1
|
Winn NC, Cappel DA, Pollack ED, Lantier L, Riveros JK, Bracy DP, Beckman JA, Wasserman DH. Increased cGMP improves microvascular exercise training adaptations in diet-induced obesity. Am J Physiol Endocrinol Metab 2025; 328:E711-E722. [PMID: 40204283 PMCID: PMC12070609 DOI: 10.1152/ajpendo.00368.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/15/2024] [Accepted: 04/02/2025] [Indexed: 04/11/2025]
Abstract
With the development of atherosclerosis, impaired microvascular function can result in diminished capacity for ambulation and is a risk factor for type 2 diabetes. Dynamic changes in vascular tone are determined, in large part, by the endothelial nitric oxide synthase (eNOS)/nitric oxide (NO)/cGMP axis. We used pharmacological gain of function of the eNOS/NO/cGMP axis in diet-induced obese (DIO) mice and reduced function in lean mice to test the hypothesis that functionality of this vascular control mechanism parallels the benefits of an exercise training regimen. DIO mice have 50% lower exercise capacity (P < 0.0001) than lean mice and were used for pharmacological gain of function. The phosphodiesterase-5a (PDE-5a) inhibitor, sildenafil, increases cGMP and was administered to DIO mice daily. In sedentary mice, neither acute nor chronic sildenafil improves exercise capacity. In contrast, chronic sildenafil synergizes with exercise training to improve performance during an incremental exercise test. Improved exercise performance was accompanied by a 40% increase in basal skeletal muscle capillary flow velocity and ∼20% increase in plasma-perfused capillary density measured via intravital microscopy. Loss of function was tested in lean mice hemizygous for endothelial cell (EC) specific eNOS creating an EC-eNOS knockdown (KD). EC-eNOS KD decreases capillary density and exercise tolerance in sedentary mice; however, it did not prevent exercise-training-induced improvements in endurance capacity. These data show that 1) increasing cGMP with sildenafil enhances microcirculatory function and exercise work tolerance that results from training; 2) eNOS KD does not prevent the microcirculatory or improvements in exercise tolerance with training. PDE-5a inhibitors combined with physical exercise are a potential mechanism for improving ambulation in patients with circulatory limitations.NEW & NOTEWORTHY This study used pharmacological gain-of-function and genetic loss-of-function approaches to test the hypothesis that the eNOS/NO/cGMP axis is central to exercise training adaptations in microcirculatory function and exercise capacity. Chronic but not acute treatment with the PDE5 inhibitor, sildenafil, synergizes with exercise training to improve performance with incremental exercise in obese mice; whereas endothelium-specific knockdown in eNOS does not blunt the microcirculatory adaptations and improvements in exercise tolerance with training.
Collapse
Affiliation(s)
- Nathan C. Winn
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - David A. Cappel
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Ethan D. Pollack
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Louise Lantier
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Mouse Metabolic Phenotyping Center, Vanderbilt University, Nashville, Tennessee, USA
| | - Jillian K. Riveros
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Deanna P. Bracy
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Joshua A. Beckman
- Division of Vascular Medicine, UT Southwestern Medical Center, Dallas, TX
| | - David H. Wasserman
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Mouse Metabolic Phenotyping Center, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
2
|
Brunmaier LAE, Ozdemir T, Walker TW. Angiogenesis: Biological Mechanisms and In Vitro Models. Ann Biomed Eng 2025:10.1007/s10439-025-03721-2. [PMID: 40210793 DOI: 10.1007/s10439-025-03721-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 03/25/2025] [Indexed: 04/12/2025]
Abstract
The translation of biomedical devices and drug research is an expensive and long process with a low probability of receiving FDA approval. Developing physiologically relevant in vitro models with human cells offers a solution to not only improving the odds of FDA approval but also to expand our ability to study complex in vivo systems in a simpler fashion. Animal models remain the standard for pre-clinical testing; however, the data from animal models is an unreliable extrapolation when anticipating a human response in clinical trials, thus contributing to the low rates of translation. In this review, we focus on in vitro vascular or angiogenic models because of the incremental role that the vascular system plays in the translation of biomedical research. The first section of this review discusses the most common angiogenic cytokines that are used in vitro to initiate angiogenesis, followed by angiogenic inhibitors where both initiators and inhibitors work to maintain vascular homeostasis. Next, we evaluate previously published in vitro models, where we evaluate capturing the physical environment for biomimetic in vitro modeling. These topics provide a foundation of parameters that must be considered to improve and achieve vascular biomimicry. Finally, we summarize these topics to suggest a path forward with the goal of engineering human in vitro models that emulate the in vivo environment and provide a platform for biomedical device and drug screening that produces data to support clinical translation.
Collapse
Affiliation(s)
- Laura A E Brunmaier
- Nanoscience and Biomedical Engineering Department, South Dakota School of Mines & Technology, 501 E St. Joseph St., Rapid City, SD, 57701, USA
| | - Tugba Ozdemir
- Nanoscience and Biomedical Engineering Department, South Dakota School of Mines & Technology, 501 E St. Joseph St., Rapid City, SD, 57701, USA
| | - Travis W Walker
- Karen M. Swindler Department of Chemical and Biological Engineering, South Dakota School of Mines & Technology, 501 E St. Joseph St., Rapid City, SD, 57701, USA.
| |
Collapse
|
3
|
Liu Y, Wu Z, Li Y, Chen Y, Zhao X, Wu M, Xia Y. Metabolic reprogramming and interventions in angiogenesis. J Adv Res 2025; 70:323-338. [PMID: 38704087 PMCID: PMC11976431 DOI: 10.1016/j.jare.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/30/2024] [Accepted: 05/01/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND Endothelial cell (EC) metabolism plays a crucial role in the process of angiogenesis. Intrinsic metabolic events such as glycolysis, fatty acid oxidation, and glutamine metabolism, support secure vascular migration and proliferation, energy and biomass production, as well as redox homeostasis maintenance during vessel formation. Nevertheless, perturbation of EC metabolism instigates vascular dysregulation-associated diseases, especially cancer. AIM OF REVIEW In this review, we aim to discuss the metabolic regulation of angiogenesis by EC metabolites and metabolic enzymes, as well as prospect the possible therapeutic opportunities and strategies targeting EC metabolism. KEY SCIENTIFIC CONCEPTS OF REVIEW In this work, we discuss various aspects of EC metabolism considering normal and diseased vasculature. Of relevance, we highlight that the implications of EC metabolism-targeted intervention (chiefly by metabolic enzymes or metabolites) could be harnessed in orchestrating a spectrum of pathological angiogenesis-associated diseases.
Collapse
Affiliation(s)
- Yun Liu
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Zifang Wu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yikun Li
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China; College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Yating Chen
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Xuan Zhao
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| | - Miaomiao Wu
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan 410128, China.
| | - Yaoyao Xia
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| |
Collapse
|
4
|
Winn NC, Cappel DA, Pollock ED, Lantier L, Riveros JK, Debrow P, Bracy DP, Beckman JA, Wasserman DH. Increased cGMP improves microvascular exercise training adaptations independent of endothelial nitric oxide synthase. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.18.612717. [PMID: 39345415 PMCID: PMC11429803 DOI: 10.1101/2024.09.18.612717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Impaired microvascular function is a hallmark of pre-diabetes. With development of atherosclerosis this impaired microvascular function can result in diminished capacity for ambulation and is a risk factor for Type 2 Diabetes. Dynamic changes in vascular tone are determined, in large part, by the eNOS/NO/cGMP axis. We used gain of function of the eNOS/NO/cGMP axis in diet-induced obese (DIO) mice and reduced function in lean mice to test the hypothesis that functionality of this vascular control mechanism parallels the benefits of an exercise training regimen. DIO mice have lower exercise capacity than lean mice and were used for pharmacological gain of function. The PDE-5a inhibitor - sildenafil - increases cGMP and was administered to DIO mice daily. In sedentary mice, we find that sildenafil does not improve exercise capacity. In contrast, it amplifies the microcirculatory effects of exercise training. Sildenafil synergizes with exercise training to improve performance during an incremental exercise test. Improved exercise performance was accompanied by increased skeletal muscle capillary flow velocity and capillary density measured via intravital microscopy. Loss of function was tested in lean mice hemizygous for endothelial cell (EC) specific eNOS creating an EC-eNOS knockdown (KD). EC-eNOS KD decreases capillary density and exercise tolerance in sedentary mice; however, it did not prevent exercise-training induced improvements in endurance capacity. These data show that 1) increasing cGMP with sildenafil enhances microcirculatory function and exercise work tolerance that results from training; 2) eNOS KD does not prevent the microcirculatory or improvements in exercise tolerance with training. PDE-5a inhibitors combined with physical exercise are a potential mechanism for improving ambulation in patients with circulatory limitations.
Collapse
Affiliation(s)
- Nathan C. Winn
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - David A. Cappel
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Ethan D. Pollock
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Louise Lantier
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Mouse Metabolic Phenotyping Center, Vanderbilt University, Nashville, Tennessee, USA
| | - Jillian K. Riveros
- Department of Molecular Metabolism; Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Payton Debrow
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Deanna P. Bracy
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Joshua A. Beckman
- Division of Vascular Medicine, UT Southwestern Medical Center, Dallas, TX
| | - David H. Wasserman
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Mouse Metabolic Phenotyping Center, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
5
|
Zhang B, Li Y, Peng A, Liu C, Lin J, Feng Y, Wan J. Association between the pan-immune-inflammation value and coronary collateral circulation in chronic total coronary occlusive patients. BMC Cardiovasc Disord 2024; 24:458. [PMID: 39198732 PMCID: PMC11351751 DOI: 10.1186/s12872-024-04139-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 08/21/2024] [Indexed: 09/01/2024] Open
Abstract
BACKGROUND Inflammation and immunity play important roles in the formation of coronary collateral circulation (CCC). The pan-immune-inflammation value (PIV) is a novel marker for evaluating systemic inflammation and immunity. The study aimed to investigate the association between the PIV and CCC formation in patients with chronic total occlusion (CTO). METHODS This retrospective study enrolled 1150 patients who were diagnosed with CTO through coronary angiographic (CAG) examinations from January 2013 to December 2021 in China. The Cohen-Rentrop criteria were used to catagorize CCC formation: good CCC formation (Rentrop grade 2-3) and poor CCC formation group (Rentrop grade 0-1). Based on the tertiles of the PIV, all patients were classified into three groups as follows: P1 group, PIV ≤ 237.56; P2 group, 237.56< PIV ≤ 575.18; and P3 group, PIV > 575.18. RESULTS A significant relationship between the PIV and the formation of CCC was observed in our study. Utilizing multivariate logistic regression and adjusting for confounding factors, the PIV emerged as an independent risk factor for poor CCC formation. Notably, the restricted cubic splines revealed a dose-response relationship between the PIV and risk of poor CCC formation. In terms of predictive accuracy, the area under the ROC curve (AUC) for PIV in anticipating poor CCC formation was 0.618 (95% CI: 0.584-0.651, P < 0.001). Furthermore, the net reclassification index (NRI) and integrated discrimination index (IDI) for PIV, concerning the prediction of poor CCC formation, were found to be 0.272 (95% CI: 0.142-0.352, P < 0.001) and 0.051 (95% CI: 0.037-0.065, P < 0.001), respectively. It's noteworthy that both the NRI and IDI values were higher for PIV compared to other inflammatory biomarkers, suggesting its superiority in predictive capacity. CONCLUSIONS PIV was associated with the formation of CCC. Notably, PIV exhibited potential as a predictor for poor CCC formation and showcased superior predictive performance compared to other complete blood count-based inflammatory biomarkers.
Collapse
Affiliation(s)
- Bing Zhang
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Ya Li
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Aihong Peng
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Cuiyi Liu
- Department of Cardiology, Xi 'An Da Xing Hospital, Xi 'an, 710000, Shanxi, China
| | - Jiesheng Lin
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Munich, Germany
- Institute for Medical Information Processing, Biometry, and Epidemiology, Faculty of Medicine, Pettenkofer School of Public Health, LMU Munich, 85764, Munich, Germany
| | - Yujia Feng
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Jing Wan
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China.
| |
Collapse
|
6
|
Cai Y, Li L, Shao C, Chen Y, Wang Z. Therapeutic Strategies for Angiogenesis Based on Endothelial Cell Epigenetics. J Cardiovasc Transl Res 2024; 17:816-827. [PMID: 38294628 DOI: 10.1007/s12265-024-10485-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 01/17/2024] [Indexed: 02/01/2024]
Abstract
With the in-depth investigation of various diseases, angiogenesis has gained increasing attention. Among the contributing factors to angiogenesis research, endothelial epigenetics has emerged as an influential player. Endothelial epigenetic therapy exerts its regulatory effects on endothelial cells by controlling gene expression, RNA, and histone modification within these cells, which subsequently promotes or inhibits angiogenesis. As a result, this therapeutic approach offers potential strategies for disease treatment. The purpose of this review is to outline the pertinent mechanisms of endothelial cell epigenetics, encompassing glycolysis, lactation, amino acid metabolism, non-coding RNA, DNA methylation, histone modification, and their connections to specific diseases and clinical applications. We firmly believe that endothelial cell epigenetics has the potential to become an integral component of precision medicine therapy, unveiling novel therapeutic targets and providing new directions and opportunities for disease treatment.
