOI: 10.1172/jci129679] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 09/28/2021] [Indexed: 12/12/2022] Open
Abstract
Epoxyeicosatrienoic acids (EETs) have potent antiinflammatory properties. Hydrolysis of EETs by soluble epoxide hydrolase/ epoxide hydrolase 2 (sEH/EPHX2) to less active diols attenuates their antiinflammatory effects. Macrophage activation is critical to many inflammatory responses; however, the role of EETs and sEH in regulating macrophage function remains unknown. Lung bacterial clearance of Streptococcus pneumoniae was impaired in Ephx2-deficient (Ephx2-/-) mice and in mice treated with an sEH inhibitor. The EET receptor antagonist EEZE restored lung clearance of S. pneumoniae in Ephx2-/- mice. Ephx2-/- mice had normal lung Il1b, Il6, and Tnfa expression levels and macrophage recruitment to the lungs during S. pneumoniae infection; however, Ephx2 disruption attenuated proinflammatory cytokine induction, Tlr2 and Pgylrp1 receptor upregulation, and Ras-related C3 botulinum toxin substrates 1 and 2 (Rac1/2) and cell division control protein 42 homolog (Cdc42) activation in PGN-stimulated macrophages. Consistent with these observations, Ephx2-/- macrophages displayed reduced phagocytosis of S. pneumoniae in vivo and in vitro. Heterologous overexpression of TLR2 and peptidoglycan recognition protein 1 (PGLYRP1) in Ephx2-/- macrophages restored macrophage activation and phagocytosis. Human macrophage function was similarly regulated by EETs. Together, these results demonstrate that EETs reduced macrophage activation and phagocytosis of S. pneumoniae through the downregulation of TLR2 and PGLYRP1 expression. Defining the role of EETs and sEH in macrophage function may lead to the development of new therapeutic approaches for bacterial diseases.
Collapse
|
9
|
Zhou Y, Murugan DD, Khan H, Huang Y, Cheang WS. Roles and Therapeutic Implications of Endoplasmic Reticulum Stress and Oxidative Stress in Cardiovascular Diseases. Antioxidants (Basel) 2021; 10:antiox10081167. [PMID: 34439415 PMCID: PMC8388996 DOI: 10.3390/antiox10081167] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/18/2021] [Accepted: 07/21/2021] [Indexed: 12/12/2022] Open
Abstract
In different pathological states that cause endoplasmic reticulum (ER) calcium depletion, altered glycosylation, nutrient deprivation, oxidative stress, DNA damage or energy perturbation/fluctuations, the protein folding process is disrupted and the ER becomes stressed. Studies in the past decade have demonstrated that ER stress is closely associated with pathogenesis of obesity, insulin resistance and type 2 diabetes. Excess nutrients and inflammatory cytokines associated with metabolic diseases can trigger or worsen ER stress. ER stress plays a critical role in the induction of endothelial dysfunction and atherosclerosis. Signaling pathways including AMP-activated protein kinase and peroxisome proliferator-activated receptor have been identified to regulate ER stress, whilst ER stress contributes to the imbalanced production between nitric oxide (NO) and reactive oxygen species (ROS) causing oxidative stress. Several drugs or herbs have been proved to protect against cardiovascular diseases (CVD) through inhibition of ER stress and oxidative stress. The present article reviews the involvement of ER stress and oxidative stress in cardiovascular dysfunction and the potential therapeutic implications.
Collapse
Affiliation(s)
- Yan Zhou
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China;
| | - Dharmani Devi Murugan
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia;
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan 23200, Pakistan;
| | - Yu Huang
- School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong 999077, China;
| | - Wai San Cheang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China;
- Correspondence: ; Tel.: +853-8822-4914
| |
Collapse
|
10
|
m6A modification promotes miR-133a repression during cardiac development and hypertrophy via IGF2BP2. Cell Death Discov 2021; 7:157. [PMID: 34226535 PMCID: PMC8257704 DOI: 10.1038/s41420-021-00552-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 05/09/2021] [Accepted: 06/03/2021] [Indexed: 12/31/2022] Open
Abstract
Both N6-methyladenosine (m6A) RNA modification and microRNAs (miRNAs) are common regulatory mechanisms for gene post-transcription by modulating mRNA stability and translation. They also share the same 3′-untranslated regions (UTRs) regions for their target gene. However, little is known about their potential interaction in cell development and biology. Here, we aimed to investigate how m6A regulates the specific miRNA repression during cardiac development and hypertrophy. Our multiple lines of bioinformatic and molecular biological evidence have shown that m6A modification on cardiac miR-133a target sequence promotes miR-133a repressive effect via AGO2-IGF2BP2 (Argonaute 2—Insulin-like growth factor 2 mRNA binding protein 2) complex. Among 139 cardiac miRNAs, only the seed sequence of miR-133a was inversely complement to m6A consensus motif “GGACH” by sequence alignment analysis. Immunofluorescence staining, luciferase reporter, and m6A-RIP (RNA immunoprecipitation) assays revealed that m6A modification facilitated miR-133a binding to and repressing their targets. The inhibition of the miR-133a on cardiac proliferation and hypertrophy could be prevented by silencing of Fto (FTO alpha-ketoglutarate dependent dioxygenase) which induced m6A modification. IGF2BP2, an m6A binding protein, physically interacted with AGO2 and increased more miR-133a accumulation on its target site, which was modified by m6A. In conclusion, our study revealed a novel and precise regulatory mechanism that the m6A modification promoted the repression of specific miRNA during heart development and hypertrophy. Targeting m6A modification might provide a strategy to repair hypertrophic gene expression induced by miR-133a.
Collapse
|
11
|
Liang C, Wang QS, Yang X, Zhu D, Sun Y, Niu N, Yao J, Dong BH, Jiang S, Tang LL, Lou J, Yu CJ, Shao Q, Wu MM, Zhang ZR. Homocysteine Causes Endothelial Dysfunction via Inflammatory Factor-Mediated Activation of Epithelial Sodium Channel (ENaC). Front Cell Dev Biol 2021; 9:672335. [PMID: 34222246 PMCID: PMC8247579 DOI: 10.3389/fcell.2021.672335] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/18/2021] [Indexed: 11/24/2022] Open
Abstract
Background Hyperhomocysteinemia (HHcy) causes cardiovascular diseases via regulating inflammatory responses. We investigated whether and how the epithelial sodium channel (ENaC), a recently identified ion channel in endothelial cells, plays a role in HHcy-induced endothelial dysfunction. Methods Cell-attached patch-clamp recording in acute split-open aortic endothelial cells, western blot, confocal imaging, and wire myograph combined with pharmacological approaches were used to determine whether HHcy-mediated inflammatory signaling leads to endothelial dysfunction via stimulating ENaC. Results The data showed that 4 weeks after L-methionine diet the levels of plasma Hcy were significantly increased and the ENaC was dramatically activated in mouse aortic endothelial cells. Administration of benzamil, a specific ENaC blocker, ameliorated L-methionine diet-induced impairment of endothelium-dependent relaxation (EDR) and reversed Hcy-induced increase in ENaC activity. Pharmacological inhibition of NADPH oxidase, reactive oxygen species (ROS), cyclooxygenase-2 (COX-2)/thromboxane B2 (TXB2), or serum/glucocorticoid regulated kinase 1 (SGK1) effectively attenuated both the Hcy-induced activation of endothelial ENaC and impairment of EDR. Our in vitro data showed that both NADPH oxidase inhibitor and an ROS scavenger reversed Hcy-induced increase in COX-2 expression in human umbilical vein endothelial cells (HUVECs). Moreover, Hcy-induced increase in expression levels of SGK-1, phosphorylated-SGK-1, and phosphorylated neural precursor cell-expressed developmentally downregulated protein 4-2 (p-Nedd4-2) in HUVECs were significantly blunted by a COX-2 inhibitor. Conclusion We show that Hcy activates endothelial ENaC and subsequently impairs EDR of mouse aorta, via ROS/COX-2-dependent activation of SGK-1/Nedd4-2 signaling. Our study provides a rational that blockade of the endothelial ENaC could be potential method to prevent and/or to treat Hcy-induced cardiovascular disease.
Collapse
Affiliation(s)
- Chen Liang
- Departments of Pharmacy and Cardiology, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang Key Laboratory for Metabolic Disorder and Cancer Related Cardiovascular Diseases, NHC Key Laboratory of Cell Transplantation, Harbin Medical University and Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Qiu-Shi Wang
- Departments of Pharmacy and Cardiology, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang Key Laboratory for Metabolic Disorder and Cancer Related Cardiovascular Diseases, NHC Key Laboratory of Cell Transplantation, Harbin Medical University and Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Xu Yang
- Departments of Pharmacy and Cardiology, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang Key Laboratory for Metabolic Disorder and Cancer Related Cardiovascular Diseases, NHC Key Laboratory of Cell Transplantation, Harbin Medical University and Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Di Zhu
- Departments of Pharmacy and Cardiology, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang Key Laboratory for Metabolic Disorder and Cancer Related Cardiovascular Diseases, NHC Key Laboratory of Cell Transplantation, Harbin Medical University and Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Yu Sun
- Departments of Pharmacy and Cardiology, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang Key Laboratory for Metabolic Disorder and Cancer Related Cardiovascular Diseases, NHC Key Laboratory of Cell Transplantation, Harbin Medical University and Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Na Niu
- Departments of Pharmacy and Cardiology, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang Key Laboratory for Metabolic Disorder and Cancer Related Cardiovascular Diseases, NHC Key Laboratory of Cell Transplantation, Harbin Medical University and Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Jie Yao
- Departments of Pharmacy and Cardiology, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang Key Laboratory for Metabolic Disorder and Cancer Related Cardiovascular Diseases, NHC Key Laboratory of Cell Transplantation, Harbin Medical University and Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Bi-Han Dong
- Departments of Pharmacy and Cardiology, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang Key Laboratory for Metabolic Disorder and Cancer Related Cardiovascular Diseases, NHC Key Laboratory of Cell Transplantation, Harbin Medical University and Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Shuai Jiang
- Departments of Pharmacy and Cardiology, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang Key Laboratory for Metabolic Disorder and Cancer Related Cardiovascular Diseases, NHC Key Laboratory of Cell Transplantation, Harbin Medical University and Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Liang-Liang Tang
- Departments of Pharmacy and Cardiology, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang Key Laboratory for Metabolic Disorder and Cancer Related Cardiovascular Diseases, NHC Key Laboratory of Cell Transplantation, Harbin Medical University and Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Jie Lou
- Departments of Pharmacy and Cardiology, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang Key Laboratory for Metabolic Disorder and Cancer Related Cardiovascular Diseases, NHC Key Laboratory of Cell Transplantation, Harbin Medical University and Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Chang-Jiang Yu
- Departments of Pharmacy and Cardiology, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang Key Laboratory for Metabolic Disorder and Cancer Related Cardiovascular Diseases, NHC Key Laboratory of Cell Transplantation, Harbin Medical University and Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Qun Shao
- Departments of Pharmacy and Cardiology, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang Key Laboratory for Metabolic Disorder and Cancer Related Cardiovascular Diseases, NHC Key Laboratory of Cell Transplantation, Harbin Medical University and Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Ming-Ming Wu
- Departments of Pharmacy and Cardiology, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang Key Laboratory for Metabolic Disorder and Cancer Related Cardiovascular Diseases, NHC Key Laboratory of Cell Transplantation, Harbin Medical University and Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Zhi-Ren Zhang
- Departments of Pharmacy and Cardiology, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang Key Laboratory for Metabolic Disorder and Cancer Related Cardiovascular Diseases, NHC Key Laboratory of Cell Transplantation, Harbin Medical University and Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| |
Collapse
|
12
|
Suppression of m6A mRNA modification by DNA hypermethylated ALKBH5 aggravates the oncological behavior of KRAS mutation/LKB1 loss lung cancer. Cell Death Dis 2021; 12:518. [PMID: 34016959 PMCID: PMC8137886 DOI: 10.1038/s41419-021-03793-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 04/30/2021] [Accepted: 05/03/2021] [Indexed: 02/04/2023]
Abstract
Oncogenic KRAS mutations combined with the loss of the LKB1 tumor-suppressor gene (KL) are strongly associated with aggressive forms of lung cancer. N6-methyladenosine (m6A) in mRNA is a crucial epigenetic modification that controls cancer self-renewal and progression. However, the regulation and role of m6A modification in this cancer are unclear. We found that decreased m6A levels correlated with the disease progression and poor survival for KL patients. The correlation was mediated by a special increase in ALKBH5 (AlkB family member 5) levels, an m6A demethylase. ALKBH5 gain- or loss-of function could effectively reverse LKB1 regulated cell proliferation, colony formation, and migration of KRAS-mutated lung cancer cells. Mechanistically, LKB1 loss upregulated ALKBH5 expression by DNA hypermethylation of the CTCF-binding motif on the ALKBH5 promoter, which inhibited CTCF binding but enhanced histone modifications, including H3K4me3, H3K9ac, and H3K27ac. This effect could successfully be rescued by LKB1 expression. ALKBH5 demethylation of m6A stabilized oncogenic drivers, such as SOX2, SMAD7, and MYC, through a pathway dependent on YTHDF2, an m6A reader protein. The above findings were confirmed in clinical KRAS-mutated lung cancer patients. We conclude that loss of LKB1 promotes ALKBH5 transcription by a DNA methylation mechanism, reduces m6A modification, and increases the stability of m6A target oncogenes, thus contributing to aggressive phenotypes of KRAS-mutated lung cancer.
