1
|
Fidanboylu M, Thomas SA. L-Arginine and asymmetric dimethylarginine (ADMA) transport across the mouse blood-brain and blood-CSF barriers: Evidence of saturable transport at both interfaces and CNS to blood efflux. PLoS One 2024; 19:e0305318. [PMID: 39446890 PMCID: PMC11501026 DOI: 10.1371/journal.pone.0305318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 10/09/2024] [Indexed: 10/26/2024] Open
Abstract
L-Arginine is the physiological substrate for the nitric oxide synthase (NOS) family, which synthesises nitric oxide (NO) in endothelial and neuronal cells. NO synthesis can be inhibited by endogenous asymmetric dimethylarginine (ADMA). NO has explicit roles in cellular signalling and vasodilation. Impaired NO bioavailability represents the central feature of endothelial dysfunction associated with vascular diseases. Interestingly, dietary supplementation with L-arginine has been shown to alleviate endothelial dysfunctions caused by impaired NO synthesis. In this study the transport kinetics of [3H]-arginine and [3H]-ADMA into the central nervous system (CNS) were investigated using physicochemical assessment and the in situ brain/choroid plexus perfusion technique in anesthetized mice. Results indicated that L-arginine and ADMA are tripolar cationic amino acids and have a gross charge at pH 7.4 of 0.981. L-Arginine (0.00149±0.00016) has a lower lipophilicity than ADMA (0.00226±0.00006) as measured using octanol-saline partition coefficients. The in situ perfusion studies revealed that [3H]-arginine and [3H]-ADMA can cross the blood-brain barrier (BBB) and the blood-CSF barrier. [3H]-Arginine (11.6nM) and [3H]-ADMA (62.5nM) having unidirectional transfer constants (Kin) into the frontal cortex of 5.84±0.86 and 2.49±0.35 μl.min-1.g-1, respectively, and into the CSF of 1.08±0.24 and 2.70±0.90 μl.min-1.g-1, respectively. In addition, multiple-time uptake studies revealed the presence of CNS-to-blood efflux of ADMA. Self- and cross-inhibition studies indicated the presence of transporters at the BBB and the blood-CSF barriers for both amino acids, which were shared to some degree. Importantly, these results are the first to demonstrate: (i) saturable transport of [3H]-ADMA at the blood-CSF barrier (choroid plexus) and (ii) a significant CNS to blood efflux of [3H]-ADMA. Our results suggest that the arginine paradox, in other words the clinical observation that NO-deficient patients respond well to oral supplementation with L-arginine even though the plasma concentration is sufficient to saturate endothelial NOS, could be related to altered ADMA transport (efflux).
Collapse
Affiliation(s)
- Mehmet Fidanboylu
- Pharmaceutical Sciences Research Division, King’s College London, London, United Kingdom
- Institute of Pharmaceutical Science, King’s College London, London, United Kingdom
| | - Sarah Ann Thomas
- Pharmaceutical Sciences Research Division, King’s College London, London, United Kingdom
- Institute of Pharmaceutical Science, King’s College London, London, United Kingdom
| |
Collapse
|
2
|
Paloschi V, Pauli J, Winski G, Wu Z, Li Z, Botti L, Meucci S, Conti P, Rogowitz F, Glukha N, Hummel N, Busch A, Chernogubova E, Jin H, Sachs N, Eckstein H, Dueck A, Boon RA, Bausch AR, Maegdefessel L. Utilization of an Artery-on-a-Chip to Unravel Novel Regulators and Therapeutic Targets in Vascular Diseases. Adv Healthc Mater 2024; 13:e2302907. [PMID: 37797407 PMCID: PMC11468405 DOI: 10.1002/adhm.202302907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/18/2023] [Indexed: 10/07/2023]
Abstract
In this study, organ-on-chip technology is used to develop an in vitro model of medium-to-large size arteries, the artery-on-a-chip (AoC), with the objective to recapitulate the structure of the arterial wall and the relevant hemodynamic forces affecting luminal cells. AoCs exposed either to in vivo-like shear stress values or kept in static conditions are assessed to generate a panel of novel genes modulated by shear stress. Considering the crucial role played by shear stress alterations in carotid arteries affected by atherosclerosis (CAD) and abdominal aortic aneurysms (AAA) disease development/progression, a patient cohort of hemodynamically relevant specimens is utilized, consisting of diseased and non-diseased (internal control) vessel regions from the same patient. Genes activated by shear stress follow the same expression pattern in non-diseased segments of human vessels. Single cell RNA sequencing (scRNA-seq) enables to discriminate the unique cell subpopulations between non-diseased and diseased vessel portions, revealing an enrichment of flow activated genes in structural cells originating from non-diseased specimens. Furthermore, the AoC served as a platform for drug-testing. It reproduced the effects of a therapeutic agent (lenvatinib) previously used in preclinical AAA studies, therefore extending the understanding of its therapeutic effect through a multicellular structure.
Collapse
Affiliation(s)
- Valentina Paloschi
- Department for Vascular and Endovascular SurgeryTechnical University of Munich80333MunichGermany
- German Center for Cardiovascular Research DZHKPartner Site Munich Heart Alliance80336BerlinGermany
| | - Jessica Pauli
- Department for Vascular and Endovascular SurgeryTechnical University of Munich80333MunichGermany
- German Center for Cardiovascular Research DZHKPartner Site Munich Heart Alliance80336BerlinGermany
| | - Greg Winski
- Department of Medicine, Cardiovascular UnitKarolinska Institute171 77StockholmSweden
| | - Zhiyuan Wu
- Department for Vascular and Endovascular SurgeryTechnical University of Munich80333MunichGermany
- Department of Vascular SurgeryBeijing HospitalNational Center of GerontologyInstitute of Geriatric MedicineChinese Academy of Medical ScienceBeijing10073P. R. China
| | - Zhaolong Li
- Department for Vascular and Endovascular SurgeryTechnical University of Munich80333MunichGermany
| | - Lorenzo Botti
- Department of Engineering and Applied SciencesUniversity of BergamoBergamo24129Italy
| | - Sandro Meucci
- Micronit MicrotechnologiesEnschede15 7521The Netherlands
| | - Pierangelo Conti
- Department of Engineering and Applied SciencesUniversity of BergamoBergamo24129Italy
| | | | - Nadiya Glukha
- Department for Vascular and Endovascular SurgeryTechnical University of Munich80333MunichGermany
| | - Nora Hummel
- Department for Vascular and Endovascular SurgeryTechnical University of Munich80333MunichGermany
| | - Albert Busch
- Department for Vascular and Endovascular SurgeryTechnical University of Munich80333MunichGermany
- Division of Vascular and Endovascular SurgeryDepartment for VisceralThoracic and Vascular SurgeryMedical Faculty Carl Gustav Carus and University HospitalTechnical University Dresden01069DresdenGermany
| | | | - Hong Jin
- Department of Medicine, Cardiovascular UnitKarolinska Institute171 77StockholmSweden
| | - Nadja Sachs
- Department for Vascular and Endovascular SurgeryTechnical University of Munich80333MunichGermany
| | - Hans‐Henning Eckstein
- Department for Vascular and Endovascular SurgeryTechnical University of Munich80333MunichGermany
| | - Anne Dueck
- German Center for Cardiovascular Research DZHKPartner Site Munich Heart Alliance80336BerlinGermany
- Institute of Pharmacology and ToxicologyTechnical University of Munich80333MunichGermany
| | - Reinier A. Boon
- Department of PhysiologyAmsterdam Cardiovascular Sciences (ACS)Amsterdam UMCVU University Medical CenterAmsterdam1081 HVThe Netherlands
- Institute of Cardiovascular RegenerationCenter of Molecular MedicineGoethe‐University60323FrankfurtGermany
- German Center for Cardiovascular Research DZHKPartner Site Frankfurt Rhine‐Main10785BerlinGermany
| | - Andreas R. Bausch
- Department of Cellular BiophysicsTechnical University of Munich80333MunichGermany
| | - Lars Maegdefessel
- Department for Vascular and Endovascular SurgeryTechnical University of Munich80333MunichGermany
- German Center for Cardiovascular Research DZHKPartner Site Munich Heart Alliance80336BerlinGermany
- Department of Medicine, Cardiovascular UnitKarolinska Institute171 77StockholmSweden
| |
Collapse
|
3
|
Dimina L, Tremblay-Franco M, Deveaux A, Tardivel C, Fouillet H, Polakof S, Martin JC, Mariotti F. Plasma Metabolome Analysis Suggests That L-Arginine Supplementation Affects Microbial Activity Resulting in a Decrease in Trimethylamine N-oxide-A Randomized Controlled Trial in Healthy Overweight Adults with Cardiometabolic Risk Factors. Curr Dev Nutr 2023; 7:102038. [PMID: 38162999 PMCID: PMC10754708 DOI: 10.1016/j.cdnut.2023.102038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 11/09/2023] [Indexed: 01/03/2024] Open
Abstract
Background The effects of supplementation with L-arginine (L-arg), the precursor of nitric oxide (NO), on vascular and cardiometabolic health have largely been explored. Whether other mechanisms of the action of L-arg exist remains unknown, as arginine metabolism is complicated. Objective We aimed to characterize the effect of low dose L-arg supplementation on overall human metabolism both in a fasting state and in response to an allostatic stress. Methods In a randomized, double-blind, crossover study, 32 healthy overweight adults (mean age 45 y) with cardiometabolic risk (fasting plasma triglycerides >150 mg/dL; waist circumference >94 cm [male] or >80 cm [female]) were treated with 1.5 g sustained-release L-arg 3 times/d (4.5 g/d) or placebo for 4 wk. On the last day of treatment, volunteers consumed a high-fat meal challenge (900 kcal, 80% as fat, 13% as carbohydrate, and 7% as protein). Plasma was collected at fasting, 2, 4, and 6 h after the challenge, and the metabolome was analyzed by high-resolution liquid chromatography-mass spectrometry. Metabolic profiles were analyzed using linear mixed models-principal component analysis. Results The challenge meal explained most of the changes in the metabolome. The overall effect of L-arg supplementation significantly explained 0.5% of the total variance, irrespective of the response to the challenge meal (P < 0.05). Among the metabolites that explain most of the L-arg effect, we found many amino acids, including branched-chain amino acids, that were decreased by L-arg supplementation. L-arg also decreased trimethylamine N-oxide (TMAO). Other changes suggest that L-arg increased methyl demand. Conclusions Analysis of the effect of 4 wk of L-arg supplementation on the metabolome reveals important effects on methyl balance and gut microbiota activity, such as a decrease in TMAO. Further studies are needed to investigate those mechanisms and the implications of these changes for long-term health.This trial was registered at clinicaltrials.gov as NCT02354794.
Collapse
Affiliation(s)
- Laurianne Dimina
- Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, Palaiseau, France
| | - Marie Tremblay-Franco
- Toxalim-Research Centre in Food Toxicology, Toulouse University, INRAE UMR 1331, ENVT, INP-Purpan, Paul Sabatier University, Toulouse, France
- Metatoul-AXIOM platform, National Infrastructure for Metabolomics and Fluxomics, MetaboHUB, Toxalim, INRAE UMR 1331, Toulouse, France
| | - Ambre Deveaux
- Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, Palaiseau, France
| | | | - Hélène Fouillet
- Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, Palaiseau, France
| | - Sergio Polakof
- Université Clermont Auvergne, INRAE, UMR 1019, Unité Nutrition Humaine, Clermont-Ferrand, France
| | | | - François Mariotti
- Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, Palaiseau, France
| |
Collapse
|
4
|
Luo W, Tang S, Xiao X, Luo S, Yang Z, Huang W, Tang S. Translation Animal Models of Diabetic Kidney Disease: Biochemical and Histological Phenotypes, Advantages and Limitations. Diabetes Metab Syndr Obes 2023; 16:1297-1321. [PMID: 37179788 PMCID: PMC10168199 DOI: 10.2147/dmso.s408170] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 04/29/2023] [Indexed: 05/15/2023] Open
Abstract
Animal models play a crucial role in studying the pathogenesis of diseases, developing new drugs, identifying disease risk markers, and improving means of prevention and treatment. However, modeling diabetic kidney disease (DKD) has posed a challenge for scientists. Although numerous models have been successfully developed, none of them can encompass all the key characteristics of human DKD. It is essential to choose the appropriate model according to the research needs, as different models develop different phenotypes and have their limitations. This paper provides a comprehensive overview of biochemical and histological phenotypes, modeling mechanisms, advantages and limitations of DKD animal models, in order to update relevant model information and provide insights and references for generating or selecting the appropriate animal models to fit different experimental needs.
Collapse
Affiliation(s)
- Wenting Luo
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan Province, People’s Republic of China
| | - Shiyun Tang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| | - Xiang Xiao
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan Province, People’s Republic of China
| | - Simin Luo
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan Province, People’s Republic of China
| | - Zixuan Yang
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan Province, People’s Republic of China
| | - Wei Huang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| | - Songqi Tang
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan Province, People’s Republic of China
| |
Collapse
|
5
|
Mason SA, Parker L, van der Pligt P, Wadley GD. Vitamin C supplementation for diabetes management: A comprehensive narrative review. Free Radic Biol Med 2023; 194:255-283. [PMID: 36526243 DOI: 10.1016/j.freeradbiomed.2022.12.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/01/2022] [Accepted: 12/05/2022] [Indexed: 12/15/2022]
Abstract
Growing evidence suggests that vitamin C supplementation may be an effective adjunct therapy in the management of people with diabetes. This paper critically reviews the current evidence on effects of vitamin C supplementation and its potential mechanisms in diabetes management. Evidence from meta-analyses of randomized controlled trials (RCTs) show favourable effects of vitamin C on glycaemic control and blood pressure that may be clinically meaningful, and mixed effects on blood lipids and endothelial function. However, evidence is mostly of low evidence certainty. Emerging evidence is promising for effects of vitamin C supplementation on some diabetes complications, particularly diabetic foot ulcers. However, there is a notable lack of robust and well-designed studies exploring effects of vitamin C as a single compound supplement on diabetes prevention and patient-important outcomes (i.e. prevention and amelioration of diabetes complications). RCTs are also required to investigate potential preventative or ameliorative effects of vitamin C on gestational diabetes outcomes. Oral vitamin C doses of 500-1000 mg per day are potentially effective, safe, and affordable for many individuals with diabetes. However, personalisation of supplementation regimens that consider factors such as vitamin C status, disease status, current glycaemic control, vitamin C intake, redox status, and genotype is important to optimize vitamin C's therapeutic effects safely. Finally, given a high prevalence of vitamin C deficiency in patients with complications, it is recommended that plasma vitamin C concentration be measured and monitored in the clinic setting.
Collapse
Affiliation(s)
- Shaun A Mason
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia.
| | - Lewan Parker
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia
| | - Paige van der Pligt
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia; Department of Nutrition and Dietetics, Western Health, Footscray, Australia
| | - Glenn D Wadley
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia
| |
Collapse
|
6
|
Młynarczyk M, Falkowska M, Micun Z, Obuchowska I, Kochanowicz J, Socha K, Konopińska J. Diet, Oxidative Stress, and Blood Serum Nutrients in Various Types of Glaucoma: A Systematic Review. Nutrients 2022; 14:nu14071421. [PMID: 35406033 PMCID: PMC9002851 DOI: 10.3390/nu14071421] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/23/2022] [Accepted: 03/25/2022] [Indexed: 12/11/2022] Open
Abstract
Glaucoma is one of the most common causes of irreversible vision loss worldwide. It is an insidious disease with a multifactorial pathogenesis. Despite progress in treatment methods, prevention and lifestyle modifications may be useful in slowing the progression of this disease. This systematic review aimed to evaluate the influence of diet, oxidative stress, and disturbances in blood serum levels of nutrients on the incidence and severity of glaucoma based on scientific reports on the role of nutrition in the pathogenesis and course of glaucoma. This paper presents an analysis of the above issues; however, further research is required to develop this topic. Future clinical trials are needed to assess the influence of nutrition and to develop nutritional management strategies for patients with glaucoma.