Collapse
Affiliation(s)
- Yue Cai
- Department of Cardiology, Affiliated Hospital of Jiangsu University, No. 438 Jiefang Road, Jingkou District, Zhenjiang, 212000, Jiangsu Province, China
| | - Lihua Li
- Department of Pathology, Affiliated Hospital of Jiangsu University, No. 438 Jiefang Road, Jingkou District, Zhenjiang, 212000, Jiangsu Province, China
| | - Chen Shao
- Department of Cardiology, Affiliated Hospital of Jiangsu University, No. 438 Jiefang Road, Jingkou District, Zhenjiang, 212000, Jiangsu Province, China
| | - Yiliu Chen
- Department of Cardiology, Affiliated Hospital of Jiangsu University, No. 438 Jiefang Road, Jingkou District, Zhenjiang, 212000, Jiangsu Province, China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, No. 438 Jiefang Road, Jingkou District, Zhenjiang, 212000, Jiangsu Province, China.
| |
Collapse
|
7
|
Alvi SB, Pracha N, Shalaan M, Dholaniya PS, Mergaye M, Sridharan D, Khan M. Fabrication and Optimization of Poly(ε-caprolactone) Microspheres Loaded with S-Nitroso-N-Acetylpenicillamine for Nitric Oxide Delivery. Biomedicines 2024; 12:1363. [PMID: 38927571 PMCID: PMC11201505 DOI: 10.3390/biomedicines12061363] [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: 03/18/2024] [Revised: 06/03/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Heart disease is one of the leading causes of death in the United States and throughout the world. While there are different techniques for reducing or preventing the impact of heart disease, nitric oxide (NO) is administered as nitroglycerin for reversing angina or chest pain. Unfortunately, due to its gaseous and short-lived half-life, NO can be difficult to study or even administer. Therefore, controlled delivery of NO is desirable for therapeutic use. In the current study, the goal was to fabricate NO-releasing microspheres (MSs) using a donor molecule, S-Nitroso-N-Acetyl penicillamine, (SNAP), and encapsulating it in poly(ε-caprolactone) (PCL) using a single-emulsion technique that can provide sustained delivery of NO to cells over time without posing any toxicity risks. Optimization of the fabrication process was performed by varying the duration of homogenization (5, 10, and 20 min) and its effect on entrapment efficiency and size. The optimized SNAP-MS had an entrapment efficiency of ˃50%. Furthermore, we developed a modified method for NO detection by using NO microsensors to detect the NO release from SNAP-MSs in real time, showing sustained release behavior. The fabricated SNAP-MSs were tested for biocompatibility with HUVECs (human umbilical vein endothelial cells), which were found to be biocompatible. Lastly, we tested the effect of controlled NO delivery to human induced pluripotent stem-derived cardiomyocytes (hiPSC-CMs) via SNAP-MSs, which showed a significant improvement in the electrophysiological parameters and alleviated anoxic stress.
Collapse
Affiliation(s)
- Syed Baseeruddin Alvi
- Division of Basic and Translational Research, Department of Emergency Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Nooruddin Pracha
- Division of Basic and Translational Research, Department of Emergency Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Mahmoud Shalaan
- Division of Basic and Translational Research, Department of Emergency Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Pankaj Singh Dholaniya
- Division of Basic and Translational Research, Department of Emergency Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad 500046, India
| | - Muhamad Mergaye
- Division of Basic and Translational Research, Department of Emergency Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Divya Sridharan
- Division of Basic and Translational Research, Department of Emergency Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Mahmood Khan
- Division of Basic and Translational Research, Department of Emergency Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH 43210, USA
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
8
|
Hricisák L, Pál É, Nagy D, Delank M, Polycarpou A, Fülöp Á, Sándor P, Sótonyi P, Ungvári Z, Benyó Z. NO Deficiency Compromises Inter- and Intrahemispheric Blood Flow Adaptation to Unilateral Carotid Artery Occlusion. Int J Mol Sci 2024; 25:697. [PMID: 38255769 PMCID: PMC10815552 DOI: 10.3390/ijms25020697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/30/2023] [Accepted: 01/01/2024] [Indexed: 01/24/2024] Open
Abstract
Carotid artery stenosis (CAS) affects approximately 5-7.5% of older adults and is recognized as a significant risk factor for vascular cognitive impairment (VCI). The impact of CAS on cerebral blood flow (CBF) within the ipsilateral hemisphere relies on the adaptive capabilities of the cerebral microcirculation. In this study, we aimed to test the hypothesis that the impaired availability of nitric oxide (NO) compromises CBF homeostasis after unilateral carotid artery occlusion (CAO). To investigate this, three mouse models exhibiting compromised production of NO were tested: NOS1 knockout, NOS1/3 double knockout, and mice treated with the NO synthesis inhibitor L-NAME. Regional CBF changes following CAO were evaluated using laser-speckle contrast imaging (LSCI). Our findings demonstrated that NOS1 knockout, NOS1/3 double knockout, and L-NAME-treated mice exhibited impaired CBF adaptation to CAO. Furthermore, genetic deficiency of one or two NO synthase isoforms increased the tortuosity of pial collaterals connecting the frontoparietal and temporal regions. In conclusion, our study highlights the significant contribution of NO production to the functional adaptation of cerebrocortical microcirculation to unilateral CAO. We propose that impaired bioavailability of NO contributes to the impaired CBF homeostasis by altering inter- and intrahemispheric blood flow redistribution after unilateral disruption of carotid artery flow.
Collapse
Affiliation(s)
- László Hricisák
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.H.); (É.P.); (D.N.); (M.D.); (A.P.); (Á.F.); (P.S.)
- HUN-REN-SU Cerebrovascular and Neurocognitive Diseases Research Group, 1094 Budapest, Hungary
| | - Éva Pál
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.H.); (É.P.); (D.N.); (M.D.); (A.P.); (Á.F.); (P.S.)
- HUN-REN-SU Cerebrovascular and Neurocognitive Diseases Research Group, 1094 Budapest, Hungary
| | - Dorina Nagy
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.H.); (É.P.); (D.N.); (M.D.); (A.P.); (Á.F.); (P.S.)
- HUN-REN-SU Cerebrovascular and Neurocognitive Diseases Research Group, 1094 Budapest, Hungary
| | - Max Delank
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.H.); (É.P.); (D.N.); (M.D.); (A.P.); (Á.F.); (P.S.)
| | - Andreas Polycarpou
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.H.); (É.P.); (D.N.); (M.D.); (A.P.); (Á.F.); (P.S.)
- Mayo Clinic, College of Medicine and Science, Rochester, MN 55905, USA
- Division of Cardiothoracic Surgery, University of Minnesota, Minneapolis, MN 55455, USA
| | - Ágnes Fülöp
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.H.); (É.P.); (D.N.); (M.D.); (A.P.); (Á.F.); (P.S.)
- HUN-REN-SU Cerebrovascular and Neurocognitive Diseases Research Group, 1094 Budapest, Hungary
| | - Péter Sándor
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.H.); (É.P.); (D.N.); (M.D.); (A.P.); (Á.F.); (P.S.)
- HUN-REN-SU Cerebrovascular and Neurocognitive Diseases Research Group, 1094 Budapest, Hungary
| | - Péter Sótonyi
- Department of Vascular and Endovascular Surgery, Semmelweis University, 1122 Budapest, Hungary;
| | - Zoltán Ungvári
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Doctoral College/Department of Public Health, Semmelweis University, 1089 Budapest, Hungary
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Zoltán Benyó
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.H.); (É.P.); (D.N.); (M.D.); (A.P.); (Á.F.); (P.S.)
- HUN-REN-SU Cerebrovascular and Neurocognitive Diseases Research Group, 1094 Budapest, Hungary
| |
Collapse
|
9
|
Joó JG, Sulyok E, Bódis J, Kornya L. Disrupted Balance of the Oxidant-Antioxidant System in the Pathophysiology of Female Reproduction: Oxidative Stress and Adverse Pregnancy Outcomes. Curr Issues Mol Biol 2023; 45:8091-8111. [PMID: 37886954 PMCID: PMC10605220 DOI: 10.3390/cimb45100511] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 09/29/2023] [Accepted: 10/02/2023] [Indexed: 10/28/2023] Open
Abstract
The significance of oxidative stress in the pathophysiology of male reproductive processes has been closely studied in the last two decades. Recently, it has become clear that oxidative stress can lead to numerous pathological conditions during female reproductive processes as well, contributing to the development of endometriosis, polycystic ovary syndrome and various forms of infertility. During pregnancy, physiological generation of reactive oxygen species (ROS) occurs in association with several developmental processes including oocyte maturation and implantation. An overproduction of ROS can lead to disturbances in fetal development and increases the risk for missed abortion, intrauterine growth restriction, pre-eclampsia, premature delivery and gestational diabetes. Our review focuses on the etiological role of the disrupted oxidant-antioxidant system during human gestation as it relates to adverse pregnancy outcomes.
Collapse
Affiliation(s)
- József Gábor Joó
- Department of Obstetrics and Gynecology, Semmelweis University, 1088 Budapest, Hungary
| | - Endre Sulyok
- Faculty of Health Sciences, University of Pécs, 7621 Pécs, Hungary
| | - József Bódis
- Faculty of Health Sciences, University of Pécs, 7621 Pécs, Hungary
| | - László Kornya
- Central Hospital of South Pest National Institute of Hematology and Infectious Diseases, 1476 Budapest, Hungary
| |
Collapse
|
10
|
Yoshida YG, Yan S, Xu H, Yang J. Novel Metal Nanomaterials to Promote Angiogenesis in Tissue Regeneration. ENGINEERED REGENERATION 2023; 4:265-276. [PMID: 37234753 PMCID: PMC10207714 DOI: 10.1016/j.engreg.2023.03.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023] Open
Abstract
Angiogenesis-the formation of new blood vessels from existing blood vessels-has drawn significant attention in medical research. New techniques have been developed to control proangiogenic factors to obtain desired effects. Two important research areas are 1) understanding cellular mechanisms and signaling pathways involved in angiogenesis and 2) discovering new biomaterials and nanomaterials with proangiogenic effects. This paper reviews recent developments in controlling angiogenesis in the context of regenerative medicine and wound healing. We focus on novel proangiogenic materials that will advance the field of regenerative medicine. Specifically, we mainly focus on metal nanomaterials. We also discuss novel technologies developed to carry these proangiogenic inorganic molecules efficiently to target sites. We offer a comprehensive overview by combining existing knowledge regarding metal nanomaterials with novel developments that are still being refined to identify new nanomaterials.
Collapse
Affiliation(s)
- Yuki G. Yoshida
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, USA
| | - Su Yan
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Hui Xu
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Jian Yang
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
11
|
Sabe SA, Scrimgeour LA, Xu CM, Sabra M, Karbasiafshar C, Aboulgheit A, Abid MR, Sellke FW. Extracellular vesicle therapy attenuates antiangiogenic signaling in ischemic myocardium of swine with metabolic syndrome. J Thorac Cardiovasc Surg 2023; 166:e5-e14. [PMID: 36244819 PMCID: PMC10023593 DOI: 10.1016/j.jtcvs.2022.09.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/25/2022] [Accepted: 09/12/2022] [Indexed: 10/14/2022]
Abstract
OBJECTIVE Our recent studies using a porcine model of metabolic syndrome (MS) and chronic myocardial ischemia show that extracellular vesicle (EV) therapy improves blood flow and arteriogenesis in ischemic myocardium, although mechanisms of these changes are unclear. We hypothesized that in the setting of MS, EV therapy would decrease antiangiogenic signaling to mediate increased blood flow to chronically ischemic myocardium. METHODS Yorkshire swine were fed a high-fat diet for 4 weeks to induce MS, then underwent placement of an ameroid constrictor to the left circumflex artery to induce chronic myocardial ischemia. Two weeks later, pigs underwent intramyocardial injection of vehicle (control, n = 6) or human bone marrow-derived EVs (n = 8). Five weeks later, left ventricular myocardium in ischemic territory was harvested. Protein expression was measured using immunoblot analysis, and data were analyzed using Wilcoxon rank sum test. Myocardial perfusion was measured with isotope-labeled microspheres, and correlation data were analyzed using Spearman rank correlation coefficient. RESULTS EV treatment was associated with decreased expression of antiangiogenic proteins, angiostatin (P < .001) and endostatin (P = .043) in ischemic myocardium compared with control. In EV-treated pigs, there was a negative correlation between blood flow to ischemic myocardium and angiostatin (rs = -0.76; P = .037), but not endostatin expression (rs = .02; P = .98). EV treatment was also associated with decreased cathepsin D, which cleaves precursors to produce angiostatin and endostatin, in ischemic myocardium (P = .020). CONCLUSIONS In the setting of MS and chronic myocardial ischemia, EV therapy is associated with decreased expression of antiangiogenic proteins, which might contribute to increased blood flow to chronically ischemic myocardium.
Collapse
Affiliation(s)
- Sharif A Sabe
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Laura A Scrimgeour
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Cynthia M Xu
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Mohamed Sabra
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Catherine Karbasiafshar
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Ahmed Aboulgheit
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - M Ruhul Abid
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Frank W Sellke
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI.
| |
Collapse
|
12
|
Wang W, Shang W, Zou J, Liu K, Liu M, Qiu X, Zhang H, Wang K, Wang N. ZNF667 facilitates angiogenesis after myocardial ischemia through transcriptional regulation of VASH1 and Wnt signaling pathway. Int J Mol Med 2022; 50:129. [PMID: 36043524 PMCID: PMC9448299 DOI: 10.3892/ijmm.2022.5185] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 08/09/2022] [Indexed: 11/24/2022] Open
Abstract
Zinc finger protein 667 (ZNF667, also referred as Mipu1), a widely expressed KRAB/C2H2-type zinc finger transcription factor, can protect against hypoxic-ischemic myocardial injury. Pro-angiogenesis is regarded as a promising strategy for the treatment of acute myocardial infarction (AMI). However, whether ZNF667 is involved in the angiogenesis following AMI remains to be elucidated. The present study reported that the expression of ZNF667 in CD31-positive endothelial cells (ECs) was upregulated in the heart of AMI mice. Hypoxic challenge (1% oxygen) promoted the mRNA and protein expression of ZNF667 in the human umbilical vein endothelial cells (HUVECs) in a time-dependent manner. Moreover, ZNF667 promoted hypoxia-induced invasion and tube formation of HUVECs. Mechanically, ZNF667 could directly bind to the promoter of anti-angiogenic gene VASH1 and inhibit its expression. Consequently, VASH1 overexpression abolished hypoxic challenge or ZNF667 overexpression-induced invasion and tube formation of HUVECs. Further bioinformatic analyses suggested that overexpression of ZNF667 or knockdown of VASH1-induced differentially expressed genes in HUVECs were greatly enriched in the Wnt signaling pathway (DAAM1, LEF1, RAC2, FRAT1, NFATc2 and WNT5A). Together, these data suggested that ZNF667 facilitates myocardial ischemia-driven angiogenesis through transcriptional repression of VASH1 and regulation of Wnt signaling pathway.
