1
|
Li X, Wang Y, Liu C, Fu G, Li J, Zhang J. Beraprost sodium attenuates the development of myocardial fibrosis after myocardial infarction by regulating GSK-3β expression in rats. Immun Inflamm Dis 2023; 11:e1050. [PMID: 38018586 PMCID: PMC10633815 DOI: 10.1002/iid3.1050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 09/21/2023] [Accepted: 10/05/2023] [Indexed: 11/30/2023] Open
Abstract
OBJECTIVE The aim of this study was to elucidate the mechanism of beraprost sodium (BPS) in the intervention of myocardial fibrosis after myocardial infarction (MI) through glycogen synthase kinase-3β (GSK-3β) and to provide new ideas for intervention in myocardial fibrosis. MATERIALS AND METHODS MI model rats given BPS and cardiac fibroblasts (CFs) treated with BPS and TGF-β. HE staining and Masson staining were used to detect the pathological changes of myocardial tissue. Fibrotic markers were detected by immunohistochemical staining. The expressions of GSK-3β, cAMP response element binding protein (CREB), and p-CREB were analyzed by qPCR and western blot analysis. EDU staining was used to detect the proliferation of CFs. The promoter activity of GSK-3β was detected by luciferase assay. Chromatin immunoprecipitation assay was used to detect the binding levels of GSK-3β promoter and Y-box binding protein 1 (YBX1). The levels of intracellular cyclic adenosine monophosphate (cAMP) were analyzed by enzyme-linked immunosorbent assay (ELISA). RESULTS After operation, BPS improved myocardial fibrosis and upregulated GSK-3β protein expression in male SD rats. BPS can down-regulate α-smooth muscle actin (α-SMA) level and up-regulate GSK-3β protein expression in CFs after TGF-β stimulation. Furthermore, GSK-3β knockdown can reverse the effect of BPS on TGF-β-activated CFs, enhance α-SMA expression, and promote the proliferation of CFs. BPS could regulate GSK-3β expression by promoting the binding of GSK-3β promoter to YBX1. BPS induced upregulation of p-CREB and cAMP, resulting in reduced fibrosis, which was reversed by the knockdown of GSK-3β or prostaglandin receptor (IPR) antagonists. CONCLUSION BPS treatment increased the binding of YBX1 to the GSK-3β promoter, and GSK-3β protein expression was upregulated, which further caused the upregulation of p-CREB and cAMP, and finally inhibited myocardial fibrosis.
Collapse
Affiliation(s)
- Xing‐Xing Li
- Department of Extracorporeal Life Support CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yun‐Zhe Wang
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Chuang Liu
- Department of Extracorporeal Life Support CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Guo‐Wei Fu
- Department of Extracorporeal Life Support CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Jun Li
- Department of Extracorporeal Life Support CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Jin‐Ying Zhang
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Henan Province′s Key Laboratory of Cardiac Injury and RepairZhengzhouChina
- Henan Province Clinical Research Center for Cardiovascular DiseasesZhengzhouChina
| |
Collapse
|
2
|
Cheng C, Zhang J, Li X, Xue F, Cao L, Meng L, Sui W, Zhang M, Zhao Y, Xi B, Yu X, Xu F, Yang J, Zhang Y, Zhang C. NPRC deletion mitigated atherosclerosis by inhibiting oxidative stress, inflammation and apoptosis in ApoE knockout mice. Signal Transduct Target Ther 2023; 8:290. [PMID: 37553374 PMCID: PMC10409771 DOI: 10.1038/s41392-023-01560-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 05/24/2023] [Accepted: 07/07/2023] [Indexed: 08/10/2023] Open
Abstract
Previous studies suggested a beneficial effect of natriuretic peptides in animal models of cardiovascular disease, but the role of natriuretic peptide receptor C (NPRC) in the pathogenesis of atherosclerosis (AS) remains unknown. This study was designed to test the hypothesis that NPRC may promote AS lesion formation and instability by enhancing oxidative stress, inflammation, and apoptosis via protein kinase A (PKA) signaling. ApoE-/- mice were fed chow or Western diet for 12 weeks and NPRC expression was significantly increased in the aortic tissues of Western diet-fed mice. Systemic NPRC knockout mice were crossed with ApoE-/- mice to generate ApoE-/-NPRC-/- mice, and NPRC deletion resulted in a significant decrease in the size and instability of aortic atherosclerotic lesions in ApoE-/-NPRC-/- versus ApoE-/- mice. In addition, endothelial cell-specific NPRC knockout attenuated atherosclerotic lesions in mice. In contrast, endothelial cell overexpression of NPRC aggravated the size and instability of atherosclerotic aortic lesions in mice. Experiments in vitro showed that NPRC knockdown in human aortic endothelial cells (HAECs) inhibited ROS production, pro-inflammatory cytokine expression and endothelial cell apoptosis, and increased eNOS expression. Furthermore, NPRC knockdown in HAECs suppressed macrophage migration, cytokine expression, and phagocytosis via its effects on endothelial cells. On the contrary, NPRC overexpression in endothelial cells resulted in opposite effects. Mechanistically, the anti-inflammation and anti-atherosclerosis effects of NPRC deletion involved activation of cAMP/PKA pathway, leading to downstream upregulated AKT1 pathway and downregulated NF-κB pathway. In conclusion, NPRC deletion reduced the size and instability of atherosclerotic lesions in ApoE-/- mice via attenuating inflammation and endothelial cell apoptosis and increasing eNOS expression by modulating cAMP/PKA-AKT1 and NF-κB pathways. Thus, targeting NPRC may provide a promising approach to the prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Cheng Cheng
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, 110004, China
| | - Jie Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaodong Li
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, 110004, China
| | - Fei Xue
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Lei Cao
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Linlin Meng
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Wenhai Sui
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Meng Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yuxia Zhao
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
- Department of Traditional Chinese Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Bo Xi
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiao Yu
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Feng Xu
- Department of Emergency Medicine, Chest Pain Center, Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Qilu Hospital, Shandong University, Jinan, China
| | - Jianmin Yang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.
| | - Yun Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.
- Cardiovascular Disease Research Center of Shandong First Medical University, Central Hospital Affiliated to Shandong First Medical University, Jinan, China.
| | - Cheng Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.
- Cardiovascular Disease Research Center of Shandong First Medical University, Central Hospital Affiliated to Shandong First Medical University, Jinan, China.
| |
Collapse
|
3
|
Mukherjee S, Sarkar AK, Lahiri A, Sengupta Bandyopadhyay S. Analysis of the interaction of a non-canonical twin half-site of Cyclic AMP-Response Element (CRE) with CRE-binding protein. Biochimie 2023; 211:25-34. [PMID: 36842626 DOI: 10.1016/j.biochi.2023.02.011] [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: 08/15/2022] [Revised: 12/23/2022] [Accepted: 02/17/2023] [Indexed: 02/26/2023]
Abstract
Differential regulation of a gene having either canonical or non-canonical cyclic AMP response element (CRE) in its promoter is primarily accomplished by its interactions with CREB (cAMP-response element binding protein). The present study aims to delineate the mechanism of the CREB-CRE interactions at the Oncostatin-M (osm) promoter by in vitro and in silico approaches. The non-canonical CREosm consists of two half-CREs separated by a short intervening sequence of 9 base pairs. In this study, in vitro binding assays revealed that out of the two CRE half-sites, the right half-CRE was indispensable for binding of CREB, while the left sequence showed weaker binding ability and specificity. Genome-wide modeling and high throughput free energy calculations for the energy-minimized models containing CREB-CREosm revealed that there was no difference in the binding of CREB to the right half of CREosm site when compared to the entire CREosm. These results were in accordance with the in vitro studies, confirming the indispensable role of the right half-CREosm site in stable complex formation with the CREB protein. Additionally, conversion of the right half-CREosm site to a canonical CRE palindrome showed stronger CREB binding, irrespective of the presence or absence of the left CRE sequence. Thus, the present study establishes an interesting insight into the interaction of CREB with a CRE variant located at the far end of a TATA-less promoter of a cytokine-encoding gene, which in turn could be involved in the regulation of transcription under specific conditions.
