1
|
Liu W, Man X, Wang Y, Wang Q, Wang Z, Qi J, Qin Q, Han B, Sun J. Tirofiban mediates neuroprotective effects in acute ischemic stroke by reducing inflammatory response. Neuroscience 2024; 555:32-40. [PMID: 39025399 DOI: 10.1016/j.neuroscience.2024.07.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 07/03/2024] [Accepted: 07/10/2024] [Indexed: 07/20/2024]
Abstract
Growing evidence suggests that neuroinflammation is a critical driver of the development, worsening, and cell death observed in acute ischemic stroke (AIS). While prior research has demonstrated that tirofiban enhances functional recovery in AIS patients by suppressing platelet aggregation, its impact and underlying mechanisms in AIS-related neuroinflammation remain elusive. The current study established an AIS mouse model employing photochemical techniques and assessed neurological function and brain infarct size using the modified neurological severity scale (mNSS) and 2,3,5-Triphenyltetrazolium chloride (TTC) staining, respectively. Tirofiban significantly reduced the volume of cerebral infarction in AIS mice, accompanied by an enhancement in their neurological functions. Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assays along with experiments assessing oxidative stress showed that tirofiban mitigated oxidative damage and apoptosis in the ischemic penumbra post-AIS. Additionally, DNA microarray analysis revealed alterations in gene expression patterns in the ischemic penumbra after tirofiban treatment. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis showed that most gene-level downregulated signaling pathways were closely related to the inflammatory response. Moreover, the protein microarray analysis revealed that tirofiban diminished the expression levels of inflammatory cytokines, such as interleukin-1 (IL-1), IL-6, and tumor necrosis factor-alpha, in the ischemic penumbra. Additionally, immunofluorescence staining showed that tirofiban regulated inflammatory responses by altering the state and phenotype of microglia. In conclusion, this study suggests that tirofiban reduces inflammatory response by regulating microglial state and phenotype and lowering the levels of inflammatory factors, providing neuroprotection in acute ischemic stroke.
Collapse
Affiliation(s)
- Wei Liu
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266003, China
| | - Xu Man
- Department of Integrative Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266003, China
| | - Yongbin Wang
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266003, China
| | - Qingqing Wang
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266003, China
| | - Zhiyuan Wang
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266003, China
| | - Jianjiao Qi
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266003, China
| | - Qiaoji Qin
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266003, China
| | - Ban Han
- Department of Neurology, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266003, China.
| | - Jinping Sun
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266003, China.
| |
Collapse
|
2
|
Keller M, Rohlf K, Glotzbach A, Leonhardt G, Lüke S, Derksen K, Demirci Ö, Göçener D, AlWahsh M, Lambert J, Lindskog C, Schmidt M, Brenner W, Baumann M, Zent E, Zischinsky ML, Hellwig B, Madjar K, Rahnenführer J, Overbeck N, Reinders J, Cadenas C, Hengstler JG, Edlund K, Marchan R. Inhibiting the glycerophosphodiesterase EDI3 in ER-HER2+ breast cancer cells resistant to HER2-targeted therapy reduces viability and tumour growth. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2023; 42:25. [PMID: 36670508 PMCID: PMC9854078 DOI: 10.1186/s13046-022-02578-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 12/20/2022] [Indexed: 01/22/2023]
Abstract
BACKGROUND Intrinsic or acquired resistance to HER2-targeted therapy is often a problem when small molecule tyrosine kinase inhibitors or antibodies are used to treat patients with HER2 positive breast cancer. Therefore, the identification of new targets and therapies for this patient group is warranted. Activated choline metabolism, characterized by elevated levels of choline-containing compounds, has been previously reported in breast cancer. The glycerophosphodiesterase EDI3 (GPCPD1), which hydrolyses glycerophosphocholine to choline and glycerol-3-phosphate, directly influences choline and phospholipid metabolism, and has been linked to cancer-relevant phenotypes in vitro. While the importance of choline metabolism has been addressed in breast cancer, the role of EDI3 in this cancer type has not been explored. METHODS EDI3 mRNA and protein expression in human breast cancer tissue were investigated using publicly-available Affymetrix gene expression microarray datasets (n = 540) and with immunohistochemistry on a tissue microarray (n = 265), respectively. A panel of breast cancer cell lines of different molecular subtypes were used to investigate expression and activity of EDI3 in vitro. To determine whether EDI3 expression is regulated by HER2 signalling, the effect of pharmacological inhibition and siRNA silencing of HER2, as well as the influence of inhibiting key components of signalling cascades downstream of HER2 were studied. Finally, the influence of silencing and pharmacologically inhibiting EDI3 on viability was investigated in vitro and on tumour growth in vivo. RESULTS In the present study, we show that EDI3 expression is highest in ER-HER2 + human breast tumours, and both expression and activity were also highest in ER-HER2 + breast cancer cell lines. Silencing HER2 using siRNA, as well as inhibiting HER2 signalling with lapatinib decreased EDI3 expression. Pathways downstream of PI3K/Akt/mTOR and GSK3β, and transcription factors, including HIF1α, CREB and STAT3 were identified as relevant in regulating EDI3 expression. Silencing EDI3 preferentially decreased cell viability in the ER-HER2 + cells. Furthermore, silencing or pharmacologically inhibiting EDI3 using dipyridamole in ER-HER2 + cells resistant to HER2-targeted therapy decreased cell viability in vitro and tumour growth in vivo. CONCLUSIONS Our results indicate that EDI3 may be a potential novel therapeutic target in patients with HER2-targeted therapy-resistant ER-HER2 + breast cancer that should be further explored.
Collapse
Affiliation(s)
- Magdalena Keller
- grid.419241.b0000 0001 2285 956XLeibniz Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany
| | - Katharina Rohlf
- grid.419241.b0000 0001 2285 956XLeibniz Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany
| | - Annika Glotzbach
- grid.419241.b0000 0001 2285 956XLeibniz Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany
| | - Gregor Leonhardt
- grid.419241.b0000 0001 2285 956XLeibniz Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany
| | - Simon Lüke
- grid.419241.b0000 0001 2285 956XLeibniz Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany
| | - Katharina Derksen
- grid.419241.b0000 0001 2285 956XLeibniz Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany
| | - Özlem Demirci
- grid.419241.b0000 0001 2285 956XLeibniz Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany
| | - Defne Göçener
- grid.419241.b0000 0001 2285 956XLeibniz Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany
| | - Mohammad AlWahsh
- grid.419243.90000 0004 0492 9407Leibniz Institut Für Analytische Wissenschaften - ISAS E.V, Dortmund, Germany ,grid.411778.c0000 0001 2162 1728Institute of Pathology and Medical Research Center (ZMF), University Medical Center Mannheim, Heidelberg University, Mannheim, Germany ,grid.443348.c0000 0001 0244 5415Department of Pharmacy, AlZaytoonah University of Jordan, Amman, Jordan
| | - Jörg Lambert
- grid.419243.90000 0004 0492 9407Leibniz Institut Für Analytische Wissenschaften - ISAS E.V, Dortmund, Germany
| | - Cecilia Lindskog
- grid.8993.b0000 0004 1936 9457Department of Immunology Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Marcus Schmidt
- grid.410607.4Department of Obstetrics and Gynecology, University Medical Center Mainz, Mainz, Germany
| | - Walburgis Brenner
- grid.410607.4Department of Obstetrics and Gynecology, University Medical Center Mainz, Mainz, Germany
| | - Matthias Baumann
- grid.505582.fPharmacology Department, Lead Discovery Center, Dortmund, Germany
| | - Eldar Zent
- grid.505582.fPharmacology Department, Lead Discovery Center, Dortmund, Germany
| | - Mia-Lisa Zischinsky
- grid.505582.fPharmacology Department, Lead Discovery Center, Dortmund, Germany
| | - Birte Hellwig
- grid.5675.10000 0001 0416 9637Department of Statistics, TU Dortmund University, Dortmund, Germany
| | - Katrin Madjar
- grid.5675.10000 0001 0416 9637Department of Statistics, TU Dortmund University, Dortmund, Germany
| | - Jörg Rahnenführer
- grid.5675.10000 0001 0416 9637Department of Statistics, TU Dortmund University, Dortmund, Germany
| | - Nina Overbeck
- grid.419241.b0000 0001 2285 956XLeibniz Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany
| | - Jörg Reinders
- grid.419241.b0000 0001 2285 956XLeibniz Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany
| | - Cristina Cadenas
- grid.419241.b0000 0001 2285 956XLeibniz Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany
| | - Jan G. Hengstler
- grid.419241.b0000 0001 2285 956XLeibniz Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany
| | - Karolina Edlund
- grid.419241.b0000 0001 2285 956XLeibniz Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany
| | - Rosemarie Marchan
- grid.419241.b0000 0001 2285 956XLeibniz Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany
| |
Collapse
|
3
|
Degjoni A, Campolo F, Stefanini L, Venneri MA. The NO/cGMP/PKG pathway in platelets: The therapeutic potential of PDE5 inhibitors in platelet disorders. J Thromb Haemost 2022; 20:2465-2474. [PMID: 35950928 PMCID: PMC9805178 DOI: 10.1111/jth.15844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 08/01/2022] [Accepted: 08/08/2022] [Indexed: 01/09/2023]
Abstract
Platelets are the "guardians" of the blood circulatory system. At sites of vessel injury, they ensure hemostasis and promote immunity and vessel repair. However, their uncontrolled activation is one of the main drivers of thrombosis. To keep circulating platelets in a quiescent state, the endothelium releases platelet antagonists including nitric oxide (NO) that acts by stimulating the intracellular receptor guanylyl cyclase (GC). The latter produces the second messenger cyclic guanosine-3',5'-monophosphate (cGMP) that inhibits platelet activation by stimulating protein kinase G, which phosphorylates hundreds of intracellular targets. Intracellular cGMP pools are tightly regulated by a fine balance between GC and phosphodiesterases (PDEs) that are responsible for the hydrolysis of cyclic nucleotides. Phosphodiesterase type 5 (PDE5) is a cGMP-specific PDE, broadly expressed in most tissues in humans and rodents. In clinical practice, PDE5 inhibitors (PDE5i) are used as first-line therapy for erectile dysfunction, pulmonary artery hypertension, and lower urinary tract symptoms. However, several studies have shown that PDE5i may ameliorate the outcome of various other conditions, like heart failure and stroke. Interestingly, NO donors and cGMP analogs increase the capacity of anti-platelet drugs targeting the purinergic receptor type Y, subtype 12 (P2Y12) receptor to block platelet aggregation, and preclinical studies have shown that PDE5i inhibits platelet functions. This review summarizes the molecular mechanisms underlying the effect of PDE5i on platelet activation and aggregation focusing on the therapeutic potential of PDE5i in platelet disorders, and the outcomes of a combined therapy with PDE5i and NO donors to inhibit platelet activation.
