1
|
Bai C, Zhang F, Yang Z, Zhang Y, Guo D, Zhang Q. Formaldehyde induced the cardiac damage by regulating the NO/cGMP signaling pathway and L-Ca 2+ channels. Toxicol Res (Camb) 2023; 12:1105-1112. [PMID: 38145098 PMCID: PMC10734627 DOI: 10.1093/toxres/tfad102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 09/25/2023] [Accepted: 10/09/2023] [Indexed: 12/26/2023] Open
Abstract
Background Formaldehyde (FA) is a common environmental pollutant that has been found to cause negative cardiovascular effects, however, the toxicological mechanism is not well understood. In this study, we investigated the molecular effects of the Nitric Oxide (NO)/cyclic Guanosine Monophosphate (cGMP) signaling pathway and L-type calcium (L-Ca2+) channels in rat hearts. Methods We designed the short-term FA exposure on the rat heart in different concentrations (0, 0.5, 3, 18 mg/m3). After 7 days of exposure, the rats were sacrificed and the rat tissues were removed for various experiments. Results Our experimental data showed that FA resulted in the upregulation NO and cGMP, especially at 18 mg/m3. Further, when exposed to high concentrations of FA, Cav1.2 and Cav1.3 expression decreased. We conclude that the NO/cGMP signaling pathway and downstream related channels can be regulated by increasing the production of NO in the low concentration group of FA. High concentration FA directly regulates L-Ca22+ channels. Conclusion This study suggests that FA damages the function of the cardiovascular system by regulating the NO/cGMP signaling pathway and L-Ca2+ channels.
Collapse
Affiliation(s)
- Caixia Bai
- College of Environment and Resource, Shanxi University, Taiyuan 030006, China
- Institute of Environmental Science, Shanxi University, Taiyuan 030006, China
| | - Fu Zhang
- College of Environment and Resource, Shanxi University, Taiyuan 030006, China
| | - Zhenhua Yang
- Institute of Environmental Science, Shanxi University, Taiyuan 030006, China
- Shanxi Laboratory for Yellow River, Shanxi University, Taiyuan 030006, China
| | - Yuexia Zhang
- Institute of Environmental Science, Shanxi University, Taiyuan 030006, China
- Shanxi Laboratory for Yellow River, Shanxi University, Taiyuan 030006, China
| | - Donggang Guo
- College of Environment and Resource, Shanxi University, Taiyuan 030006, China
- Shanxi Laboratory for Yellow River, Shanxi University, Taiyuan 030006, China
| | - Quanxi Zhang
- College of Environment and Resource, Shanxi University, Taiyuan 030006, China
- Institute of Environmental Science, Shanxi University, Taiyuan 030006, China
- Shanxi Laboratory for Yellow River, Shanxi University, Taiyuan 030006, China
| |
Collapse
|
2
|
Nasef M, Ben Turkia H, Haider Ali AM, Mahdawi E, Nair A. To What Extent Does Arginine Reduce the Risk of Developing Necrotizing Enterocolitis? Cureus 2023; 15:e45813. [PMID: 37876383 PMCID: PMC10591459 DOI: 10.7759/cureus.45813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/21/2023] [Indexed: 10/26/2023] Open
Abstract
Necrotizing enterocolitis (NEC) and neonatal sepsis are polar opposite diseases that are commonly encountered in the NICU. Concerning the frequency of these pathologies, NEC is regarded as being a much rarer condition, whereas neonatal sepsis is slightly more commonly encountered. However, neonatal sepsis can present with varying clinical presentations and, if caught late, can be detrimental to the patient. Many different modes of therapies have been studied for both conditions at different levels of pathologies, from a microscopic to a macroscopic level, leading to an assessment of treatment approaches. With the different ongoing treatment protocols being studied, one such therapy under investigation that does stand out is the use of L-arginine in both conditions. The L-arginine, being an essential amino acid, has many basic biological roles in developing neonates. It mainly involves the production of nitric oxide (NO), a potent vasodilator, which is particularly important in the development of vasculature in almost every organ. In premature infants, poorly developed vasculature makes them more susceptible to injury, therefore increasing the risk of diseases such as NEC and the severity of diseases such as neonatal sepsis. By assessing the uses of L-arginine and its application towards treating conditions like NEC and neonatal sepsis, we aim to identify its potential benefits as a treatment and its potential applications in clinical practice by understanding its basic functions and role in the pathophysiology of NEC and neonatal sepsis.
Collapse
Affiliation(s)
- Minoosh Nasef
- Neonatology, King Hamad University Hospital, Muharraq, BHR
| | | | | | - Esam Mahdawi
- Obstetrics and Gynaecology, King Hamad University Hospital, Muharraq, BHR
| | - Arun Nair
- Pediatrics, Saint Peter's University Hospital, Somerset, USA
| |
Collapse
|
3
|
OZTAN O, ŞİMŞİR C, TUTKUN L, HOCAOGLU A, DENİZ S, TÜRKSOY VA, ALAGÜNEY ME. Investigation of the relationship between placenta trace element levels and methylated arginines. JOURNAL OF HEALTH SCIENCES AND MEDICINE 2021. [DOI: 10.32322/jhsm.962489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
4
|
Dall'Asta A, Kumar S. Prelabor and intrapartum Doppler ultrasound to predict fetal compromise. Am J Obstet Gynecol MFM 2021; 3:100479. [PMID: 34496306 DOI: 10.1016/j.ajogmf.2021.100479] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 08/23/2021] [Accepted: 08/30/2021] [Indexed: 12/17/2022]
Abstract
According to current estimates, over 20% of the 4 million neonatal deaths occurring globally every year are related to intrapartum hypoxic complications that happen as a result of uterine contractions against a background of inadequate placental function. Most of such intrapartum complications occur among apparently uncomplicated term pregnancies. Available evidence suggests that current risk-assessment strategies do not adequately identify many of the fetuses vulnerable to periods of intermittent hypoxia that characterize human labor. In this review, we discuss the data available on Doppler ultrasound for the evaluation of placental function before and during labor in appropriately grown fetuses; we also discuss the current strategies for ultrasound-based risk stratification, the physiology of intrapartum compromise, and the potential future treatments to prevent fetal distress in labor and reduce perinatal complications related to birth asphyxia.
Collapse
Affiliation(s)
- Andrea Dall'Asta
- Obstetrics and Gynecology Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy (Dr Dall'Asta); Department of Metabolism, Digestion and Reproduction, Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom (Dr Dall'Asta).
| | - Sailesh Kumar
- Mater Research Institute, University of Queensland, Queensland, Australia (Dr Kumar); Faculty of Medicine, The University of Queensland, Queensland, Australia (Dr Kumar)
| |
Collapse
|
5
|
Visniauskas B, Perry JC, Gomes GN, Nogueira-Pedro A, Paredes-Gamero EJ, Tufik S, Chagas JR. Intermittent hypoxia changes the interaction of the kinin-VEGF system and impairs myocardial angiogenesis in the hypertrophic heart. Physiol Rep 2021; 9:e14863. [PMID: 33991464 PMCID: PMC8123545 DOI: 10.14814/phy2.14863] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/29/2021] [Accepted: 04/13/2021] [Indexed: 12/12/2022] Open
Abstract
Intermittent hypoxia (IH) is a feature of obstructive sleep apnea (OSA), a condition highly associated with hypertension-related cardiovascular diseases. Repeated episodes of IH contribute to imbalance of angiogenic growth factors in the hypertrophic heart, which is key in the progression of cardiovascular complications. In particular, the interaction between vascular endothelial growth factor (VEGF) and the kallikrein-kinin system (KKS) is essential for promoting angiogenesis. However, researchers have yet to investigate experimental models of IH that reproduce OSA, myocardial angiogenesis, and expression of KKS components. We examined temporal changes in cardiac angiogenesis in a mouse IH model. Adult male C57BI/6 J mice were implanted with Matrigel plugs and subjected to IH for 1-5 weeks with subsequent weekly histological evaluation of vascularization. Expression of VEGF and KKS components was also evaluated. After 3 weeks, in vivo myocardial angiogenesis and capillary density were decreased, accompanied by a late increase of VEGF and its type 2 receptor. Furthermore, IH increased left ventricular myocardium expression of the B2 bradykinin receptor, while reducing mRNA levels of B1 receptor. These results suggest that in IH, an unexpected response of the VEGF and KKS systems could explain the reduced capillary density and impaired angiogenesis in the hypoxic heart, with potential implications in hypertrophic heart malfunction.
Collapse
Affiliation(s)
- Bruna Visniauskas
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Juliana C Perry
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Guiomar N Gomes
- Departmento de Fisiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | | | - Sergio Tufik
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Jair R Chagas
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil.,Departamento de Biofísica, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
6
|
Shakeri H, Boen JRA, De Moudt S, Hendrickx JO, Leloup AJA, Jacobs G, De Meyer GRY, De Keulenaer GW, Guns PJDF, Segers VFM. Neuregulin-1 compensates for endothelial nitric oxide synthase deficiency. Am J Physiol Heart Circ Physiol 2021; 320:H2416-H2428. [PMID: 33989083 DOI: 10.1152/ajpheart.00914.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Endothelial cells (ECs) secrete different paracrine signals that modulate the function of adjacent cells; two examples of these paracrine signals are nitric oxide (NO) and neuregulin-1 (NRG1), a cardioprotective growth factor. Currently, it is undetermined whether one paracrine factor can compensate for the loss of another. Herein, we hypothesized that NRG1 can compensate for endothelial NO synthase (eNOS) deficiency. We characterized eNOS null and wild-type (WT) mice by cardiac ultrasound and histology and we determined circulating NRG1 levels. In a separate experiment, eight groups of mice were divided into four groups of eNOS null mice and WT mice; half of the mice received angiotensin II (ANG II) to induce a more severe phenotype. Mice were randomized to daily injections with NRG1 or vehicle for 28 days. eNOS deficiency increased NRG1 plasma levels, indicating that ECs increase their NRG1 expression when NO production is deleted. eNOS deficiency also increased blood pressure, lowered heart rate, induced cardiac fibrosis, and affected diastolic function. In eNOS null mice, ANG II administration not only increased cardiac fibrosis but also induced cardiac hypertrophy and renal fibrosis. NRG1 administration prevented cardiac and renal hypertrophy and fibrosis caused by ANG II infusion and eNOS deficiency. Moreover, Nrg1 expression in the myocardium is shown to be regulated by miR-134. This study indicates that administration of endothelium-derived NRG1 can compensate for eNOS deficiency in the heart and kidneys.NEW & NOTEWORTHY ECs compensate for eNOS deficiency by increasing the secretion of NRG1. NRG1 administration prevents cardiac and renal hypertrophy and fibrosis caused by ANG II infusion and eNOS deficiency. NRG1 expression is regulated by miR-134.
Collapse
Affiliation(s)
- Hadis Shakeri
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Jente R A Boen
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium.,Research Group Cardiovascular Diseases, GENCOR Department, University of Antwerp, Antwerp, Belgium
| | - Sofie De Moudt
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Jhana O Hendrickx
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Arthur J A Leloup
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Griet Jacobs
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Guido R Y De Meyer
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Gilles W De Keulenaer
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium.,Hartcentrum ZNA, Antwerp, Belgium
| | | | - Vincent F M Segers
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium.,Department of Cardiology, University Hospital Antwerp, Edegem, Belgium
| |
Collapse
|
7
|
Liu M, Iosef C, Rao S, Domingo-Gonzalez R, Fu S, Snider P, Conway SJ, Umbach GS, Heilshorn SC, Dewi RE, Dahl MJ, Null DM, Albertine KH, Alvira CM. Transforming Growth Factor-induced Protein Promotes NF-κB-mediated Angiogenesis during Postnatal Lung Development. Am J Respir Cell Mol Biol 2021; 64:318-330. [PMID: 33264084 PMCID: PMC7909333 DOI: 10.1165/rcmb.2020-0153oc] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 10/29/2020] [Indexed: 12/23/2022] Open
Abstract
Pulmonary angiogenesis is a key driver of alveolarization. Our prior studies showed that NF-κB promotes pulmonary angiogenesis during early alveolarization. However, the mechanisms regulating temporal-specific NF-κB activation in the pulmonary vasculature are unknown. To identify mechanisms that activate proangiogenic NF-κB signaling in the developing pulmonary vasculature, proteomic analysis of the lung secretome was performed using two-dimensional difference gel electrophoresis. NF-κB activation and angiogenic function was assessed in primary pulmonary endothelial cells (PECs) and TGFBI (transforming growth factor-β-induced protein)-regulated genes identified using RNA sequencing. Alveolarization and pulmonary angiogenesis was assessed in wild-type and Tgfbi null mice exposed to normoxia or hyperoxia. Lung TGFBI expression was determined in premature lambs supported by invasive and noninvasive respiratory support. Secreted factors from the early alveolar, but not the late alveolar or adult lung, promoted proliferation and migration in quiescent, adult PECs. Proteomic analysis identified TGFBI as one protein highly expressed by the early alveolar lung that promoted PEC migration by activating NF-κB via αvβ3 integrins. RNA sequencing identified Csf3 as a TGFBI-regulated gene that enhances nitric oxide production in PECs. Loss of TGFBI in mice exaggerated the impaired pulmonary angiogenesis induced by chronic hyperoxia, and TGFBI expression was disrupted in premature lambs with impaired alveolarization. Our studies identify TGFBI as a developmentally regulated protein that promotes NF-κB-mediated angiogenesis during early alveolarization by enhancing nitric oxide production. We speculate that dysregulation of TGFBI expression may contribute to diseases marked by impaired alveolar and vascular growth.