Collapse
Affiliation(s)
- Maryla Młynarczyk
- Department of Ophthalmology, Medical University of Białystok, M. Skłodowskiej-Curie 24a, 15-276 Białystok, Poland; (M.M.); (Z.M.); (I.O.)
| | - Martyna Falkowska
- Department of Bromatology, Faculty of Pharmacy with the Division of Laboratory Medicine, Medical University of Białystok, Mickiewicza 2D, 15-222 Białystok, Poland; (M.F.); (K.S.)
| | - Zuzanna Micun
- Department of Ophthalmology, Medical University of Białystok, M. Skłodowskiej-Curie 24a, 15-276 Białystok, Poland; (M.M.); (Z.M.); (I.O.)
| | - Iwona Obuchowska
- Department of Ophthalmology, Medical University of Białystok, M. Skłodowskiej-Curie 24a, 15-276 Białystok, Poland; (M.M.); (Z.M.); (I.O.)
| | - Jan Kochanowicz
- Department of Neurology, Medical University of Białystok, M. Skłodowskiej-Curie 24a, 15-276 Białystok, Poland;
| | - Katarzyna Socha
- Department of Bromatology, Faculty of Pharmacy with the Division of Laboratory Medicine, Medical University of Białystok, Mickiewicza 2D, 15-222 Białystok, Poland; (M.F.); (K.S.)
| | - Joanna Konopińska
- Department of Ophthalmology, Medical University of Białystok, M. Skłodowskiej-Curie 24a, 15-276 Białystok, Poland; (M.M.); (Z.M.); (I.O.)
- Correspondence: ; Tel.: +48-600471666
| |
Collapse
|
7
|
Lau M, Sealy B, Combes V, Morsch M, Garcia-Bennett AE. Enhanced Antioxidant Effects of the Anti-Inflammatory Compound Probucol when Released from Mesoporous Silica Particles. Pharmaceutics 2022; 14:pharmaceutics14030502. [PMID: 35335878 PMCID: PMC8953917 DOI: 10.3390/pharmaceutics14030502] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 02/17/2022] [Accepted: 02/21/2022] [Indexed: 12/10/2022] Open
Abstract
Brain endothelial cells mediate the function and integrity of the blood brain barrier (BBB) by restricting its permeability and exposure to potential toxins. However, these cells are highly susceptible to cellular damage caused by oxidative stress and inflammation. Consequent disruption to the integrity of the BBB can lead to the pathogenesis of neurodegenerative diseases. Drug compounds with antioxidant and/or anti-inflammatory properties therefore have the potential to preserve the structure and function of the BBB. In this work, we demonstrate the enhanced antioxidative effects of the compound probucol when loaded within mesoporous silica particles (MSP) in vitro and in vivo zebrafish models. The dissolution kinetics were significantly enhanced when released from MSPs. An increased reduction in lipopolysaccharide (LPS)-induced reactive oxygen species (ROS), cyclooxygenase (COX) enzyme activity and prostaglandin E2 production was measured in human brain endothelial cells treated with probucol-loaded MSPs. Furthermore, the LPS-induced permeability across an endothelial cell monolayer by paracellular and transcytotic mechanisms was also reduced at lower concentrations compared to the antioxidant ascorbic acid. Zebrafish pre-treated with probucol-loaded MSPs reduced hydrogen peroxide-induced ROS to control levels after 24-h incubation, at significantly lower concentrations than ascorbic acid. We provide compelling evidence that the encapsulation of antioxidant and anti-inflammatory compounds within MSPs can enhance their release, enhance their antioxidant effects properties, and open new avenues for the accelerated suppression of neuroinflammation.
Collapse
Affiliation(s)
- Michael Lau
- School of Natural Sciences, Macquarie University, Sydney, NSW 2109, Australia;
| | - Benjamin Sealy
- Malaria and Microvesicles Research Group, School of Life Science, Faculty of Science, University of Technology Sydney, Ultimo, Sydney, NSW 2007, Australia; (B.S.); (V.C.)
| | - Valery Combes
- Malaria and Microvesicles Research Group, School of Life Science, Faculty of Science, University of Technology Sydney, Ultimo, Sydney, NSW 2007, Australia; (B.S.); (V.C.)
| | - Marco Morsch
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia;
| | - Alfonso E. Garcia-Bennett
- School of Natural Sciences, Macquarie University, Sydney, NSW 2109, Australia;
- Australian Research Council Industrial Transformation Training Centre for Facilitated Advancement of Australia’s Bioactives (FAAB), Macquarie University, Sydney, NSW 2109, Australia
- Correspondence:
| |
Collapse
|
8
|
Lucas R, Hadizamani Y, Enkhbaatar P, Csanyi G, Caldwell RW, Hundsberger H, Sridhar S, Lever AA, Hudel M, Ash D, Ushio-Fukai M, Fukai T, Chakraborty T, Verin A, Eaton DC, Romero M, Hamacher J. Dichotomous Role of Tumor Necrosis Factor in Pulmonary Barrier Function and Alveolar Fluid Clearance. Front Physiol 2022; 12:793251. [PMID: 35264975 PMCID: PMC8899333 DOI: 10.3389/fphys.2021.793251] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 11/30/2021] [Indexed: 02/04/2023] Open
Abstract
Alveolar-capillary leak is a hallmark of the acute respiratory distress syndrome (ARDS), a potentially lethal complication of severe sepsis, trauma and pneumonia, including COVID-19. Apart from barrier dysfunction, ARDS is characterized by hyper-inflammation and impaired alveolar fluid clearance (AFC), which foster the development of pulmonary permeability edema and hamper gas exchange. Tumor Necrosis Factor (TNF) is an evolutionarily conserved pleiotropic cytokine, involved in host immune defense against pathogens and cancer. TNF exists in both membrane-bound and soluble form and its mainly -but not exclusively- pro-inflammatory and cytolytic actions are mediated by partially overlapping TNFR1 and TNFR2 binding sites situated at the interface between neighboring subunits in the homo-trimer. Whereas TNFR1 signaling can mediate hyper-inflammation and impaired barrier function and AFC in the lungs, ligand stimulation of TNFR2 can protect from ventilation-induced lung injury. Spatially distinct from the TNFR binding sites, TNF harbors within its structure a lectin-like domain that rather protects lung function in ARDS. The lectin-like domain of TNF -mimicked by the 17 residue TIP peptide- represents a physiological mediator of alveolar-capillary barrier protection. and increases AFC in both hydrostatic and permeability pulmonary edema animal models. The TIP peptide directly activates the epithelial sodium channel (ENaC) -a key mediator of fluid and blood pressure control- upon binding to its α subunit, which is also a part of the non-selective cation channel (NSC). Activity of the lectin-like domain of TNF is preserved in complexes between TNF and its soluble TNFRs and can be physiologically relevant in pneumonia. Antibody- and soluble TNFR-based therapeutic strategies show considerable success in diseases such as rheumatoid arthritis, psoriasis and inflammatory bowel disease, but their chronic use can increase susceptibility to infection. Since the lectin-like domain of TNF does not interfere with TNF's anti-bacterial actions, while exerting protective actions in the alveolar-capillary compartments, it is currently evaluated in clinical trials in ARDS and COVID-19. A more comprehensive knowledge of the precise role of the TNFR binding sites versus the lectin-like domain of TNF in lung injury, tissue hypoxia, repair and remodeling may foster the development of novel therapeutics for ARDS.
Collapse
Affiliation(s)
- Rudolf Lucas
- Vascular Biology Center, Augusta University, Augusta, GA, United States,Department of Pharmacology and Toxicology, Augusta University, Augusta, GA, United States,Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States,*Correspondence: Rudolf Lucas,
| | - Yalda Hadizamani
- Lungen-und Atmungsstiftung Bern, Bern, Switzerland,Pneumology, Clinic for General Internal Medicine, Lindenhofspital Bern, Bern, Switzerland
| | - Perenlei Enkhbaatar
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, United States
| | - Gabor Csanyi
- Vascular Biology Center, Augusta University, Augusta, GA, United States,Department of Pharmacology and Toxicology, Augusta University, Augusta, GA, United States
| | - Robert W. Caldwell
- Department of Pharmacology and Toxicology, Augusta University, Augusta, GA, United States
| | - Harald Hundsberger
- Department of Medical Biotechnology, University of Applied Sciences, Krems, Austria,Department of Dermatology, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Supriya Sridhar
- Vascular Biology Center, Augusta University, Augusta, GA, United States
| | - Alice Ann Lever
- Vascular Biology Center, Augusta University, Augusta, GA, United States,Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Martina Hudel
- Institute for Medical Microbiology, Justus-Liebig University, Giessen, Germany
| | - Dipankar Ash
- Vascular Biology Center, Augusta University, Augusta, GA, United States
| | - Masuko Ushio-Fukai
- Vascular Biology Center, Augusta University, Augusta, GA, United States,Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Tohru Fukai
- Vascular Biology Center, Augusta University, Augusta, GA, United States,Department of Pharmacology and Toxicology, Augusta University, Augusta, GA, United States,Charlie Norwood Veterans Affairs Medical Center, Augusta, GA, United States
| | - Trinad Chakraborty
- Institute for Medical Microbiology, Justus-Liebig University, Giessen, Germany
| | - Alexander Verin
- Vascular Biology Center, Augusta University, Augusta, GA, United States,Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Douglas C. Eaton
- Department of Medicine, School of Medicine, Emory University, Atlanta, GA, United States
| | - Maritza Romero
- Vascular Biology Center, Augusta University, Augusta, GA, United States,Department of Pharmacology and Toxicology, Augusta University, Augusta, GA, United States,Department of Anesthesiology and Perioperative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Jürg Hamacher
- Lungen-und Atmungsstiftung Bern, Bern, Switzerland,Pneumology, Clinic for General Internal Medicine, Lindenhofspital Bern, Bern, Switzerland,Medical Clinic V-Pneumology, Allergology, Intensive Care Medicine, and Environmental Medicine, Faculty of Medicine, University Medical Centre of the Saarland, Saarland University, Homburg, Germany,Institute for Clinical & Experimental Surgery, Faculty of Medicine, Saarland University, Homburg, Germany,Jürg Hamacher,
| |
Collapse
|
9
|
Williams IM, Wasserman DH. Capillary Endothelial Insulin Transport: The Rate-limiting Step for Insulin-stimulated Glucose Uptake. Endocrinology 2022; 163:6462374. [PMID: 34908124 PMCID: PMC8758342 DOI: 10.1210/endocr/bqab252] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Indexed: 11/19/2022]
Abstract
The rate-limiting step for skeletal muscle glucose uptake is transport from microcirculation to muscle interstitium. Capillary endothelium poses a barrier that delays the onset of muscle insulin action. Defining physiological barriers that control insulin access to interstitial space is difficult because of technical challenges that confront study of microscopic events in an integrated physiological system. Two physiological variables determine muscle insulin access. These are the number of perfused capillaries and the permeability of capillary walls to insulin. Disease states associated with capillary rarefaction are closely linked to insulin resistance. Insulin permeability through highly resistant capillary walls of muscle poses a significant barrier to insulin access. Insulin may traverse the endothelium through narrow intercellular junctions or vesicular trafficking across the endothelial cell. Insulin is large compared with intercellular junctions, making this an unlikely route. Transport by endothelial vesicular trafficking is likely the primary route of transit. Studies in vivo show movement of insulin is not insulin receptor dependent. This aligns with single-cell transcriptomics that show the insulin receptor is not expressed in muscle capillaries. Work in cultured endothelial cell lines suggest that insulin receptor activation is necessary for endothelial insulin transit. Controversies remain in the understanding of transendothelial insulin transit to muscle. These controversies closely align with experimental approaches. Control of circulating insulin accessibility to skeletal muscle is an area that remains ripe for discovery. Factors that impede insulin access to muscle may contribute to disease and factors that accelerate access may be of therapeutic value for insulin resistance.
Collapse
Affiliation(s)
- Ian M Williams
- Department of Molecular Physiology and Biophysics and Vanderbilt Mouse Metabolic Phenotyping Center, Vanderbilt University, Nashville, TN 37232-0615, USA
| | - David H Wasserman
- Department of Molecular Physiology and Biophysics and Vanderbilt Mouse Metabolic Phenotyping Center, Vanderbilt University, Nashville, TN 37232-0615, USA
- Correspondence: David H. Wasserman, PhD, Light Hall Rm. 702, Vanderbilt University, Nashville, TN 37232-0615, USA.
| |
Collapse
|
10
|
Kwiatkowski G, Bar A, Jasztal A, Chłopicki S. MRI-based in vivo detection of coronary microvascular dysfunction before alterations in cardiac function induced by short-term high-fat diet in mice. Sci Rep 2021; 11:18915. [PMID: 34556779 PMCID: PMC8460671 DOI: 10.1038/s41598-021-98401-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 08/26/2021] [Indexed: 02/07/2023] Open
Abstract
Endothelial dysfunction is one of the hallmarks of vascular abnormalities in metabolic diseases and has been repeatedly demonstrated in coronary and peripheral circulation in mice fed high-fat diet (HFD), particularly after long-term HFD. However, the temporal relationship between development of coronary microvascular endothelial dysfunction and deterioration in diastolic and systolic cardiac function after short-term feeding with HFD has not yet been studied. This study aimed to correlate the changes in coronary microvascular endothelial function and global cardiac performance indices in vivo after short-term feeding with HFD in mice. Short-term feeding with a HFD (60% fat + 1% cholesterol) resulted in severely impaired coronary microvascular function, as evidenced by the diminished effect of nitric oxide synthase inhibition (by L-NAME) assessed using T1 mapping via in vivo MRI. Deterioration of coronary microvascular function was detected as early as after 7 days of HFD and further declined after 8 weeks on a HFD. HFD-induced coronary microvascular dysfunction was not associated with impaired myocardial capillary density and was present before systemic insulin resistance assessed by a glucose tolerance test. Basal coronary flow and coronary reserve, as assessed using the A2A adenosine receptor agonist regadenoson, were also not altered in HFD-fed mice. Histological analysis did not reveal cardiomyocyte hypertrophy or fibrosis. Increased lipid accumulation in cardiomyocytes was detected as early as after 7 days of HFD and remained at a similar level at 8 weeks on a HFD. Multiparametric cardiac MRI revealed a reduction in systolic heart function, including decreased ejection rate, increased end-systolic volume and decreased myocardial strain in diastole with impaired ejection fraction, but not until 4 weeks of HFD. Short-term feeding with HFD resulted in early endothelial dysfunction in coronary microcirculation that preceded alteration in cardiac function and systemic insulin resistance.
Collapse
Affiliation(s)
- Grzegorz Kwiatkowski
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, ul. Bobrzynskiego 14, 30-348, Kraków, Poland
| | - Anna Bar
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, ul. Bobrzynskiego 14, 30-348, Kraków, Poland
| | - Agnieszka Jasztal
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, ul. Bobrzynskiego 14, 30-348, Kraków, Poland
| | - Stefan Chłopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, ul. Bobrzynskiego 14, 30-348, Kraków, Poland.