Collapse
|
13
|
Jiang Q, Wang L, Si X, Tian JL, Zhang Y, Gui HL, Li B, Tan DH. Current progress on the mechanisms of hyperhomocysteinemia-induced vascular injury and use of natural polyphenol compounds. Eur J Pharmacol 2021; 905:174168. [PMID: 33984300 DOI: 10.1016/j.ejphar.2021.174168] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 04/09/2021] [Accepted: 05/05/2021] [Indexed: 12/28/2022]
Abstract
Cardiovascular disease is one of the most common diseases in the elderly population, and its incidence has rapidly increased with the prolongation of life expectancy. Hyperhomocysteinemia is an independent risk factor for various cardiovascular diseases, including atherosclerosis, and damage to vascular function plays an initial role in its pathogenesis. This review presents the latest knowledge on the mechanisms of vascular injury caused by hyperhomocysteinemia, including oxidative stress, endoplasmic reticulum stress, protein N-homocysteinization, and epigenetic modification, and discusses the therapeutic targets of natural polyphenols. Studies have shown that natural polyphenols in plants can reduce homocysteine levels and regulate DNA methylation by acting on oxidative stress and endoplasmic reticulum stress-related signaling pathways, thus improving hyperhomocysteinemia-induced vascular injury. Natural polyphenols obtained via daily diet are safer and have more practical significance in the prevention and treatment of chronic diseases than traditional drugs.
Collapse
Affiliation(s)
- Qiao Jiang
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning 110866, China.
| | - Li Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Lihu Road 1800, Wuxi 214122, China.
| | - Xu Si
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning 110866, China.
| | - Jin-Long Tian
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning 110866, China.
| | - Ye Zhang
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning 110866, China.
| | - Hai-Long Gui
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning 110866, China.
| | - Bin Li
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning 110866, China.
| | - De-Hong Tan
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning 110866, China.
| |
Collapse
|
14
|
Luo J, Hu S, Fu M, Luo L, Li Y, Li W, Cai Y, Dong R, Yang Y, Tu L, Xu X. Inhibition of soluble epoxide hydrolase alleviates insulin resistance and hypertension via downregulation of SGLT2 in the mouse kidney. J Biol Chem 2021; 296:100667. [PMID: 33864813 PMCID: PMC8131320 DOI: 10.1016/j.jbc.2021.100667] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 04/07/2021] [Accepted: 04/13/2021] [Indexed: 12/09/2022] Open
Abstract
The epoxyeicosatrienoic acid (EET) exerts beneficial effects on insulin resistance and/or hypertension. EETs could be readily converted to less biological active diols by soluble epoxide hydrolase (sEH). However, whether sEH inhibition can ameliorate the comorbidities of insulin resistance and hypertension and the underlying mechanisms of this relationship are unclear. In this study, C57BL/6 mice were rendered hypertensive and insulin resistant through a high-fat and high-salt (HF–HS) diet. The sEH inhibitor, 1-trifluoromethoxyphenyl-3-(1-propionylpiperidin-4-yl) urea (TPPU), was used to treat mice (1 mg/kg/day) for 8 weeks, followed by analysis of metabolic parameters. The expression of sEH and the sodium–glucose cotransporter 2 (SGLT2) was markedly upregulated in the kidneys of mice fed an HF–HS diet. We found that TPPU administration increased kidney EET levels, improved insulin resistance, and reduced hypertension. Furthermore, TPPU treatment prevented upregulation of SGLT2 and the associated increased urine volume and the excretion of urine glucose and urine sodium. Importantly, TPPU alleviated renal inflammation. In vitro, human renal proximal tubule epithelial cells (HK-2 cells) were used to further investigate the underlying mechanism. We observed that 14,15-EET or sEH knockdown or inhibition prevented the upregulation of SGLT2 upon treatment with palmitic acid or NaCl by inhibiting the inhibitory kappa B kinase α/β/NF-κB signaling pathway. In conclusion, sEH inhibition by TPPU alleviated insulin resistance and hypertension induced by an HF–HS diet in mice. The increased urine excretion of glucose and sodium was mediated by decreased renal SGLT2 expression because of inactivation of the inhibitory kappa B kinase α/β/NF-κB–induced inflammatory response.
Collapse
Affiliation(s)
- Jinlan Luo
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Division of Cardiology and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuiqing Hu
- Division of Cardiology and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Menglu Fu
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liman Luo
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuanyuan Li
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenhua Li
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yueting Cai
- Division of Cardiology and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ruolan Dong
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Yang
- Division of Endocrinology and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ling Tu
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China.
| | - Xizhen Xu
- Division of Cardiology and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China.
| |
Collapse
|
15
|
Yu W, Li S, Wu H, Hu P, Chen L, Zeng C, Tong X. Endothelial Nox4 dysfunction aggravates atherosclerosis by inducing endoplasmic reticulum stress and soluble epoxide hydrolase. Free Radic Biol Med 2021; 164:44-57. [PMID: 33418110 DOI: 10.1016/j.freeradbiomed.2020.12.450] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 12/17/2020] [Accepted: 12/29/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND AND AIMS Our previous findings have demonstrated the protective effect of endothelial Nox4-based NADPH oxidase on atherosclerosis. One of the possible mechanisms is the inhibition of soluble epoxide hydrolase (sEH), a proinflammatory and atherogenic factor. Our goal was to investigate whether in vivo inhibition of sEH by 1-trifluoromethoxyphenyl-3-(1-propionylpiperidin-4-yl) urea (TPPU) alleviates endothelial Nox4 dysfunction caused atherosclerosis and the regulatory mechanism of endothelial Nox4 on sEH. METHODS & results: We used endothelial human Nox4 dominant-negative (EDN) transgenic mice in ApoE deficient background to mimic the dysfunction of endothelial Nox4 in atherosclerosis-prone conditions. In EDN aortic endothelium, sEH and the inflammatory marker vascular cell adhesion molecule 1 (VCAM1) were upregulated. TPPU reduced atherosclerotic lesions in EDN mice. In EDN endothelial cells (ECs), the endoplasmic reticulum (ER) stress markers (BIP, IRE1α, phosphorylation of PERK, ATF6) were upregulated, and they can be suppressed by ER stress inhibitor 4-phenyl butyric acid (4-PBA). In EDN ECs, 4-PBA downregulated the expression of sEH and VCAM1, suppressed inflammation, and its application in vivo reduced atherosclerotic lesions of EDN mice. CONCLUSIONS Endothelial Nox4 dysfunction upregulated sEH to enhance inflammation, probably by its induction of ER stress. Inhibition of ER stress or sEH is beneficial to alleviate atherosclerosis caused by endothelial Nox4 dysfunction.
Collapse
Affiliation(s)
- Weimin Yu
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Innovative Drug Research Centre, Chongqing University, Chongqing, 401331, China
| | - Siqi Li
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Innovative Drug Research Centre, Chongqing University, Chongqing, 401331, China
| | - Haixia Wu
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Innovative Drug Research Centre, Chongqing University, Chongqing, 401331, China
| | - Pingping Hu
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Innovative Drug Research Centre, Chongqing University, Chongqing, 401331, China.
| | - Lili Chen
- Wuhan Easy Diagnosis Biomedicine Co., Ltd, Wuhan, 430075, China
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, 400042, China.
| | - Xiaoyong Tong
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Innovative Drug Research Centre, Chongqing University, Chongqing, 401331, China.
| |
Collapse
|
16
|
The Multifaceted Role of Epoxide Hydrolases in Human Health and Disease. Int J Mol Sci 2020; 22:ijms22010013. [PMID: 33374956 PMCID: PMC7792612 DOI: 10.3390/ijms22010013] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/18/2020] [Accepted: 12/19/2020] [Indexed: 12/12/2022] Open
Abstract
Epoxide hydrolases (EHs) are key enzymes involved in the detoxification of xenobiotics and biotransformation of endogenous epoxides. They catalyze the hydrolysis of highly reactive epoxides to less reactive diols. EHs thereby orchestrate crucial signaling pathways for cell homeostasis. The EH family comprises 5 proteins and 2 candidate members, for which the corresponding genes are not yet identified. Although the first EHs were identified more than 30 years ago, the full spectrum of their substrates and associated biological functions remain partly unknown. The two best-known EHs are EPHX1 and EPHX2. Their wide expression pattern and multiple functions led to the development of specific inhibitors. This review summarizes the most important points regarding the current knowledge on this protein family and highlights the particularities of each EH. These different enzymes can be distinguished by their expression pattern, spectrum of associated substrates, sub-cellular localization, and enzymatic characteristics. We also reevaluated the pathogenicity of previously reported variants in genes that encode EHs and are involved in multiple disorders, in light of large datasets that were made available due to the broad development of next generation sequencing. Although association studies underline the pleiotropic and crucial role of EHs, no data on high-effect variants are confirmed to date.
Collapse
|
17
|
Zhang X, Huang Z, Xie Z, Chen Y, Zheng Z, Wei X, Huang B, Shan Z, Liu J, Fan S, Chen J, Zhao F. Homocysteine induces oxidative stress and ferroptosis of nucleus pulposus via enhancing methylation of GPX4. Free Radic Biol Med 2020; 160:552-565. [PMID: 32896601 DOI: 10.1016/j.freeradbiomed.2020.08.029] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 08/19/2020] [Accepted: 08/30/2020] [Indexed: 12/16/2022]
Abstract
Homocysteine (Hcy) is an amino acid involved in gene methylation. Plasma concentration of Hcy is elevated in the pathological condition hyperhomocysteinemia (HHcy), which increases the risk of disorders of the vascular, nervous and musculoskeletal systems, including chondrocyte dysfunction. The present study aimed to explore the role of Hcy in intervertebral disc degeneration (IVDD), using a range of techniques. A clinical epidemiological study showed that HHcy is an independent risk factor for human IVDD. Cell culture using rat nucleus pulposus cells showed that Hcy promotes a degenerative cell phenotype (involving increased oxidative stress and cell death by ferroptosis) which is mediated by upregulated methylation of GPX4. An in-vivo mouse 'puncture' model of IVDD showed that folic acid (which is used to treat HHcy in humans) reduced the ability of diet-induced HHcy to promote IVDD. We conclude that Hcy upregulates oxidative stress and ferroptosis in the nucleus pulposus via enhancing GPX4 methylation, and is a new contributing factor in IVDD.
Collapse
Affiliation(s)
- Xuyang Zhang
- Department of Orthopaedics, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, PR China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, PR China.
| | - Zhaobo Huang
- Department of Orthopaedics, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, PR China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, PR China.
| | - Ziang Xie
- Department of Orthopaedics, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, PR China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, PR China.
| | - Yilei Chen
- Department of Orthopaedics, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, PR China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, PR China.
| | - Zeyu Zheng
- Department of Orthopaedics, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, PR China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, PR China.
| | - Xiao'an Wei
- Department of Orthopaedics, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, PR China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, PR China.
| | - Bao Huang
- Department of Orthopaedics, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, PR China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, PR China.
| | - Zhi Shan
- Department of Orthopaedics, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, PR China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, PR China.
| | - Junhui Liu
- Department of Orthopaedics, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, PR China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, PR China.
| | - Shunwu Fan
- Department of Orthopaedics, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, PR China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, PR China.
| | - Jian Chen
- Department of Orthopaedics, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, PR China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, PR China.
| | - Fengdong Zhao
- Department of Orthopaedics, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, PR China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, PR China.
| |
Collapse
|
18
|
Xiao MZ, Liu JM, Xian CL, Chen KY, Liu ZQ, Cheng YY. Therapeutic potential of ALKB homologs for cardiovascular disease. Biomed Pharmacother 2020; 131:110645. [PMID: 32942149 DOI: 10.1016/j.biopha.2020.110645] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 08/05/2020] [Accepted: 08/16/2020] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the leading causes of human death. Recently, ALKB homologs, including ALKBH1-8 and FTO, have been found to have a variety of biological functions, such as histone demethylation, RNA demethylation, and DNA demethylation. These functions may regulate the physiological and pathological processes of CVDs, including inflammation, oxidative stress, cell apoptosis, and mitochondrial, endothelial, and fat metabolism dysfunction. In the present review, we summarize the biological functions of ALKB homologs and the relationship between the ALKB homologs and CVDs. Importantly, we discuss the roles of ALKB homologs in the regulation of oxidative stress, inflammation, autophagy, and DNA damage in CVDs, as well as the practical applications of ALKB homologs inhibitors or agonists in treating CVDs. In conclusion, the ALKBH family might be a promising target for CVDs therapy.