Collapse
Affiliation(s)
- Min Liu
- Department of Pediatrics, Center for Excellence in Pulmonary Biology, and
| | - Cristiana Iosef
- Department of Pediatrics, Center for Excellence in Pulmonary Biology, and
| | - Shailaja Rao
- Department of Pediatrics, Center for Excellence in Pulmonary Biology, and
| | | | - Sha Fu
- Department of Pediatrics, Center for Excellence in Pulmonary Biology, and
- Liuyang People’s Hospital, Hunan, China
| | - Paige Snider
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana
| | - Simon J. Conway
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana
| | - Gray S. Umbach
- Department of Pediatrics, Center for Excellence in Pulmonary Biology, and
- University of Texas Southwestern Medical School, Dallas, Texas; and
| | - Sarah C. Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, California
| | - Ruby E. Dewi
- Department of Materials Science and Engineering, Stanford University, Stanford, California
| | - Mar J. Dahl
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah
| | - Donald M. Null
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah
| | - Kurt H. Albertine
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah
| | - Cristina M. Alvira
- Department of Pediatrics, Center for Excellence in Pulmonary Biology, and
| |
Collapse
|
8
|
Yang X, Li X, Luo M, Guo Y, Li C, Lv D, Cheng Z, Huang L, Shang FF, Huang B, Shen J, Luo S, Yan J. Tubeimoside I promotes angiogenesis via activation of eNOS-VEGF signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2021; 267:113642. [PMID: 33264658 DOI: 10.1016/j.jep.2020.113642] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/19/2020] [Accepted: 11/23/2020] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tubeimoside I (TBM) is a triterpenoid saponin purified from tubeimu (tuber of Bolbostemma paniculatum (Maxim.) Franquet). In traditional Chinese medicine, tubeimu had been used to treat acute mastitis, snake bites, detoxication, inflammatory diseases, and tumors for over 1000 years. AIM OF THE STUDY This study aimed to investigate whether TBM could promote angiogenesis and how to promote angiogenesis. MATERIALS AND METHODS In vivo, the pro-angiogenic effects of TBM were examined using the hindlimb ischemia model. After the ischemia operation, 1 mg/kg/day TBM was given via intraperitoneal injection for 28 days and the recovery of blood flow was monitored by Doppler scanner every 7 days. The capillary density in gastrocnemius muscle was detected by immunofluorescence. Expression of related proteins were determined by western blotting. In vitro, the pro-angiogenic effects of TBM on HUVECs were examined by Cell Counting Kit-8, scratch assay, endothelial cell tube formation assay and western blotting. RESULTS TBM improved recovery from hindlimb ischemia in C57BL/6 mice. TBM promoted endothelial cell viability, migration and tube formation in HUVECs. TBM could activate eNOS-VEGF signaling pathway by enhancing expression of eNOS. And TBM's pro-angiogenesis effects could be abolished by L-NAME (an inhibitor of eNOS). CONCLUSIONS TBM promoted angiogenesis via the activation of eNOS-VEGF signaling pathway and TBM could be a novel agent for therapeutic angiogenesis in ischemic diseases.
Collapse
Affiliation(s)
- Xiyang Yang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China; Institute of Life Sciences, Chongqing Medical University, Chongqing, 400010, China
| | - Xingbing Li
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China; Institute of Life Sciences, Chongqing Medical University, Chongqing, 400010, China
| | - Minghao Luo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China; Institute of Life Sciences, Chongqing Medical University, Chongqing, 400010, China
| | - Yongzheng Guo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China; Institute of Life Sciences, Chongqing Medical University, Chongqing, 400010, China
| | - Chang Li
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China; Institute of Life Sciences, Chongqing Medical University, Chongqing, 400010, China
| | - Dingyi Lv
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China; Institute of Life Sciences, Chongqing Medical University, Chongqing, 400010, China
| | - Zhe Cheng
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China; Institute of Life Sciences, Chongqing Medical University, Chongqing, 400010, China
| | - Longxiang Huang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China; Institute of Life Sciences, Chongqing Medical University, Chongqing, 400010, China
| | - Fei-Fei Shang
- Institute of Life Sciences, Chongqing Medical University, Chongqing, 400010, China
| | - Bi Huang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Jian Shen
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China; Institute of Life Sciences, Chongqing Medical University, Chongqing, 400010, China
| | - Suxin Luo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China; Institute of Life Sciences, Chongqing Medical University, Chongqing, 400010, China.
| | - Jianghong Yan
- Institute of Life Sciences, Chongqing Medical University, Chongqing, 400010, China.
| |
Collapse
|
9
|
Complementary Effect of Maternal Sildenafil and Fetal Tracheal Occlusion Improves Lung Development in the Rabbit Model of Congenital Diaphragmatic Hernia. Ann Surg 2020; 275:e586-e595. [PMID: 33055583 DOI: 10.1097/sla.0000000000003943] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
OBJECTIVE To evaluate the effect of combining antenatal sildenafil with fetal tracheal occlusion (TO) in fetal rabbits with surgically induced congenital diaphragmatic hernia (CDH). BACKGROUND Although antenatal sildenafil administration rescues vascular abnormalities in lungs of fetal rabbits with CDH, it only partially improves airway morphometry. We hypothesized that we could additionally stimulate lung growth by combining this medical treatment with fetal TO. METHODS CDH was created on gestational day (GD)23 (n=54). Does were randomized to receive either sildenafil 10 mg/kg/d or placebo by subcutaneous injection from GD24 to GD30. On GD28, fetuses were randomly assigned to TO or sham neck dissection. At term (GD30) fetuses were delivered, ventilated, and finally harvested for histological and molecular analyses. Unoperated littermates served as controls. RESULTS The lung-to-body-weight ratio was significantly reduced in sham-CDH fetuses either (1.2 ± 0.3% vs 2.3 ± 0.3% in controls, P=0.0003). Sildenafil had no effect on this parameter, while CDH fetuses undergoing TO had a lung-to-body-weight ratio comparable to that of controls (2.5 ± 0.8%, P<0.0001). Sildenafil alone induced an improvement in the mean terminal bronchiolar density (2.5 ± 0.8 br/mm vs 3.5 ± 0.9 br/mm, P=0.043) and lung mechanics (static elastance 61 ± 36 cmH2O /mL vs 113 ± 40 cmH2O/mL, P=0.008), but both effects were more pronounced in fetuses undergoing additional TO (2.1 ± 0.8 br/mm, P=0.001 and 31 ± 9 cmH2O/mL, P<0.0001 respectively). Both CDH-sham and CDH-TO fetuses treated with placebo had an increased medial wall thickness of peripheral pulmonary vessels (41.9 ± 2.9% and 41.8 ± 3.2%, vs 24.0 ± 2.9% in controls, P<0.0001). CDH fetuses treated with sildenafil, either with or without TO, had a medial thickness in the normal range (29.4% ± 2.6%). Finally, TO reduced gene expression of vascular endothelial growth factor and surfactant protein A and B, but this effect was counteracted by sildenafil. CONCLUSION In the rabbit model for CDH, the combination of maternal sildenafil and TO has a complementary effect on vascular and parenchymal lung development.
Collapse
|
10
|
Russo FM, De Bie F, Hodges R, Flake A, Deprest J. Sildenafil for Antenatal Treatment of Congenital Diaphragmatic Hernia: From Bench to Bedside. Curr Pharm Des 2020; 25:601-608. [PMID: 30894101 DOI: 10.2174/1381612825666190320151856] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 03/18/2019] [Indexed: 01/19/2023]
Abstract
BACKGROUND Persistent pulmonary hypertension (PPH) is one of the main causes of mortality and morbidity in infants affected by congenital diaphragmatic hernia (CDH). Since the structural changes that lead to PPH take place already in utero, a treatment starting in the prenatal phase may prevent the occurrence of this complication. OBJECTIVE To summarize the development process of antenatal sildenafil for CDH. METHODS The pharmacokinetics and efficacy of sildenafil have been assessed in the rat and the rabbit model. The transfer of the drug through the human placenta has been measured with the ex-vivo placenta perfusion model. Results from this experiment are being incorporated in a pregnancy-physiologically based pharmacokinetic (p- PBPK) model. A phase I-IIb placental transfer and safety study is ongoing. RESULTS Sildenafil administration to pregnant rats and rabbits led to therapeutic foetal drug levels without maternal and foetal toxicity, although it was associated with impaired vascular development in foetuses with nonhypoplastic lungs. Peak concentrations and 24-hour exposure were higher in pregnant rabbits compared to nonpregnant ones. In rat and rabbit foetuses with CDH, sildenafil rescued the lung vascular anomalies and partially improved parenchymal development. Sildenafil crossed the human placenta at a high rate ex-vivo, independently from the initial maternal concentration. CONCLUSION There is preclinical evidence that maternally administered sildenafil prevents the vascular changes that lead to PPH in CDH newborns. The phase I/IIb clinical study together with the p-PBPK model will define the maternal dose needed for a therapeutic effect in the foetus. Foetal safety will be investigated both in the clinical study and in the sheep. The final step will be a multicentre, randomized, placebo-controlled trial.
Collapse
Affiliation(s)
- Francesca M Russo
- Cluster Woman and Child, Department of Development and Regeneration, Biomedical Sciences, KU Leuven, Leuven, Belgium
| | - Felix De Bie
- Cluster Woman and Child, Department of Development and Regeneration, Biomedical Sciences, KU Leuven, Leuven, Belgium
| | - Ryan Hodges
- The Ritchie Centre, Hudson Institute for Medical Research, Department of Obstetrics and Gynaecology, Monash University, Melbourne, Victoria, Australia
| | - Alan Flake
- Center for Fetal Research, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Jan Deprest
- Cluster Woman and Child, Department of Development and Regeneration, Biomedical Sciences, KU Leuven, Leuven, Belgium.,Department of Obstetrics and Gynecology, Institute of Women's Health, University College London, London, United Kingdom
| |
Collapse
|
11
|
Liu T, Mukosera GT, Blood AB. The role of gasotransmitters in neonatal physiology. Nitric Oxide 2019; 95:29-44. [PMID: 31870965 DOI: 10.1016/j.niox.2019.12.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 11/07/2019] [Accepted: 12/17/2019] [Indexed: 12/12/2022]
Abstract
The gasotransmitters, nitric oxide (NO), hydrogen sulfide (H2S), and carbon monoxide (CO), are endogenously-produced volatile molecules that perform signaling functions throughout the body. In biological tissues, these small, lipid-permeable molecules exist in free gaseous form for only seconds or less, and thus they are ideal for paracrine signaling that can be controlled rapidly by changes in their rates of production or consumption. In addition, tissue concentrations of the gasotransmitters are influenced by fluctuations in the level of O2 and reactive oxygen species (ROS). The normal transition from fetus to newborn involves a several-fold increase in tissue O2 tensions and ROS, and requires rapid morphological and functional adaptations to the extrauterine environment. This review summarizes the role of gasotransmitters as it pertains to newborn physiology. Particular focus is given to the vasculature, ventilatory, and gastrointestinal systems, each of which uniquely illustrate the function of gasotransmitters in the birth transition and newborn periods. Moreover, given the relative lack of studies on the role that gasotransmitters play in the newborn, particularly that of H2S and CO, important gaps in knowledge are highlighted throughout the review.
Collapse
Affiliation(s)
- Taiming Liu
- Department of Pediatrics, Division of Neonatology, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA
| | - George T Mukosera
- Department of Pediatrics, Division of Neonatology, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA
| | - Arlin B Blood
- Department of Pediatrics, Division of Neonatology, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA; Lawrence D. Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA.
| |
Collapse
|
12
|
Impact of Arginine Nutrition and Metabolism during Pregnancy on Offspring Outcomes. Nutrients 2019; 11:nu11071452. [PMID: 31252534 PMCID: PMC6682918 DOI: 10.3390/nu11071452] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 06/24/2019] [Accepted: 06/26/2019] [Indexed: 12/12/2022] Open
Abstract
By serving as a precursor for the synthesis of nitric oxide, polyamines, and other molecules with biological importance, arginine plays a key role in pregnancy and fetal development. Arginine supplementation is a potential therapy for treating many human diseases. An impaired arginine metabolic pathway during gestation might produce long-term morphological or functional changes in the offspring, namely, developmental programming to increase vulnerability to developing a variety of non-communicable diseases (NCDs) in later life. In contrast, reprogramming is a strategy that shifts therapeutic interventions from adulthood to early-life, in order to reverse the programming processes, which might counterbalance the rising epidemic of NCDs. This review presented the role of arginine synthesis and metabolism in pregnancy. We also provided evidence for the links between an impaired arginine metabolic pathway and the pathogenesis of compromised pregnancy and fetal programming. This was followed by reprogramming strategies targeting the arginine metabolic pathway, to prevent the developmental programming of NCDs. Despite emerging evidence from experimental studies showing that targeting the arginine metabolic pathway has promise as a reprogramming strategy in pregnancy to prevent NCDs in the offspring, these results need further clinical application.
Collapse
|
13
|
Rain M, Chaudhary H, Kukreti R, Thinlas T, Mohammad G, Pasha Q. Elevated Vasodilatory Cyclases and Shorter Telomere Length Contribute to High-Altitude Pulmonary Edema. High Alt Med Biol 2018; 19:60-68. [PMID: 29443612 DOI: 10.1089/ham.2017.0136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Rain, Manjari, Himanshi Chaudhary, Ritushree Kukreti, Tashi Thinlas, Ghulam Mohammad, and Qadar Pasha. Elevated vasodilatory cyclases and shorter telomere length contribute to high-altitude pulmonary edema. High Alt Med Biol. 19:60-68, 2018. AIM High-altitude (HA) genetics is complex with respect to health and disease (HA pulmonary edema i.e., HAPE). Based on the widely recognized fact that oxidative stress is a major trigger of several physiological processes, this study was designed to establish the significance of vasodilatory cyclases and telomere length in HA physiology. The study was performed in three groups, namely HAPE-free sojourners (HAPE-f, n = 150), HAPE patients (HAPE-p, n = 150), and healthy highland natives or highlanders (HLs, n = 150). Variations in soluble guanylyl cyclase β1-subunit (GUCY1B3) and adenylyl cyclase type 6 (ADCY6) were genotyped by the SNaPshot method and/or Fluidigm SNP type genotyping. Plasma GUCY1B3 and ADCY6 levels were estimated using ELISA, and relative telomere length was estimated by qRT-PCR. RESULTS The rs7638AA genotype was over-represented in HLs compared with HAPE-f and HAPE-p (p = 0.035 and p = 0.012, respectively). Similarly, the rs7638A allele was prevalent in HLs compared with both groups, but significance was attained against HAPE-p (p = 0.012). Significantly elevated plasma levels of GUCY1B3 and ADCY6 were obtained in HAPE-p compared with HAPE-f (p = 0.001 and p = 0.006, respectively) and HLs (p = 3.31E-05 and p = 0.05, respectively). Shorter telomere length was observed in HAPE-p compared with HAPE-f (p > 0.05) and HLs (p = 0.017). CONCLUSION Elevated cyclases and shorter telomere length associate with HAPE pathophysiology.