Collapse
Affiliation(s)
- Anisa Degjoni
- Department of Experimental MedicineSapienza University of RomeRomeItaly
| | - Federica Campolo
- Department of Experimental MedicineSapienza University of RomeRomeItaly
| | - Lucia Stefanini
- Department of Translational and Precision MedicineSapienza University of RomeRomeItaly
| | - Mary Anna Venneri
- Department of Experimental MedicineSapienza University of RomeRomeItaly
| |
Collapse
|
4
|
Chen YF, Stampley JE, Irving BA, Dugas TR. Chronic Nucleoside Reverse Transcriptase Inhibitors Disrupt Mitochondrial Homeostasis and Promote Premature Endothelial Senescence. Toxicol Sci 2020; 172:445-456. [PMID: 31545371 DOI: 10.1093/toxsci/kfz203] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Combination antiretroviral therapy (cART) has improved the life expectancy of HIV patients, thus increasing the number of people living with HIV (PLWH). However, cardiovascular diseases (CVD) are now one of the most prevalent causes of death among PLWH. Nucleoside reverse transcriptase inhibitors (NRTIs) are the backbone of cART, and the emtricitabine (FTC) and tenofovir disoproxil fumarate (TDF) coformulation is commonly used. In prior studies, acute NRTI treatment-induced endothelial dysfunction, increased reactive oxygen species production, and mitophagic activity, suggesting that mitochondrial dysfunction may be critical to NRTI-induced endothelial dysfunction. Mitochondrial dysfunction plays a causal role in endothelial senescence, whereas premature endothelial senescence can promote the development of CVD. We hypothesize that for chronic NRTI treatment, a disruption in mitochondrial homeostasis leads to premature endothelial senescence and predisposes PLWH to CVD. We used human aortic endothelial cells (HAEC) and HIV-1 transgenic (Tg26) mice to test the interrelationship between mitochondrial and vascular dysfunction after chronic NRTI treatment in vitro and in vivo. Mitochondrial DNA copy number was decreased in late-passage HAEC treated with NRTIs, and senescence-associated β-galactosidase accumulation was elevated. In late-passage HAEC, NRTIs decreased the activity of Parkin-mediated mitophagy. In Tg26 mice treated with FTC, plasma nitrite levels were decreased. Endothelium-dependent vasodilation in NRTI-treated Tg26 mice was also reduced. Our work suggests that long-term use of NRTI may disrupt mitochondrial homeostasis, induce premature endothelial senescence, and impair vascular function.
Collapse
Affiliation(s)
- Yi-Fan Chen
- Comparative Biomedical Sciences, LSU School of Veterinary Medicine, Baton Rouge, Louisiana 70808
| | - James E Stampley
- College of Human Sciences and Education, LSU School of Kinesiology, Baton Rouge, Louisiana 70803
| | - Brian A Irving
- College of Human Sciences and Education, LSU School of Kinesiology, Baton Rouge, Louisiana 70803.,Pennington Biomedical Research Center, Baton Rouge, Louisiana, 70808
| | - Tammy R Dugas
- Comparative Biomedical Sciences, LSU School of Veterinary Medicine, Baton Rouge, Louisiana 70808
| |
Collapse
|
5
|
Flora GD, Nayak MK. A Brief Review of Cardiovascular Diseases, Associated Risk Factors and Current Treatment Regimes. Curr Pharm Des 2020; 25:4063-4084. [PMID: 31553287 DOI: 10.2174/1381612825666190925163827] [Citation(s) in RCA: 191] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 09/16/2019] [Indexed: 12/22/2022]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of premature death and disability in humans and their incidence is on the rise globally. Given their substantial contribution towards the escalating costs of health care, CVDs also generate a high socio-economic burden in the general population. The underlying pathogenesis and progression associated with nearly all CVDs are predominantly of atherosclerotic origin that leads to the development of coronary artery disease, cerebrovascular disease, venous thromboembolism and, peripheral vascular disease, subsequently causing myocardial infarction, cardiac arrhythmias or stroke. The aetiological risk factors leading to the onset of CVDs are well recognized and include hyperlipidaemia, hypertension, diabetes, obesity, smoking and, lack of physical activity. They collectively represent more than 90% of the CVD risks in all epidemiological studies. Despite high fatality rate of CVDs, the identification and careful prevention of the underlying risk factors can significantly reduce the global epidemic of CVDs. Beside making favorable lifestyle modifications, primary regimes for the prevention and treatment of CVDs include lipid-lowering drugs, antihypertensives, antiplatelet and anticoagulation therapies. Despite their effectiveness, significant gaps in the treatment of CVDs remain. In this review, we discuss the epidemiology and pathology of the major CVDs that are prevalent globally. We also determine the contribution of well-recognized risk factors towards the development of CVDs and the prevention strategies. In the end, therapies for the control and treatment of CVDs are discussed.
Collapse
Affiliation(s)
- Gagan D Flora
- Department of Internal Medicine, University of Iowa, Iowa City, IA 52242, United States
| | - Manasa K Nayak
- Department of Internal Medicine, University of Iowa, Iowa City, IA 52242, United States
| |
Collapse
|
6
|
Kim JW, Lee JB, Lee SH. Effect and Mechanism of Phosphodiesterase Inhibitors on Trabecular Outflow. KOREAN JOURNAL OF OPHTHALMOLOGY 2019; 33:414-421. [PMID: 31612651 PMCID: PMC6791954 DOI: 10.3341/kjo.2019.0057] [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: 05/21/2019] [Revised: 06/10/2019] [Accepted: 07/15/2019] [Indexed: 11/23/2022] Open
Abstract
PURPOSE Phosphodiesterase (PDE) inhibitors increase matrix metalloproteinase (MMP) production by inhibiting re-uptake of adenosine and may potentiate nitric oxide (NO) activity. This study was performed to investigate the effects and mechanisms of PDE inhibitors on trabecular outflow in cultured human trabecular meshwork cells (HTMCs). METHODS Primary HTMC cultures were exposed to 0, 20, and 50 μM dipyridamole (DPD) or theophylline (TPN). Permeability through the HTMC monolayer was assessed using carboxyfluorescein. The production of NO was assessed using the Griess assay and MMP-2 levels were measured via Western blotting. RESULTS DPD significantly increased permeability accompanied with increased nitrite concentration and MMP-2 levels (all p < 0.05). TPN increased nitrite but did not affect permeability or MMP-2 levels significantly (p > 0.05). When treated with DPD and TPN together, both permeability and nitrite production were increased; however, MMP-2 levels showed no difference compared to DPD exposure alone (p > 0.05). CONCLUSIONS DPD increased trabecular permeability accompanied with increased nitrite production and MMP-2 levels. PDE inhibitors may increase trabecular outflow by increasing MMP-2 levels and by potentiating NO activity through cyclic GMP in HTMC.
Collapse
Affiliation(s)
- Jae Woo Kim
- Department of Ophthalmology, Daegu Catholic University School of Medicine, Daegu, Korea.
| | - Jong Been Lee
- Department of Ophthalmology, Daegu Catholic University School of Medicine, Daegu, Korea
| | - So Hyung Lee
- Department of Ophthalmology, Daegu Catholic University School of Medicine, Daegu, Korea
| |
Collapse
|
7
|
Tanaka DM, de Oliveira LFL, Marin-Neto JA, Romano MMD, de Carvalho EEV, de Barros Filho ACL, Ribeiro FFF, Cabeza JM, Lopes CD, Fabricio CG, Kesper N, Moreira HT, Wichert-Ana L, Schmidt A, Higuchi MDL, Cunha-Neto E, Simões MV. Prolonged dipyridamole administration reduces myocardial perfusion defects in experimental chronic Chagas cardiomyopathy. J Nucl Cardiol 2019; 26:1569-1579. [PMID: 29392628 DOI: 10.1007/s12350-018-1198-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 12/20/2017] [Indexed: 12/27/2022]
Abstract
BACKGROUND Myocardial perfusion defects (MPD) due to coronary microvascular dysfunction is frequent in chronic Chagas cardiomyopathy (CCC) and may be involved with development of myocardial damage. We investigated whether MPD precedes left ventricular systolic dysfunction and tested the hypothesis that prolonged use of dipyridamole (DIPY) could reduce MPD in an experimental model of CCC in hamsters. METHODS AND RESULTS We investigated female hamsters 6-months after T. cruzi infection (baseline condition) and control animals, divided into T. cruzi-infected animals treated with DIPY (CH + DIPY) or placebo (CH + PLB); and uninfected animals treated with DIPY (CO + DIPY) or placebo (CO + PLB). The animals were submitted to echocardiogram and rest SPECT-Sestamibi-Tc99m myocardial perfusion scintigraphy. Next, the animals were treated with DIPY (4 mg/kg bid, intraperitoneal) or saline for 30 days, and reevaluated with the same imaging methods. At baseline, the CH + PLB and CH + DIPY groups showed larger areas of perfusion defect (13.2 ± 13.2% and 17.3 ± 13.2%, respectively) compared with CO + PLB and CO + DIPY (3.8 ± 2.2% e 3.5 ± 2.7%, respectively), P < .05. After treatment, we observed: reduction of perfusion defects only in the CH + DIPY group (17.3 ± 13.2% to 6.8 ± 7.6%, P = .001) and reduction of LVEF in CH + DIPY and CH + PLB groups (from 65.3 ± 9.0% to 53.6 ± 6.9% and from 69.3 ± 5.0% to 54.4 ± 8.6%, respectively, P < .001). Quantitative histology revealed greater extents of inflammation and interstitial fibrosis in both Chagas groups, compared with control group (P < .001), but no difference between Chagas groups (P > .05). CONCLUSIONS The prolonged use of DIPY in this experimental model of CCC has reduced the rest myocardial perfusion defects, supporting the notion that those areas correspond to viable hypoperfused myocardium.