Collapse
Affiliation(s)
- Ming-Zhu Xiao
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China; School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Jia-Ming Liu
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China; School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Cui-Ling Xian
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China; School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Keng-Yu Chen
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China; The Second Affiliated Hospital of Guangdong Pharmaceutical University, Yunfu, 527300, China
| | - Zhong-Qiu Liu
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China.
| | - Yuan-Yuan Cheng
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China.
| |
Collapse
|
19
|
Guo Y, Pei Y, Li K, Cui W, Zhang D. DNA N 6-methyladenine modification in hypertension. Aging (Albany NY) 2020; 12:6276-6291. [PMID: 32283543 PMCID: PMC7185115 DOI: 10.18632/aging.103023] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 03/02/2020] [Indexed: 02/06/2023]
Abstract
DNA methylation has a role in the pathogenesis of essential hypertension. DNA N6-methyladenine (6mA) modification as a novel adenine methylation exists in human tissues, but whether it plays a role in hypertension development remains unclear. Here, we reported that the global 6mA DNA level in leukocytes was significantly reduced in patients with hypertension and was reversed with successful treatment. Age, systolic blood pressure, and serum total cholesterol and high-density lipoprotein levels were associated with decreased leukocyte 6mA DNA level. Elevated ALKBH1 (AlkB homolog 1), a demethylase of 6mA, level mediated this dynamic change in 6mA level in leukocytes and vascular smooth muscle cells in hypertension mouse and rat models. Knockdown of ALKBH1 suppressed angiotensin II-induced vascular smooth muscle phenotype transformation, proliferation and migration. ALKBH1-6mA directly and negatively regulated hypoxia inducible factor 1 α (HIF1α), which responded to angiotensin II-induced vascular remodeling. Collectively, our results demonstrate a potential epigenetic role for ALKBH1-6mA regulation in hypertension development, diagnosis and treatment.
Collapse
Affiliation(s)
- Ye Guo
- Department of Laboratory Medicine, Peking Union Medical College Hospital and Peking Union Medical College, Beijing 100021, PR China
| | - Yuqing Pei
- State Key Laboratory of Molecular Oncology, Department of Clinical Laboratory, National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, PR China
| | - Kexin Li
- State Key Laboratory of Molecular Oncology, Department of Clinical Laboratory, National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, PR China
| | - Wei Cui
- State Key Laboratory of Molecular Oncology, Department of Clinical Laboratory, National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, PR China
| | - Donghong Zhang
- Center for Molecular and Translational Medicine, Georgia State University, Research Science Center, Atlanta, GA 30303, USA
| |
Collapse
|
20
|
Liu G, Wu F, Jiang X, Que Y, Qin Z, Hu P, Lee KSS, Yang J, Zeng C, Hammock BD, Tong X. Inactivation of Cys 674 in SERCA2 increases BP by inducing endoplasmic reticulum stress and soluble epoxide hydrolase. Br J Pharmacol 2020; 177:1793-1805. [PMID: 31758704 DOI: 10.1111/bph.14937] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/29/2019] [Accepted: 10/30/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND AND PURPOSE The kidney is essential in regulating sodium homeostasis and BP. The irreversible oxidation of Cys674 (C674) in the sarcoplasmic/endoplasmic reticulum calcium ATPase 2 (SERCA2) is increased in the renal cortex of hypertensive mice. Whether inactivation of C674 promotes hypertension is unclear. Here we have investigated the effects on BP of the inactivation of C674, and its role in the kidney. EXPERIMENTAL APPROACH We used heterozygous SERCA2 C674S knock-in (SKI) mice, where half of C674 was substituted by serine, to represent partial irreversible oxidation of C674. The BP, urine volume, and urine composition of SKI mice and their littermate wild-type (WT) mice were measured. The kidneys were collected for cell culture, Na+ /K+ -ATPase activity, protein expression, and immunohistological analysis. KEY RESULTS Compared with WT mice, SKI mice had higher BP, lower urine volume and sodium excretion, up-regulated endoplasmic reticulum (ER) stress markers and soluble epoxide hydrolase (sEH), and down-regulated dopamine D1 receptors in renal cortex and cells from renal proximal tubule. ER stress and sEH were mutually regulated, and both upstream of D1 receptors. Inhibition of ER stress or sEH up-regulated expression of D1 receptors, decreased the activity of Na+ /K+ -ATPase, increased sodium excretion, and lowered BP in SKI mice. CONCLUSIONS AND IMPLICATIONS The inactivation of SERCA2 C674 promotes the development of hypertension by inducing ER stress and sEH. Our study highlights the importance of C674 redox status in BP control and the contribution of SERCA2 to sodium homeostasis and BP in the kidney.
Collapse
Affiliation(s)
- Gang Liu
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Fuhua Wu
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Xiaoli Jiang
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Yumei Que
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Zhexue Qin
- Department of Cardiovascular Diseases, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Pingping Hu
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Kin Sing Stephen Lee
- Department of Entomology & UCD Comprehensive Cancer Center, University of California-Davis, Davis, California.,Department of Pharmacology & Toxicology, Michigan State University, East Lansing, Michigan
| | - Jian Yang
- Department of Clinical Nutrition, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Bruce D Hammock
- Department of Entomology & UCD Comprehensive Cancer Center, University of California-Davis, Davis, California
| | - Xiaoyong Tong
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| |
Collapse
|
21
|
Soluble epoxide hydrolase inhibitor protects against blood-brain barrier dysfunction in a mouse model of type 2 diabetes via the AMPK/HO-1 pathway. Biochem Biophys Res Commun 2020; 524:354-359. [PMID: 32001002 DOI: 10.1016/j.bbrc.2020.01.085] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 01/15/2020] [Indexed: 12/11/2022]
Abstract
Diabetes mellitus is a metabolic disorder that can lead to blood-brain barrier (BBB) disruption and cognitive decline. However, the mechanisms of BBB breakdown in diabetes are still unclear. Soluble epoxide hydrolase (sEH) is an enzyme that degrades epoxyeicosatrienoic acids (EETs), which have multiple protective effects on vascular structure and functions. In the current study, we showed increased vascular permeability of the BBB, which was accompanied by upregulation of sEH and downregulation of 14,15-EET. Moreover, the sEH inhibitor t-AUCB restored diabetic BBB integrity in vivo, and 14,15-EET prevented ROS accumulation and MEC injury in vitro. t-AUCB or 14,15-EET treatment provoked AMPK/HO-1 activation under diabetic conditions in vivo and in vitro. Thus, we suggest that decreased EET degradation by sEH inhibition might be a potential therapeutic approach to attenuate the progression of BBB injury in diabetic mice via AMPK/HO-1 pathway activation.
Collapse
|
22
|
Association of N 6-methyladenine DNA with plaque progression in atherosclerosis via myocardial infarction-associated transcripts. Cell Death Dis 2019; 10:909. [PMID: 31797919 PMCID: PMC6892866 DOI: 10.1038/s41419-019-2152-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 11/01/2019] [Accepted: 11/05/2019] [Indexed: 01/14/2023]
Abstract
Modification of the novel N6-methyladenine (m6A) DNA implicates this epigenetic mark in human malignant disease, but its role in atherosclerosis (AS) is largely unknown. Here, we found that the leukocyte level of m6A but not 5mC DNA modification was decreased with increasing of carotid plaque size and thickness in 207 AS patients as compared with 142 sex- and age-matched controls. Serum low-density lipoprotein (LDL) and leukocyte m6A levels were associated with the progression of carotid plaque size and thickness. Both LDL level and plaque thickness were also independently and negatively related to m6A level. Reduced m6A level was further confirmed in leukocytes and endothelium in western diet-induced AS mice and in oxidized-LDL (ox-LDL)-treated human endothelium and monocyte cells. Decreased m6A level was closely related to the upregulation of AlkB homolog 1 (ALKBH1), the demethylase of m6A. Silencing of ALKBH1 or hypoxia-inducible factor 1α (HIF1α) could rescue the ox-LDL–increased level of MIAT, a hypoxia-response gene. Mechanically, ox-LDL induced HIF1α for transfer into the nucleus. Nuclear HIF1α bound to the ALKBH1-demethylated MIAT promoter and transcriptionally upregulated its expression. Therefore, elevated ALKBH1 level in endothelium and leukocytes reduced m6A level, which is a novel and sensitive biomarker for AS progression.
Collapse
|
23
|
Zhou H, Huang C, Liu R, Liu C, Ma C, Ren X. Lack of association between serum homocysteine level and middle cerebral artery stenosis. Brain Behav 2019; 9:e01297. [PMID: 31225691 PMCID: PMC6710193 DOI: 10.1002/brb3.1297] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 03/29/2019] [Accepted: 04/06/2019] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVES Lowering homocysteine (HCY) has beneficial effects on vascular events in primary prevention but not in secondary prevention. Research on serum HCY level and middle cerebral artery (MCA) stenosis is lacking. The purpose of the study was to determine the association between these factors and provide more evidence for the prevention of ischemic stroke. METHODS A total of 412 patients (35-93 years old) in the Neurology Department were recruited. Data of clinical and biochemical vascular risk factors were collected. MCA stenosis, including M1, M2, and M3, was determined by brain magnetic resonance angiography (MRA) and classified into stenosis or no stenosis. The differences and associations were analyzed by relevant statistical methods. RESULTS There was no significant difference (p = 0.325) in HCY levels between the MCA stenosis and no stenosis groups at baseline. Logistic regression analysis demonstrated that there was no significant association between HCY levels and MCA stenosis (p = 0.447). After the two groups were matched for age and sex, there was still no difference (p = 0.540 for males and 0.061 for females) or association (p = 0.709 for males and 0.098 for females). In addition, we found that ischemic stroke was more prevalent in the MCA stenosis group and uric acid was higher in males with MCA stenosis. CONCLUSIONS The study indicates a lack of association between serum HCY level and MCA stenosis, which may partially explain the negative results of secondary prevention clinical trials focused on lowering serum HCY level. Future studies on HCY reduction should focus more on primary prevention of ischemic stroke.
Collapse
Affiliation(s)
- Haitao Zhou
- Neurology Department, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, China
| | - Chao Huang
- Neurology Department, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, China
| | - Ruihua Liu
- Neurology Department, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, China
| | - Chao Liu
- Neurology Department, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, China
| | - Congmin Ma
- Neurology Department, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, China
| | - Xiangyang Ren
- Neurology Department, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, China
| |
Collapse
|
24
|
Dysregulation of Epigenetic Mechanisms of Gene Expression in the Pathologies of Hyperhomocysteinemia. Int J Mol Sci 2019; 20:ijms20133140. [PMID: 31252610 PMCID: PMC6651274 DOI: 10.3390/ijms20133140] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 06/21/2019] [Accepted: 06/25/2019] [Indexed: 02/07/2023] Open
Abstract
Hyperhomocysteinemia (HHcy) exerts a wide range of biological effects and is associated with a number of diseases, including cardiovascular disease, dementia, neural tube defects, and cancer. Although mechanisms of HHcy toxicity are not fully uncovered, there has been a significant progress in their understanding. The picture emerging from the studies of homocysteine (Hcy) metabolism and pathophysiology is a complex one, as Hcy and its metabolites affect biomolecules and processes in a tissue- and sex-specific manner. Because of their connection to one carbon metabolism and editing mechanisms in protein biosynthesis, Hcy and its metabolites impair epigenetic control of gene expression mediated by DNA methylation, histone modifications, and non-coding RNA, which underlies the pathology of human disease. In this review we summarize the recent evidence showing that epigenetic dysregulation of gene expression, mediated by changes in DNA methylation and histone N-homocysteinylation, is a pathogenic consequence of HHcy in many human diseases. These findings provide new insights into the mechanisms of human disease induced by Hcy and its metabolites, and suggest therapeutic targets for the prevention and/or treatment.