Collapse
Affiliation(s)
- Manjari Rain
- 1 CSIR-Institute of Genomics and Integrative Biology , Delhi, India .,2 Academy of Scientific and Innovative Research , New Delhi, India
| | | | - Ritushree Kukreti
- 1 CSIR-Institute of Genomics and Integrative Biology , Delhi, India .,2 Academy of Scientific and Innovative Research , New Delhi, India
| | - Tashi Thinlas
- 3 Department of Medicine, Sonam Norboo Memorial Hospital , Leh, Ladakh, Jammu and Kashmir, India
| | - Ghulam Mohammad
- 3 Department of Medicine, Sonam Norboo Memorial Hospital , Leh, Ladakh, Jammu and Kashmir, India
| | - Qadar Pasha
- 1 CSIR-Institute of Genomics and Integrative Biology , Delhi, India .,4 Indian Council of Medical Research , New Delhi, India
| |
Collapse
|
14
|
Zhang XN, Ma ZJ, Wang Y, Sun B, Guo X, Pan CQ, Chen LM. Angelica Dahurica ethanolic extract improves impaired wound healing by activating angiogenesis in diabetes. PLoS One 2017; 12:e0177862. [PMID: 28542422 PMCID: PMC5443501 DOI: 10.1371/journal.pone.0177862] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 05/04/2017] [Indexed: 12/26/2022] Open
Abstract
Abnormal angiogenesis plays an important role in impaired wound healing and development of chronic wounds in diabetes mellitus. Angelica dahurica radix is a common traditional Chinese medicine with wide spectrum medicinal effects. In this study, we analyzed the potential roles of Angelica dahurica ethanolic extract (ADEE) in correcting impaired angiogenesis and delayed wound healing in diabetes by using streptozotocin-induced diabetic rats. ADEE treatment accelerated diabetic wound healing through inducing angiogenesis and granulation tissue formation. The angiogenic property of ADEE was subsequently verified ex vivo using aortic ring assays. Furthermore, we investigated the in vitro angiogenic activity of ADEE and its underlying mechanisms using human umbilical vein endothelial cells. ADEE treatment induced HUVECs proliferation, migration, and tube formation, which are typical phenomena of angiogenesis, in dose-dependent manners. These effects were associated with activation of angiogenic signal modulators, including extracellular signal-regulated kinase 1/2 (ERK1/2), Akt, endothelial nitric oxide synthase (eNOS) as well as increased NO production, and independent of affecting VEGF expression. ADEE-induced angiogenic events were inhibited by the MEK inhibitor PD98059, the PI3K inhibitor Wortmannin, and the eNOS inhibitor L-NAME. Our findings highlight an angiogenic role of ADEE and its ability to protect against impaired wound healing, which may be developed as a promising therapy for impaired angiogenesis and delayed wound healing in diabetes.
Collapse
Affiliation(s)
- Xiao-Na Zhang
- From the Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Ze-Jun Ma
- From 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Key Laboratory of Hormone and Development, Ministry of Health, Metabolic Disease Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, People's Republic of China
| | - Ying Wang
- From 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Key Laboratory of Hormone and Development, Ministry of Health, Metabolic Disease Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, People's Republic of China
| | - Bei Sun
- From 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Key Laboratory of Hormone and Development, Ministry of Health, Metabolic Disease Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, People's Republic of China
| | - Xin Guo
- From 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Key Laboratory of Hormone and Development, Ministry of Health, Metabolic Disease Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, People's Republic of China
| | - Cong-Qing Pan
- From 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Key Laboratory of Hormone and Development, Ministry of Health, Metabolic Disease Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, People's Republic of China
| | - Li-Ming Chen
- From 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Key Laboratory of Hormone and Development, Ministry of Health, Metabolic Disease Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, People's Republic of China
| |
Collapse
|
15
|
Treatment with Sildenafil and Donepezil Improves Angiogenesis in Experimentally Induced Critical Limb Ischemia. BIOMED RESEARCH INTERNATIONAL 2017; 2017:9532381. [PMID: 28243607 PMCID: PMC5294352 DOI: 10.1155/2017/9532381] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 11/13/2016] [Accepted: 11/23/2016] [Indexed: 12/23/2022]
Abstract
Objectives. In this study, we aimed to demonstrate the role of sildenafil (an antagonist of phosphodiesterase type 5 (PDE-5)) and donepezil (a specific and reversible inhibitor of acetylcholinesterase (Ach)) in increasing ischemia-induced angiogenesis. Method. Critical limb ischemia was induced by ligation of the common femoral artery followed by ligation of the common iliac artery. The operated animals were divided into 3 groups: receiving sildenafil, receiving donepezil, and surgery alone; the contralateral lower limb was used as a negative control. The results were controlled based on clinical score and Doppler ultrasound. Gastrocnemius muscle samples were taken from all animals, both from the ischemic and nonischemic limb and were used for histopathological and immunohistochemical examination for the evaluation of the number of nuclei/field, endothelial cells (CD31), dividing cells (Ki-67), and vascular endothelial growth factor (VEGFR-3). Results. An increasing tendency of the number of nuclei/field with time was observed both in the case of sildenafil and donepezil treatment. The formation of new capillaries (the angiogenesis process) was more strongly influenced by donepezil treatment compared to sildenafil or no treatment. This treatment significantly influenced the capillary/fiber ratio, which was increased compared to untreated ligated animals. Sildenafil treatment led to a gradual increase in the number of dividing cells, which was significantly compared to the negative control group and compared to the ligation control group. The same effect (increase in the number of Ki-67 positive cells) was more obvious in the case of donepezil treatment. Conclusion. Donepezil treatment has a better effect in ligation-induced ischemia compared to sildenafil, promoting angiogenesis in the first place, and also arteriogenesis.
Collapse
|
16
|
Chongsatientam A, Yimlamai T. Therapeutic Pulsed Ultrasound Promotes Revascularization and Functional Recovery of Rat Skeletal Muscle after Contusion Injury. ULTRASOUND IN MEDICINE & BIOLOGY 2016; 42:2938-2949. [PMID: 27665217 DOI: 10.1016/j.ultrasmedbio.2016.08.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 07/26/2016] [Accepted: 08/02/2016] [Indexed: 06/06/2023]
Abstract
The mechanism by which therapeutic pulsed ultrasound (TPU) promotes the repair of damaged gastrocnemius muscle was investigated. Male Wistar rats were divided into uninjured, sham-treated injured and TPU-treated injured (TPU) groups. Injury was induced by mass-drop technique. TPU was applied to the injured muscle for 5 min, daily, started at day 1 post-injury and continuing for 3, 7 and 14 d. For 3 d post-injury, a significant reduction in muscle force was observed in both the sham-treated injured and TPU groups. TPU treatment significantly increased recovery force of the injured muscle after day 7 post-injury. This effect of TPU is associated with increased centronucleated fibers and cross-sectional area, mRNA expression of the vascular endothelial growth factor and capillary density of the regenerated fibers, but not with mRNA expression of nitric oxide synthase. We conclude that TPU hastens muscle recovery, at least in part, by upregulating angiogenesis.
Collapse
Affiliation(s)
| | - Tossaporn Yimlamai
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
17
|
Li Y, Li L, Li Z, Sheng J, Zhang X, Feng D, Zhang X, Yin F, Wang A, Wang F. Tat PTD-Endostatin-RGD: A novel protein with anti-angiogenesis effect in retina via eye drops. Biochim Biophys Acta Gen Subj 2016; 1860:2137-47. [PMID: 27233450 DOI: 10.1016/j.bbagen.2016.05.031] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 05/16/2016] [Accepted: 05/21/2016] [Indexed: 01/04/2023]
Abstract
BACKGROUND Diabetic retinopathy is a leading cause of blindness. The objective was to design a novel fusion protein, Tat PTD-Endostatin-RGD, to treat retinal neovascularization via eye drops instead of traditional intravitreal injection trepapeutical methods. METHOD The anti-angiogenesis ability was evaluated in vitro by chick embryo chorioallantoic membrane assay, wound healing assay and tube formation assay. Corneal barrier and blood-retina barrier were constructed in vitro to investigate the penetration ability of Tat PTD-Endostatin-RGD. Western blot was used to detect the integrin αvβ3 expression level in rat retina microvascular endothelial cells which was stimulated by S-nitroso-N-acetylpenicillamine. The binding affinity of Tat PTD-Endostatin-RGD to integrin αvβ3 was investigated by evaluating the penetration ability on blood-retina barriers treated with S-nitroso-N-acetylpenicillamine. The pharmacodynamics and efficacy analysis were further carried out in the oxygen-induced retinopathy model in vivo. In addition, the pharmacokinetic profile via eye drops was studied on a C57BL/6 mice model. RESULT Tat PTD-Endostatin-RGD showed high anti-angiogenesis activity and high ability to penetrate these two barriers in vitro. The Western blot results indicated S-nitroso-N-acetylpenicillamine upregulated the expression level of integrin αvβ3 in a dose-dependent manner. Tat PTD-Endostatin-RGD showed a high affinity to rat retina microvascular endothelial cells treated with S-nitroso-N-acetylpenicillamine. The results showed that Tat PTD-Endostatin-RGD could inhibit abnormal angiogenesis in retina via eye drops. CONCLUSION Tat PTD-Endostatin-RGD showed high penetration ability through ocular barriers, bound specifically to integrin αvβ3 and effectively inhibited the abnormal angiogenesis. GENERAL SIGNIFICANCE Tat PTD-Endostatin-RGD represents a potent novel drug applied via eye drops for fundus oculi neovascularization diseases.
Collapse
Affiliation(s)
- Yan Li
- Key Laboratory of Chemical Biology of Natural Products (Ministry of Education), Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Lian Li
- Key Laboratory of Chemical Biology of Natural Products (Ministry of Education), Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Zhiwei Li
- Department of Ophthalmology, Provincial Hospital Affiliated to Shandong University, Jinan 250012, China
| | - Juzheng Sheng
- Key Laboratory of Chemical Biology of Natural Products (Ministry of Education), Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Xinke Zhang
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Danyang Feng
- Key Laboratory of Chemical Biology of Natural Products (Ministry of Education), Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Xu Zhang
- Key Laboratory of Chemical Biology of Natural Products (Ministry of Education), Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Fengxin Yin
- Key Laboratory of Chemical Biology of Natural Products (Ministry of Education), Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Aijun Wang
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Fengshan Wang
- Key Laboratory of Chemical Biology of Natural Products (Ministry of Education), Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China; National Glycoengineering Research Center, Shandong University, Jinan 250012, China.
| |
Collapse
|
18
|
Russo FM, Toelen J, Eastwood MP, Jimenez J, Miyague AH, Vande Velde G, DeKoninck P, Himmelreich U, Vergani P, Allegaert K, Deprest J. Transplacental sildenafil rescues lung abnormalities in the rabbit model of diaphragmatic hernia. Thorax 2016; 71:517-25. [PMID: 26987998 DOI: 10.1136/thoraxjnl-2015-207949] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 02/20/2016] [Indexed: 12/20/2022]
Abstract
INTRODUCTION The management of congenital diaphragmatic hernia (DH) would benefit from an antenatal medical therapy, which addresses both lung hypoplasia and persistent pulmonary hypertension. We aimed at evaluating the pulmonary effects of sildenafil in the fetal rabbit model for DH. METHODS We performed a dose-finding study to achieve therapeutic fetal plasmatic concentrations without toxicity following maternal sildenafil administration. Subsequently, DH fetuses were randomly exposed to transplacental placebo or sildenafil 10 mg/kg/day from gestational day 24 until examination at term (day 30). Efficacy measures were ipsilateral pulmonary vascular and airway morphometry, micro-CT-based branching analysis, Doppler flow in the main pulmonary artery and postnatal lung mechanics. RESULTS Fetal sildenafil plasmatic concentration was above the minimal therapeutic level for at least 22 h/day without maternal and fetal side effects. The placebo-exposed DH fetuses had increased wall thickness in peripheral pulmonary vessels and significantly less fifth-order vessels compared with controls (CTR). Sildenafil-exposed DH fetuses, instead, had a medial and adventitial thickness in peripheral pulmonary vessels in the normal range and normal vascular branching. Fetal pulmonary artery Doppler showed a reduction of pulmonary vascular resistances both in DH and in CTR fetuses treated by sildenafil compared with the placebo-treated ones. Sildenafil also reversed the mean terminal bronchiolar density to normal and improved lung mechanics, yet without measurable impact on lung-to-bodyweight ratio. CONCLUSIONS In the rabbit model for DH, antenatal sildenafil rescues vascular branching and architecture, reduces pulmonary vascular resistances and also improves airway morphometry and respiratory mechanics.
