51
|
Review of clinical applications of nitric oxide-containing air-plasma gas flow generated by Plason device. CLINICAL PLASMA MEDICINE 2020. [DOI: 10.1016/j.cpme.2020.100112] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
52
|
Zippusch S, Helms F, Lau S, Klingenberg M, Schrimpf C, Haverich A, Wilhelmi M, Böer U. Perfusion promotes endothelialized pore formation in high concentration fibrin gels otherwise unsuitable for tube development. Int J Artif Organs 2020; 44:130-138. [PMID: 32611278 DOI: 10.1177/0391398820936700] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Vascularization of tissue engineered implants is crucial for their survival and integration in the recipient's body. Pre-vascularized, fibrin-based implants offer a solution since low concentration fibrin hydrogels (1 mg/mL) have been shown to promote tube formation of endothelial cells in co-culture with adipogenic stem cells. However, higher fibrinogen concentrations (> 20 mg/mL) enabling the fabrication of stable implants are necessary.We here characterized fibrin gels of 1-30 mg/mL for their rheological properties and whether they support tube formation of endothelial cell-adipogenic stem cell co-cultures for up to 7 days. Moreover, 20 mg/mL gels containing preformed channels and endothelial cell-adipogenic stem cell co-culture were perfused continuously in a customized flow chamber with 3.9 dyn/cm2 for 12 days and analyzed for capillary formation.Rheology of fibrin gels showed increasing stability proportional to fibrinogen concentration with 20 mg/mL gels having a storage module of 465 Pa. Complex tube networks stable for 7 days were observed at 1-5 mg/mL gels whereas higher concentrations showed initial sprouting only. However, perfusion of 20 mg/mL fibrin gels resulted in endothelialized pore formation in several layers of the gel with endothelial cell-adipogenic stem cell co-culture.Thus, perfusion supports the formation of capillary-like structures in fibrin gels that are too dense for spontaneous tube formation under static conditions. Future studies are necessary to further increase pore density and to investigate proper nutrition of tissue-specific target cells in the scaffold.
Collapse
Affiliation(s)
- Sarah Zippusch
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover Medical School, Hannover, Germany.,Division for Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Florian Helms
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover Medical School, Hannover, Germany
| | - Skadi Lau
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover Medical School, Hannover, Germany.,Division for Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Melanie Klingenberg
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover Medical School, Hannover, Germany.,Division for Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Claudia Schrimpf
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover Medical School, Hannover, Germany.,Division for Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Axel Haverich
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover Medical School, Hannover, Germany.,Division for Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Mathias Wilhelmi
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover Medical School, Hannover, Germany.,Division for Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Ulrike Böer
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover Medical School, Hannover, Germany.,Division for Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| |
Collapse
|
53
|
Wang L, Erişen DE, Yang K, Zhang B, Guan H, Chen S. Anticoagulation and antibacterial functional coating on vascular implant interventional medical catheter. J Biomed Mater Res B Appl Biomater 2020; 108:2868-2877. [PMID: 32420689 DOI: 10.1002/jbm.b.34618] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 03/30/2020] [Accepted: 04/13/2020] [Indexed: 01/12/2023]
Abstract
Vascular implant interventional medical catheter will contact with blood firstly after implantation. The anticoagulation and antibacterial functions of this device will determine the success or failure. Copper (Cu) has been verified to possess multi-biofunctions, but it was challenging to add the Cu metal to most materials. Take advantage of its functionality; Cu has been grafted on the material surface to improve the anticoagulation function and accelerate endothelialization. In this study, a Cu-bearing chitosan coating was prepared on the catheter to endow the anticoagulation and anti-infection functions. Besides, properties characterization and functional evaluation of the coated medical catheter were investigated. Dynamic blood clotting and platelet adhesion tests were carried out to evaluate the anticoagulation property. Besides this, the antibacterial test was used to estimate the anti-infection function. The surface energy and Cu ions release from the coating were detected and calculated by contact angles and immersion tests, respectively. The results of attenuated total reflectance Fourier transform infrared spectroscopy (ATR-FTIR) and X-ray photoelectron spectroscopy (XPS) revealed that Cu ions were grafted in the chitosan coating. Thermogravimetric analysis (TA) result showed the concentration of Cu ions in the coating. The results of dynamic blood clotting, platelet adhesion, and antibacterial tests revealed that Cu grafted in chitosan would improve the blood compatibility and anti-infection property. The surface properties and Cu ions release behavior of Cu-bearing coating revealed the reasons for multi-biofunctions. This study indicated that the Cu-bearing chitosan coating could endow the vascular implant interventional device anticoagulation and anti-infection functions, which has excellent potential for clinical application.
Collapse
Affiliation(s)
- Lirong Wang
- College of Chemistry, Liaoning University, Shenyang, China
| | - Deniz E Erişen
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang, China.,University of Science and Technology of China, Hefei, China
| | - Ke Yang
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang, China
| | - Bingchun Zhang
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang, China
| | - Hongyu Guan
- College of Chemistry, Liaoning University, Shenyang, China
| | - Shanshan Chen
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang, China
| |
Collapse
|
54
|
Schall N, Garcia JJ, Kalyanaraman H, China SP, Lee JJ, Sah RL, Pfeifer A, Pilz RB. Protein kinase G1 regulates bone regeneration and rescues diabetic fracture healing. JCI Insight 2020; 5:135355. [PMID: 32315291 DOI: 10.1172/jci.insight.135355] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 04/08/2020] [Indexed: 01/16/2023] Open
Abstract
Bone fractures are a major cause of morbidity and mortality, particularly in patients with diabetes, who have a high incidence of fractures and exhibit poor fracture healing. Coordinated expression of osteoblast-derived vascular endothelial growth factor (VEGF) and bone morphogenic proteins (BMPs) is essential for fracture repair. The NO/cGMP/protein kinase G (PKG) signaling pathway mediates osteoblast responses to estrogens and mechanical stimulation, but the pathway's role in bone regeneration is unknown. Here, we used a mouse cortical-defect model to simulate bone fractures and studied osteoblast-specific PKG1-knockout and diabetic mice. The knockout mice had normal bone microarchitecture but after injury exhibited poor bone regeneration, with decreased osteoblasts, collagen deposition, and microvessels in the bone defect area. Primary osteoblasts and tibiae from the knockout mice expressed low amounts of Vegfa and Bmp2/4 mRNAs, and PKG1 was required for cGMP-stimulated expression of these genes. Diabetic mice also demonstrated low Vegfa and Bmp2/4 expression in bone and impaired bone regeneration after injury; notably, the cGMP-elevating agent cinaciguat restored Vegfa and BMP2/4 expression and full bone healing. We conclude that PKG1 is a key orchestrator of VEGF and BMP signaling during bone regeneration and propose pharmacological PKG activation as a novel therapeutic approach to enhance fracture healing.
Collapse
Affiliation(s)
- Nadine Schall
- Department of Medicine, University of California, San Diego, La Jolla, California, USA.,Institute for Pharmacology and Toxicology, University of Bonn, Bonn, Germany
| | - Julian J Garcia
- Department of Medicine, University of California, San Diego, La Jolla, California, USA.,Department of Bioengineering, University of California, San Diego, La Jolla, California, USA
| | - Hema Kalyanaraman
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Shyamsundar Pal China
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Jenna J Lee
- Department of Bioengineering, University of California, San Diego, La Jolla, California, USA
| | - Robert L Sah
- Department of Bioengineering, University of California, San Diego, La Jolla, California, USA
| | - Alexander Pfeifer
- Institute for Pharmacology and Toxicology, University of Bonn, Bonn, Germany
| | - Renate B Pilz
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
55
|
Wei X, Guan L, Fan P, Liu X, Liu R, Liu Y, Bai H. Direct Current Electric Field Stimulates Nitric Oxide Production and Promotes NO-Dependent Angiogenesis: Involvement of the PI3K/Akt Signaling Pathway. J Vasc Res 2020; 57:195-205. [PMID: 32375152 DOI: 10.1159/000506517] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 02/13/2020] [Indexed: 02/05/2023] Open
Abstract
Electric fields (EFs) promote angiogenesis in vitro and in vivo. These results indicate the feasibility of the application of EFs to modulate angiogenesis. Nitric oxide (NO) derived from endothelial nitric oxide synthase (eNOS) is an important regulator of angiogenesis. However, the role of direct current EFs in eNOS activity and expression in association with angiogenesis of endothelial cells has not been investigated. In the present study, we stimulated human umbilical vein endothelial cells (HUVECs) with EFs and evaluated the activity and expression of eNOS. EFs induced significant phosphorylation of eNOS, upregulation of the expression of eNOS protein, and an increase in NO production from HUVECs. L-NAME, a specific inhibitor of eNOS, abolished EF-induced HUVEC angiogenesis. EFs stimulated Akt activation. Inhibition of PI3K activity inhibited EF-mediated Akt and eNOS activation and inhibited NO production in the endothelial cells. Moreover, EFs stimulated HUVEC proliferation and enhanced the S phase cell population of the cell cycle. We conclude that EFs stimulate eNOS activation and NO production via a PI3K/Akt-dependent pathway. Thus, activation of eNOS appears to be one of the key signaling pathways necessary for EF-mediated angiogenesis. These novel findings suggest that NO signaling may have an important role in EF-mediated endothelial cell function.
Collapse
Affiliation(s)
- Xing Wei
- Laboratory of Genetic Disease and Perinatal Medicine and Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Linbo Guan
- Laboratory of Genetic Disease and Perinatal Medicine and Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Ping Fan
- Laboratory of Genetic Disease and Perinatal Medicine and Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Xinghui Liu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Rui Liu
- Division of Peptides Related to Human Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Liu
- Department of Biochemistry and Molecular Biology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, China
| | - Huai Bai
- Laboratory of Genetic Disease and Perinatal Medicine and Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China,
| |
Collapse
|
56
|
Abstract
Cardiovascular diseases (CVDs) pose a serious threat to human health, which are characterized by high disability and mortality rate globally such as myocardial infarction (MI), atherosclerosis, and heart failure. Although stem cells transplantation and growth factors therapy are promising, their low survival rate and loss at the site of injury are major obstacles to this therapy. Recently, the development of hydrogel scaffold materials provides a new way to solve this problem, which have shown the potential to treat CVD. Among these scaffold materials, environmentally responsive hydrogels have great prospects in repairing the microenvironment of cardiovascular tissues and vascular regeneration. They provide a new method for the treatment of cardiovascular tissue repair and space-time control for the release of various therapeutic drugs, including small-molecule drugs, growth factors, and stem cells. Herein, this article reviews the occurrence and current treatment of CVD, as well as the repair of cardiovascular injury by several environmental responsive hydrogels systems currently used, mainly focusing on the delivery of growth factors or the application of cell therapy to revascularization. In addition, we will also discuss the enormous potential and personal perspectives of environmentally responsive hydrogels in cardiovascular repair.
Collapse
|
57
|
Menger MM, Nalbach L, Roma LP, Körbel C, Wrublewsky S, Glanemann M, Laschke MW, Menger MD, Ampofo E. Erythropoietin accelerates the revascularization of transplanted pancreatic islets. Br J Pharmacol 2020; 177:1651-1665. [PMID: 31721150 DOI: 10.1111/bph.14925] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 10/30/2019] [Accepted: 11/03/2019] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND AND PURPOSE Pancreatic islet transplantation is a promising therapeutic approach for Type 1 diabetes. A major prerequisite for the survival of grafted islets is a rapid revascularization after transplantation. Erythropoietin (EPO), the primary regulator of erythropoiesis, has been shown to promote angiogenesis. Therefore, we investigated in this study whether EPO improves the revascularization of transplanted islets. EXPERIMENTAL APPROACH Islets from FVB/N mice were transplanted into dorsal skinfold chambers of recipient animals, which were daily treated with an intraperitoneal injection of EPO (500 IU·kg-1 ) or vehicle (control) throughout an observation period of 14 days. In a second set of experiments, animals were only pretreated with EPO over a 6-day period prior to islet transplantation. The revascularization of the grafts was assessed by repetitive intravital fluorescence microscopy and immunohistochemistry. In addition, a streptozotocin-induced diabetic mouse model was used to study the effect of EPO-pretreatment on the endocrine function of the grafts. KEY RESULTS EPO treatment slightly accelerated the revascularization of the islet grafts. This effect was markedly more pronounced in EPO-pretreated animals, resulting in significantly higher numbers of engrafted islets and an improved perfusion of endocrine tissue without affecting systemic haematocrit levels when compared with controls. Moreover, EPO-pretreatment significantly accelerated the recovery of normoglycaemia in diabetic mice after islet transplantation. CONCLUSION AND IMPLICATIONS These findings demonstrate that, particularly, short-term EPO-pretreatment represents a promising therapeutic approach to improve the outcome of islet transplantation, without an increased risk of thromboembolic events.