Collapse
|
25
|
Liang H, Xie X, Song X, Huang M, Su T, Chang X, Liang B, Huang D. Orphan nuclear receptor NR4A1 suppresses hyperhomocysteinemia-induced hepatic steatosis in vitro and in vivo. FEBS Lett 2019; 593:1061-1071. [PMID: 30973961 DOI: 10.1002/1873-3468.13384] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 03/20/2019] [Accepted: 04/08/2019] [Indexed: 02/05/2023]
Abstract
Homocysteine (Hcy) is associated with nonalcoholic fatty liver disease (NAFLD). orphan nuclear receptor subfamily 4 group A member 1 (NR4A1) is involved in hepatic lipid metabolism. However, the potential role of NR4A1 in Hcy-associated NAFLD remains elusive. We aimed to elucidate the regulation of NR4A1 and its significance in Hcy-induced NAFLD. Hcy induced steatosis and elevated the expression of CD36 and FATP2 in HepG2 cells. Furthermore, Hcy enhanced p300 and decreased HDAC7 recruitment to the NR4A1 promoter, resulting in histone H3K27 hyperacetylation and NR4A1 upregulation. Moreover, NR4A1 depletion not only mimicked but also exaggerated the effects of Hcy on steatosis, whereas NR4A1 agonist Cytosporone B (CsnB) blocked Hcy-induced steatosis. In hyperhomocysteinemia (HHcy) mice, CsnB attenuated HHcy-induced hepatic steatosis. Thus, Hcy transiently and rapidly induces NR4A1 expression to reduce Hcy-induced steatosis.
Collapse
Affiliation(s)
- Hongjin Liang
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, China
- Department of Rheumatology, the First Affiliated Hospital, Shantou University Medical College, China
| | - Xina Xie
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, China
- Health Science Center, Institute of Translational Medicine, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, China
| | - Xuhong Song
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, China
| | - Meihui Huang
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, China
- Department of Pathology and Central Laboratory, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, China
| | - Ting Su
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, China
| | - Xiaolan Chang
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, China
| | - Bin Liang
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, China
| | - Dongyang Huang
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, China
| |
Collapse
|
26
|
Yao L, Cao B, Cheng Q, Cai W, Ye C, Liang J, Liu W, Tan L, Yan M, Li B, He J, Hwang SH, Zhang X, Wang C, Ai D, Hammock BD, Zhu Y. Inhibition of soluble epoxide hydrolase ameliorates hyperhomocysteinemia-induced hepatic steatosis by enhancing β-oxidation of fatty acid in mice. Am J Physiol Gastrointest Liver Physiol 2019; 316:G527-G538. [PMID: 30789748 PMCID: PMC6483021 DOI: 10.1152/ajpgi.00148.2018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Hepatic steatosis is the beginning phase of nonalcoholic fatty liver disease, and hyperhomocysteinemia (HHcy) is a significant risk factor. Soluble epoxide hydrolase (sEH) hydrolyzes epoxyeicosatrienoic acids (EETs) and other epoxy fatty acids, attenuating their cardiovascular protective effects. However, the involvement of sEH in HHcy-induced hepatic steatosis is unknown. The current study aimed to explore the role of sEH in HHcy-induced lipid disorder. We fed 6-wk-old male mice a chow diet or 2% (wt/wt) high-metnionine diet for 8 wk to establish the HHcy model. A high level of homocysteine induced lipid accumulation in vivo and in vitro, which was concomitant with the increased activity and expression of sEH. Treatment with a highly selective specific sEH inhibitor (0.8 mg·kg-1·day-1 for the animal model and 1 μM for cells) prevented HHcy-induced lipid accumulation in vivo and in vitro. Inhibition of sEH activated the peroxisome proliferator-activated receptor-α (PPAR-α), as evidenced by elevated β-oxidation of fatty acids and the expression of PPAR-α target genes in HHcy-induced hepatic steatosis. In primary cultured hepatocytes, the effect of sEH inhibition on PPAR-α activation was further confirmed by a marked increase in PPAR-response element luciferase activity, which was reversed by knock down of PPAR-α. Of note, 11,12-EET ligand dependently activated PPAR-α. Thus increased sEH activity is a key determinant in the pathogenesis of HHcy-induced hepatic steatosis, and sEH inhibition could be an effective treatment for HHcy-induced hepatic steatosis. NEW & NOTEWORTHY In the current study, we demonstrated that upregulation of soluble epoxide hydrolase (sEH) is involved in the hyperhomocysteinemia (HHcy)-caused hepatic steatosis in an HHcy mouse model and in murine primary hepatocytes. Improving hepatic steatosis in HHcy mice by pharmacological inhibition of sEH to activate peroxisome proliferator-activated receptor-α was ligand dependent, and sEH could be a potential therapeutic target for the treatment of nonalcoholic fatty liver disease.
Collapse
Affiliation(s)
- Liu Yao
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Boyang Cao
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Qian Cheng
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Wenbin Cai
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Chenji Ye
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Jing Liang
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Wenli Liu
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Lu Tan
- 2Department of Laboratory Animal Science and Technology, Tianjin Medical University, Tianjin, China
| | - Meng Yan
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Bochuan Li
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Jinlong He
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Sung Hee Hwang
- 3Department of Entomology and Nematology and University of California, Davis Comprehensive Cancer Center, Davis, California
| | - Xu Zhang
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Chunjiong Wang
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Ding Ai
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Bruce D. Hammock
- 3Department of Entomology and Nematology and University of California, Davis Comprehensive Cancer Center, Davis, California
| | - Yi Zhu
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| |
Collapse
|
27
|
Wu L, Zhang D, Zhou L, Pei Y, Zhuang Y, Cui W, Chen J. FUN14 domain-containing 1 promotes breast cancer proliferation and migration by activating calcium-NFATC1-BMI1 axis. EBioMedicine 2019; 41:384-394. [PMID: 30803933 PMCID: PMC6442990 DOI: 10.1016/j.ebiom.2019.02.032] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 02/15/2019] [Accepted: 02/15/2019] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND FUN14 domain-containing 1 (FUNDC1), as a novel member of mitochondria-associated endoplasmic reticulum (ER) membranes associates with mitochondrial division and mitophagy. However, the expression profile and functional roles of FUNDC1 remain largely unclear in human cancer biology, including breast cancer (BC). METHODS Immunohistochemistry and western blot analysis were used to determine the expression of FUNDC1 and BMI1 polycomb ring finger oncogene (BMI1). CCK8, cell counting and transwell assays were used to analyze cell proliferation, migration and invasion, respectively. Luciferase reporter and chromatin immunoprecipitation (ChIP) assays were used to detect the transcriptional regulation of Nuclear factor of activated T-cells, cytoplasmic 1 (NFATC1). The prognostic merit of NFATC1 expression was assessed by Kaplan-Meier assay. FINDINGS Immunohistochemistry revealed strong immunostaining for FUNDC1 in cytoplasmic and nuclear membrane distribution in BC tissues as compared with normal breast epithelium. Kaplan-Meier survival analysis showed worse outcome for BC patients with high FUNDC1 expression. In vitro assay of gain- and loss-of-function of FUNDC1 suggested that FUNDC1 could stimulate BC cell proliferation, migration and invasion. Furthermore, elevated FUNDC1 level promoted Ca2+ cytosol influx from ER and extracellular, as well as NFATC1 nuclear translocation and activity. Nuclear NFATC1 bound to the BMI1 gene promoter and transcriptionally upregulated its expression. Notably, BMI1 overexpression could rescue the loss of function of FUNDC1. Co-expression of FUNDC1 and BMI1 in BC patients predicted worse prognosis than without either expression. INTERPRETATION FUNDC1 might promote BC progression by activating the Ca2+-NFATC1-BMI1 axis. This pathway may be promising for developing multiple targets for BC therapy.
Collapse
Affiliation(s)
- Lianpin Wu
- Department of Cardiology, The Second Affiliated Hospital of Wenzhou Medical University, 109 Xueyuan Road, Wenzhou 325027, Zhejiang, PR China
| | - Donghong Zhang
- Center for Molecular and Translational Medicine, Georgia State University, Research Science Center, 157 Decatur St SE, Atlanta, GA 30303. USA
| | - Li Zhou
- Department of Gynecological Oncology, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou 515031, PR China
| | - Yuqing Pei
- Clinical Laboratory, National Cancer Center/National Clinical Research Center for Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, PR China
| | - Yixuan Zhuang
- Oncological Research Lab, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou 515031, PR China
| | - Wei Cui
- Clinical Laboratory, National Cancer Center/National Clinical Research Center for Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, PR China
| | - Jiongyu Chen
- Oncological Research Lab, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou 515031, PR China.
| |
Collapse
|
28
|
Liu X, Qin Z, Liu C, Song M, Luo X, Zhao H, Qian D, Chen J, Huang L. Nox4 and soluble epoxide hydrolase synergistically mediate homocysteine-induced inflammation in vascular smooth muscle cells. Vascul Pharmacol 2019; 120:106544. [PMID: 30610956 DOI: 10.1016/j.vph.2019.01.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 12/05/2018] [Accepted: 01/01/2019] [Indexed: 01/03/2023]
Abstract
BACKGROUND Hyperhomocysteinemia leads to a vascular smooth muscle cell (VSMC) inflammatory response. Meanwhile, Nox4 dependent reactive oxygen species (ROS) signaling and soluble epoxide hydrolase (sEH)/epoxyeicosatrienoic acids (EETs) are both involved in vascular inflammation. Herein, we hypothesized that Nox4 and soluble epoxide hydrolase cross regulated during homocysteine-induced VSMC inflammation. METHODS AND RESULTS In cultured VSMCs, the expression of the inflammatory factors VCAM1 and ICAM1 was measured by real-time PCR and Western blotting, while supernatant MCP1 was measured by ELISA. Upon VSMC stimulation with 50 μΜ homocysteine, we observed the VCAM1 and ICAM1 mRNA levels were increased by 1.15 and 1.0 folds, respectively. The MCP1 levels in the supernatant of cultured VSMCs treated with 100 μΜ increased to 1.76 folds. As expected, homocysteine induced Nox4 expression and Nox4-dependent ROS generation. The sEH expression was also upregulated in the presence of homocysteine in a dose-dependent manner. Furthermore, we knocked down Nox4 with siRNA. Knockdown of Nox4 decreased ROS generation and homocysteine-induced sEH expression. Overexpression of Nox4 with an adenovirus stimulated sEH expression. Similarly, knockdown or chemical inhibition of sEH blunted the upregulation of Nox4 by homocysteine. In vivo, in homocysteine-fed mice, concomitant upregulation of Nox4 and sEH was associated with increased VCAM1 and ICAM1 expression in the aortic wall. CONCLUSIONS The inflammatory response induced by homocysteine in VSMCs was accompanied by Nox4 and sEH upregulation. Nox4 and soluble epoxide hydrolase synergistically contribute to homocysteine-induced inflammation.
Collapse
Affiliation(s)
- Xi Liu
- Department of Cardiology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Zhexue Qin
- Department of Cardiology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Chuan Liu
- Department of Cardiology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Mingbao Song
- Department of Cardiology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Xiaolin Luo
- Department of Cardiology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Hongqing Zhao
- Department of Cardiology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Dehui Qian
- Department of Cardiology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Jianfei Chen
- Department of Cardiology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Lan Huang
- Department of Cardiology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China.
| |
Collapse
|
29
|
Chen L, Lei L, Li T, Yan J, Jiang J. A20 alleviates the vascular remodeling induced by homocysteine. Am J Transl Res 2018; 10:3991-4003. [PMID: 30662645 PMCID: PMC6325502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 10/22/2018] [Indexed: 06/09/2023]
Abstract
Hyperhomocysteinemia is an independent risk factor for multiple cardiovascular diseases. The pathogenesis of homocysteine-induced vascular remodeling has not yet been elucidated. In vivo, we established hyperhomocysteinemia model with high L-methionine diet and found that the accumulation of macrophages, proliferation of VSMCs and decreased expression of A20 in the aorta of mice fed with high methionine diet. In vitro, we found that the overexpression of A20 suppressed the nuclear translocation of NF-kappaB p65 and attenuated homocysteine-induced proliferation and migration of VSMCs. However, down-regulation of A20 reversed the protective effects above. Moreover, A20 attenuated homocysteine-induced vascular remodeling by alleviating the activation of inflammation and suppressing the proliferation and migration of VSMCs through enhanced nuclear translocation of IRF3 and binding to PPAR-γ.