Collapse
Affiliation(s)
- Wenmei Wang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410008, P.R. China
| | - Weite Shang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410008, P.R. China
| | - Jiang Zou
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410008, P.R. China
| | - Ke Liu
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410008, P.R. China
| | - Meidong Liu
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410008, P.R. China
| | - Xiaoqin Qiu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Huali Zhang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410008, P.R. China
| | - Kangkai Wang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410008, P.R. China
| | - Nian Wang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
13
|
Chen L, Wu Z, Yang L, Chen Y, Wang W, Cheng L, Li C, Lv D, Xia L, Chen J, Tang L, Zhang LI, Zhang S, Luo J. Nitric oxide in multikinase inhibitor-induced hand-foot skin reaction. Transl Res 2022; 245:82-98. [PMID: 35189405 DOI: 10.1016/j.trsl.2022.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 01/17/2022] [Accepted: 02/14/2022] [Indexed: 11/13/2022]
Abstract
Hand-foot skin reaction (HFSR) is the most debilitating and prevalent side effect caused by multikinase inhibitors (MKIs) that share vascular endothelial growth factor receptor (VEGFR) as the common inhibition target, such as sorafenib, regorafenib, axitinib, etc. Though not life-threatening, HFSR can significantly deteriorate patients' quality of life and jeopardize the continuity of cancer therapy. Despite years of efforts, there are no FDA-approved treatments for HFSR and the understanding of the precise pathogenic mechanism is still limited. In this study, we hypothesized that nitric oxide has the potential therapeutic effect to reverse the toxicity caused by MKI through upregulation of several VEGF/VEGFR downstream signaling pathways. We found that glyceryl trinitrate (GTN), a nitric oxide donor, could stimulate cell proliferation, migration, and protect cells from apoptosis induced by MKIs in vitro. Local application of GTN mitigated tissue damage in a rat model, while not impacting the anti-tumor effect of the MKI in HepG2 tumor-bearing mice. Finally, GTN ointment alleviated cutaneous damages and improved quality of life in 6 HFSR patients. Our study proposed and validated the mechanism to counteract VEGFR inhibition, providing GTN as the potential treatment to MKI-induced HFSR, which may further improve the therapeutic window of various MKI based cancer therapies.
Collapse
Affiliation(s)
- Leying Chen
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Zhaoyu Wu
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Linan Yang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yuyun Chen
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Wenhong Wang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Liting Cheng
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Chong Li
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Dazhao Lv
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Liangyong Xia
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Jia Chen
- Department of Dermatopathology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lina Tang
- Department of Oncology, the 6th People' Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China.
| | - L I Zhang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China..
| | - Shiyi Zhang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China..
| | - Jie Luo
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China..
| |
Collapse
|
14
|
Brandt MM, Cheng C, Merkus D, Duncker DJ, Sorop O. Mechanobiology of Microvascular Function and Structure in Health and Disease: Focus on the Coronary Circulation. Front Physiol 2022; 12:771960. [PMID: 35002759 PMCID: PMC8733629 DOI: 10.3389/fphys.2021.771960] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/11/2021] [Indexed: 12/19/2022] Open
Abstract
The coronary microvasculature plays a key role in regulating the tight coupling between myocardial perfusion and myocardial oxygen demand across a wide range of cardiac activity. Short-term regulation of coronary blood flow in response to metabolic stimuli is achieved via adjustment of vascular diameter in different segments of the microvasculature in conjunction with mechanical forces eliciting myogenic and flow-mediated vasodilation. In contrast, chronic adjustments in flow regulation also involve microvascular structural modifications, termed remodeling. Vascular remodeling encompasses changes in microvascular diameter and/or density being largely modulated by mechanical forces acting on the endothelium and vascular smooth muscle cells. Whereas in recent years, substantial knowledge has been gathered regarding the molecular mechanisms controlling microvascular tone and how these are altered in various diseases, the structural adaptations in response to pathologic situations are less well understood. In this article, we review the factors involved in coronary microvascular functional and structural alterations in obstructive and non-obstructive coronary artery disease and the molecular mechanisms involved therein with a focus on mechanobiology. Cardiovascular risk factors including metabolic dysregulation, hypercholesterolemia, hypertension and aging have been shown to induce microvascular (endothelial) dysfunction and vascular remodeling. Additionally, alterations in biomechanical forces produced by a coronary artery stenosis are associated with microvascular functional and structural alterations. Future studies should be directed at further unraveling the mechanisms underlying the coronary microvascular functional and structural alterations in disease; a deeper understanding of these mechanisms is critical for the identification of potential new targets for the treatment of ischemic heart disease.
Collapse
Affiliation(s)
- Maarten M Brandt
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Caroline Cheng
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.,Division of Internal Medicine and Dermatology, Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, Netherlands
| | - Daphne Merkus
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.,Walter Brendel Center of Experimental Medicine (WBex), LMU Munich, Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance (MHA), Munich, Germany
| | - Dirk J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Oana Sorop
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
15
|
Vascular Effects of Low-Dose ACE2 Inhibitor MLN-4760—Benefit or Detriment in Essential Hypertension? Biomedicines 2021; 10:biomedicines10010038. [PMID: 35052717 PMCID: PMC8773407 DOI: 10.3390/biomedicines10010038] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 12/17/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infects host cells through angiotensin-converting enzyme 2 (ACE2). Concurrently, the product of ACE2 action, angiotensin 1–7 (Ang 1–7), binds to Mas receptors within the cardiovascular system and provides protective effects. Therefore, it is crucial to reveal the role of ACE2 inhibition, especially within pre-existing cardiovascular pathologies. In our study, we imitated the action of SARS-CoV-2 in organisms using the low dose of the ACE2 inhibitor MLN-4760 with the aim of investigating to what degree ACE2 inhibition is detrimental to the cardiovascular system of spontaneously hypertensive rats (SHRs), which represent a model of human essential hypertension. Our study revealed the complex action of MLN-4760 in SHRs. On the one hand, we found that MLN-4760 had (1) (pro)obesogenic effects that negatively correlated with alternative renin-angiotensin system activity and Ang 1–7 in plasma, (2) negative effects on ACE1 inhibitor (captopril) action, (3) detrimental effects on the small arteries function and (4) anti-angiogenic effect in the model of chick chorioallantoic membrane. On the other hand, MLN-4760 induced compensatory mechanisms involving strengthened Mas receptor-, nitric oxide- and hydrogen sulfide-mediated signal transduction in the aorta, which was associated with unchanged blood pressure, suggesting beneficial action of MLN-4760 when administered at a low dose.
Collapse
|
16
|
Kuryata OV, Kushnir YS, Nedzvetsky VS, Korsa VV, Tykhomyrov AA. Serum Levels of the Biomarkers Associated with Astrocytosis, Neurodegeneration, and Demyelination: Neurological Benefits of Citicoline Treatment of Patients with Ischemic Stroke and Atrial Fibrillation. NEUROPHYSIOLOGY+ 2021; 53:2-12. [PMID: 34866692 PMCID: PMC8630515 DOI: 10.1007/s11062-021-09907-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Indexed: 01/31/2023]
Abstract
Ischemic stroke is a main complication of atrial fibrillation (cardiac arrhythmia). The aim of our study was to estimate the effects of citicoline (CDP-choline) therapy on the levels of circulating neurospecific protein markers in serum of the patients with ischemic stroke and atrial fibrillation. Fiftyfour patients (mean age 76 years) treated with citicoline in a dose of 2.0 g daily intravenously for 12 to 14 days in addition to basic treatment formed the examined group. Thirty-two patients (mean age 68.5 years) obtained only standard therapy and formed the control group. Serum levels of neuronal and glial protein markers, including glial fibrillary acidic protein (GFAP), a neurofilament light subunit (NF-L), myelin basic protein (MBP), and ionized calcium-binding adaptor molecule 1 (Iba1), were measured in patients of both groups before and after treatment; an immunoblotting technique followed by densitometry analysis were used. Supplementary citicoline treatment provided significant reductions of the levels of GFAP (33%, P = 0.034), NF-L (27%, P = 0.019), and MBP (32%, P = 0.018), as compared to the initial values, while there were no marked changes in the studied parameters in the control group. The results obtained allow us to hypothesize that therapeutic benefit of citicoline in patients with ischemic stroke and atrial fibrillation can be mediated through increasing neuronal viability, protecting against axonal injury, decreasing the level of reactive astrogliosis, preventing deficiencies in the blood-brain integrity, and reducing the intensity of demyelination. However, citicoline administration exerted no effect on the blood content of microglial marker Iba-1, thus possibly preserving an important functional significance of microglia, which is needed to resolve local inflammation and clear cellular debris, and also provide protective factors to reduce cell injury in the ischemic brain. The obtained results indicate that serum levels of neurospecific biomarkers are significant and clinically relevant indices of the efficiency of treatment of the above-mentioned pathologies and can be used for further investigations of the stroke pathophysiology and molecular mechanisms of nootropic-mediated neuroprotection.
Collapse
Affiliation(s)
| | | | - V. S. Nedzvetsky
- Bingöl University, Bingöl, Turkey
- Oles Honchar Dnipro National University, Dnipro, Ukraine
| | - V. V. Korsa
- Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - A. A. Tykhomyrov
- Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| |
Collapse
|
17
|
Yang B, Li G, Liu J, Li X, Zhang S, Sun F, Liu W. Nanotechnology for Age-Related Macular Degeneration. Pharmaceutics 2021; 13:pharmaceutics13122035. [PMID: 34959316 PMCID: PMC8705006 DOI: 10.3390/pharmaceutics13122035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/04/2021] [Accepted: 11/22/2021] [Indexed: 01/12/2023] Open
Abstract
Age-related macular degeneration (AMD) is a degenerative eye disease that is the leading cause of irreversible vision loss in people 50 years and older. Today, the most common treatment for AMD involves repeated intravitreal injections of anti-vascular endothelial growth factor (VEGF) drugs. However, the existing expensive therapies not only cannot cure this disease, they also produce a variety of side effects. For example, the number of injections increases the cumulative risk of endophthalmitis and other complications. Today, a single intravitreal injection of gene therapy products can greatly reduce the burden of treatment and improve visual effects. In addition, the latest innovations in nanotherapy provide the best drug delivery alternative for the treatment of AMD. In this review, we discuss the development of nano-drug delivery systems and gene therapy strategies for AMD in recent years. In addition, we discuss some novel targeting strategies and the potential application of these delivery methods in the treatment of AMD. Finally, we also propose that the combination of CRISPR/Cas9 technology with a new non-viral delivery system may be promising as a therapeutic strategy for the treatment of AMD.
Collapse
Affiliation(s)
- Bo Yang
- Department of Anesthesiology, The Second Hospital of Jilin University, Changchun 130012, China;
- School of Life Sciences, Jilin University, Changchun 130012, China; (G.L.); (J.L.); (X.L.); (S.Z.); (F.S.)
| | - Ge Li
- School of Life Sciences, Jilin University, Changchun 130012, China; (G.L.); (J.L.); (X.L.); (S.Z.); (F.S.)
| | - Jiaxin Liu
- School of Life Sciences, Jilin University, Changchun 130012, China; (G.L.); (J.L.); (X.L.); (S.Z.); (F.S.)
| | - Xiangyu Li
- School of Life Sciences, Jilin University, Changchun 130012, China; (G.L.); (J.L.); (X.L.); (S.Z.); (F.S.)
| | - Shixin Zhang
- School of Life Sciences, Jilin University, Changchun 130012, China; (G.L.); (J.L.); (X.L.); (S.Z.); (F.S.)
| | - Fengying Sun
- School of Life Sciences, Jilin University, Changchun 130012, China; (G.L.); (J.L.); (X.L.); (S.Z.); (F.S.)
| | - Wenhua Liu
- Department of Anesthesiology, The Second Hospital of Jilin University, Changchun 130012, China;
- Correspondence:
| |
Collapse
|
18
|
Decreased parasite burden and altered host response in children with sickle cell anemia and severe anemia with malaria. Blood Adv 2021; 5:4710-4720. [PMID: 34470050 PMCID: PMC8759120 DOI: 10.1182/bloodadvances.2021004704] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 05/21/2021] [Indexed: 11/20/2022] Open
Abstract
Plasmodium falciparum malaria causes morbidity and mortality in African children with sickle cell anemia (SCA), but comparisons of host responses to P falciparum between children with SCA (homozygous sickle cell disease/hemoglobin SS [HbSS]) and normal hemoglobin genotype/hemoglobin AA (HbAA) are limited. We assessed parasite biomass and plasma markers of inflammation and endothelial activation in children with HbAA (n = 208) or HbSS (n = 22) who presented with severe anemia and P falciparum parasitemia to Mulago Hospital in Kampala, Uganda. Genotyping was performed at study completion. No child had known SCA at enrollment. Children with HbSS did not differ from children with HbAA in peripheral parasite density, but had significantly lower sequestered parasite biomass. Children with HbSS had greater leukocytosis but significantly lower concentrations of several plasma inflammatory cytokines, including tumor necrosis factor α (TNF-α). In contrast, children with HbSS had threefold greater concentrations of angiopoietin-2 (Angpt-2), a marker of endothelial dysregulation associated with mortality in severe malaria. Lower TNF-α concentrations were associated with increased risk of postdischarge mortality or readmission, whereas higher Angpt-2 concentrations were associated with increased risk of recurrent clinical malaria. Children with SCA have decreased parasite sequestration and inflammation but increased endothelial dysregulation during severe anemia with P falciparum parasitemia, which may ameliorate acute infectious complications but predispose to harmful long-term sequelae.