Collapse
Affiliation(s)
- Maximilian M Menger
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Lisa Nalbach
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Leticia P Roma
- Biophysics Department, Center for Human and Molecular Biology, Saarland University, Homburg/Saar, Germany
| | - Christina Körbel
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Selina Wrublewsky
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Matthias Glanemann
- Department for General, Visceral, Vascular and Pediatric Surgery, Saarland University, Homburg/Saar, Germany
| | - Matthias W Laschke
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Michael D Menger
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Emmanuel Ampofo
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| |
Collapse
|
58
|
Omori K, Tachikawa M, Hirose S, Taii A, Akanuma SI, Hosoya KI, Terasaki T. Developmental changes in transporter and receptor protein expression levels at the rat blood-brain barrier based on quantitative targeted absolute proteomics. Drug Metab Pharmacokinet 2020; 35:117-123. [DOI: 10.1016/j.dmpk.2019.09.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 09/17/2019] [Accepted: 09/24/2019] [Indexed: 02/02/2023]
|
59
|
Yang SS, Oh JM, Chun S, Kim BS, Kim CS, Lee J. Tauroursodeoxycholic acid induces angiogenic activity in endothelial cells and accelerates bone regeneration. Bone 2020; 130:115073. [PMID: 31626993 DOI: 10.1016/j.bone.2019.115073] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 07/08/2019] [Accepted: 09/17/2019] [Indexed: 12/31/2022]
Abstract
Angiogenesis is a crucial process during bone tissue regeneration. The aim of this study was to investigate the angiogenic activity and the potentiation of bone regeneration via angiogenesis using tauroursodeoxycholic acid (TUDCA) in vitro and in vivo. We investigated the effect of TUDCA on proliferation and angiogenic differentiation in human umbilical vein endothelial cells (HUVECs) and the associated signaling pathway. Proliferation was determined using crystal violet assay. Angiogenic effects were evaluated based on cell migration and tube formation. In order to explore TUDCA-signaling pathways, phosphorylation of mitogen activated protein kinase, protein kinase B (AKT), and endothelial nitric oxide synthase (eNOS) was determined using western blot. Furthermore, in vivo bone formation and angiogenesis were determined using a New Zealand outbred albino rabbit calvarial defect model, while angiogenesis and bone formation were evaluated using micro-CT and histological analysis. Our results show that TUDCA significantly increased cell proliferation. Moreover, TUDCA enhanced cell migration and tube formation in HUVECs. TUDCA increased the phosphorylation of AKT, ERK1/2, c-Jun N-terminal kinase, and eNOS. Specific inhibitors of ERK1/2 (PD98059), JNK (SP600125), and AKT (AKT1/2) inhibited the TUDCA-induced migration and tube formation, while the p38 inhibitor (SB203580) did not. The in vivo study used TUDCA to accelerate new blood vessel formation and promoted bone formation in rabbit calvarial defect model. These results indicate that TUDCA plays a critical role in enhancing the angiogenesis of endothelial cells and in vivo new bone regeneration. The use of TUDCA may contribute to the regeneration of bone tissue by improving angiogenesis.
Collapse
Affiliation(s)
- Sun-Sik Yang
- Department of Oral and Maxillofacial Surgery, College of Dentistry, Wonkwang University 77 Dunsan-ro, Seo-gu, Daejeon 302-120, Republic of Korea
| | - Jung-Mi Oh
- Department of Physiology, Chonbuk National Medical School, Jeonju-si, Jeollabuk-do 54896, Republic of Korea
| | - Sungkun Chun
- Department of Physiology, Chonbuk National Medical School, Jeonju-si, Jeollabuk-do 54896, Republic of Korea
| | - Beom-Su Kim
- Carbon Nano Convergence Technology Center for Next Generation Engineers (CNN), Chonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeollabuk-do 54896, Republic of Korea.
| | - Cheol Sang Kim
- Department of Bionanosystem Engineering, Graduate School, Chonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeollabuk-do 54896, Republic of Korea; Division of Mechanical Design Engineering, Chonbuk National University, Jeonju-si, Jeollabuk-do 54896, Republic of Korea.
| | - Jun Lee
- Department of Oral and Maxillofacial Surgery, College of Dentistry, Wonkwang University 77 Dunsan-ro, Seo-gu, Daejeon 302-120, Republic of Korea.
| |
Collapse
|
60
|
Antibacterial and osteoinductive biomacromolecules composite electrospun fiber. Int J Biol Macromol 2020; 143:958-967. [DOI: 10.1016/j.ijbiomac.2019.09.156] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 09/19/2019] [Accepted: 09/23/2019] [Indexed: 11/19/2022]
|
61
|
Zhu X, Gao Y. 17O NMR spectroscopy-assisted in vitro bioactivity studies of the intermediates formed via Na 2S and RSNO cross-linking reactions. RSC Adv 2020; 10:39617-39626. [PMID: 35515380 PMCID: PMC9057446 DOI: 10.1039/d0ra05054g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 10/22/2020] [Indexed: 11/22/2022] Open
Abstract
The cross-linking reaction between sulfide and S-nitrosothiol moieties has been intensively investigated and thionitrite/thionitrous acid (SNO−/HSNO) as well as nitrosopersulfide (SSNO−) were reported to be the intermediates that could serve as reservoirs for nitric oxide (NO). However, debate still exists regarding the stability and biological activity of SNO−/HSNO and SSNO−. In order to investigate the chemical properties and biological activity of SNO− and SSNO−, we set out to re-characterize the reaction intermediates using UV-Vis and 15N NMR spectroscopy techniques, as well as a new 17O NMR approach. The effects of SNO− and SSNO− on cellular NO and cGMP levels were assessed via cell culture experiments, and also the effects of SNO− and SSNO− on cell proliferation, migration, and capillary-like structure formation were evaluated with human umbilical vein endothelial cells (HUVEC). Through this work, the characteristic peaks and half-lives of SNO− and SSNO− were elucidated under various preparation conditions. The biological assays demonstrated that SSNO− increased the cellular NO and cGMP levels and also facilitated cell proliferation, migration and stimulated angiogenesis, while in contrast SNO− did not exhibit these effects. By using UV-Vis, 15N NMR and 17O NMR spectroscopy techniques, we characterized the intermediates (SSNO− and SNO−) obtained from RSNO and Na2S cross-linking reaction. We found that SSNO− could serve as NO reservoir in cell culture experiments.![]()
Collapse
Affiliation(s)
- Xingyu Zhu
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education
- School of Life Sciences
- Jilin University
- Changchun 130012
- China
| | - Yin Gao
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education
- School of Life Sciences
- Jilin University
- Changchun 130012
- China
| |
Collapse
|
62
|
Sung YC, Jin PR, Chu LA, Hsu FF, Wang MR, Chang CC, Chiou SJ, Qiu JT, Gao DY, Lin CC, Chen YS, Hsu YC, Wang J, Wang FN, Yu PL, Chiang AS, Wu AYT, Ko JJS, Lai CPK, Lu TT, Chen Y. Delivery of nitric oxide with a nanocarrier promotes tumour vessel normalization and potentiates anti-cancer therapies. NATURE NANOTECHNOLOGY 2019; 14:1160-1169. [PMID: 31740794 DOI: 10.1038/s41565-019-0570-3] [Citation(s) in RCA: 264] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 10/03/2019] [Indexed: 05/28/2023]
Abstract
Abnormal tumour vasculature has a significant impact on tumour progression and response to therapy. Nitric oxide (NO) regulates angiogenesis and maintains vascular homeostasis and, thus, can be delivered to normalize tumour vasculature. However, a NO-delivery system with a prolonged half-life and a sustained release mechanism is currently lacking. Here we report the development of NanoNO, a nanoscale carrier that enables sustained NO release to efficiently deliver NO into hepatocellular carcinoma. Low-dose NanoNO normalizes tumour vessels and improves the delivery and effectiveness of chemotherapeutics and tumour necrosis factor-related, apoptosis-inducing, ligand-based therapy in both primary tumours and metastases. Furthermore, low-dose NanoNO reprogrammes the immunosuppressive tumour microenvironment toward an immunostimulatory phenotype, thereby improving the efficacy of cancer vaccine immunotherapy. Our findings demonstrate the ability of nanoscale NO delivery to efficiently reprogramme tumour vasculature and immune microenvironments to overcome resistance to cancer therapy, resulting in a therapeutic benefit.
Collapse
Affiliation(s)
- Yun-Chieh Sung
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, Taiwan
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Pei-Ru Jin
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, Taiwan
| | - Li-An Chu
- Brain Research Center, National Tsing Hua University, Hsinchu, Taiwan
| | - Fu-Fei Hsu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Mei-Ren Wang
- Department of Chemistry, Chung Yuan Christian University, Taoyuan, Taiwan
| | - Chih-Chun Chang
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, Taiwan
| | - Show-Jen Chiou
- Department of Applied Chemistry, National Chiayi University, Chiayi, Taiwan
| | - Jiantai Timothy Qiu
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Biomedical Sciences, School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Dong-Yu Gao
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, Taiwan
| | - Chu-Chi Lin
- Department of Biomedical Sciences, School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Sing Chen
- Department of Biomedical Sciences, School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yi-Chiung Hsu
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan
| | - Jane Wang
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Fu-Nien Wang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Pei-Lun Yu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Ann-Shyn Chiang
- Brain Research Center, National Tsing Hua University, Hsinchu, Taiwan
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli, Taiwan
| | - Anthony Yan-Tang Wu
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, Taiwan
- Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Academia Sinica, Taipei, Taiwan
- Department and Graduate Institute of Pharmacology, National Taiwan University, Taipei, Taiwan
| | - John Jun-Sheng Ko
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, Taiwan
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, Taiwan
| | - Charles Pin-Kuang Lai
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, Taiwan
- Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Academia Sinica, Taipei, Taiwan
- Genome and Systems Biology Degree Program, National Taiwan University and Academia Sinica, Taipei, Taiwan
| | - Tsai-Te Lu
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, Taiwan.