Collapse
Affiliation(s)
| | | | | | | | - Eduardo Elias Vieira de Carvalho
- Department of Applied Physical Therapy, Institute of Health Sciences, Federal University of Triangulo Mineiro, Minas Gerais, Brazil
| | | | | | | | - Carla Duque Lopes
- Medical School of Ribeirao Preto, University of São Paulo, Sao Paulo, Brazil
| | | | - Norival Kesper
- Instituto de Medicina Tropical, Faculty of Medicine, University os Sao Paulo, Sao Paulo, Brazil
| | | | - Lauro Wichert-Ana
- Medical School of Ribeirao Preto, University of São Paulo, Sao Paulo, Brazil
| | - André Schmidt
- Medical School of Ribeirao Preto, University of São Paulo, Sao Paulo, Brazil
| | | | - Edécio Cunha-Neto
- Heart Institute (InCor), Faculty of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | | |
Collapse
|
8
|
Lana D, Ugolini F, Melani A, Nosi D, Pedata F, Giovannini MG. The neuron-astrocyte-microglia triad in CA3 after chronic cerebral hypoperfusion in the rat: Protective effect of dipyridamole. Exp Gerontol 2017; 96:46-62. [PMID: 28606482 DOI: 10.1016/j.exger.2017.06.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 06/07/2017] [Accepted: 06/08/2017] [Indexed: 12/01/2022]
Abstract
We investigated the quantitative and morphofunctional alterations of neuron-astrocyte-microglia triads in CA3 hippocampus, in comparison to CA1, after 2 Vessel Occlusion (2VO) and the protective effect of dipyridamole. We evaluated 3 experimental groups: sham-operated rats (sham, n=15), 2VO-operated rats treated with vehicle (2VO-vehicle, n=15), and 2VO-operated rats treated with dipyridamole from day 0 to day 7 (2VO-dipyridamole, n=15), 90days after 2VO. We analyzed Stratum Pyramidalis (SP), Stratum Lucidum (SL) and Stratum Radiatum (SR) of CA3. 1) ectopic neurons increased in SL and SR of 2VO-vehicle, and 2VO-dipyridamole rats; 2) apoptotic neurons increased in SP of 2VO-vehicle rats and dipyridamole reverted this effect; 3) astrocytes increased in SP, SL and SR of 2VO-vehicle and 2VO-dipyridamole rats; 4) TNF-α expression increased in astrocytes, blocked by dipyridamole, and in dendrites in SR of 2VO-vehicle rats; 5) total microglia increased in SL and SR of 2VO-vehicle and 2VO-dipyridamole rats; 6) triads increased in SR of 2VO-vehicle rats and dipyridamole reverted this effect. Microglia cooperated with astrocytes to phagocytosis of apoptotic neurons and debris, and engulfed ectopic non-fragmented neurons in SL of 2VO-vehicle and 2VO-dipyridamole rats, through a new mechanism called phagoptosis. CA3 showed a better adaptive capacity than CA1 to the ischemic insult, possibly due to the different behaviour of astrocytes and microglial cells. Dipyridamole had neuroprotective effects.
Collapse
Affiliation(s)
- Daniele Lana
- Department of Health Sciences, Section of Pharmacology and Clinical Oncology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy.
| | - Filippo Ugolini
- Department of Health Sciences, Section of Pharmacology and Clinical Oncology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy.
| | - Alessia Melani
- Department of NEUROFARBA, Section of Pharmacology and Toxicology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy.
| | - Daniele Nosi
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla, 50139 Firenze, Italy.
| | - Felicita Pedata
- Department of NEUROFARBA, Section of Pharmacology and Toxicology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy.
| | - Maria Grazia Giovannini
- Department of Health Sciences, Section of Pharmacology and Clinical Oncology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy.
| |
Collapse
|
9
|
Chen M, Fan H, Ledford BT, Farah Z, Barron C, Liu Z, He JQ. Impacts of femoral artery and vein excision versus femoral artery excision on the hindlimb ischemic model in CD-1 mice. Microvasc Res 2017; 110:48-55. [DOI: 10.1016/j.mvr.2016.12.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 12/15/2016] [Accepted: 12/16/2016] [Indexed: 01/10/2023]
|
10
|
Yuan S, Kevil CG. Nitric Oxide and Hydrogen Sulfide Regulation of Ischemic Vascular Remodeling. Microcirculation 2016; 23:134-45. [PMID: 26381654 DOI: 10.1111/micc.12248] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 09/13/2015] [Indexed: 12/22/2022]
Abstract
Blockage or restriction of blood flow through conduit arteries results in tissue ischemia downstream of the disturbed area. Local tissues can adapt to this challenge by stimulating vascular remodeling through angiogenesis and arteriogenesis thereby restoring blood perfusion and removal of wastes. Multiple molecular mechanisms of vascular remodeling during ischemia have been identified and extensively studied. However, therapeutic benefits from these findings and insights are limited due to the complexity of various signaling networks and a lack of understanding central metabolic regulators governing these responses. The gasotransmitters NO and H2 S have emerged as master regulators that influence multiple molecular targets necessary for ischemic vascular remodeling. In this review, we discuss how NO and H2 S are individually regulated under ischemia, what their roles are in angiogenesis and arteriogenesis, and how their interaction controls ischemic vascular remodeling.
Collapse
Affiliation(s)
- Shuai Yuan
- Departments of Pathology, Molecular and Cellular Physiology, and Cell Biology and Anatomy, LSU Health Shreveport, Shreveport, Louisiana, USA
| | - Christopher G Kevil
- Departments of Pathology, Molecular and Cellular Physiology, and Cell Biology and Anatomy, LSU Health Shreveport, Shreveport, Louisiana, USA
| |
Collapse
|
11
|
Lana D, Iovino L, Nosi D, Wenk GL, Giovannini MG. The neuron-astrocyte-microglia triad involvement in neuroinflammaging mechanisms in the CA3 hippocampus of memory-impaired aged rats. Exp Gerontol 2016; 83:71-88. [PMID: 27466072 DOI: 10.1016/j.exger.2016.07.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 06/23/2016] [Accepted: 07/20/2016] [Indexed: 01/08/2023]
Abstract
We examined the effects of inflammaging on memory encoding, and qualitative and quantitative modifications on proinflammatory proteins, apoptosis, neurodegeneration and morphological changes of neuron-astrocyte-microglia triads in CA3 Stratum Pyramidale (SP), Stratum Lucidum (SL) and Stratum Radiatum (SR) of young (3months) and aged rats (20months). Aged rats showed short-term memory impairments in the inhibitory avoidance task, increased expression of iNOS and activation of p38MAPK in SP, increase of apoptotic neurons in SP and of ectopic neurons in SL, and decrease of CA3 pyramidal neurons. The number of astrocytes and their branches length decreased in the three CA3 subregions of aged rats, with morphological signs of clasmatodendrosis. Total and activated microglia increased in the three CA3 subregions of aged rats. In aged rats CA3, astrocytes surrounded ectopic degenerating neurons forming "micro scars" around them. Astrocyte branches infiltrated the neuronal cell body, and, together with activated microglia formed "triads". In the triads, significantly more numerous in CA3 SL and SR of aged rats, astrocytes and microglia cooperated in fragmentation and phagocytosis of ectopic neurons. Inflammaging-induced modifications of astrocytes and microglia in CA3 of aged rats may help clearing neuronal debris derived from low-grade inflammation and apoptosis. These events might be common mechanisms underlying many neurodegenerative processes. The frequency to which they appear might depend upon, or might be the cause of, the burden and severity of neurodegeneration. Targeting the triads may represent a therapeutic strategy which may control inflammatory processes and spread of further cellular damage to neighboring cells.
Collapse
Affiliation(s)
- Daniele Lana
- Department of Health Sciences, Section of Pharmacology and Clinical Oncology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy.
| | - Ludovica Iovino
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, Viale Morgagni 63 and Section of Anatomy and Histology, Largo Brambilla 3, University of Florence, 50134 Firenze, Italy.
| | - Daniele Nosi
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, Viale Morgagni 63 and Section of Anatomy and Histology, Largo Brambilla 3, University of Florence, 50134 Firenze, Italy.
| | - Gary L Wenk
- Department of Psychology, The Ohio State University, OH, USA..
| | - Maria Grazia Giovannini
- Department of Health Sciences, Section of Pharmacology and Clinical Oncology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy.
| |
Collapse
|
12
|
Sharma AK, Kumar A, Taneja G, Nagaich U, Deep A, Rajput SK. Synthesis and preliminary therapeutic evaluation of copper nanoparticles against diabetes mellitus and -induced micro- (renal) and macro-vascular (vascular endothelial and cardiovascular) abnormalities in rats. RSC Adv 2016. [DOI: 10.1039/c6ra03890e] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Current study synthesized and investigated the effect of low-dose copper nanoparticles (CuNPs) against diabetes mellitus and -induced experimental micro- (nephropathy) and macro-vascular (cardio and endothelium) complications.
Collapse
Affiliation(s)
- Arun K. Sharma
- Cardiovascular Pharmacology Division
- Department of Pharmacology
- Amity Institute of Pharmacy
- Amity University
- Noida
| | - Ashish Kumar
- Department of Nanomedicine and Pharmaceutics
- Amity University
- Noida
- India
| | - Gaurav Taneja
- Cardiovascular Pharmacology Division
- Department of Pharmacology
- Amity Institute of Pharmacy
- Amity University
- Noida
| | - Upendra Nagaich
- Department of Nanomedicine and Pharmaceutics
- Amity University
- Noida
- India
| | - Aakash Deep
- Department of Pharmaceutical Chemistry
- Chaudhary Bansi Lal University
- Bhiwani 127021
- India
| | - Satyendra K. Rajput
- Cardiovascular Pharmacology Division
- Department of Pharmacology
- Amity Institute of Pharmacy
- Amity University
- Noida
| |
Collapse
|
13
|
Heuslein JL, Meisner JK, Li X, Song J, Vincentelli H, Leiphart RJ, Ames EG, Blackman BR, Blackman BR, Price RJ. Mechanisms of Amplified Arteriogenesis in Collateral Artery Segments Exposed to Reversed Flow Direction. Arterioscler Thromb Vasc Biol 2015; 35:2354-65. [PMID: 26338297 PMCID: PMC4618717 DOI: 10.1161/atvbaha.115.305775] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 08/14/2015] [Indexed: 12/27/2022]
Abstract
OBJECTIVE Collateral arteriogenesis, the growth of existing arterial vessels to a larger diameter, is a fundamental adaptive response that is often critical for the perfusion and survival of tissues downstream of chronic arterial occlusion(s). Shear stress regulates arteriogenesis; however, the arteriogenic significance of reversed flow direction, occurring in numerous collateral artery segments after femoral artery ligation, is unknown. Our objective was to determine if reversed flow direction in collateral artery segments differentially regulates endothelial cell signaling and arteriogenesis. APPROACH AND RESULTS Collateral segments experiencing reversed flow direction after femoral artery ligation in C57BL/6 mice exhibit increased pericollateral macrophage recruitment, amplified arteriogenesis (30% diameter and 2.8-fold conductance increases), and remarkably permanent (12 weeks post femoral artery ligation) remodeling. Genome-wide transcriptional analyses on human umbilical vein endothelial cells exposed to reversed flow conditions mimicking those occurring in vivo yielded 10-fold more significantly regulated transcripts, as well as enhanced activation of upstream regulators (nuclear factor κB [NFκB], vascular endothelial growth factor, fibroblast growth factor-2, and transforming growth factor-β) and arteriogenic canonical pathways (protein kinase A, phosphodiesterase, and mitogen-activated protein kinase). Augmented expression of key proarteriogenic molecules (Kruppel-like factor 2 [KLF2], intercellular adhesion molecule 1, and endothelial nitric oxide synthase) was also verified by quantitative real-time polymerase chain reaction, leading us to test whether intercellular adhesion molecule 1 or endothelial nitric oxide synthase regulate amplified arteriogenesis in flow-reversed collateral segments in vivo. Interestingly, enhanced pericollateral macrophage recruitment and amplified arteriogenesis was attenuated in flow-reversed collateral segments after femoral artery ligation in intercellular adhesion molecule 1(-/-) mice; however, endothelial nitric oxide synthase(-/-) mice showed no such differences. CONCLUSIONS Reversed flow leads to a broad amplification of proarteriogenic endothelial signaling and a sustained intercellular adhesion molecule 1-dependent augmentation of arteriogenesis. Further investigation of the endothelial mechanotransduction pathways activated by reversed flow may lead to more effective and durable therapeutic options for arterial occlusive diseases.