- Chair of Pharmacology, Faculty of Medicine, Jagiellonian University Medical College, Grzegorzecka 16, 31-531, Kraków, Poland.
| |
Collapse
|
11
|
Adams JA, Lopez JR, Uryash A, Sackner MA. Whole body periodic acceleration (pGz) improves endotoxin induced cardiomyocyte contractile dysfunction and attenuates the inflammatory response in mice. Heliyon 2021; 7:e06444. [PMID: 33748496 PMCID: PMC7970274 DOI: 10.1016/j.heliyon.2021.e06444] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 11/10/2020] [Accepted: 03/04/2021] [Indexed: 11/01/2022] Open
Abstract
Sepsis-induces myocardial contractile dysfunction. We previously showed that whole body periodic acceleration (pGz), the sinusoidal motion of the supine body head-foot ward direction significantly improves survival and decreases microvascular permeability in a lethal model of sepsis. We tested the hypothesis that pGz improves LPS induced cardiomyocyte contractile dysfunction and decreases LPS pro-inflammatory cytokine response when applied pre- or post-treatment. Isolated cardiomyocytes were obtained from mice that received LPS who had been pre-treated with pGz for three days (pGz-LPS) or control. Peak shortening (PS), maximal velocity of shortening (+dL/dt), and relengthening (-dL/dt) as well as diastolic intracellular calcium concentration ([Ca+2]d), sodium ([Na+]d), reactive oxygen species (ROS), and cardiac troponin (cTnT) production were measured. LPS decreased PS, +dL/dt, and -dL/dt, by 37%, 41% and 35% change respectively (p < 0.01), increased [Ca+2]d, [Na+]d, ROS, and cTnT by 343%, 122%, 298%, and 610% change respectively (p < 0.01) compared to control. pGz pre-treatment attenuated the parameters mentioned above. In a separate cohort, the effects of a lethal dose of LPS on protein expression of nitric oxide synthases (iNOS, eNOS, nNOS), pro- and anti-inflammatory cytokines in hearts of mice was studied in pre-treated with pGz for three days prior to LPS (pGz-LPS) and post-treated with pGz 30 min after LPS (LPS-pGz) were determined. LPS increased expression of early and late iNOS and decreased expression of eNOS, phosphorylated eNOS (p-eNOS), and nNOS. Both pre- and post-treatment with pGz markedly reduced early and late pro-inflammatory surge. Therefore, pre- and post-treatment with pGz improves LPS-induced cardiomyocyte dysfunction, decreases iNOS expression, and increases cytoprotective eNOS and nNOS, with decreased pro-inflammatory response. Such results have potential for translation to benefit outcomes in human sepsis.
Collapse
Affiliation(s)
- Jose A Adams
- Division of Neonatology, Mount Sinai Medical Center, Miami Beach, FL, USA
| | - Jose R Lopez
- Department of Research, Mount Sinai Medical Center, Miami Beach, FL, USA
| | - Arkady Uryash
- Division of Neonatology, Mount Sinai Medical Center, Miami Beach, FL, USA
| | - Marvin A Sackner
- Department of Medicine, Mount Sinai Medical Center, Miami Beach, FL, USA
| |
Collapse
|
12
|
Tao QR, Chu YM, Wei L, Tu C, Han YY. Antiangiogenic therapy in diabetic nephropathy: A double‑edged sword (Review). Mol Med Rep 2021; 23:260. [PMID: 33655322 PMCID: PMC7893700 DOI: 10.3892/mmr.2021.11899] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 01/21/2021] [Indexed: 02/06/2023] Open
Abstract
Diabetes and the associated complications are becoming a serious global threat and an increasing burden to human health and the healthcare systems. Diabetic nephropathy (DN) is the primary cause of end-stage kidney disease. Abnormal angiogenesis is well established to be implicated in the morphology and pathophysiology of DN. Factors that promote or inhibit angiogenesis serve an important role in DN. In the present review, the current issues associated with the vascular disease in DN are highlighted, and the challenges in the development of treatments are discussed.
Collapse
Affiliation(s)
- Qian-Ru Tao
- Department of Nephrology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213000, P.R. China
| | - Ying-Ming Chu
- Department of Integrated Traditional Chinese Medicine, Peking University First Hospital, Beijing 100034, P.R. China
| | - Lan Wei
- Department of Internal Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213000, P.R. China
| | - Chao Tu
- Department of Internal Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213000, P.R. China
| | - Yuan-Yuan Han
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, Yunnan 650118, P.R. China
| |
Collapse
|
13
|
Reina-Torres E, De Ieso ML, Pasquale LR, Madekurozwa M, van Batenburg-Sherwood J, Overby DR, Stamer WD. The vital role for nitric oxide in intraocular pressure homeostasis. Prog Retin Eye Res 2020; 83:100922. [PMID: 33253900 DOI: 10.1016/j.preteyeres.2020.100922] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 11/13/2020] [Accepted: 11/23/2020] [Indexed: 02/07/2023]
Abstract
Catalyzed by endothelial nitric oxide (NO) synthase (eNOS) activity, NO is a gaseous signaling molecule maintaining endothelial and cardiovascular homeostasis. Principally, NO regulates the contractility of vascular smooth muscle cells and permeability of endothelial cells in response to either biochemical or biomechanical cues. In the conventional outflow pathway of the eye, the smooth muscle-like trabecular meshwork (TM) cells and Schlemm's canal (SC) endothelium control aqueous humor outflow resistance, and therefore intraocular pressure (IOP). The mechanisms by which outflow resistance is regulated are complicated, but NO appears to be a key player as enhancement or inhibition of NO signaling dramatically affects outflow function; and polymorphisms in NOS3, the gene that encodes eNOS modifies the relation between various environmental exposures and glaucoma. Based upon a comprehensive review of past foundational studies, we present a model whereby NO controls a feedback signaling loop in the conventional outflow pathway that is sensitive to changes in IOP and its oscillations. Thus, upon IOP elevation, the outflow pathway tissues distend, and the SC lumen narrows resulting in increased SC endothelial shear stress and stretch. In response, SC cells upregulate the production of NO, relaxing neighboring TM cells and increasing permeability of SC's inner wall. These IOP-dependent changes in the outflow pathway tissues reduce the resistance to aqueous humor drainage and lower IOP, which, in turn, diminishes the biomechanical signaling on SC. Similar to cardiovascular pathogenesis, dysregulation of the eNOS/NO system leads to dysfunctional outflow regulation and ocular hypertension, eventually resulting in primary open-angle glaucoma.
Collapse
Affiliation(s)
| | | | - Louis R Pasquale
- Eye and Vision Research Institute of New York Eye and Ear Infirmary at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | - Darryl R Overby
- Department of Bioengineering, Imperial College London, London, UK.
| | - W Daniel Stamer
- Department of Ophthalmology, Duke University, Durham, NC, USA.
| |
Collapse
|
14
|
Tawada M, Ito Y, Banshodani M, Yamashita M, Shintaku S, Sun T, Suzuki Y, Kinashi H, Kubo Y, Ando M, Yamaguchi M, Katsuno T, Mizuno M, Kawanishi H. Vasculopathy plays an important role during the development and relapse of encapsulating peritoneal sclerosis with conventional peritoneal dialysis solutions. Nephrol Dial Transplant 2020; 36:1519-1526. [DOI: 10.1093/ndt/gfaa073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Indexed: 12/21/2022] Open
Abstract
Abstract
Background
Encapsulating peritoneal sclerosis (EPS) is an uncommon but life-threatening complication of peritoneal dialysis (PD) therapy. The causative factors of EPS remain unclear. Pathological studies of the peritoneum affected by EPS and relationships with clinical factors including PD solutions remain lacking. The objective of this study was to examine peritoneal samples from EPS patients and to identify the associations of peritoneal pathology with different clinical factors.
Methods
Peritoneal specimens were obtained at the time of surgical enterolysis in Tsuchiya General Hospital from 1993 to 2016. A total of 223 PD patients were enrolled and analyzed. Tissues were fixed with formalin and processed with hematoxylin and eosin and Masson’s trichrome staining, as well as immunohistochemical staining for CD31 and CD68.
Results
Evaluations could be made in 174 patients who received surgical enterolysis. Conventional or pH-neutral low-glucose degradation product PD solutions were utilized during PD treatment. The conventional PD solution group showed less angiogenesis (P = 0.013) but more severe vasculopathy, in the form of a lower ratio of luminal diameter to vessel diameter (L/V ratio) (P < 0.001) in association with longer PD treatment. Multivariate Cox proportional hazard models revealed that L/V ratio (per 0.1 increase, hazard ratio = 0.88, 95% confidence interval 0.77–0.99, P = 0.047) was significantly associated with a lower incidence of EPS relapse. In contrast, most of the cases in the pH-neutral solution group showed milder vasculopathy.
Conclusions
The pathology of EPS differed between conventional and pH-neutral solution groups. Vasculopathy was related to the development and relapse of EPS in the conventional solution group.
Collapse
Affiliation(s)
- Mitsuhiro Tawada
- Department of Nephrology and Rheumatology, Aichi Medical University, Nagakute, Japan
- Department of Nephrology and Renal Replacement Therapy, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuhiko Ito
- Department of Nephrology and Rheumatology, Aichi Medical University, Nagakute, Japan
| | - Masataka Banshodani
- Department of Surgery and Artificial Organs, Akane-Foundation, Tsuchiya General Hospital, Hiroshima, Japan
| | - Masahiro Yamashita
- Department of Surgery and Artificial Organs, Akane-Foundation, Tsuchiya General Hospital, Hiroshima, Japan
| | - Sadanori Shintaku
- Department of Surgery and Artificial Organs, Akane-Foundation, Tsuchiya General Hospital, Hiroshima, Japan
| | - Ting Sun
- Department of Nephrology and Renal Replacement Therapy, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuhiro Suzuki
- Department of Nephrology and Renal Replacement Therapy, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroshi Kinashi
- Department of Nephrology and Rheumatology, Aichi Medical University, Nagakute, Japan
| | - Yoko Kubo
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masahiko Ando
- Data Coordinating Center, Department of Advanced Medicine, Nagoya University Hospital, Nagoya, Japan
| | - Makoto Yamaguchi
- Department of Nephrology and Rheumatology, Aichi Medical University, Nagakute, Japan
| | - Takayuki Katsuno
- Department of Nephrology and Rheumatology, Aichi Medical University, Nagakute, Japan
| | - Masashi Mizuno
- Department of Nephrology and Renal Replacement Therapy, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hideki Kawanishi
- Department of Surgery and Artificial Organs, Akane-Foundation, Tsuchiya General Hospital, Hiroshima, Japan
| |
Collapse
|
15
|
Tiruppathi C, Regmi SC, Wang DM, Mo GCH, Toth PT, Vogel SM, Stan RV, Henkemeyer M, Minshall RD, Rehman J, Malik AB. EphB1 interaction with caveolin-1 in endothelial cells modulates caveolae biogenesis. Mol Biol Cell 2020; 31:1167-1182. [PMID: 32238105 PMCID: PMC7353165 DOI: 10.1091/mbc.e19-12-0713] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/24/2020] [Accepted: 03/25/2020] [Indexed: 12/16/2022] Open
Abstract
Caveolae, the cave-like structures abundant in endothelial cells (ECs), are important for multiple signaling processes such as production of nitric oxide and caveolae-mediated intracellular trafficking. Using superresolution microscopy, fluorescence resonance energy transfer, and biochemical analysis, we observed that the EphB1 receptor tyrosine kinase constitutively interacts with caveolin-1 (Cav-1), the key structural protein of caveolae. Activation of EphB1 with its ligand Ephrin B1 induced EphB1 phosphorylation and the uncoupling EphB1 from Cav-1 and thereby promoted phosphorylation of Cav-1 by Src. Deletion of Cav-1 scaffold domain binding (CSD) motif in EphB1 prevented EphB1 binding to Cav-1 as well as Src-dependent Cav-1 phosphorylation, indicating the importance of CSD in the interaction. We also observed that Cav-1 protein expression and caveolae numbers were markedly reduced in ECs from EphB1-deficient (EphB1-/-) mice. The loss of EphB1 binding to Cav-1 promoted Cav-1 ubiquitination and degradation, and hence the loss of Cav-1 was responsible for reducing the caveolae numbers. These studies identify the crucial role of EphB1/Cav-1 interaction in the biogenesis of caveolae and in coordinating the signaling function of Cav-1 in ECs.
Collapse
Affiliation(s)
- Chinnaswamy Tiruppathi
- Departments of Pharmacology, The University of Illinois College of Medicine, Chicago, IL 60612
- The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612
| | - Sushil C. Regmi
- Departments of Pharmacology, The University of Illinois College of Medicine, Chicago, IL 60612
| | - Dong-Mei Wang
- Departments of Pharmacology, The University of Illinois College of Medicine, Chicago, IL 60612
| | - Gary C. H. Mo
- Departments of Pharmacology, The University of Illinois College of Medicine, Chicago, IL 60612
| | - Peter T. Toth
- Departments of Pharmacology, The University of Illinois College of Medicine, Chicago, IL 60612
| | - Stephen M. Vogel
- Departments of Pharmacology, The University of Illinois College of Medicine, Chicago, IL 60612
| | - Radu V. Stan
- Department of Pathology, Geisel School of Medicine, Dartmouth College, Hanover, NH 03755
| | - Mark Henkemeyer
- Departments of Neuroscience and Developmental Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Richard D. Minshall
- Departments of Pharmacology, The University of Illinois College of Medicine, Chicago, IL 60612
- Anesthesiology, The University of Illinois College of Medicine, Chicago, IL 60612
- The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612
| | - Jalees Rehman
- Departments of Pharmacology, The University of Illinois College of Medicine, Chicago, IL 60612
| | - Asrar B. Malik
- Departments of Pharmacology, The University of Illinois College of Medicine, Chicago, IL 60612
- The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612
| |
Collapse
|
16
|
Papaefthymiou A, Doulberis M, Katsinelos P, Liatsos C, Polyzos SA, Kotronis G, Papanikolaou K, Kountouras J. Impact of nitric oxide's bidirectional role on glaucoma: focus onHelicobacter pylori–related nitrosative stress. Ann N Y Acad Sci 2020; 1465:10-28. [DOI: 10.1111/nyas.14253] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 09/07/2019] [Accepted: 09/17/2019] [Indexed: 12/12/2022]
Affiliation(s)
| | - Michael Doulberis
- Department of Gastroenterology and HepatologyUniversity of Zurich Zurich Switzerland
- Department of Internal Medicine, Second Medical Clinic, Ippokration HospitalAristotle University of Thessaloniki Thessaloniki Macedonia Greece
| | - Panagiotis Katsinelos
- Department of Internal Medicine, Second Medical Clinic, Ippokration HospitalAristotle University of Thessaloniki Thessaloniki Macedonia Greece
| | - Christos Liatsos
- Department of Gastroenterology401 General Military Hospital of Athens Athens Greece
| | - Stergios A. Polyzos
- Department of Internal Medicine, Second Medical Clinic, Ippokration HospitalAristotle University of Thessaloniki Thessaloniki Macedonia Greece
- First Department of Pharmacology, School of MedicineAristotle University of Thessaloniki Thessaloniki Macedonia Greece
| | - Georgios Kotronis
- Department of Internal MedicineAgios Pavlos General Hospital Thessaloniki Macedonia Greece
| | - Katerina Papanikolaou
- Department of Internal Medicine, Second Medical Clinic, Ippokration HospitalAristotle University of Thessaloniki Thessaloniki Macedonia Greece
| | - Jannis Kountouras
- Department of Internal Medicine, Second Medical Clinic, Ippokration HospitalAristotle University of Thessaloniki Thessaloniki Macedonia Greece
| |
Collapse
|
17
|
Dunne M, Regenold M, Allen C. Hyperthermia can alter tumor physiology and improve chemo- and radio-therapy efficacy. Adv Drug Deliv Rev 2020; 163-164:98-124. [PMID: 32681862 DOI: 10.1016/j.addr.2020.07.007] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/07/2020] [Accepted: 07/10/2020] [Indexed: 12/20/2022]
Abstract
Hyperthermia has demonstrated clinical success in improving the efficacy of both chemo- and radio-therapy in solid tumors. Pre-clinical and clinical research studies have demonstrated that targeted hyperthermia can increase tumor blood flow and increase the perfused fraction of the tumor in a temperature and time dependent manner. Changes in tumor blood circulation can produce significant physiological changes including enhanced vascular permeability, increased oxygenation, decreased interstitial fluid pressure, and reestablishment of normal physiological pH conditions. These alterations in tumor physiology can positively impact both small molecule and nanomedicine chemotherapy accumulation and distribution within the tumor, as well as the fraction of the tumor susceptible to radiation therapy. Hyperthermia can trigger drug release from thermosensitive formulations and further improve the accumulation, distribution, and efficacy of chemotherapy.