- Department of Chemistry, Chung Yuan Christian University, Taoyuan, Taiwan.
| | - Yunching Chen
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
63
|
Corliss BA, Delalio LJ, Stevenson Keller TC, Keller AS, Keller DA, Corliss BH, Beers JM, Peirce SM, Isakson BE. Vascular Expression of Hemoglobin Alpha in Antarctic Icefish Supports Iron Limitation as Novel Evolutionary Driver. Front Physiol 2019; 10:1389. [PMID: 31780954 PMCID: PMC6861181 DOI: 10.3389/fphys.2019.01389] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 10/24/2019] [Indexed: 12/16/2022] Open
Abstract
Frigid temperatures of the Southern Ocean are known to be an evolutionary driver in Antarctic fish. For example, many fish have reduced red blood cell (RBC) concentration to minimize vascular resistance. Via the oxygen-carrying protein hemoglobin, RBCs contain the vast majority of the body's iron, which is known to be a limiting nutrient in marine ecosystems. Since lower RBC levels also lead to reduced iron requirements, we hypothesize that low iron availability was an additional evolutionary driver of Antarctic fish speciation. Antarctic Icefish of the family Channichthyidae are known to have an extreme alteration of iron metabolism due to loss of RBCs and two iron-binding proteins, hemoglobin and myoglobin. Loss of hemoglobin is considered a maladaptive trait allowed by relaxation of predator selection since extreme adaptations are required to compensate for the loss of oxygen-carrying capacity. However, iron dependency minimization may have driven hemoglobin loss instead of a random evolutionary event. Given the variety of functions that hemoglobin serves in the endothelium, we suspected the protein corresponding to the 3' truncated Hbα fragment (Hbα-3'f) that was not genetically excluded by icefish may still be expressed as a protein. Using whole mount confocal microscopy, we show that Hbα-3'f is expressed in the vascular endothelium of icefish retina, suggesting this Hbα fragment may still serve an important role in the endothelium. These observations support a novel hypothesis that iron minimization could have influenced icefish speciation with the loss of the iron-binding portion of Hbα in Hbα-3'f, as well as hemoglobin β and myoglobin.
Collapse
Affiliation(s)
- Bruce A Corliss
- Biomedical Engineering Department, University of Virginia, Charlottesville, VA, United States
| | - Leon J Delalio
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - T C Stevenson Keller
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United States.,Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Alexander S Keller
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | | | - Bruce H Corliss
- Graduate School of Oceanography, University of Rhode Island, Kingston, RI, United States
| | - Jody M Beers
- Department of Biology, College of Charleston, Charleston, SC, United States
| | - Shayn M Peirce
- Biomedical Engineering Department, University of Virginia, Charlottesville, VA, United States
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United States.,Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, VA, United States
| |
Collapse
|
64
|
|
65
|
Abstract
Background The study of the mechanisms controlling wound healing is an attractive area within the field of biology, with it having a potentially significant impact on the health sector given the current medical burden associated with healing in the elderly population. Healing is a complex process and includes many steps that are regulated by coding and noncoding RNAs, proteins and other molecules. Nitric oxide (NO) is one of these small molecule regulators and its function has already been associated with inflammation and angiogenesis during adult healing. Results Our results showed that NO is also an essential component during embryonic scarless healing and acts via a previously unknown mechanism. NO is mainly produced during the early phase of healing and it is crucial for the expression of genes associated with healing. However, we also observed a late phase of healing, which occurs for several hours after wound closure and takes place under the epidermis and includes tissue remodelling that is dependent on NO. We also found that the NO is associated with multiple cellular metabolic pathways, in particularly the glucose metabolism pathway. This is particular noteworthy as the use of NO donors have already been found to be beneficial for the treatment of chronic healing defects (including those associated with diabetes) and it is possible that its mechanism of action follows those observed during embryonic wound healing. Conclusions Our study describes a new role of NO during healing, which may potentially translate to improved therapeutic treatments, especially for individual suffering with problematic healing.
Collapse
|
66
|
Heidary F, Gharebaghi R, Ghasemi H, Mahdavi MRV, Ghaffarpour S, Naghizadeh MM, Ghazanfari T. Angiogenesis modulatory factors in subjects with chronic ocular complications of Sulfur Mustard exposure: A case-control study. Int Immunopharmacol 2019; 76:105843. [PMID: 31629219 DOI: 10.1016/j.intimp.2019.105843] [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: 06/19/2019] [Revised: 08/17/2019] [Accepted: 08/19/2019] [Indexed: 11/18/2022]
Abstract
BACKGROUND Chronic ocular complications of Sulfur Mustard (SM) exposure leads to severe ocular morbidity during time. The aim of this study was to compare serum levels of Interleukin 17 (IL-17), IL-12, vascular endothelial growth factor (VEGF)-C, VEGF-D and nitric oxide (NO) in SM-exposed patients versus the control group and to measure tear concentration of VEGF-C only in the SM-exposed group. METHODS In this prospective case control, 128 SM-exposed patients and 31 healthy control subjects were included. In the case group ocular manifestations were classified to three subgroups of mild (19 cases), moderate (31 cases) and severe (78 cases) forms of disease. Serum levels of IL-17, IL-12, NO, VEGF-C and VEGF-D, in all subjects and tear concentration of VEGF-C in SM-exposed group was evaluated. RESULTS All subjects were male and mean ± standard deviation (SD) of age in the case and control groups were 44.9 ± 8.8 and 40.9 ± 10.1 years, respectively. Except for significantly lower serum level of IL-17 (p < 0.001) and NO (p = 0.003), other values were not significantly different. The tear concentration of VEGF-C and serum level of IL-12 were not different between subgroups in the SM-exposed group, yet were significantly lower among those with abnormally dilated and tortuous conjunctival vessels and corneal pannus, respectively (p = 0.01, p = 0.015). CONCLUSIONS Exposure to SM significantly reduced serum level of IL-17 and NO in the delayed phase, yet did not influence VEGF-C; VEGF-D or IL-12.
Collapse
Affiliation(s)
- Fatemeh Heidary
- Immunoregulation Research Center, Shahed University, Tehran 3319118651, Iran
| | - Reza Gharebaghi
- International Virtual Ophthalmic Research Center, Tehran, Iran
| | - Hassan Ghasemi
- Department of Ophthalmology, Shahed University, Tehran 3319118651, Iran
| | | | - Sara Ghaffarpour
- Immunoregulation Research Center, Shahed University, Tehran 3319118651, Iran
| | - Mohammad Mehdi Naghizadeh
- Non Communicable Diseases Research Center, Fasa University of Medical Science, Fasa 7461686688, Iran
| | - Tooba Ghazanfari
- Immunoregulation Research Center, Shahed University, Tehran 3319118651, Iran.
| |
Collapse
|
67
|
Hays E, Bonavida B. Nitric Oxide-Mediated Enhancement and Reversal of Resistance of Anticancer Therapies. Antioxidants (Basel) 2019; 8:E407. [PMID: 31533363 PMCID: PMC6769868 DOI: 10.3390/antiox8090407] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 09/11/2019] [Accepted: 09/13/2019] [Indexed: 12/13/2022] Open
Abstract
In the last decade, immune therapies against human cancers have emerged as a very effective therapeutic strategy in the treatment of various cancers, some of which are resistant to current therapies. Although the clinical responses achieved with many therapeutic strategies were significant in a subset of patients, another subset remained unresponsive initially, or became resistant to further therapies. Hence, there is a need to develop novel approaches to treat those unresponsive patients. Several investigations have been reported to explain the underlying mechanisms of immune resistance, including the anti-proliferative and anti-apoptotic pathways and, in addition, the increased expression of the transcription factor Yin-Yang 1 (YY1) and the programmed death ligand 1 (PD-L1). We have reported that YY1 leads to immune resistance through increasing HIF-1α accumulation and PD-L1 expression. These mechanisms inhibit the ability of the cytotoxic T-lymphocytes to mediate their cytotoxic functions via the inhibitory signal delivered by the PD-L1 on tumor cells to the PD-1 receptor on cytotoxic T-cells. Thus, means to override these resistance mechanisms are needed to sensitize the tumor cells to both cell killing and inhibition of tumor progression. Treatment with nitric oxide (NO) donors has been shown to sensitize many types of tumors to chemotherapy, immunotherapy, and radiotherapy. Treatment of cancer cell lines with NO donors has resulted in the inhibition of cancer cell activities via, in part, the inhibition of YY1 and PD-L1. The NO-mediated inhibition of YY1 was the result of both the inhibition of the upstream NF-κB pathway as well as the S-nitrosylation of YY1, leading to both the downregulation of YY1 expression as well as the inhibition of YY1-DNA binding activity, respectively. Also, treatment with NO donors induced the inhibition of YY1 and resulted in the inhibition of PD-L1 expression. Based on the above findings, we propose that treatment of tumor cells with the combination of NO donors, at optimal noncytotoxic doses, and anti-tumor cytotoxic effector cells or other conventional therapies will result in a synergistic anticancer activity and tumor regression.
Collapse
Affiliation(s)
- Emily Hays
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California Los Angeles, CA 90095, USA.
| | - Benjamin Bonavida
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California Los Angeles, CA 90095, USA.
| |
Collapse
|
68
|
Trac D, Hoffman JR, Bheri S, Maxwell JT, Platt MO, Davis ME. Predicting Functional Responses of Progenitor Cell Exosome Potential with Computational Modeling. Stem Cells Transl Med 2019; 8:1212-1221. [PMID: 31385648 PMCID: PMC6811701 DOI: 10.1002/sctm.19-0059] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 06/17/2019] [Indexed: 01/08/2023] Open
Abstract
Congenital heart disease can lead to severe right ventricular heart failure (RVHF). We have shown that aggregated c‐kit+ progenitor cells (CPCs) can improve RVHF repair, likely due to exosome‐mediated effects. Here, we demonstrate that miRNA content from monolayer (2D) and aggregated (3D) CPC exosomes can be related to in vitro angiogenesis and antifibrosis responses using partial least squares regression (PLSR). PLSR reduced the dimensionality of the data set to the top 40 miRNAs with the highest weighted coefficients for the in vitro biological responses. Target pathway analysis of these top 40 miRNAs demonstrated significant fit to cardiac angiogenesis and fibrosis pathways. Although the model was trained on in vitro data, we demonstrate that the model can predict angiogenesis and fibrosis responses to exosome treatment in vivo with a strong correlation with published in vivo responses. These studies demonstrate that PLSR modeling of exosome miRNA content has the potential to inform preclinical trials and predict new promising CPC therapies. stem cells translational medicine2019;8:1212–1221
Collapse
Affiliation(s)
- David Trac
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia, USA
| | - Jessica R Hoffman
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Sruti Bheri
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia, USA
| | - Joshua T Maxwell
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Manu O Platt
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia, USA
| | - Michael E Davis
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia, USA.,Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA.,Children's Heart Research & Outcomes (HeRO) Center, Children's Healthcare of Atlanta & Emory University, Atlanta, Georgia, USA
| |
Collapse
|
69
|
Ahmed Alshwafy RY, Dahy AA, Warad I, Mahfouz RM. Synthesis and DFT calculations of new ruthenium(II) nitrosyl complexes using cis-fac-dichlorotetrakis(dimethylsulfoxide)ruthenium(II) precursor and different oximes as sources of nitrosyl ligand. J COORD CHEM 2019. [DOI: 10.1080/00958972.2019.1647340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
| | | | - Ismail Warad
- Chemistry Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Refaat M. Mahfouz
- Chemistry Department, Faculty of Science, Assiut University, Assiut, Egypt
| |
Collapse
|
70
|
Pant J, Pedaparthi S, Hopkins SP, Goudie MJ, Douglass ME, Handa H. Antibacterial and Cellular Response Toward a Gasotransmitter-Based Hybrid Wound Dressing. ACS Biomater Sci Eng 2019; 5:4002-4012. [DOI: 10.1021/acsbiomaterials.9b00737] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Jitendra Pant
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, Georgia 30602, United States
| | - Shriya Pedaparthi
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, Georgia 30602, United States
| | - Sean P. Hopkins
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, Georgia 30602, United States
| | - Marcus J. Goudie
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, Georgia 30602, United States
| | - Megan E. Douglass
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, Georgia 30602, United States
| | - Hitesh Handa
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, Georgia 30602, United States
| |
Collapse
|
71
|
Hirunpattarasilp C, Attwell D, Freitas F. The role of pericytes in brain disorders: from the periphery to the brain. J Neurochem 2019; 150:648-665. [PMID: 31106417 DOI: 10.1111/jnc.14725] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/15/2019] [Accepted: 05/15/2019] [Indexed: 12/13/2022]
Abstract
It is becoming increasingly apparent that disorders of the brain microvasculature contribute to many neurological disorders. In recent years it has become clear that a major player in these events is the capillary pericyte which, in the brain, is now known to control the blood-brain barrier, regulate blood flow, influence immune cell entry and be crucial for angiogenesis. In this review we consider the under-explored possibility that peripheral diseases which affect the microvasculature, such as hypertension, kidney disease and diabetes, produce central nervous system (CNS) dysfunction by mechanisms affecting capillary pericytes within the CNS. We highlight how cellular messengers produced peripherally can act via signalling pathways within CNS pericytes to reshape blood vessels, restrict blood flow or compromise blood-brain barrier function, thus causing neuronal dysfunction. Increased understanding of how renin-angiotensin, Rho-kinase and PDGFRβ signalling affect CNS pericytes may suggest novel therapeutic approaches to reducing the CNS effects of peripheral disorders.