Collapse
Affiliation(s)
- Joshua L Heuslein
- From the Departments of Biomedical Engineering (J.L.H., J.K.M., X.L., J.S., H.V., R.J.L., E.G.A., R.J.P.), Molecular Physiology and Biological Physics (E.G.A.), Radiology (R.J.P.), and Radiation Oncology (R.J.P.), University of Virginia, Charlottesville; and HemoShear Therapeutics LLC, Charlottesville, VA (B.R.B.)
| | - Joshua K Meisner
- From the Departments of Biomedical Engineering (J.L.H., J.K.M., X.L., J.S., H.V., R.J.L., E.G.A., R.J.P.), Molecular Physiology and Biological Physics (E.G.A.), Radiology (R.J.P.), and Radiation Oncology (R.J.P.), University of Virginia, Charlottesville; and HemoShear Therapeutics LLC, Charlottesville, VA (B.R.B.)
| | - Xuanyue Li
- From the Departments of Biomedical Engineering (J.L.H., J.K.M., X.L., J.S., H.V., R.J.L., E.G.A., R.J.P.), Molecular Physiology and Biological Physics (E.G.A.), Radiology (R.J.P.), and Radiation Oncology (R.J.P.), University of Virginia, Charlottesville; and HemoShear Therapeutics LLC, Charlottesville, VA (B.R.B.)
| | - Ji Song
- From the Departments of Biomedical Engineering (J.L.H., J.K.M., X.L., J.S., H.V., R.J.L., E.G.A., R.J.P.), Molecular Physiology and Biological Physics (E.G.A.), Radiology (R.J.P.), and Radiation Oncology (R.J.P.), University of Virginia, Charlottesville; and HemoShear Therapeutics LLC, Charlottesville, VA (B.R.B.)
| | - Helena Vincentelli
- From the Departments of Biomedical Engineering (J.L.H., J.K.M., X.L., J.S., H.V., R.J.L., E.G.A., R.J.P.), Molecular Physiology and Biological Physics (E.G.A.), Radiology (R.J.P.), and Radiation Oncology (R.J.P.), University of Virginia, Charlottesville; and HemoShear Therapeutics LLC, Charlottesville, VA (B.R.B.)
| | - Ryan J Leiphart
- From the Departments of Biomedical Engineering (J.L.H., J.K.M., X.L., J.S., H.V., R.J.L., E.G.A., R.J.P.), Molecular Physiology and Biological Physics (E.G.A.), Radiology (R.J.P.), and Radiation Oncology (R.J.P.), University of Virginia, Charlottesville; and HemoShear Therapeutics LLC, Charlottesville, VA (B.R.B.)
| | - Elizabeth G Ames
- From the Departments of Biomedical Engineering (J.L.H., J.K.M., X.L., J.S., H.V., R.J.L., E.G.A., R.J.P.), Molecular Physiology and Biological Physics (E.G.A.), Radiology (R.J.P.), and Radiation Oncology (R.J.P.), University of Virginia, Charlottesville; and HemoShear Therapeutics LLC, Charlottesville, VA (B.R.B.)
| | - Brett R Blackman
- From the Departments of Biomedical Engineering (J.L.H., J.K.M., X.L., J.S., H.V., R.J.L., E.G.A., R.J.P.), Molecular Physiology and Biological Physics (E.G.A.), Radiology (R.J.P.), and Radiation Oncology (R.J.P.), University of Virginia, Charlottesville; and HemoShear Therapeutics LLC, Charlottesville, VA (B.R.B.)
| | | | - Richard J Price
- From the Departments of Biomedical Engineering (J.L.H., J.K.M., X.L., J.S., H.V., R.J.L., E.G.A., R.J.P.), Molecular Physiology and Biological Physics (E.G.A.), Radiology (R.J.P.), and Radiation Oncology (R.J.P.), University of Virginia, Charlottesville; and HemoShear Therapeutics LLC, Charlottesville, VA (B.R.B.).
| |
Collapse
|
14
|
Marques SM, Castro PR, Campos PP, Viana CTR, Parreiras PM, Ferreira MAN, Andrade SP. Genetic strain differences in the development of peritoneal fibroproliferative processes in mice. Wound Repair Regen 2015; 22:381-9. [PMID: 24844337 DOI: 10.1111/wrr.12177] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 02/28/2014] [Indexed: 12/17/2022]
Abstract
Fibroproliferative processes are regulated by a wide variety of tissue components and genetic factors. However, whether there are genetic differences in peritoneal fibroproliferative tissue formation, with consequent differences in response to drug treatment, is unclear. We characterize the influence of the genetic background on peritoneal fibroproliferative tissue induced by sponge implants in DBA/1, Swiss, C57BL/6, and BALB/c mouse strains. In addition, responses to dipyridamole in the implants were evaluated. Angiogenesis, assessed by intra-implant hemoglobin content, was highest in Swiss mice, whereas levels of vascular endothelial growth factor were highest in C57BL/6 mice. The levels of pro-inflammatory cytokines and of inflammatory enzymes (myeloperoxidase- and N-acetyl-β-D-glucosaminidase) were also strain-related. The pro-fibrogenic markers transforming growth factor beta-1 and collagen were lowest in implants placed in DBA/1 mice, whereas those in C57BL/6 mice had the highest levels. Differential sensitivity to dipyridamole was also observed, with this compound being pro-angiogenic in implants placed in DBA/1 mice but antiangiogenic in implants placed in Swiss. An overall anti-inflammatory response was observed in the inbred strains. Antifibrogenic effects were observed only in implants placed in C57BL/6 mice. These important strain-related differences in the development of peritoneal fibrosis and in response to dipyridamole must be considered in the design and analysis of studies on fibrogenesis in mice.
Collapse
Affiliation(s)
- Suzane M Marques
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | | | | | | | | | | |
Collapse
|
15
|
Cabiati M, Burchielli S, Matteucci M, Svezia B, Panchetti L, Caselli C, Prescimone T, Morales MA, Del Ry S. Dipyridamole-induced C-type natriuretic peptide mRNA overexpression in a minipig model of pacing-induced left ventricular dysfunction. Peptides 2015; 64:67-73. [PMID: 25613228 DOI: 10.1016/j.peptides.2015.01.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 01/09/2015] [Accepted: 01/09/2015] [Indexed: 12/23/2022]
Abstract
Dipyridamole (DP) restores ischemic tissue blood flow stimulating angiogenesis in eNOS-dependent pathways. C-type natriuretic peptide (CNP) is expected to mimic the migration-stimulatory effect of NO via a cGMP-dependent mechanism. Aim of this study was to assess the role of concomitant treatment with DP on CNP levels in blood and myocardial tissue of minipigs with left ventricular dysfunction (LVD) induced by pacing at 200bpm in the right ventricular apex. Minipigs with DP therapy (DP+, n=4) or placebo (DP-, n=4) and controls (C-SHAM, n=4) underwent 2D-EchoDoppler examination and blood collection before and after 4 weeks of pacing, when cardiac tissue was collected. Histological/immunohistochemical analyses were performed. CNP levels were determined by radioimmunoassay; cardiac CNP, BNP, natriuretic receptors expression by Real-Time PCR. After pacing, cardiac parameters resulted less impaired in DP+ compared to DP-. Histological sections presented normal morphology while the arteriolar density resulted: C-SHAM: 9.0±1.2; DP-: 4.9±0.3; DP+: 6.5±0.6number/mm(2); C-SHAM vs DP- and DP+ p=0.004, p=0.04, respectively. CNP mRNA resulted lower in DP- compared to C-SHAM and DP+ as well as NPR-B (p=0.011, DP- vs DP+). Both NPR-A/NPR-C mRNA expressions were significantly (p<0.001) lower both in DP- and DP+ compared to C-SHAM. BNP mRNA was higher in LVD. CNP plasma levels showed a similar trend with respect to gene expression (C-SHAM: 30.5±15; DP-: 18.6±5.5; DP+: 21.2±4.7pg/ml). These data suggest that DP may serve as a preconditioning agent to increase the protective CNP-mediated endocrine response in LVD. This response, mediated by its specific receptor NPR-B, may offer new insights into molecular targets for treatment of LVD.