Collapse
Affiliation(s)
- Francesca M Russo
- Cluster Organ Systems, Department of Development and Regeneration, Biomedical Sciences, KU Leuven, Leuven, Belgium Department of Obstetrics and Gynecology, University Hospitals Leuven, Leuven, Belgium
| | - Jaan Toelen
- Cluster Organ Systems, Department of Development and Regeneration, Biomedical Sciences, KU Leuven, Leuven, Belgium Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
| | - M Patrice Eastwood
- Cluster Organ Systems, Department of Development and Regeneration, Biomedical Sciences, KU Leuven, Leuven, Belgium
| | - Julio Jimenez
- Cluster Organ Systems, Department of Development and Regeneration, Biomedical Sciences, KU Leuven, Leuven, Belgium Department of Obstetrics and Gynecology, Clinica Alemana, Santiago, Chile
| | - Andre Hadyme Miyague
- Cluster Organ Systems, Department of Development and Regeneration, Biomedical Sciences, KU Leuven, Leuven, Belgium
| | - Greetje Vande Velde
- Department of Imaging and Pathology, Biomedical MRI/MoSAIC, KU Leuven, Leuven, Belgium
| | - Philip DeKoninck
- Cluster Organ Systems, Department of Development and Regeneration, Biomedical Sciences, KU Leuven, Leuven, Belgium
| | - Uwe Himmelreich
- Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
| | - Patrizia Vergani
- Department of Obstetrics and Gynecology, University of Milano-Bicocca, Monza, Italy
| | - Karel Allegaert
- Cluster Organ Systems, Department of Development and Regeneration, Biomedical Sciences, KU Leuven, Leuven, Belgium Department of Neonatalogy, University Hospitals Leuven, Leuven, Belgium
| | - Jan Deprest
- Cluster Organ Systems, Department of Development and Regeneration, Biomedical Sciences, KU Leuven, Leuven, Belgium Department of Obstetrics and Gynecology, University Hospitals Leuven, Leuven, Belgium Department of Obstetrics and Gynecology, Institute of Women's Health, University College London, London, UK
| |
Collapse
|
19
|
Pathological Left Ventricular Hypertrophy and Stem Cells: Current Evidence and New Perspectives. Stem Cells Int 2015; 2016:5720758. [PMID: 26798360 PMCID: PMC4699040 DOI: 10.1155/2016/5720758] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Revised: 08/17/2015] [Accepted: 09/06/2015] [Indexed: 12/17/2022] Open
Abstract
Left ventricular hypertrophy (LVH) is a strong predictor of adverse cardiovascular outcomes. It is the result of complex mechanisms that include not only an increase in protein synthesis and cell size but also proliferating cardiac progenitor cells and the influx of bone marrow-derived cells developing into cardiomyocytes. Stem and progenitor cells are known to contribute to the renewal of adult mammalian cardiomyocytes in case of myocardial injury or pressure and volume overload. They are activated in LVH and play a regulatory role in myocardial repair. They have high proliferative potential and secrete numerous cytokines, growth factors, and microRNAs that play important roles in cell differentiation, cardiac remodeling, and neovascularization. They are mobilized in response to either mechanical or chemical stimuli, hormones, or pharmacologic agents. Another important source of progenitor cells is the epicardial layer. It appears that precursor cells migrate from the epicardium to the myocardium in order to interact with myocardial cells. In addition, migratory cells participate in the formation of almost all cardiac structures in myocardial hypertrophy. Although the pathophysiological mechanisms are still obscure and further studies are required, their properties may open the door to regenerative cell therapy for the prevention of adverse remodeling.
Collapse
|
20
|
Hutchinson TE, Patel JM. Peptide-stimulated angiogenesis: Role of lung endothelial caveolar signaling and nitric oxide. Nitric Oxide 2015; 51:43-51. [PMID: 26537637 DOI: 10.1016/j.niox.2015.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 09/29/2015] [Accepted: 10/26/2015] [Indexed: 10/22/2022]
Abstract
Endothelial nitric oxide (NO) synthase (eNOS)-derived NO plays a critical role in the modulation of angiogenesis in the pulmonary vasculature. We recently reported that an eleven amino acid (SSWRRKRKESS) cell penetrating synthetic peptide (P1) activates caveolar signaling, caveloae/eNOS dissociation, and enhance NO production in lung endothelial cells (EC). This study examines whether P1 promote angiogenesis via modulation of caveolar signaling and the level of NO generation in EC and pulmonary artery (PA) segments. P1-enhanced tube formation and cell sprouting were abolished by caveolae disruptor Filipin (FIL) in EC and PA, respectively. P1 enhanced eNOS activity and angiogenesis were attenuated by inhibition of eNOS as well as PLCγ-1, PKC-α but not PI3K-mediated caveolar signaling in intact EC and/or PA. P1 failed to enhance the catalytic activity of eNOS and angiogenesis in caveolae disrupted EC by FIL. Lower (0.01 mM) concentration of NOC-18 enhanced angiogenesis without inhibition of eNOS activity whereas higher concentration of NOC-18 (1.0 mM) inhibited eNOS activity and angiogenesis in EC. Inhibition of eNOS by l-NAME in the presence of P1 resulted in near total loss of tube formation in EC. Although P1 enhanced angiogenesis mimicked only by lower concentrations of NO generated by NOC-18, this response is independent of caveolar signaling/integrity. These results suggest that P1-enhanced angiogenesis is regulated by dynamic process involving caveolar signaling-mediated increased eNOS/NO activity or by the direct exposure to NOC-18 generating only physiologic range of NO independent of caveolae in lung EC and PA segments.
Collapse
Affiliation(s)
- Tarun E Hutchinson
- Department of Medicine, University of Florida College of Medicine, Gainesville, FL 32608-1197, USA
| | - Jawaharlal M Patel
- Department of Medicine, University of Florida College of Medicine, Gainesville, FL 32608-1197, USA; Research Service, North Florida/South Georgia Veterans Health System, Gainesville, FL 32608-1197, USA.
| |
Collapse
|
21
|
Campbell KA, Li X, Biendarra SM, Terzic A, Nelson TJ. Nos3-/- iPSCs model concordant signatures of in utero cardiac pathogenesis. J Mol Cell Cardiol 2015; 87:228-36. [PMID: 26344701 DOI: 10.1016/j.yjmcc.2015.08.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 08/28/2015] [Accepted: 08/29/2015] [Indexed: 01/17/2023]
Abstract
BACKGROUND Through genome-wide transcriptional comparisons, this study interrogates the capacity of in vitro differentiation of induced pluripotent stem cells (iPSCs) to accurately model pathogenic signatures of developmental cardiac defects. METHODS AND RESULTS Herein, we studied the molecular etiology of cardiac defects in Nos3(-/-) mice via transcriptional analysis of stage-matched embryonic tissues and iPSC-derived cells. In vitro comparisons of differentiated cells were calibrated to in utero benchmarks of health and disease. Integrated systems biology analysis of WT and Nos3(-/-) transcriptional profiles revealed 50% concordant expression patterns between in utero embryonic tissues and ex vivo iPSC-derived cells. In particular, up-regulation of glucose metabolism (p-value=3.95×10(-12)) and down-regulation of fatty acid metabolism (p-value=6.71×10(-12)) highlight a bioenergetic signature of early Nos3 deficiency during cardiogenesis that can be recapitulated in iPSC-derived differentiated cells. CONCLUSIONS The in vitro concordance of early Nos3(-/-) disease signatures supports the utility of iPSCs as a cellular model of developmental heart defects. Moreover, this study supports the use of iPSCs as a platform to pinpoint initial stages of congenital cardiac pathogenesis.
Collapse
Affiliation(s)
- Katherine A Campbell
- Department of Molecular Pharmacology and Experimental Therapeutics, 200 First Street SW, Mayo Clinic, Rochester, MN 55905, USA; Center for Regenerative Medicine, 200 First Street SW, Mayo Clinic, Rochester, MN 55905, USA; Division of General Internal Medicine, 200 First Street SW, Mayo Clinic, Rochester, MN 55905, USA
| | - Xing Li
- Department of Health Sciences Research, 200 First Street SW, Mayo Clinic, Rochester, MN 55905, USA; Division of Biomedical Statistics and Informatics, 200 First Street SW, Mayo Clinic, Rochester, MN 55905, USA
| | - Sherri M Biendarra
- Department of Molecular Pharmacology and Experimental Therapeutics, 200 First Street SW, Mayo Clinic, Rochester, MN 55905, USA
| | - Andre Terzic
- Department of Molecular Pharmacology and Experimental Therapeutics, 200 First Street SW, Mayo Clinic, Rochester, MN 55905, USA; Center for Regenerative Medicine, 200 First Street SW, Mayo Clinic, Rochester, MN 55905, USA; Division of Cardiovascular Diseases, 200 First Street SW, Mayo Clinic, Rochester, MN 55905, USA; Department of Medical Genetics, 200 First Street SW, Mayo Clinic, Rochester, MN 55905, USA
| | - Timothy J Nelson
- Department of Molecular Pharmacology and Experimental Therapeutics, 200 First Street SW, Mayo Clinic, Rochester, MN 55905, USA; Center for Regenerative Medicine, 200 First Street SW, Mayo Clinic, Rochester, MN 55905, USA; Division of Cardiovascular Diseases, 200 First Street SW, Mayo Clinic, Rochester, MN 55905, USA; Division of General Internal Medicine, 200 First Street SW, Mayo Clinic, Rochester, MN 55905, USA; Center for Transplantation and Clinical Regeneration, 200 First Street SW, Mayo Clinic, Rochester, MN 55905, USA; Division of Pediatric Cardiology,200 First Street SW, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
22
|
Bassareo PP, Mussap M, Bassareo V, Flore G, Mercuro G. Nitrergic system and plasmatic methylarginines: Evidence of their role in the perinatal programming of cardiovascular diseases. Clin Chim Acta 2015; 451:21-7. [PMID: 26004093 DOI: 10.1016/j.cca.2015.05.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 05/17/2015] [Indexed: 01/28/2023]
Abstract
Atherosclerosis, in turn preceded by endothelial dysfunction, underlies a series of important cardiovascular diseases. Reduced bioavailability of endothelial nitric oxide, by increasing vascular tone and promoting platelet aggregation, leukocyte adhesion, and smooth muscle cell proliferation, plays a key role in the onset of the majority of cardiovascular diseases. In addition, high blood levels of asymmetric dimethylarginine, a potent inhibitor of nitric oxide synthesis, are associated with future development of adverse cardiovascular events and cardiac death. Recent reports have demonstrated that another methylarginine, i.e., symmetric dimethylarginine, is also involved in the onset of endothelial dysfunction and hypertension. Almost a decade ago, prematurity at birth and intrauterine growth retardation were first associated with a potential negative influence on the cardiovascular apparatus, thus constituting risk factors or leading to early onset of cardiovascular diseases. This condition is referred to as cardiovascular perinatal programming. Accordingly, cardiovascular morbidity and mortality are higher among former preterm adults than in those born at term. The aim of this paper was to undertake a comprehensive literature review focusing on cellular and biochemical mechanisms resulting in both reduced nitric oxide bioavailability and increased methylarginine levels in subjects born preterm. Evidence of the involvement of these compounds in the perinatal programming of cardiovascular risk are also discussed.
Collapse
Affiliation(s)
- Pier Paolo Bassareo
- Department of Cardiovascular and Neurological Sciences, University of Cagliari, Cagliari, Italy.
| | - Michele Mussap
- Department of Laboratory Medicine, IRCCS San Martino-IST, University Hospital, National Institute for Cancer Research, Genova, Italy
| | - Valentina Bassareo
- Department of Toxicology, CNR Institute of Neuroscience, Section of Cagliari, National Institute of Neuroscience, University of Cagliari, Cagliari, Italy
| | - Giovanna Flore
- Department of Cardiovascular and Neurological Sciences, University of Cagliari, Cagliari, Italy
| | - Giuseppe Mercuro
- Department of Cardiovascular and Neurological Sciences, University of Cagliari, Cagliari, Italy
| |
Collapse
|
23
|
Popov TM, Goranova T, Stancheva G, Kaneva R, Dikov T, Chalakov I, Rangachev J, Konov D, Todorov S, Stoyanov O, Mitev V. Relative quantitative expression of hypoxia-inducible factor-1α, -2α and -3α, and vascular endothelial growth factor A in laryngeal carcinoma. Oncol Lett 2015; 9:2879-2885. [PMID: 26137164 DOI: 10.3892/ol.2015.3070] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 03/05/2015] [Indexed: 01/07/2023] Open
Abstract
The aim of the present study was to determine the relative quantitative expression of hypoxia-inducible factor (HIF)-1α, -2α and -3α, and VEGF-A in laryngeal carcinoma. A total of 63 patients with carcinoma of the larynx were enrolled in the study. Total RNA was isolated from fresh, frozen normal and tumor tissues of each patient, and quantitative polymerase chain reaction was performed. HIF-1α was upregulated in the majority of patients (44 patients; 69.84%). By contrast, only 7 (11.11%) patients from the whole group displayed HIF-2α overexpression, while the HIF-3α isoform was silenced in the majority of patients (48 patients, 76.19%). A small group of 5 (7.94%) patients exhibited significant overexpression of the HIF-3α isoform. VEGF-A expression was significantly higher (P<0.05) in patients with upregulated HIF-1α (2.72±1.41 RQ) compared with patients without upregulated HIF-1α (1.86±1.46 RQ). There was a moderate positive correlation between mRNA expression levels of HIF-1α and VEGF-A (rs=0.392; P<0.005). To the best of our knowledge, this study is first to report quantitative data with regard to the expression of all three HIF isoforms in malignant neoplasms. The findings suggest the existence of specific phenotypes of HIF expression in laryngeal carcinoma, where the HIF switch is absent.