Collapse
|
19
|
Sharma M, Rana U, Joshi C, Michalkiewicz T, Afolayan A, Parchur A, Joshi A, Teng RJ, Konduri GG. Decreased Cyclic Guanosine Monophosphate-Protein Kinase G Signaling Impairs Angiogenesis in a Lamb Model of Persistent Pulmonary Hypertension of the Newborn. Am J Respir Cell Mol Biol 2021; 65:555-567. [PMID: 34185619 PMCID: PMC8641848 DOI: 10.1165/rcmb.2020-0434oc] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 06/25/2021] [Indexed: 11/24/2022] Open
Abstract
Impaired angiogenesis function in pulmonary artery endothelial cells (PAEC) contributes to persistent pulmonary hypertension of the newborn (PPHN). Decreased nitric oxide (NO) amounts in PPHN lead to impaired mitochondrial biogenesis and angiogenesis in the lung; the mechanisms remain unclear. We hypothesized that decreased cyclic guanosine monophosphate (cGMP)-PKG (protein kinase G) signaling downstream of NO leads to decreased mitochondrial biogenesis and angiogenesis in PPHN. PPHN was induced by ductus arteriosus constriction from 128-136 days' gestation in fetal lambs. Control animals were gestation-matched lambs that did not undergo ductal constriction. PAEC isolated from PPHN lambs were treated with the sGC (soluble guanylate cyclase) activator cinaciguat, the PKG activator 8-bromo-cGMP, or the PDE-V (PDE type V) inhibitor sildenafil. Lysates were immunoblotted for mitochondrial transcription factors and electron transport chain C-I (complex I), C-II, C-III, C-IV, and C-V proteins. The in vitro angiogenesis of PAEC was evaluated by using tube-formation and scratch-recovery assays. cGMP concentrations were measured by using an enzyme immunoassay. Fetal lambs with ductal constriction were given sildenafil or control saline through continuous infusion in utero, and the lung histology, capillary counts, vessel density, and right ventricular pressure were assessed at birth. PPHN PAEC showed decreased mitochondrial transcription factor levels, electron transport chain protein levels, and in vitro tube formation and cell migration; these were restored by cinaciguat, 8-bromo-cGMP, and sildenafil. Cinaciguat and sildenafil increased cGMP concentrations in PPHN PAEC. Radial alveolar and capillary counts and vessel density were lower in PPHN lungs, and the right ventricular pressure and Fulton Index were higher in PPHN lungs; these were improved by in utero sildenafil infusion. cGMP-PKG signaling is a potential therapeutic target to restore decreased mitochondrial biogenesis and angiogenesis in PPHN.
Collapse
Affiliation(s)
- Megha Sharma
- Division of Neonatology, Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Ujala Rana
- Division Neonatology, Department of Pediatrics, Medical College of Wisconsin and Children’s Research Institute, Children’s Wisconsin, Wauwatosa, Wisconsin; and
| | - Chintamani Joshi
- Division Neonatology, Department of Pediatrics, Medical College of Wisconsin and Children’s Research Institute, Children’s Wisconsin, Wauwatosa, Wisconsin; and
| | - Teresa Michalkiewicz
- Division Neonatology, Department of Pediatrics, Medical College of Wisconsin and Children’s Research Institute, Children’s Wisconsin, Wauwatosa, Wisconsin; and
| | - Adeleye Afolayan
- Division Neonatology, Department of Pediatrics, Medical College of Wisconsin and Children’s Research Institute, Children’s Wisconsin, Wauwatosa, Wisconsin; and
| | - Abdul Parchur
- Center for Imaging, Department of Radiology, Medical College of Wisconsin, Wauwatosa, Wisconsin
| | - Amit Joshi
- Center for Imaging, Department of Radiology, Medical College of Wisconsin, Wauwatosa, Wisconsin
| | - Ru-Jeng Teng
- Division Neonatology, Department of Pediatrics, Medical College of Wisconsin and Children’s Research Institute, Children’s Wisconsin, Wauwatosa, Wisconsin; and
| | - Girija G. Konduri
- Division Neonatology, Department of Pediatrics, Medical College of Wisconsin and Children’s Research Institute, Children’s Wisconsin, Wauwatosa, Wisconsin; and
| |
Collapse
|
20
|
Phoswa WN, Khaliq OP. The Role of Oxidative Stress in Hypertensive Disorders of Pregnancy (Preeclampsia, Gestational Hypertension) and Metabolic Disorder of Pregnancy (Gestational Diabetes Mellitus). OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5581570. [PMID: 34194606 PMCID: PMC8184326 DOI: 10.1155/2021/5581570] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 04/25/2021] [Indexed: 11/17/2022]
Abstract
Purpose of the Review.To highlight the role of oxidative stress in hypertensive disorders of pregnancy (HDP) and metabolic disorders of pregnancy (gestational diabetes mellitus). Recent Findings. In both preeclampsia (PE) and gestational hypertension (GH), oxidative stress leads to inadequate placental perfusion thus resulting in a hypoxic placenta, which generally leads to the activation of maternal systemic inflammatory response. In PE, this causes inflammation in the kidneys and leads to proteinuria. A proteinuria marker known as urinary 8-oxoGuo excretion is expressed in preeclampsia. In GDM, oxidative stress plays a role in the pathogenesis of the disease, as a result of over secretion of insulin during pregnancy. This uncontrolled secretion of insulin results in the production of lipid peroxidation factors that also mask the secretion of antioxidants. Therefore, ROS becomes abundant at cellular level and prevents the cells from transporting glucose to body tissues. Summary. There is a need for more research investigating the role of oxidative stress, especially in obstetrics-related conditions. More studies are required in order to understand the difference between the pathogenesis and pathophysiology of PE versus GH since investigations on the differences in genetic aspects of each condition are lacking. Furthermore, research to improve diagnostic procedures for GDM in pregnancy is needed.
Collapse
Affiliation(s)
- Wendy N. Phoswa
- Department of Life and Consumer Sciences, University of South Africa (UNISA), Science Campus, Private Bag X6, Florida, Roodepoort 1710, South Africa
| | - Olive P. Khaliq
- Department of Obstetrics and Gynaecology and Women's Health and HIV Research Group, Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
21
|
Stamerra CA, Del Pinto R, di Giosia P, Ferri C, Sahebkar A. Anderson-Fabry Disease: From Endothelial Dysfunction to Emerging Therapies. Adv Pharmacol Pharm Sci 2021; 2021:5548445. [PMID: 34095851 PMCID: PMC8137293 DOI: 10.1155/2021/5548445] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 05/07/2021] [Indexed: 12/30/2022] Open
Abstract
The Anderson-Fabry disease is a rare, X-linked, multisystemic, progressive lysosomal storage disease caused by α-galactosidase A total or partial deficiency. The resulting syndrome is mainly characterized by early-onset autonomic neuropathy and life-threatening multiorgan involvement, including renal insufficiency, heart disease, and early stroke. The enzyme deficiency leads to tissue accumulation of the glycosphingolipid globotriaosylceramide and its analogues, but the mechanisms linking such accumulation to organ damage are only partially understood. In contrast, enzyme replacement and chaperone therapies are already fully available to patients and allow substantial amelioration of quality and quantity of life. Substrate reduction, messenger ribonucleic acid (mRNA)-based, and gene therapies are also on the horizon. In this review, the clinical scenario and molecular aspects of Anderson-Fabry disease are described, along with updates on disease mechanisms and emerging therapies.
Collapse
Affiliation(s)
- Cosimo A. Stamerra
- University of L'Aquila, Department of Life, Health and Environmental Sciences, Building Delta 6-San Salvatore Hospital, Via Vetoio, Coppito, L'Aquila 67100, Italy
| | - Rita Del Pinto
- University of L'Aquila, Department of Life, Health and Environmental Sciences, Building Delta 6-San Salvatore Hospital, Via Vetoio, Coppito, L'Aquila 67100, Italy
| | - Paolo di Giosia
- University of L'Aquila, Department of Life, Health and Environmental Sciences, Building Delta 6-San Salvatore Hospital, Via Vetoio, Coppito, L'Aquila 67100, Italy
| | - Claudio Ferri
- University of L'Aquila, Department of Life, Health and Environmental Sciences, Building Delta 6-San Salvatore Hospital, Via Vetoio, Coppito, L'Aquila 67100, Italy
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
22
|
do Monte FA, Ahuja N, Awad KR, Pan Z, Young S, Kim HKW, Aswath P, Brotto M, Varanasi VG. Silicon Oxynitrophosphide Nanoscale Coating Enhances Antioxidant Marker-Induced Angiogenesis During in vivo Cranial Bone-Defect Healing. JBMR Plus 2021; 5:e10425. [PMID: 33869985 PMCID: PMC8046063 DOI: 10.1002/jbm4.10425] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 09/18/2020] [Accepted: 10/01/2020] [Indexed: 11/08/2022] Open
Abstract
Critical-sized bone defects are challenging to heal because of the sudden and large volume of lost bone. Fixative plates are often used to stabilize defects, yet oxidative stress and delayed angiogenesis are contributing factors to poor biocompatibility and delayed bone healing. This study tests the angiogenic and antioxidant properties of amorphous silicon oxynitrophosphide (SiONPx) nanoscale-coating material on endothelial cells to regenerate vascular tissue in vitro and in bone defects. in vitro studies evaluate the effect of silicon oxynitride (SiONx) and two different SiONPx compositions on human endothelial cells exposed to ROS (eg, hydrogen peroxide) that simulates oxidative stress conditions. in vivo studies using adult male Sprague Dawley rats (approximately 450 g) were performed to compare a bare plate, a SiONPx-coated implant plate, and a sham control group using a rat standard-sized calvarial defect. Results from this study showed that plates coated with SiONPx significantly reduced cell death, and enhanced vascular tubule formation and matrix deposition by upregulating angiogenic and antioxidant expression (eg, vascular endothelial growth factor A, angiopoetin-1, superoxide dismutase 1, nuclear factor erythroid 2-related factor 2, and catalase 1). Moreover, endothelial cell markers (CD31) showed a significant tubular structure in the SiONPx coating group compared with an empty and uncoated plate group. This reveals that atomic doping of phosphate into the nanoscale coating of SiONx produced markedly elevated levels of antioxidant and angiogenic markers that enhance vascular tissue regeneration. This study found that SiONPx or SiONx nanoscale-coated materials enhance antioxidant expression, angiogenic marker expression, and reduce ROS levels needed for accelerating vascular tissue regeneration. These results further suggest that SiONPx nanoscale coating could be a promising candidate for titanium plate for rapid and enhanced cranial bone-defect healing. © 2020 The Authors. JBMR Plus published by Wiley Periodicals LLC. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Felipe A do Monte
- Department of BioengineeringUniversity of Texas at ArlingtonArlingtonTXUSA
- Center for Excellence in Hip DisordersTexas Scottish Rite HospitalDallasTXUSA
| | - Neelam Ahuja
- Bone‐Muscle Research CenterUniversity of Texas at ArlingtonArlingtonTXUSA
| | - Kamal R Awad
- Bone‐Muscle Research CenterUniversity of Texas at ArlingtonArlingtonTXUSA
- Department of Materials Science and EngineeringUniversity of Texas at ArlingtonArlingtonTXUSA
| | - Zui Pan
- Bone‐Muscle Research CenterUniversity of Texas at ArlingtonArlingtonTXUSA
| | - Simon Young
- Department of Oral and Maxillofacial SurgeryThe University of Texas Health Science Center at Houston, School of DentistryHoustonTXUSA
| | - Harry KW Kim
- Center for Excellence in Hip DisordersTexas Scottish Rite HospitalDallasTXUSA
- Department of Orthopedic SurgeryUniversity of Texas Southwestern Medical Center at DallasDallasTXUSA
| | - Pranesh Aswath
- Department of Materials Science and EngineeringUniversity of Texas at ArlingtonArlingtonTXUSA
| | - Marco Brotto
- Bone‐Muscle Research CenterUniversity of Texas at ArlingtonArlingtonTXUSA
| | - Venu G Varanasi
- Bone‐Muscle Research CenterUniversity of Texas at ArlingtonArlingtonTXUSA
- Department of Materials Science and EngineeringUniversity of Texas at ArlingtonArlingtonTXUSA
| |
Collapse
|
23
|
De Melo N, Murrell L, Islam MT, Titman JJ, Macri-Pellizzeri L, Ahmed I, Sottile V. Tailoring Pyro-and Orthophosphate Species to Enhance Stem Cell Adhesion to Phosphate Glasses. Int J Mol Sci 2021; 22:ijms22020837. [PMID: 33467686 PMCID: PMC7829838 DOI: 10.3390/ijms22020837] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/11/2021] [Accepted: 01/13/2021] [Indexed: 12/29/2022] Open
Abstract
Phosphate-based glasses (PBGs) offer significant therapeutic potential due to their bioactivity, controllable compositions, and degradation rates. Several PBGs have already demonstrated their ability to support direct cell growth and in vivo cytocompatibility for bone repair applications. This study investigated development of PBG formulations with pyro- and orthophosphate species within the glass system (40 − x)P2O5·(16 + x)CaO·20Na2O·24MgO (x = 0, 5, 10 mol%) and their effect on stem cell adhesion properties. Substitution of phosphate for calcium revealed a gradual transition within the glass structure from Q2 to Q0 phosphate species. Human mesenchymal stem cells were cultured directly onto discs made from three PBG compositions. Analysis of cells seeded onto the discs revealed that PBG with higher concentration of pyro- and orthophosphate content (61% Q1 and 39% Q0) supported a 4.3-fold increase in adhered cells compared to glasses with metaphosphate connectivity (49% Q2 and 51% Q1). This study highlights that tuning the composition of PBGs to possess pyro- and orthophosphate species only, enables the possibility to control cell adhesion performance. PBGs with superior cell adhesion profiles represent ideal candidates for biomedical applications, where cell recruitment and support for tissue ingrowth are of critical importance for orthopaedic interventions.