Collapse
Affiliation(s)
- Chanawee Hirunpattarasilp
- Department of Neuroscience, Andrew Huxley Building, University College London, Physiology & Pharmacology, Gower Street, London, UK
| | - David Attwell
- Department of Neuroscience, Andrew Huxley Building, University College London, Physiology & Pharmacology, Gower Street, London, UK
| | - Felipe Freitas
- Department of Neuroscience, Andrew Huxley Building, University College London, Physiology & Pharmacology, Gower Street, London, UK
| |
Collapse
|
72
|
Kang Y, Kim J, Park J, Lee YM, Saravanakumar G, Park KM, Choi W, Kim K, Lee E, Kim C, Kim WJ. Tumor vasodilation by N-Heterocyclic carbene-based nitric oxide delivery triggered by high-intensity focused ultrasound and enhanced drug homing to tumor sites for anti-cancer therapy. Biomaterials 2019; 217:119297. [PMID: 31255980 DOI: 10.1016/j.biomaterials.2019.119297] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 06/17/2019] [Accepted: 06/20/2019] [Indexed: 11/16/2022]
Abstract
Nitric oxide (NO) is widely known as an effective vasodilator at low concentrations. Drug delivery systems combined with NO can dilate blood vessels surrounding tumor tissues, and the drug accumulation in tumors is accelerated by the enhanced permeability and retention effect, leading to an improvement in the anti-tumor effect. N-heterocyclic carbene-based NO donors (e.g., 1,3-bis-(2,4,6-trimethylphenyl)imidazolylidene nitric oxide (IMesNO) have been developed for stable NO storing in air and water, and NO release by thermolysis. Herein, we demonstrated on-demand NO release by high-intensity focused ultrasound (HIFU) as a stimulus, which generated high heat and exerted an ablation effect when treated in vivo. We demonstrated IMesNO to be a HIFU-responsive NO donor and its potential application in vivo using IMesNO-loaded micelles. Moreover, IMesNO-loaded micelles mixed with drug-loaded micelles (IMesNO/DOX@MCs) showed acceleration of drug accumulation in tumor sites and enhanced tumor growth inhibition. Thus, our findings suggest a potential clinical bioapplication of NO-releasing drug-loaded micelles owing to the therapeutic function of NO and HIFU treatment for anti-cancer therapy.
Collapse
Affiliation(s)
- Youngnam Kang
- School of Interdisciplinary Bioscience and Bioengineering, POSTECH (Pohang University of Science and Technology), Pohang 37673, Republic of Korea
| | - Jeesu Kim
- Department of Creative IT Engineering and Electrical Engineering, POSTECH (Pohang University of Science and Technology), Pohang 37673, Republic of Korea
| | - Junbeom Park
- Center for Self-Assembly and Complexity (CSC), Institute for Basic Science (IBS), Pohang 37673, Republic of Korea; Department of Chemistry, POSTECH (Pohang University of Science and Technology), Pohang 37673, Republic of Korea
| | - Yeong Mi Lee
- Department of Chemistry, POSTECH (Pohang University of Science and Technology), Pohang 37673, Republic of Korea
| | - Gurusamy Saravanakumar
- Department of Chemistry, POSTECH (Pohang University of Science and Technology), Pohang 37673, Republic of Korea
| | - Kyeng Min Park
- Center for Self-Assembly and Complexity (CSC), Institute for Basic Science (IBS), Pohang 37673, Republic of Korea
| | - Wonseok Choi
- Department of Creative IT Engineering and Electrical Engineering, POSTECH (Pohang University of Science and Technology), Pohang 37673, Republic of Korea
| | - Kimoon Kim
- Center for Self-Assembly and Complexity (CSC), Institute for Basic Science (IBS), Pohang 37673, Republic of Korea; Department of Chemistry, POSTECH (Pohang University of Science and Technology), Pohang 37673, Republic of Korea.
| | - Eunsung Lee
- Center for Self-Assembly and Complexity (CSC), Institute for Basic Science (IBS), Pohang 37673, Republic of Korea; Department of Chemistry, POSTECH (Pohang University of Science and Technology), Pohang 37673, Republic of Korea.
| | - Chulhong Kim
- Department of Creative IT Engineering and Electrical Engineering, POSTECH (Pohang University of Science and Technology), Pohang 37673, Republic of Korea.
| | - Won Jong Kim
- School of Interdisciplinary Bioscience and Bioengineering, POSTECH (Pohang University of Science and Technology), Pohang 37673, Republic of Korea; Department of Chemistry, POSTECH (Pohang University of Science and Technology), Pohang 37673, Republic of Korea.
| |
Collapse
|
73
|
1,2-Dihydroxyxanthone: Effect on A375-C5 Melanoma Cell Growth Associated with Interference with THP-1 Human Macrophage Activity. Pharmaceuticals (Basel) 2019; 12:ph12020085. [PMID: 31167479 PMCID: PMC6630936 DOI: 10.3390/ph12020085] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/01/2019] [Accepted: 06/03/2019] [Indexed: 12/13/2022] Open
Abstract
Xanthones have been suggested as prospective candidates for cancer treatment. 1,2- dihydroxyxanthone (1,2-DHX) is known to interfere with the growth of several cancer cell lines. We investigated the effects of 1,2-DHX on the growth of the A375-C5 melanoma cell line and THP-1 human macrophage activity. 1,2-DHX showed a moderate growth inhibition of A375-C5 melanoma cells (concentration that causes a 50% inhibition of cell growth (GI50) = 55.0 ± 2.3 µM), but strongly interfered with THP-1 human macrophage activity. Supernatants from lipopolysaccharide (LPS)-stimulated THP-1 macrophage cultures exposed to 1,2-DHX significantly increased growth inhibition of A375-C5 cells, when compared to supernatants from untreated LPS-stimulated macrophages or to direct treatment with 1,2-DHX only. 1,2-DHX decreased THP-1 secretion of interleukin-1β (IL-1β) and interleukin-10 (IL-10), but stimulated tumor necrosis factor-α (TNF-α) and transforming growth factor-β1 (TGF-β1) production. This xanthone also inhibited nitric oxide (NO) production by RAW 264.7 murine macrophages, possibly through inhibition of inducible NO synthase production. In conclusion, these findings suggest a potential impact of 1,2-DHX in melanoma treatment, not only due to a direct effect on cancer cells but also by modulation of macrophage activity.
Collapse
|
74
|
Augustine R, Prasad P, Khalaf IMN. Therapeutic angiogenesis: From conventional approaches to recent nanotechnology-based interventions. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 97:994-1008. [DOI: 10.1016/j.msec.2019.01.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 12/06/2018] [Accepted: 01/02/2019] [Indexed: 12/27/2022]
|
75
|
Bellafiore M, Battaglia G, Bianco A, Palma A. Expression Pattern of Angiogenic Factors in Healthy Heart in Response to Physical Exercise Intensity. Front Physiol 2019; 10:238. [PMID: 30984008 PMCID: PMC6447665 DOI: 10.3389/fphys.2019.00238] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 02/22/2019] [Indexed: 12/22/2022] Open
Abstract
Recently, many studies showing the regeneration potential of both cardiac and hematopoietic stem cells in adult heart following injury were definitively retracted by the literature. Therefore, stimulating myocardial angiogenesis becomes to be important for preventing cardiovascular diseases. Regular endurance exercise has been reported to induce capillary growth in healthy and diseased myocardium resulting in cardioprotective phenotype. Previously, we demonstrated a significantly increased capillary proliferation in mouse hearts following 30 and 45 days of endurance training. In the present study, we examined the localization and expression pattern of vascular endothelial growth factor receptors (VEGFR-1/Flt-1 and VEGFR-2/Flk-1), hypoxia-inducible factor-1α (HIF-1α), and inducible nitric oxide synthase (iNOS) in heart neocapillarization in response to a mild, moderate, and high intensity of endurance training. Sixty-three Swiss male mice were divided into four untrained control groups and three groups trained for 15 (T15), 30 (T30), and 45 (T45) days with a gradually increasing intensity on a treadmill. We observed the localization of studied proteins with immunostaining and their expression level with Western blot analyses. We found that VEGFR-2/Flk-1 expression progressively increased in trained groups compared with controls, while VEGFR-1/Flt-1 and HIF-1α were higher in T15 than in controls, T30, and T45 animals. Differently, iNOS levels enhanced after 15 and 30 days of exercise. The localization of these factors was not altered by exercise. The results showed that the expression of VEGFR-1/Flt-1, VEGFR-2/Flk-1, HIF-1α, and iNOS is differently regulated in cardiac angiogenesis according to the exercise intensity. VEGFR-1/Flt-1 and HIF-1α are upregulated by a mild intensity exercise, while VEGFR-2/Flk-1 progressively enhances with increasing workload. Differently, iNOS protein is modulated by a moderate intensity exercise. VEGF pathway appears to be involved in exercise-related angiogenesis in heart and VEGF might act in a paracrine and endocrine manner. Understanding this relationship is important for developing exercise strategies to protect the heart by insults.
Collapse
Affiliation(s)
- Marianna Bellafiore
- Department of Psychology, Educational Science and Human Movement, University of Palermo, Palermo, Italy
| | - Giuseppe Battaglia
- Department of Psychology, Educational Science and Human Movement, University of Palermo, Palermo, Italy
| | - Antonino Bianco
- Department of Psychology, Educational Science and Human Movement, University of Palermo, Palermo, Italy
| | - Antonio Palma
- Department of Psychology, Educational Science and Human Movement, University of Palermo, Palermo, Italy
| |
Collapse
|
76
|
Li SJ, Zhou DY, Li Y, Liu HW, Wu P, Ou-Yang J, Jiang WL, Li CY. Efficient Two-Photon Fluorescent Probe for Imaging of Nitric Oxide during Endoplasmic Reticulum Stress. ACS Sens 2018; 3:2311-2319. [PMID: 30375854 DOI: 10.1021/acssensors.8b00567] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Nitric oxide (NO) is a vital gaseous signal molecule and plays an important role in diverse physiological and pathological processes including regulation of vascular functions. Endoplasmic reticulum (ER) stress is caused by the accumulation of misfolded or unfolded protein in the ER. Besides, ER stress induced by NO can be involved in the pathogenesis of various vascular diseases. Unfortunately, to the best of our knowledge, no ER-targeting probe for NO is reported to study the relationship between ER stress and the level of NO in a biological system. Herein, an ER-targeted fluorescent probe named ER-Nap-NO for imaging of NO is designed and synthesized. ER-Nap-NO consists of three main parts: naphthalimide (two-photon fluorophore), o-phenylenediamino (NO recognition group), and methyl sulfonamide (ER-targetable group). The probe itself is nonfluorescent because a photoinduced electron transfer (PET) process exists. After the addition of NO, the PET process is inhibited and thus strong fluorescence is released. Moreover, the response mechanism is confirmed by 1H NMR and mass spectra and DFT calculation in detail. In addition, from the experimental results, we can conclude that the probe displays several obvious advantages including high sensitivity, selectivity, and ER-targetable ability. Based on these excellent properties, the probe is used for the two-photon imaging of exogenous and endogenous NO in ER of living cells. Most importantly, the ER-targetable probe has potential capability as a tool for investigating the level of NO during tunicamycin-induced ER stress in cells and tissues, which is beneficial for revealing the role of NO in ER-associated vascular diseases.