Collapse
Affiliation(s)
| | - Teodora Goranova
- Department of Medical Chemistry and Biochemistry and Molecular Medicine Center, Medical University of Sofia, Sofia 1680, Bulgaria
| | - Gergana Stancheva
- Department of Medical Chemistry and Biochemistry and Molecular Medicine Center, Medical University of Sofia, Sofia 1680, Bulgaria
| | - Radka Kaneva
- Department of Medical Chemistry and Biochemistry and Molecular Medicine Center, Medical University of Sofia, Sofia 1680, Bulgaria
| | - Tihomir Dikov
- Department of Pathology, Medical University of Sofia, Sofia 1680, Bulgaria
| | - Ivan Chalakov
- Department of Ear, Nose and Throat, Medical University of Sofia, Sofia 1680, Bulgaria
| | - Julian Rangachev
- Department of Ear, Nose and Throat, Medical University of Sofia, Sofia 1680, Bulgaria
| | - Dimitar Konov
- Department of Ear, Nose and Throat, Medical University of Sofia, Sofia 1680, Bulgaria
| | - Spiridon Todorov
- Department of Ear, Nose and Throat, Medical University of Sofia, Sofia 1680, Bulgaria
| | - Orlin Stoyanov
- Department of Ear, Nose and Throat, Medical University of Sofia, Sofia 1680, Bulgaria
| | - Vanio Mitev
- Department of Medical Chemistry and Biochemistry and Molecular Medicine Center, Medical University of Sofia, Sofia 1680, Bulgaria
| |
Collapse
|
24
|
Involvement of activation of C-met signaling pathway in CD151-induced HUVECs angiogenesis. ACTA ACUST UNITED AC 2015; 35:35-41. [PMID: 25673190 DOI: 10.1007/s11596-015-1385-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Revised: 10/27/2014] [Indexed: 12/16/2022]
Abstract
CD151 is a member of the tetraspanin family that is implicated as a promoter of pathological or physiological angiogenesis. C-Met is expressed on a variety of cells including vascular endothelial cells (VECs) and up-regulated during angiogenesis. In this study, we investigated whether CD151 regulated migration, proliferation, tube formation and angiogenesis of human umbilical VECs (HUVECs) with activation of C-Met. Moreover, we studied whether CD151 could affect the angiogenic molecules such as nitric oxide (NO), vascular cell adhesion molecule-1 (VCAM-1) and vascular endothelial growth factor (VEGF). The expression of CD151 was determined by Western blotting. The cell proliferation assay was performed using the cell counting kit-8 (CCK-8) method and cell migration was assessed in microchemotaxis chambers by using fetal bovine serum (FBS) as the chemotactic stimulus. The angiogenic molecules were evaluated using ELISA. The NO level was detected using NO detection kit. The potential involvement of various signaling pathways was explored using relevant antibodies. We found that proliferation, migration and tube formation of HUVECs were promoted by CD151 with activation of C-Met, FAK and CDC42, while they were suppressed with CD151 knockdown by RNAi. Similarly, the levels of NO, VCAM-1 and VEGF in HUVECs were increased by CD151, but they were inhibited with CD151 knockdown by RNAi. These data suggested that CD151 could promote migration, proliferation, tube formation and angiogenesis of HUVECs, which was possibly related to the C-Met signaling pathways.
Collapse
|
25
|
Faulkner A, Purcell R, Hibbert A, Latham S, Thomson S, Hall WL, Wheeler-Jones C, Bishop-Bailey D. A thin layer angiogenesis assay: a modified basement matrix assay for assessment of endothelial cell differentiation. BMC Cell Biol 2014; 15:41. [PMID: 25476021 PMCID: PMC4263020 DOI: 10.1186/s12860-014-0041-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 10/30/2014] [Indexed: 12/28/2022] Open
Abstract
Background Basement matrices such as Matrigel™ and Geltrex™ are used in a variety of cell culture assays of anchorage-dependent differentiation including endothelial cell tube formation assays. The volumes of matrix recommended for these assays (approximately 150 μl/cm2) are costly, limit working distances for microscopy, and require cell detachment for subsequent molecular analysis. Here we describe the development and validation of a thin-layer angiogenesis (TLA) assay for assessing the angiogenic potential of endothelial cells that overcomes these limitations. Results Geltrex™ basement matrix at 5 μl/cm2 in 24-well (10 μl) or 96-well (2 μl) plates supports endothelial cell differentiation into tube-like structures in a comparable manner to the standard larger volumes of matrix. Since working distances are reduced, high-resolution single cell microscopy, including DIC and confocal imaging, can be used readily. Using MitoTracker dye we now demonstrate, for the first time, live mitochondrial dynamics and visualise the 3-dimensional network of mitochondria present in differentiated endothelial cells. Using a standard commercial total RNA extraction kit (Qiagen) we also show direct RNA extraction and RT-qPCR from differentiated endothelial cells without the need to initially detach cells from their supporting matrix. Conclusions We present here a new thin-layer assay (TLA) for measuring the anchorage-dependent differentiation of endothelial cells into tube-like structures which retains all the characteristics of the traditional approach but with the added benefit of a greatly lowered cost and better compatibility with other techniques, including RT-qPCR and high-resolution microscopy. Electronic supplementary material The online version of this article (doi:10.1186/s12860-014-0041-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ashton Faulkner
- Comparative Biomedical Sciences, Royal Veterinary College, University of London Royal College Street, London, NW1 0TU, UK.
| | - Robert Purcell
- Comparative Biomedical Sciences, Royal Veterinary College, University of London Royal College Street, London, NW1 0TU, UK.
| | - Andrew Hibbert
- Comparative Biomedical Sciences, Royal Veterinary College, University of London Royal College Street, London, NW1 0TU, UK.
| | - Sally Latham
- Comparative Biomedical Sciences, Royal Veterinary College, University of London Royal College Street, London, NW1 0TU, UK.
| | - Scott Thomson
- Comparative Biomedical Sciences, Royal Veterinary College, University of London Royal College Street, London, NW1 0TU, UK.
| | - Wendy L Hall
- Diabetes and Nutritional Sciences Division, School of Medicine, King's College London, 150 Stamford Street, London, SE1 9NH, UK.
| | - Caroline Wheeler-Jones
- Comparative Biomedical Sciences, Royal Veterinary College, University of London Royal College Street, London, NW1 0TU, UK.
| | - David Bishop-Bailey
- Comparative Biomedical Sciences, Royal Veterinary College, University of London Royal College Street, London, NW1 0TU, UK.
| |
Collapse
|
26
|
Chun JN, Cho M, So I, Jeon JH. The protective effects of Schisandra chinensis fruit extract and its lignans against cardiovascular disease: A review of the molecular mechanisms. Fitoterapia 2014; 97:224-33. [DOI: 10.1016/j.fitote.2014.06.014] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 06/19/2014] [Accepted: 06/20/2014] [Indexed: 02/06/2023]
|
27
|
Wang X, Meng H, Chen P, Yang N, Lu X, Wang ZM, Gao W, Zhou N, Zhang M, Xu Z, Chen B, Tao Z, Wang L, Yang Z, Zhu T. Beneficial effects of muscone on cardiac remodeling in a mouse model of myocardial infarction. Int J Mol Med 2014; 34:103-11. [PMID: 24807380 PMCID: PMC4072338 DOI: 10.3892/ijmm.2014.1766] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 04/23/2014] [Indexed: 11/06/2022] Open
Abstract
Musk has been traditionally used in East Asia to alleviate the symptoms of angina pectoris. However, it remains unclear as to whether muscone, the main active ingredient of musk, has any beneficial effects on persistent myocardial ischemia in vivo. The aim of the present study was to investigate whether muscone can improve cardiac function and attenuate myocardial remodeling following myocardial infarction (MI) in mice. Mice were subjected to permanent ligation of the left anterior descending coronary artery to induce MI, and then randomly treated with muscone (2 mg/kg/day) or the vehicle (normal saline) for 3 weeks. Sham-operated mice were used as controls and were also administered the vehicle (normal saline). Treatment with muscone significantly improved cardiac function and exercise tolerance, as evidenced by the decrease in the left ventricular end-systolic diameter, left ventricular end-diastolic diameter, as well as an increase in the left ventricular ejection fraction, left ventricular fractional shortening and time to exhaustion during swimming. Pathological and morphological assessments indicated that treatment with muscone alleviated myocardial fibrosis, collagen deposition and improved the heart weight/body weight ratio. Muscone inhibited the inflammatory response by reducing the expression of transforming growth factor (TGF)‑β1, tumor necrosis factor (TNF)-α, interleukin (IL)-1β and nuclear factor (NF)-κB. Treatment with muscone also reduced myocardial apoptosis by enhancing Bcl-2 and suppressing Bax expression. Muscone also induced the phosphorylation of protein kinase B (Akt) and endothelial nitric oxide synthase (eNOS). Our results demonstrate that muscone ameliorates cardiac remodeling and dysfunction induced by MI by exerting anti-fibrotic, anti-inflammatory and anti-apoptotic effects in the ischemic myocardium.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Haoyu Meng
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Pengsheng Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Naiquan Yang
- Department of Cardiology, Huai'an Second People's Hospital Affiliated to Xuzhou Medical College, Huai'an, Jiangsu 223002, P.R. China
| | - Xin Lu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Ze-Mu Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Wei Gao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Ningtian Zhou
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Min Zhang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Zhihui Xu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Bo Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Zhengxian Tao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Liangsheng Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Zhijian Yang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Tiebin Zhu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
28
|
Hale AT, Tian H, Anih E, Recio FO, Shatat MA, Johnson T, Liao X, Ramirez-Bergeron DL, Proweller A, Ishikawa M, Hamik A. Endothelial Kruppel-like factor 4 regulates angiogenesis and the Notch signaling pathway. J Biol Chem 2014; 289:12016-12028. [PMID: 24599951 PMCID: PMC4002108 DOI: 10.1074/jbc.m113.530956] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 02/18/2014] [Indexed: 01/08/2023] Open
Abstract
Regulation of endothelial cell biology by the Notch signaling pathway (Notch) is essential to vascular development, homeostasis, and sprouting angiogenesis. Although Notch determines cell fate and differentiation in a wide variety of cells, the molecular basis of upstream regulation of Notch remains poorly understood. Our group and others have implicated the Krüppel-like factor family of transcription factors as critical regulators of endothelial function. Here, we show that Krüppel-like factor 4 (KLF4) is a central regulator of sprouting angiogenesis via regulating Notch. Using a murine model in which KLF4 is overexpressed exclusively in the endothelium, we found that sustained expression of KLF4 promotes ineffective angiogenesis leading to diminished tumor growth independent of endothelial cell proliferation or cell cycling effects. These tumors feature increased vessel density yet are hypoperfused, leading to tumor hypoxia. Mechanistically, we show that KLF4 differentially regulates expression of Notch receptors, ligands, and target genes. We also demonstrate that KLF4 limits cleavage-mediated activation of Notch1. Finally, we rescue Notch target gene expression and the KLF4 sprouting angiogenesis phenotype by supplementation of DLL4 recombinant protein. Identification of this hitherto undiscovered role of KLF4 implicates this transcription factor as a critical regulator of Notch, tumor angiogenesis, and sprouting angiogenesis.
Collapse
Affiliation(s)
- Andrew T Hale
- Case Cardiovascular Research Institute and Harrington Heart & Vascular Institute, University Hospitals Case Medical Center, Cleveland, Ohio 44106; Department of Medicine, Case Western Reserve University School of Medicine and University Hospitals Case Medical Center, Cleveland, Ohio 44106
| | - Hongmei Tian
- Case Cardiovascular Research Institute and Harrington Heart & Vascular Institute, University Hospitals Case Medical Center, Cleveland, Ohio 44106; Department of Medicine, Case Western Reserve University School of Medicine and University Hospitals Case Medical Center, Cleveland, Ohio 44106
| | - Ejike Anih
- Case Cardiovascular Research Institute and Harrington Heart & Vascular Institute, University Hospitals Case Medical Center, Cleveland, Ohio 44106; Department of Medicine, Case Western Reserve University School of Medicine and University Hospitals Case Medical Center, Cleveland, Ohio 44106
| | - Fernando O Recio
- Case Cardiovascular Research Institute and Harrington Heart & Vascular Institute, University Hospitals Case Medical Center, Cleveland, Ohio 44106
| | - Mohammad A Shatat
- Case Cardiovascular Research Institute and Harrington Heart & Vascular Institute, University Hospitals Case Medical Center, Cleveland, Ohio 44106; Division of Pulmonary, Critical Care and Sleep Medicine
| | - Trent Johnson
- Case Cardiovascular Research Institute and Harrington Heart & Vascular Institute, University Hospitals Case Medical Center, Cleveland, Ohio 44106
| | - Xudong Liao
- Case Cardiovascular Research Institute and Harrington Heart & Vascular Institute, University Hospitals Case Medical Center, Cleveland, Ohio 44106; Department of Medicine, Case Western Reserve University School of Medicine and University Hospitals Case Medical Center, Cleveland, Ohio 44106
| | - Diana L Ramirez-Bergeron
- Case Cardiovascular Research Institute and Harrington Heart & Vascular Institute, University Hospitals Case Medical Center, Cleveland, Ohio 44106; Department of Medicine, Case Western Reserve University School of Medicine and University Hospitals Case Medical Center, Cleveland, Ohio 44106
| | - Aaron Proweller
- Case Cardiovascular Research Institute and Harrington Heart & Vascular Institute, University Hospitals Case Medical Center, Cleveland, Ohio 44106; Department of Medicine, Case Western Reserve University School of Medicine and University Hospitals Case Medical Center, Cleveland, Ohio 44106
| | - Masakazu Ishikawa
- Case Cardiovascular Research Institute and Harrington Heart & Vascular Institute, University Hospitals Case Medical Center, Cleveland, Ohio 44106; Department of Medicine, Case Western Reserve University School of Medicine and University Hospitals Case Medical Center, Cleveland, Ohio 44106
| | - Anne Hamik
- Case Cardiovascular Research Institute and Harrington Heart & Vascular Institute, University Hospitals Case Medical Center, Cleveland, Ohio 44106; Department of Medicine, Case Western Reserve University School of Medicine and University Hospitals Case Medical Center, Cleveland, Ohio 44106; Division of Cardiovascular Medicine, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, Ohio 44106.
| |
Collapse
|
29
|
Chen P, Pang S, Yang N, Meng H, Liu J, Zhou N, Zhang M, Xu Z, Gao W, Chen B, Tao Z, Wang L, Yang Z. Beneficial effects of schisandrin B on the cardiac function in mice model of myocardial infarction. PLoS One 2013; 8:e79418. [PMID: 24260217 PMCID: PMC3832629 DOI: 10.1371/journal.pone.0079418] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 09/16/2013] [Indexed: 12/22/2022] Open
Abstract
The fruit of Schisandra chinensis has been used in the traditional Chinese medicine for thousands of years. Accumulating evidence suggests that Schisandrin B (Sch B) has cardioprotection effect on myocardial ischemia invitro. However, it is unclear whether Sch B has beneficial effects on continuous myocardial ischemia in vivo. The aim of the present study was to investigate whether Sch B could improve cardiac function and attenuate myocardial remodeling after myocardial infarction (MI) in mice. Mice model of MI was established by permanent ligation of the left anterior descending (LAD) coronary artery. Then the MI mice were randomly treated with Sch B or vehicle alone. After treatment for 3 weeks, Sch B could increase survival rate, improve heart function and decrease infarct size compared with vehicle. Moreover, Sch B could down-regulate some inflammatory cytokines, activate eNOS pathway, inhibit cell apoptosis, and enhance cell proliferation. Further in vitro study on H9c2 cells showed similar effects of Sch B on prevention of hypoxia-induced inflammation and cell apoptosis. Taken together, our results demonstrate that Sch B can reduce inflammation, inhibit apoptosis, and improve cardiac function after ischemic injury. It represents a potential novel therapeutic approach for treatment of ischemic heart disease.