Collapse
|
18
|
He F, Zhang D, Chen Q, Zhao Y, Wu L, Li Z, Zhang C, Jiang Z, Wang Y. Angiopoietin‐Tie signaling in kidney diseases: an updated review. FEBS Lett 2019; 593:2706-2715. [PMID: 31380564 DOI: 10.1002/1873-3468.13568] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 07/30/2019] [Accepted: 08/01/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Fang‐Fang He
- Department of Nephrology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Di Zhang
- Department of Nephrology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Qing Chen
- Department of Hepatobiliary Surgery Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Yi Zhao
- Department of Nephrology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Liang Wu
- Department of Nephrology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Zhen‐Qiong Li
- Department of Nephrology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Chun Zhang
- Department of Nephrology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Zhao‐Hua Jiang
- Department of Plastic and Reconstructive Surgery Shanghai Ninth People's Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Yu‐Mei Wang
- Department of Nephrology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| |
Collapse
|
19
|
Guequén A, Zamorano P, Córdova F, Koning T, Torres A, Ehrenfeld P, Boric MP, Salazar-Onfray F, Gavard J, Durán WN, Quezada C, Sarmiento J, Sánchez FA. Interleukin-8 Secreted by Glioblastoma Cells Induces Microvascular Hyperpermeability Through NO Signaling Involving S-Nitrosylation of VE-Cadherin and p120 in Endothelial Cells. Front Physiol 2019; 10:988. [PMID: 31440166 PMCID: PMC6694439 DOI: 10.3389/fphys.2019.00988] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 07/18/2019] [Indexed: 01/08/2023] Open
Abstract
Glioblastoma is a highly aggressive brain tumor, characterized by the formation of dysfunctional blood vessels and a permeable endothelial barrier. S-nitrosylation, a post-translational modification, has been identified as a regulator of endothelial function. In this work we explored whether S-nitrosylation induced by glioblastoma tumors regulates the endothelial function. As proof of concept, we observed that S-nitrosylation is present in the tumoral microenvironment of glioblastoma in two different animal models. Subsequently, we measured S nitrosylation and microvascular permeability in EAhy296 endothelial cells and in cremaster muscle. In vitro, conditioned medium from the human glioblastoma cell line U87 activates endothelial nitric oxide synthase, causes VE-cadherin- S-nitrosylation and induces hyperpermeability. Blocking Interleukin-8 (IL-8) in the conditioned medium inhibited S-nitrosylation of VE-cadherin and hyperpermeability. Recombinant IL-8 increased endothelial permeability by activating eNOS, S-nitrosylation of VE-cadherin and p120, internalization of VE-cadherin and disassembly of adherens junctions. In vivo, IL-8 induced S-nitrosylation of VE-cadherin and p120 and conditioned medium from U87 cells caused hyperpermeability in the mouse cremaster muscle. We conclude that eNOS signaling induced by glioma cells-secreted IL-8 regulates endothelial barrier function in the context of glioblastoma involving S-nitrosylation of VE-cadherin and p120. Our results suggest that inhibiting S-nitrosylation may be an effective way to control and/or block damage to the endothelial barrier and prevent cancer progression.
Collapse
Affiliation(s)
- Anita Guequén
- Instituto de Inmunología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - Patricia Zamorano
- Instituto de Inmunología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - Francisco Córdova
- Instituto de Inmunología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - Tania Koning
- Instituto de Inmunología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - Angelo Torres
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| | - Pamela Ehrenfeld
- Instituto de Histología, Anatomía y Patología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
- Centro Interdisciplinario de Estudios del Sistema Nervioso (CISNe), Universidad Austral de Chile, Valdivia, Chile
| | - Mauricio P. Boric
- Departamento de Fisiología, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Flavio Salazar-Onfray
- Instituto Milenio de Inmunología e Inmunoterapia, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Julie Gavard
- Team SOAP, Signaling in Oncogenesis, Angiogenesis and Permeability, INSERM, CNRS, Institut de Cancérologie de l’Ouest, Université de Nantes, Nantes, France
| | - Walter N. Durán
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, The State University of New Jersey, Newark, NJ, United States
| | - Claudia Quezada
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| | - José Sarmiento
- Instituto de Fisiología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - Fabiola A. Sánchez
- Instituto de Inmunología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
- Centro Interdisciplinario de Estudios del Sistema Nervioso (CISNe), Universidad Austral de Chile, Valdivia, Chile
| |
Collapse
|
20
|
Mierke CT. The matrix environmental and cell mechanical properties regulate cell migration and contribute to the invasive phenotype of cancer cells. REPORTS ON PROGRESS IN PHYSICS. PHYSICAL SOCIETY (GREAT BRITAIN) 2019; 82:064602. [PMID: 30947151 DOI: 10.1088/1361-6633/ab1628] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
The minimal structural unit of a solid tumor is a single cell or a cellular compartment such as the nucleus. A closer look inside the cells reveals that there are functional compartments or even structural domains determining the overall properties of a cell such as the mechanical phenotype. The mechanical interaction of these living cells leads to the complex organization such as compartments, tissues and organs of organisms including mammals. In contrast to passive non-living materials, living cells actively respond to the mechanical perturbations occurring in their microenvironment during diseases such as fibrosis and cancer. The transformation of single cancer cells in highly aggressive and hence malignant cancer cells during malignant cancer progression encompasses the basement membrane crossing, the invasion of connective tissue, the stroma microenvironments and transbarrier migration, which all require the immediate interaction of the aggressive and invasive cancer cells with the surrounding extracellular matrix environment including normal embedded neighboring cells. All these steps of the metastatic pathway seem to involve mechanical interactions between cancer cells and their microenvironment. The pathology of cancer due to a broad heterogeneity of cancer types is still not fully understood. Hence it is necessary to reveal the signaling pathways such as mechanotransduction pathways that seem to be commonly involved in the development and establishment of the metastatic and mechanical phenotype in several carcinoma cells. We still do not know whether there exist distinct metastatic genes regulating the progression of tumors. These metastatic genes may then be activated either during the progression of cancer by themselves on their migration path or in earlier stages of oncogenesis through activated oncogenes or inactivated tumor suppressor genes, both of which promote the metastatic phenotype. In more detail, the adhesion of cancer cells to their surrounding stroma induces the generation of intracellular contraction forces that deform their microenvironments by alignment of fibers. The amplitude of these forces can adapt to the mechanical properties of the microenvironment. Moreover, the adhesion strength of cancer cells seems to determine whether a cancer cell is able to migrate through connective tissue or across barriers such as the basement membrane or endothelial cell linings of blood or lymph vessels in order to metastasize. In turn, exposure of adherent cancer cells to physical forces, such as shear flow in vessels or compression forces around tumors, reinforces cell adhesion, regulates cell contractility and restructures the ordering of the local stroma matrix that leads subsequently to secretion of crosslinking proteins or matrix degrading enzymes. Hence invasive cancer cells alter the mechanical properties of their microenvironment. From a mechanobiological point-of-view, the recognized physical signals are transduced into biochemical signaling events that guide cellular responses such as cancer progression after the malignant transition of cancer cells from an epithelial and non-motile phenotype to a mesenchymal and motile (invasive) phenotype providing cellular motility. This transition can also be described as the physical attempt to relate this cancer cell transitional behavior to a T1 phase transition such as the jamming to unjamming transition. During the invasion of cancer cells, cell adaptation occurs to mechanical alterations of the local stroma, such as enhanced stroma upon fibrosis, and therefore we need to uncover underlying mechano-coupling and mechano-regulating functional processes that reinforce the invasion of cancer cells. Moreover, these mechanisms may also be responsible for the awakening of dormant residual cancer cells within the microenvironment. Physicists were initially tempted to consider the steps of the cancer metastasis cascade as single events caused by a single mechanical alteration of the overall properties of the cancer cell. However, this general and simple view has been challenged by the finding that several mechanical properties of cancer cells and their microenvironment influence each other and continuously contribute to tumor growth and cancer progression. In addition, basement membrane crossing, cell invasion and transbarrier migration during cancer progression is explained in physical terms by applying physical principles on living cells regardless of their complexity and individual differences of cancer types. As a novel approach, the impact of the individual microenvironment surrounding cancer cells is also included. Moreover, new theories and models are still needed to understand why certain cancers are malignant and aggressive, while others stay still benign. However, due to the broad variety of cancer types, there may be various pathways solely suitable for specific cancer types and distinct steps in the process of cancer progression. In this review, physical concepts and hypotheses of cancer initiation and progression including cancer cell basement membrane crossing, invasion and transbarrier migration are presented and discussed from a biophysical point-of-view. In addition, the crosstalk between cancer cells and a chronically altered microenvironment, such as fibrosis, is discussed including the basic physical concepts of fibrosis and the cellular responses to mechanical stress caused by the mechanically altered microenvironment. Here, is highlighted how biophysical approaches, both experimentally and theoretically, have an impact on classical hallmarks of cancer and fibrosis and how they contribute to the understanding of the regulation of cancer and its progression by sensing and responding to the physical environmental properties through mechanotransduction processes. Finally, this review discusses various physical models of cell migration such as blebbing, nuclear piston, protrusive force and unjamming transition migration modes and how they contribute to cancer progression. Moreover, these cellular migration modes are influenced by microenvironmental perturbances such as fibrosis that can induce mechanical alterations in cancer cells, which in turn may impact the environment. Hence, the classical hallmarks of cancer need to be refined by including biomechanical properties of cells, cell clusters and tissues and their microenvironment to understand mechano-regulatory processes within cancer cells and the entire organism.
Collapse
|
21
|
Sun W, Gao Y, Ding Y, Cao Y, Chen J, Lv G, Lu J, Yu B, Peng M, Xu H, Sun Y. Catalpol ameliorates advanced glycation end product-induced dysfunction of glomerular endothelial cells via regulating nitric oxide synthesis by inducible nitric oxide synthase and endothelial nitric oxide synthase. IUBMB Life 2019; 71:1268-1283. [PMID: 30861639 DOI: 10.1002/iub.2032] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 02/17/2019] [Indexed: 12/11/2022]
Abstract
Catalpol (Cat.) is an iridoid glucoside extracted from the root of Rehmannia glutinosa Libosch. In this study, we investigated whether Cat. could protect the mouse glomerular endothelial cells against the deleterious effect induced by advanced glycation end products (AGEs) and explored potential mechanisms. We found that 10 μM Cat. showed a protective effect on dead cells stimulated by AGEs. Cat. significantly decreased the expression of p-NF-κBp65 and inducible nitric oxide synthase (iNOS) and increased the expression of phosphorylated-endothelial nitric oxide synthase (p-eNOS; Ser1177), PI3K, p-Akt (Thr308), and total-Akt. Moreover, Cat. restored the integrity of glomerular endothelial barrier by increasing endothelial tight gap junction protein and ameliorated the endothelial hyperpermeability induced by AGEs via modulating the nitric oxide (NO) production. Additionally, Cat. attenuated the massive release of NO induced by AGEs, inhibiting the macrophage infiltration by modulating the NO production, accompanied by the decrease in the release of monocyte chemoattractant protein-1 and intercellular cell adhesion molecule-1 in vitro. Therefore, Cat. ameliorated AGEs-induced endothelial dysfunction via inhibiting the NF-κB/iNOS pathway and activating the PI3K/Akt/eNOS pathway. © 2019 IUBMB Life, 71(9):1268-1283, 2019.
Collapse
Affiliation(s)
- Weixiang Sun
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China.,Department of Pharmacology, School of Pharmacy, Hanlin College, Nanjing University of Chinese Medicine, Taizhou, People's Republic of China
| | - Yuyan Gao
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Yushi Ding
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Ying Cao
- Department of Pharmacology, School of Pharmacy, Hanlin College, Nanjing University of Chinese Medicine, Taizhou, People's Republic of China
| | - Jing Chen
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Gaohong Lv
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Jinfu Lu
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Bin Yu
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Meilin Peng
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Huiqin Xu
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China.,Department of Pharmacology, School of Pharmacy, Hanlin College, Nanjing University of Chinese Medicine, Taizhou, People's Republic of China
| | - Yun Sun
- Department of Pharmacology, School of Pharmacy, Hanlin College, Nanjing University of Chinese Medicine, Taizhou, People's Republic of China
| |
Collapse
|
22
|
Adams JA, Uryash A, Lopez JR, Sackner MA. Whole body periodic acceleration improves survival and microvascular leak in a murine endotoxin model. PLoS One 2019; 14:e0208681. [PMID: 30682019 PMCID: PMC6347233 DOI: 10.1371/journal.pone.0208681] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 01/08/2019] [Indexed: 01/19/2023] Open
Abstract
Sepsis is a life threatening condition which produces multi-organ dysfunction with profound circulatory and cellular derangements. Administration of E.Coli endotoxin (LPS) produces systemic inflammatory effects of sepsis including disruption of endothelial barrier, and if severe enough death. Whole body periodic acceleration (pGz) is the headward-footward motion of the body. pGz has been shown to induce pulsatile shear stress to the endothelium, thereby releasing vascular and cardio protective mediators. The purpose of this study was to determine whether or not pGz performed as a pre-treatment or post-treatment strategy improves survival in a lethal murine endotoxin model.This study was designed as a prospective randomized controlled study in mice. pGz was performed in mice as pre-treatment (pGz-LPS, 3 days prior to LPS), post-treatment (LPS- pGz, 30 min after LPS) strategies or Control (LPS-CONT), in a lethal murine model of endotoxemia. Endotoxemia was induced with intraperitoneal injection of E.Coli LPS (40mg/kg). In a separate group of mice, a nonspecific nitric oxide synthase inhibitor (L-NAME) was provided in their drinking water and pGz-LPS and LPS-pGz performed to determine the effect of nitric oxide (NO) inhibition on survival. In another subset of mice, micro vascular leakage was determined. Behavioral scoring around the clock was performed in all mice at 30 min intervals after LPS administration, until 48 hrs. survival or death. LPS induced 100% mortality in LPS-CONT animals by 30 hrs. In contrast, survival to 48 hrs. occurred in 60% of pGz-LPS and 80% of LPS-pGz. L-NAME abolished the survival effects of pGz. Microvascular leakage was markedly reduced in both pre and post pGz treated animals and was associated with increased tyrosine kinase endothelial-enriched tunica interna endothelial cell kinase 2 (TIE2) receptor and its phosphorylation (p-TIE2). In a murine model of lethal endotoxemia, pGz performed as a pre or post treatment strategy significantly improved survival, and markedly reduced microvascular leakage. The effect was modulated, in part, by NO since a non-selective inhibitor of NO abolished the pGz survival effect.