Collapse
Affiliation(s)
- Song-Jiao Li
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan 411105, People’s Republic of China
| | - Dong-Ye Zhou
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan 411105, People’s Republic of China
| | - Yongfei Li
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan 411105, People’s Republic of China
- College of Chemical Engineering, Xiangtan University, Xiangtan 411105, People’s Republic of China
| | - Hong-Wen Liu
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan 411105, People’s Republic of China
| | - Ping Wu
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan 411105, People’s Republic of China
| | - Juan Ou-Yang
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan 411105, People’s Republic of China
| | - Wen-Li Jiang
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan 411105, People’s Republic of China
| | - Chun-Yan Li
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan 411105, People’s Republic of China
| |
Collapse
|
77
|
Serin M, Altinel D, Leblebici C, Biltekin B, Huseyinbas O, Kurt Yazar S, Irmak F, Sonmez A, Bayramicli M. Subdermal nitrous oxide delivery increases skin microcirculation and random flap survival in rats. J Plast Surg Hand Surg 2018; 53:37-44. [PMID: 30371138 DOI: 10.1080/2000656x.2018.1531013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Random skin flaps are essential tools in reconstructive surgery. In this study, we investigated the effect of subdermal nitrous oxide (N2O) application on random flap survival. In this experimental study, we used 21 female rats in three groups. In the N2O and air groups, gases were administrated under the proposed dorsal flap areas daily for seven days. Following the treatment period, flaps were raised and inserted back into their place from the dorsal skin. In the control group, the flaps were elevated and inserted back to their place without any pretreatment. Calculation of necrotic flap areas, histological examination and microangiography was performed to evaluate the results 7 days after the flap surgery. The average of necrotic flap area in the N2O, air and control group was 13.45%, 37.67% and 46.43%, respectively. (N2O vs air p = .044; N2O vs control p = .003). The average number of capillary formations identified in the histological analysis was 7.0 ± 1.58, 3.75 ± 2.36 and 4.4 ± 0.54 in the N2O, air and control group, respectively. (N2O vs air p = .017; N2O vs control p = .037). The average number of capillary structures identified in the angiography images were 6.3 ± 1.52, 1.6 ± 1.15 and 1.3 ± 0.57 in the N2O, air and control group, respectively. (N2O vs air p = .04; N2O vs control p = .02). We conclude that subdermal N2O application increases random flap survival through an increase in the skin microcirculation and could be promising for future clinical applications.
Collapse
Affiliation(s)
- Merdan Serin
- a Department of Plastic and Reconstructive Surgery , Istanbul Training and Research Hospital, University of Health Sciences , Istanbul , Turkey
| | - Dincer Altinel
- a Department of Plastic and Reconstructive Surgery , Istanbul Training and Research Hospital, University of Health Sciences , Istanbul , Turkey
| | - Cem Leblebici
- b Department of Pathology , Istanbul Training and Research Hospital, University of Health Sciences , Istanbul , Turkey
| | - Burcu Biltekin
- c Department of Histology and Embryology , Cerrahpasa Medical School, Istanbul University , Istanbul , Turkey
| | - Onder Huseyinbas
- d Animal Research Laboratory , Istanbul Bezmialem University Medical School , Istanbul , Turkey
| | - Sevgi Kurt Yazar
- a Department of Plastic and Reconstructive Surgery , Istanbul Training and Research Hospital, University of Health Sciences , Istanbul , Turkey
| | - Fatih Irmak
- e Department of Plastic and Reconstructive Surgery , Istanbul Sisli Etfal Training and Research Hospital, University of Health Sciences , Istanbul , Turkey
| | - Ahmet Sonmez
- f Private practice in Plastic and Reconstructive Surgery , Istanbul , Turkey
| | - Mehmet Bayramicli
- g Department of Plastic and Reconstructive Surgery , Marmara University School of Medicine , Istanbul , Turkey
| |
Collapse
|
78
|
Kashyap D, Sharma A, Tuli HS, Sak K, Garg VK, Buttar HS, Setzer WN, Sethi G. Apigenin: A natural bioactive flavone-type molecule with promising therapeutic function. J Funct Foods 2018. [DOI: 10.1016/j.jff.2018.07.037] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
79
|
Edmonds ME, Bodansky HJ, Boulton AJM, Chadwick PJ, Dang CN, D'Costa R, Johnston A, Kennon B, Leese G, Rajbhandari SM, Serena TE, Young MJ, Stewart JE, Tucker AT, Carter MJ. Multicenter, randomized controlled, observer-blinded study of a nitric oxide generating treatment in foot ulcers of patients with diabetes-ProNOx1 study. Wound Repair Regen 2018; 26:228-237. [PMID: 29617058 DOI: 10.1111/wrr.12630] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 03/26/2018] [Indexed: 12/24/2022]
Abstract
The aim of this multicenter, prospective, observer-blinded, parallel group, randomized controlled trial was to assess the safety and efficacy of EDX110, a nitric oxide generating medical device, in the treatment of diabetic foot ulcers in a patient group reflecting "real world" clinical practice compared against optimal standard care. Participants were recruited from ten hospital sites in multidisciplinary foot ulcer clinics. The ulcers were full thickness, with an area of 25-2,500 mm2 and either a palpable pedal pulse or ankle brachial pressure index > 0.5. Infected ulcers were included. Treatment lasted 12 weeks, or until healed, with a 12-week follow-up period. Both arms were given optimal debridement, offloading and antimicrobial treatment, the only difference being the fixed used of EDX110 as the wound dressing in the EDX110 group. 135 participants were recruited with 148 ulcers (EDX110-75; Control-73), 30% of which were clinically infected at baseline. EDX110 achieved its primary endpoint by attaining a median Percentage Area Reduction of 88.6% compared to 46.9% for the control group (p = 0.016) at 12 weeks in the intention-to-treat population. There was no significant difference between wound size reduction achieved by EDX110 after 4 weeks and the wound size reduction achieved in the control group after 12 weeks. EDX110 was well tolerated. Thirty serious adverse events were reported (12 in the EDX110 group, of which 4 were related to the ulcer; 18 in the control group, of which 10 were related and 1 possibly related to the ulcer), with significant reduction in serious adverse events related to the ulcer in EDX group. There was no significant difference in adverse events. This study, in a real world clinical foot ulcer population, demonstrates the ability of EDX110 to improve healing, as measured by significantly reducing the ulcer area, compared to current best clinical practice.
Collapse
Affiliation(s)
- Michael E Edmonds
- Diabetic Foot Clinic, King's College Hospital, London, United Kingdom
| | - Harvey J Bodansky
- Diabetes Centre, St James's University Hospital, Leeds, West Yorkshire, United Kingdom
| | - Andrew J M Boulton
- Manchester Diabetes Centre, Manchester Royal Infirmary, Manchester, United Kingdom
| | - Paul J Chadwick
- Podiatry and Foot Health, Salford Royal Hospital (NHS) Foundation Trust, Salford, United Kingdom
| | - Cuong N Dang
- Diabetes Centre, North Manchester General Hospital, Manchester, United Kingdom
| | - Ryan D'Costa
- Diabetes Centre, Pinderfields Hospital, Wakefield, United Kingdom
| | - Atholl Johnston
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Brian Kennon
- Diabetes Centre, Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - Graham Leese
- Diabetes Clinic, Ninewells Hospital, Dundee, United Kingdom
| | - Satyan M Rajbhandari
- Diabetes Centre, Chorley & South Ribble Hospital, Preston Road, University of Central Lancashire, Chorley, United Kingdom
| | | | - Matthew J Young
- Diabetic Foot Clinic, Royal Infirmary of Edinburgh, Edinburgh, United Kingdom
| | - Joanne E Stewart
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Arthur T Tucker
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | | |
Collapse
|
80
|
Srivastava A, Shukla V, Tiwari D, Gupta J, Kumar S, Kumar A. Targeted therapy of chronic liver diseases with the inhibitors of angiogenesis. Biomed Pharmacother 2018; 105:256-266. [PMID: 29859468 DOI: 10.1016/j.biopha.2018.05.102] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 05/09/2018] [Accepted: 05/21/2018] [Indexed: 01/09/2023] Open
Abstract
Angiogenesis appears to be intrinsically associated with the progression of chronic liver diseases, which eventually leads to the development of cirrhosis and related complications, including hepatocellular carcinoma. Several studies have suggested that this association is relevant for chronic liver disease (CLD) progression, with angiogenesis. The fact that angiogenesis plays a pivotal role in CLDs gives rise to new opportunities for treating CLDs. Inhibitor of angiogenesis has proved effective for the treatment of patients suffering from CLD. However, it is limited in diagnosis. The last decade has witnessed a plethora of publications which elucidate the potential of angiogenesis inhibitors for the therapy of CLD. The close relationship between the progression of CLDs and angiogenesis emphasizes the need for anti-angiogenic therapy to block/slow down CLD progression. The present review summarizes all these discussions, the results of the related studies carried out to date and the future prospects in this field. We discuss liver angiogenesis in normal and pathophysiologic conditions with a focus on the role and future use of angiogenic factors as second-line treatment of CLD. This review compiles relevant findings and offers opinions that have emerged in last few years relating liver angiogenesis and its treatment using anti-angiogenic factors.
Collapse
Affiliation(s)
- Ankita Srivastava
- Faculty of Biotechnology, Institute of Biosciences and Technology, Shri Ramswaroop Memorial University, Barabanki, Uttar Pradesh, India
| | - Vanistha Shukla
- Faculty of Biotechnology, Institute of Biosciences and Technology, Shri Ramswaroop Memorial University, Barabanki, Uttar Pradesh, India
| | - Deepika Tiwari
- Faculty of Biotechnology, Institute of Biosciences and Technology, Shri Ramswaroop Memorial University, Barabanki, Uttar Pradesh, India
| | - Jaya Gupta
- Faculty of Biotechnology, Institute of Biosciences and Technology, Shri Ramswaroop Memorial University, Barabanki, Uttar Pradesh, India
| | - Sunil Kumar
- Faculty of Biotechnology, Institute of Biosciences and Technology, Shri Ramswaroop Memorial University, Barabanki, Uttar Pradesh, India.
| | - Awanish Kumar
- Department of Biotechnology, National Institute of Technology, Raipur, Chhattisgarh, India.
| |
Collapse
|
81
|
Saghiri MA, Asatourian A, Nguyen EH, Wang S, Sheibani N. Hydrogel Arrays and Choroidal Neovascularization Models for Evaluation of Angiogenic Activity of Vital Pulp Therapy Biomaterials. J Endod 2018; 44:773-779. [PMID: 29602530 PMCID: PMC6300997 DOI: 10.1016/j.joen.2018.02.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 01/29/2018] [Accepted: 02/06/2018] [Indexed: 12/22/2022]
Abstract
INTRODUCTION This study intended to evaluate the angiogenic properties of vital pulp therapy materials including white mineral trioxide aggregate (WMTA), calcium hydroxide (Ca[OH]2), Geristore (Den-Mat, Santa Maria, CA), and nano WMTA biomaterials. METHODS WMTA, Ca(OH)2, Geristore, and nano WMTA disks were prepared, dispersed into 2 mL Milli-Q (Millipore, ThermoFisher, Hanover Park, IL) distilled water, and centrifuged to obtain 2 mL supernatant elution. Thirty-five wells of polyethylene glycol hydrogel arrays were prepared and divided into 5 groups of 7 (n = 7). Mice molar endothelial cells (ECs) were placed on hydrogel arrays. The elution prepared from each sample was diluted in growth medium (1:3) and added to the hydrogel arrays. The EC medium alone was used for the control. For the choroidal neovascularization (CNV) model, thirty-five 6-week-old female mice were lasered and divided into 5 groups, and elution from each sample (2 μL) or saline (control) was delivered by intravitreal injection on the day of the laser treatment and 1 week later. The mean number of nodes, the total length of the branches in the hydrogel arrays, and the mean area of CNV were calculated using ImageJ software (National Institutes of Health, Bethesda, MD) and analyzed by 1-way analysis of variance and post hoc Tukey honest significant difference tests. RESULTS The comparison of results regarding the number of nodes showed the values of control > Geristore > nano WMTA > WMTA > Ca(OH)2. Regarding the total branch length and the CNV area, the comparison of results showed values of Geristore > control > nano WMTA > WMTA > Ca(OH)2. CONCLUSIONS All tested materials showed minimal antiangiogenic activity, whereas Geristore and nano WMTA showed a higher proangiogenic activity than WMTA and Ca(OH)2.