Collapse
Affiliation(s)
- Pengsheng Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Sisi Pang
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Naiquan Yang
- Department of Cardiology, Huai’an Second People’s Hospital Affiliated to Xuzhou Medical College, Huai’an, China
| | - Haoyu Meng
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jia Liu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Clinical Diabetes Centre of Jiangsu Province, Nanjing Medical University, Nanjing, China
| | - Ningtian Zhou
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Min Zhang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhihui Xu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Gao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Bo Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhengxian Tao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Liansheng Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhijian Yang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- * E-mail:
| |
Collapse
|
30
|
Carnicer R, Crabtree MJ, Sivakumaran V, Casadei B, Kass DA. Nitric oxide synthases in heart failure. Antioxid Redox Signal 2013; 18:1078-99. [PMID: 22871241 PMCID: PMC3567782 DOI: 10.1089/ars.2012.4824] [Citation(s) in RCA: 122] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Accepted: 08/07/2012] [Indexed: 12/22/2022]
Abstract
SIGNIFICANCE The regulation of myocardial function by constitutive nitric oxide synthases (NOS) is important for the maintenance of myocardial Ca(2+) homeostasis, relaxation and distensibility, and protection from arrhythmia and abnormal stress stimuli. However, sustained insults such as diabetes, hypertension, hemodynamic overload, and atrial fibrillation lead to dysfunctional NOS activity with superoxide produced instead of NO and worse pathophysiology. RECENT ADVANCES Major strides in understanding the role of normal and abnormal constitutive NOS in the heart have revealed molecular targets by which NO modulates myocyte function and morphology, the role and nature of post-translational modifications of NOS, and factors controlling nitroso-redox balance. Localized and differential signaling from NOS1 (neuronal) versus NOS3 (endothelial) isoforms are being identified, as are methods to restore NOS function in heart disease. CRITICAL ISSUES Abnormal NOS signaling plays a key role in many cardiac disorders, while targeted modulation may potentially reverse this pathogenic source of oxidative stress. FUTURE DIRECTIONS Improvements in the clinical translation of potent modulators of NOS function/dysfunction may ultimately provide a powerful new treatment for many hearts diseases that are fueled by nitroso-redox imbalance.
Collapse
Affiliation(s)
- Ricardo Carnicer
- Department of Cardiovascular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Mark J. Crabtree
- Department of Cardiovascular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Vidhya Sivakumaran
- Division of Cardiology, Department of Medicine, Johns Hopkins University Medical Institutions, Baltimore, Maryland
| | - Barbara Casadei
- Department of Cardiovascular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - David A. Kass
- Division of Cardiology, Department of Medicine, Johns Hopkins University Medical Institutions, Baltimore, Maryland
| |
Collapse
|
31
|
Kulandavelu S, Whiteley KJ, Bainbridge SA, Qu D, Adamson SL. Endothelial NO Synthase Augments Fetoplacental Blood Flow, Placental Vascularization, and Fetal Growth in Mice. Hypertension 2013; 61:259-66. [DOI: 10.1161/hypertensionaha.112.201996] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
It is not known whether eNOS deficiency in the mother or the conceptus (ie, placenta and fetus) causes fetal growth restriction in mice lacking the endothelial NO synthase gene (eNOS knockout [KO]). We hypothesized that eNOS sustains fetal growth by maintaining low fetoplacental vascular tone and promoting fetoplacental vascularity and that this is a conceptus effect and is independent of maternal genotype. We found that eNOS deficiency blunted fetal growth, and blunted the normal increase in umbilical blood flow and umbilical venous diameter and the decrease in umbilical arterial Resistance Index in late gestation (14.5–17.5 days) in eNOS KO relative to C57Bl/6J controls. On day 17.5, fetoplacental capillary lobule length and capillary density in vascular corrosion casts were reduced in eNOS KO placentas. Reduced vascularization may be a result of decreased vascular endothelial growth factor mRNA and protein expression in eNOS KO placentas at this stage. These factors, combined with significant anemia found in eNOS KO fetuses, would be anticipated to reduce fetal oxygen delivery and contribute to the fetal tissue hypoxia that was detected in the heart, lung, kidney, and liver by immunohistochemistry using pimonidazole. Although maternal eNOS deficiency impairs uteroplacental adaptations to pregnancy, maternal genotype was not a significant factor affecting growth in heterozygous conceptuses. This indicates that fetal growth restriction was primarily caused by conceptus eNOS deficiency. In mice, placental hemodynamic and vascular changes with gestation and growth restriction showed strong parallels with human pregnancy. Thus, the eNOS KO model could provide insights into the pathogenesis of human intrauterine growth restriction.
Collapse
Affiliation(s)
- Shathiyah Kulandavelu
- From the Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada (S.K., K.J.W., S.A.B., D.Q., S.L.A.); and the Departments of Physiology, and Obstetrics and Gynaecology, University of Toronto, Toronto, Ontario, Canada (S.K., S.A.B., S.L.A.)
| | - Kathie J. Whiteley
- From the Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada (S.K., K.J.W., S.A.B., D.Q., S.L.A.); and the Departments of Physiology, and Obstetrics and Gynaecology, University of Toronto, Toronto, Ontario, Canada (S.K., S.A.B., S.L.A.)
| | - Shannon A. Bainbridge
- From the Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada (S.K., K.J.W., S.A.B., D.Q., S.L.A.); and the Departments of Physiology, and Obstetrics and Gynaecology, University of Toronto, Toronto, Ontario, Canada (S.K., S.A.B., S.L.A.)
| | - Dawei Qu
- From the Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada (S.K., K.J.W., S.A.B., D.Q., S.L.A.); and the Departments of Physiology, and Obstetrics and Gynaecology, University of Toronto, Toronto, Ontario, Canada (S.K., S.A.B., S.L.A.)
| | - S. Lee Adamson
- From the Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada (S.K., K.J.W., S.A.B., D.Q., S.L.A.); and the Departments of Physiology, and Obstetrics and Gynaecology, University of Toronto, Toronto, Ontario, Canada (S.K., S.A.B., S.L.A.)
| |
Collapse
|
32
|
Hirose M, Takeishi Y, Nakada T, Shimojo H, Kashihara T, Nishio A, Suzuki S, Mende U, Matsumoto K, Matsushita N, Taira E, Sato F, Yamada M. Nicorandil prevents Gαq-induced progressive heart failure and ventricular arrhythmias in transgenic mice. PLoS One 2012; 7:e52667. [PMID: 23285142 PMCID: PMC3527603 DOI: 10.1371/journal.pone.0052667] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Accepted: 11/19/2012] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Beneficial effects of nicorandil on the treatment of hypertensive heart failure (HF) and ischemic heart disease have been suggested. However, whether nicorandil has inhibitory effects on HF and ventricular arrhythmias caused by the activation of G protein alpha q (Gα(q)) -coupled receptor (GPCR) signaling still remains unknown. We investigated these inhibitory effects of nicorandil in transgenic mice with transient cardiac expression of activated Gα(q) (Gα(q)-TG). METHODOLOGY/PRINCIPAL FINDINGS Nicorandil (6 mg/kg/day) or vehicle was chronically administered to Gα(q)-TG from 8 to 32 weeks of age, and all experiments were performed in mice at the age of 32 weeks. Chronic nicorandil administration prevented the severe reduction of left ventricular fractional shortening and inhibited ventricular interstitial fibrosis in Gα(q)-TG. SUR-2B and SERCA2 gene expression was decreased in vehicle-treated Gα(q)-TG but not in nicorandil-treated Gα(q)-TG. eNOS gene expression was also increased in nicorandil-treated Gα(q)-TG compared with vehicle-treated Gα(q)-TG. Electrocardiogram demonstrated that premature ventricular contraction (PVC) was frequently (more than 20 beats/min) observed in 7 of 10 vehicle-treated Gα(q)-TG but in none of 10 nicorandil-treated Gα(q)-TG. The QT interval was significantly shorter in nicorandil-treated Gα(q)-TG than vehicle-treated Gα(q)-TG. Acute nicorandil administration shortened ventricular monophasic action potential duration and reduced the number of PVCs in Langendorff-perfused Gα(q)-TG mouse hearts. Moreover, HMR1098, a blocker of cardiac sarcolemmal K(ATP) channels, significantly attenuated the shortening of MAP duration induced by nicorandil in the Gα(q)-TG heart. CONCLUSIONS/SIGNIFICANCE These findings suggest that nicorandil can prevent the development of HF and ventricular arrhythmia caused by the activation of GPCR signaling through the shortening of the QT interval, action potential duration, the normalization of SERCA2 gene expression. Nicorandil may also improve the impaired coronary circulation during HF.
Collapse
Affiliation(s)
- Masamichi Hirose
- Department of Molecular and Cellular Pharmacology, Iwate Medical University School of Pharmaceutical Sciences, Shiwa, Iwate, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Huang LT, Hsieh CS, Chang KA, Tain YL. Roles of nitric oxide and asymmetric dimethylarginine in pregnancy and fetal programming. Int J Mol Sci 2012. [PMID: 23203083 PMCID: PMC3509599 DOI: 10.3390/ijms131114606] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Nitric oxide (NO) regulates placental blood flow and actively participates in trophoblast invasion and placental development. Asymmetric dimethylarginine (ADMA) can inhibit NO synthase, which generates NO. ADMA has been associated with uterine artery flow disturbances such as preeclampsia. Substantial experimental evidence has reliably supported the hypothesis that an adverse in utero environment plays a role in postnatal physiological and pathophysiological programming. Growing evidence suggests that the placental nitrergic system is involved in epigenetic fetal programming. In this review, we discuss the roles of NO and ADMA in normal and compromised pregnancies as well as the link between placental insufficiency and epigenetic fetal programming.
Collapse
Affiliation(s)
- Li-Tung Huang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 800, Taiwan; E-Mail:
- Department of Traditional Chinese Medicine, Chang Gung University, Linkow 333, Taiwan
| | - Chih-Sung Hsieh
- Department of Pediatric Surgery, Pingtung Christian Hospital, Pingtung 900, Taiwan; E-Mail:
- Department of Nursing, MeiHo University, Pingtung 900, Taiwan
| | - Kow-Aung Chang
- Department of Anesthesiology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 800, Taiwan; E-Mail:
| | - You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 800, Taiwan; E-Mail:
- Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 800, Taiwan
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +886-975056995; Fax: +886-7-7338009
| |
Collapse
|
34
|
Liu Y, Lu X, Xiang FL, Poelmann RE, Gittenberger-de Groot AC, Robbins J, Feng Q. Nitric oxide synthase-3 deficiency results in hypoplastic coronary arteries and postnatal myocardial infarction. Eur Heart J 2012; 35:920-31. [PMID: 23048191 DOI: 10.1093/eurheartj/ehs306] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
AIMS Hypoplastic coronary artery disease is a rare congenital abnormality that is associated with sudden cardiac death. However, molecular mechanisms responsible for this disease are not clear. The aim of the present study was to assess the role of nitric oxide synthase-3 (NOS3) in the pathogenesis of hypoplastic coronary arteries. METHODS AND RESULTS Wild-type (WT), NOS3(-/-), and a novel cardiac-specific NOS3 overexpression mouse model were employed. Deficiency in NOS3 resulted in coronary artery hypoplasia in foetal mice and spontaneous myocardial infarction in postnatal hearts. Coronary artery diameters, vessel density, and volume were significantly decreased in NOS3(-/-) mice at postnatal day 0. In addition, NOS3(-/-) mice showed a significant increase in the ventricular wall thickness, myocardial volume, and cardiomyocyte cell size compared with WT mice. Lack of NOS3 also down-regulated the expression of Gata4, Wilms tumour-1, vascular endothelial growth factor, basic fibroblast growth factor and erythropoietin, and inhibited migration of epicardial cells. These abnormalities and hypoplastic coronary arteries in the NOS3(-/-) mice were completely rescued by the cardiac-specific overexpression of NOS3. CONCLUSION Nitric oxide synthase-3 is required for coronary artery development and deficiency in NOS3 leads to hypoplastic coronary arteries.
Collapse
Affiliation(s)
- Yin Liu
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada N6A 5C1
| | | | | | | | | | | | | |
Collapse
|
35
|
NOing the heart: role of nitric oxide synthase-3 in heart development. Differentiation 2012; 84:54-61. [PMID: 22579300 DOI: 10.1016/j.diff.2012.04.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Revised: 04/03/2012] [Accepted: 04/10/2012] [Indexed: 01/30/2023]
Abstract
Congenital heart disease is the most common birth defect in humans. Identifying factors that are critical to embryonic heart development could further our understanding of the disease and lead to new strategies of its prevention and treatment. Nitric oxide synthase-3 (NOS3) or endothelial nitric oxide synthase (eNOS) is known for many important biological functions including vasodilation, vascular homeostasis and angiogenesis. Over the past decade, studies from our lab and others have shown that NOS3 is required during heart development. More specifically, deficiency in NOS3 results in congenital septal defects, cardiac hypertrophy and postnatal heart failure. In addition, NOS3 is pivotal to the morphogenesis of major coronary arteries and myocardial capillary development. Interestingly, these effects of NOS3 are mediated through induction of transcription and growth factors that are crucial in the formation of coronary arteries. Finally, deficiency in NOS3 results in high incidences of bicuspid aortic valves, a disease in humans that often leads to complications with age including aortic valve stenosis or regurgitation, endocarditis, aortic aneurysm formation, and aortic dissection. In summary, these data suggest NOS3 plays a critical role in embryonic heart development and morphogenesis of coronary arteries and aortic valves.