Collapse
Affiliation(s)
- Liang Chen
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei Province, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and TechnologyWuhan 430030, Hubei Province, China
| | - Lei Lei
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei Province, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and TechnologyWuhan 430030, Hubei Province, China
| | - Tianyu Li
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei Province, China
| | - Jiangtao Yan
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei Province, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and TechnologyWuhan 430030, Hubei Province, China
| | - Jiangang Jiang
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei Province, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and TechnologyWuhan 430030, Hubei Province, China
| |
Collapse
|
30
|
Wu X, Zhang L, Miao Y, Yang J, Wang X, Wang CC, Feng J, Wang L. Homocysteine causes vascular endothelial dysfunction by disrupting endoplasmic reticulum redox homeostasis. Redox Biol 2018; 20:46-59. [PMID: 30292945 PMCID: PMC6174864 DOI: 10.1016/j.redox.2018.09.021] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 09/18/2018] [Accepted: 09/26/2018] [Indexed: 02/07/2023] Open
Abstract
Endothelial dysfunction induced by hyperhomocysteinemia (HHcy) plays a critical role in vascular pathology. However, little is known about the role of endoplasmic reticulum (ER) redox homeostasis in HHcy-induced endothelial dysfunction. Here, we show that Hcy induces ER oxidoreductin-1α (Ero1α) expression with ER stress and inflammation in human umbilical vein endothelial cells and in the arteries of HHcy mice. Hcy upregulates Ero1α expression by promoting binding of hypoxia-inducible factor 1α to the ERO1A promoter. Notably, Hcy rather than other thiol agents markedly increases the GSH/GSSG ratio in the ER, therefore allosterically activating Ero1α to produce H2O2 and trigger ER oxidative stress. By contrast, the antioxidant pathway mediated by ER glutathione peroxidase 7 (GPx7) is downregulated in HHcy mice. Ero1α knockdown and GPx7 overexpression protect the endothelium from HHcy-induced ER oxidative stress and inflammation. Our work suggests that targeting ER redox homeostasis could be used as an intervention for HHcy-related vascular diseases.
Collapse
Affiliation(s)
- Xun Wu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lihui Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yütong Miao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China
| | - Juan Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China
| | - Xian Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China
| | - Chih-Chen Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Juan Feng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China.
| | - Lei Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
31
|
Peng L, Huang YT, Chen J, Zhuang YX, Zhang F, Chen JY, Zhou L, Zhang DH. Osthole sensitizes with radiotherapy to suppress tumorigenesis of human nasopharyngeal carcinoma in vitro and in vivo. Cancer Manag Res 2018; 10:5471-5477. [PMID: 30519095 PMCID: PMC6233473 DOI: 10.2147/cmar.s182798] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Radiotherapy is one of the most comment and useful treatment for nasopharyngeal carcinoma (NPC), but the radioresistance remains a major obstacle. Osthole, a natural coumarin derivative, has been shown to have anti-tumor and anti-inflammatory activity. However, the relationship between osthole and NPC treatment, especially for radiotherapy, is still elusive. METHODS Osthole with or without X ray radiotherapy treated with CNE2 cells, a human EC cell line. Cell viability, proliferation, migration and apoptosis were measured by MTT, colony formation, Annexin V/PI double staining, Transwell assay, respectively. NPC tumor models were established on BALB/c nude mice by subcutaneously injection of CNE2 cells and the effect of osthole and radiotherapy on tumor growth in vivo was studied. RESULTS We found that in a dose-dependent manner, osthole could individually, and synergistically with radiotherapy, reduce NPC cell (CNE2) viability, proliferation, migration, and invasion, and induce apoptosis, respectively. This effect of anti-tumor growth and induction of apoptosis was further confirmed in mice induced by subcutaneously injection with CNE2 cells and following treated with osthole or/and radiation. CONCLUSION Osthole increases the effect of radiotherapy on anti-human nasopharyngeal cancer.
Collapse
Affiliation(s)
- Lin Peng
- Clinical Laboratory, Cancer Hospital of Shantou University Medical College, Shantou 515031, People's Republic of China
| | - Yi-Teng Huang
- Health Care Center, The First Affiliated Hospital of Shantou University Medical College. Shantou 515041, People's Republic of China
| | - Jian Chen
- Department of Radiotherapy, Cancer Hospital of Shantou University Medical College, Shantou 515031, People's Republic of China
| | - Yi-Xuan Zhuang
- Department of Radiotherapy, Cancer Hospital of Shantou University Medical College, Shantou 515031, People's Republic of China
| | - Fan Zhang
- Oncological Research Lab, Cancer Hospital of Shantou University Medical College, Shantou 515031, People's Republic of China,
| | - Jiong-Yu Chen
- Oncological Research Lab, Cancer Hospital of Shantou University Medical College, Shantou 515031, People's Republic of China,
| | - Li Zhou
- Department of Gynecological Oncology, Cancer Hospital of Shantou University Medical College, Shantou 515031, People's Repulic of China
| | - Dong-Hong Zhang
- Department of Cardiology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang, People's Repulic of China,
| |
Collapse
|
32
|
Ma L, Yan M, Kong X, Jiang Y, Zhao T, Zhao H, Liu Q, Zhang H, Liu P, Cao Y, Li P. Association of EPHX2 R287Q Polymorphism with Diabetic Nephropathy in Chinese Type 2 Diabetic Patients. J Diabetes Res 2018; 2018:2786470. [PMID: 29629376 PMCID: PMC5832179 DOI: 10.1155/2018/2786470] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 10/11/2017] [Indexed: 01/19/2023] Open
Abstract
The aim of this study was to investigate the relationship between EPHX2 rs751141 (R287Q polymorphism) and diabetic nephropathy (DN) in Chinese type 2 diabetes (T2D). This case-control study explored the association between EPHX2 rs751141 and DN in a total of 870 Chinese T2D patients (406 T2D patients with DN and 464 T2D patients without DN). DNA was extracted from peripheral leukocytes of the patients and rs751141 was genotyped. The A allele frequency of rs751141 was significantly lower in DN patients (20.94%) compared with non-DN controls (27.8%) (P = 0.001), and the A allele of rs751141 was associated with a significantly lower risk of DN after adjustment for multiple covariates in the additive genetic model (OR = 0.68, 95% CI = 0.52-0.88, P = 0.004). Significant association between rs751141 and homocysteine (Hcy) level on the risk of DN was observed, indicating that in patients with the highest Hcy levels, the A allele showed marked association with lower risk of DN in all three genetic models. In conclusion, the A allele of exonic polymorphism in EPHX2 rs751141 is negatively associated with the incidence of DN in the Chinese T2D population, which could be modulated by Hcy level status.
Collapse
Affiliation(s)
- Liang Ma
- Clinical Laboratory, China-Japan Friendship Hospital, Beijing, China
| | - Meihua Yan
- Beijing Key Lab Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Science, China-Japan Friendship Hospital, Beijing, China
| | - Xiaomu Kong
- Department of Endocrinology, China-Japan Friendship Hospital, Beijing, China
| | - Yongwei Jiang
- Clinical Laboratory, China-Japan Friendship Hospital, Beijing, China
| | - Tingting Zhao
- Beijing Key Lab Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Science, China-Japan Friendship Hospital, Beijing, China
| | - Hailing Zhao
- Beijing Key Lab Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Science, China-Japan Friendship Hospital, Beijing, China
| | - Qian Liu
- Clinical Laboratory, China-Japan Friendship Hospital, Beijing, China
| | - Haojun Zhang
- Beijing Key Lab Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Science, China-Japan Friendship Hospital, Beijing, China
| | - Peng Liu
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yongtong Cao
- Clinical Laboratory, China-Japan Friendship Hospital, Beijing, China
| | - Ping Li
- Beijing Key Lab Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Science, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
33
|
Zhang D, Wang Y, Lu P, Wang P, Yuan X, Yan J, Cai C, Chang CP, Zheng D, Wu B, Zhou B. REST regulates the cell cycle for cardiac development and regeneration. Nat Commun 2017; 8:1979. [PMID: 29215012 PMCID: PMC5719406 DOI: 10.1038/s41467-017-02210-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Accepted: 11/13/2017] [Indexed: 12/17/2022] Open
Abstract
Despite the importance of cardiomyocyte proliferation in cardiac development and regeneration, the mechanisms that promote cardiomyocyte cell cycle remain incompletely understood. RE1 silencing transcription factor (REST) is a transcriptional repressor of neuronal genes. Here we show that REST also regulates the cardiomyocyte cell cycle. REST binds and represses the cell cycle inhibitor gene p21 and is required for mouse cardiac development and regeneration. Rest deletion de-represses p21 and inhibits the cardiomyocyte cell cycle and proliferation in embryonic or regenerating hearts. By contrast, REST overexpression in cultured cardiomyocytes represses p21 and increases proliferation. We further show that p21 knockout rescues cardiomyocyte cell cycle and proliferation defects resulting from Rest deletion. Our study reveals a REST-p21 regulatory axis as a mechanism for cell cycle progression in cardiomyocytes, which might be exploited therapeutically to enhance cardiac regeneration.
Collapse
Affiliation(s)
- Donghong Zhang
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Yidong Wang
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Pengfei Lu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Ping Wang
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Xinchun Yuan
- Department of Medical Ultrasound, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Jianyun Yan
- Department of Developmental and Regenerative Biology, The Black Family Stem Cell Institute, and The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Chenleng Cai
- Department of Developmental and Regenerative Biology, The Black Family Stem Cell Institute, and The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ching-Pin Chang
- Department of Medicine, Indian University School of Medicine, Indianapolis, IN, 46202, USA
| | - Deyou Zheng
- Departments of Genetics, Neurology and Neuroscience, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Bingruo Wu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Bin Zhou
- Departments of Genetics, Pediatrics, and Medicine (Cardiology), The Wilf Cardiovascular Research Institute, The Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
- Department of Cardiology of First Affiliated Hospital and State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
| |
Collapse
|
34
|
Therapeutic potential of omega-3 fatty acid-derived epoxyeicosanoids in cardiovascular and inflammatory diseases. Pharmacol Ther 2017; 183:177-204. [PMID: 29080699 DOI: 10.1016/j.pharmthera.2017.10.016] [Citation(s) in RCA: 142] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Numerous benefits have been attributed to dietary long-chain omega-3 polyunsaturated fatty acids (n-3 LC-PUFAs), including protection against cardiac arrhythmia, triglyceride-lowering, amelioration of inflammatory, and neurodegenerative disorders. This review covers recent findings indicating that a variety of these beneficial effects are mediated by "omega-3 epoxyeicosanoids", a class of novel n-3 LC-PUFA-derived lipid mediators, which are generated via the cytochrome P450 (CYP) epoxygenase pathway. CYP enzymes, previously identified as arachidonic acid (20:4n-6; AA) epoxygenases, accept eicosapentaenoic acid (20:5n-3; EPA) and docosahexaenoic acid (22:6n-3; DHA), the major fish oil n-3 LC-PUFAs, as efficient alternative substrates. In humans and rodents, dietary EPA/DHA supplementation causes a profound shift of the endogenous CYP-eicosanoid profile from AA- to EPA- and DHA-derived metabolites, increasing, in particular, the plasma and tissue levels of 17,18-epoxyeicosatetraenoic acid (17,18-EEQ) and 19,20-epoxydocosapentaenoic acid (19,20-EDP). Based on preclinical studies, these omega-3 epoxyeicosanoids display cardioprotective, vasodilatory, anti-inflammatory, and anti-allergic properties that contribute to the beneficial effects of n-3 LC-PUFAs in diverse disease conditions ranging from cardiac disease, bronchial disorders, and intraocular neovascularization, to allergic intestinal inflammation and inflammatory pain. Increasing evidence also suggests that background nutrition as well as genetic and disease state-related factors could limit the response to EPA/DHA-supplementation by reducing the formation and/or enhancing the degradation of omega-3 epoxyeicosanoids. Recently, metabolically robust synthetic analogs mimicking the biological activities of 17,18-EEQ have been developed. These drug candidates may overcome limitations of dietary EPA/DHA supplementation and provide novel options for the treatment of cardiovascular and inflammatory diseases.