Collapse
Affiliation(s)
- Nigel De Melo
- School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK; (N.D.M.); (L.M.-P.)
| | - Lauren Murrell
- Advanced Materials Research Group, Faculty of Engineering, University of Nottingham, Nottingham NG7 2RD, UK; (L.M.); (M.T.I.)
| | - Md Towhidul Islam
- Advanced Materials Research Group, Faculty of Engineering, University of Nottingham, Nottingham NG7 2RD, UK; (L.M.); (M.T.I.)
- Department of Applied Chemistry and Chemical Engineering, Faculty of Engineering, Noakhali Science and Technology University, Noakhali 3814, Bangladesh
| | - Jeremy J. Titman
- School of Chemistry, University of Nottingham, Nottingham NG7 2RD, UK;
| | - Laura Macri-Pellizzeri
- School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK; (N.D.M.); (L.M.-P.)
| | - Ifty Ahmed
- Advanced Materials Research Group, Faculty of Engineering, University of Nottingham, Nottingham NG7 2RD, UK; (L.M.); (M.T.I.)
- Correspondence: (I.A.); (V.S.)
| | - Virginie Sottile
- School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK; (N.D.M.); (L.M.-P.)
- Department of Molecular Medicine, The University of Pavia, 27100 Pavia, Italy
- Correspondence: (I.A.); (V.S.)
| |
Collapse
|
24
|
Abstract
Endothelial cell (EC) metabolism is important for health and disease. Metabolic pathways, such as glycolysis, fatty acid oxidation, and amino acid metabolism, determine vasculature formation. These metabolic pathways have different roles in securing the production of energy and biomass and the maintenance of redox homeostasis in vascular migratory tip cells, proliferating stalk cells, and quiescent phalanx cells, respectively. Emerging evidence demonstrates that perturbation of EC metabolism results in EC dysfunction and vascular pathologies. Here, we summarize recent insights into EC metabolic pathways and their deregulation in vascular diseases. We further discuss the therapeutic implications of targeting EC metabolism in various pathologies.
Collapse
Affiliation(s)
- Xuri Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China; ,
| | - Anil Kumar
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China; ,
| | - Peter Carmeliet
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China; , .,Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven B-3000, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven B-3000, Belgium
| |
Collapse
|
25
|
Kim DH, Meza CA, Clarke H, Kim JS, Hickner RC. Vitamin D and Endothelial Function. Nutrients 2020; 12:E575. [PMID: 32098418 PMCID: PMC7071424 DOI: 10.3390/nu12020575] [Citation(s) in RCA: 185] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 02/19/2020] [Accepted: 02/19/2020] [Indexed: 12/21/2022] Open
Abstract
Vitamin D is known to elicit a vasoprotective effect, while vitamin D deficiency is a risk factor for endothelial dysfunction (ED). ED is characterized by reduced bioavailability of a potent endothelium-dependent vasodilator, nitric oxide (NO), and is an early event in the development of atherosclerosis. In endothelial cells, vitamin D regulates NO synthesis by mediating the activity of the endothelial NO synthase (eNOS). Under pathogenic conditions, the oxidative stress caused by excessive production of reactive oxygen species (ROS) facilitates NO degradation and suppresses NO synthesis, consequently reducing NO bioavailability. Vitamin D, however, counteracts the activity of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase which produces ROS, and improves antioxidant capacity by enhancing the activity of antioxidative enzymes such as superoxide dismutase. In addition to ROS, proinflammatory mediators such as TNF-α and IL-6 are risk factors for ED, restraining NO and eNOS bioactivity and upregulating the expression of various atherosclerotic factors through the NF-κB pathway. These proinflammatory activities are inhibited by vitamin D by suppressing NF-κB signaling and production of proinflammatory cytokines. In this review, we discuss the diverse activities of vitamin D in regulating NO bioavailability and endothelial function.
Collapse
Affiliation(s)
- Do-Houn Kim
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL 32306, USA; (D.-H.K.); (C.A.M.); (H.C.); (J.-S.K.)
- Center for Advancing Exercise and Nutrition Research on Aging, Florida State University, Tallahassee, FL 32306, USA
| | - Cesar A. Meza
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL 32306, USA; (D.-H.K.); (C.A.M.); (H.C.); (J.-S.K.)
| | - Holly Clarke
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL 32306, USA; (D.-H.K.); (C.A.M.); (H.C.); (J.-S.K.)
| | - Jeong-Su Kim
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL 32306, USA; (D.-H.K.); (C.A.M.); (H.C.); (J.-S.K.)
- Center for Advancing Exercise and Nutrition Research on Aging, Florida State University, Tallahassee, FL 32306, USA
- Institute of Sports Sciences and Medicine, College of Human Sciences, Florida State University, Tallahassee, FL 32306, USA
| | - Robert C. Hickner
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL 32306, USA; (D.-H.K.); (C.A.M.); (H.C.); (J.-S.K.)
- Institute of Sports Sciences and Medicine, College of Human Sciences, Florida State University, Tallahassee, FL 32306, USA
- Department of Biokinetics, Exercise and Leisure Sciences, School of Health Sciences, University of KwaZulu-Natal, Westville 4041, South Africa
| |
Collapse
|
26
|
Hulin JA, Gubareva EA, Jarzebska N, Rodionov RN, Mangoni AA, Tommasi S. Inhibition of Dimethylarginine Dimethylaminohydrolase (DDAH) Enzymes as an Emerging Therapeutic Strategy to Target Angiogenesis and Vasculogenic Mimicry in Cancer. Front Oncol 2020; 9:1455. [PMID: 31993367 PMCID: PMC6962312 DOI: 10.3389/fonc.2019.01455] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 12/05/2019] [Indexed: 01/01/2023] Open
Abstract
The small free radical gas nitric oxide (NO) plays a key role in various physiological and pathological processes through enhancement of endothelial cell survival and proliferation. In particular, NO has emerged as a molecule of interest in carcinogenesis and tumor progression due to its crucial role in various cancer-related events including cell invasion, metastasis, and angiogenesis. The dimethylarginine dimethylaminohydrolase (DDAH) family of enzymes metabolize the endogenous nitric oxide synthase (NOS) inhibitors, asymmetric dimethylarginine (ADMA) and monomethyl arginine (L-NMMA), and are thus key for maintaining homeostatic control of NO. Dysregulation of the DDAH/ADMA/NO pathway resulting in increased local NO availability often promotes tumor growth, angiogenesis, and vasculogenic mimicry. Recent literature has demonstrated increased DDAH expression in tumors of different origins and has also suggested a potential ADMA-independent role for DDAH enzymes in addition to their well-studied ADMA-mediated influence on NO. Inhibition of DDAH expression and/or activity in cell culture models and in vivo studies has indicated the potential therapeutic benefit of this pathway through inhibition of both angiogenesis and vasculogenic mimicry, and strategies for manipulating DDAH function in cancer are currently being actively pursued by several research groups. This review will thus provide a timely discussion on the expression, regulation, and function of DDAH enzymes in regard to angiogenesis and vasculogenic mimicry, and will offer insight into the therapeutic potential of DDAH inhibition in cancer based on preclinical studies.
Collapse
Affiliation(s)
- Julie-Ann Hulin
- Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Ekaterina A Gubareva
- N.N. Petrov National Medical Research Center of Oncology, Saint Petersburg, Russia
| | - Natalia Jarzebska
- Division of Angiology, Department of Internal Medicine III, University Center for Vascular Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,Department of Anesthesiology and Intensive Care Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Roman N Rodionov
- Division of Angiology, Department of Internal Medicine III, University Center for Vascular Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Arduino A Mangoni
- Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Sara Tommasi
- Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| |
Collapse
|
27
|
do Monte FA, Awad KR, Ahuja N, Kim HK, Aswath P, Brotto M, Varanasi VG. Amorphous Silicon Oxynitrophosphide-Coated Implants Boost Angiogenic Activity of Endothelial Cells. Tissue Eng Part A 2020; 26:15-27. [PMID: 31044666 PMCID: PMC6983748 DOI: 10.1089/ten.tea.2019.0051] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 04/29/2019] [Indexed: 12/29/2022] Open
Abstract
Lack of osteointegration is a major cause of aseptic loosening and failure of implants used in bone replacement. Implants coated with angiogenic biomaterials can improve osteointegration and potentially reduce these complications. Silicon- and phosphorus-based materials have been shown to upregulate expression of angiogenic factors and improve endothelial cell functions. In the present study, we hypothesize that implants coated with amorphous silica-based coatings in the form of silicon oxynitrophosphide (SiONP) by using plasma-enhanced chemical vapor deposition (PECVD) technique could enhance human umbilical vein endothelial cell angiogenic properties in vitro. The tested groups were: glass coverslip (GCS), tissue culture plate, SiON, SiONP1 (O: 7.3 at %), and SiONP2 (O: 14.2 at %) implants. The SiONP2 composition demonstrated 3.5-fold more fibronectin deposition than the GCS (p < 0.001). The SiONP2 group also presented a significant improvement in the capillary tubule length and thickness compared with the other groups (p < 0.01). At 24 h, we observed at least a twofold upregulation of vascular endothelial growth factor A, hypoxia-inducible factor-1α, angiopoietin-1, and nesprin-2, more evident in the SiONP1 and SiONP2 groups. In conclusion, the studied amorphous silica-coated implants, especially the SiONP2 composition, could enhance the endothelial cell angiogenic properties in vitro and may induce faster osteointegration and healing. Impact Statement In this study, we report for the first time the significant enhancement of human umbilical vein endothelial cell angiogenic properties (in vitro) by the amorphous silica-based coatings in the form of silicon oxynitrophosphide (SiONP). The SiONP2 demonstrated 3.5-fold more fibronectin deposition than the glass coverslip and presented a significant improvement in the capillary tubule length and thickness. At 24 h, SiONP reported twofold upregulation of vascular endothelial growth factor A, hypoxia-inducible factor-1α, angiopoietin-1, and nesprin-2. The studied amorphous silica-coated implants enhance the endothelial cell angiogenic properties in vitro and may induce faster osteointegration and healing.
Collapse
Affiliation(s)
- Felipe A. do Monte
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas
- Center for Excellence in Hip Disorders, Texas Scottish Rite Hospital, Dallas, Texas
| | - Kamal R. Awad
- Department of Materials Science and Engineering, University of Texas at Arlington, Arlington, Texas
- Bone-Muscle Research Center, College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, Texas
| | - Neelam Ahuja
- Bone-Muscle Research Center, College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, Texas
| | - Harry K.W. Kim
- Center for Excellence in Hip Disorders, Texas Scottish Rite Hospital, Dallas, Texas
- Department of Orthopedic Surgery, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas
| | - Pranesh Aswath
- Department of Materials Science and Engineering, University of Texas at Arlington, Arlington, Texas
| | - Marco Brotto
- Bone-Muscle Research Center, College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, Texas
| | - Venu G. Varanasi
- Department of Materials Science and Engineering, University of Texas at Arlington, Arlington, Texas
- Bone-Muscle Research Center, College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, Texas
| |
Collapse
|
28
|
RIBEIRO DV, FONSECA LDS, MOREIRA RHR, CHAIN CP, ABREU MLT, DALÓLIO FS, OLIVEIRA AF, GARBOSSA CAP. The economic viability of L-arginine supplementation in diets for sows in the lactation phase. REVISTA BRASILEIRA DE SAÚDE E PRODUÇÃO ANIMAL 2020. [DOI: 10.1590/s1519-9940210412020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
ABSTRACT The objective of this study was to evaluate the economic viability of L-arginine supplementation in diets for sows during the lactation phase. A descriptive case study was carried out to identify the economic viability of the supplementation of L-arginine in the lactation ration, in relation to the control diet, on the productive performance of hyper prolific sows and their litters. The study was carried out on a commercial farm located in the state of Minas Gerais. It analyzed feasibility criteria such as the marginal physical product (MPPg) and gross margin (GM) in the scenario of technical and economic efficiency, which were compared to the GM in the control group. Technical efficiency was established at 0.71% of L-arginine in daily feed intake according to previous fieldwork by the authors, while economic efficiency obtained in this study was 0.43% supplementation. The relationship between the L-arginine price and the sale price of the piglets directly determined the economic viability, compared to the control group. Thus, initially considering the price of a piglet as R$ 9.76/kg, the cost of L-arginine should not exceed 6.61 times this value; that is, it is estimated at up to R$ 64.50. Therefore, at the final price of L-arginine of R$ 54.88, a piglet should be sold at a price of R$ 8.30/kg to make the supplementation of L-arginine in the diet of lactation-phase swine economically possible.