Collapse
Affiliation(s)
- Jose A. Adams
- Mt. Sinai Medical Center Division of Neonatology, Miami Beach, FL, United States of America
- * E-mail:
| | - Arkady Uryash
- Mt. Sinai Medical Center Division of Neonatology, Miami Beach, FL, United States of America
| | - Jose R. Lopez
- Department of Research, Mount Sinai Medical Center, Miami Beach, FL, United States of America
| | - Marvin A. Sackner
- Emeritus Director Medical Services, Mount Sinai Medical Center, Miami Beach, FL, United States of America
| |
Collapse
|
23
|
Williams IM, McClatchey PM, Bracy DP, Valenzuela FA, Wasserman DH. Acute Nitric Oxide Synthase Inhibition Accelerates Transendothelial Insulin Efflux In Vivo. Diabetes 2018; 67:1962-1975. [PMID: 30002132 PMCID: PMC6152344 DOI: 10.2337/db18-0288] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 07/03/2018] [Indexed: 12/22/2022]
Abstract
Before insulin can stimulate glucose uptake in muscle, it must be delivered to skeletal muscle (SkM) through the microvasculature. Insulin delivery is determined by SkM perfusion and the rate of movement of insulin across the capillary endothelium. The endothelium therefore plays a central role in regulating insulin access to SkM. Nitric oxide (NO) is a key regulator of endothelial function and stimulates arterial vasodilation, which increases SkM perfusion and the capillary surface area available for insulin exchange. The effects of NO on transendothelial insulin efflux (TIE), however, are unknown. We hypothesized that acute reduction of endothelial NO would reduce TIE. However, intravital imaging of TIE in mice revealed that reduction of NO by l-NG-nitro-l-arginine methyl ester (l-NAME) enhanced the rate of TIE by ∼30% and increased total extravascular insulin delivery. This accelerated TIE was associated with more rapid insulin-stimulated glucose lowering. Sodium nitroprusside, an NO donor, had no effect on TIE in mice. The effects of l-NAME on TIE were not due to changes in blood pressure alone, as a direct-acting vasoconstrictor (phenylephrine) did not affect TIE. These results demonstrate that acute NO synthase inhibition increases the permeability of capillaries to insulin, leading to an increase in delivery of insulin to SkM.
Collapse
Affiliation(s)
- Ian M Williams
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| | - P Mason McClatchey
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| | - Deanna P Bracy
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
- Mouse Metabolic Phenotyping Center, Vanderbilt University, Nashville, TN
| | | | - David H Wasserman
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
- Mouse Metabolic Phenotyping Center, Vanderbilt University, Nashville, TN
| |
Collapse
|
24
|
Mira E, Carmona-Rodríguez L, Pérez-Villamil B, Casas J, Fernández-Aceñero MJ, Martínez-Rey D, Martín-González P, Heras-Murillo I, Paz-Cabezas M, Tardáguila M, Oury TD, Martín-Puig S, Lacalle RA, Fabriás G, Díaz-Rubio E, Mañes S. SOD3 improves the tumor response to chemotherapy by stabilizing endothelial HIF-2α. Nat Commun 2018; 9:575. [PMID: 29422508 PMCID: PMC5805714 DOI: 10.1038/s41467-018-03079-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 01/18/2018] [Indexed: 02/08/2023] Open
Abstract
One drawback of chemotherapy is poor drug delivery to tumor cells, due in part to hyperpermeability of the tumor vasculature. Extracellular superoxide dismutase (SOD3) is an antioxidant enzyme usually repressed in the tumor milieu. Here we show that specific SOD3 re-expression in tumor-associated endothelial cells (ECs) increases doxorubicin (Doxo) delivery into and chemotherapeutic effect on tumors. Enhanced SOD3 activity fostered perivascular nitric oxide accumulation and reduced vessel leakage by inducing vascular endothelial cadherin (VEC) transcription. SOD3 reduced HIF prolyl hydroxylase domain protein activity, which increased hypoxia-inducible factor-2α (HIF-2α) stability and enhanced its binding to a specific VEC promoter region. EC-specific HIF-2α ablation prevented both the SOD3-mediated increase in VEC transcription and the enhanced Doxo effect. SOD3, VEC, and HIF-2α levels correlated positively in primary colorectal cancers, which suggests a similar interconnection of these proteins in human malignancy. Tumour vasculature influences drug delivery. Here, the authors show that SOD3 re-expression enhances doxorubicin delivery and effects through normalization of tumour vasculature via the HIF-2a/VE-cadherin pathway.
Collapse
Affiliation(s)
- Emilia Mira
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Darwin, 3, Madrid, 28049, Spain
| | - Lorena Carmona-Rodríguez
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Darwin, 3, Madrid, 28049, Spain
| | - Beatriz Pérez-Villamil
- Genomics and Microarray Laboratory, Medical Oncology & Surgical Pathology Departments, Instituto de Investigación Sanitaria San Carlos Hospital Clínico San Carlos, Univ. Complutense de Madrid, CIBERONC, Profesor Martín Lagos, S/N, Madrid, 28040, Spain
| | - Josefina Casas
- Department of Biomedicinal Chemistry, Institute of Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18-26, Barcelona, 08034, Spain
| | - María Jesús Fernández-Aceñero
- Genomics and Microarray Laboratory, Medical Oncology & Surgical Pathology Departments, Instituto de Investigación Sanitaria San Carlos Hospital Clínico San Carlos, Univ. Complutense de Madrid, CIBERONC, Profesor Martín Lagos, S/N, Madrid, 28040, Spain
| | - Diego Martínez-Rey
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Darwin, 3, Madrid, 28049, Spain
| | - Paula Martín-González
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Darwin, 3, Madrid, 28049, Spain
| | - Ignacio Heras-Murillo
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Darwin, 3, Madrid, 28049, Spain
| | - Mateo Paz-Cabezas
- Genomics and Microarray Laboratory, Medical Oncology & Surgical Pathology Departments, Instituto de Investigación Sanitaria San Carlos Hospital Clínico San Carlos, Univ. Complutense de Madrid, CIBERONC, Profesor Martín Lagos, S/N, Madrid, 28040, Spain
| | - Manuel Tardáguila
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Darwin, 3, Madrid, 28049, Spain.,Genetics Institute, University of Florida, 2033 Mowry Road, Gainesville, FL, 32610, USA
| | - Tim D Oury
- Department of Pathology, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, PA, 15261, USA
| | - Silvia Martín-Puig
- Myocardial Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares, Calle de Melchor Fernández Almagro, 3, Madrid, 28029, Spain
| | - Rosa Ana Lacalle
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Darwin, 3, Madrid, 28049, Spain
| | - Gemma Fabriás
- Department of Biomedicinal Chemistry, Institute of Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18-26, Barcelona, 08034, Spain
| | - Eduardo Díaz-Rubio
- Genomics and Microarray Laboratory, Medical Oncology & Surgical Pathology Departments, Instituto de Investigación Sanitaria San Carlos Hospital Clínico San Carlos, Univ. Complutense de Madrid, CIBERONC, Profesor Martín Lagos, S/N, Madrid, 28040, Spain
| | - Santos Mañes
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Darwin, 3, Madrid, 28049, Spain.
| |
Collapse
|
25
|
Roca F, Iacob M, Remy-Jouet I, Bellien J, Joannides R. Evidence for a Role of Vascular Endothelium in the Control of Arterial Wall Viscosity in Humans. Hypertension 2018; 71:143-150. [DOI: 10.1161/hypertensionaha.117.09870] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 06/28/2017] [Accepted: 10/16/2017] [Indexed: 11/16/2022]
Affiliation(s)
- Frederic Roca
- From the Department of Pharmacology, Rouen University Hospital, France (F.R., M.I., J.B., R.J.); Inserm U1096, Normandie Univ, UNIROUEN, France (F.R., M.I., I.R.-J., J.B., R.J.); Institute for Research and Innovation in Biomedicine, University of Rouen, France (F.R., I.R.-J., J.B., R.J.); and Clinical Investigation Center CIC-CRB 1404, Rouen University Hospital, France (F.R., J.B., R.J.)
| | - Michele Iacob
- From the Department of Pharmacology, Rouen University Hospital, France (F.R., M.I., J.B., R.J.); Inserm U1096, Normandie Univ, UNIROUEN, France (F.R., M.I., I.R.-J., J.B., R.J.); Institute for Research and Innovation in Biomedicine, University of Rouen, France (F.R., I.R.-J., J.B., R.J.); and Clinical Investigation Center CIC-CRB 1404, Rouen University Hospital, France (F.R., J.B., R.J.)
| | - Isabelle Remy-Jouet
- From the Department of Pharmacology, Rouen University Hospital, France (F.R., M.I., J.B., R.J.); Inserm U1096, Normandie Univ, UNIROUEN, France (F.R., M.I., I.R.-J., J.B., R.J.); Institute for Research and Innovation in Biomedicine, University of Rouen, France (F.R., I.R.-J., J.B., R.J.); and Clinical Investigation Center CIC-CRB 1404, Rouen University Hospital, France (F.R., J.B., R.J.)
| | - Jeremy Bellien
- From the Department of Pharmacology, Rouen University Hospital, France (F.R., M.I., J.B., R.J.); Inserm U1096, Normandie Univ, UNIROUEN, France (F.R., M.I., I.R.-J., J.B., R.J.); Institute for Research and Innovation in Biomedicine, University of Rouen, France (F.R., I.R.-J., J.B., R.J.); and Clinical Investigation Center CIC-CRB 1404, Rouen University Hospital, France (F.R., J.B., R.J.)
| | - Robinson Joannides
- From the Department of Pharmacology, Rouen University Hospital, France (F.R., M.I., J.B., R.J.); Inserm U1096, Normandie Univ, UNIROUEN, France (F.R., M.I., I.R.-J., J.B., R.J.); Institute for Research and Innovation in Biomedicine, University of Rouen, France (F.R., I.R.-J., J.B., R.J.); and Clinical Investigation Center CIC-CRB 1404, Rouen University Hospital, France (F.R., J.B., R.J.)
| |
Collapse
|
26
|
The Relation Between Endothelial Nitric Oxide Synthase Polymorphisms and Normal Tension Glaucoma. J Glaucoma 2017; 26:1030-1035. [PMID: 28777225 DOI: 10.1097/ijg.0000000000000751] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
PURPOSE We investigated whether polymorphisms of the endothelial NO synthase (eNOS) gene are associated with normal tension glaucoma (NTG). We also investigated whether the eNOS polymorphisms are associated with NTG subgroups [NTG with and without optic disc hemorrhage (DH)]. METHODS A total of 251 patients with NTG and 245 healthy volunteers were enrolled in this study. DNA from peripheral blood leukocytes was extracted, and the genotypes of 4 polymorphisms (rs2070744, rs1549758, rs1799983, and rs2566514) in the eNOS gene were determined using restriction fragment length polymorphism and the SNaPshot method. The primary outcome was to investigate the relation between eNOS polymorphisms and NTG. The secondary outcome was to compare the frequencies of the polymorphic genotypes among the NTG subgroups. Bonferroni correction was used to adjust for type I error. RESULTS In all subjects, the genotype distribution was in accordance with Hardy-Weinberg equilibrium. None of the 4 polymorphisms showed any significant difference in the frequencies of alleles or genotypes between the NTG patients and controls. In the further analysis comparing the genotypic frequencies between NTG with DH and normal controls, the CC/CT genotype of rs2070744 was significantly associated with DH in NTG patients (genotypic association test, P-value=0.0041). On the multiple logistic regression analysis adjusted for covariates such as sex and age, the NTG with DH was associated with polymorphic genotypes of rs2070744 with a borderline significance (additive genetic model, P=0.0070). CONCLUSIONS Our results indicates that eNOS rs2070744 can be associated with NTG patients with DH. This finding suggests that the eNOS polymorphism may be a genetic risk factor in the development of DH in NTG patients.
Collapse
|
27
|
Aliancy J, Stamer WD, Wirostko B. A Review of Nitric Oxide for the Treatment of Glaucomatous Disease. Ophthalmol Ther 2017; 6:221-232. [PMID: 28584936 PMCID: PMC5693832 DOI: 10.1007/s40123-017-0094-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Indexed: 12/21/2022] Open
Abstract
Glaucoma is the leading cause of irreversible blindness worldwide, affecting 64.3 million people. An estimated 60.5 million people are affected by primary open angle glaucoma globally, and this will increase to 111.8 million by 2040. The definition of glaucoma has evolved greatly over time. Although multiple risk factors such as ischemia, inflammation, myopia, race, age and low ocular perfusion pressure may play a role, intraocular pressure (IOP) is still the main risk factor we can easily identify and modify. Currently, both medical and surgical interventions aim to reduce IOP. Effective IOP reduction controls and prevents the progression in many cases of glaucoma. Although this multifactorial disease's true pathophysiology is difficult to elucidate, physiologic mediators including nitric oxide (NO) are being evaluated as novel ways to impact progression by both lowering IOP and improving optic nerve head perfusion. Latanoprostene bunod 0.024% is an emerging therapeutic agent that has shown promise in clinical trials. As a nitric oxide-donating prostaglandin F2-alpha receptor agonist, it has proven to effectively, and with good tolerability, reduce IOP in glaucoma and ocular hypertensive patients. Latanoprostene bunod capitalizes on NO's ability to modulate the conventional aqueous humor outflow system, directly improving outflow through the trabecular meshwork, Schlemm's canal and distal scleral vessels. Importantly, targeting the conventional outflow tissues with NO-donating drugs represents an opportunity to restore outflow function, which will most likely have a beneficial consequence of additional IOP-lowering effects with dampening of diurnal and other IOP fluctuations, the benefit of a healthy trabecular meshwork.
Collapse
Affiliation(s)
- Joah Aliancy
- Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - W Daniel Stamer
- Department of Ophthalmology, Duke University, Durham, NC, USA
| | | |
Collapse
|
28
|
Palygin O, Ilatovskaya DV, Levchenko V, Endres BT, Geurts AM, Staruschenko A. Nitric oxide production by glomerular podocytes. Nitric Oxide 2017; 72:24-31. [PMID: 29128399 DOI: 10.1016/j.niox.2017.11.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 09/27/2017] [Accepted: 11/07/2017] [Indexed: 01/18/2023]
Abstract
Nitric Oxide (NO), a potent vasodilator and vital signaling molecule, has been shown to contribute to the regulation of glomerular ultrafiltration. However, whether changes in NO occur in podocytes during the pathogenesis of salt-sensitive hypertension has not yet been thoroughly examined. We showed here that podocytes produce NO, and further hypothesized that hypertensive animals would exhibit reduced NO production in these cells in response to various paracrine factors, which might contribute to the damage of glomeruli filtration barrier and development of proteinuria. To test this, we isolated glomeruli from the kidneys of Dahl salt-sensitive (SS) rats fed a low salt (LS; 0.4% NaCl) or high salt (HS; 4% NaCl, 3 weeks) diets and loaded podocytes with either a combination of NO and Ca2+ fluorophores (DAF-FM and Fura Red, respectively) or DAF-FM alone. Changes in fluorescence were observed with confocal microscopy in response to adenosine triphosphate (ATP), angiotensin II (Ang II), and hydrogen peroxide (H2O2). Application of Ang II resulted in activation of both NO and intracellular calcium ([Ca2+]i) transients. In contrast, ATP promoted [Ca2+]i transients, but did not have any effects on NO production. SS rats fed a HS diet for 3 weeks demonstrated impaired NO production: the response to Ang II or H2O2 in podocytes of glomeruli isolated from SS rats fed a HS diet was significantly reduced compared to rats fed a LS diet. Therefore, glomerular podocytes from hypertensive rats showed a diminished NO release in response to Ang II or oxidative stress, suggesting that podocytic NO signaling is dysfunctional in this condition and likely contributes to the development of kidney injury.