Collapse
|
36
|
dl-3n-Butylphthalide Promotes Angiogenesis Via the Extracellular Signal-regulated Kinase 1/2 and Phosphatidylinositol 3-Kinase/Akt-endothelial Nitric Oxide Synthase Signaling Pathways. J Cardiovasc Pharmacol 2012; 59:352-62. [DOI: 10.1097/fjc.0b013e3182443e74] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
37
|
Mujoo K, Krumenacker JS, Murad F. Nitric oxide-cyclic GMP signaling in stem cell differentiation. Free Radic Biol Med 2011; 51:2150-7. [PMID: 22019632 PMCID: PMC3232180 DOI: 10.1016/j.freeradbiomed.2011.09.037] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Revised: 09/27/2011] [Accepted: 09/29/2011] [Indexed: 12/15/2022]
Abstract
The nitric oxide-cyclic GMP (NO-cGMP) pathway mediates important physiological functions associated with various integrative body systems including the cardiovascular and nervous systems. Furthermore, NO regulates cell growth, survival, apoptosis, proliferation, and differentiation at the cellular level. To understand the significance of the NO-cGMP pathway in development and differentiation, studies have been conducted both in developing embryos and in stem cells. Manipulation of the NO-cGMP pathway, by employing activators and inhibitors as pharmacological probes, and genetic manipulation of NO signaling components have implicated the involvement of this pathway in the regulation of stem cell differentiation. This review focuses on some of the work pertaining to the role of NO-cGMP in the differentiation of stem cells into cells of various lineages, particularly into myocardial cells, and in stem cell-based therapy.
Collapse
Affiliation(s)
- Kalpana Mujoo
- Brown Foundation Institute of Molecular Medicine, Texas Therapeutics Institute, University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| | | | | |
Collapse
|
38
|
Kazakov A, Müller P, Jagoda P, Semenov A, Böhm M, Laufs U. Endothelial nitric oxide synthase of the bone marrow regulates myocardial hypertrophy, fibrosis, and angiogenesis. Cardiovasc Res 2011; 93:397-405. [DOI: 10.1093/cvr/cvr305] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
|
39
|
Krause B, Hanson M, Casanello P. Role of nitric oxide in placental vascular development and function. Placenta 2011; 32:797-805. [PMID: 21798594 PMCID: PMC3218217 DOI: 10.1016/j.placenta.2011.06.025] [Citation(s) in RCA: 152] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Revised: 06/28/2011] [Accepted: 06/29/2011] [Indexed: 11/27/2022]
Abstract
Nitric oxide (NO) is one of the most pleiotropic signaling molecules at systemic and cellular levels, participating in vascular tone regulation, cellular respiration, proliferation, apoptosis and gene expression. Indeed NO actively participates in trophoblast invasion, placental development and represents the main vasodilator in this tissue. Despite the large number of studies addressing the role of NO in the placenta, its participation in placental vascular development and the effect of altered levels of NO on placental function remains to be clarified. This review draws a time-line of the participation of NO throughout placental vascular development, from the differentiation of vascular precursors to the consolidation of vascular function are considered. The influence of NO on cell types involved in the origin of the placental vasculature and the expression and function of the nitric oxide synthases (NOS) throughout pregnancy are described. The developmental processes involved in the placental vascular bed are considered, such as the participation of NO in placental vasculogenesis and angiogenesis through VEGF and Angiopoietin signaling molecules. The role of NO in vascular function once the placental vascular tree has developed, in normal pregnancy as well as in pregnancy-related diseases, is then discussed.
Collapse
Affiliation(s)
- B.J. Krause
- Division of Obstetrics and Gynecology, School of Medicine, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, Chile
| | - M.A. Hanson
- Institute of Developmental Sciences, Academic Unit of Human Development & Health, Faculty of Medicine, University of Southampton, SO16 6YD, UK
| | - P. Casanello
- Division of Obstetrics and Gynecology, School of Medicine, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, Chile
| |
Collapse
|
40
|
Bartsch C, Bekhite MM, Wolheim A, Richter M, Ruhe C, Wissuwa B, Marciniak A, Müller J, Heller R, Figulla HR, Sauer H, Wartenberg M. NADPH oxidase and eNOS control cardiomyogenesis in mouse embryonic stem cells on ascorbic acid treatment. Free Radic Biol Med 2011; 51:432-43. [PMID: 21570463 DOI: 10.1016/j.freeradbiomed.2011.04.029] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Revised: 04/14/2011] [Accepted: 04/15/2011] [Indexed: 02/07/2023]
Abstract
Ascorbic acid (AA) increases cardiomyogenesis of embryonic stem (ES) cells. Herein we show that treatment of mouse ES cells with AA enhanced cardiac differentiation accompanied by an upregulation of the NADPH oxidase isoforms NOX2 and NOX4, phosphorylation of endothelial nitric oxide synthase (eNOS), and cyclic GMP (cGMP) formation, indicating that reactive oxygen species (ROS) as well as nitric oxide (NO) may be involved in cardiomyogenesis. In whole mount embryoid bodies as well as isolated Flk-1-positive (Flk-1(+)) cardiovascular progenitor cells ROS elevation by AA was observed in early stages of differentiation (Days 4-7), and absent at Day 10. In contrast NO generation following incubation with AA was absent at Day 4 and increased at Days 7 and 10. AA-mediated cardiomyogenesis was blunted by the NADPH oxidase inhibitors diphenylen iodonium (DPI) and apocynin, the free radical scavengers N-(2-mercaptopropionyl)-glycine (NMPG) and ebselen, and the NOS inhibitor L-NAME. Downregulation of NOX4 by short hairpin RNA (shRNA) resulted in significant inhibition of cardiomyogenesis and abolished the stimulation of MHC-ß and MLC2v gene expression observed on AA treatment. Our data demonstrate that AA stimulates cardiomyocyte differentiation from ES cells by signaling pathways that involve ROS generated at early stages and NO at late stages of cardiomyogenesis.
Collapse
Affiliation(s)
- Caroline Bartsch
- Department of Internal Medicine I, Cardiology Division, Friedrich Schiller University, Jena, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Luong C, Rey-Perra J, Vadivel A, Gilmour G, Sauve Y, Koonen D, Walker D, Todd KG, Gressens P, Kassiri Z, Nadeem K, Morgan B, Eaton F, Dyck JR, Archer SL, Thébaud B. Antenatal sildenafil treatment attenuates pulmonary hypertension in experimental congenital diaphragmatic hernia. Circulation 2011; 123:2120-31. [PMID: 21537000 DOI: 10.1161/circulationaha.108.845909] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Lung hypoplasia and persistent pulmonary hypertension of the newborn limit survival in congenital diaphragmatic hernia (CDH). Unlike other diseases resulting in persistent pulmonary hypertension of the newborn, infants with CDH are refractory to inhaled nitric oxide (NO). Nitric oxide mediates pulmonary vasodilatation at birth in part via cyclic GMP production. Phosphodiesterase type 5 (PDE5) limits the effects of NO by inactivation of cyclic GMP. Because of the limited success in postnatal management of CDH, we hypothesized that antenatal PDE5 inhibition would attenuate pulmonary artery remodeling in experimental nitrofen-induced CDH. METHODS AND RESULTS Nitrofen administered at embryonic day 9.5 to pregnant rats resulted in a 60% incidence of CDH in the offspring and recapitulated features seen in human CDH, including structural abnormalities (lung hypoplasia, decreased pulmonary vascular density, pulmonary artery remodeling, right ventricular hypertrophy), and functional abnormalities (decreased pulmonary artery relaxation in response to the NO donor 2-(N,N-diethylamino)-diazenolate-2-oxide). Antenatal sildenafil administered to the pregnant rat from embryonic day 11.5 to embryonic day 20.5 crossed the placenta, increased fetal lung cyclic GMP and decreased active PDE5 expression. Antenatal sildenafil improved lung structure, increased pulmonary vessel density, reduced right ventricular hypertrophy, and improved postnatal NO donor 2-(N,N-diethylamino)-diazenolate-2-oxide-induced pulmonary artery relaxation. This was associated with increased lung endothelial NO synthase and vascular endothelial growth factor protein expression. Antenatal sildenafil had no adverse effect on retinal structure/function and brain development. CONCLUSIONS Antenatal sildenafil improves pathological features of persistent pulmonary hypertension of the newborn in experimental CDH and does not alter the development of other PDE5-expressing organs. Given the high mortality/morbidity of CDH, the potential benefit of prenatal PDE5 inhibition in improving the outcome for infants with CDH warrants further studies.
Collapse
Affiliation(s)
- Christina Luong
- Department of Pediatrics, Women and Children Health Research Institute, Cardiovascular Research Group, University of Alberta, Edmonton, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Ou ZJ, Chang FJ, Luo D, Liao XL, Wang ZP, Zhang X, Xu YQ, Ou JS. Endothelium-derived microparticles inhibit angiogenesis in the heart and enhance the inhibitory effects of hypercholesterolemia on angiogenesis. Am J Physiol Endocrinol Metab 2011; 300:E661-8. [PMID: 21245463 DOI: 10.1152/ajpendo.00611.2010] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Therapeutic angiogenesis remains unsuccessful in coronary artery disease. It is known that plasma endothelium-derived microparticles (EMPs) are increased in coronary artery disease and that hypercholesterolemia can inhibit angiogenesis. We evaluated the relationship between EMPs and hypercholesterolemia in the impairment of angiogenesis. EMPs isolated from human umbilical vein endothelial cells were injected into low-density lipoprotein receptor-null (LDLr(-/-)) mice fed a Western diet for 2 wk and C57BL6 mice for 6 h or were directly added to the tissue culture media. Hearts isolated from mice were sectioned and cultured, and endothelial tube formation was measured. The expression and phosphorylation of endothelial NO synthase (eNOS) and the generation of NO in the hearts were determined. Angiogenesis was inhibited by pathophysiological concentrations of EMPs but not physiological concentrations of EMPs in hearts from C57BL6 mice. However, angiogenesis was inhibited by EMPs at both physiological and pathophysiological concentrations of EMPs in hearts from hypercholesterolemic LDLr(-/-) mice. Pathophysiological concentrations of EMPs decreased eNOS phosphorylation at Ser(1177) and NO generation without altering eNOS expression in hearts from C57BL6 mice. Both physiological and pathophysiological concentrations of EMPs decreased not only eNOS phosphorylation at Ser(1177) and NO generation, but eNOS expression in hypercholesterolemic hearts from LDLr(-/-) mice. These data demonstrated that pathophysiological concentrations of EMPs could inhibit angiogenesis in hearts by decreasing eNOS activity. EMPs and hypercholesterolemia mutually enhanced their inhibitory effect of angiogenesis by inducing eNOS dysfunction. Our findings suggest a novel mechanism by which hypercholesterolemia impairs angiogenesis.
Collapse
Affiliation(s)
- Zhi-Jun Ou
- Division of Hypertension and Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Khazaei M, Fallahzadeh AR, Sharifi MR, Afsharmoghaddam N, Javanmard SH, Salehi E. Effects of diabetes on myocardial capillary density and serum angiogenesis biomarkers in male rats. Clinics (Sao Paulo) 2011; 66:1419-24. [PMID: 21915494 PMCID: PMC3161222 DOI: 10.1590/s1807-59322011000800019] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Accepted: 04/26/2011] [Indexed: 01/10/2023] Open
Abstract
INTRODUCTION Cardiovascular disease is one of the main causes of mortality and morbidity in diabetic patients. This study evaluated the effects of diabetes on myocardial capillary density and several serum angiogenic factors including nitric oxide, vascular endothelial growth factor, and soluble vascular endothelial growth factor receptors. METHODS Twelve male rats were divided into two groups: control and diabetic (n = 6 each). Diabetes was induced with a single dose of streptozotocin (50 mg/kg). After 21 days, capillary density in the myocardial tissue was evaluated using immunohistochemical staining and is reported as capillaries per mm². Blood samples were collected before and after the induction of diabetes. RESULTS In the diabetic group, serum nitric oxide and soluble vascular endothelial growth factor receptor 2 concentrations were lower than the levels in the control group, while the level of soluble vascular endothelial growth factor receptor 1 was significantly higher. There was no significant change in the serum vascular endothelial growth factor concentration between the diabetic and control groups; however, the ratio of vascular endothelial growth factor to vascular endothelial growth factor receptor 1 was significantly lower in the diabetic animals. The myocardial capillary density was also lower in the diabetic group compared with the control group (1549 ± 161 vs. 2156 ± 202/mm², respectively). CONCLUSION Reduced serum nitric oxide and vascular endothelial growth factor receptor 2 levels, increased serum vascular endothelial growth factor receptor 1 levels and a lower vascular endothelial growth factor to vascular endothelial growth factor receptor 1 ratio may be responsible for the decreased myocardial capillary density in diabetic rats.