Collapse
|
29
|
Stam K, Cai Z, van der Velde N, van Duin R, Lam E, van der Velden J, Hirsch A, Duncker DJ, Merkus D. Cardiac remodelling in a swine model of chronic thromboembolic pulmonary hypertension: comparison of right vs. left ventricle. J Physiol 2019; 597:4465-4480. [PMID: 31194256 PMCID: PMC6852085 DOI: 10.1113/jp277896] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 06/07/2019] [Indexed: 12/11/2022] Open
Abstract
KEY POINTS Right ventricle (RV) function is the most important determinant of survival and quality of life in patients with chronic thromboembolic pulmonary hypertension (CTEPH). The changes in right and left ventricle gene expression that contribute to ventricular remodelling are incompletely investigated. RV remodelling in our CTEPH swine model is associated with increased expression of the genes involved in inflammation (TGFβ), oxidative stress (ROCK2, NOX1 and NOX4), and apoptosis (BCL2 and caspase-3). Alterations in ROCK2 expression correlated inversely with RV contractile reserve during exercise. Since ROCK2 has been shown to be involved in hypertrophy, oxidative stress, fibrosis and endothelial dysfunction, ROCK2 inhibition may present a viable therapeutic target in CTEPH. ABSTRACT Right ventricle (RV) function is the most important determinant of survival and quality of life in patients with chronic thromboembolic pulmonary hypertension (CTEPH). The present study investigated whether the increased cardiac afterload is associated with (i) cardiac remodelling and hypertrophic signalling; (ii) changes in angiogenic factors and capillary density; and (iii) inflammatory changes associated with oxidative stress and interstitial fibrosis. CTEPH was induced in eight chronically instrumented swine by chronic nitric oxide synthase inhibition and up to five weekly pulmonary embolizations. Nine healthy swine served as a control. After 9 weeks, RV function was assessed by single beat analysis of RV-pulmonary artery (PA) coupling at rest and during exercise, as well as by cardiac magnetic resonance imaging. Subsequently, the heart was excised and RV and left ventricle (LV) tissues were processed for molecular and histological analyses. Swine with CTEPH exhibited significant RV hypertrophy in response to the elevated PA pressure. RV-PA coupling was significantly reduced, correlated inversely with pulmonary vascular resistance and did not increase during exercise in CTEPH swine. Expression of genes associated with hypertrophy (BNP), inflammation (TGFβ), oxidative stress (ROCK2, NOX1 and NOX4), apoptosis (BCL2 and caspase-3) and angiogenesis (VEGFA) were increased in the RV of CTEPH swine and correlated inversely with RV-PA coupling during exercise. In the LV, only significant changes in ROCK2 gene-expression occurred. In conclusion, RV remodelling in our CTEPH swine model is associated with increased expression of genes involved in inflammation and oxidative stress, suggesting that these processes contribute to RV remodelling and dysfunction in CTEPH and hence represent potential therapeutic targets.
Collapse
Affiliation(s)
- Kelly Stam
- Department of Cardiology, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Zongye Cai
- Department of Cardiology, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Nikki van der Velde
- Department of Cardiology, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Richard van Duin
- Department of Cardiology, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Esther Lam
- Department of Cardiology, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Jolanda van der Velden
- Amsterdam UMCVrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular SciencesAmsterdamThe Netherlands
| | - Alexander Hirsch
- Department of Cardiology, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Dirk J Duncker
- Department of Cardiology, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Daphne Merkus
- Department of Cardiology, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| |
Collapse
|
30
|
Stanisic J, Koricanac G, Kostic M, Stojiljkovic M, Culafic T, Romic S, Tepavcevic S. Low-intensity exercise in the prevention of cardiac insulin resistance-related inflammation and disturbances in NOS and MMP-9 regulation in fructose-fed ovariectomized rats. Appl Physiol Nutr Metab 2019; 44:1219-1229. [PMID: 30897341 DOI: 10.1139/apnm-2018-0785] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Exercise is important nonpharmacological treatment for improvement of insulin sensitivity in menopause. However, its effect on menopausal cardiac insulin resistance is needing further research. We investigated protective effects of low-intensity exercise on cardiac insulin signaling, inflammation, regulation of nitric oxide synthase (NOS) and matrix metalloproteinase 9 (MMP-9) in ovariectomized (OVX) Wistar rats, submitted to 10% fructose solution for 9 weeks. OVX rats were divided into control, sedentary fructose, and exercise fructose groups. Measurements of physical and biochemical characteristics were carried out to evaluate metabolic syndrome development. Messenger RNA and protein levels and phosphorylation of cardiac insulin signaling molecules, endothelial and inducible NOS (eNOS and iNOS), p65 subunit of nuclear factor κB (NFκB), tumor necrosis factor α (TNF-α), suppressor of cytokine signaling 3 (SOCS3), and MMP-9 were analyzed. Fructose increased insulin level, homeostasis model assessment (HOMA) index, and visceral adipose tissue weight, while low-intensity exercise prevented insulin level and HOMA index increase. Fructose also decreased cardiac pAkt (Ser473), peNOS (Ser1177) and increased insulin receptor substrate 1 (IRS1) phosphorylation at Ser307, pNFκB (Ser276) and NFκB and MMP-9 content, without any effect on iNOS, protein-tyrosine phosphatase 1B, TNF-α, and SOCS3. Exercise prevented changes in pIRS1 (Ser307), pAkt (Ser473), peNOS (Ser1177), pNFκB (Ser276), and NFκB expression. In addition, exercise increased pIRS1 (Tyr632), pAkt (Thr308), and eNOS expression. Low-intensity exercise prevented cardiac insulin signaling disarrangement in fructose-fed OVX rats and therefore eNOS dysfunction, as well as pro-inflammatory signaling activation, without effect on tissue remodeling, suggesting physical training as a way to reduce cardiovascular risk.
Collapse
Affiliation(s)
- Jelena Stanisic
- Laboratory for Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Belgrade, Republic of Serbia.,Laboratory for Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Belgrade, Republic of Serbia
| | - Goran Koricanac
- Laboratory for Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Belgrade, Republic of Serbia.,Laboratory for Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Belgrade, Republic of Serbia
| | - Milan Kostic
- Laboratory for Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Belgrade, Republic of Serbia.,Laboratory for Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Belgrade, Republic of Serbia
| | - Mojca Stojiljkovic
- Laboratory for Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Belgrade, Republic of Serbia.,Laboratory for Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Belgrade, Republic of Serbia
| | - Tijana Culafic
- Laboratory for Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Belgrade, Republic of Serbia.,Laboratory for Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Belgrade, Republic of Serbia
| | - Snjezana Romic
- Laboratory for Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Belgrade, Republic of Serbia.,Laboratory for Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Belgrade, Republic of Serbia
| | - Snezana Tepavcevic
- Laboratory for Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Belgrade, Republic of Serbia.,Laboratory for Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Belgrade, Republic of Serbia
| |
Collapse
|
31
|
Teuwen LA, Geldhof V, Carmeliet P. How glucose, glutamine and fatty acid metabolism shape blood and lymph vessel development. Dev Biol 2019; 447:90-102. [DOI: 10.1016/j.ydbio.2017.12.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 10/26/2017] [Accepted: 12/01/2017] [Indexed: 12/18/2022]
|
32
|
Monte F, Cebe T, Ripperger D, Ighani F, Kojouharov HV, Chen BM, Kim HKW, Aswath PB, Varanasi VG. Ionic silicon improves endothelial cells' survival under toxic oxidative stress by overexpressing angiogenic markers and antioxidant enzymes. J Tissue Eng Regen Med 2018; 12:2203-2220. [PMID: 30062712 PMCID: PMC6508967 DOI: 10.1002/term.2744] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 06/11/2018] [Accepted: 07/17/2018] [Indexed: 11/10/2022]
Abstract
Oxidative stress, induced by harmful levels of reactive oxygen species, is a common occurrence that impairs proper bone defect vascular healing through the impairment of endothelial cell function. Ionic silicon released from silica-based biomaterials, can upregulate hypoxia-inducible factor-1α (HIF-1α). Yet it is unclear whether ionic Si can restore endothelial cell function under oxidative stress conditions. Therefore, we hypothesized that ionic silicon can help improve human umbilical vein endothelial cells' (HUVECs') survival under toxic oxidative stress. In this study, we evaluated the ionic jsilicon effect on HUVECs viability, proliferation, migration, gene expression, and capillary tube formation under normal conditions and under harmful hydrogen peroxide levels. We demonstrated that 0.5-mM Si4+ significantly enhanced angiogenesis in HUVECs under normal condition (p < 0.05). HUVECs exposed to 0.5-mM Si4+ presented a morphological change, even without the bed of Matrigel, and formed significantly more tube-like structures than the control (p < 0.001). In addition, 0.5-mM Si4+ enhanced cell viability in HUVECs under harmful H2 O2 levels. HIF-1α, vascular endothelial growth factor-A, and vascular endothelial growth factor receptor-2 were overexpressed more than twofold in silicon-treated HUVECs, under normal and toxic H2 O2 conditions. Moreover, the HUVECs were treated with 0.5-mM Si4+ overexpressed superoxide dismutase-1 (SOD-1), catalase-1 (Cat-1), and nitric oxide synthase-3 (NOS3) under normal and oxidative stress environment (p < 0.01). A computational model was used for explaining the antioxidant effect of Si4+ in endothelial cells and human periosteum cells by SOD-1 enhancement. In conclusion, we demonstrated that 0.5-mM Si4+ can recover the HUVECs' viability under oxidative stress conditions by reducing cell death and upregulating expression of angiogenic and antioxidant factors.
Collapse
Affiliation(s)
- Felipe Monte
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas
- Center for Excellence in Hip Disorders, Texas Scottish Rite Hospital, Dallas, Texas
| | - Tugba Cebe
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas
| | | | - Fareed Ighani
- Texas A&M University College of Dentistry, Dallas, Texas
| | | | - Benito M. Chen
- Department of Mathematics, University of Texas at Arlington, Arlington, Texas
| | - Harry K. W. Kim
- Center for Excellence in Hip Disorders, Texas Scottish Rite Hospital, Dallas, Texas
- Department of Orthopedic Surgery, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas
| | - Pranesh B. Aswath
- Department of Materials Science and Engineering, University of Texas at Arlington, Arlington, Texas
| | - Venu G. Varanasi
- Department of Materials Science and Engineering, University of Texas at Arlington, Arlington, Texas
- Department of College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, Texas
| |
Collapse
|
33
|
Loso J, Lund N, Avanesov M, Muschol N, Lezius S, Cordts K, Schwedhelm E, Patten M. Serum Biomarkers of Endothelial Dysfunction in Fabry Associated Cardiomyopathy. Front Cardiovasc Med 2018; 5:108. [PMID: 30159316 PMCID: PMC6104487 DOI: 10.3389/fcvm.2018.00108] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 07/17/2018] [Indexed: 12/13/2022] Open
Abstract
Background: Fabry disease (FD) is characterized by early development of vasculopathy and endothelial dysfunction. However, it is unclear whether these findings also play a pivotal role in cardiac manifestation. As Fabry cardiomyopathy (FC) is the leading cause of death in FD, we aimed to gather a better insight in pathological mechanisms of the disease. Methods: Serum samples were obtained from 17 healthy controls, 15 FD patients with and 7 without FC. FC was defined by LV wall thickening of >12 mm in cardiac magnetic resonance imaging and serum level of proBNP, high sensitive Troponin T (hsT), and globotriaosylsphingosine (lyso-GB3) were obtained. A multiplex ELISA-Assay for 23 different angiogenesis markers was performed in pooled samples. Markers showing significant differences among groups were further analyzed in single samples using specific Elisa antibody assays. L-homoarginine (hArg), L-arginine, asymmetric (ADMA), and symmetric Dimethylarginine (SDMA) were quantified by liquid chromatography—mass spectrometry. Results: Angiostatin and matrix metalloproteinase 9 (MMP-9) were elevated in FD patients compared to controls independently of the presence of FC (angiostatin: 98 ± 25 vs. 75 ± 15 ng/mL; p = 0.001; MMP-9: 8.0 ± 3.4 vs. 5.0 ± 2.4 μg/mL; p = 0.002). SDMA concentrations were highest in patients with FC (0.90 ± 0.64 μmol/l) compared to patients without (0.57 ± 0.10 μmol/l; p = 0.027) and vs. controls (0.58 ± 0.12 μmol/l; p = 0.006) and was positively correlated with indexed LV-mass (r = 0.61; p = 0.003), hsT (r = 0.56, p = 0.008), and lyso-Gb3 (r = 0.53, p = 0.013). Accordingly, the ratio of L-homoarginine to SDMA (hArg/SDMA) was lowest in patients with FC (2.63 ± 1.78) compared to controls (4.16 ± 1.44; p = 0.005). For L-arginine, hArg and ADMA no significant differences among groups could be detected, although a trend toward higher ADMA and lower hArg levels could be observed in the FC group. Furthermore, a significant relationship between kidney and cardiac function could be revealed (p = 0.045). Conclusion: Elevated MMP-9 and angiostatin levels suggest an increased extracellular matrix turnover in FD patients. Furthermore, endothelial dysfunction may also be involved in FC, as SDMA and hArg/SDMA are altered in these patients.