Collapse
Affiliation(s)
- Srimoyee Mukherjee
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, 92 A.P.C. Road, Kolkata, 700009, India
| | - Aditya Kumar Sarkar
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, 92 A.P.C. Road, Kolkata, 700009, India
| | - Ansuman Lahiri
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, 92 A.P.C. Road, Kolkata, 700009, India
| | - Sumita Sengupta Bandyopadhyay
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, 92 A.P.C. Road, Kolkata, 700009, India.
| |
Collapse
|
4
|
Montanaro R, Vellecco V, Torregrossa R, Casillo GM, Manzo OL, Mitidieri E, Bucci M, Castaldo S, Sorrentino R, Whiteman M, Smimmo M, Carriero F, Terrazzano G, Cirino G, d'Emmanuele di Villa Bianca R, Brancaleone V. Hydrogen sulfide donor AP123 restores endothelial nitric oxide-dependent vascular function in hyperglycemia via a CREB-dependent pathway. Redox Biol 2023; 62:102657. [PMID: 36913800 PMCID: PMC10025109 DOI: 10.1016/j.redox.2023.102657] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 03/03/2023] [Indexed: 03/06/2023] Open
Abstract
Diabetes is associated with severe vascular complications involving the impairment of endothelial nitric oxide synthase (eNOS) as well as cystathionine γ-lyase (CSE) activity. eNOS function is suppressed in hyperglycaemic conditions, resulting in reduced NO bioavailability, which is paralleled by reduced levels of hydrogen sulfide (H2S). Here we have addressed the molecular basis of the interplay between the eNOS and CSE pathways. We tested the impact of H2S replacement by using the mitochondrial-targeted H2S donor AP123 in isolated vessels and cultured endothelial cells in high glucose (HG) environment, at concentrations not causing any vasoactive effect per se. Aorta exposed to HG displayed a marked reduction of acetylcholine (Ach)-induced vasorelaxation that was restored by the addition of AP123 (10 nM). In HG condition, bovine aortic endothelial cells (BAEC) showed reduced NO levels, downregulation of eNOS expression, and suppression of CREB activation (p-CREB). Similar results were obtained by treating BAEC with propargylglycine (PAG), an inhibitor of CSE. AP123 treatment rescued eNOS expression, as well as NO levels, and restored p-CREB expression in both the HG environment and the presence of PAG. This effect was mediated by a PI3K-dependent activity since wortmannin (PI3K inhibitor) blunted the rescuing effects operated by the H2S donor. Experiments performed in the aorta of CSE-/- mice confirmed that reduced levels of H2S not only negatively affect the CREB pathway but also impair Ach-induced vasodilation, significantly ameliorated by AP123. We have demonstrated that the endothelial dysfunction due to HG involves H2S/PI3K/CREB/eNOS route, thus highlighting a novel aspect of the H2S/NO interplay in the vasoactive response.
Collapse
Affiliation(s)
| | - Valentina Vellecco
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | | | - Gian Marco Casillo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Onorina Laura Manzo
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, USA
| | - Emma Mitidieri
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Mariarosaria Bucci
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy.
| | - Sigismondo Castaldo
- U.O.C.Ricerca Formazione & Cooperazione Internazionale, A.O.R.N." Antonio Cardarelli", Naples, Italy
| | - Raffaella Sorrentino
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine and Surgery, University of Naples Federico II, 80131, Naples, Italy
| | | | - Martina Smimmo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Flavia Carriero
- Department of Science, University of Basilicata, Potenza, Italy
| | | | - Giuseppe Cirino
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | | | | |
Collapse
|
5
|
Fontes MT, Arruda-Junior DF, dos Santos DS, Ribeiro-Silva JC, Antônio EL, Tucci PF, Rossoni LV, Girardi AC. Dipeptidyl peptidase 4 inhibition rescues PKA-eNOS signaling and suppresses aortic hypercontractility in male rats with heart failure. Life Sci 2023; 323:121648. [PMID: 37001807 DOI: 10.1016/j.lfs.2023.121648] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 03/11/2023] [Accepted: 03/28/2023] [Indexed: 03/31/2023]
Abstract
AIMS Vascular dysfunction and elevated circulating dipeptidyl peptidase 4 (DPP4) activity are both reported to be involved in the progression of heart failure (HF). While the cardiac benefits of DPP4 inhibitors (DPP4i) have been extensively studied, little is known about the effects of DPP4i on vascular dysfunction in nondiabetic HF. This study tested the hypothesis that vildagliptin (DPP4i) mitigates aortic hyperreactivity in male HF rats. MATERIALS AND METHODS Male Wistar rats were subjected to left ventricle (LV) radiofrequency ablation to HF induction or sham operation (SO). Six weeks after surgery, radiofrequency-ablated rats who developed HF were treated with vildagliptin (120 mg⸱kg-1⸱day-1) or vehicle for 4 weeks. Thoracic aorta reactivity, dihydroethidium fluorescence, immunoblotting experiments, and enzyme-linked immunosorbent assays were performed. KEY FINDINGS DPP4i ameliorated the hypercontractility of HF aortas to the α-adrenoceptor agonist phenylephrine towards SO levels. In HF, the reduced endothelium and nitric oxide (NO) anticontractile effect on phenylephrine response was restored by DPP4i. At the molecular level, this vasoprotective effect of DPP4i was accompanied by (i) reduced oxidative stress and NADPH oxidase 2 (Nox2) expression, (ii) enhanced total endothelial nitric oxide synthase (eNOS) expression and phosphorylation at Ser1177, and (iii) increased PKA activation, which acts upstream of eNOS. Additionally, DPP4i restored the higher serum angiotensin II concentration towards SO. SIGNIFICANCE Our data demonstrate that DPP4i ameliorates aortic hypercontractility, most likely by enhancing NO bioavailability, showing that the DPP4i-induced cardioprotection in male HF may arise from effects not only in the heart but also in conductance arteries.
Collapse
|
6
|
Mutation in FBXO32 causes dilated cardiomyopathy through up-regulation of ER-stress mediated apoptosis. Commun Biol 2021; 4:884. [PMID: 34272480 PMCID: PMC8285540 DOI: 10.1038/s42003-021-02391-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 06/23/2021] [Indexed: 02/06/2023] Open
Abstract
Endoplasmic reticulum (ER) stress induction of cell death is implicated in cardiovascular diseases. Sustained activation of ER-stress induces the unfolded protein response (UPR) pathways, which in turn activate three major effector proteins. We previously reported a missense homozygous mutation in FBXO32 (MAFbx, Atrogin-1) causing advanced heart failure by impairing autophagy. In the present study, we performed transcriptional profiling and biochemical assays, which unexpectedly revealed a reduced activation of UPR effectors in patient mutant hearts, while a strong up-regulation of the CHOP transcription factor and of its target genes are observed. Expression of mutant FBXO32 in cells is sufficient to induce CHOP-associated apoptosis, to increase the ATF2 transcription factor and to impair ATF2 ubiquitination. ATF2 protein interacts with FBXO32 in the human heart and its expression is especially high in FBXO32 mutant hearts. These findings provide a new underlying mechanism for FBXO32-mediated cardiomyopathy, implicating abnormal activation of CHOP. These results suggest alternative non-canonical pathways of CHOP activation that could be considered to develop new therapeutic targets for the treatment of FBXO32-associated DCM. Al-Yacoub et al. investigate the consequences of FBXO32 mutation on dilated cardiomyopathy. ER stress, abnormal CHOP activation and CHOP-induced apoptosis with no UPR effector activation are found to underlie the FBXO32 mutation induced cardiomyopathy, suggesting an alternative pathway that can be considered to develop new therapeutic targets for its treatment.
Collapse
|
7
|
Wei C, Meng L, Zhang Y. miR-450a-5p Eliminates MGO-Induced Insulin Resistance via Targeting CREB. Int J Stem Cells 2020; 13:46-54. [PMID: 32114742 PMCID: PMC7119216 DOI: 10.15283/ijsc19088] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 11/21/2019] [Accepted: 12/30/2019] [Indexed: 02/06/2023] Open
Abstract
Background and Objectives miR-450a-5p was involved in fat formation, however, its role in insulin resistance remains unclear. This study investigated the effects of miR-450a-5p on endothelial cells, with the aim of finding a potential target for diabetes mellitus. Methods and Results Human umbilical vein endothelial cells (HUVECs) were treated with low-glucose, high-glucose, methylglyoxal (MGO), and insulin alone or in combination with MGO. The expression of miR-450a-5p in treated cells was measured by quantitative real-time polymerase chain reaction (qRT-PCR) assays. The cell activity, migration and fat formation were determined by MTT experiments, Transwell assay and oil red O staining. The expressions of eNOS/AKT pathway-related proteins in cells were assessed by Western blot (WB) analysis. Furthermore, the target gene of miR-450a-5p was analyzed by double-luciferase reporter analysis, and its effects on eNOS/AKT pathway were estimated. We found that the expression of miR-450a-5p was decreased obviously in endothelial cells treated with high-glucose and MGO. In vitro cell experiments showed that MGO could not only promote the activity of endothelial cells, but also accelerate cell migration and fat accumulation, which, however, could be reversed by up-regulation of miR-450a-5p. Moreover, MGO inhibited eNOS/AKT pathway activation and NO release mediated by insulin, and such effects were reversed by up-regulation of miR-450a-5p. Furthermore, CREB was the target gene for miR-450a-5p, had an activation effect on the eNOS/AKT pathway. Conclusions Up-regulated miR-450a-5p eliminates MGO-induced insulin resistance via targeting CREB, and therefore could be used as a potential target to improve insulin resistance and treat patients with diabetes-related diseases.