Collapse
Affiliation(s)
- Majid Khazaei
- Department of Physiology, Isfahan University of Medical Sciences, Isfahan, Iran.
| | | | | | | | | | | |
Collapse
|
44
|
Nakano-Doi A, Nakagomi T, Fujikawa M, Nakagomi N, Kubo S, Lu S, Yoshikawa H, Soma T, Taguchi A, Matsuyama T. Bone marrow mononuclear cells promote proliferation of endogenous neural stem cells through vascular niches after cerebral infarction. Stem Cells 2010; 28:1292-302. [PMID: 20517983 DOI: 10.1002/stem.454] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Increasing evidence shows that administration of bone marrow mononuclear cells (BMMCs) is a potential treatment for various ischemic diseases, such as ischemic stroke. Although angiogenesis has been considered primarily responsible for the effect of BMMCs, their direct contribution to endothelial cells (ECs) by being a functional elements of vascular niches for neural stem/progenitor cells (NSPCs) has not been considered. Herein, we examine whether BMMCs affected the properties of ECs and NSPCs, and whether they promoted neurogenesis and functional recovery after stroke. We compared i.v. transplantations 1 x 10(6) BMMCs and phosphate-buffered saline in mice 2 days after cortical infarction. Systemically administered BMMCs preferentially accumulated at the postischemic cortex and peri-infarct area in brains; cell proliferation of ECs (angiogenesis) at these regions was significantly increased in BMMCs-treated mice compared with controls. We also found that endogenous NSPCs developed in close proximity to ECs in and around the poststroke cortex and that ECs were essential for proliferation of these ischemia-induced NSPCs. Furthermore, BMMCs enhanced proliferation of NSPCs as well as ECs. Proliferation of NSPCs was suppressed by additional treatment with endostatin (known to inhibit proliferation of ECs) following BMMCs transplantation. Subsequently, neurogenesis and functional recovery were also promoted in BMMCs-treated mice compared with controls. These results suggest that BMMCs can contribute to the proliferation of endogenous ischemia-induced NSPCs through vascular niche regulation, which includes regulation of endothelial proliferation. In addition, these results suggest that BMMCs transplantation has potential as a novel therapeutic option in stroke treatment.
Collapse
Affiliation(s)
- Akiko Nakano-Doi
- Institute for Advanced Medical Sciences, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Kuehl K, Loffredo C, Lammer EJ, Iovannisci DM, Shaw GM. Association of congenital cardiovascular malformations with 33 single nucleotide polymorphisms of selected cardiovascular disease-related genes. ACTA ACUST UNITED AC 2010; 88:101-10. [PMID: 19764075 DOI: 10.1002/bdra.20630] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Clark (1996) proposed that abnormal blood flow is related to some congenital cardiovascular malformations (CCVMs), particularly CCVM with obstruction to blood flow. Our hypothesis is that CCVMs may relate to genes that affect blood coagulation or flow. We studied whether polymorphisms of such genes are related to CCVMs; previous association of these SNPs to conotruncal CCVMs is described. METHODS We assessed risk of pulmonary stenosis (PS, N = 120), atrial septal defect (ASD, N = 108), aortic stenosis (AS, N = 36), and coarctation of the aorta (CoAo, N = 64), associated with 33 candidate genes, selected for their relationship to blood flow affected by homocysteine metabolism, coagulation, cell-cell interaction, inflammation, or blood pressure regulation. RESULTS Effects were specific to cardiac phenotype and race. CoAo was associated with MTHFR (-667) C>T (odds ratio [OR] for TT 3.5, 95% confidence limits [CI] 1.4-8.6). AS was associated with a polymorphism of SERPINE1, G5>G4, OR = 5.6 for the homozygote with 95% CI 1.4-22.9. Unique polymorphisms were associated with increased risk of ASD and PS: NPPA 664G>A with ASD (OR of 2.4, 95%CI 1.3-4.4) and NOS3 (-690) C>T with PS (OR 6.1; 95% CI 1.6-22.6 in the African American population only). For ASD, the NPPA (-664) G>A SNP there was increased risk from the variant genotype only in maternal smokers (OR 2.6; 95% CI 1.0-7.2). CONCLUSIONS Genes affecting vascular function and coagulation appear to be promising candidates for the etiology of cardiac malformations and warrant further study.
Collapse
Affiliation(s)
- Karen Kuehl
- Children's National Heart Institute, Washington, DC 20010, USA.
| | | | | | | | | |
Collapse
|
46
|
Abstract
Fenofibrate have been illustrated to stimulate nitric oxide (NO) pathway, which plays pivotal roles in neovascularization. Here, we evaluated the effect of fenofibrate on neovascularization using a murine ischemic hindlimb model. C57BL/6J mice were treated with fenofibrate and/or NG-nitro-l-arginine methyl ester hydrochloride (l-NAME) for 28 days after ischemia operation. We exploited a concentration of L-NAME that did not affect blood pressure levels but suppress NO activity. Limb blood perfusion and capillary density in ischemic limb, serum NO levels, and aortic NOS activity were significantly increased by fenofibrate treatment when compared with the untreatment group. And, these effects were abolished by coadministration of L-NAME. Fenofibrate treatment significantly lowered serum triglyceride levels. Cotreatment of L-NAME did not inhibit serum triglyceride level, lowering effect of fenofibrate. These results suggested that the lowering in serum triglyceride levels is not involved in the improvement of neovascularization. In an in vitro experiment, fenofibrate stimulated NOS activity in human umbilical vein endothelial cells. Also, fenofibrate stimulated in vitro angiogenesis, and this effect was abolished by coincubation with L-NAME. In conclusions, fenofibrate enhanced neovascularization in a murine hindlimb ischemia model. The mechanism is most likely through activation of NO pathway in endothelial cells.
Collapse
|
47
|
Uriu-Adams JY, Scherr RE, Lanoue L, Keen CL. Influence of copper on early development: prenatal and postnatal considerations. Biofactors 2010; 36:136-52. [PMID: 20232410 DOI: 10.1002/biof.85] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Copper (Cu) is an essential nutrient whose requirement is increased during pregnancy and lactation. These represent times of critical growth and development, and the fetus and neonate are particularly vulnerable to deficiencies of this nutrient. Genetic mutations that predispose the offspring to inadequate stores of Cu can be life threatening as is observed in children with Menkes disease. During the last decade, severe Cu deficiency, once thought to be a rare condition, has been reported in the literature at an increasing frequency. Secondary Cu deficiencies can be induced by a variety of ways such as excessive zinc or iron intake, certain drugs, and bariatric surgery. Premature and low birth weight infants can be born with low Cu stores. A number of mechanisms can contribute to the teratogenicity of Cu including decreased activity of select cuproenzymes, increased oxidative stress, decreased nitric oxide availability, altered iron metabolism, abnormal extracellular matrix protein crosslinking, decreased angiogenesis and altered cell signaling among others. The brain, heart, and vessels as well as tissues such as lung, skin and hair, and systems including the skeletal, immune, and blood systems, are negatively affected by suboptimal Cu during development. Additionally, persistent structural, biochemical, and functional adverse effects in the offspring are noted even when Cu supplementation is initiated after birth, supporting the concept that adequate Cu nutriture during pregnancy and lactation is critical for normal development. Although Cu-containing IUDs are an effective method for increasing intrauterine Cu concentrations and for reducing the risk of pregnancy, high amounts of dietary Cu are not thought to represent a direct developmental risk.
Collapse
Affiliation(s)
- Janet Y Uriu-Adams
- Department of Nutrition, University of California, Davis, Davis, CA 95616, USA.
| | | | | | | |
Collapse
|
48
|
de Waard MC, van Haperen R, Soullié T, Tempel D, de Crom R, Duncker DJ. Beneficial effects of exercise training after myocardial infarction require full eNOS expression. J Mol Cell Cardiol 2010; 48:1041-9. [PMID: 20153335 DOI: 10.1016/j.yjmcc.2010.02.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2009] [Revised: 01/30/2010] [Accepted: 02/02/2010] [Indexed: 02/09/2023]
Abstract
Exercise training attenuates left ventricular (LV) dysfunction after myocardial infarction (MI). It could be speculated that these effects of exercise are mediated by increased endothelial NO synthase (eNOS) activity. In the present study we tested the hypothesis that eNOS plays a critical role in the exercise-induced amelioration of LV dysfunction after MI. MI or sham was induced in eNOS(-/-), eNOS(+/-) and eNOS(+/+) mice. After 8 weeks of voluntary wheel running (approximately 7 km/day in all groups) or sedentary housing, global cardiac function was determined in vivo and (immuno)histochemistry was performed to assess cardiomyocyte size, fibrosis, capillary density and apoptosis in remote myocardium. At baseline eNOS(-/-) mice had higher mean aortic pressure compared to eNOS(+/-) and eNOS(+/+) mice, but had normal global cardiac function. MI resulted in marked LV remodeling, including cardiomyocyte hypertrophy and a reduction in capillary density, increased fibrosis and apoptosis, as well as LV systolic and diastolic dysfunction to the same extent in all genotypes. In eNOS(+/+) MI mice exercise abolished fibrosis and apoptosis in the remote myocardium, attenuated LV systolic dysfunction and ameliorated pulmonary congestion. These beneficial effects were lost in eNOS(+/-) and eNOS(-/-) mice, while LV systolic dysfunction and pulmonary congestion in eNOS(+/-) mice were exacerbated by exercise. In conclusion, the beneficial effects of exercise after MI on LV remodeling and dysfunction depend critically on endogenous eNOS. The observation that the lack of one eNOS allele is sufficient to negate all beneficial effects of exercise, strongly suggests that exercise depends on full eNOS expression.
Collapse
Affiliation(s)
- Monique C de Waard
- Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus MC, University Medical Center Rotterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
49
|
Kakinuma Y, Furihata M, Akiyama T, Arikawa M, Handa T, Katare RG, Sato T. Donepezil, an acetylcholinesterase inhibitor against Alzheimer's dementia, promotes angiogenesis in an ischemic hindlimb model. J Mol Cell Cardiol 2009; 48:680-93. [PMID: 19962381 DOI: 10.1016/j.yjmcc.2009.11.010] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2009] [Revised: 11/07/2009] [Accepted: 11/10/2009] [Indexed: 11/25/2022]
Abstract
Our recent studies have indicated that acetylcholine (ACh) protects cardiomyocytes from prolonged hypoxia through activation of the PI3K/Akt/HIF-1alpha/VEGF pathway and that cardiomyocyte-derived VEGF promotes angiogenesis in a paracrine fashion. These results suggest that a cholinergic system plays a role in modulating angiogenesis. Therefore, we assessed the hypothesis that the cholinergic modulator donepezil, an acetylcholinesterase inhibitor utilized in Alzheimer's disease, exhibits beneficial effects, especially on the acceleration of angiogenesis. We evaluated the effects of donepezil on angiogenic properties in vitro and in vivo, using an ischemic hindlimb model of alpha7 nicotinic receptor-deleted mice (alpha7 KO) and wild-type mice (WT). Donepezil activated angiogenic signals, i.e., HIF-1alpha and VEGF expression, and accelerated tube formation in human umbilical vein endothelial cells (HUVECs). ACh and nicotine upregulated signal transduction with acceleration of tube formation, suggesting that donepezil promotes a common angiogenesis pathway. Moreover, donepezil-treated WT exhibited rich capillaries with enhanced VEGF and PCNA endothelial expression, recovery from impaired tissue perfusion, prevention of ischemia-induced muscular atrophy with sustained surface skin temperature in the limb, and inhibition of apoptosis independent of the alpha7 receptor. Donepezil exerted comparably more effects in alpha7 KO in terms of angiogenesis, tissue perfusion, biochemical markers, and surface skin temperature. Donepezil concomitantly elevated VEGF expression in intracardiac endothelial cells of WT and alpha7 KO and further increased choline acetyltransferase (ChAT) protein expression, which is critical for ACh synthesis in endothelial cells. The present study concludes that donepezil can act as a therapeutic tool to accelerate angiogenesis in cardiovascular disease patients.
Collapse
Affiliation(s)
- Yoshihiko Kakinuma
- Department of Cardiovascular Control, Kochi Medical School, Nankoku, Kochi 783-8505, Japan.
| | | | | | | | | | | | | |
Collapse
|
50
|
Majumder S, Gupta R, Reddy H, Sinha S, Muley A, Kolluru GK, Chatterjee S. Cadmium attenuates bradykinin-driven nitric oxide production by interplaying with the localization pattern of endothelial nitric oxide synthase. Biochem Cell Biol 2009; 87:605-20. [PMID: 19767824 DOI: 10.1139/o09-018] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Cadmium, a ubiquitous heavy metal, interferes with endothelial functions and angiogenesis. Bradykinin is a Ca-mobilizing soluble peptide that acts via nitric oxide to promote vasodilation and capillary permeability. The objective of the present study was to explore the Cd implications in bradykinin-dependent endothelial functions. An egg yolk angiogenesis model was employed to evaluate the effect of Cd on bradykinin-induced angiogenesis. The results demonstrate that 100 nmol/L Cd attenuated bradykinin-dependent angiogenesis. The results of the in vitro wound healing and tube formation assays by using EAhy 926, a transformed endothelial cell line, suggest that Cd blocked bradykinin-mediated endothelial migration and tube formation by 38% and 67%, respectively, while nitric oxide supplementation could reverse the effect of Cd on bradykinin-induced endothelial migration by 94%. The detection of nitric oxide by using a DAF-2DA fluorescent probe, Griess assay, and ultrasensitive electrode suggests that Cd blocked bradykinin-induced nitric oxide production. Fluorescence imaging of eNOS-GFP transfected endothelial cells, immunofluorescence, and Western blot studies of Cd and bradykinin-treated cells show that Cd interfered with the localization pattern of eNOS, which possibly attenuates nitric oxide production in part. Additionally, Ca imaging of Cd- and bradykinin-treated cells suggests that Cd blocked bradykinin-dependent Ca influx into the cells, thus partially blocking Ca-dependent nitric oxide production in endothelial cells. The results of this study conclude that Cd blunted the effect of bradykinin by interfering with the Ca-associated NOS activity specifically by impeding subcellular trafficking of eNOS.
Collapse
Affiliation(s)
- Syamantak Majumder
- Vascular Biology Lab, Life Sciences Division, AU-KBC Research Centre, MIT Campus, Anna University, Chennai 600044, Tamil Nadu, India
| | | | | | | | | | | | | |
Collapse
|