Collapse
Affiliation(s)
- Oleg Palygin
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Daria V Ilatovskaya
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Vladislav Levchenko
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Bradley T Endres
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Aron M Geurts
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | | |
Collapse
|
29
|
Cui SX, Epstein FH. MRI assessment of coronary microvascular endothelial nitric oxide synthase function using myocardial T 1 mapping. Magn Reson Med 2017; 79:2246-2253. [PMID: 28782150 DOI: 10.1002/mrm.26870] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 06/28/2017] [Accepted: 07/19/2017] [Indexed: 01/07/2023]
Abstract
PURPOSE Endothelial nitric oxide synthase (eNOS) plays a central role in regulating vascular tone, blood flow, and microvascular permeability. Endothelial dysfunction, including eNOS dysfunction, is an early biomarker of vascular disease. This study aimed to show that myocardial T1 mapping during nitric oxide synthase (NOS) inhibition could assess coronary microvascular eNOS function. METHODS Wild-type mice, eNOS-/- mice, and wild-type mice fed a high-fat diet underwent T1 mapping at baseline and for 20 min after injection of NG -nitro-L-arginine methyl ester (LNAME), a NOS inhibitor. First-pass perfusion MRI was performed in wild-type mice at baseline and 5 min after LNAME injection. RESULTS T1 mapping detected an increase in myocardial T1 5 min after an injection of 4 mg/kg LNAME compared with baseline in control mice (T1 = 1515 ± 30 ms with LNAME versus T1 = 1402 ± 30 ms at baseline, P < 0.05). No change in myocardial T1 after LNAME injection was observed in eNOS-/- mice. The change in T1 after LNAME injection was less in high-fat-diet mice (ΔT1 = 31 ± 14 ms at 12 weeks of diet and ΔT1 = 16 ± 17 ms at 18 weeks of diet) compared with mice fed a standard diet (ΔT1 = 113 ± 15 ms), with P < 0.05. First-pass MRI measured similar perfusion at baseline and 5 min after LNAME injection. CONCLUSIONS NOS inhibition causes an increase in myocardial T1 in healthy mice, and this effect is mediated through eNOS. T1 mapping during NOS inhibition detects coronary microvascular eNOS dysfunction in high-fat-diet mice. T1 mapping during NOS inhibition may be useful in preclinical studies aiming to investigate mechanisms underlying and therapies for coronary microvascular eNOS dysfunction. Magn Reson Med 79:2246-2253, 2018. © 2017 International Society for Magnetic Resonance in Medicine.
Collapse
Affiliation(s)
- Sophia X Cui
- Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Frederick H Epstein
- Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA.,Radiology, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
30
|
Czikora I, Alli AA, Sridhar S, Matthay MA, Pillich H, Hudel M, Berisha B, Gorshkov B, Romero MJ, Gonzales J, Wu G, Huo Y, Su Y, Verin AD, Fulton D, Chakraborty T, Eaton DC, Lucas R. Epithelial Sodium Channel-α Mediates the Protective Effect of the TNF-Derived TIP Peptide in Pneumolysin-Induced Endothelial Barrier Dysfunction. Front Immunol 2017; 8:842. [PMID: 28785264 PMCID: PMC5519615 DOI: 10.3389/fimmu.2017.00842] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 07/04/2017] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Streptococcus pneumoniae is a major etiologic agent of bacterial pneumonia. Autolysis and antibiotic-mediated lysis of pneumococci induce release of the pore-forming toxin, pneumolysin (PLY), their major virulence factor, which is a prominent cause of acute lung injury. PLY inhibits alveolar liquid clearance and severely compromises alveolar-capillary barrier function, leading to permeability edema associated with pneumonia. As a consequence, alveolar flooding occurs, which can precipitate lethal hypoxemia by impairing gas exchange. The α subunit of the epithelial sodium channel (ENaC) is crucial for promoting Na+ reabsorption across Na+-transporting epithelia. However, it is not known if human lung microvascular endothelial cells (HL-MVEC) also express ENaC-α and whether this subunit is involved in the regulation of their barrier function. METHODS The presence of α, β, and γ subunits of ENaC and protein phosphorylation status in HL-MVEC were assessed in western blotting. The role of ENaC-α in monolayer resistance of HL-MVEC was examined by depletion of this subunit by specific siRNA and by employing the TNF-derived TIP peptide, a specific activator that directly binds to ENaC-α. RESULTS HL-MVEC express all three subunits of ENaC, as well as acid-sensing ion channel 1a (ASIC1a), which has the capacity to form hybrid non-selective cation channels with ENaC-α. Both TIP peptide, which specifically binds to ENaC-α, and the specific ASIC1a activator MitTx significantly strengthened barrier function in PLY-treated HL-MVEC. ENaC-α depletion significantly increased sensitivity to PLY-induced hyperpermeability and in addition, blunted the protective effect of both the TIP peptide and MitTx, indicating an important role for ENaC-α and for hybrid NSC channels in barrier function of HL-MVEC. TIP peptide blunted PLY-induced phosphorylation of both calmodulin-dependent kinase II (CaMKII) and of its substrate, the actin-binding protein filamin A (FLN-A), requiring the expression of both ENaC-α and ASIC1a. Since non-phosphorylated FLN-A promotes ENaC channel open probability and blunts stress fiber formation, modulation of this activity represents an attractive target for the protective actions of ENaC-α in both barrier function and liquid clearance. CONCLUSION Our results in cultured endothelial cells demonstrate a previously unrecognized role for ENaC-α in strengthening capillary barrier function that may apply to the human lung. Strategies aiming to activate endothelial NSC channels that contain ENaC-α should be further investigated as a novel approach to improve barrier function in the capillary endothelium during pneumonia.
Collapse
Affiliation(s)
- Istvan Czikora
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Abdel A Alli
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, United States.,Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| | - Supriya Sridhar
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Michael A Matthay
- Cardiovascular Research Institute, UCSF, San Francisco, CA, United States
| | - Helena Pillich
- Institute for Medical Microbiology, Justus-Liebig University, Giessen, Germany
| | - Martina Hudel
- Institute for Medical Microbiology, Justus-Liebig University, Giessen, Germany
| | - Besim Berisha
- Institute for Medical Microbiology, Justus-Liebig University, Giessen, Germany
| | - Boris Gorshkov
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Maritza J Romero
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States.,Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Joyce Gonzales
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Guangyu Wu
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Yuqing Huo
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States.,Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Yunchao Su
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Alexander D Verin
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States.,Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - David Fulton
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States.,Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Trinad Chakraborty
- Institute for Medical Microbiology, Justus-Liebig University, Giessen, Germany
| | - Douglas C Eaton
- Department of Physiology, Emory University School of Medicine, Atlanta, GA, United States
| | - Rudolf Lucas
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States.,Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States.,Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
31
|
Hwang SJ, Kim JW. Effect of Tetrahydrozoline on the Permeability of Trabecular Meshwork Cell Monolayer. JOURNAL OF THE KOREAN OPHTHALMOLOGICAL SOCIETY 2017. [DOI: 10.3341/jkos.2017.58.1.69] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- Seok Jin Hwang
- Department of Ophthalmology, Catholic University of Daegu School of Medicine, Daegu, Korea
| | - Jae Woo Kim
- Department of Ophthalmology, Catholic University of Daegu School of Medicine, Daegu, Korea
| |
Collapse
|
32
|
RhoA S-nitrosylation as a regulatory mechanism influencing endothelial barrier function in response to G +-bacterial toxins. Biochem Pharmacol 2016; 127:34-45. [PMID: 28017778 DOI: 10.1016/j.bcp.2016.12.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 12/19/2016] [Indexed: 01/05/2023]
Abstract
Disruption of the endothelial barrier in response to Gram positive (G+) bacterial toxins is a major complication of acute lung injury (ALI) and can be further aggravated by antibiotics which stimulate toxin release. The integrity of the pulmonary endothelial barrier is mediated by the balance of disruptive forces such as the small GTPase RhoA, and protective forces including endothelium-derived nitric oxide (NO). How NO protects against the barrier dysfunction is incompletely understood and our goal was to determine whether NO and S-nitrosylation can modulate RhoA activity and whether this mechanism is important for G+ toxin-induced microvascular permeability. We found that the G+ toxin listeriolysin-O (LLO) increased RhoA activity and that NO and S-NO donors inhibit RhoA activity. RhoA was robustly S-nitrosylated as determined by biotin-switch and mercury column analysis. MS revealed that three primary cysteine residues are S-nitrosylated including cys16, cys20 and cys159. Mutation of these residues to serine diminished S-nitrosylation to endogenous NO and mutant RhoA was less sensitive to inhibition by S-NO. G+-toxins stimulated the denitrosylation of RhoA which was not mediated by S-nitrosoglutathione reductase (GSNOR), thioredoxin (TRX) or thiol-dependent enzyme activity but was instead stimulated directly by elevated calcium levels. Calcium-promoted the direct denitrosylation of WT but not mutant RhoA and mutant RhoA adenovirus was more effective than WT in disrupting the barrier integrity of human lung microvascular endothelial cells. In conclusion, we reveal a novel mechanism by which NO and S-nitrosylation reduces RhoA activity which may be of significance in the management of pulmonary endothelial permeability induced by G+-toxins.
Collapse
|
33
|
Mamiya A, Kitano H, Kokubun S, Hidai C. Activation peptide of coagulation factor IX regulates endothelial permeability. Transl Res 2016; 177:70-84.e5. [PMID: 27392935 DOI: 10.1016/j.trsl.2016.06.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 06/11/2016] [Accepted: 06/15/2016] [Indexed: 01/07/2023]
Abstract
Endothelial hyperpermeability is involved in several critical illnesses, and its regulatory mechanisms have been intensively investigated. It was recently reported that the activation peptide of coagulation factor IX enhances cell matrix and intercellular adhesion. The aim of this study was to investigate the role of activation peptide of coagulation factor IX in intercellular adhesion of endothelial cells and evaluate its effects on endothelial permeability. In the presence of activation peptide, cells spread with lamellipodium-like broad protrusions multidirectionally, increasing the area of adhesion to matrix by 16% within 30 minutes. In intercellular adhesion, treatment with activation peptide induced overlapping of adjacent cell edges and remodeling of intercellular adhesion sites, with colocalization of the adherens junction proteins VE-cadherin and β-catenin and a marker protein of the lateral border recycling compartment, PECAM. Activation peptide decreased gaps between cells by 66% in cultured endothelial cells and suppressed increased endothelial cell monolayer permeability induced by interleukin-1β in a dose-dependent manner. Treatment with activation peptide decreased eNOS protein expression and altered its subcellular distribution, decreasing intracellular cGMP. An analogue of cGMP suppressed the effects of activation peptide on cell spreading. In addition, the effect of activation peptide on hyperpermeability was investigated in mice injected with lipopolysaccharide. Intravenous injection of lipopolysaccharide increased lung weight by 28%, and treatment with activation peptide significantly suppressed the increase in lung weight to 5%. Our results indicate that activation peptide of factor IX regulates endothelial intercellular adhesion and thus could be used in the treatment of vascular hyperpermeability.
Collapse
Affiliation(s)
- Atsushi Mamiya
- Department of Otorhinolaryngology-Head and Neck Surgery, Nihon University School of Medicine, Tokyo, Japan
| | - Hisataka Kitano
- Department of Otorhinolaryngology-Head and Neck Surgery, Nihon University School of Medicine, Tokyo, Japan
| | - Shinichiro Kokubun
- Department of Biomedical Science, Nihon University School of Medicine, Tokyo, Japan
| | - Chiaki Hidai
- Department of Biomedical Science, Nihon University School of Medicine, Tokyo, Japan.
| |
Collapse
|
34
|
Gao F, Artham S, Sabbineni H, Al-Azayzih A, Peng XD, Hay N, Adams RH, Byzova TV, Somanath PR. Akt1 promotes stimuli-induced endothelial-barrier protection through FoxO-mediated tight-junction protein turnover. Cell Mol Life Sci 2016; 73:3917-33. [PMID: 27113546 PMCID: PMC5023469 DOI: 10.1007/s00018-016-2232-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 04/12/2016] [Accepted: 04/14/2016] [Indexed: 11/28/2022]
Abstract
Vascular permeability regulated by the vascular endothelial growth factor (VEGF) through endothelial-barrier junctions is essential for inflammation. Mechanisms regulating vascular permeability remain elusive. Although 'Akt' and 'Src' have been implicated in the endothelial-barrier regulation, it is puzzling how both agents that protect and disrupt the endothelial-barrier activate these kinases to reciprocally regulate vascular permeability. To delineate the role of Akt1 in endothelial-barrier regulation, we created endothelial-specific, tamoxifen-inducible Akt1 knockout mice and stable ShRNA-mediated Akt1 knockdown in human microvascular endothelial cells. Akt1 loss leads to decreased basal and angiopoietin1-induced endothelial-barrier resistance, and enhanced VEGF-induced endothelial-barrier breakdown. Endothelial Akt1 deficiency resulted in enhanced VEGF-induced vascular leakage in mice ears, which was rescued upon re-expression with Adeno-myrAkt1. Furthermore, co-treatment with angiopoietin1 reversed VEGF-induced vascular leakage in an Akt1-dependent manner. Mechanistically, our study revealed that while VEGF-induced short-term vascular permeability is independent of Akt1, its recovery is reliant on Akt1 and FoxO-mediated claudin expression. Pharmacological inhibition of FoxO transcription factors rescued the defective endothelial barrier due to Akt1 deficiency. Here we provide novel insights on the endothelial-barrier protective role of VEGF in the long term and the importance of Akt1-FoxO signaling on tight-junction stabilization and prevention of vascular leakage through claudin expression.
Collapse
Affiliation(s)
- Fei Gao
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, USA
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Sandeep Artham
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Harika Sabbineni
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Ahmad Al-Azayzih
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, USA
- College of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Xiao-Ding Peng
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Nissim Hay
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Ralf H Adams
- Max Plank Institute of Molecular Biomedicine, Röntgenstraße 20, Münster, Germany
| | - Tatiana V Byzova
- Department of Molecular Cardiology, Joseph J. Jacob's Center for Thrombosis and Vascular Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Payaningal R Somanath
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, USA.
- Department of Medicine and Vascular Biology Center, Augusta University, HM1200, Augusta, GA, 30912, USA.
| |
Collapse
|
35
|
Dolinina J, Sverrisson K, Rippe A, Öberg CM, Rippe B. Nitric oxide synthase inhibition causes acute increases in glomerular permeability in vivo, dependent upon reactive oxygen species. Am J Physiol Renal Physiol 2016; 311:F984-F990. [PMID: 27681559 DOI: 10.1152/ajprenal.00152.2016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 09/21/2016] [Indexed: 11/22/2022] Open
Abstract
There is increasing evidence that the permeability of the glomerular filtration barrier (GFB) is partly regulated by a balance between the bioavailability of nitric oxide (NO) and that of reactive oxygen species (ROS). It has been postulated that normal or moderately elevated NO levels protect the GFB from permeability increases, whereas ROS, through reducing the bioavailability of NO, have the opposite effect. We tested the tentative antagonism between NO and ROS on glomerular permeability in anaesthetized Wistar rats, in which the left ureter was cannulated for urine collection while simultaneously blood access was achieved. Rats were systemically infused with either l-NAME or l-NAME together with the superoxide scavenger Tempol, or together with l-arginine or the NO-donor DEA-NONOate, or the cGMP agonist 8-bromo-cGMP. To measure glomerular sieving coefficients (theta, θ) to Ficoll, rats were infused with FITC-Ficoll 70/400 (mol/radius 10-80 Å). Plasma and urine samples were analyzed by high-performance size-exclusion chromatography (HPSEC) for determination of θ for Ficoll repeatedly during up to 2 h. l-NAME increased θ for Ficoll70Å from 2.27 ± 1.30 × 10-5 to 8.46 ± 2.06 × 10-5 (n = 6, P < 0.001) in 15 min. Tempol abrogated these increases in glomerular permeability and an inhibition was also observed with l-arginine and with 8-bromo-cGMP. In conclusion, acute NO synthase inhibition in vivo by l-NAME caused rapid increases in glomerular permeability, which could be reversed by either an ROS antagonist or by activating the guanylyl cyclase-cGMP pathway. The data strongly suggest a protective effect of NO in maintaining normal glomerular permeability in vivo.