Collapse
Affiliation(s)
- Cuifeng Wei
- Department of Endocrinology, Jingmen No. 1 People's Hospital, Jingmen, China
| | - Li Meng
- Department of Endocrinology, Jingmen No. 1 People's Hospital, Jingmen, China
| | - Yuting Zhang
- Department of Endocrinology, Jingmen No. 1 People's Hospital, Jingmen, China
| |
Collapse
|
8
|
Vigorelli V, Rurali E, Carugo S, Pompilio G, Vinci MC. Sensitive and quantitative method to evaluate DNA methylation of the positive regulatory domains (PRDI, PRDII) and cAMP response element (CRE) in human endothelial nitric oxide synthase promoter. Nitric Oxide 2019; 92:41-48. [PMID: 31421231 DOI: 10.1016/j.niox.2019.08.005] [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: 05/20/2019] [Revised: 08/07/2019] [Accepted: 08/08/2019] [Indexed: 11/28/2022]
Abstract
Nitric oxide plays a prominent role in the cardiovascular system and much attention has been devoted in the last years on deciphering the regulation of human endothelial nitric oxide synthase (eNOS) expression. Epigenetic-based mechanisms have a key role in the eNOS expression and their pathologic perturbations may have profound effects on the steady state RNA levels in the endothelium. The human eNOS promoter lacks a canonical TATA box and it does not contain a proximal CpG island. A differentially DNA methylated region (DMR) in the native eNOS proximal promoter is involved in gene expression regulation. Here we describe a quantitative, sensitive and cost-effective method that, relying on a novel normalization strategy, allows the quantification of DNA methylation status of the positive regulatory domains (PRDI, PRDII) and cAMP response element (CRE) in human eNOS promoter. This technique will enable to explore the functional relevance of DNA methylation perturbations of eNOS promoter both under pathological and physiological conditions.
Collapse
Affiliation(s)
- V Vigorelli
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino - IRCCS, Milan, Italy
| | - E Rurali
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino - IRCCS, Milan, Italy
| | - S Carugo
- Cardiology Unit, ASST Santi Paolo e Carlo and Department of Health Sciences, University of Milan, Milan, Italy
| | - G Pompilio
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino - IRCCS, Milan, Italy; Dipartimento di Scienze Cliniche e di Comunità, Università degli Studi di Milano, Milan, Italy
| | - M C Vinci
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino - IRCCS, Milan, Italy.
| |
Collapse
|
9
|
Strassheim D, Karoor V, Stenmark K, Verin A, Gerasimovskaya E. A current view of G protein-coupled receptor - mediated signaling in pulmonary hypertension: finding opportunities for therapeutic intervention. ACTA ACUST UNITED AC 2018; 2. [PMID: 31380505 PMCID: PMC6677404 DOI: 10.20517/2574-1209.2018.44] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Pathological vascular remodeling is observed in various cardiovascular diseases including pulmonary hypertension (PH), a disease of unknown etiology that has been characterized by pulmonary artery vasoconstriction, right ventricular hypertrophy, vascular inflammation, and abnormal angiogenesis in pulmonary circulation. G protein-coupled receptors (GPCRs) are the largest family in the genome and widely expressed in cardiovascular system. They regulate all aspects of PH pathophysiology and represent therapeutic targets. We overview GPCRs function in vasoconstriction, vasodilation, vascular inflammation-driven remodeling and describe signaling cross talk between GPCR, inflammatory cytokines, and growth factors. Overall, the goal of this review is to emphasize the importance of GPCRs as critical signal transducers and targets for drug development in PH.
Collapse
Affiliation(s)
- Derek Strassheim
- Departments of Medicine, University of Colorado Denver, Aurora, CO 80045, USA
| | - Vijaya Karoor
- Departments of Medicine, University of Colorado Denver, Aurora, CO 80045, USA.,Cardiovascular and Pulmonary Research laboratories, University of Colorado Denver, Aurora, CO 80045, USA
| | - Kurt Stenmark
- Cardiovascular and Pulmonary Research laboratories, University of Colorado Denver, Aurora, CO 80045, USA.,Department of Pediatrics, Pulmonary and Critical Care Medicine, University of Colorado Denver, Aurora, CO 80045, USA
| | - Alexander Verin
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA
| | - Evgenia Gerasimovskaya
- Cardiovascular and Pulmonary Research laboratories, University of Colorado Denver, Aurora, CO 80045, USA.,Department of Pediatrics, Pulmonary and Critical Care Medicine, University of Colorado Denver, Aurora, CO 80045, USA
| |
Collapse
|
10
|
Interleukin-1β is associated with coronary endothelial dysfunction in patients with mTOR-inhibitor-eluting stent implantation. Heart Vessels 2017; 32:823-832. [PMID: 28116487 DOI: 10.1007/s00380-017-0947-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 01/06/2017] [Indexed: 01/28/2023]
Abstract
Implantation of mammalian target of rapamycin (mTOR)-inhibitor drug-eluting stents (DESs) impairs coronary endothelial function. There are no known non-invasive biomarkers of coronary endothelial dysfunction. We aimed to assess the association between serum interleukin-1beta (IL-1β) and coronary endothelial dysfunction in patients with mTOR-inhibitor DES implantation and to investigate the association between the mTOR pathway and IL-1β. We enrolled 35 patients who had implanted DESs for coronary artery disease. At a 10-month follow-up, peripheral venous blood samples were collected to measure IL-1β levels. Coronary endothelial dysfunction was evaluated by intracoronary infusion of incremental doses of acetylcholine. Serum IL-1β levels were significantly associated with the magnitude of vasoconstriction to acetylcholine at the segment distal (P < 0.05) but not proximal to the stent. Serum IL-1β levels were positively correlated with stent length (P < 0.05). To examine the direct effects of mTOR inhibition on IL-1β release, sirolimus was incubated in cultured human umbilical vein endothelial cells (HUVECs) or coronary artery smooth muscle cells (CASMCs). Sirolimus directly increased IL-1β mRNA expression (P < 0.01) and enhanced IL-1β release into the culture media (P < 0.01) in CASMCs, but not in HUVECs. Inhibition of mTOR triggers IL-1β release through transcriptional activation in CASMCs. Serum IL-1β levels are a potential biomarker for mTOR-inhibitor DES-associated coronary endothelial dysfunction.
Collapse
|
11
|
Mateuszuk L, Jasztal A, Maslak E, Gasior-Glogowska M, Baranska M, Sitek B, Kostogrys R, Zakrzewska A, Kij A, Walczak M, Chlopicki S. Antiatherosclerotic Effects of 1-Methylnicotinamide in Apolipoprotein E/Low-Density Lipoprotein Receptor-Deficient Mice: A Comparison with Nicotinic Acid. J Pharmacol Exp Ther 2016; 356:514-24. [PMID: 26631491 PMCID: PMC6047228 DOI: 10.1124/jpet.115.228643] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 11/30/2015] [Indexed: 01/06/2023] Open
Abstract
1-Methylnicotinamide (MNA), the major endogenous metabolite of nicotinic acid (NicA), may partially contribute to the vasoprotective properties of NicA. Here we compared the antiatherosclerotic effects of MNA and NicA in apolipoprotein E (ApoE)/low-density lipoprotein receptor (LDLR)-deficient mice. ApoE/LDLR(-/-) mice were treated with MNA or NicA (100 mg/kg). Plaque size, macrophages, and cholesterol content in the brachiocephalic artery, endothelial function in the aorta, systemic inflammation, platelet activation, as well as the concentration of MNA and its metabolites in plasma and urine were measured. MNA and NicA reduced atherosclerotic plaque area, plaque inflammation, and cholesterol content in the brachiocephalic artery. The antiatherosclerotic actions of MNA and NicA were associated with improved endothelial function, as evidenced by a higher concentration of 6-keto-prostaglandin F1 α and nitrite/nitrate in the aortic ring effluent, inhibition of platelets (blunted thromboxane B2 generation), and inhibition of systemic inflammation (lower plasma concentration of serum amyloid P, haptoglobin). NicA treatment resulted in an approximately 2-fold higher concentration of MNA and its metabolites in urine and a 4-fold higher nicotinamide/MNA ratio in plasma, compared with MNA treatment. In summary; MNA displays pronounced antiatherosclerotic action in ApoE/LDLR(-/-) mice, an effect associated with an improvement in prostacyclin- and nitric oxide-dependent endothelial function, inhibition of platelet activation, inhibition of inflammatory burden in plaques, and diminished systemic inflammation. Despite substantially higher MNA availability after NicA treatment, compared with an equivalent dose of MNA, the antiatherosclerotic effect of NicA was not stronger. We suggest that detrimental effects of NicA or its metabolites other than MNA may limit beneficial effects of NicA-derived MNA.