Collapse
Affiliation(s)
- Jefferson Loso
- Department of General and Interventional Cardiology, University Heart Center Hamburg, Hamburg, Germany
| | - Natalie Lund
- Department of General and Interventional Cardiology, University Heart Center Hamburg, Hamburg, Germany
| | - Maxim Avanesov
- Department of Diagnostic and Interventional Radiology, University Medical Center Hamburg- Eppendorf, Hamburg, Germany
| | - Nicole Muschol
- Department of Pediatrics, University Medical Center Hamburg- Eppendorf, Hamburg, Germany
| | - Susanne Lezius
- Department of Medical Biometry and Epidemiology, University Medical Center Hamburg- Eppendorf, Hamburg, Germany
| | - Kathrin Cordts
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg- Eppendorf, Hamburg, Germany.,DZHK (German Center for Cardiovascular Research e.V.), Hamburg, Germany
| | - Edzard Schwedhelm
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg- Eppendorf, Hamburg, Germany.,DZHK (German Center for Cardiovascular Research e.V.), Hamburg, Germany
| | - Monica Patten
- Department of General and Interventional Cardiology, University Heart Center Hamburg, Hamburg, Germany.,DZHK (German Center for Cardiovascular Research e.V.), Hamburg, Germany
| |
Collapse
|
34
|
Draoui N, de Zeeuw P, Carmeliet P. Angiogenesis revisited from a metabolic perspective: role and therapeutic implications of endothelial cell metabolism. Open Biol 2018; 7:rsob.170219. [PMID: 29263247 PMCID: PMC5746547 DOI: 10.1098/rsob.170219] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 11/28/2017] [Indexed: 12/16/2022] Open
Abstract
Endothelial cell (EC) metabolism has lately emerged as a novel and promising therapeutic target to block vascular dysregulation associated with diseases like cancer and blinding eye disease. Glycolysis, fatty acid oxidation (FAO) and, more recently, glutamine/asparagine metabolism emerged as key regulators of EC metabolism, able to impact angiogenesis in health and disease. ECs are highly glycolytic as they require ATP and biomass for vessel sprouting. Notably, a regulator of the glycolytic pathway, 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3, controls vessel sprouting during the angiogenic switch and its inhibition in tumour ECs leads to vessel normalization, thereby reducing metastasis and ameliorating chemotherapy. Moreover, FAO promotes EC proliferation through DNA synthesis, and plays an essential role in lymphangiogenesis via epigenetic regulation of histone acetylation. Pathological angiogenesis was decreased upon blockade of carnitine palmitoyltransferase 1, a regulator of FAO in ECs. More recently, metabolism of glutamine, in conjunction with asparagine, was reported to maintain EC sprouting through TCA anaplerosis, redox homeostasis, mTOR activation and endoplasmic stress control. Inactivation or blockade of glutaminase 1, which hydrolyses glutamine into ammonia and glutamate, impairs angiogenesis in health and disease, while silencing of asparagine synthetase reduces vessel sprouting in vitro. In this review, we summarize recent insights into EC metabolism and discuss therapeutic implications of targeting EC metabolism.
Collapse
Affiliation(s)
- Nihed Draoui
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven 3000, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, KU Leuven, Campus Gasthuisberg O&N4, Herestraat 49-912, Leuven 3000, Belgium
| | - Pauline de Zeeuw
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven 3000, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, KU Leuven, Campus Gasthuisberg O&N4, Herestraat 49-912, Leuven 3000, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven 3000, Belgium .,Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, KU Leuven, Campus Gasthuisberg O&N4, Herestraat 49-912, Leuven 3000, Belgium
| |
Collapse
|
35
|
Wang YB, Li Y, Deng YB, Liu YN, Zhang J, Sun J, Zhu Y, Li L, Tang QY, Zhou W. Enlarged Size and Impaired Elastic Properties of the Ascending Aorta are Associated with Endothelial Dysfunction and Elevated Plasma Matrix Metalloproteinase-2 Level in Patients with Bicuspid Aortic Valve. ULTRASOUND IN MEDICINE & BIOLOGY 2018; 44:955-962. [PMID: 29472114 DOI: 10.1016/j.ultrasmedbio.2018.01.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 11/17/2017] [Accepted: 01/12/2018] [Indexed: 06/08/2023]
Abstract
The aim of this study was to test whether enlarged size and impaired elastic properties of the ascending aorta are associated with impaired endothelial function and increases in plasma matrix metalloproteinase (MMP)-2 concentrations in patients with bicuspid aortic valve (BAV) without significant valvular dysfunction. The size and the elasticity of the ascending aorta and the flow-mediated vasodilation (FMD) in the brachial artery in response to hyperemia were evaluated with 2-D echocardiography and high-frequency linear ultrasound in 42 patients with BAV without significant valvular dysfunction and 30 age- and sex-matched healthy controls. In the BAV group, diastolic ascending aortic diameter (AoD) (32.1 ± 8.1 mm vs. 25.3 ± 3.6 mm, p <0.001) and aortic stiffness index (8.0 ± 5.3 vs. 4.0 ± 1.8, p <0.001) were significantly higher, and aortic strain (7.4 ± 3.6% vs. 11.1 ± 3.0%, p <0.001) and aortic distensibility (7.4 ± 4.1 × 10-6cm2/dyn vs. 11.1 ± 4.3 × 10-6cm2/dyn, p <0.001) were significantly lower than those in the control group. The BAV group also had lower FMD (6.5 ± 2.2% vs. 11.9 ± 2.7%, p <0.001) and higher plasma MMP-2 levels (226.7 ± 55.0 ng/mL vs. 177.0 ± 45.3 ng/mL, p <0.001) compared with the control group. In the BAV group, AoD, aortic strain, aortic stiffness index and aortic distensibility significantly correlated with FMD and MMP-2 (all p <0.05). The multivariable linear regression analysis further indicated that FMD and MMP-2 were independently associated with AoD (β = -1.1, p = 0.005, and β = 0.09, p <0.001, respectively). These findings suggest that enlarged size and impaired elastic properties of the ascending aorta are associated with endothelial dysfunction and elevated plasma MMP-2 level in patients with BAV without significant valvular dysfunction. FMD and plasma MMP-2 level are the significant and independent predictors of dilation of the ascending aorta in patients with BAV.
Collapse
Affiliation(s)
- Yi-Bin Wang
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Li
- Department of Medical Ultrasound, the First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - You-Bin Deng
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Ya-Ni Liu
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Zhang
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Sun
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Zhu
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Li
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiao-Ying Tang
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Zhou
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
36
|
Abstract
Receptor signaling relays on intracellular events amplified by secondary and tertiary messenger molecules. In cardiomyocytes and smooth muscle cells, cyclic AMP (cAMP) and subsequent calcium (Ca2+) fluxes are the best characterized receptor-regulated signaling events. However, most of receptors able to modify contractility and other intracellular responses signal through a variety of other messengers, and whether these signaling events are interconnected has long remained unclear. For example, the PI3K (phosphoinositide 3-kinase) pathway connected to the production of the lipid second messenger PIP3/PtdIns(3,4,5)P3 (phosphatidylinositol (3,4,5)-trisphosphate) is potentially involved in metabolic regulation, activation of hypertrophy, and survival pathways. Recent studies, highlighted in this review, started to interconnect PI3K pathway activation to Ca2+ signaling. This interdependency, by balancing contractility with metabolic control, is crucial for cells of the cardiovascular system and is emerging to play key roles in disease development. Better understanding of the interplay between Ca2+ and PI3K signaling is, thus, expected to provide new ground for therapeutic intervention. This review explores the emerging molecular mechanisms linking Ca2+ and PI3K signaling in health and disease.
Collapse
Affiliation(s)
- Alessandra Ghigo
- From the Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Italy (A.G., M. Li, E.H.); and INSERM U1048, I2MC and Université Toulouse III, France (M. Laffargue)
| | - Muriel Laffargue
- From the Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Italy (A.G., M. Li, E.H.); and INSERM U1048, I2MC and Université Toulouse III, France (M. Laffargue)
| | - Mingchuan Li
- From the Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Italy (A.G., M. Li, E.H.); and INSERM U1048, I2MC and Université Toulouse III, France (M. Laffargue)
| | - Emilio Hirsch
- From the Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Italy (A.G., M. Li, E.H.); and INSERM U1048, I2MC and Université Toulouse III, France (M. Laffargue).
| |
Collapse
|
37
|
Rationale and design of DUAL study: Doxycycline to Upgrade response in light chain (AL) amyloidosis (DUAL): A phase 2 pilot study of a two-pronged approach of prolonged doxycycline with plasma cell-directed therapy in the treatment of AL amyloidosis. Contemp Clin Trials Commun 2017; 8:33-38. [PMID: 29696194 PMCID: PMC5898504 DOI: 10.1016/j.conctc.2017.08.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Revised: 08/15/2017] [Accepted: 08/16/2017] [Indexed: 12/19/2022] Open
Abstract
Light chain (AL) amyloidosis is a plasma cell neoplasm associated with insoluble fibril deposition from clonal immunoglobulin chains systemically. The disease is associated with high early mortality and morbidity owing to advanced organ deposition as well as lack of proven de-fibrillogenic therapies. Pre-clinical and retrospective clinical data suggests that doxycycline has benefit in AL amyloidosis. The ongoing DUAL study is a single center, open label, phase 2 study in which patients with AL amyloidosis who are undergoing clone-directed therapy for the underlying neoplasm with oral doxycycline given for 1 year to test the hypothesis that prolonged doxycycline use will be safe, feasible, and lead to reduced early mortality in systemic AL amyloidosis and hasten organ amyloid response. Clinical follow up visits will occur at monthly intervals for systemic AL patients and at 3 monthly intervals for localized AL patients. Blood tests will be collected during these time points for hematologic response assessment. Organ testing will be conducted at 3 monthly intervals and radiologic testing will be conducted at 6 monthly intervals. Research blood samples will be collected at baseline, 6 and 12 months. Other correlative studies include matrix metalloproteinases (MMP), tissue inhibitor of metalloproteinases (TIMP) testing and patient-reported outcomes.
Collapse
|
38
|
Zhang Y, Liu NM, Wang Y, Youn JY, Cai H. Endothelial cell calpain as a critical modulator of angiogenesis. Biochim Biophys Acta Mol Basis Dis 2017; 1863:1326-1335. [PMID: 28366876 DOI: 10.1016/j.bbadis.2017.03.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 03/04/2017] [Accepted: 03/28/2017] [Indexed: 12/13/2022]
Abstract
Calpains are a family of calcium-dependent non-lysosomal cysteine proteases. In particular, calpains residing in the endothelial cells play important roles in angiogenesis. It has been shown that calpain activity can be increased in endothelial cells by growth factors, primarily vascular endothelial growth factor (VEGF). VEGF/VEGFR2 induces calpain 2 dependent activation of PI3K/AMPK/Akt/eNOS pathway, and consequent nitric oxide production and physiological angiogenesis. Under pathological conditions such as tumor angiogenesis, endothelial calpains can be activated by hypoxia. This review focuses on the molecular regulatory mechanisms of calpain activation, and the newly identified mechanistic roles and downstream signaling events of calpains in physiological angiogenesis, and in the conditions of pathological tumor angiogenesis and diabetic wound healing, as well as retinopathy and atherosclerosis that are also associated with an increase in calpain activity. Further discussed include the differential strategies of modulating angiogenesis through manipulating calpain expression/activity in different pathological settings. Targeted limitation of angiogenesis in cancer and targeted promotion of angiogenesis in diabetic wound healing via modulations of calpains and calpain-dependent signaling mechanisms are of significant translational potential. Emerging strategies of tissue-specific targeting, environment-dependent targeting, and genome-targeted editing may turn out to be effective regimens for targeted manipulation of angiogenesis through calpain pathways, for differential treatments including both attenuation of tumor angiogenesis and potentiation of diabetic angiogenesis.
Collapse
Affiliation(s)
- Yixuan Zhang
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA; Division of Cardiology, Department Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA
| | - Norika Mengchia Liu
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA; Division of Cardiology, Department Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA
| | - Yongchen Wang
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA; Division of Cardiology, Department Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA
| | - Ji Youn Youn
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA; Division of Cardiology, Department Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA
| | - Hua Cai
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA; Division of Cardiology, Department Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA.
| |
Collapse
|
39
|
Abstract
The heart is uniquely responsible for providing its own blood supply through the coronary circulation. Regulation of coronary blood flow is quite complex and, after over 100 years of dedicated research, is understood to be dictated through multiple mechanisms that include extravascular compressive forces (tissue pressure), coronary perfusion pressure, myogenic, local metabolic, endothelial as well as neural and hormonal influences. While each of these determinants can have profound influence over myocardial perfusion, largely through effects on end-effector ion channels, these mechanisms collectively modulate coronary vascular resistance and act to ensure that the myocardial requirements for oxygen and substrates are adequately provided by the coronary circulation. The purpose of this series of Comprehensive Physiology is to highlight current knowledge regarding the physiologic regulation of coronary blood flow, with emphasis on functional anatomy and the interplay between the physical and biological determinants of myocardial oxygen delivery. © 2017 American Physiological Society. Compr Physiol 7:321-382, 2017.
Collapse
Affiliation(s)
- Adam G Goodwill
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN
| | - Gregory M Dick
- California Medical Innovations Institute, 872 Towne Center Drive, Pomona, CA
| | - Alexander M Kiel
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN
- Weldon School of Biomedical Engineering, Purdue University, 206 S Martin Jischke Drive, Lafayette, IN
| | - Johnathan D Tune
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
40
|
Unthank JL, Sheridan KM, Dalsing MC. Collateral Growth in the Peripheral Circulation: A Review. Vasc Endovascular Surg 2016; 38:291-313. [PMID: 15306947 DOI: 10.1177/153857440403800401] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Arterial occlusive diseases are a major cause of morbidity and death in the United States. The enlargement of pre-existing vessels, which bypass the site of arterial occlusion, provide a natural way for the body to compensate for such obstructions. Individuals differ in their capacity to develop collateral vessels. In recent years much attention has been focused upon therapy to promote collateral development, primarily using individual growth factors. Such studies have had mixed results. Persistent controversies exist regarding the initiating stimuli, the processes involved in enlargement, the specific vessels that should be targeted, and the most appropriate terminology. Consequently, it is now recognized that more research is needed to extend our knowledge of the complex process of collateral growth. This basic science review addresses five questions essential in understanding current problems in collateral growth research and the development of therapeutic interventions.