Collapse
Affiliation(s)
| | | | - Anna Rippe
- Department of Nephrology, Lund University, Lund, Sweden
| | - Carl M Öberg
- Department of Nephrology, Lund University, Lund, Sweden
| | - Bengt Rippe
- Department of Nephrology, Lund University, Lund, Sweden
| |
Collapse
|
36
|
Abstract
Diabetic nephropathy is the main cause of end-stage renal failure in the Western world. In diabetes, metabolic and haemodynamic perturbations disrupt the integrity of the glomerular filtration barrier, leading to ultrastructural alterations of the glomeruli, including podocyte foot process fusion and detachment, glomerular basement membrane thickening, reduced endothelial cell glycocalyx, and mesangial extracellular matrix accumulation and glomerulosclerosis, ultimately leading to albuminuria and end-stage renal disease. Many vascular growth factors, such as angiopoietins, are implicated in glomerular biology. In normal physiology angiopoietins regulate the function of the glomerular filtration barrier. When they are dysregulated, however, as they are in diabetes, they drive the cellular mechanisms that mediate diabetic glomerular pathology. Modulation of angiopoietins expression and signalling has been proposed as a tool to correct the cellular mechanisms involved in the pathophysiology of diabetic microvascular disease, such as retinopathy in humans. Future work might evaluate whether this novel therapeutic approach should be extended to diabetic kidney disease.
Collapse
Affiliation(s)
- Luigi Gnudi
- Unit for Metabolic Medicine, Cardiovascular Division, Faculty of Life Science & Medicine, King's College London, 3rd Floor Franklin-Wilkins Building, Waterloo Campus, Stamford Street, London, SE1 9RT, UK.
| |
Collapse
|
37
|
Olszewska-Pazdrak B, McVicar SD, Rayavara K, Moya SM, Kantara C, Gammarano C, Olszewska P, Fuller GM, Sower LE, Carney DH. Nuclear Countermeasure Activity of TP508 Linked to Restoration of Endothelial Function and Acceleration of DNA Repair. Radiat Res 2016; 186:162-74. [PMID: 27388041 DOI: 10.1667/rr14409.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
There is increasing evidence that radiation-induced damage to endothelial cells and loss of endothelial function may contribute to both acute radiation syndromes and long-term effects of whole-body nuclear irradiation. Therefore, several drugs are being developed to mitigate the effects of nuclear radiation, most of these drugs will target and protect or regenerate leukocytes and platelets. Our laboratory has demonstrated that TP508, a 23-amino acid thrombin peptide, activates endothelial cells and stem cells to revascularize and regenerate tissues. We now show that TP508 can mitigate radiation-induced damage to endothelial cells in vitro and in vivo. Our in vitro results demonstrate that human endothelial cells irradiation attenuates nitric oxide (NO) signaling, disrupts tube formation and induces DNA double-strand breaks (DSB). TP508 treatment reverses radiation effects on NO signaling, restores tube formation and accelerates the repair of radiation-induced DSB. The radiation-mitigating effects of TP508 on endothelial cells were also seen in CD-1 mice where systemic injection of TP508 stimulated endothelial cell sprouting from aortic explants after 8 Gy irradiation. Systemic doses of TP508 that mitigated radiation-induced endothelial cell damage, also significantly increased survival of CD-1 mice when injected 24 h after 8.5 Gy exposure. These data suggest that increased survival observed with TP508 treatment may be due to its effects on vascular and microvascular endothelial cells. Our study supports the usage of a regenerative drug such as TP508 to activate endothelial cells as a countermeasure for mitigating the effects of nuclear radiation.
Collapse
Affiliation(s)
- Barbara Olszewska-Pazdrak
- a Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas and
| | - Scott D McVicar
- a Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas and
| | | | - Stephanie M Moya
- a Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas and
| | - Carla Kantara
- a Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas and.,b Chrysalis BioTherapeutics, Inc., Galveston, Texas
| | - Chris Gammarano
- a Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas and
| | - Paulina Olszewska
- a Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas and
| | | | | | - Darrell H Carney
- a Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas and.,b Chrysalis BioTherapeutics, Inc., Galveston, Texas
| |
Collapse
|
38
|
Tawada M, Ito Y, Hamada C, Honda K, Mizuno M, Suzuki Y, Sakata F, Terabayashi T, Matsukawa Y, Maruyama S, Imai E, Matsuo S, Takei Y. Vascular Endothelial Cell Injury Is an Important Factor in the Development of Encapsulating Peritoneal Sclerosis in Long-Term Peritoneal Dialysis Patients. PLoS One 2016; 11:e0154644. [PMID: 27119341 PMCID: PMC4847858 DOI: 10.1371/journal.pone.0154644] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 04/15/2016] [Indexed: 12/22/2022] Open
Abstract
Background and Objectives Encapsulating peritoneal sclerosis (EPS) is a rare but serious and life-threatening complication of peritoneal dialysis (PD). However, the precise pathogenesis remains unclear; in addition, predictors and early diagnostic biomarkers for EPS have not yet to be established. Methods Eighty-three peritoneal membrane samples taken at catheter removal were examined to identify pathological characteristics of chronic peritoneal deterioration, which promotes EPS in patients undergoing long-term PD treatment with low occurrence of peritonitis. Results According to univariable logistic regression analysis of the pathological findings, thickness of the peritoneal membrane (P = 0.045), new membrane formation score (P = 0.006), ratio of luminal diameter to vessel diameter (L/V ratio, P<0.001), presence of CD31-negative vessels (P = 0.021), fibrin deposition (P<0.001), and collagen volume fraction (P = 0.018) were associated with EPS development. In analyses of samples with and without EPS matched for PD treatment period, non-diabetes, and PD solution, univariable analysis identified L/V ratio (per 0.1 increase: odds ratio (OR) 0.44, P = 0.003) and fibrin deposition (OR 6.35, P = 0.027) as the factors associated with EPS. L/V ratio was lower in patients with fibrin exudation than in patients without fibrin exudation. Conclusions These findings suggest that damage to vascular endothelial cells, as represented by low L/V ratio, could be a predictive finding for the development of EPS, particularly in long-term PD patients unaffected by peritonitis.
Collapse
Affiliation(s)
- Mitsuhiro Tawada
- Department of Nephrology and Renal Replacement Therapy, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuhiko Ito
- Department of Nephrology and Renal Replacement Therapy, Nagoya University Graduate School of Medicine, Nagoya, Japan
- * E-mail:
| | - Chieko Hamada
- Department of Nephrology, Juntendo University, Tokyo, Japan
| | - Kazuho Honda
- Department of Pathology, Tokyo Women’s Medical University, Tokyo, Japan
| | - Masashi Mizuno
- Department of Nephrology and Renal Replacement Therapy, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuhiro Suzuki
- Department of Nephrology and Renal Replacement Therapy, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Fumiko Sakata
- Department of Nephrology and Renal Replacement Therapy, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takeshi Terabayashi
- Department of Nephrology and Renal Replacement Therapy, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshihisa Matsukawa
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shoichi Maruyama
- Department of Nephrology and Renal Replacement Therapy, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Enyu Imai
- Department of Nephrology and Renal Replacement Therapy, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Seiichi Matsuo
- Department of Nephrology and Renal Replacement Therapy, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshifumi Takei
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
39
|
Abstract
Kidney glomeruli ultrafilter blood to generate urine and they are dysfunctional in a variety of kidney diseases. There are two key vascular growth factor families implicated in glomerular biology and function, namely the vascular endothelial growth factors (VEGFs) and the angiopoietins (Angpt). We present examples showing not only how these molecules help generate and maintain healthy glomeruli but also how they drive disease when their expression is dysregulated. Finally, we review how manipulating VEGF and Angpt signalling may be used to treat glomerular disease.
Collapse
|
40
|
Abstract
A multifunctional microRNA, miR-155, has been recently recognized as an important modulator of numerous biological processes. In our previous in vitro studies, miR-155 was identified as a potential regulator of the endothelial morphogenesis. The present study demonstrates that in vivo inhibition of miR-155 supports cerebral vasculature after experimental stroke. Intravenous injections of a specific miR-155 inhibitor were initiated at 48 h after mouse distal middle cerebral artery occlusion (dMCAO). Microvasculature in peri-infarct area, infarct size, and animal functional recovery were assessed at 1, 2, and 3 weeks after dMCAO. Using in vivo two-photon microscopy, we detected improved blood flow and microvascular integrity in the peri-infarct area of miR-155 inhibitor-injected mice. Electron microscopy revealed that, in contrast to the control group, these animals demonstrated well preserved capillary tight junctions (TJs). Western blot analysis data indicate that improved TJ integrity in the inhibitor-injected animals could be associated with stabilization of the TJ protein ZO-1 and mediated by the miR-155 target protein Rheb. MRI analysis showed significant (34%) reduction of infarct size in miR-155 inhibitor-injected animals at 21 d after dMCAO. Reduced brain injury was confirmed by electron microscopy demonstrating decreased neuronal damage in the peri-infarct area of stroke. Preservation of brain tissue was reflected in efficient functional recovery of inhibitor-injected animals. Based on our findings, we propose that in vivo miR-155 inhibition after ischemia supports brain microvasculature, reduces brain tissue damage, and improves the animal functional recovery. Significance statement: In the present study, we investigated an effect of the in vivo inhibition of a microRNA, miR-155, on brain recovery after experimental cerebral ischemia. To our knowledge, this is the first report describing the efficiency of intravenous anti-miRNA injections in a mouse model of ischemic stroke. The role of miRNAs in poststroke revascularization has been unexplored and in vivo regulation of miRNAs during the subacute phase of stroke has not yet been proposed. Our investigation introduces a new and unexplored approach to cerebral regeneration: regulation of poststroke angiogenesis and recovery through direct modulation of specific miRNA activity. We expect that our findings will lead to the development of novel strategies for regulating neurorestorative processes in the postischemic brain.
Collapse
|
41
|
Chang JYH, Stamer WD, Bertrand J, Read AT, Marando CM, Ethier CR, Overby DR. Role of nitric oxide in murine conventional outflow physiology. Am J Physiol Cell Physiol 2015; 309:C205-14. [PMID: 26040898 DOI: 10.1152/ajpcell.00347.2014] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 05/30/2015] [Indexed: 11/22/2022]
Abstract
Elevated intraocular pressure (IOP) is the main risk factor for glaucoma. Exogenous nitric oxide (NO) decreases IOP by increasing outflow facility, but whether endogenous NO production contributes to the physiological regulation of outflow facility is unclear. Outflow facility was measured by pressure-controlled perfusion in ex vivo eyes from C57BL/6 wild-type (WT) or transgenic mice expressing human endothelial NO synthase (eNOS) fused to green fluorescent protein (GFP) superimposed on the endogenously expressed murine eNOS (eNOS-GFPtg). In WT mice, exogenous NO delivered by 100 μM S-nitroso-N-acetylpenicillamine (SNAP) increased outflow facility by 62 ± 28% (SD) relative to control eyes perfused with the inactive SNAP analog N-acetyl-d-penicillamine (NAP; n = 5, P = 0.016). In contrast, in eyes from eNOS-GFPtg mice, SNAP had no effect on outflow facility relative to NAP (-9 ± 4%, P = 0.40). In WT mice, the nonselective NOS inhibitor N(G)-nitro-l-arginine methyl ester (l-NAME, 10 μM) decreased outflow facility by 36 ± 13% (n = 5 each, P = 0.012), but 100 μM l-NAME had no detectable effect on outflow facility (-16 ± 5%, P = 0.22). An eNOS-selective inhibitor (cavtratin, 50 μM) decreased outflow facility by 19 ± 12% in WT (P = 0.011) and 39 ± 25% in eNOS-GFPtg (P = 0.014) mice. In the conventional outflow pathway of eNOS-GFPtg mice, eNOS-GFP expression was localized to endothelial cells lining Schlemm's canal and the downstream vessels, with no apparent expression in the trabecular meshwork. These results suggest that endogenous NO production by eNOS within endothelial cells of Schlemm's canal or downstream vessels contributes to the physiological regulation of aqueous humor outflow facility in mice, representing a viable strategy to more successfully lower IOP in glaucoma.
Collapse
Affiliation(s)
- Jason Y H Chang
- Department of Bioengineering, Imperial College London, London, United Kingdom; Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina
| | - W Daniel Stamer
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina
| | - Jacques Bertrand
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - A Thomas Read
- Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, Ontario, Canada; and
| | - Catherine M Marando
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - C Ross Ethier
- Department of Bioengineering, Imperial College London, London, United Kingdom; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia
| | - Darryl R Overby
- Department of Bioengineering, Imperial College London, London, United Kingdom;
| |
Collapse
|
42
|
Comparative proteomics of cerebrospinal fluid reveals a predictive model for differential diagnosis of pneumococcal, meningococcal, and enteroviral meningitis, and novel putative therapeutic targets. BMC Genomics 2015; 16 Suppl 5:S11. [PMID: 26040285 PMCID: PMC4460676 DOI: 10.1186/1471-2164-16-s5-s11] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Meningitis is the inflammation of the meninges in response to infection or chemical agents. While aseptic meningitis, most frequently caused by enteroviruses, is usually benign with a self-limiting course, bacterial meningitis remains associated with high morbidity and mortality rates, despite advances in antimicrobial therapy and intensive care. Fast and accurate differential diagnosis is crucial for assertive choice of the appropriate therapeutic approach for each form of meningitis. METHODS We used 2D-PAGE and mass spectrometry to identify the cerebrospinal fluid proteome specifically related to the host response to pneumococcal, meningococcal, and enteroviral meningitis. The disease-specific proteome signatures were inspected by pathway analysis. RESULTS Unique cerebrospinal fluid proteome signatures were found to the three aetiological forms of meningitis investigated, and a qualitative predictive model with four protein markers was developed for the differential diagnosis of these diseases. Nevertheless, pathway analysis of the disease-specific proteomes unveiled that Kallikrein-kinin system may play a crucial role in the pathophysiological mechanisms leading to brain damage in bacterial meningitis. Proteins taking part in this cellular process are proposed as putative targets to novel adjunctive therapies. CONCLUSIONS Comparative proteomics of cerebrospinal fluid disclosed candidate biomarkers, which were combined in a qualitative and sequential predictive model with potential to improve the differential diagnosis of pneumococcal, meningococcal and enteroviral meningitis. Moreover, we present the first evidence of the possible implication of Kallikrein-kinin system in the pathophysiology of bacterial meningitis.