Collapse
MESH Headings
- Animals
- Cardiac Pacing, Artificial
- Dipyridamole/pharmacology
- Dipyridamole/therapeutic use
- Disease Models, Animal
- Heart/drug effects
- Natriuretic Peptide, C-Type/genetics
- Natriuretic Peptide, C-Type/metabolism
- Protective Agents/pharmacology
- Protective Agents/therapeutic use
- RNA, Messenger/metabolism
- Swine
- Swine, Miniature
- Up-Regulation
- Ventricular Dysfunction, Left/drug therapy
- Ventricular Dysfunction, Left/etiology
- Ventricular Dysfunction, Left/genetics
- Ventricular Dysfunction, Left/metabolism
Collapse
Affiliation(s)
- M Cabiati
- CNR Institute of Clinical Physiology, CNR, Italy
| | | | - M Matteucci
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | - B Svezia
- CNR Institute of Clinical Physiology, CNR, Italy; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | - L Panchetti
- Fondazione Toscana G. Monasterio, Pisa, Italy
| | - C Caselli
- CNR Institute of Clinical Physiology, CNR, Italy
| | - T Prescimone
- CNR Institute of Clinical Physiology, CNR, Italy
| | - M A Morales
- CNR Institute of Clinical Physiology, CNR, Italy
| | - S Del Ry
- CNR Institute of Clinical Physiology, CNR, Italy.
| |
Collapse
|
16
|
Lana D, Melani A, Pugliese AM, Cipriani S, Nosi D, Pedata F, Giovannini MG. The neuron-astrocyte-microglia triad in a rat model of chronic cerebral hypoperfusion: protective effect of dipyridamole. Front Aging Neurosci 2014; 6:322. [PMID: 25505884 PMCID: PMC4245920 DOI: 10.3389/fnagi.2014.00322] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 11/04/2014] [Indexed: 11/13/2022] Open
Abstract
Chronic cerebral hypoperfusion during aging may cause progressive neurodegeneration as ischemic conditions persist. Proper functioning of the interplay between neurons and glia is fundamental for the functional organization of the brain. The aim of our research was to study the pathophysiological mechanisms, and particularly the derangement of the interplay between neurons and astrocytes-microglia with the formation of "triads," in a model of chronic cerebral hypoperfusion induced by the two-vessel occlusion (2VO) in adult Wistar rats (n = 15). The protective effect of dipyridamole given during the early phases after 2VO (4 mg/kg/day i.v., the first 7 days after 2VO) was verified (n = 15). Sham-operated rats (n = 15) were used as controls. Immunofluorescent triple staining of neurons (NeuN), astrocytes (GFAP), and microglia (IBA1) was performed 90 days after 2VO. We found significantly higher amount of "ectopic" neurons, neuronal debris and apoptotic neurons in CA1 Str. Radiatum and Str. Pyramidale of 2VO rats. In CA1 Str. Radiatum of 2VO rats the amount of astrocytes (cells/mm(2)) did not increase. In some instances several astrocytes surrounded ectopic neurons and formed a "micro scar" around them. Astrocyte branches could infiltrate the cell body of ectopic neurons, and, together with activated microglia cells formed the "triads." In the triad, significantly more numerous in CA1 Str. Radiatum of 2VO than in sham rats, astrocytes and microglia cooperated in the phagocytosis of ectopic neurons. These events might be common mechanisms underlying many neurodegenerative processes. The frequency to which they appear might depend upon, or might be the cause of, the burden and severity of neurodegeneration. Dypiridamole significantly reverted all the above described events. The protective effect of chronic administration of dipyridamole might be a consequence of its vasodilatory, antioxidant and anti-inflammatory role during the early phases after 2VO.
Collapse
Affiliation(s)
- Daniele Lana
- Section of Pharmacology and Clinical Oncology, Department of Health Sciences, University of Florence Florence, Italy
| | - Alessia Melani
- Section of Pharmacology and Toxicology, Department of NEUROFARBA, University of Florence Florence, Italy
| | - Anna Maria Pugliese
- Section of Pharmacology and Toxicology, Department of NEUROFARBA, University of Florence Florence, Italy
| | | | - Daniele Nosi
- Department of Experimental and Clinical Medicine, University of Florence Florence, Italy
| | - Felicita Pedata
- Section of Pharmacology and Toxicology, Department of NEUROFARBA, University of Florence Florence, Italy
| | - Maria Grazia Giovannini
- Section of Pharmacology and Clinical Oncology, Department of Health Sciences, University of Florence Florence, Italy
| |
Collapse
|
17
|
Liu S, Lai L, Zuo Q, Dai F, Wu L, Wang Y, Zhou Q, Liu J, Liu J, Li L, Lin Q, Creighton CJ, Costello MG, Huang S, Jia C, Liao L, Luo H, Fu J, Liu M, Yi Z, Xiao J, Li X. PKA turnover by the REGγ-proteasome modulates FoxO1 cellular activity and VEGF-induced angiogenesis. J Mol Cell Cardiol 2014; 72:28-38. [PMID: 24560667 PMCID: PMC4237316 DOI: 10.1016/j.yjmcc.2014.02.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Revised: 02/04/2014] [Accepted: 02/11/2014] [Indexed: 01/08/2023]
Abstract
The REGγ-proteasome serves as a short-cut for the destruction of certain intact mammalian proteins in the absence of ubiquitin- and ATP. The biological roles of the proteasome activator REGγ are not completely understood. Here we demonstrate that REGγ controls degradation of protein kinase A catalytic subunit-α (PKAca) both in primary human umbilical vein endothelial cells (HUVECs) and mouse embryonic fibroblast cells (MEFs). Accumulation of PKAca in REGγ-deficient HUVECs or MEFs results in phosphorylation and nuclear exclusion of the transcription factor FoxO1, indicating that REGγ is involved in preserving FoxO1 transcriptional activity. Consequently, VEGF-induced expression of the FoxO1 responsive genes, VCAM-1 and E-Selectin, was tightly controlled by REGγ in a PKA dependent manner. Functionally, REGγ is crucial for the migration of HUVECs. REGγ(-/-) mice display compromised VEGF-instigated neovascularization in cornea and aortic ring models. Implanted matrigel plugs containing VEGF in REGγ(-/-) mice induced fewer capillaries than in REGγ(+/+) littermates. Taken together, our study identifies REGγ as a novel angiogenic factor that plays an important role in VEGF-induced expression of VCAM-1 and E-Selectin by antagonizing PKA signaling. Identification of the REGγ-PKA-FoxO1 pathway in endothelial cells (ECs) provides another potential target for therapeutic intervention in vascular diseases.
Collapse
Affiliation(s)
- Shuang Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China; Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China; Department of Hematology, Guangdong No. 2 Provincial People's Hospital, No.1 Shiliugang Rd, Guangzhou, Guangdong 510317, China
| | - Li Lai
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China; Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Qiuhong Zuo
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China; Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Fujun Dai
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China; Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Lin Wu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China; Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Yan Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China; Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Qingxia Zhou
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China; Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Jian Liu
- Department of Molecular and Cellular Biology, Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Jiang Liu
- Institute of Aging Research, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 310036, China
| | - Lei Li
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China; Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Qingxiang Lin
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China; Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Chad J Creighton
- Department of Medicine, Dan L. Duncan Cancer Center Division of Biostatistics, Baylor College of Medicine, Houston, TX, USA
| | - Myra Grace Costello
- Department of Molecular and Cellular Biology, Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Shixia Huang
- Department of Molecular and Cellular Biology, Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Caifeng Jia
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China; Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Lujian Liao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China; Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Honglin Luo
- The James Hogg Research Centre for Cardiovascular and Pulmonary Research, University of British Columbia-St. Paul's Hospital, 1081 Burrard St., Vancouver, British Columbia V6Z 1Y6, Canada
| | - Junjiang Fu
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Luzhou Medical College, Luzhou 646000, China
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China; Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Zhengfang Yi
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China; Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China.
| | - Jianru Xiao
- Department of Orthopaedic Oncology, Changzheng Hospital, The Second Military Medical University, 415 Fengyang Road, Shanghai 200003, China.
| | - Xiaotao Li
- Department of Molecular and Cellular Biology, Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Orthopaedic Oncology, Changzheng Hospital, The Second Military Medical University, 415 Fengyang Road, Shanghai 200003, China; Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China.
| |
Collapse
|
18
|
Balakumar P, Nyo YH, Renushia R, Raaginey D, Oh AN, Varatharajan R, Dhanaraj SA. Classical and pleiotropic actions of dipyridamole: Not enough light to illuminate the dark tunnel? Pharmacol Res 2014; 87:144-50. [PMID: 24861566 DOI: 10.1016/j.phrs.2014.05.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 05/01/2014] [Accepted: 05/13/2014] [Indexed: 12/01/2022]
Abstract
Dipyridamole is a platelet inhibitor indicated for the secondary prevention of transient ischemic attack. It inhibits the enzyme phosphodiesterase, elevates cAMP and cGMP levels and prevents platelet aggregation. Dipyridamole inhibits the cellular uptake of adenosine into red blood cells, platelets and endothelial cells that results in increased extracellular availability of adenosine, leading to modulation of cardiovascular function. The antiplatelet action of dipyridamole might offer therapeutic benefits in secondary stroke prevention in combination with aspirin. Inflammation and oxidative stress play an important role in atherosclerosis and thrombosis development, leading to stroke progression. Studies demonstrated anti-inflammatory, anti-oxidant and anti-proliferative actions of dipyridamole. These pleiotropic potentials of dipyridamole might contribute to improved therapeutic outcomes when used with aspirin in preventing secondary stroke. Dipyridamole was documented as a coronary vasodilator 5 decades ago. The therapeutic failure of dipyridamole as a coronary vasodilator is linked with induction of 'coronary steal' phenomenon in which by dilating resistance vessels in non-ischemic zone, dipyridamole diverts the already reduced blood flow away from the area of ischemic myocardium. Dipyridamole at high-dose could cause a marked 'coronary steal' effect. Dipyridamole, however, at low-dose could have a minimal hemodynamic effect. Low-dose dipyridamole treatment has a therapeutic potential in partially preventing diabetes mellitus-induced experimental vascular endothelial and renal abnormalities by enhancing endothelial nitric oxide signals and inducing renovascular reduction of oxidative stress. In spite of plenteous research on dipyridamole's use in clinics, its precise clinical application is still obscure. This review sheds lights on pleiotropic pharmacological actions and therapeutic potentials of dipyridamole.