Collapse
|
35
|
Altered Protein Expression of Cardiac CYP2J and Hepatic CYP2C, CYP4A, and CYP4F in a Mouse Model of Type II Diabetes-A Link in the Onset and Development of Cardiovascular Disease? Pharmaceutics 2017; 9:pharmaceutics9040044. [PMID: 29023376 PMCID: PMC5750650 DOI: 10.3390/pharmaceutics9040044] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 09/30/2017] [Accepted: 10/06/2017] [Indexed: 12/23/2022] Open
Abstract
Arachidonic acid can be metabolized by cytochrome P450 (CYP450) enzymes in a tissue- and cell-specific manner to generate vasoactive products such as epoxyeicosatrienoic acids (EETs-cardioprotective) and hydroxyeicosatetraenoic acids (HETEs-cardiotoxic). Type II diabetes is a well-recognized risk factor for developing cardiovascular disease. A mouse model of Type II diabetes (C57BLKS/J-db/db) was used. After sacrifice, livers and hearts were collected, washed, and snap frozen. Total proteins were extracted. Western blots were performed to assess cardiac CYP2J and hepatic CYP2C, CYP4A, and CYP4F protein expression, respectively. Significant decreases in relative protein expression of cardiac CYP2J and hepatic CYP2C were observed in Type II diabetes animals compared to controls (CYP2J: 0.80 ± 0.03 vs. 1.05 ± 0.06, n = 20, p < 0.001); (CYP2C: 1.56 ± 0.17 vs. 2.21 ± 0.19, n = 19, p < 0.01). In contrast, significant increases in relative protein expression of both hepatic CYP4A and CYP4F were noted in Type II diabetes mice compared to controls (CYP4A: 1.06 ± 0.09 vs. 0.18 ± 0.01, n = 19, p < 0.001); (CYP4F: 2.53 ± 0.22 vs. 1.10 ± 0.07, n = 19, p < 0.001). These alterations induced by Type II diabetes in the endogenous pathway (CYP450) of arachidonic acid metabolism may increase the risk for cardiovascular disease by disrupting the fine equilibrium between cardioprotective (CYP2J/CYP2C-generated) and cardiotoxic (CYP4A/CYP4F-generated) metabolites of arachidonic acid.
Collapse
|
36
|
Mandaviya PR, Aïssi D, Dekkers KF, Joehanes R, Kasela S, Truong V, Stolk L, Heemst DV, Ikram MA, Lindemans J, Slagboom PE, Trégouët DA, Uitterlinden AG, Wei C, Wells P, Gagnon F, van Greevenbroek MM, Heijmans BT, Milani L, Morange PE, van Meurs JB, Heil SG. Homocysteine levels associate with subtle changes in leukocyte DNA methylation: an epigenome-wide analysis. Epigenomics 2017; 9:1403-1422. [PMID: 28990796 DOI: 10.2217/epi-2017-0038] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
AIM Homocysteine (Hcy) is a sensitive marker of one-carbon metabolism. Higher Hcy levels have been associated with global DNA hypomethylation. We investigated the association between plasma Hcy and epigenome-wide DNA methylation in leukocytes. METHODS Methylation was measured using Illumina 450 k arrays in 2035 individuals from six cohorts. Hcy-associated differentially methylated positions and regions were identified using meta-analysis. RESULTS Three differentially methylated positions cg21607669 (SLC27A1), cg26382848 (AJUBA) and cg10701000 (KCNMA1) at chromosome 19, 14 and 10, respectively, were significantly associated with Hcy. In addition, we identified 68 Hcy-associated differentially methylated regions, the most significant of which was a 1.8-kb spanning domain (TNXB/ATF6B) at chromosome 6. CONCLUSION We identified novel epigenetic loci associated with Hcy levels, of which specific role needs to be further validated.
Collapse
Affiliation(s)
- Pooja R Mandaviya
- Department of Clinical Chemistry, Erasmus University Medical Center, Rotterdam, The Netherlands.,Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Dylan Aïssi
- Sorbonne Universités, UPMC Univ. Paris 06, INSERM, UMR_S 1166, Team Genomics & Pathophysiology of Cardiovascular Diseases, Paris, France.,ICAN Institute for Cardiometabolism & Nutrition, Paris, France
| | - Koen F Dekkers
- Molecular Epidemiology Section, Department of Medical Statistics & Bioinformatics, Leiden University Medical Center, Leiden, The Netherlands
| | - Roby Joehanes
- Institute for Aging Research, Hebrew SeniorLife, Harvard Medical School, Boston, MA, USA
| | - Silva Kasela
- Estonian Genome Center, University of Tartu, Tartu, Estonia.,Institute of Molecular & Cell Biology, University of Tartu, Tartu, Estonia
| | - Vinh Truong
- Division of Epidemiology, Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - Lisette Stolk
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Diana van Heemst
- Department of Gerontology & Geriatrics Section, Leiden University Medical Center, Leiden, The Netherlands
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jan Lindemans
- Department of Clinical Chemistry, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - P Eline Slagboom
- Molecular Epidemiology Section, Department of Medical Statistics & Bioinformatics, Leiden University Medical Center, Leiden, The Netherlands
| | - David-Alexandre Trégouët
- Sorbonne Universités, UPMC Univ. Paris 06, INSERM, UMR_S 1166, Team Genomics & Pathophysiology of Cardiovascular Diseases, Paris, France.,ICAN Institute for Cardiometabolism & Nutrition, Paris, France
| | - André G Uitterlinden
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Chen Wei
- Department of Epidemiology, Tulane University, New Orleans, LA, USA
| | - Phil Wells
- Department of Medicine, Ottawa Hospital Research Institute, Ottawa, Canada
| | - France Gagnon
- Division of Epidemiology, Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - Marleen Mj van Greevenbroek
- Department of Internal Medicine & School for Cardiovascular Diseases (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - Bastiaan T Heijmans
- Molecular Epidemiology Section, Department of Medical Statistics & Bioinformatics, Leiden University Medical Center, Leiden, The Netherlands
| | - Lili Milani
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Pierre-Emmanuel Morange
- Laboratory of Haematology, La Timone Hospital, Marseille, France.,Institut National pour la Santé et la Recherche Médicale (INSERM), Unité Mixte de Recherche en Santé (UMR_S) 1062, Nutrition Obesity & Risk of Thrombosis, Aix-Marseille University, Marseille, France
| | - Joyce Bj van Meurs
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Sandra G Heil
- Department of Clinical Chemistry, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | |
Collapse
|
37
|
Fu Y, Wang X, Kong W. Hyperhomocysteinaemia and vascular injury: advances in mechanisms and drug targets. Br J Pharmacol 2017; 175:1173-1189. [PMID: 28836260 DOI: 10.1111/bph.13988] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 07/27/2017] [Accepted: 08/12/2017] [Indexed: 12/14/2022] Open
Abstract
Homocysteine is a sulphur-containing non-proteinogenic amino acid. Hyperhomocysteinaemia (HHcy), the pathogenic elevation of plasma homocysteine as a result of an imbalance of its metabolism, is an independent risk factor for various vascular diseases, such as atherosclerosis, hypertension, vascular calcification and aneurysm. Treatments aimed at lowering plasma homocysteine via dietary supplementation with folic acids and vitamin B are more effective in preventing vascular disease where the population has a normally low folate consumption than in areas with higher dietary folate. To date, the mechanisms of HHcy-induced vascular injury are not fully understood. HHcy increases oxidative stress and its downstream signalling pathways, resulting in vascular inflammation. HHcy also causes vascular injury via endoplasmic reticulum stress. Moreover, HHcy up-regulates pathogenic genes and down-regulates protective genes via DNA demethylation and methylation respectively. Homocysteinylation of proteins induced by homocysteine also contributes to vascular injury by modulating intracellular redox state and altering protein function. Furthermore, HHcy-induced vascular injury leads to neuronal damage and disease. Also, an HHcy-activated sympathetic system and HHcy-injured adipose tissue also cause vascular injury, thus demonstrating the interactions between the organs injured by HHcy. Here, we have summarized the recent developments in the mechanisms of HHcy-induced vascular injury, which are further considered as potential therapeutic targets in this condition. LINKED ARTICLES This article is part of a themed section on Spotlight on Small Molecules in Cardiovascular Diseases. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.8/issuetoc.
Collapse
Affiliation(s)
- Yi Fu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Health Science Center, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Xian Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Health Science Center, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Health Science Center, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| |
Collapse
|
38
|
Soluble epoxide hydrolase activation by S -nitrosation contributes to cardiac ischemia–reperfusion injury. J Mol Cell Cardiol 2017; 110:70-79. [DOI: 10.1016/j.yjmcc.2017.07.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 07/14/2017] [Accepted: 07/19/2017] [Indexed: 12/16/2022]
|
39
|
Hyperhomocysteinemia results from and promotes hepatocellular carcinoma via CYP450 metabolism by CYP2J2 DNA methylation. Oncotarget 2017; 8:15377-15392. [PMID: 28030819 PMCID: PMC5362492 DOI: 10.18632/oncotarget.14165] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 11/24/2016] [Indexed: 12/03/2022] Open
Abstract
Hyperhomocysteinemia (HHcy) can result from liver disease or dysfunction and further alters intracellular lipid metabolism. Cytochrome P450 (CYP) arachidonic acid epoxygenases are expressed in human cancers and promote cancer metastasis. This study explored the interaction of Hcy and CYP450 metabolism in hepatocellular carcinoma (HCC). The levels of 4-epoxyeicosatrienoic acid (EET) isomers and their generative enzyme CYP2J2 level as well as intracellular Hcy level were higher in 42 cases of HCC than in paired non-tumor tissue. Elevated Hcy-decreased DNA methylation on SP1/AP1 binding motifs and enhancement on the CYP2J2 promoter via ERK1/2 signaling was essential for CYP2J2 upregulation and EET metabolism. Increased Hcy level enhanced the neoplastic cellular phenotype, which was reversed by CYP2J2 knockdown in vitro. Furthermore, tumor growth and size as well as patterns of CYP2J2 expression and DNA demethylation were increased with HHcy in mice induced orthotopically by 2% (wt/wt) L-methionine with or without folate deficiency. Moreover, the effect was attenuated by shRNA knockdown of CYP2J2. Thus, HHcy results from but can also promote hepatocarcingenesis via CYP450-EET metabolism by crosstalk of DNA demethylation of CYP2J2 and ERK1/2 signaling.
Collapse
|
40
|
Xie HH, Li J, Li PQ, Zhang AA, Li Y, Wang YZ, Xie DX, Xie XD. A genetic variant in a homocysteine metabolic gene that increases the risk of congenital cardiac septal defects in Han Chinese populations. IUBMB Life 2017; 69:700-705. [PMID: 28834160 DOI: 10.1002/iub.1651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 06/13/2017] [Accepted: 06/20/2017] [Indexed: 11/06/2022]
Abstract
Elevated homocysteine levels are known to be a risk factor for congenital cardiac septal defects (CCSDs), but the mechanism underlying this effect is unknown. The genetic variants that were significantly associated with circulating homocysteine concentrations have been systematically identified through the genome-wide association studies of one-carbon core metabolites. To examine the role of the genome-wide significant homocysteine related variants in the occurrence of CCSDs, we investigated the association between these variants and CCSDs in Han Chinese populations. Five variants of the genome-wide significant homocysteine-related genes were selected for analysis in two stages of case-controlled studies with a total of 904 CCSD patients and 997 controls. SYT9 expression was detected in human cardiovascular tissue using qRT-PCR. The intronic variant rs11041321 of the SYT9 gene was associated with an increased risk of developing CCSDs in both the separate and combined case-controlled studies. Combined samples from the two stage cohorts had a significant elevation in CCSD risk for the T allele (OR = 1.43, P = 2.6 × 10-6 ), CT genotype and TT genotype (CT: OR = 1.30, TT: OR = 2.21; P = 1 × 10-4 ) compared with the wild-type C allele and CC genotype, respectively. The risky T allele carriers exhibited decreased SYT9 mRNA expression, compared with wild-type C allele carriers. The intronic SYT9 variant rs11041321, which exhibits a significant genome-wide association with circulating homocysteine, was associated with the occurrence of CCSDs. This finding helps to characterize the unexpected role of SYT9 in homocysteine metabolism and the development of CCSDs, which further highlighted the interplay of diet, genetics, and human birth defects. © 2017 IUBMB Life, 69(9):700-705, 2017.