Collapse
|
43
|
Kim HY, Kim JW. Effect of Nitric Oxide on the Permeability of Trabecular Meshwork Cell Monolayer. JOURNAL OF THE KOREAN OPHTHALMOLOGICAL SOCIETY 2015. [DOI: 10.3341/jkos.2015.56.5.771] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- Hyun Yeon Kim
- Department of Ophthalmology, Catholic University of Daegu School of Medicine, Daegu, Korea
| | - Jae Woo Kim
- Department of Ophthalmology, Catholic University of Daegu School of Medicine, Daegu, Korea
| |
Collapse
|
44
|
Lucas R, Fulton D, Caldwell RW, Romero MJ. Arginase in the vascular endothelium: friend or foe? Front Immunol 2014; 5:589. [PMID: 25452758 PMCID: PMC4233309 DOI: 10.3389/fimmu.2014.00589] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 11/04/2014] [Indexed: 02/05/2023] Open
Affiliation(s)
- Rudolf Lucas
- Vascular Biology Center, Medical College of Georgia, Georgia Regents University , Augusta, GA , USA ; Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University , Augusta, GA , USA
| | - David Fulton
- Vascular Biology Center, Medical College of Georgia, Georgia Regents University , Augusta, GA , USA ; Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University , Augusta, GA , USA
| | - Robert William Caldwell
- Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University , Augusta, GA , USA
| | - Maritza J Romero
- Vascular Biology Center, Medical College of Georgia, Georgia Regents University , Augusta, GA , USA ; Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University , Augusta, GA , USA
| |
Collapse
|
45
|
Ashpole NE, Overby DR, Ethier CR, Stamer WD. Shear stress-triggered nitric oxide release from Schlemm's canal cells. Invest Ophthalmol Vis Sci 2014; 55:8067-76. [PMID: 25395486 DOI: 10.1167/iovs.14-14722] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Endothelial nitric oxide (NO) synthase is regulated by shear stress. At elevated intraocular pressures when the Schlemm's canal (SC) begins to collapse, shear stress is comparable with that in large arteries. We investigated the relationship between NO production and shear stress in cultured human SC cells. METHODS Schlemm's canal endothelial cells isolated from three normal and two glaucomatous human donors were seeded into Ibidi flow chambers at confluence, cultured for 7 days, and subjected to steady shear stress (0.1 or 10 dynes/cm(2)) for 6, 24, or 168 hours. Cell alignment with flow direction was monitored, and NO production was measured using 4-amino-5-methylamino-2',7'-difluorofluorescein (DAF-FM) and Griess reagents. Human trabecular meshwork (TM) and umbilical vein endothelial cells (HUVECs) were used as controls. RESULTS Normal SC strains aligned with the direction of flow by 7 days. Comparing 0.1 vs. 10 dynes/cm(2), NO levels increased by 82% at 24 hours and 8-fold after 7 days by DAF-FM, and similar results were obtained with Griess reagent. Shear responses by SC cells at 24 hours were comparable with HUVECs, and greater than TM cells, which appeared shear-insensitive. Nitric oxide production by SC cells was detectable as early as 6 hours and was inhibited by 100 μM nitro-L-arginine methyl ester. Two glaucomatous SC cell strains were either unresponsive or lifted from the plate in the face of shear. CONCLUSIONS Shear stress triggers NO production in human SC cells, similar to other vascular endothelia. Increased shear stress and NO production during SC collapse at elevated intraocular pressures may in part mediate IOP homeostasis.
Collapse
Affiliation(s)
- Nicole E Ashpole
- Biomedical Engineering, Duke University, Durham, North Carolina, United States
| | - Darryl R Overby
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - C Ross Ethier
- Department of Bioengineering, Imperial College London, London, United Kingdom Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, United States
| | - W Daniel Stamer
- Ophthalmology Department, Duke University, Durham, North Carolina, United States
| |
Collapse
|
46
|
Wu F, Szczepaniak WS, Shiva S, Liu H, Wang Y, Wang L, Wang Y, Kelley EE, Chen AF, Gladwin MT, McVerry BJ. Nox2-dependent glutathionylation of endothelial NOS leads to uncoupled superoxide production and endothelial barrier dysfunction in acute lung injury. Am J Physiol Lung Cell Mol Physiol 2014; 307:L987-97. [PMID: 25326583 DOI: 10.1152/ajplung.00063.2014] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Microvascular barrier integrity is dependent on bioavailable nitric oxide (NO) produced locally by endothelial NO synthase (eNOS). Under conditions of limited substrate or cofactor availability or by enzymatic modification, eNOS may become uncoupled, producing superoxide in lieu of NO. This study was designed to investigate how eNOS-dependent superoxide production contributes to endothelial barrier dysfunction in inflammatory lung injury and its regulation. C57BL/6J mice were challenged with intratracheal LPS. Bronchoalveolar lavage fluid was analyzed for protein accumulation, and lung tissue homogenate was assayed for endothelial NOS content and function. Human lung microvascular endothelial cell (HLMVEC) monolayers were exposed to LPS in vitro, and barrier integrity and superoxide production were measured. Biopterin species were quantified, and coimmunoprecipitation (Co-IP) assays were performed to identify protein interactions with eNOS that putatively drive uncoupling. Mice exposed to LPS demonstrated eNOS-dependent increased alveolar permeability without evidence for altered canonical NO signaling. LPS-induced superoxide production and permeability in HLMVEC were inhibited by the NOS inhibitor nitro-l-arginine methyl ester, eNOS-targeted siRNA, the eNOS cofactor tetrahydrobiopterin, and superoxide dismutase. Co-IP indicated that LPS stimulated the association of eNOS with NADPH oxidase 2 (Nox2), which correlated with augmented eNOS S-glutathionylation both in vitro and in vivo. In vitro, Nox2-specific inhibition prevented LPS-induced eNOS modification and increases in both superoxide production and permeability. These data indicate that eNOS uncoupling contributes to superoxide production and barrier dysfunction in the lung microvasculature after exposure to LPS. Furthermore, the results implicate Nox2-mediated eNOS-S-glutathionylation as a mechanism underlying LPS-induced eNOS uncoupling in the lung microvasculature.
Collapse
Affiliation(s)
- Feng Wu
- University of Pittsburgh School of Medicine Department of Medicine, Division of Pulmonary Allergy and Critical Care Medicine, Pittsburgh, Pennsylvania
| | - William S Szczepaniak
- University of Pittsburgh School of Medicine Department of Medicine, Division of Pulmonary Allergy and Critical Care Medicine, Pittsburgh, Pennsylvania
| | - Sruti Shiva
- University of Pittsburgh Vascular Medicine Institute, Pittsburgh, Pennsylvania; University of Pittsburgh School of Medicine Department of Pharmacology, Pittsburgh, Pennsylvania
| | - Huanbo Liu
- University of Pittsburgh School of Medicine Department of Surgery, Pittsburgh, Pennsylvania
| | - Yinna Wang
- University of Pittsburgh Vascular Medicine Institute, Pittsburgh, Pennsylvania
| | - Ling Wang
- University of Pittsburgh Vascular Medicine Institute, Pittsburgh, Pennsylvania
| | - Ying Wang
- University of Pittsburgh School of Medicine Department of Medicine, Division of Pulmonary Allergy and Critical Care Medicine, Pittsburgh, Pennsylvania
| | - Eric E Kelley
- University of Pittsburgh Vascular Medicine Institute, Pittsburgh, Pennsylvania; University of Pittsburgh School of Medicine Department of Anesthesiology, Pittsburgh, Pennsylvania
| | - Alex F Chen
- University of Pittsburgh Vascular Medicine Institute, Pittsburgh, Pennsylvania; University of Pittsburgh School of Medicine Department of Surgery, Pittsburgh, Pennsylvania
| | - Mark T Gladwin
- University of Pittsburgh School of Medicine Department of Medicine, Division of Pulmonary Allergy and Critical Care Medicine, Pittsburgh, Pennsylvania; University of Pittsburgh Vascular Medicine Institute, Pittsburgh, Pennsylvania
| | - Bryan J McVerry
- University of Pittsburgh School of Medicine Department of Medicine, Division of Pulmonary Allergy and Critical Care Medicine, Pittsburgh, Pennsylvania; University of Pittsburgh Vascular Medicine Institute, Pittsburgh, Pennsylvania;
| |
Collapse
|
47
|
Chen F, Kumar S, Yu Y, Aggarwal S, Gross C, Wang Y, Chakraborty T, Verin AD, Catravas JD, Lucas R, Black SM, Fulton DJR. PKC-dependent phosphorylation of eNOS at T495 regulates eNOS coupling and endothelial barrier function in response to G+ -toxins. PLoS One 2014; 9:e99823. [PMID: 25020117 PMCID: PMC4096401 DOI: 10.1371/journal.pone.0099823] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 05/19/2014] [Indexed: 11/30/2022] Open
Abstract
Gram positive (G+) infections make up ∼50% of all acute lung injury cases which are characterized by extensive permeability edema secondary to disruption of endothelial cell (EC) barrier integrity. A primary cause of increased permeability are cholesterol-dependent cytolysins (CDCs) of G+-bacteria, such as pneumolysin (PLY) and listeriolysin-O (LLO) which create plasma membrane pores, promoting Ca2+-influx and activation of PKCα. In human lung microvascular endothelial cells (HLMVEC), pretreatment with the nitric oxide synthase (NOS) inhibitor, ETU reduced the ability of LLO to increase microvascular cell permeability suggesting an endothelial nitric oxide synthase (eNOS)-dependent mechanism. LLO stimulated superoxide production from HLMVEC and this was prevented by silencing PKCα or NOS inhibition suggesting a link between these pathways. Both LLO and PLY stimulated eNOS T495 phosphorylation in a PKC-dependent manner. Expression of a phosphomimetic T495D eNOS (human isoform) resulted in increased superoxide and diminished nitric oxide (NO) production. Transduction of HLMVEC with an active form of PKCα resulted in the robust phosphorylation of T495 and increased peroxynitrite production, indicative of eNOS uncoupling. To determine the mechanisms underlying eNOS uncoupling, HLMVEC were stimulated with LLO and the amount of hsp90 and caveolin-1 bound to eNOS determined. LLO stimulated the dissociation of hsp90, and in particular, caveolin-1 from eNOS. Both hsp90 and caveolin-1 have been shown to influence eNOS uncoupling and a peptide mimicking the scaffolding domain of caveolin-1 blocked the ability of PKCα to stimulate eNOS-derived superoxide. Collectively, these results suggest that the G+ pore-forming toxins promote increased EC permeability via activation of PKCα, phosphorylation of eNOS-T495, loss of hsp90 and caveolin-1 binding which collectively promote eNOS uncoupling and the production of barrier disruptive superoxide.
Collapse
Affiliation(s)
- Feng Chen
- Department of Forensic Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
- Vascular Biology Center Medical College of Georgia at Georgia Regents University, Augusta, Georgia, United States of America
| | - Sanjiv Kumar
- Vascular Biology Center Medical College of Georgia at Georgia Regents University, Augusta, Georgia, United States of America
| | - Yanfang Yu
- Vascular Biology Center Medical College of Georgia at Georgia Regents University, Augusta, Georgia, United States of America
| | - Saurabh Aggarwal
- Vascular Biology Center Medical College of Georgia at Georgia Regents University, Augusta, Georgia, United States of America
| | - Christine Gross
- Vascular Biology Center Medical College of Georgia at Georgia Regents University, Augusta, Georgia, United States of America
| | - Yusi Wang
- Vascular Biology Center Medical College of Georgia at Georgia Regents University, Augusta, Georgia, United States of America
| | - Trinad Chakraborty
- Institute for Medical Microbiology, Justus Liebig University, Giessen, Germany
| | - Alexander D. Verin
- Vascular Biology Center Medical College of Georgia at Georgia Regents University, Augusta, Georgia, United States of America
| | - John D. Catravas
- Old Dominion University, Norfolk, Virginia, United States of America
| | - Rudolf Lucas
- Vascular Biology Center Medical College of Georgia at Georgia Regents University, Augusta, Georgia, United States of America
- Department of Pharmacology, Medical College of Georgia at Georgia Regents University, Augusta, Georgia, United States of America
| | - Stephen M. Black
- Vascular Biology Center Medical College of Georgia at Georgia Regents University, Augusta, Georgia, United States of America
| | - David J. R. Fulton
- Vascular Biology Center Medical College of Georgia at Georgia Regents University, Augusta, Georgia, United States of America
- Department of Pharmacology, Medical College of Georgia at Georgia Regents University, Augusta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
48
|
Aggarwal S, Gross CM, Kumar S, Dimitropoulou C, Sharma S, Gorshkov BA, Sridhar S, Lu Q, Bogatcheva NV, Jezierska-Drutel AJ, Lucas R, Verin AD, Catravas JD, Black SM. Dimethylarginine dimethylaminohydrolase II overexpression attenuates LPS-mediated lung leak in acute lung injury. Am J Respir Cell Mol Biol 2014; 50:614-25. [PMID: 24134589 DOI: 10.1165/rcmb.2013-0193oc] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Acute lung injury (ALI) is a severe hypoxemic respiratory insufficiency associated with lung leak, diffuse alveolar damage, inflammation, and loss of lung function. Decreased dimethylaminohydrolase (DDAH) activity and increases in asymmetric dimethylarginine (ADMA), together with exaggerated oxidative/nitrative stress, contributes to the development of ALI in mice exposed to LPS. Whether restoring DDAH function and suppressing ADMA levels can effectively ameliorate vascular hyperpermeability and lung injury in ALI is unknown, and was the focus of this study. In human lung microvascular endothelial cells, DDAH II overexpression prevented the LPS-dependent increase in ADMA, superoxide, peroxynitrite, and protein nitration. DDAH II also attenuated the endothelial barrier disruption associated with LPS exposure. Similarly, in vivo, we demonstrated that the targeted overexpression of DDAH II in the pulmonary vasculature significantly inhibited the accumulation of ADMA and the subsequent increase in oxidative/nitrative stress in the lungs of mice exposed to LPS. In addition, augmenting pulmonary DDAH II activity before LPS exposure reduced lung vascular leak and lung injury and restored lung function when DDAH activity was increased after injury. Together, these data suggest that enhancing DDAH II activity may prove a useful adjuvant therapy to treat patients with ALI.
Collapse
Affiliation(s)
- Saurabh Aggarwal
- Pulmonary Disease Program, Vascular Biology Center, Georgia Regents University, Augusta, Georgia
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Barvitenko NN, Aslam M, Filosa J, Matteucci E, Nikinmaa M, Pantaleo A, Saldanha C, Baskurt OK. Tissue oxygen demand in regulation of the behavior of the cells in the vasculature. Microcirculation 2014; 20:484-501. [PMID: 23441854 DOI: 10.1111/micc.12052] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Accepted: 02/19/2013] [Indexed: 12/20/2022]
Abstract
The control of arteriolar diameters in microvasculature has been in the focus of studies on mechanisms matching oxygen demand and supply at the tissue level. Functionally, important vascular elements include EC, VSMC, and RBC. Integration of these different cell types into functional units aimed at matching tissue oxygen supply with tissue oxygen demand is only achieved when all these cells can respond to the signals of tissue oxygen demand. Many vasoactive agents that serve as signals of tissue oxygen demand have their receptors on all these types of cells (VSMC, EC, and RBC) implying that there can be a coordinated regulation of their behavior by the tissue oxygen demand. Such functions of RBC as oxygen carrying by Hb, rheology, and release of vasoactive agents are considered. Several common extra- and intracellular signaling pathways that link tissue oxygen demand with control of VSMC contractility, EC permeability, and RBC functioning are discussed.
Collapse
|
50
|
Meredith ME, Qu ZC, May JM. Ascorbate reverses high glucose- and RAGE-induced leak of the endothelial permeability barrier. Biochem Biophys Res Commun 2014; 445:30-5. [PMID: 24472555 DOI: 10.1016/j.bbrc.2014.01.078] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 01/20/2014] [Indexed: 02/07/2023]
Abstract
High glucose concentrations due to diabetes increase leakage of plasma constituents across the endothelial permeability barrier. We sought to determine whether vitamin C, or ascorbic acid (ascorbate), could reverse such high glucose-induced increases in endothelial barrier permeability. Human umbilical vein endothelial cells and two brain endothelial cell lines cultured at 25 mM glucose showed increases in endothelial barrier permeability to radiolabeled inulin compared to cells cultured at 5mM glucose. Acute loading of the cells for 30-60 min with ascorbate before the permeability assay prevented the high glucose-induced increase in permeability and decreased basal permeability at 5mM glucose. High glucose-induced barrier leakage was mediated largely by activation of the receptor for advanced glycation end products (RAGE), since it was prevented by RAGE blockade and mimicked by RAGE ligands. Intracellular ascorbate completely prevented RAGE ligand-induced increases in barrier permeability. The high glucose-induced increase in endothelial barrier permeability was also acutely decreased by several cell-penetrant antioxidants, suggesting that at least part of the ascorbate effect could be due to its ability to act as an antioxidant.
Collapse
Affiliation(s)
- M Elizabeth Meredith
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232-0475, United States
| | - Zhi-Chao Qu
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232-0475, United States
| | - James M May
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232-0475, United States; Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232-0475, United States.
| |
Collapse
|