Collapse
Affiliation(s)
- Pitchai Balakumar
- Pharmacology Unit, Faculty of Pharmacy, Asian Institute of Medicine, Science and Technology (AIMST) University, Semeling, 08100 Bedong, Kedah Darul Aman, Malaysia.
| | - Ying Hui Nyo
- Pharmacology Unit, Faculty of Pharmacy, Asian Institute of Medicine, Science and Technology (AIMST) University, Semeling, 08100 Bedong, Kedah Darul Aman, Malaysia
| | - Raja Renushia
- Pharmacology Unit, Faculty of Pharmacy, Asian Institute of Medicine, Science and Technology (AIMST) University, Semeling, 08100 Bedong, Kedah Darul Aman, Malaysia
| | - Devarajan Raaginey
- Pharmacology Unit, Faculty of Pharmacy, Asian Institute of Medicine, Science and Technology (AIMST) University, Semeling, 08100 Bedong, Kedah Darul Aman, Malaysia
| | - Ann Nah Oh
- Pharmacology Unit, Faculty of Pharmacy, Asian Institute of Medicine, Science and Technology (AIMST) University, Semeling, 08100 Bedong, Kedah Darul Aman, Malaysia
| | - Rajavel Varatharajan
- Pharmacology Unit, Faculty of Pharmacy, Asian Institute of Medicine, Science and Technology (AIMST) University, Semeling, 08100 Bedong, Kedah Darul Aman, Malaysia
| | - Sokkalingam A Dhanaraj
- Pharmaceutical Technology Unit, Faculty of Pharmacy, AIMST University, Semeling, 08100 Bedong, Kedah Darul Aman, Malaysia
| |
Collapse
|
19
|
Sharma AK, Khanna D, Balakumar P. Low-dose dipyridamole treatment partially prevents diabetes mellitus-induced vascular endothelial and renal abnormalities in rats. Int J Cardiol 2014; 172:530-2. [DOI: 10.1016/j.ijcard.2014.01.053] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 01/12/2014] [Indexed: 10/25/2022]
|
20
|
Bir SC, Pattillo CB, Pardue S, Kolluru GK, Shen X, Giordano T, Kevil CG. Nitrite anion therapy protects against chronic ischemic tissue injury in db/db diabetic mice in a NO/VEGF-dependent manner. Diabetes 2014; 63:270-81. [PMID: 24009258 PMCID: PMC4179307 DOI: 10.2337/db13-0890] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Nitrite anion has been demonstrated to be a prodrug of nitric oxide (NO) with positive effects on tissue ischemia/reperfusion injury, cytoprotection, and vasodilation. However, effects of nitrite anion therapy for ischemic tissue vascular remodeling during diabetes remain unknown. We examined whether sodium nitrite therapy altered ischemic revascularization in BKS-Lepr(db/db) mice subjected to permanent unilateral femoral artery ligation. Sodium nitrite therapy completely restored ischemic hind limb blood flow compared with nitrate or PBS therapy. Importantly, delayed nitrite therapy 5 days after ischemia restored ischemic limb blood flow in aged diabetic mice. Restoration of blood flow was associated with increases in ischemic tissue angiogenesis activity and cell proliferation. Moreover, nitrite but not nitrate therapy significantly prevented ischemia-mediated tissue necrosis in aged mice. Nitrite therapy significantly increased ischemic tissue vascular endothelial growth factor (VEGF) protein expression that was essential for nitrite-mediated reperfusion of ischemic hind limbs. Nitrite significantly increased ischemic tissue NO bioavailability along with concomitant reduction of superoxide formation. Lastly, nitrite treatment also significantly stimulated hypoxic endothelial cell proliferation and migration in the presence of high glucose in an NO/VEGF-dependent manner. These results demonstrate that nitrite therapy effectively stimulates ischemic tissue vascular remodeling in the setting of metabolic dysfunction that may be clinically useful.
Collapse
Affiliation(s)
- Shyamal C. Bir
- Department of Pathology, Louisiana State University Health Sciences Center–Shreveport, Shreveport, LA
| | - Christopher B. Pattillo
- Department of Pathology, Louisiana State University Health Sciences Center–Shreveport, Shreveport, LA
| | - Sibile Pardue
- Department of Pathology, Louisiana State University Health Sciences Center–Shreveport, Shreveport, LA
| | - Gopi K. Kolluru
- Department of Pathology, Louisiana State University Health Sciences Center–Shreveport, Shreveport, LA
| | - Xinggui Shen
- Department of Pathology, Louisiana State University Health Sciences Center–Shreveport, Shreveport, LA
| | | | - Christopher G. Kevil
- Department of Pathology, Louisiana State University Health Sciences Center–Shreveport, Shreveport, LA
- TheraVasc Inc., Cleveland, OH
- Corresponding author: Christopher G. Kevil,
| |
Collapse
|
21
|
Peter EA, Shen X, Shah SH, Pardue S, Glawe JD, Zhang WW, Reddy P, Akkus NI, Varma J, Kevil CG. Plasma free H2S levels are elevated in patients with cardiovascular disease. J Am Heart Assoc 2013; 2:e000387. [PMID: 24152982 PMCID: PMC3835249 DOI: 10.1161/jaha.113.000387] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Hydrogen sulfide (H2S) has been implicated in regulating cardiovascular pathophysiology in experimental models. However, there is a paucity of information regarding the levels of H2S in health and cardiovascular disease. In this study we examine the levels of H2S in patients with cardiovascular disease as well as bioavailability of nitric oxide and inflammatory indicators. METHODS AND RESULTS Patients over the age of 40 undergoing coronary or peripheral angiography were enrolled in the study. Ankle brachial index (ABI) measurement, measurement of plasma-free H2S and total nitric oxide (NO), thrombospondin-1 (TSP-1), Interleukin-6 (IL-6), and soluble intercellular adhesion molecule-1 (sICAM-1) levels were performed. Patients with either coronary artery disease alone (n = 66), peripheral arterial disease (PAD) alone (n = 13), or any vascular disease (n = 140) had higher plasma-free H2S levels compared to patients without vascular disease (n = 53). Plasma-free H2S did not distinguish between disease in different vascular beds; however, total NO levels were significantly reduced in PAD patients and the ratio of plasma free H2S to NO was significantly greater in patients with PAD. Lastly, plasma IL-6, ICAM-1, and TSP-1 levels did not correlate with H2S or NO bioavailability in either vascular disease condition. CONCLUSIONS Findings reported in this study reveal that plasma-free H2S levels are significantly elevated in vascular disease and identify a novel inverse relationship with NO bioavailability in patients with peripheral arterial disease.
Collapse
Affiliation(s)
- Elvis A Peter
- Section of Cardiology, LSU Health Shreveport, Shreveport, LA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Bir SC, Shen X, Kavanagh TJ, Kevil CG, Pattillo CB. Control of angiogenesis dictated by picomolar superoxide levels. Free Radic Biol Med 2013; 63:135-42. [PMID: 23685287 PMCID: PMC3732119 DOI: 10.1016/j.freeradbiomed.2013.05.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Revised: 05/08/2013] [Accepted: 05/09/2013] [Indexed: 12/12/2022]
Abstract
Control of vascular insufficiencies due to various cardiovascular pathologies is important for developing specific and effective treatments. Fluctuations in oxidative stress significantly alter the progression of angiogenesis under physiological and pathological conditions. However, the precise amount of reactive oxygen species (ROS) required to influence subsequent signaling pathways for ischemic angiogenesis remains undefined. Here, we have determined the effect of ROS-mediated molecular mechanisms on angiogenesis in a murine model of peripheral artery disease using Gclm mutant mice (a model of compromised glutathione synthesis and therefore reduced antioxidant capacity). Left femoral artery ligation and excision were performed in Gclm WT (+/+), heterozygous (+/-), and null (-/-) mice. Blood flow (laser Doppler), angiogenic index (CD31/DAPI), and proliferation index (Ki67/DAPI) were significantly increased in Gclm(+/-) mice but not in Gclm(+/+) or Gclm(-/-) mice. Measurements of reactive oxygen species suggest that the amount of superoxide required to stimulate angiogenesis after the induction of ischemia is 9.82 pmol/mg of tissue. Protein carbonyl levels increased in a manner consistent with increasing oxidative stress. Superoxide and protein carbonyl levels were reduced by the addition of the nitroxide tempol, a known superoxide dismutase mimetic. Finally, restoration of blood flow in Gclm(+/-) mice was attenuated by a VEGF164 aptamer, verifying that slightly elevated levels of ROS restore blood flow by stimulating endothelial cell proliferation through a VEGF-dependent pathway. The results of this study reveal new information on the amount of ROS necessary for angiogenic activity and provide the foundation of critical redox parameters for vascular remodeling responses. The information obtained from this study on vascular ischemia, using a model of decreased antioxidant capacity, has provided insight into the control of revascularization and is a step forward in our ability to regulate angiogenic therapies.
Collapse
Affiliation(s)
- Shyamal C Bir
- Department of Pathology, LSU Health Sciences Center-Shreveport, Shreveport, LA 71104
| | - Xinggui Shen
- Department of Pathology, LSU Health Sciences Center-Shreveport, Shreveport, LA 71104
| | - Terrance J Kavanagh
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98195
| | - Christopher G Kevil
- Department of Pathology, LSU Health Sciences Center-Shreveport, Shreveport, LA 71104
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center-Shreveport, Shreveport, LA 71104
| | - Christopher B Pattillo
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center-Shreveport, Shreveport, LA 71104
- Correspondence to: Christopher B. Pattillo, Ph.D., Department of Molecular and Cellular Physiology, LSU Health Sciences Center-Shreveport, 1501 Kings Hwy, Shreveport, LA 71130, , Phone: (318) 675-6974, Fax: (318) 675-6005
| |
Collapse
|
23
|
Kamga Pride C, Mo L, Quesnelle K, Dagda RK, Murillo D, Geary L, Corey C, Portella R, Zharikov S, St Croix C, Maniar S, Chu CT, Khoo NKH, Shiva S. Nitrite activates protein kinase A in normoxia to mediate mitochondrial fusion and tolerance to ischaemia/reperfusion. Cardiovasc Res 2013; 101:57-68. [PMID: 24081164 DOI: 10.1093/cvr/cvt224] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AIMS Nitrite (NO2(-)), a dietary constituent and nitric oxide (NO) oxidation product, mediates cardioprotection after ischaemia/reperfusion (I/R) in a number of animal models when administered during ischaemia or as a pre-conditioning agent hours to days prior to the ischaemic episode. When present during ischaemia, the reduction of nitrite to bioactive NO by deoxygenated haem proteins accounts for its protective effects. However, the mechanism of nitrite-induced pre-conditioning, a normoxic response which does not appear to require reduction of nitrite to NO, remains unexplored. METHODS AND RESULTS Using a model of hypoxia/reoxygenation (H/R) in cultured rat H9c2 cardiomyocytes, we demonstrate that a transient (30 min) normoxic nitrite treatment significantly attenuates cell death after a hypoxic episode initiated 1 h later. Mechanistically, this protection depends on the activation of protein kinase A, which phosphorylates and inhibits dynamin-related protein 1, the predominant regulator of mitochondrial fission. This results morphologically, in the promotion of mitochondrial fusion and functionally in the augmentation of mitochondrial membrane potential and superoxide production. We identify AMP kinase (AMPK) as a downstream target of the mitochondrial reactive oxygen species (ROS) generated and show that its oxidation and subsequent phosphorylation are essential for cytoprotection, as scavenging of ROS prevents AMPK activation and inhibits nitrite-mediated protection after H/R. The protein kinase A-dependent protection mediated by nitrite is reproduced in an intact isolated rat heart model of I/R. CONCLUSIONS These data are the first to demonstrate nitrite-dependent normoxic modulation of both mitochondrial morphology and function and reveal a novel signalling pathway responsible for nitrite-mediated cardioprotection.