Collapse
Affiliation(s)
- Han-Hui Xie
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Jiong Li
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China.,Gansu Cardiovascular Institute, Lanzhou, 730050, China
| | - Pei-Qiang Li
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - An-An Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Yi Li
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Yan-Zhen Wang
- Gansu Cardiovascular Institute, Lanzhou, 730050, China
| | - Ding-Xiong Xie
- Gansu Cardiovascular Institute, Lanzhou, 730050, China.,The Second Hospital of Lanzhou University, Lanzhou, 730030, China
| | - Xiao-Dong Xie
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China.,Gansu Cardiovascular Institute, Lanzhou, 730050, China
| |
Collapse
|
41
|
Zhang D, Wu B, Wang P, Wang Y, Lu P, Nechiporuk T, Floss T, Greally JM, Zheng D, Zhou B. Non-CpG methylation by DNMT3B facilitates REST binding and gene silencing in developing mouse hearts. Nucleic Acids Res 2017; 45:3102-3115. [PMID: 27956497 PMCID: PMC5389556 DOI: 10.1093/nar/gkw1258] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Revised: 10/25/2016] [Accepted: 12/01/2016] [Indexed: 12/12/2022] Open
Abstract
The dynamic interaction of DNA methylation and transcription factor binding in regulating spatiotemporal gene expression is essential for embryogenesis, but the underlying mechanisms remain understudied. In this study, using mouse models and integration of in vitro and in vivo genetic and epigenetic analyses, we show that the binding of REST (repressor element 1 (RE1) silencing transcription factor; also known as NRSF) to its cognate RE1 sequences is temporally regulated by non-CpG methylation. This process is dependent on DNA methyltransferase 3B (DNMT3B) and leads to suppression of adult cardiac genes in developing hearts. We demonstrate that DNMT3B preferentially mediates non-CpG methylation of REST-targeted genes in the developing heart. Downregulation of DNMT3B results in decreased non-CpG methylation of RE1 sequences, reduced REST occupancy, and consequently release of the transcription suppression during later cardiac development. Together, these findings reveal a critical gene silencing mechanism in developing mammalian hearts that is regulated by the dynamic interaction of DNMT3B-mediated non-CpG methylation and REST binding.
Collapse
Affiliation(s)
- Donghong Zhang
- Departments of Genetics, Pediatrics, and Medicine (Cardiology), Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Bingruo Wu
- Departments of Genetics, Pediatrics, and Medicine (Cardiology), Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Ping Wang
- Department of Neurology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Yidong Wang
- Departments of Genetics, Pediatrics, and Medicine (Cardiology), Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Pengfei Lu
- Departments of Genetics, Pediatrics, and Medicine (Cardiology), Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Tamilla Nechiporuk
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Thomas Floss
- German Research Center for Environmental Health, Neuherberg, Germany
| | - John M. Greally
- Departments of Genetics, Medicine (Hematology), and Pediatrics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Deyou Zheng
- Departments of Genetics, Neurology, and Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Bin Zhou
- Departments of Genetics, Pediatrics, and Medicine (Cardiology), Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
42
|
Vanhoutte PM, Shimokawa H, Feletou M, Tang EHC. Endothelial dysfunction and vascular disease - a 30th anniversary update. Acta Physiol (Oxf) 2017; 219:22-96. [PMID: 26706498 DOI: 10.1111/apha.12646] [Citation(s) in RCA: 581] [Impact Index Per Article: 83.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 10/27/2015] [Accepted: 12/17/2015] [Indexed: 02/06/2023]
Abstract
The endothelium can evoke relaxations of the underlying vascular smooth muscle, by releasing vasodilator substances. The best-characterized endothelium-derived relaxing factor (EDRF) is nitric oxide (NO) which activates soluble guanylyl cyclase in the vascular smooth muscle cells, with the production of cyclic guanosine monophosphate (cGMP) initiating relaxation. The endothelial cells also evoke hyperpolarization of the cell membrane of vascular smooth muscle (endothelium-dependent hyperpolarizations, EDH-mediated responses). As regards the latter, hydrogen peroxide (H2 O2 ) now appears to play a dominant role. Endothelium-dependent relaxations involve both pertussis toxin-sensitive Gi (e.g. responses to α2 -adrenergic agonists, serotonin, and thrombin) and pertussis toxin-insensitive Gq (e.g. adenosine diphosphate and bradykinin) coupling proteins. New stimulators (e.g. insulin, adiponectin) of the release of EDRFs have emerged. In recent years, evidence has also accumulated, confirming that the release of NO by the endothelial cell can chronically be upregulated (e.g. by oestrogens, exercise and dietary factors) and downregulated (e.g. oxidative stress, smoking, pollution and oxidized low-density lipoproteins) and that it is reduced with ageing and in the course of vascular disease (e.g. diabetes and hypertension). Arteries covered with regenerated endothelium (e.g. following angioplasty) selectively lose the pertussis toxin-sensitive pathway for NO release which favours vasospasm, thrombosis, penetration of macrophages, cellular growth and the inflammatory reaction leading to atherosclerosis. In addition to the release of NO (and EDH, in particular those due to H2 O2 ), endothelial cells also can evoke contraction of the underlying vascular smooth muscle cells by releasing endothelium-derived contracting factors. Recent evidence confirms that most endothelium-dependent acute increases in contractile force are due to the formation of vasoconstrictor prostanoids (endoperoxides and prostacyclin) which activate TP receptors of the vascular smooth muscle cells and that prostacyclin plays a key role in such responses. Endothelium-dependent contractions are exacerbated when the production of nitric oxide is impaired (e.g. by oxidative stress, ageing, spontaneous hypertension and diabetes). They contribute to the blunting of endothelium-dependent vasodilatations in aged subjects and essential hypertensive and diabetic patients. In addition, recent data confirm that the release of endothelin-1 can contribute to endothelial dysfunction and that the peptide appears to be an important contributor to vascular dysfunction. Finally, it has become clear that nitric oxide itself, under certain conditions (e.g. hypoxia), can cause biased activation of soluble guanylyl cyclase leading to the production of cyclic inosine monophosphate (cIMP) rather than cGMP and hence causes contraction rather than relaxation of the underlying vascular smooth muscle.
Collapse
Affiliation(s)
- P. M. Vanhoutte
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy; Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong City Hong Kong
| | - H. Shimokawa
- Department of Cardiovascular Medicine; Tohoku University; Sendai Japan
| | - M. Feletou
- Department of Cardiovascular Research; Institut de Recherches Servier; Suresnes France
| | - E. H. C. Tang
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy; Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong City Hong Kong
- School of Biomedical Sciences; Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong City Hong Kong
| |
Collapse
|
43
|
Ma L, Jiang Y, Kong X, Yan M, Zhao T, Zhao H, Liu Q, Zhang H, Cao Y, Li P. Synergistic Effect of the MTHFR C677T and EPHX2 G860A Polymorphism on the Increased Risk of Ischemic Stroke in Chinese Type 2 Diabetic Patients. J Diabetes Res 2017; 2017:6216205. [PMID: 28409162 PMCID: PMC5376931 DOI: 10.1155/2017/6216205] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 02/14/2017] [Accepted: 03/07/2017] [Indexed: 01/19/2023] Open
Abstract
The aim of this study was to investigate the relationship between the combined effect of MTHFR C677T (rs1801133) and EPHX2 G860A (rs751141) polymorphism and ischemic stroke in Chinese T2DM patients. This case-control study included a total of 626 Chinese T2DM patients (236 T2DM patients with ischemic stroke and 390 T2DM patients without ischemic stroke). The rs1801133 and rs751141 were genotyped using real-time polymerase chain reaction. Statistical analysis was performed with SPSS 17.0. Results showed that the combined effect of MTHFR TT and EPHX2 GG or GA + AA genotype has a higher risk of ischemic stroke compared with the control group (combined effect of MTHFR CC and EPHX2 GA + AA genotypes; OR = 3.46 and OR = 3.42, resp.; P = .001 and P = .002, resp.). The A allele showed marked association with a lower risk of ischemic stroke in patients with the lowest Hcy levels under additive, recessive, and dominant genetic models (OR = 0.45, OR = 0.11, and OR = 0.44, resp.; P = .002, P = .035, and P = .008, resp.), which was not observed in medium or high Hcy level groups. In conclusion, the T allele of rs1801133 and the G allele of rs751141 may be risk factors of ischemic stroke in the Chinese T2DM population.
Collapse
Affiliation(s)
- Liang Ma
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Clinical Laboratory, China-Japan Friendship Hospital, Beijing, China
- Beijing Key Lab Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Science, China-Japan Friendship Hospital, Beijing, China
| | - Yongwei Jiang
- Clinical Laboratory, China-Japan Friendship Hospital, Beijing, China
| | - Xiaomu Kong
- Department of Endocrinology, China-Japan Friendship Hospital, Beijing, China
| | - Meihua Yan
- Beijing Key Lab Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Science, China-Japan Friendship Hospital, Beijing, China
| | - Tingting Zhao
- Beijing Key Lab Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Science, China-Japan Friendship Hospital, Beijing, China
| | - Hailing Zhao
- Beijing Key Lab Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Science, China-Japan Friendship Hospital, Beijing, China
| | - Qian Liu
- Clinical Laboratory, China-Japan Friendship Hospital, Beijing, China
| | - Haojun Zhang
- Beijing Key Lab Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Science, China-Japan Friendship Hospital, Beijing, China
| | - Yongtong Cao
- Clinical Laboratory, China-Japan Friendship Hospital, Beijing, China
- *Yongtong Cao: and
| | - Ping Li
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Key Lab Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Science, China-Japan Friendship Hospital, Beijing, China
- *Ping Li:
| |
Collapse
|
44
|
Epoxygenated Fatty Acids Inhibit Retinal Vascular Inflammation. Sci Rep 2016; 6:39211. [PMID: 27966642 PMCID: PMC5155241 DOI: 10.1038/srep39211] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 11/21/2016] [Indexed: 01/28/2023] Open
Abstract
The objective of the present study was to assess the effect of elevating epoxygenated fatty acids on retinal vascular inflammation. To stimulate inflammation we utilized TNFα, a potent pro-inflammatory mediator that is elevated in the serum and vitreous of diabetic patients. In TNFα-stimulated primary human retinal microvascular endothelial cells, total levels of epoxyeicosatrienoic acids (EETs), but not epoxydocosapentaenoic acids (EDPs), were significantly decreased. Exogenous addition of 11,12-EET or 19,20-EDP when combined with 12-(3-adamantane-1-yl-ureido)-dodecanoic acid (AUDA), an inhibitor of epoxide hydrolysis, inhibited VCAM-1 and ICAM-1 expression and protein levels; conversely the diol product of 19,20-EDP hydrolysis, 19,20-DHDP, induced VCAM1 and ICAM1 expression. 11,12-EET and 19,20-EDP also inhibited leukocyte adherence to human retinal microvascular endothelial cell monolayers and leukostasis in an acute mouse model of retinal inflammation. Our results indicate that this inhibition may be mediated through an indirect effect on NFκB activation. This is the first study demonstrating a direct comparison of EET and EDP on vascular inflammatory endpoints, and we have confirmed a comparable efficacy from each isomer, suggesting a similar mechanism of action. Taken together, these data establish that epoxygenated fatty acid elevation will inhibit early pathology related to TNFα-induced inflammation in retinal vascular diseases.
Collapse
|
45
|
Zhang X, Liu X, Luo J, Xiao W, Ye X, Chen M, Li Y, Zhang GJ. Notch3 inhibits epithelial-mesenchymal transition by activating Kibra-mediated Hippo/YAP signaling in breast cancer epithelial cells. Oncogenesis 2016; 5:e269. [PMID: 27841855 PMCID: PMC5141289 DOI: 10.1038/oncsis.2016.67] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Revised: 09/06/2016] [Accepted: 09/19/2016] [Indexed: 02/05/2023] Open
Abstract
Invasion, metastasis and chemoresistance are leading causes of death in breast cancer patients. A vital change of epithelial cells, epithelial-mesenchymal transition (EMT), is involved in these processes. Unfortunately, the molecular mechanisms controlling EMT remain to be elucidated. Our previous studies have shown that ectopic N3ICD expression inhibits EMT in MDA-MB-231, a triple-negative breast cancer (TNBC) epithelial cell line. To decipher the mechanism, we performed in-depth studies. Specifically, we found that overexpressing N3ICD transcriptionally upregulated the expression of Kibra, an upstream member of the Hippo pathway. Correspondingly, we also observed that phosphorylated Hippo pathway core kinases, including Lats1/2 and MST1/2, were increased and decreased by overexpressing and knocking down Notch3, respectively. Furthermore, we found that the oncogenic transcriptional coactivator yes-associated protein (YAP), which is negatively regulated by the Hippo pathway, was inhibited by overexpressing N3ICD in breast cancer epithelial cells. The ability of Kibra to inhibit EMT has been previously reported. We thus speculated that Notch3 inhibition of EMT is mediated by upregulated Kibra. To verify this hypothesis, a rescue experiment was performed. Evidently, the ability of Notch3 to inhibit EMT can be countered by knocking down Kibra expression. These data suggest that Notch3 inhibits EMT by activating the Hippo/YAP pathway by upregulating Kibra in breast cancer epithelial cells, and Kibra may be a downstream effector of Notch3. These findings deepen our understanding of EMT in both development and disease, and will undoubtedly help to provide new therapeutic strategies for interfering with cancer invasion and metastasis, especially for TNBC.