Collapse
Affiliation(s)
- Mohammad Ali Saghiri
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin; Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin.
| | - Armen Asatourian
- Sector of Angiogenesis Regenerative Medicine, Dr Hajar Afsar Lajevardi Research Cluster, Shiraz, Iran
| | - Eric H Nguyen
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin; Human Models for Analysis of Pathways (HMAPs) Center, University of Wisconsin, Madison, Wisconsin
| | - Shoujian Wang
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Nader Sheibani
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin; Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin; Human Models for Analysis of Pathways (HMAPs) Center, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|
82
|
Chen CY, Lin WC, Wong KL, Cheng KS, Leung YM, Yang SE. Gossypol stimulates opening of a Ca2+
- and Na+
-permeable but Ni2+
- and Co2+
-impermeable pore in bEND.3 endothelial cells. Clin Exp Pharmacol Physiol 2018; 45:788-796. [PMID: 29498086 DOI: 10.1111/1440-1681.12929] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 02/13/2018] [Accepted: 02/17/2018] [Indexed: 12/29/2022]
Affiliation(s)
- Cing-Yu Chen
- School of Pharmacy; China Medical University; Taichung Taiwan
| | - Wen-Chuan Lin
- School of Pharmacy; China Medical University; Taichung Taiwan
| | - Kar-Lok Wong
- Department of Anesthesiology; China Medical University Hospital; Taichung Taiwan
| | - Ka-Shun Cheng
- Department of Anesthesiology; China Medical University Hospital; Taichung Taiwan
- Department of Anesthesiology; The Qingdao University Yuhuangding Hospital; Yantai Shandong China
| | - Yuk-Man Leung
- Department of Physiology; China Medical University; Taichung Taiwan
| | - Shu-Er Yang
- Department of Beauty Science and Graduate Institute of Beauty Science and Technology; Chienkuo Technology University; Changhua Taiwan
| |
Collapse
|
83
|
Basavarajappa HD, Sulaiman RS, Qi X, Shetty T, Sheik Pran Babu S, Sishtla KL, Lee B, Quigley J, Alkhairy S, Briggs CM, Gupta K, Tang B, Shadmand M, Grant MB, Boulton ME, Seo SY, Corson TW. Ferrochelatase is a therapeutic target for ocular neovascularization. EMBO Mol Med 2018; 9:786-801. [PMID: 28377496 PMCID: PMC5452042 DOI: 10.15252/emmm.201606561] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Ocular neovascularization underlies major blinding eye diseases such as “wet” age‐related macular degeneration (AMD). Despite the successes of treatments targeting the vascular endothelial growth factor (VEGF) pathway, resistant and refractory patient populations necessitate discovery of new therapeutic targets. Using a forward chemical genetic approach, we identified the heme synthesis enzyme ferrochelatase (FECH) as necessary for angiogenesis in vitro and in vivo. FECH is overexpressed in wet AMD eyes and murine choroidal neovascularization; siRNA knockdown of Fech or partial loss of enzymatic function in the Fechm1Pas mouse model reduces choroidal neovascularization. FECH depletion modulates endothelial nitric oxide synthase function and VEGF receptor 2 levels. FECH is inhibited by the oral antifungal drug griseofulvin, and this compound ameliorates choroidal neovascularization in mice when delivered intravitreally or orally. Thus, FECH inhibition could be used therapeutically to block ocular neovascularization.
Collapse
Affiliation(s)
- Halesha D Basavarajappa
- Eugene and Marilyn Glick Eye Institute and Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Rania S Sulaiman
- Eugene and Marilyn Glick Eye Institute and Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Xiaoping Qi
- Eugene and Marilyn Glick Eye Institute and Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Trupti Shetty
- Eugene and Marilyn Glick Eye Institute and Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sardar Sheik Pran Babu
- Eugene and Marilyn Glick Eye Institute and Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kamakshi L Sishtla
- Eugene and Marilyn Glick Eye Institute and Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Bit Lee
- College of Pharmacy, Gachon University, Incheon, South Korea
| | - Judith Quigley
- Eugene and Marilyn Glick Eye Institute and Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sameerah Alkhairy
- Eugene and Marilyn Glick Eye Institute and Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Christian M Briggs
- Eugene and Marilyn Glick Eye Institute and Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kamna Gupta
- Eugene and Marilyn Glick Eye Institute and Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Buyun Tang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Mehdi Shadmand
- Eugene and Marilyn Glick Eye Institute and Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Maria B Grant
- Eugene and Marilyn Glick Eye Institute and Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Michael E Boulton
- Eugene and Marilyn Glick Eye Institute and Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Seung-Yong Seo
- College of Pharmacy, Gachon University, Incheon, South Korea
| | - Timothy W Corson
- Eugene and Marilyn Glick Eye Institute and Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA .,Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
84
|
Mandal P. Insight of nitric oxide signaling: A potential biomarker with multifaceted complex mechanism in colorectal carcinogenesis. Biochem Biophys Res Commun 2018; 495:1766-1768. [DOI: 10.1016/j.bbrc.2017.12.051] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 12/08/2017] [Indexed: 10/25/2022]
|
85
|
Developmental changes of l-arginine transport at the blood-brain barrier in rats. Microvasc Res 2017; 117:16-21. [PMID: 29247719 DOI: 10.1016/j.mvr.2017.12.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 11/25/2017] [Accepted: 12/12/2017] [Indexed: 11/23/2022]
Abstract
l-Arginine is required for regulating synapse formation/patterning and angiogenesis in the developing brain. We hypothesized that this requirement would be met by increased transporter-mediated supply across the blood-brain barrier (BBB). Thus, the purpose of this work was to test the idea that elevation of blood-to-brain l-arginine transport across the BBB in the postnatal period coincides with up-regulation of cationic acid transporter 1 (CAT1) expression in developing brain capillaries. We found that the apparent brain-to-plasma concentration ratio (Kp, app) of l-arginine after intravenous administration during the first and second postnatal weeks was 2-fold greater than that at the adult stage. Kp, app of l-serine was also increased at the first postnatal week. In contrast, Kp, app of d-mannitol, a passively BBB-permeable molecule, did not change, indicating that increased transport of l-arginine and l-serine is not due to BBB immaturity. Double immunohistochemical staining of CAT1 and a marker protein, glucose transporter 1, revealed that CAT1 was localized on both luminal and abluminal membranes of brain capillary endothelial cells during the developmental and adult stages. A dramatic increase in CAT1 expression in the brain was seen at postnatal day 7 (P7) and day 14 (P14) and the expression subsequently decreased as the brain matured. In accordance with this, intense immunostaining of CAT1 was observed in brain capillaries at P7 and P14. These findings strongly support our hypothesis and suggest that the supply of blood-born l-arginine to the brain via CAT1 at the BBB plays a key role in meeting the elevated demand for l-arginine in postnatal brain.
Collapse
|
86
|
Weitnauer M, Petry A, BelAiba R, Görlach A. Inhibition of endothelial nitric oxyde synthase increases capillary formation via Rac1-dependent induction of hypoxia-inducible factor-1α and plasminogen activator inhibitor-1. Thromb Haemost 2017; 108:849-62. [DOI: 10.1160/th12-04-0277] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Accepted: 09/10/2012] [Indexed: 12/24/2022]
Abstract
SummaryDisruption of endothelial homeostasis results in endothelial dysfunction, characterised by a dysbalance between nitric oxide (NO) and reactive oxygen species (ROS) levels often accompanied by a prothrombotic and proproliferative state. The serine protease thrombin not only is instrumental in formation of the fibrin clot, but also exerts direct effects on the vessel wall by activating proliferative and angiogenic responses. In endothelial cells, thrombin can induce NO as well as ROS levels. However, the relative contribution of these reactive species to the angiogenic response towards thrombin is not completely clear. Since plasminogen activator inhibitor-1 (PAI-1), a direct target of the proangiogenic transcription factors hypoxia-inducible factors (HIFs), exerts prothrombotic and proangiogenic activities we investigated the role of ROS and NO in the regulation of HIF-1α, PAI-1 and capillary formation in response to thrombin. Thrombin enhanced the formation of NO as well as ROS generation involving the GTPase Rac1 in endothelial cells. Rac1-dependent ROS formation promoted induction of HIF-1α, PAI-1 and capillary formation by thrombin, while NO reduced ROS bioavailability and subsequently limited induction of HIF-1α, PAI-1 and the angiogenic response. Importantly, thrombin activation of Rac1 was diminished by NO, but enhanced by ROS. Thus, our findings show that capillary formation induced by thrombin via Rac1-dependent activation of HIF-1 and PAI-1 is limited by the concomitant release of NO which reduced ROS bioavailability. Rac1 activity is sensitive to ROS and NO, thereby playing an essential role in fine tuning the endothelial response to thrombin.
Collapse
|
87
|
Jin S, Qi X, Wang T, Ren L, Yang K, Zhong H. In vitro study of stimulation effect on endothelialization by a copper bearing cobalt alloy. J Biomed Mater Res A 2017; 106:561-569. [PMID: 28984065 DOI: 10.1002/jbm.a.36263] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 08/29/2017] [Accepted: 09/12/2017] [Indexed: 01/03/2023]
Abstract
Endothelialization is an important process after stenting in coronary artery. Recovery of the injured site timely can reduce the neointima formation and platelet absorbance, leading to a lower risk of in-stent restenosis. Copper is known to be critical in vascular construction. Thus a combination of copper with stent materials is a meaningful attempt. A copper bearing L605-Cu cobalt alloy was prepared and its effect on human umbilical vein endothelial cells (HUVECs) was evaluated in vitro in this study. It was found that HUVECs attached and stretched better on the surface of L605-Cu compared with L605, and the apoptosis of cells was decreased simultaneously. The migration and tube formation of HUVECs were also enhanced by the extract of L605-Cu. Furthermore, L605-Cu increased the mRNA expression of VEGF in HUVECs significantly. However it had no effect on the secretion of NO or mRNA expression of eNOS. The result of blood clotting test indicated that L605-Cu had better blood compatibility. These results above have demonstrated that the L605-Cu alloy is promising to be a new stent material with function of accelerating endothelialization. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 561-569, 2018.