Collapse
Affiliation(s)
- Lukasz Mateuszuk
- Jagiellonian Centre for Experimental Therapeutics (Ł.M., A.J., E.M., M.G.-G., M.B., B.S., A.Z., A.K., M.W., S.C.) and Faculty of Chemistry (M.B.), Jagiellonian University, Krakow, Poland; Department of Toxicology, Faculty of Pharmacy (A.K., M.W.) and Chair of Pharmacology (S.C.), Jagiellonian University Medical College, Krakow, Poland; and Department of Human Nutrition, Faculty of Food Technology, University of Agriculture, Krakow, Poland (R.K.)
| | - Agnieszka Jasztal
- Jagiellonian Centre for Experimental Therapeutics (Ł.M., A.J., E.M., M.G.-G., M.B., B.S., A.Z., A.K., M.W., S.C.) and Faculty of Chemistry (M.B.), Jagiellonian University, Krakow, Poland; Department of Toxicology, Faculty of Pharmacy (A.K., M.W.) and Chair of Pharmacology (S.C.), Jagiellonian University Medical College, Krakow, Poland; and Department of Human Nutrition, Faculty of Food Technology, University of Agriculture, Krakow, Poland (R.K.)
| | - Edyta Maslak
- Jagiellonian Centre for Experimental Therapeutics (Ł.M., A.J., E.M., M.G.-G., M.B., B.S., A.Z., A.K., M.W., S.C.) and Faculty of Chemistry (M.B.), Jagiellonian University, Krakow, Poland; Department of Toxicology, Faculty of Pharmacy (A.K., M.W.) and Chair of Pharmacology (S.C.), Jagiellonian University Medical College, Krakow, Poland; and Department of Human Nutrition, Faculty of Food Technology, University of Agriculture, Krakow, Poland (R.K.)
| | - Marlena Gasior-Glogowska
- Jagiellonian Centre for Experimental Therapeutics (Ł.M., A.J., E.M., M.G.-G., M.B., B.S., A.Z., A.K., M.W., S.C.) and Faculty of Chemistry (M.B.), Jagiellonian University, Krakow, Poland; Department of Toxicology, Faculty of Pharmacy (A.K., M.W.) and Chair of Pharmacology (S.C.), Jagiellonian University Medical College, Krakow, Poland; and Department of Human Nutrition, Faculty of Food Technology, University of Agriculture, Krakow, Poland (R.K.)
| | - Malgorzata Baranska
- Jagiellonian Centre for Experimental Therapeutics (Ł.M., A.J., E.M., M.G.-G., M.B., B.S., A.Z., A.K., M.W., S.C.) and Faculty of Chemistry (M.B.), Jagiellonian University, Krakow, Poland; Department of Toxicology, Faculty of Pharmacy (A.K., M.W.) and Chair of Pharmacology (S.C.), Jagiellonian University Medical College, Krakow, Poland; and Department of Human Nutrition, Faculty of Food Technology, University of Agriculture, Krakow, Poland (R.K.)
| | - Barbara Sitek
- Jagiellonian Centre for Experimental Therapeutics (Ł.M., A.J., E.M., M.G.-G., M.B., B.S., A.Z., A.K., M.W., S.C.) and Faculty of Chemistry (M.B.), Jagiellonian University, Krakow, Poland; Department of Toxicology, Faculty of Pharmacy (A.K., M.W.) and Chair of Pharmacology (S.C.), Jagiellonian University Medical College, Krakow, Poland; and Department of Human Nutrition, Faculty of Food Technology, University of Agriculture, Krakow, Poland (R.K.)
| | - Renata Kostogrys
- Jagiellonian Centre for Experimental Therapeutics (Ł.M., A.J., E.M., M.G.-G., M.B., B.S., A.Z., A.K., M.W., S.C.) and Faculty of Chemistry (M.B.), Jagiellonian University, Krakow, Poland; Department of Toxicology, Faculty of Pharmacy (A.K., M.W.) and Chair of Pharmacology (S.C.), Jagiellonian University Medical College, Krakow, Poland; and Department of Human Nutrition, Faculty of Food Technology, University of Agriculture, Krakow, Poland (R.K.)
| | - Agnieszka Zakrzewska
- Jagiellonian Centre for Experimental Therapeutics (Ł.M., A.J., E.M., M.G.-G., M.B., B.S., A.Z., A.K., M.W., S.C.) and Faculty of Chemistry (M.B.), Jagiellonian University, Krakow, Poland; Department of Toxicology, Faculty of Pharmacy (A.K., M.W.) and Chair of Pharmacology (S.C.), Jagiellonian University Medical College, Krakow, Poland; and Department of Human Nutrition, Faculty of Food Technology, University of Agriculture, Krakow, Poland (R.K.)
| | - Agnieszka Kij
- Jagiellonian Centre for Experimental Therapeutics (Ł.M., A.J., E.M., M.G.-G., M.B., B.S., A.Z., A.K., M.W., S.C.) and Faculty of Chemistry (M.B.), Jagiellonian University, Krakow, Poland; Department of Toxicology, Faculty of Pharmacy (A.K., M.W.) and Chair of Pharmacology (S.C.), Jagiellonian University Medical College, Krakow, Poland; and Department of Human Nutrition, Faculty of Food Technology, University of Agriculture, Krakow, Poland (R.K.)
| | - Maria Walczak
- Jagiellonian Centre for Experimental Therapeutics (Ł.M., A.J., E.M., M.G.-G., M.B., B.S., A.Z., A.K., M.W., S.C.) and Faculty of Chemistry (M.B.), Jagiellonian University, Krakow, Poland; Department of Toxicology, Faculty of Pharmacy (A.K., M.W.) and Chair of Pharmacology (S.C.), Jagiellonian University Medical College, Krakow, Poland; and Department of Human Nutrition, Faculty of Food Technology, University of Agriculture, Krakow, Poland (R.K.)
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (Ł.M., A.J., E.M., M.G.-G., M.B., B.S., A.Z., A.K., M.W., S.C.) and Faculty of Chemistry (M.B.), Jagiellonian University, Krakow, Poland; Department of Toxicology, Faculty of Pharmacy (A.K., M.W.) and Chair of Pharmacology (S.C.), Jagiellonian University Medical College, Krakow, Poland; and Department of Human Nutrition, Faculty of Food Technology, University of Agriculture, Krakow, Poland (R.K.)
| |
Collapse
|
12
|
Kawasaki Y, Yokobayashi E, Sakamoto K, Tenma E, Takaki H, Chiba Y, Otashiro T, Ishihara M, Yonezawa S, Sugiyama A, Natori Y. Angiostatin prevents IL-1β-induced down-regulation of eNOS expression by inhibiting the NF-κB cascade. J Pharmacol Sci 2015; 129:200-4. [DOI: 10.1016/j.jphs.2015.10.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 09/17/2015] [Accepted: 09/24/2015] [Indexed: 12/30/2022] Open
|
13
|
LI CHANGYI, YANG LINGCHAO, GUO KAI, WANG YUEPENG, LI YIGANG. Mitogen-activated protein kinase phosphatase-1: A critical phosphatase manipulating mitogen-activated protein kinase signaling in cardiovascular disease (Review). Int J Mol Med 2015; 35:1095-102. [DOI: 10.3892/ijmm.2015.2104] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 01/29/2015] [Indexed: 11/06/2022] Open
|
14
|
Shatanawi A, Lemtalsi T, Yao L, Patel C, Caldwell RB, Caldwell RW. Angiotensin II limits NO production by upregulating arginase through a p38 MAPK-ATF-2 pathway. Eur J Pharmacol 2014; 746:106-14. [PMID: 25446432 DOI: 10.1016/j.ejphar.2014.10.042] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 10/08/2014] [Accepted: 10/18/2014] [Indexed: 01/11/2023]
Abstract
Enhanced vascular arginase activity can impair endothelium-dependent vasorelaxation by decreasing l-arginine availability to endothelial nitric oxide (NO) synthase, thereby reducing NO production and uncoupling NOS function. Elevated angiotensin II (Ang II) is a key component of endothelial dysfunction in many cardiovascular diseases and has been linked to elevated arginase activity. In this study we explored the signaling pathway leading to increased arginase expression/activity in response to Ang II in bovine aortic endothelial cells (BAEC). Our previous studies indicate involvement of p38 mitogen activated protein kinase (MAPK) in Ang II-induced arginase upregulation and reduced NO production. In this study, we further investigated the Ang II-transcriptional regulation of arginase 1 in endothelial cells. Our results indicate the involvement of ATF-2 transcription factor of the AP1 family in arginase 1 upregulation and in limiting NO production. Using small interfering RNA (siRNA) targeting ATF-2, we showed that this transcription factor is required for Ang II-induced arginase 1 gene upregulation and increased arginase 1 expression and activity, leading to reduced NO production. Electrophoretic mobility shift assay and chromatin immunoprecipitation assay further confirmed the involvement of ATF-2. Moreover, our data indicate that p38 MAPK phosphorylates ATF-2 in response to Ang II. Collectively, our results indicate that Ang II increases endothelial arginase activity/expression through a p38 MAPK/ATF-2 pathway leading to reduced endothelial NO production. These signaling steps might be therapeutic targets for preventing vascular endothelial dysfunction associated with elevated arginase activity/expression.