Collapse
Affiliation(s)
- X Zhang
- Changjiang Scholar's Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - X Liu
- Changjiang Scholar's Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - J Luo
- Changjiang Scholar's Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - W Xiao
- Changjiang Scholar's Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - X Ye
- Changjiang Scholar's Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - M Chen
- Changjiang Scholar's Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Y Li
- Changjiang Scholar's Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, China
- Changjiang Scholar's Laboratory, Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou 515041, China. E-mail: or
| | - G-J Zhang
- Changjiang Scholar's Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, China
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, China
- Changjiang Scholar's Laboratory, Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou 515041, China. E-mail: or
| |
Collapse
|
46
|
Zhang K, Liu Y, Liu X, Chen J, Cai Q, Wang J, Huang H. Apocynin improving cardiac remodeling in chronic renal failure disease is associated with up-regulation of epoxyeicosatrienoic acids. Oncotarget 2016; 6:24699-708. [PMID: 26322503 PMCID: PMC4694789 DOI: 10.18632/oncotarget.5084] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 08/07/2015] [Indexed: 12/28/2022] Open
Abstract
Cardiac remodeling is one of the most common cardiac abnormalities and associated with a high mortality in chronic renal failure (CRF) patients. Apocynin, a nicotinamide-adenine dinucleotide phosphate (NADPH) oxidase inhibitor, has been showed cardio-protective effects. However, whether apocynin can improve cardiac remodeling in CRF and what is the underlying mechanism are unclear. In the present study, we enrolled 94 participants. In addition, we used 5/6 nephrectomized rats to mimic cardiac remodeling in CRF. Serum levels of epoxyeicosatrienoic acids (EETs) and its mainly metabolic enzyme-soluble epoxide hydrolase (sEH) were measured. The results showed that the serum levels of EETs were significantly decreased in renocardiac syndrome participants (P < 0.05). In 5/6 nephrectomized CRF model, the ratio of left ventricular weight / body weight, left ventricular posterior wall thickness, and cardiac interstitial fibrosis were significantly increased while ejection fraction significantly decreased (P < 0.05). All these effects could partly be reversed by apocynin. Meanwhile, we found during the process of cardiac remodeling in CRF, apocynin significantly increased the reduced serum levels of EETs and decreased the mRNA and protein expressions of sEH in the heart (P < 0.05). Our findings indicated that the protective effect of apocynin on cardiac remodeling in CRF was associated with the up-regulation of EETs. EETs may be a new mediator for the injury of kidney-heart interactions.
Collapse
Affiliation(s)
- Kun Zhang
- Department of Cardiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.,Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou 510120, China
| | - Yu Liu
- Department of Cardiology, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530000, China
| | - Xiaoqiang Liu
- Department of Clinical Laboratory, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Jie Chen
- Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou 510120, China.,Department of Radiation Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Qingqing Cai
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Sun Yat-Sen University, Guangzhou 510120, China
| | - Jingfeng Wang
- Department of Cardiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.,Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou 510120, China
| | - Hui Huang
- Department of Cardiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.,Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou 510120, China
| |
Collapse
|
47
|
Yao L, Wang C, Zhang X, Peng L, Liu W, Zhang X, Liu Y, He J, Jiang C, Ai D, Zhu Y. Hyperhomocysteinemia activates the aryl hydrocarbon receptor/CD36 pathway to promote hepatic steatosis in mice. Hepatology 2016; 64:92-105. [PMID: 26928949 DOI: 10.1002/hep.28518] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 02/20/2016] [Indexed: 01/19/2023]
Abstract
UNLABELLED Hyperhomocysteinemia (HHcy) is associated with liver diseases such as fatty liver and hepatic fibrosis; however, the underlying mechanism is still largely unknown. The current study aimed to explore the signaling pathway involved in HHcy-induced hepatic steatosis (HS). C57BL/6 mice were fed a high-methionine diet (HMD) for 4 and 8 weeks to establish the HHcy mouse model. Compared to a chow diet, the HMD induced hepatic steatosis and elevated hepatic expression of CD36, a fatty acid transport protein. The increased CD36 expression was associated with activation of the aryl hydrocarbon receptor (AHR). In primary cultured hepatocytes, high levels of homocysteine (Hcy) treatment up-regulated CD36 and increased subsequent lipid uptake; both were significantly attenuated by small interfering RNA (siRNA) knockdown of CD36 and AHR. Chromatin immunoprecipitation assay revealed that Hcy promoted binding of AHR to the CD36 promoter, and transient transfection assay demonstrated markedly increased activity of the AHR response element by Hcy, which was ligand dependent. Mass spectrometry revealed significantly increased hepatic content of lipoxin A4 (LXA4 ), a metabolite of arachidonic acid, in HMD-fed mice. Furthermore, overexpression of 15-oxoprostaglandin 13-reductase 1, a LXA4 inactivation enzyme, inhibited Hcy-induced AHR activation, lipid uptake, and lipid accumulation. Moreover, LXA4 -induced up-regulation of CD36 and lipid uptake was inhibited by AHR siRNA in vitro in hepatocytes. Finally, treatment with an AHR antagonist reversed HHcy-induced lipid accumulation by inhibiting the AHR-CD36 pathway in mice. CONCLUSION HHcy activates the AHR-CD36 pathway by increasing hepatic LXA4 content, which results in hepatic steatosis. (Hepatology 2016;64:92-105).
Collapse
Affiliation(s)
- Liu Yao
- Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Chunjiong Wang
- Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Xu Zhang
- Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Liyuan Peng
- Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Wenli Liu
- Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Xuejiao Zhang
- Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Yajin Liu
- Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Jinlong He
- Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China
| | - Ding Ai
- Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Yi Zhu
- Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| |
Collapse
|
48
|
He J, Wang C, Zhu Y, Ai D. Soluble epoxide hydrolase: A potential target for metabolic diseases. J Diabetes 2016; 8:305-13. [PMID: 26621325 DOI: 10.1111/1753-0407.12358] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 11/16/2015] [Accepted: 11/22/2015] [Indexed: 12/12/2022] Open
Abstract
Epoxyeicosatrienoic acids (EETs), important lipid mediators derived from arachidonic acid, have many beneficial effects in metabolic diseases, including atherosclerosis, hypertension, cardiac hypertrophy, diabetes, non-alcoholic fatty liver disease, and kidney disease. Epoxyeicosatrienoic acids can be further hydrolyzed to less active diols by the enzyme soluble epoxide hydrolase (sEH). Increasing evidence suggests that inhibition of sEH increases levels of EETs, which have anti-inflammatory effects and can prevent the development of hypertension, atherosclerosis, heart failure, fatty liver, and multiple organ fibrosis. Arachidonic acid is the most abundant omega-6 polyunsaturated fatty acid (PUFA) and shares the same set of enzymes with omega-3 PUFAs, such as docosahexaenoic acid and eicosapentaenoic acid. The omega-3 PUFAs and metabolites, such as regioisomeric epoxyeicosatetraenoic acids and epoxydocosapentaenoic acids, have been reported to have strong vasodilatory and anti-inflammatory effects. Therefore, sEH may be a potential therapeutic target for metabolic disorders. In this review, we focus on our and other recent studies of the functions of sEH, including the effects of its eicosanoid products from both omega-3 and omega-6 PUFAs, in various metabolic diseases. We also discuss the possible cellular and molecular mechanisms underlying the regulation of sEH.
Collapse
Affiliation(s)
- Jinlong He
- Collaborative Innovation Center of Tianjin for Medical Epigenetics, Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Chunjiong Wang
- Collaborative Innovation Center of Tianjin for Medical Epigenetics, Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Yi Zhu
- Collaborative Innovation Center of Tianjin for Medical Epigenetics, Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Ding Ai
- Collaborative Innovation Center of Tianjin for Medical Epigenetics, Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| |
Collapse
|
49
|
Wang Q, Huo L, He J, Ding W, Su H, Tian D, Welch C, Hammock BD, Ai D, Zhu Y. Soluble epoxide hydrolase is involved in the development of atherosclerosis and arterial neointima formation by regulating smooth muscle cell migration. Am J Physiol Heart Circ Physiol 2015; 309:H1894-903. [PMID: 26453326 DOI: 10.1152/ajpheart.00289.2015] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 08/18/2015] [Indexed: 02/05/2023]
Abstract
Epoxyeicosatrienoic acids (EETs) have beneficial effects on cardiovascular disease. Soluble epoxide hydrolase (sEH) metabolizes EETs to less active diols, thus diminishing their biological activity. sEH inhibitors can suppress the progression of atherosclerotic lesions in animal models. However, the regulation of sEH in vascular smooth muscle cells (VSMCs) and role of sEH in patients with atherosclerosis have not been evaluated. We hypothesize that sEH in VSMCs plays a pivotal role in atherosclerosis and injury-induced neointima formation. In this study, sEH expression in human autopsy atherosclerotic plaque was determined by immunohistochemistry. In cultured rat and human VSMCs, the phenotypic switching marker and sEH expression induced by platelet-derived growth factor-BB (PDGF-BB) were examined by Western blot analysis. Carotid-artery balloon injury was performed after adenovirus-mediated overexpression of sEH or oral administration of a potent sEH inhibitor in Sprague-Dawley rats. sEH was highly expressed in VSMCs of the intima and media within human atherosclerotic plaque. In vitro, PDGF-BB upregulated the expression in VSMCs after transcription and promoted cell proliferation and migration; the latter effect could be largely attenuated by an sEH inhibitor. Adenovirus-mediated overexpression of sEH could mimic the effect of PDGF-BB and induce VSMC proliferation and migration. In vivo, the sEH inhibitor led to a significant decrease in injury-induced neointima formation in a rat carotid-artery injury model. These data establish the effect of sEH expression on atherosclerotic progression and vascular remodeling after injury, thus identifying a novel integrative role for sEH in VSMC phenotypic modulation and migration. Blocking sEH activity may be a potential therapeutic approach for ameliorating vascular occlusive disease.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Animals
- Atherosclerosis/enzymology
- Atherosclerosis/genetics
- Atherosclerosis/pathology
- Atherosclerosis/prevention & control
- Becaplermin
- Carotid Artery Injuries/enzymology
- Carotid Artery Injuries/genetics
- Carotid Artery Injuries/pathology
- Carotid Artery Injuries/therapy
- Cell Dedifferentiation
- Cell Movement/drug effects
- Cell Proliferation
- Cells, Cultured
- Coronary Artery Disease/enzymology
- Coronary Artery Disease/genetics
- Coronary Artery Disease/pathology
- Disease Models, Animal
- Disease Progression
- Eicosanoids/metabolism
- Enzyme Inhibitors/pharmacology
- Epoxide Hydrolases/antagonists & inhibitors
- Epoxide Hydrolases/genetics
- Epoxide Hydrolases/metabolism
- Female
- Humans
- Male
- Middle Aged
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/pathology
- Neointima
- Phenotype
- Phenylurea Compounds/pharmacology
- Piperidines/pharmacology
- Proto-Oncogene Proteins c-sis/pharmacology
- Rats, Sprague-Dawley
- Signal Transduction
- Time Factors
- Transfection
- Vascular Remodeling
- Young Adult
Collapse
Affiliation(s)
- Qingjie Wang
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong, China; Department of Physiology and Pathophysiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Leijun Huo
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Jinlong He
- Department of Physiology and Pathophysiology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Medical University, Tianjin, China
| | - Wenshuang Ding
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Hang Su
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Dongping Tian
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Carrie Welch
- Division of Molecular Medicine, Department of Medicine, Columbia University, New York, New York; and
| | - Bruce D Hammock
- Department of Entomology and Comprehensive Cancer Center, University of California, Davis, California
| | - Ding Ai
- Department of Physiology and Pathophysiology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Medical University, Tianjin, China
| | - Yi Zhu
- Department of Physiology and Pathophysiology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Medical University, Tianjin, China;
| |
Collapse
|
50
|
Xue SS, He JL, Zhang X, Liu YJ, Xue FX, Wang CJ, Ai D, Zhu Y. Metabolomic analysis revealed the role of DNA methylation in the balance of arachidonic acid metabolism and endothelial activation. Biochim Biophys Acta Mol Cell Biol Lipids 2015; 1851:1317-26. [DOI: 10.1016/j.bbalip.2015.07.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 06/19/2015] [Accepted: 07/06/2015] [Indexed: 01/26/2023]
|