Collapse
Affiliation(s)
- Shujing Jin
- School of Materials Science and Engineering, Dalian University of Technology, Dalian, 116024, China.,Institute of Metal Research, Chinese Academy of Sciences, 72 Wenhua Road, Shenyang, 110016, China
| | - Xun Qi
- Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.,Key Laboratory of Diagnostic Imaging and Interventional Radiology of Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Tongmin Wang
- School of Materials Science and Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Ling Ren
- Institute of Metal Research, Chinese Academy of Sciences, 72 Wenhua Road, Shenyang, 110016, China
| | - Ke Yang
- Institute of Metal Research, Chinese Academy of Sciences, 72 Wenhua Road, Shenyang, 110016, China
| | - Hongshan Zhong
- Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.,Key Laboratory of Diagnostic Imaging and Interventional Radiology of Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| |
Collapse
|
88
|
Nemati S, Rezabakhsh A, Khoshfetrat AB, Nourazarian A, Biray Avci Ç, Goker Bagca B, Alizadeh Sardroud H, Khaksar M, Ahmadi M, Delkhosh A, Sokullu E, Rahbarghazi R. Alginate-gelatin encapsulation of human endothelial cells promoted angiogenesis in in vivo and in vitro milieu. Biotechnol Bioeng 2017; 114:2920-2930. [DOI: 10.1002/bit.26395] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 07/31/2017] [Accepted: 08/01/2017] [Indexed: 01/04/2023]
Affiliation(s)
- Sorour Nemati
- Chemical Engineering Faculty; Sahand University of Technology; Tabriz Iran
- Stem Cell Research Center; Tabriz University of Medical Sciences; Tabriz Iran
| | - Aysa Rezabakhsh
- Department of Pharmacology and Toxicology; Faculty of Pharmacy; Tabriz University of Medical Sciences; Tabriz Iran
| | | | - Alireza Nourazarian
- Department of Biochemistry and Clinical Laboratories; Faculty of Medicine; Tabriz University of Medical Sciences; Tabriz Iran
| | - Çığır Biray Avci
- Department of Medical Biology; Faculty of Medicine; Ege University; Izmir Turkey
| | - Bakiye Goker Bagca
- Department of Medical Biology; Faculty of Medicine; Ege University; Izmir Turkey
| | | | - Majid Khaksar
- Stem Cell Research Center; Tabriz University of Medical Sciences; Tabriz Iran
| | - Mahdi Ahmadi
- Department of Physiology; Faculty of Medicine; Tabriz University of Medical Sciences; Tabriz Iran
| | - Aref Delkhosh
- Stem Cell Research Center; Tabriz University of Medical Sciences; Tabriz Iran
| | - Emel Sokullu
- Izmir Katip Celebi University; Bioengineering Department; Izmir Turkey
- Harvard Medical School; Division of Biomedical Engineering at Brigham and Women's Hospital, Harvard-MIT Health Sciences and Technology; Cambridge MA
| | - Reza Rahbarghazi
- Stem Cell Research Center; Tabriz University of Medical Sciences; Tabriz Iran
- Department of Applied Cell Sciences; Faculty of Advanced Medical Sciences; Tabriz University of Medical Sciences; Tabriz Iran
| |
Collapse
|
89
|
The Role of Nitric Oxide from Neurological Disease to Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1007:71-88. [PMID: 28840553 DOI: 10.1007/978-3-319-60733-7_5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
90
|
Kurusamy S, López-Maderuelo D, Little R, Cadagan D, Savage AM, Ihugba JC, Baggott RR, Rowther FB, Martínez-Martínez S, Arco PGD, Murcott C, Wang W, Francisco Nistal J, Oceandy D, Neyses L, Wilkinson RN, Cartwright EJ, Redondo JM, Armesilla AL. Selective inhibition of plasma membrane calcium ATPase 4 improves angiogenesis and vascular reperfusion. J Mol Cell Cardiol 2017; 109:38-47. [PMID: 28684310 DOI: 10.1016/j.yjmcc.2017.07.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 06/12/2017] [Accepted: 07/03/2017] [Indexed: 02/04/2023]
Abstract
AIMS Ischaemic cardiovascular disease is a major cause of morbidity and mortality worldwide. Despite promising results from pre-clinical animal models, VEGF-based strategies for therapeutic angiogenesis have yet to achieve successful reperfusion of ischaemic tissues in patients. Failure to restore efficient VEGF activity in the ischaemic organ remains a major problem in current pro-angiogenic therapeutic approaches. Plasma membrane calcium ATPase 4 (PMCA4) negatively regulates VEGF-activated angiogenesis via inhibition of the calcineurin/NFAT signalling pathway. PMCA4 activity is inhibited by the small molecule aurintricarboxylic acid (ATA). We hypothesize that inhibition of PMCA4 with ATA might enhance VEGF-induced angiogenesis. METHODS AND RESULTS We show that inhibition of PMCA4 with ATA in endothelial cells triggers a marked increase in VEGF-activated calcineurin/NFAT signalling that translates into a strong increase in endothelial cell motility and blood vessel formation. ATA enhances VEGF-induced calcineurin signalling by disrupting the interaction between PMCA4 and calcineurin at the endothelial-cell membrane. ATA concentrations at the nanomolar range, that efficiently inhibit PMCA4, had no deleterious effect on endothelial-cell viability or zebrafish embryonic development. However, high ATA concentrations at the micromolar level impaired endothelial cell viability and tubular morphogenesis, and were associated with toxicity in zebrafish embryos. In mice undergoing experimentally-induced hindlimb ischaemia, ATA treatment significantly increased the reperfusion of post-ischaemic limbs. CONCLUSIONS Our study provides evidence for the therapeutic potential of targeting PMCA4 to improve VEGF-based pro-angiogenic interventions. This goal will require the development of refined, highly selective versions of ATA, or the identification of novel PMCA4 inhibitors.
Collapse
Affiliation(s)
- Sathishkumar Kurusamy
- Cardiovascular Molecular Pharmacology Laboratory, School of Pharmacy, University of Wolverhampton, Wolverhampton, UK
| | - Dolores López-Maderuelo
- Gene Regulation in Cardiovascular Remodelling and Inflammation Group, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; CIBERCV, Spain
| | - Robert Little
- Division of Cardiovascular Sciences, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, UK
| | - David Cadagan
- Cardiovascular Molecular Pharmacology Laboratory, School of Pharmacy, University of Wolverhampton, Wolverhampton, UK
| | - Aaron M Savage
- Department of Infection, Immunity & Cardiovascular Disease & Bateson Centre, University of Sheffield, UK
| | - Jude C Ihugba
- Cardiovascular Molecular Pharmacology Laboratory, School of Pharmacy, University of Wolverhampton, Wolverhampton, UK
| | - Rhiannon R Baggott
- Cardiovascular Molecular Pharmacology Laboratory, School of Pharmacy, University of Wolverhampton, Wolverhampton, UK
| | - Farjana B Rowther
- Brain Tumor UK Neuro-oncology Research Centre, University of Wolverhampton, Wolverhampton, UK
| | - Sara Martínez-Martínez
- Gene Regulation in Cardiovascular Remodelling and Inflammation Group, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; CIBERCV, Spain
| | - Pablo Gómez-Del Arco
- Gene Regulation in Cardiovascular Remodelling and Inflammation Group, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; CIBERCV, Spain; Department of Molecular Biology, Universidad Autonoma de Madrid (C.B.M.S.O.), Madrid, Spain
| | - Clare Murcott
- Cardiovascular Molecular Pharmacology Laboratory, School of Pharmacy, University of Wolverhampton, Wolverhampton, UK
| | - Weiguang Wang
- Oncology Laboratory, Research Institute in Healthcare Science, Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton, UK
| | - J Francisco Nistal
- Cardiovascular Surgery, Hospital Universitario Marqués de Valdecilla, IDIVAL, Facultad de Medicina, Universidad de Cantabria, Santander, Spain
| | - Delvac Oceandy
- Division of Cardiovascular Sciences, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, UK
| | - Ludwig Neyses
- Division of Cardiovascular Sciences, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, UK; University of Luxembourg, Luxembourg
| | - Robert N Wilkinson
- Department of Infection, Immunity & Cardiovascular Disease & Bateson Centre, University of Sheffield, UK
| | - Elizabeth J Cartwright
- Division of Cardiovascular Sciences, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, UK
| | - Juan Miguel Redondo
- Gene Regulation in Cardiovascular Remodelling and Inflammation Group, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; CIBERCV, Spain.
| | - Angel Luis Armesilla
- Cardiovascular Molecular Pharmacology Laboratory, School of Pharmacy, University of Wolverhampton, Wolverhampton, UK; CIBERCV, Spain.
| |
Collapse
|
91
|
Pant J, Goudie MJ, Hopkins S, Brisbois EJ, Handa H. Tunable Nitric Oxide Release from S-Nitroso-N-acetylpenicillamine via Catalytic Copper Nanoparticles for Biomedical Applications. ACS APPLIED MATERIALS & INTERFACES 2017; 9:15254-15264. [PMID: 28409633 PMCID: PMC8007131 DOI: 10.1021/acsami.7b01408] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
The quest for novel therapies to prevent bacterial infections and blood clots (thrombosis) is of utmost importance in biomedical research due to the exponential growth in the cases of thrombosis and blood infections and the emergence of multi-drug-resistant strains of bacteria. Endogenous nitric oxide (NO) is a cellular signaling molecule that plays a pivotal role in host immunity against pathogens, prevention of clotting, and regulation of systemic blood pressure, among several other biological functions. The physiological effect of NO is dose dependent, which necessitates the study of its tunable release kinetics, which is the objective of this study. In the present study, polymer composites were fabricated by incorporating S-nitroso-N-acetylpenicillamine (SNAP) in a medical-grade polymer, Carbosil, and top-coated with varying concentrations of catalytic copper nanoparticles (Cu-NPs). The addition of the Cu-NPs increased the NO release, as well as the overall antimicrobial activity via the oligodynamic effect of Cu. SNAP (10 wt %) composites without Cu-NP coatings showed a NO flux of 1.32 ± 0.6 × 10-10 mol min-1 cm-2, whereas Cu-NP-incorporated SNAP films exhibited fluxes of 4.48 ± 0.5 × 10-10, 4.84 ± 0.3 × 10-10, and 11.7 ± 3.6 × 10-10 mol min-1 cm-2 with 1, 3, and 5 wt % Cu-NPs, respectively. This resulted in a significant reduction (up to 99.8%) in both gram-positive and gram-negative bacteria, with very low platelet adhesion (up to 92% lower) as compared to that of the corresponding controls. Copper leachates from the SNAP films were detected using the inductively coupled plasma-mass spectrometry technique and were found to be significantly lower in concentration than the recommended safety limit by the FDA. The cell viability test performed on mouse fibroblast 3T3 cells provided supportive evidence for the biocompatibility of the material in vitro.
Collapse
Affiliation(s)
- Jitendra Pant
- School of Chemical, Materials and Biomedical Engineering, University of Georgia, Athens, USA
| | - Marcus J. Goudie
- School of Chemical, Materials and Biomedical Engineering, University of Georgia, Athens, USA
| | - Sean Hopkins
- School of Chemical, Materials and Biomedical Engineering, University of Georgia, Athens, USA
| | | | - Hitesh Handa
- School of Chemical, Materials and Biomedical Engineering, University of Georgia, Athens, USA
- Corresponding author: Hitesh Handa, School of Chemical, Materials, and Biomedical Engineering, University of Georgia, 220 Riverbend Road, Athens, GA 30605, Telephone: (706) 542-8109,
| |
Collapse
|
92
|
Dong X, Chang J, Li H. Bioglass promotes wound healing through modulating the paracrine effects between macrophages and repairing cells. J Mater Chem B 2017; 5:5240-5250. [DOI: 10.1039/c7tb01211j] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Bioglass stimulates macrophages to switch to the M2 phenotype and modulates the paracrine effects between macrophages and repairing cells.
Collapse
Affiliation(s)
- Xin Dong
- Med-X Research Institute
- School of Biomedical Engineering
- Shanghai Jiao Tong University
- Shanghai 200030
- China
| | - Jiang Chang
- Med-X Research Institute
- School of Biomedical Engineering
- Shanghai Jiao Tong University
- Shanghai 200030
- China
| | - Haiyan Li
- Med-X Research Institute
- School of Biomedical Engineering
- Shanghai Jiao Tong University
- Shanghai 200030
- China
| |
Collapse
|
93
|
Barui AK, Nethi SK, Patra CR. Investigation of the role of nitric oxide driven angiogenesis by zinc oxide nanoflowers. J Mater Chem B 2017; 5:3391-3403. [DOI: 10.1039/c6tb03323g] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Zinc oxide nanoflowers (ZONF) induce the generation of intracellular ROS that stimulates the phosphorylation of Akt and p38MAPK leading to activation of eNOS to produce NO, triggering angiogenesis in a cGMP dependent manner.