Collapse
Affiliation(s)
- Alia Shatanawi
- Department of Pharmacology, Faculty of Medicine, The University of Jordan, Amman 11942, Jordan; Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA.
| | - Tahira Lemtalsi
- Vascular Biology Center, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA
| | - Lin Yao
- Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA
| | - Chintan Patel
- Vascular Biology Center, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA
| | - Ruth B Caldwell
- Vascular Biology Center, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA; VA Medical Center, Augusta, GA 30912, USA
| | - R William Caldwell
- Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA
| |
Collapse
|
15
|
Benard G, Trian T, Bellance N, Berger P, Lavie J, Espil-Taris C, Rocher C, Eimer-Bouillot S, Goizet C, Nouette-Gaulain K, Letellier T, Lacombe D, Rossignol R. Adaptative capacity of mitochondrial biogenesis and of mitochondrial dynamics in response to pathogenic respiratory chain dysfunction. Antioxid Redox Signal 2013; 19:350-65. [PMID: 22369111 DOI: 10.1089/ars.2011.4244] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
AIMS Cellular energy homeostasy relies on mitochondrial plasticity, the molecular determinants of which are multiple. Yet, the relative contribution of and possible cooperation between mitochondrial biogenesis and morphogenesis to cellular energy homeostasy remains elusive. Here we analyzed the adaptative capacity of mitochondrial content and dynamics in muscle biopsies of patients with a complex IV defect, and in skin fibroblasts challenged with complex IV inhibition. RESULTS We observed a biphasic variation of the mitochondrial content upon complex IV inhibition in muscle biopsies and in skin fibroblasts. Adjustment of mitochondrial content for respiratory maintenance was blocked by using a dominant negative form of CREB (CREB-M1) and by L-NAME, a blocker of NO production. Accordingly, cells treated with KCN 6 μM showed higher levels of phospho-CREB, PGC1α mRNA, eNOS mRNA, and mtTFA mRNA. We also observed the increased expression of the fission protein DRP1 during fibroblasts adaptation, as well as mitochondrial ultrastructural defects indicative of increased fission in patients muscle micrographs. Accordingly, the expression of a dominant negative form of DRP1 (K38A mutant) reduced the biogenic response in fibroblasts challenged with 6 μM KCN. INNOVATION Our findings indicate that mitochondrial biogenesis and mitochondrial fission cooperate to promote cellular adaptation to respiratory chain inhibition. CONCLUSIONS Our data show for the first time that DRP1 intervenes during the initiation of the mitochondrial adaptative response to respiratory chain defects. The evidenced pathway of mitochondrial adaptation to respiratory chain deficiency provides a safety mechanism against mitochondrial dysfunction.
Collapse
Affiliation(s)
- Giovanni Benard
- Université Bordeaux, Maladies Rares: Génétique et Métabolisme (MRGM), EA 4576, F-33000 Bordeaux, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Yamaguchi S, Inada C, Tamura M, Sato N, Yamada M, Itaba S, Okazaki S, Matsuura H, Fujii S, Matsuda F, Goto Y, Mochizuki H, Kurumatani H, Miyamoto M. Beraprost sodium improves survival rates in anti-glomerular basement membrane glomerulonephritis and 5/6 nephrectomized chronic kidney disease rats. Eur J Pharmacol 2013; 714:325-31. [PMID: 23911885 DOI: 10.1016/j.ejphar.2013.07.032] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Revised: 07/05/2013] [Accepted: 07/16/2013] [Indexed: 11/16/2022]
Abstract
Beraprost sodium, a stable prostacyclin analog, was showed to improve survival rates in two different rat models, anti-glomerular basement membrane (GBM) glomerulonephritis (GN) and 5/6 nephrectomized (Nx) chronic kidney disease (CKD) rats. In the anti-GBM rat, beraprost sodium (0.2 and 0.6 mg/kg/day) improved survival rate (hazard ratio for beraprost sodium 0.6 mg/kg/day group, 0.10; 95% confidence interval, 0.01 to 0.68). Subsequently, in the 5/6 Nx CKD rat, beraprost sodium (0.6 mg/kg/day) improved survival rate (hazard ratio for beraprost sodium, 0.46; 95% confidence interval, 0.23 to 0.92), serum creatinine doubling time and the slope of the reciprocal of serum creatinine. In the anti-GBM GN rats, beraprost sodium suppressed the serum accumulation of representative uremic toxins such as indoxyl sulfate. Furthermore, beraprost sodium inhibited human aortic endothelial cell (HAEC) injury induced by indoxyl sulfate, indicating that beraprost sodium might have a protective effect against cardiovascular damage due to CKD. These results show that beraprost sodium can improve the survival rates in two rat models of anti-GBM GN and 5/6 Nx CKD rats by protecting endothelial cells and thereby ameliorating decreased renal function. Therefore, clinical studies are needed in patients with chronic kidney failure to determine whether beraprost sodium will become a useful medication in CKD.
Collapse
Affiliation(s)
- Shinichi Yamaguchi
- Pharmacology Laboratory, Pharmaceutical Research Laboratories, Toray Industries, Inc., Kanagawa 248 8555, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Desch M, Hackmayer G, Todorov VT. Identification of ATF2 as a transcriptional regulator of renin gene. Biol Chem 2013; 393:93-100. [PMID: 22628303 DOI: 10.1515/bc-2011-157] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2011] [Accepted: 10/07/2011] [Indexed: 11/15/2022]
Abstract
The cAMP response element (enhCRE) in the distal enhancer regulatory region of renin gene is believed to play a major role in the control of renin transcription. enhCRE binds the CRE-binding protein (CREB), which is the main transcription factor target of cAMP signaling. Using the mouse renin-producing cell line As4.1 we found that activating transcription factor-2 (ATF2) also binds to enhCRE. N-terminal phosphorylation of ATF2, which controls its transactivation, is associated with downregulation of renin gene expression by the cytokine tumor necrosis factor-α (TNFα). The ubiquitin proteasome inhibitor MG132 also phosphorylates ATF2 and inhibits renin expression. Knockdown of ATF2 attenuated the suppression of renin gene expression by MG132, thus demonstrating that ATF2 mediates the inhibitory effect of MG132. In addition, MG132 increased the DNA-binding of ATF2 as well as the ratio of bound ATF2 to CREB. Using ATF2- and CREB-Gal4 fusion protein constructs coupled with luciferase reporter system we showed that ATF2 has a weaker transactivating capacity than CREB. These data suggest that ATF2 represses renin expression by drifting the transcriptional control of renin gene away from CREB. Accordingly, TNFα completely abrogated the cAMP-dependent stimulation of renin gene expression.
Collapse
Affiliation(s)
- Michael Desch
- Institute of Physiology, University of Regensburg, D93040 Regensburg, Germany
| | | | | |
Collapse
|
18
|
Shin S, Kim KJ, Chang HJ, Lee BW, Yang WI, Cha BS, Choi D. The effect of oral prostaglandin analogue on painful diabetic neuropathy: a double-blind, randomized, controlled trial. Diabetes Obes Metab 2013; 15:185-8. [PMID: 22974254 DOI: 10.1111/dom.12010] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2011] [Revised: 10/11/2011] [Accepted: 07/02/2012] [Indexed: 12/29/2022]
Abstract
This study was performed to assess the efficacy of beraprost sodium (BPS) in painful diabetic peripheral neuropathy (DPN) in type 2 diabetes mellitus (T2DM) patients. In this randomized clinical trial, 99 T2DM patients (41% male, age 60 ± 6 years) with DPN but without evidence of peripheral artery disease were randomized to receive either BPS (40 µg, tid) or placebo for 8 weeks. The primary end point was the improvement of the total symptom score (TSS), temperature rebound (TR) and nadir to peak (NP) above baseline. After 8 weeks treatment, the change of TSS in the BPS group showed a significant improvement compared to the placebo group (2.80 ± 2.48 vs. 1.60 ± 1.94 points, p = 0.009). Furthermore, the number of patients who showed signs of improvement in TSS and the proportion of patients with 50% relief of symptom was also significantly greater in the BPS group than in the placebo group (83.7 vs. 62%, p = 0.015, 36.2 vs. 14%, p = 0.009, respectively). In conclusion, treatment with BPS significantly improved TSS over an 8-week period.
Collapse
Affiliation(s)
- S Shin
- Division of Cardiology, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|
19
|
Clenbuterol upregulates histone demethylase JHDM2a via the β2-adrenoceptor/cAMP/PKA/p-CREB signaling pathway. Cell Signal 2012; 24:2297-306. [PMID: 22820505 DOI: 10.1016/j.cellsig.2012.07.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Revised: 06/20/2012] [Accepted: 07/16/2012] [Indexed: 01/19/2023]
Abstract
BACKGROUND β(2)-Adrenergic receptor (β(2)-AR) signaling activated by the agonist clenbuterol is important in the metabolism of muscle and adipose cells. Additionally, the significant role of histone demethylase JHDM2a in regulating metabolic gene expression was also recently demonstrated in Jhdm2a(-/-) mice. To elucidate the molecular mechanism involved in clenbuterol-induced adipocyte reduction from an epigenetic perspective, this study focused on cAMP-responsive element binding protein (CREB) to determine whether JHDM2a is regulated by the β(2)-AR/cAMP/protein kinase A (PKA) signaling pathway. RESULTS In porcine tissues treated with clenbuterol, JHDM2a expression was upregulated, and in porcine cells, expression of exogenous CREB led to increased JHDM2a expression. In addition, changes in JHDM2a expression were coincident with variations in the phosphorylation of CREB and p-CREB/CBP interaction in porcine and human cells treated with drugs known to modify the β(2)-AR/cAMP/PKA pathway. Finally, binding assays demonstrated that CREB regulated JHDM2a by binding directly to the CRE site nearest to the transcription start site. CONCLUSION Our results reveal that clenbuterol activates the β(2)-AR signaling pathway upstream of JHDM2a and that CREB acts as an intermediate link regulated by cAMP-PKA to induce activity of the JHDM2a promoter. These findings suggest that clenbuterol decreases adipose cell size and increases muscle fiber size in porcine tissues by virtue of JHDM2a-mediated demethylation, which regulates downstream metabolic and related genes.