Collapse
Affiliation(s)
- Joseph L Unthank
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | | | | |
Collapse
|
41
|
Ranjbar K, Rahmani-Nia F, Shahabpour E. Aerobic training and l-arginine supplementation promotes rat heart and hindleg muscles arteriogenesis after myocardial infarction. J Physiol Biochem 2016; 72:393-404. [PMID: 27121159 DOI: 10.1007/s13105-016-0480-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 03/09/2016] [Indexed: 12/18/2022]
Abstract
Arteriogenesis is a main defense mechanism to prevent heart and local tissues dysfunction in occlusive artery disease. TGF-β and angiostatin have a pivotal role in arteriogenesis. We tested the hypothesis that aerobic training and l-arginine supplementation promotes cardiac and skeletal muscles arteriogenesis after myocardial infarction (MI) parallel to upregulation of TGF-β and downregulation of angiostatin. For this purpose, 4 weeks after LAD occlusion, 50 male Wistar rats were randomly distributed into five groups: (1) sham surgery without MI (sham, n = 10), (2) control-MI (Con-MI, n = 10), (3) l-arginine-MI (La-MI, n = 10), (4) exercise training-MI (Ex-MI, n = 10), and (5) exercise and l-arginine-MI (Ex + La-MI). Exercise training groups running on a treadmill for 10 weeks with moderate intensity. Rats in the l-arginine-treated groups drank water containing 4 % l-arginine. Arteriolar density with different diameters (11-25, 26-50, 51-75, and 76-150 μm), TGF-β, and angiostatin gene expression were measured in cardiac (area at risk) and skeletal (soleus and gastrocnemius) muscles. Smaller arterioles decreased in cardiac after MI. Aerobic training and l-arginine increased the number of cardiac arterioles with 11-25 and 26-50 μm diameters parallel to TGF-β overexpression. In gastrocnemius muscle, the number of arterioles/mm(2) was only increased in the 11 to 25 μm in response to training with and without l-arginine parallel to angiostatin downregulation. Soleus arteriolar density with different size was not different between experimental groups. Results showed that 10 weeks aerobic exercise training and l-arginine supplementation promotes arteriogenesis of heart and gastrocnemius muscles parallel to overexpression of TGF-β and downregulation of angiostatin in MI rats.
Collapse
Affiliation(s)
- Kamal Ranjbar
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences, University of Guilan, Rasht, Iran
| | - Farhad Rahmani-Nia
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences, University of Guilan, Rasht, Iran.
| | - Elham Shahabpour
- Exercise Physiology Department, Faculty of Physical Education and Sport Science, Shiraz University, Fars, Iran
| |
Collapse
|
42
|
Ranjbar K, Nazem F, Nazari A, Gholami M, Nezami AR, Ardakanizade M, Sohrabi M, Ahmadvand H, Mottaghi M, Azizi Y. Synergistic effects of nitric oxide and exercise on revascularisation in the infarcted ventricle in a murine model of myocardial infarction. EXCLI JOURNAL 2016; 14:1104-15. [PMID: 26869868 PMCID: PMC4746998 DOI: 10.17179/excli2015-510] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 09/07/2015] [Indexed: 12/13/2022]
Abstract
It has been shown that density of microvessels decreases in the left ventricular after myocardial infarction (MI). The change of angiogenic and angiostatic factors as the main factors in revascularisation after exercise training in area at risk is not determined yet in MI. Therefore, the aim of the present study was the effect of exercise training and L-arginine supplementation on area at risk angiogenesis in myocardial infarction rat. Four weeks after surgery (Left Anterior Descending Coronary artery Ligation), myocardial infarction rats were divided into 4 groups: Sedentary rats (Sed-MI); L-arginine supplementation (La-MI); Exercise training (Ex-MI) and Exercise + L-arginine (Ex+La). Exercise training (ET) lasted for 10 weeks at 17 m/min for 10-50 min day(-1). Rats in the L-arginine-treated groups drank water containing 4 % L-arginine. After ET and L-arginine supplementation, ventricular function was evaluated and angiogenic and angiostatic indices were measured at ~1 mm from the edge of scar tissue (area at risk). Statistical analysis revealed that gene expression of VEGF as an angiogenic factor, angiostatin as an angiostatic factor and caspase-3 at area at risk decrease significantly in response to exercise training compared to the sedentary group. The capillary and arteriolar density in the Ex groups were significantly higher than those of the Sed groups. Compared to the Ex-MI group, the Ex+La group showed a markedly increase in capillary to fiber ratio. No significant differences were found in infarct size among the four groups, but cardiac function increased in response to exercise. Exercise training increases revascularization at area at risk by reduction of angiostatin. L-arginine supplementation causes additional effects on exercise-induced angiogenesis by preventing more reduction of VEGF gene expression in response to exercise. These improvements, in turn, increase left ventricular systolic function and decrease mortality in myocardial infarction rats.
Collapse
Affiliation(s)
- Kamal Ranjbar
- Department of Sport Physiology, Faculty of Physical Education and Sport Sciences, Bu-Ali Sina University, Hamedan, Iran
| | - Farzad Nazem
- Department of Sport Physiology, Faculty of Physical Education and Sport Sciences, Bu-Ali Sina University, Hamedan, Iran
| | - Afshin Nazari
- Department of Physiology, Razi Herbal Medicine Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Mohammadreza Gholami
- Department of Anatomy, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Ali Reza Nezami
- Department of cardiology, Shahid madani hospital, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Malihe Ardakanizade
- Department of Sport Physiology, Faculty of Physical Education and Sport Sciences, Bu-Ali Sina University, Hamedan, Iran
| | - Maryam Sohrabi
- Department of Anatomy, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hasan Ahmadvand
- Department of Biochemistry, Faculty of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Mohammad Mottaghi
- Department of Anatomy, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Yaser Azizi
- Department of Physiology, Physiology research center, School of Medicine, Iran Universty of Medical Sciences, Tehran, Iran
| |
Collapse
|
43
|
Corliss BA, Azimi MS, Munson J, Peirce SM, Murfee WL. Macrophages: An Inflammatory Link Between Angiogenesis and Lymphangiogenesis. Microcirculation 2016; 23:95-121. [PMID: 26614117 PMCID: PMC4744134 DOI: 10.1111/micc.12259] [Citation(s) in RCA: 241] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 11/23/2015] [Indexed: 12/14/2022]
Abstract
Angiogenesis and lymphangiogenesis often occur in response to tissue injury or in the presence of pathology (e.g., cancer), and it is these types of environments in which macrophages are activated and increased in number. Moreover, the blood vascular microcirculation and the lymphatic circulation serve as the conduits for entry and exit for monocyte-derived macrophages in nearly every tissue and organ. Macrophages both affect and are affected by the vessels through which they travel. Therefore, it is not surprising that examination of macrophage behaviors in both angiogenesis and lymphangiogenesis has yielded interesting observations that suggest macrophages may be key regulators of these complex growth and remodeling processes. In this review, we will take a closer look at macrophages through the lens of angiogenesis and lymphangiogenesis, examining how their dynamic behaviors may regulate vessel sprouting and function. We present macrophages as a cellular link that spatially and temporally connects angiogenesis with lymphangiogenesis, in both physiological growth and in pathological adaptations, such as tumorigenesis. As such, attempts to therapeutically target macrophages in order to affect these processes may be particularly effective, and studying macrophages in both settings will accelerate the field's understanding of this important cell type in health and disease.
Collapse
Affiliation(s)
- Bruce A. Corliss
- Department of Biomedical Engineering, 415 Lane Road, University of Virginia, Charlottesville, VA 22908
| | - Mohammad S. Azimi
- Department of Biomedical Engineering, 500 Lindy Boggs Energy Center, Tulane University, New Orleans, LA 70118
| | - Jenny Munson
- Department of Biomedical Engineering, 415 Lane Road, University of Virginia, Charlottesville, VA 22908
| | - Shayn M. Peirce
- Department of Biomedical Engineering, 415 Lane Road, University of Virginia, Charlottesville, VA 22908
| | - Walter Lee Murfee
- Department of Biomedical Engineering, 500 Lindy Boggs Energy Center, Tulane University, New Orleans, LA 70118
| |
Collapse
|
44
|
Korenovskii YV, El'chaninova SA. Effects of S-Nitrosoglutathione on the Expression of MMP-1 mRNA in HT1080 Cells during Hypoxic Hypoxia. Bull Exp Biol Med 2016; 160:319-21. [PMID: 26742737 DOI: 10.1007/s10517-016-3160-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Indexed: 12/01/2022]
Abstract
Synthetic antioxidant S-nitrosoglutathione suppressed the expression of MMP-1 mRNA in HT1080 cells exposed to hypoxic hypoxia; hyperexpression of superoxide dismutase 2 increased and hyperexpression of catalase inhibited the expression of MMP-1 mRNA in HT1080 cells.
Collapse
Affiliation(s)
- Yu V Korenovskii
- Department of Biochemistry and Clinical Laboratory Diagnostics, Altay State Medical University, Ministry of Health of the Russian Federation, Barnaul, Russia.
| | - S A El'chaninova
- Department of Biochemistry and Clinical Laboratory Diagnostics, Altay State Medical University, Ministry of Health of the Russian Federation, Barnaul, Russia
| |
Collapse
|
45
|
Karimian J, Khazaei M, Shekarchizadeh P. Effect of Resistance Training on Capillary Density Around Slow and Fast Twitch Muscle Fibers in Diabetic and Normal Rats. Asian J Sports Med 2015; 6:e24040. [PMID: 26715966 PMCID: PMC4691304 DOI: 10.5812/asjsm.24040] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 03/04/2014] [Indexed: 01/22/2023] Open
Abstract
Background: It is well accepted that skeletal muscle conforms to exercise stimulus by increasing capillary density and angiogenesis, but there is less evidence regarding the effect of resistance training on capillary density in flexor hallucis longus (FHL) and soleus muscle. Objectives: In this study, we evaluated the effect of resistance training on capillary density around soleus and FHL muscles in type 1 diabetic rats. Materials and Methods: Thirty-six male rats were divided into four groups: (1) control; (2) diabetic; (3) diabetic trained and (4) control trained (n = 9 each). A Single intraperitoneal injection of Streptozotocin at a dose of 55 mg/kg was used for induction of diabetes. The rats in the trained group undertook one training session per day for 3 days/week. Training was done with the use of a 1 meter high ladder inclined at 80°. After 4 weeks, the plasma nitrite concentrations were measured. Capillary/fiber ratio was determined around soleus and FHL muscles by immunohistochemistry. Results: Plasma Nitric Oxide (NO) concentration was increased after resistance training in diabetic animals (P < 0.05). Capillary/fiber ratio around the soleus muscle of diabetic group was more than control rats. Resistance training did not alter capillary/fiber ratio in diabetic animals (1.00 ± 0.6 vs. 1.07 ± 0.07, respectively). Capillary/fiber ratio around FHL muscle was significantly different between diabetic and control and did not alter after exercise (diabetes: 1.1702 ± 0.09; diabetic trained: 1.1714 ± 0.08; control: 0.79 ± 0.08; control trained: 0.73 ± 0.03). There was a positive correlation between plasma NO concentration and capillary density in the soleus muscle (R2 = 0.65). Conclusions: Resistance training could not improve capillary/fiber ratio in soleus and FHL muscle of diabetic animals in spite of increase in some angiogenic factors including NO.
Collapse
Affiliation(s)
- Jahangir Karimian
- Department of School Management And Medical Informatics, Isfahan University of Medical Sciences, Isfahan, IR Iran
| | - Majid Khazaei
- Neurogenic Inflammation Research Centre and Department of Physiology, School of medicine, Mashhad University of Medical Sciences, Mashhad, IR Iran
| | - Parivash Shekarchizadeh
- Department of School Management And Medical Informatics, Isfahan University of Medical Sciences, Isfahan, IR Iran
- Corresponding author: Parivash Shekarchizadeh, Department of School Management and Medical Informatics, Isfahan University of Medical Sciences, Isfahan, IR Iran. Tel: +98-3117922026, E-mail:
| |
Collapse
|
46
|
Functional role of inorganic trace elements in angiogenesis—Part I: N, Fe, Se, P, Au, and Ca. Crit Rev Oncol Hematol 2015; 96:129-42. [DOI: 10.1016/j.critrevonc.2015.05.010] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 04/15/2015] [Accepted: 05/12/2015] [Indexed: 01/08/2023] Open
|
47
|
An Inflammatory Polymorphisms Risk Scoring System for the Differentiation of Ischemic Stroke Subtypes. Mediators Inflamm 2015; 2015:569714. [PMID: 26355258 PMCID: PMC4556875 DOI: 10.1155/2015/569714] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 07/13/2015] [Accepted: 07/15/2015] [Indexed: 11/18/2022] Open
Abstract
UNLABELLED Inflammation has been associated with atherothrombotic stroke and recently with cardioembolic stroke. Different genetic risk factors have been specifically associated with the subtypes of ischemic stroke (cardioembolic, atherothrombotic, and lacunar). However, there are no studies that have generated genetic risk scores for the different subtypes of ischemic stroke using polymorphisms associated with inflammation. METHODS We have analyzed 68 polymorphisms of 30 inflammatory mediator genes in 2,685 subjects: 1,987 stroke cases and 698 controls. We generated a genetic scoring system with the most significant polymorphisms weighted by the odds ratio of every polymorphism and taken into consideration the stroke subtype. RESULTS Three polymorphisms, rs1205 (CRP gene), rs1800779, and rs2257073 (NOS3 gene), were associated with cardioembolic stroke (p value <0.05). The score generated was only associated with the cardioembolic stroke subtype (p value: 0.001) and was replicated in an independent cohort (p value: 0.017). The subjects with the highest score presented a cardioembolic stroke in 92.2% of the cases (p value: 0.002). CONCLUSION The genetics of inflammatory markers is more closely associated with cardioembolic strokes than with atherothrombotic or lacunar strokes. The genetic risk scoring system could be useful in the prediction and differentiation of ischemic stroke; however, it might be specific to particular ischemic stroke subtypes.
Collapse
|
48
|
Tykhomyrov AA, Nedzvetsky VS, Bardachenko NI, Grinenko TV, Kuryata OV. Statin treatment decreases serum angiostatin levels in patients with ischemic heart disease. Life Sci 2015; 134:22-9. [DOI: 10.1016/j.lfs.2015.05.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 04/22/2015] [Accepted: 05/17/2015] [Indexed: 11/25/2022]
|
49
|
|
50
|
|