Collapse
|
24
|
Xue SY, Hebert VY, Hayes DM, Robinson CN, Glover M, Dugas TR. Nucleoside reverse transcriptase inhibitors induce a mitophagy-associated endothelial cytotoxicity that is reversed by coenzyme Q10 cotreatment. Toxicol Sci 2013; 134:323-34. [PMID: 23640862 DOI: 10.1093/toxsci/kft105] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Cardiovascular complications have been documented in HIV-1 infected populations, and antiretroviral therapy may play a role. Nucleoside reverse transcriptase inhibitors (NRTIs) are antiretrovirals known to induce mitochondrial damage in endothelial cells, culminating in endothelial dysfunction, an initiating event in atherogenesis. Though the mechanism for NRTI-induced endothelial toxicity is not yet clear, our prior work suggested that a mitochondrial oxidative stress may be involved. To further delineate the mechanism of toxicity, endothelial cells were treated with NRTIs of varying subclasses, and the level of reactive oxygen species (ROS) and mitochondrial function were assessed. To test whether rescue of mitochondrial electron transport attenuated NRTI-induced endothelial cytotoxicity, in some cases, cells were cotreated with the electron transport cofactor coenzyme Q10 (Q10). At 4-6h, NRTIs increased levels of ROS but decreased the activities of electron transport chain complexes I-IV, levels of ATP and the NAD/NADH ratio. Moreover, nitric oxide levels were decreased, whereas endothelin-1 release was increased. Q10 abolished NRTI-induced mitochondria injury and effects on endothelial agonist production. Interestingly, in cells treated with NRTIs only, markers for mitochondrial toxicity returned to baseline levels by 18-24h, suggesting a compensatory mechanism for clearing damaged mitochondria. Using confocal microscopy, with confirmation utilizing the autophagy and mitophagy markers LC-3 and Nix, respectively, we observed autophagy of mitochondria at 8-10h after treatment. Q10 prevented NRTI-mediated increase in LC-3. These findings suggest that NRTI-induced mitophagy may be involved in NRTI-induced endothelial dysfunction and that this damage likely results from oxidant injury. Further, Q10 supplementation could potentially prevent NRTI-induced endothelial dysfunction.
Collapse
Affiliation(s)
- Stephen Y Xue
- Department of Pharmacology, Toxicology, and Neuroscience, LSU Health Sciences Center in Shreveport, Shreveport, Louisiana 71130, USA
| | | | | | | | | | | |
Collapse
|
25
|
Bryan NS. Pharmacological therapies, lifestyle choices and nitric oxide deficiency: A perfect storm. Pharmacol Res 2012; 66:448-56. [DOI: 10.1016/j.phrs.2012.09.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Revised: 08/28/2012] [Accepted: 09/15/2012] [Indexed: 01/05/2023]
|
26
|
High-throughput screening for bioactive molecules using primary cell culture of transgenic zebrafish embryos. Cell Rep 2012; 2:695-704. [PMID: 22999940 DOI: 10.1016/j.celrep.2012.08.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Revised: 06/01/2012] [Accepted: 08/15/2012] [Indexed: 11/20/2022] Open
Abstract
Transgenic zebrafish embryos expressing tissue-specific green fluorescent protein (GFP) can provide an unlimited supply of primary embryonic cells. Agents that promote the differentiation of these cells may be beneficial for therapeutics. We report a high-throughput approach for screening small molecules that regulate cell differentiation using lineage-specific GFP transgenic zebrafish embryonic cells. After validating several known regulators of the differentiation of endothelial and other cell types, we performed a screen for proangiogenic molecules using undifferentiated primary cells from flk1-GFP transgenic zebrafish embryos. Cells were grown in 384-well plates with 12,128 individual small molecules, and GFP expression was analyzed by means of an automated imaging system, which allowed us to screen thousands of compounds weekly. As a result, 23 molecules were confirmed to enhance angiogenesis, and 11 of them were validated to promote the proliferation of mammalian human umbilical vascular endothelial cells and induce Flk1+ cells from murine embryonic stem cells. We demonstrated the general applicability of this strategy by analyzing additional cell lineages using zebrafish expressing GFP in pancreatic, cardiac, and dopaminergic cells.
Collapse
|
27
|
Sampaio FP, Castro PR, Marques SM, Campos PP, Ferreira MAND, Andrade SP. Genetic background determines inflammatory angiogenesis response to dipyridamole in mice. Exp Biol Med (Maywood) 2012; 237:1084-92. [DOI: 10.1258/ebm.2012.012066] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Inflammation and angiogenesis, key components of fibrovascular tissue growth, exhibit considerable variability among species and strains. We investigated whether the response of inbred and outbred mice strains to dipyridamole (DP) on these processes would present similar variability. The effects of the drug on blood vessel formation, inflammatory cell recruitment, collagen deposition and cytokine production were determined on the fibroproliferative tissue induced by sponge implants in Swiss and Balb/c mice. Angiogenesis as assessed by hemoglobin (Hb) and vascular endothelial growth factor (VEGF) concentrations differed between the strains. Swiss implants had the highest Hb content but the lowest VEGF concentrations. Systemic DP treatment exerted an antiangiogenic effect on Balb/c implants but an proangiogenic effect on Swiss implants. The inflammatory enzyme activities myeloperoxidase (six-fold higher in Balb/c implants) and N-acetyl- β-d-glucosaminidase were reduced by the treatment in Balb/c implants only. Nitrite concentrations were also higher in Balb/c implants by 40% after DP treatment. Tumor necrosis factor-alpha levels were similar in the implants of both strains and were not reduced by DP. Transforming growth factor β-1 levels and collagen deposition also varied between the strains. The inbred strain had similar levels of the cytokine but implants of Swiss mice presented more collagen. DP treatment reduced collagen deposition in Balb/c implants only. Our data showing the influence of the genetic background on marked heterogeneity of inflammatory angiogenesis components and differential sensitivity to DP may provide some answers to clinical evidence for resistance to angiogenic therapy.
Collapse
Affiliation(s)
| | | | | | - Paula Peixoto Campos
- General Pathology, Institute of Biological Sciences, Federal University of Minas Gerais, Av. Antônio Carlos 6627-Campus Pampulha, Cx Post 468, CEP 31270-901, Belo Horizonte/MG, Brazil
| | - Mônica Alves Neves Diniz Ferreira
- General Pathology, Institute of Biological Sciences, Federal University of Minas Gerais, Av. Antônio Carlos 6627-Campus Pampulha, Cx Post 468, CEP 31270-901, Belo Horizonte/MG, Brazil
| | | |
Collapse
|
28
|
Castiglione N, Rinaldo S, Giardina G, Stelitano V, Cutruzzolà F. Nitrite and nitrite reductases: from molecular mechanisms to significance in human health and disease. Antioxid Redox Signal 2012; 17:684-716. [PMID: 22304560 DOI: 10.1089/ars.2011.4196] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Nitrite, previously considered physiologically irrelevant and a simple end product of endogenous nitric oxide (NO) metabolism, is now envisaged as a reservoir of NO to be activated in response to oxygen (O(2)) depletion. In the first part of this review, we summarize and compare the mechanisms of nitrite-dependent production of NO in selected bacteria and in eukaryotes. Bacterial nitrite reductases, which are copper or heme-containing enzymes, play an important role in the adaptation of pathogens to O(2) limitation and enable microrganisms to survive in the human body. In mammals, reduction of nitrite to NO under hypoxic conditions is carried out in tissues and blood by an array of metalloproteins, including heme-containing proteins and molybdenum enzymes. In humans, tissues play a more important role in nitrite reduction, not only because most tissues produce more NO than blood, but also because deoxyhemoglobin efficiently scavenges NO in blood. In the second part of the review, we outline the significance of nitrite in human health and disease and describe the recent advances and pitfalls of nitrite-based therapy, with special attention to its application in cardiovascular disorders, inflammation, and anti-bacterial defence. It can be concluded that nitrite (as well as nitrate-rich diet for long-term applications) may hold promise as therapeutic agent in vascular dysfunction and ischemic injury, as well as an effective compound able to promote angiogenesis.
Collapse
Affiliation(s)
- Nicoletta Castiglione
- Department of Biochemical Sciences, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | | | | | | | | |
Collapse
|
29
|
Bir SC, Pattillo CB, Pardue S, Kolluru GK, Docherty J, Goyette D, Dvorsky P, Kevil CG. Nitrite anion stimulates ischemic arteriogenesis involving NO metabolism. Am J Physiol Heart Circ Physiol 2012; 303:H178-88. [PMID: 22610173 DOI: 10.1152/ajpheart.01086.2010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Nitric oxide (NO) is a potential regulator of ischemic vascular remodeling, and as such therapies augmenting its bioavailability may be useful for the treatment of ischemic tissue diseases. Here we examine the effect of administering the NO prodrug sodium nitrite on arteriogenesis activity during established tissue ischemia. Chronic hindlimb ischemia was induced by permanent unilateral femoral artery and vein ligation. Five days postligation; animals were randomized to control PBS or sodium nitrite (165 μg/kg) therapy twice daily. In situ vascular remodeling was measured longitudinally using SPY angiography and Microfil vascular casting. Delayed sodium nitrite therapy rapidly increased ischemic limb arterial vessel diameter and branching in a NO-dependent manner. SPY imaging angiography over time showed that nitrite therapy enhanced ischemic gracillis collateral vessel formation from the profunda femoris to the saphenous artery. Immunofluorescent staining of smooth muscle cell actin also confirmed that sodium nitrite therapy increased arteriogenesis in a NO-dependent manner. The NO prodrug sodium nitrite significantly increases arteriogenesis and reperfusion of established severe chronic tissue ischemia.
Collapse
Affiliation(s)
- Shyamal C Bir
- Department of Pathology Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71130, USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Allen JD, Giordano T, Kevil CG. Nitrite and nitric oxide metabolism in peripheral artery disease. Nitric Oxide 2012; 26:217-22. [PMID: 22426034 PMCID: PMC3360821 DOI: 10.1016/j.niox.2012.03.003] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Revised: 02/10/2012] [Accepted: 03/06/2012] [Indexed: 11/20/2022]
Abstract
Peripheral artery disease (PAD) represents a burgeoning form of cardiovascular disease associated with significant clinical morbidity and increased 5 year cardiovascular disease mortality. It is characterized by impaired blood flow to the lower extremities, claudication pain and severe exercise intolerance. Pathophysiological factors contributing to PAD include atherosclerosis, endothelial cell dysfunction, and defective nitric oxide metabolite physiology and biochemistry that collectively lead to intermittent or chronic tissue ischemia. Recent work from our laboratories is revealing that nitrite/nitrate anion and nitric oxide metabolism plays an important role in modulating functional and pathophysiological responses during this disease. In this review, we discuss experimental and clinical findings demonstrating that nitrite anion acts to ameliorate numerous pathophysiological events associated with PAD and chronic tissue ischemia. We also highlight future directions for this promising line of therapy.