Collapse
|
20
|
Si H, Yu J, Jiang H, Lum H, Liu D. Phytoestrogen genistein up-regulates endothelial nitric oxide synthase expression via activation of cAMP response element-binding protein in human aortic endothelial cells. Endocrinology 2012; 153:3190-8. [PMID: 22669896 PMCID: PMC3791435 DOI: 10.1210/en.2012-1076] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
We previously reported that genistein, a phytoestrogen, up-regulates endothelial nitric oxide synthase (eNOS) and prevents hypertension in rats that are independent of estrogen signaling machinery. However, how genistein regulates eNOS expression is unknown. In the present study, we show that genistein enhanced eNOS expression and NO synthesis in primary human aortic endothelial cells. Inhibition of extracellular signal regulated kinase, phosphoinositol-3 kinase, or protein kinase C did not affect genistein-enhanced eNOS expression and NO synthesis. However, chemical inhibition of protein kinase A (PKA) or adenoviral transfer of the specific endogenous PKA inhibitor gene completely abolished PKA activity and genistein-stimulated eNOS expression and NO production. Accordingly, genistein induced PKA activity and subsequent phosphorylation of cAMP response element (CRE)-binding protein (CREB) at Ser133. Suppression of CREB by small interfering RNA transfection abolished genistein-enhanced eNOS expression and NO production. Consistently, deletion of the CRE site within human eNOS promoter eliminated genistein-stimulated eNOS promoter activity. These findings provide the first evidence to our knowledge that genistein may play a beneficial role in vascular function through targeting the PKA/CREB/eNOS/NO signaling pathway.
Collapse
Affiliation(s)
- Hongwei Si
- Department of Human Nutrition, Foods, and Exercise, College of Agriculture and Life Sciences, Virginia Tech, Blacksburg, Virginia 24061, USA
| | | | | | | | | |
Collapse
|
21
|
Exogenous control of the expression of Group I CD1 molecules competent for presentation of microbial nonpeptide antigens to human T lymphocytes. Clin Dev Immunol 2011; 2011:790460. [PMID: 21603161 PMCID: PMC3095450 DOI: 10.1155/2011/790460] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Revised: 01/12/2011] [Accepted: 01/19/2011] [Indexed: 12/21/2022]
Abstract
Group I CD1 (CD1a, CD1b, and CD1c) glycoproteins expressed on immature and mature dendritic cells present nonpeptide antigens (i.e., lipid or glycolipid molecules mainly of microbial origin) to T cells. Cytotoxic CD1-restricted T lymphocytes recognizing mycobacterial lipid antigens were found in tuberculosis patients. However, thanks to a complex interplay between mycobacteria and CD1 system, M. tuberculosis possesses a successful tactic based, at least in part, on CD1 downregulation to evade CD1-dependent immunity. On the ground of these findings, it is reasonable to hypothesize that modulation of CD1 protein expression by chemical, biological, or infectious agents could influence host's immune reactivity against M. tuberculosis-associated lipids, possibly affecting antitubercular resistance. This scenario prompted us to perform a detailed analysis of the literature concerning the effect of external agents on Group I CD1 expression in order to obtain valuable information on the possible strategies to be adopted for driving properly CD1-dependent immune functions in human pathology and in particular, in human tuberculosis.
Collapse
|
22
|
Fang F, Yang Y, Yuan Z, Gao Y, Zhou J, Chen Q, Xu Y. Myocardin-related transcription factor A mediates OxLDL-induced endothelial injury. Circ Res 2011; 108:797-807. [PMID: 21330600 DOI: 10.1161/circresaha.111.240655] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
RATIONALE Atherosclerosis proceeds through a multistep reaction that begins with endothelial injury caused by a host of stress signals, among which oxidized low-density lipoprotein (oxLDL) plays a critical role. OxLDL disrupts normal functionality of the endothelium by upregulating adhesion molecules (eg, ICAM-1) and concomitantly downregulating endothelial nitric oxide synthase (eNOS) expression. The transcriptional modulator that mediates the cellular response to oxLDL remains largely obscure. OBJECTIVE Our goal was to determine whether myocardin-related transcription factor (MRTF)-A, a key protein involved in the transcriptional regulation of smooth muscle cell phenotype, is responsible for the endothelial injury by oxLDL, and, if so, how MRTF-A promotes the proatherogenic agenda initiated by oxLDL. METHODS AND RESULTS OxLDL stimulated the expression of MRTF-A in endothelial cells as evidenced by Western blotting and immunofluorescence. Overexpression of MRTF-A synergistically enhanced the induction of ICAM-1 and suppression of eNOS by oxLDL. In contrast, disruption of MRTF-A, either by small interfering RNA or dominant negative mutation, abrogated the pathogenic program triggered by oxLDL. Finally, chromatin immunoprecipitation assays indicate that oxLDL preferentially augmented MRTF-A binding to ICAM-1 and eNOS promoters and that MRTF-A drove differential epigenetic alterations taking place on these promoters in response to oxLDL. CONCLUSIONS Therefore, our data provide the first demonstration that MRTF-A is critically linked to pivotal pathophysiological events in the vascular endothelium.
Collapse
Affiliation(s)
- Fei Fang
- Nanjing Medical University, 140 Hanzhong Rd., Nanjing, Jiangsu, China.
| | | | | | | | | | | | | |
Collapse
|
23
|
Smolková K, Plecitá-Hlavatá L, Bellance N, Benard G, Rossignol R, Ježek P. Waves of gene regulation suppress and then restore oxidative phosphorylation in cancer cells. Int J Biochem Cell Biol 2010; 43:950-68. [PMID: 20460169 DOI: 10.1016/j.biocel.2010.05.003] [Citation(s) in RCA: 171] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2009] [Revised: 03/05/2010] [Accepted: 05/04/2010] [Indexed: 12/17/2022]
Abstract
We posit the following hypothesis: Independently of whether malignant tumors are initiated by a fundamental reprogramming of gene expression or seeded by stem cells, "waves" of gene expression that promote metabolic changes occur during carcinogenesis, beginning with oncogene-mediated changes, followed by hypoxia-induced factor (HIF)-mediated gene expression, both resulting in the highly glycolytic "Warburg" phenotype and suppression of mitochondrial biogenesis. Because high proliferation rates in malignancies cause aglycemia and nutrient shortage, the third (second oncogene) "wave" of adaptation stimulates glutaminolysis, which in certain cases partially re-establishes oxidative phosphorylation; this involves the LKB1-AMPK-p53, PI3K-Akt-mTOR axes and MYC dysregulation. Oxidative glutaminolysis serves as an alternative pathway compensating for cellular ATP. Together with anoxic glutaminolysis it provides pyruvate, lactate, and the NADPH pool (alternatively to pentose phosphate pathway). Retrograde signaling from revitalized mitochondria might constitute the fourth "wave" of gene reprogramming. In turn, upon reversal of the two Krebs cycle enzymes, glutaminolysis may partially (transiently) function even during anoxia, thereby further promoting malignancy. The history of the carcinogenic process within each malignant tumor determines the final metabolic phenotype of the selected surviving cells, resulting in distinct cancer bioenergetic phenotypes ranging from the highly glycolytic "classic Warburg" to partial or enhanced oxidative phosphorylation. We discuss the bioenergetically relevant functions of oncogenes, the involvement of mitochondrial biogenesis/degradation in carcinogenesis, the yet unexplained Crabtree effect of instant glucose blockade of respiration, and metabolic signaling stemming from the accumulation of succinate, fumarate, pyruvate, lactate, and oxoglutarate by interfering with prolyl hydroxylase domain enzyme-mediated hydroxylation of HIFα prolines.
Collapse
Affiliation(s)
- Katarína Smolková
- Department of Membrane Transport Biophysics, Institute of Physiology, vvi, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | | | | | | | | | | |
Collapse
|
24
|
Protective effects of prostaglandin E1 on human umbilical vein endothelial cell injury induced by hydrogen peroxide. Acta Pharmacol Sin 2010; 31:485-92. [PMID: 20305680 DOI: 10.1038/aps.2010.23] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
AIM To investigate the protective effects of prostaglandin E(1) (PGE(1)) against H(2)O(2)-induced oxidative damage on human umbilical vein endothelial cells (HUVECs). METHODS HUVECs were pretreated with PGE(1) (0.25, 0.50, and 1.00 micromol/L) for 24 h and exposed to H(2)O(2) (200 micromol/L) for 12 h, and cell viability was measured by the MTT assay. LDH, NO, SOD, GSH-Px, MDA, ROS, and apoptotic percentage were determined. eNOS expression was measured by Western blotting and real-time PCR. RESULTS PGE(1) (0.25-1.00 micromol/L) was able to markedly restore the viability of HUVECs under oxidative stress, and scavenged intracellular reactive oxygen species induced by H(2)O(2). PGE(1) also suppressed the production of lipid peroxides, such as MDA, restored the activities of endogenous antioxidants including SOD and GSH-Px, and inhibited cell apoptosis. In addition, PGE(1) significantly increased NO content, eNOS protein, and mRNA expression. CONCLUSION PGE(1) effectively protected endothelial cells against oxidative stress induced by H(2)O(2), an activity that might depend on the up-regulation of NO expression.