Collapse
Affiliation(s)
- Ayan Kumar Barui
- Department of Chemical Biology
- CSIR-Indian Institute of Chemical Technology
- Hyderabad – 500007
- India
- Academy of Scientific and Innovative Research (AcSIR)
| | - Susheel Kumar Nethi
- Department of Chemical Biology
- CSIR-Indian Institute of Chemical Technology
- Hyderabad – 500007
- India
- Academy of Scientific and Innovative Research (AcSIR)
| | - Chitta Ranjan Patra
- Department of Chemical Biology
- CSIR-Indian Institute of Chemical Technology
- Hyderabad – 500007
- India
- Academy of Scientific and Innovative Research (AcSIR)
| |
Collapse
|
94
|
Mao Y, Wang J, Yu F, Li Z, Li H, Guo C, Fan X. Ghrelin protects against palmitic acid or lipopolysaccharide-induced hepatocyte apoptosis through inhibition of MAPKs/iNOS and restoration of Akt/eNOS pathways. Biomed Pharmacother 2016; 84:305-313. [DOI: 10.1016/j.biopha.2016.09.043] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 09/12/2016] [Accepted: 09/12/2016] [Indexed: 01/24/2023] Open
|
95
|
Kamaleddin MA. The paradoxical pro- and antiangiogenic actions of resveratrol: therapeutic applications in cancer and diabetes. Ann N Y Acad Sci 2016; 1386:3-15. [PMID: 27880855 DOI: 10.1111/nyas.13283] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 09/24/2016] [Accepted: 09/29/2016] [Indexed: 02/06/2023]
Abstract
Resveratrol, a polyphenol found in grapes, peanuts, and red wine, plays different roles in diseases such as cancer and diabetes. Existing information indicates that resveratrol provides cardioprotection, as evidenced by superior postischemic ventricular recovery, reduced myocardial infarct size, and decreased number of apoptotic cardiomyocytes associated with resveratrol treatment in animal models. Cardiovascular benefits are experienced in humans with routine but not acute consumption of red wine. In this concise review, the paradoxical pro- and antiangiogenic effects of resveratrol are described, and different roles for resveratrol in the formation of new blood vessels are explained through different mechanisms. It is hypothesized that the effects of resveratrol on different cell types are not only dependent on its concentration but also on the physical and chemical conditions surrounding cells. The findings discussed herein shed light on potential therapeutic proapoptotic and antiangiogenic applications of low-dose resveratrol treatment in the prevention and treatment of different diseases.
Collapse
|
96
|
Puca AA, Spinetti G, Vono R, Vecchione C, Madeddu P. The genetics of exceptional longevity identifies new druggable targets for vascular protection and repair. Pharmacol Res 2016; 114:169-174. [PMID: 27818232 DOI: 10.1016/j.phrs.2016.10.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 09/15/2016] [Accepted: 10/31/2016] [Indexed: 12/23/2022]
Abstract
Therapeutic angiogenesis is a relatively new medical strategy in the field of cardiovascular diseases. The underpinning concept is that angiogenic growth factors or proangiogenic cells could be exploited therapeutically in cardiovascular patients to enhance native revascularization responses to an ischemic insult, thereby accelerating tissue healing. The initial enthusiasm generated by preclinical studies has been tempered by the modest success of clinical trials assessing therapeutic angiogenesis. Similarly, proangiogenic cell therapy has so far not maintained the original promises. Intriguingly, the current trend is to consider regeneration as a prerogative of the youngest organism. Consequentially, the embryonic and foetal models are attracting much attention for clinical translation into corrective modalities in the adulthood. Scientists seem to undervalue the lesson from Mother Nature, e.g. all humans are born young but very few achieve the goal of an exceptional healthy longevity. Either natural experimentation is driven by a supreme intelligence or stochastic phenomena, one has to accept the evidence that healthy longevity is the fruit of an evolutionary process lasting million years. It is therefore extremely likely that results of this natural experimentation are more reliable and translatable than the intensive, but very short human investigation on mechanisms governing repair and regeneration. With this preamble in mind, here we propose to shift the focus from the very beginning to the very end of human life and thus capture the secret of prolonged health span to improve well-being in the adulthood.
Collapse
Affiliation(s)
- Annibale A Puca
- IRCCS MultiMedica, Milan, Italy; University of Salerno, Salerno, Italy
| | | | | | - Carmine Vecchione
- University of Salerno, Salerno, Italy; IRCCS Neuromed, Pozzilli, Italy
| | | |
Collapse
|
97
|
Abstract
Nitric oxide has been implicated in numerous biological processes, particularly those involved with the cardiovascular system. Nitric oxide production is closely regulated and influenced by a number of factors in both health and disease. Nitric oxide is involved in maintaining the vascular system in its healthy, nondiseased state by producing vasorelaxation which enhances blood flow and prevents both leukocyte and platelet adhesion to the vascular wall. Dysfunctional endothelial cell nitric oxide production has been implicated in a number of disease states, including hypertension and atherosclerosis, and has been associated with adverse cardiac events. Various recent therapies may exert their beneficial effects in part by enhancing endothelial nitric oxide bloavallability. Nitric oxide has been used therapeutically in a number of cardiorespiratory disease states. An improved understanding of the pathologic processes underlying these diseases has resulted in several alternative agents being investigated and used clinically.
Collapse
Affiliation(s)
- Stuart M. Lowson
- Department of Anesthesiology and Surgical-Trauma ICU Co-Director, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
98
|
Rey-Funes M, Larrayoz IM, Fernández JC, Contartese DS, Rolón F, Inserra PIF, Martínez-Murillo R, López-Costa JJ, Dorfman VB, Martínez A, Loidl CF. Methylene blue prevents retinal damage in an experimental model of ischemic proliferative retinopathy. Am J Physiol Regul Integr Comp Physiol 2016; 310:R1011-9. [PMID: 26984891 DOI: 10.1152/ajpregu.00266.2015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 03/11/2016] [Indexed: 11/22/2022]
Abstract
Perinatal asphyxia induces retinal lesions, generating ischemic proliferative retinopathy, which may result in blindness. Previously, we showed that the nitrergic system was involved in the physiopathology of perinatal asphyxia. Here we analyze the application of methylene blue, a well-known soluble guanylate cyclase inhibitor, as a therapeutic strategy to prevent retinopathy. Male rats (n = 28 per group) were treated in different ways: 1) control group comprised born-to-term animals; 2) methylene blue group comprised animals born from pregnant rats treated with methylene blue (2 mg/kg) 30 and 5 min before delivery; 3) perinatal asphyxia (PA) group comprised rats exposed to perinatal asphyxia (20 min at 37°C); and 4) methylene blue-PA group comprised animals born from pregnant rats treated with methylene blue (2 mg/kg) 30 and 5 min before delivery, and then the pups were subjected to PA as above. For molecular studies, mRNA was obtained at different times after asphyxia, and tissue was collected at 30 days for morphological and biochemical analysis. Perinatal asphyxia produced significant gliosis, angiogenesis, and thickening of the inner retina. Methylene blue treatment reduced these parameters. Perinatal asphyxia resulted in a significant elevation of the nitrergic system as shown by NO synthase (NOS) activity assays, Western blotting, and (immuno)histochemistry for the neuronal isoform of NOS and NADPH-diaphorase activity. All these parameters were also normalized by the treatment. In addition, methylene blue induced the upregulation of the anti-angiogenic peptide, pigment epithelium-derived factor. Application of methylene blue reduced morphological and biochemical parameters of retinopathy. This finding suggests the use of methylene blue as a new treatment to prevent or decrease retinal damage in the context of ischemic proliferative retinopathy.
Collapse
Affiliation(s)
- Manuel Rey-Funes
- Laboratorio de Neuropatología Experimental, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis," Facultad de Medicina, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Ignacio M Larrayoz
- Angiogenesis Study Group, Center for Biomedical Research of La Rioja, Logroño, Spain;
| | - Juan C Fernández
- Primera Cátedra de Farmacología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Daniela S Contartese
- Laboratorio de Neuropatología Experimental, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis," Facultad de Medicina, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Federico Rolón
- Laboratorio de Neuropatología Experimental, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis," Facultad de Medicina, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Pablo I F Inserra
- Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y Diagnóstico, Universidad Maimónides, Buenos Aires, Argentina
| | - Ricardo Martínez-Murillo
- Neurovascular Research Group, Department of Molecular, Cellular and Developmental Neurobiology, Instituto Cajal, Consejo Superior Investigaciones Científicas, Madrid, Spain; and
| | - Juan J López-Costa
- Laboratorio de Neuropatología Experimental, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis," Facultad de Medicina, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Verónica B Dorfman
- Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y Diagnóstico, Universidad Maimónides, Buenos Aires, Argentina
| | - Alfredo Martínez
- Angiogenesis Study Group, Center for Biomedical Research of La Rioja, Logroño, Spain
| | - César F Loidl
- Laboratorio de Neuropatología Experimental, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis," Facultad de Medicina, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina; Laboratorio de Neurociencia, Facultad de Ciencias Médicas, Universidad Católica de Cuyo, San Juan, Argentina
| |
Collapse
|
99
|
Yu H, Peng J, Xu Y, Chang J, Li H. Bioglass Activated Skin Tissue Engineering Constructs for Wound Healing. ACS APPLIED MATERIALS & INTERFACES 2016; 8:703-715. [PMID: 26684719 DOI: 10.1021/acsami.5b09853] [Citation(s) in RCA: 149] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Wound healing is a complicated process, and fibroblast is a major cell type that participates in the process. Recent studies have shown that bioglass (BG) can stimulate fibroblasts to secrete a multitude of growth factors that are critical for wound healing. Therefore, we hypothesize that BG can stimulate fibroblasts to have a higher bioactivity by secreting more bioactive growth factors and proteins as compared to untreated fibroblasts, and we aim to construct a bioactive skin tissue engineering graft for wound healing by using BG activated fibroblast sheet. Thus, the effects of BG on fibroblast behaviors were studied, and the bioactive skin tissue engineering grafts containing BG activated fibroblasts were applied to repair the full skin lesions on nude mouse. Results showed that BG stimulated fibroblasts to express some critical growth factors and important proteins including vascular endothelial growth factor, basic fibroblast growth factor, epidermal growth factor, collagen I, and fibronectin. In vivo results revealed that fibroblasts in the bioactive skin tissue engineering grafts migrated into wound bed, and the migration ability of fibroblasts was stimulated by BG. In addition, the bioactive BG activated fibroblast skin tissue engineering grafts could largely increase the blood vessel formation, enhance the production of collagen I, and stimulate the differentiation of fibroblasts into myofibroblasts in the wound site, which would finally accelerate wound healing. This study demonstrates that the BG activated skin tissue engineering grafts contain more critical growth factors and extracellular matrix proteins that are beneficial for wound healing as compared to untreated fibroblast cell sheets.
Collapse
Affiliation(s)
- Hongfei Yu
- Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University , 1954 Huashan Road, Shanghai 200030, China
| | - Jinliang Peng
- School of Pharmacy, Shanghai Jiao Tong University , 800 Dongchuan Road, Shanghai 200240, China
| | - Yuhong Xu
- School of Pharmacy, Shanghai Jiao Tong University , 800 Dongchuan Road, Shanghai 200240, China
| | - Jiang Chang
- Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University , 1954 Huashan Road, Shanghai 200030, China
- Shanghai Institute of Ceramics, Chinese Academy of Sciences , 1295 Dingxi Road, Shanghai 200050, China
| | - Haiyan Li
- Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University , 1954 Huashan Road, Shanghai 200030, China
| |
Collapse
|
100
|
Krishnapati LS, Ghaskadbi S. Insulin induces the expression of FGF2 but does not synergize with it during angiogenesis. Microvasc Res 2016; 103:55-63. [DOI: 10.1016/j.mvr.2015.11.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 11/04/2015] [Accepted: 11/04/2015] [Indexed: 10/22/2022]
|