Collapse
|
25
|
Sato N, Kaneko M, Tamura M, Kurumatani H. The prostacyclin analog beraprost sodium ameliorates characteristics of metabolic syndrome in obese Zucker (fatty) rats. Diabetes 2010; 59:1092-100. [PMID: 20068136 PMCID: PMC2844818 DOI: 10.2337/db09-1432] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2009] [Accepted: 12/21/2009] [Indexed: 11/13/2022]
Abstract
OBJECTIVE The prostacyclin analog, beraprost sodium (BPS), was examined for its potential to improve the symptoms of obesity-type diabetes (i.e., hyperglycemia, hyperinsulinemia, dyslipidemia, histopathologic changes, and diabetic complications). RESEARCH DESIGN AND METHODS Obese Zucker rats, an experimental model of genetic obesity-induced type 2 diabetes, were repeatedly administered BPS at oral doses of 0.2 or 0.6 mg x kg(-1) x day(-1) b.i.d. for 12 weeks, and serum chemistry, urinalysis, and histopathologic examination were performed. RESULTS BPS dose-dependently suppressed serum glucose, insulin, triglyceride, and cholesterol levels in obese animals. In oral glucose tolerance test, BPS suppressed the post-glucose-loading elevation of serum glucose in a dose-dependent manner. Urinary N-acetyl-beta-D-glucosaminidase was significantly lower in BPS-treated obese animals compared with control animals, although no significant differences were observed in urinary protein levels between the BPS-treated groups and the control group. In addition, histopathologic examination revealed significant protective effects of BPS against renal disorder in obese animals. Histopathologically, BPS also inhibited the progression of hepatic steatosis, hypertrophy of adipose tissue, and pancreatic fibrosis. Furthermore, thermographic analysis of the hind limb sole skin surface indicated a significant increase in temperature in BPS-treated animals, compared with control animals, which was likely due to improved blood circulation by administration of BPS. CONCLUSIONS BPS suppressed the pathogenesis and development of diabetes and its complication, nephropathy, which was presumably accompanied by improving glucose intolerance and insulin resistance in obese Zucker rats.
Collapse
Affiliation(s)
- Nahoko Sato
- The First Pharmacology Laboratory, Pharmaceutical Research Labs, Toray Industries, Kamakura, Kanagawa, Japan.
| | | | | | | |
Collapse
|
26
|
Multi-site control and regulation of mitochondrial energy production. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2010; 1797:698-709. [PMID: 20226160 DOI: 10.1016/j.bbabio.2010.02.030] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2009] [Revised: 02/19/2010] [Accepted: 02/24/2010] [Indexed: 12/21/2022]
Abstract
With the extraordinary progress of mitochondrial science and cell biology, novel biochemical pathways have emerged as strategic points of bioenergetic regulation and control. They include mitochondrial fusion, fission and organellar motility along microtubules and microfilaments (mitochondrial dynamics), mitochondrial turnover (biogenesis and degradation), and mitochondrial phospholipids synthesis. Yet, much is still unknown about the mutual interaction between mitochondrial energy state, biogenesis, dynamics and degradation. Meanwhile, clinical research into metabolic abnormalities in tumors as diverse as renal carcinoma, glioblastomas, paragangliomas or skin leiomyomata, has designated new genes, oncogenes and oncometabolites involved in the regulation of cellular and mitochondrial energy production. Furthermore, the examination of rare neurological diseases such as Charcot-Marie Tooth type 2a, Autosomal Dominant Optic Atrophy, Lethal Defect of Mitochondrial and Peroxisomal Fission, or Spastic Paraplegia suggested involvement of MFN2, OPA1/3, DRP1 or Paraplegin, in the auxiliary control of mitochondrial energy production. Lastly, advances in the understanding of mitochondrial apoptosis have suggested a supplementary role for Bcl2 or Bax in the regulation of mitochondrial respiration and dynamics, which has fostered the investigation of alternative mechanisms of energy regulation. In this review, we discuss the regulatory mechanisms of cellular and mitochondrial energy production, and we emphasize the importance of the study of rare neurological diseases in addition to more common disorders such as cancer, for the fundamental understanding of cellular and mitochondrial energy production.
Collapse
|
27
|
Jin Y, Calvert TJ, Chen B, Chicoine LG, Joshi M, Bauer JA, Liu Y, Nelin LD. Mice deficient in Mkp-1 develop more severe pulmonary hypertension and greater lung protein levels of arginase in response to chronic hypoxia. Am J Physiol Heart Circ Physiol 2010; 298:H1518-28. [PMID: 20173047 DOI: 10.1152/ajpheart.00813.2009] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The mitogen-activated protein (MAP) kinases are involved in cellular responses to many stimuli, including hypoxia. MAP kinase signaling is regulated by a family of phosphatases that include MAP kinase phosphatase-1 (MKP-1). We hypothesized that mice lacking the Mkp-1 gene would have exaggerated chronic hypoxia-induced pulmonary hypertension. Wild-type (WT) and Mkp-1(-/-) mice were exposed to either 4 wk of normoxia or hypobaric hypoxia. Following chronic hypoxia, both genotypes demonstrated elevated right ventricular pressures, right ventricular hypertrophy as demonstrated by the ratio of the right ventricle to the left ventricle plus septum weights [RV(LV + S)], and greater vascular remodeling. However, the right ventricular systolic pressures, the RV/(LV + S), and the medial wall thickness of 100- to 300-microm vessels was significantly greater in the Mkp-1(-/-) mice than in the WT mice following 4 wk of hypobaric hypoxia. Chronic hypoxic exposure caused no detectable change in eNOS protein levels in the lungs in either genotype; however, Mkp-1(-/-) mice had lower levels of eNOS protein and lower lung NO production than did WT mice. No iNOS protein was detected in the lungs by Western blotting in any condition in either genotype. Both arginase I and arginase II protein levels were greater in the lungs of hypoxic Mkp-1(-/-) mice than those in hypoxic WT mice. Lung levels of proliferating cell nuclear antigen were greater in hypoxic Mkp-1(-/-) than in hypoxic WT mice. These data are consistent with the concept that MKP-1 acts to restrain hypoxia-induced arginase expression and thereby reduces vascular remodeling and the severity of pulmonary hypertension.
Collapse
Affiliation(s)
- Yi Jin
- The Research Institute at Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Kawabe JI, Ushikubi F, Hasebe N. Prostacyclin in Vascular Diseases - Recent Insights and Future Perspectives -. Circ J 2010; 74:836-43. [DOI: 10.1253/circj.cj-10-0195] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Jun-ichi Kawabe
- Departments of Cardiovascular Regeneration and Innovation, Asahikawa Medical College
| | | | - Naoyuki Hasebe
- Departments of Cardiovascular Regeneration and Innovation, Asahikawa Medical College
- Department of Internal Medicine, Cardiovascular, Respiratory and Neurology Division, Asahikawa Medical College
| |
Collapse
|
29
|
Gasparri F, Galvani A. Image-based high-content reporter assays: limitations and advantages. DRUG DISCOVERY TODAY. TECHNOLOGIES 2010; 7:e1-e94. [PMID: 24103681 DOI: 10.1016/j.ddtec.2010.04.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
|
30
|
Strassheim D, Riddle SR, Burke DL, Geraci MW, Stenmark KR. Prostacyclin inhibits IFN-gamma-stimulated cytokine expression by reduced recruitment of CBP/p300 to STAT1 in a SOCS-1-independent manner. THE JOURNAL OF IMMUNOLOGY 2009; 183:6981-8. [PMID: 19915063 DOI: 10.4049/jimmunol.0901045] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Increasing evidence indicates that pulmonary arterial hypertension is a vascular inflammatory disease. Prostacyclin (PGI(2)) is widely used to treat pulmonary arterial hypertension and is believed to benefit patients largely through vasodilatory effects. PGI(2) is also increasingly believed to have anti-inflammatory effects, including decreasing leukocyte cytokine production, yet few mechanistic details exist to explain how these effects are mediated at the transcriptional level. Because activated monocytes are critical sources of MCP-1 and other cytokines in cardiovascular inflammation, we examined the effects of iloprost on IFN-gamma- and IL-6-stimulated cytokine production in human monocytes. We found that iloprost inhibited IFN-gamma- and IL-6-induced MCP-1, IL-8, RANTES, and TNF-alpha production in monocytes, indicating wide-ranging anti-inflammatory action. We found that activation of STAT1 was critical for IFN-gamma-induced MCP-1 production and demonstrated that iloprost inhibited STAT1 activation by several actions as follows: 1) iloprost inhibited the phosphorylation of STAT1-S727 in the transactivation domain, thereby reducing recruitment of the histone acetylase and coactivator CBP/p300 to STAT1; 2) iloprost selectively inhibited activation of JAK2 but not JAK1, both responsible for activation of STAT1 via phosphorylation of STAT1-Y701, resulting in reduced nuclear recruitment and activation of STAT1; and 3) SOCS-1, which normally terminates IFN-gamma-signaling, was not involved in iloprost-mediated inhibition of STAT1, indicating divergence from the classical pathway for terminating IFN-gamma-signaling. We conclude that PGI(2) exerts anti-inflammatory action by inhibiting STAT1-induced cytokine production, in part by targeting the transactivation domain-induced recruitment of the histone acetylase CBP/p300.
Collapse
Affiliation(s)
- Derek Strassheim
- Pediatric Critical CareMedicine, University of Colorado Denver, Research Complex II, Room 6490, 12700 East 19th Avenue, Aurora, CO 80045, USA.
| | | | | | | | | |
Collapse
|
31
|
Abstract
AMPK (AMP-activated protein kinase) is a key regulator of cellular energy because of its capacity to detect changes in the concentration of AMP. Recent evidence, however, indicates the existence of alternative mechanisms of activation of this protein. Mitochondrial ROS (reactive oxygen species), generated as a result of the interaction between nitric oxide and mitochondrial cytochrome c oxidase, activate AMPKα1 in HUVECs (human umbilical-vein endothelial cells) at a low oxygen concentration (i.e. 3%). This activation is independent of changes in AMP. In the present study we show, using HUVECs in which AMPKα1 has been silenced, that this protein is responsible for the expression of genes involved in antioxidant defence, such as manganese superoxide dismutase, catalase, γ-glutamylcysteine synthase and thioredoxin. Furthermore, peroxisome proliferator-activated-coactivator-1, cAMP-response-element-binding protein and Foxo3a (forkhead transcription factor 3a) are involved in this signalling pathway. In addition, we show that silencing AMPKα1 in cells results in a reduced mitochondrial and eNOS (endothelial NO synthase) content, reduced cell proliferation, increased accumulation of ROS and apoptosis. Thus AMPKα1 in HUVECs regulates both their mitochondrial content and their antioxidant defences. Pharmacological activation of AMPKα1 in the vascular endothelium may be beneficial in conditions such as metabolic syndrome, Type 2 diabetes and atherosclerosis, not only because of its bioenergetic effects but also because of its ability to counteract oxidative stress.
Collapse
|
32
|
Takenaka M, Machida N, Ida N, Satoh N, Kurumatani H, Yamane Y. Effect of beraprost sodium (BPS) in a new rat partial unilateral ureteral obstruction model. Prostaglandins Leukot Essent Fatty Acids 2009; 80:263-7. [PMID: 19464862 DOI: 10.1016/j.plefa.2009.03.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2008] [Revised: 12/17/2008] [Accepted: 03/25/2009] [Indexed: 12/19/2022]
Abstract
Unilateral ureteral obstruction (UUO) is a representative model for investigating the common mechanism of decreasing renal function in chronic renal failure. In this study, we present a new partial UUO model in adult rats and evaluated the effect of beraprost sodium (BPS: stable prostaglandin I(2) (PGI(2)) analog). We could make reproductive and uniform partial UUO by ligating the left ureter together with a 0.5 mm diameter stainless steel wire with nylon thread, and withdrawing the stainless wire. One week later, the ureteral obstruction was released. After 3 weeks from the release of UUO, all animals of control group, without BPS administration, developed basophilic degeneration of tubular epithelium, tubular dilatation and interstitial fibrosis. The areas of tubular degeneration and fibrosis were significantly reduced in the BPS group, orally administered BPS 300 microg/kg twice a day from the next day of the release of obstruction, than in control group. In conclusion, we can established the adult rat partial UUO-release model and revealed that BPS can inhibit renal tubular damage and tubulointerstitial fibrosis.
Collapse
Affiliation(s)
- Masahiko Takenaka
- Takenaka Animal Hospital, 3-10-3 Zao-cho, Fukuyama, Hiroshima 721-0971, Japan
| | | | | | | | | | | |
Collapse
|
33
|
Deng X, Liu H, Huang J, Cheng L, Keller ET, Parsons SJ, Hu CD. Ionizing radiation induces prostate cancer neuroendocrine differentiation through interplay of CREB and ATF2: implications for disease progression. Cancer Res 2009; 68:9663-70. [PMID: 19047143 DOI: 10.1158/0008-5472.can-08-2229] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Radiation therapy is a first-line treatment for prostate cancer patients with localized tumors. Although some patients respond well to the treatment, approximately 10% of low-risk and up to 60% of high-risk prostate cancer patients experience recurrent tumors. However, the molecular mechanisms underlying tumor recurrence remain largely unknown. Here we show that fractionated ionizing radiation (IR) induces differentiation of LNCaP prostate cancer cells into neuroendocrine (NE)-like cells, which are known to be implicated in prostate cancer progression, androgen-independent growth, and poor prognosis. Further analyses revealed that two cyclic AMP-responsive element binding transcription factors, cyclic AMP-response element binding protein (CREB) and activating transcription factor 2 (ATF2), function as a transcriptional activator and a repressor, respectively, of NE-like differentiation and that IR induces NE-like differentiation by increasing the nuclear content of phospho-CREB and cytoplasmic accumulation of ATF2. Consistent with this notion, stable expression of a nonphosphorylatable CREB or a constitutively nuclear-localized ATF2 in LNCaP cells inhibits IR-induced NE-like differentiation. IR-induced NE-like morphologies are reversible, and three IR-resistant clones isolated from dedifferentiated cells have acquired the ability to proliferate and lost the NE-like cell properties. In addition, these three IR-resistant clones exhibit differential responses to IR- and androgen depletion-induced NE-like differentiation. However, they are all resistant to cell death induced by IR and the chemotherapeutic agent docetaxel and to androgen depletion-induced growth inhibition. These results suggest that radiation therapy-induced NE-like differentiation may represent a novel pathway by which prostate cancer cells survive the treatment and contribute to tumor recurrence.
Collapse
Affiliation(s)
- Xuehong Deng
- Department of Medicinal Chemistry and Molecular Pharmacology and the Purdue Cancer Center, Purdue University, West Lafayette, Indiana 47907, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Chinnappan D, Qu X, Xiao D, Ratnasari A, Weber HC. Human gastrin-releasing peptide receptor gene regulation requires transcription factor binding at two distinct CRE sites. Am J Physiol Gastrointest Liver Physiol 2008; 295:G153-G162. [PMID: 18483184 PMCID: PMC2494719 DOI: 10.1152/ajpgi.00036.2008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Ectopic expression of the gastrin-releasing peptide (GRP) receptor (GRP-R) occurs frequently in human malignancies of the gastrointestinal tract. Owing to paracrine and autocrine interaction with its specific high-affinity ligand GRP, tumor cell proliferation, migration, and invasion might ensue. Here we provide the first insights regarding molecular mechanisms of GRP-R regulation in gastrointestinal cancer cells. We identified by EMSA and chromatin immunoprecipitation assays two cAMP response element (CRE) binding sites that recruited transcription factor CRE binding protein (CREB) to the human GRP-R promoter. Transfection studies with a wild-type human GRP-R promoter reporter and corresponding CRE mutants showed that both CRE sites are critical for basal transcriptional activation in gastrointestinal cancer cells. Forced expression of cAMP-dependent effectors CREB and PKA resulted in robust upregulation of human GRP-R transcriptional activity, and this overexpression strictly required intact wild-type CRE sites. Direct cAMP stimulation with forskolin resulted in enhanced human GRP-R promoter activity only in HuTu-80 cells, but not in Caco-2 cells, coinciding with forskolin-induced CREB phosphorylation occurring only in HuTu-80 but not Caco-2 cells. In summary, CREB is a critical regulator of human GRP-R expression in gastrointestinal cancer and might be activated through different upstream intracellular pathways.
Collapse
Affiliation(s)
- Dharmaraj Chinnappan
- Boston University School of Medicine, Section of Gastroenterology, Boston, Massachusetts
| | - Xiangping Qu
- Boston University School of Medicine, Section of Gastroenterology, Boston, Massachusetts
| | - Dongmei Xiao
- Boston University School of Medicine, Section of Gastroenterology, Boston, Massachusetts
| | - Anita Ratnasari
- Boston University School of Medicine, Section of Gastroenterology, Boston, Massachusetts
| | - H. Christian Weber
- Boston University School of Medicine, Section of Gastroenterology, Boston, Massachusetts
| |
Collapse
|
35
|
Kubota T, Kubota N, Kadowaki T. [Endothelial function and insulin resistance]. Nihon Yakurigaku Zasshi 2008; 131:85-88. [PMID: 18277005 DOI: 10.1254/fpj.131.85] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
|
36
|
Colmone A, Li S, Wang CR. Activating Transcription Factor/cAMP Response Element Binding Protein Family Member Regulated Transcription of CD1A. THE JOURNAL OF IMMUNOLOGY 2006; 177:7024-32. [PMID: 17082618 DOI: 10.4049/jimmunol.177.10.7024] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
CD1a has a unique expression pattern among Ag-presenting molecules, expressed specifically on cortical thymocytes and APCs. As autoimmune disease, infection, and tumors can all result in alteration of CD1a expression, we are attempting to characterize the transcriptional regulation, and thus shed some light on specific expression, of CD1A. In this study, we have identified a minimal proximal promoter region required for CD1A transcription. Computer searches within this region identified numerous potential binding sites for lymphoid-specific transcription factors, including the ETS transcription factors, C/EBP, GATA, and CREB. Deletion and site-specific mutant analysis revealed a critical role of a potential cAMP response element (CRE) 965 bp upstream of the CD1A translation start site. Two activating transcription factor (ATF)/CREB family members, CREB-1 and ATF-2, are able to bind this site in vitro and in vivo. Notably, activation of ATF/CREB family members decreases CD1A transcription, while decrease in ATF-2 expression results in increased CD1A RNA level. The fact that these factors also bind the CD1A promoter in human monocytes strongly suggests a role for ATF/CREB family members in regulation of CD1A expression.
Collapse
Affiliation(s)
- Angela Colmone
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA
| | | | | |
Collapse
|