Collapse
Affiliation(s)
- Jason D. Allen
- Department of Medicine, Duke University Medical Center, Durham, NC
| | | | | |
Collapse
|
31
|
Schirmer SH, Werner CM, Laufs U, Bohm M. Nitric oxide-donating statins: a new concept to boost the lipid-independent effects. Cardiovasc Res 2012; 94:395-7. [DOI: 10.1093/cvr/cvs148] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
32
|
Bir SC, Xiong Y, Kevil CG, Luo J. Emerging role of PKA/eNOS pathway in therapeutic angiogenesis for ischaemic tissue diseases. Cardiovasc Res 2012; 95:7-18. [PMID: 22492672 DOI: 10.1093/cvr/cvs143] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Although an abundant amount of research has been devoted to the study of angiogenesis, its precise mechanisms are incompletely understood. Numerous clinical trials focused on therapeutic angiogenesis for the treatment of tissue ischaemia have not been as successful as those of preclinical studies. Thus, additional studies are needed to better understand critical molecular mechanisms regulating ischaemic neovascularization to identify novel therapeutic agents. Nitric oxide (NO) plays a central role in ischaemic neovascularization through the generation of cyclic guanosine monophosphate (cGMP) and the activation of several other signalling responses. Accumulated evidence suggests that endothelial protein kinase A/endothelial NO synthase (PKA/eNOS) signalling may play an important role in ischaemic disorders by promoting neovascularization. This review highlights recent advances in the role of the PKA/eNOS and NO-cGMP-kinase cascade pathway in ischaemic neovascularization. We also discuss molecular relationships of PKA/eNOS with other angiogenic pathways and explore the possibility of activation of the NO/nitrite endocrine system as potential therapeutic targets for ischaemic angiogenesis.
Collapse
Affiliation(s)
- Shyamal C Bir
- Department of Pathology, LSU Health Sciences Center-Shreveport, LA, USA
| | | | | | | |
Collapse
|
33
|
Srivastava K, Bath PMW, Bayraktutan U. Current therapeutic strategies to mitigate the eNOS dysfunction in ischaemic stroke. Cell Mol Neurobiol 2012; 32:319-36. [PMID: 22198555 DOI: 10.1007/s10571-011-9777-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Accepted: 11/29/2011] [Indexed: 12/22/2022]
Abstract
Impairment of endothelial nitric oxide synthase (eNOS) activity is implicated in the pathogenesis of endothelial dysfunction in many diseases including ischaemic stroke. The modulation of eNOS during and/or following ischaemic injury often represents a futile compensatory mechanism due to a significant decrease in nitric oxide (NO) bioavailability coupled with dramatic increases in the levels of reactive oxygen species that further neutralise NO. However, applications of a number of therapeutic agents alone or in combination have been shown to augment eNOS activity under a variety of pathological conditions by potentiating the expression and/or activity of Akt/eNOS/NO pathway components. The list of these therapeutic agents include NO donors, statins, angiotensin-converting enzyme inhibitors, calcium channel blockers, phosphodiesterase-3 inhibitors, aspirin, dipyridamole and ellagic acid. While most of these compounds exhibit anti-platelet properties and are able to up-regulate eNOS expression in endothelial cells and platelets, others suppress eNOS uncoupling and tetrahydrobiopterin (an eNOS stabiliser) oxidation. As the number of therapeutic molecules that modulate the expression and activity of eNOS increases, further detailed research is required to reveal their mode of action in preventing and/or reversing the endothelial dysfunction.
Collapse
Affiliation(s)
- Kirtiman Srivastava
- Division of Stroke, Clinical Sciences Building, Nottingham City Hospital Campus, The University of Nottingham, Nottingham, UK.
| | | | | |
Collapse
|
34
|
Kolluru GK, Bir SC, Kevil CG. Endothelial dysfunction and diabetes: effects on angiogenesis, vascular remodeling, and wound healing. Int J Vasc Med 2012; 2012:918267. [PMID: 22611498 PMCID: PMC3348526 DOI: 10.1155/2012/918267] [Citation(s) in RCA: 325] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Accepted: 10/18/2011] [Indexed: 02/06/2023] Open
Abstract
Diabetes mellitus (DM) is a chronic metabolic disorder characterized by inappropriate hyperglycemia due to lack of or resistance to insulin. Patients with DM are frequently afflicted with ischemic vascular disease or wound healing defect. It is well known that type 2 DM causes amplification of the atherosclerotic process, endothelial cell dysfunction, glycosylation of extracellular matrix proteins, and vascular denervation. These complications ultimately lead to impairment of neovascularization and diabetic wound healing. Therapeutic angiogenesis remains an attractive treatment modality for chronic ischemic disorders including PAD and/or diabetic wound healing. Many experimental studies have identified better approaches for diabetic cardiovascular complications, however, successful clinical translation has been limited possibly due to the narrow therapeutic targets of these agents or the lack of rigorous evaluation of pathology and therapeutic mechanisms in experimental models of disease. This paper discusses the current body of evidence identifying endothelial dysfunction and impaired angiogenesis during diabetes.
Collapse
Affiliation(s)
| | | | - Christopher G. Kevil
- Department of Pathology, LSU Health Sciences Center-Shreveport, 1501 Kings Highway, Shreveport, LA 71130, USA
| |
Collapse
|
35
|
Gresele P, Momi S, Falcinelli E. Anti-platelet therapy: phosphodiesterase inhibitors. Br J Clin Pharmacol 2012; 72:634-46. [PMID: 21649691 DOI: 10.1111/j.1365-2125.2011.04034.x] [Citation(s) in RCA: 203] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Inhibition of platelet aggregation can be achieved either by the blockade of membrane receptors or by interaction with intracellular signalling pathways. Cyclic adenosine 3',5'-monophosphate (cAMP) and cyclic guanosine 3',5'-monophosphate (cGMP) are two critical intracellular second messengers provided with strong inhibitory activity on fundamental platelet functions. Phosphodiesterases (PDEs), by catalysing the hydrolysis of cAMP and cGMP, limit the intracellular levels of cyclic nucleotides, thus regulating platelet function. The inhibition of PDEs may therefore exert a strong platelet inhibitory effect. Platelets possess three PDE isoforms (PDE2, PDE3 and PDE5), with different selectivity for cAMP and cGMP. Several nonselective or isoenzyme-selective PDE inhibitors have been developed, and some of them have entered clinical use as antiplatelet agents. This review focuses on the effect of PDE2, PDE3 and PDE5 inhibitors on platelet function and on the evidence for an antithrombotic action of some of them, and in particular of dipyridamole and cilostazol.
Collapse
Affiliation(s)
- Paolo Gresele
- Department of Internal Medicine, Section of Internal and Cardiovascular Medicine, University of Perugia, Italy.
| | | | | |
Collapse
|
36
|
Abstract
There are two primary modes of platelet inhibition: blockade of membrane receptors or neutralization of intracellular pathways. Both means of inhibition have proven benefits in the prevention and resolution of atherothrombotic events. With regard to intracellular inhibition, phosphodiesterases (PDEs) are fundamental for platelet function. Platelets possess several PDEs (PDE2, PDE3 and PDE5) that catalyze the hydrolysis of cyclic adenosine 3'-5'-monophosphate (cAMP) and cyclic guanosine 3'-5'-monophosphate (cGMP), thereby limiting the levels of intracellular nucleotides. PDE inhibitors, such as cilostazol and dipyridamole, dampen platelet function by increasing cAMP and cGMP levels. This review focuses on the roles of PDE inhibitors in modulating platelet function, with particular attention paid to drugs that have anti-platelet clinical indications.
Collapse
|
37
|
Schirmer SH, Degen A, Baumhäkel M, Custodis F, Schuh L, Kohlhaas M, Friedrich E, Bahlmann F, Kappl R, Maack C, Böhm M, Laufs U. Heart-rate reduction by If-channel inhibition with ivabradine restores collateral artery growth in hypercholesterolemic atherosclerosis. Eur Heart J 2011; 33:1223-31. [DOI: 10.1093/eurheartj/ehr255] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
38
|
Kevil CG, Kolluru GK, Pattillo CB, Giordano T. Inorganic nitrite therapy: historical perspective and future directions. Free Radic Biol Med 2011; 51:576-93. [PMID: 21619929 PMCID: PMC4414241 DOI: 10.1016/j.freeradbiomed.2011.04.042] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Revised: 04/26/2011] [Accepted: 04/27/2011] [Indexed: 12/24/2022]
Abstract
Over the past several years, investigators studying nitric oxide (NO) biology and metabolism have come to learn that the one-electron oxidation product of NO, nitrite anion, serves as a unique player in modulating tissue NO bioavailability. Numerous studies have examined how this oxidized metabolite of NO can act as a salvage pathway for maintaining NO equivalents through multiple reduction mechanisms in permissive tissue environments. Moreover, it is now clear that nitrite anion production and distribution throughout the body can act in an endocrine manner to augment NO bioavailability, which is important for physiological and pathological processes. These discoveries have led to renewed hope and efforts for an effective NO-based therapeutic agent through the unique action of sodium nitrite as an NO prodrug. More recent studies also indicate that sodium nitrate may also increase plasma nitrite levels via the enterosalivary circulatory system resulting in nitrate reduction to nitrite by microorganisms found within the oral cavity. In this review, we discuss the importance of nitrite anion in several disease models along with an appraisal of sodium nitrite therapy in the clinic, potential caveats of such clinical uses, and future possibilities for nitrite-based therapies.
Collapse
Affiliation(s)
- Christopher G Kevil
- Department of Pathology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71130, USA.
| | | | | | | |
Collapse
|
39
|
Grb-2-associated binder 1 (Gab1) regulates postnatal ischemic and VEGF-induced angiogenesis through the protein kinase A-endothelial NOS pathway. Proc Natl Acad Sci U S A 2011; 108:2957-62. [PMID: 21282639 DOI: 10.1073/pnas.1009395108] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The intracellular signaling mechanisms underlying postnatal angiogenesis are incompletely understood. Herein we show that Grb-2-associated binder 1 (Gab1) plays a critical role in ischemic and VEGF-induced angiogenesis. Endothelium-specific Gab1 KO (EGKO) mice displayed impaired angiogenesis in the ischemic hindlimb despite normal induction of VEGF expression. Matrigel plugs with VEGF implanted in EGKO mice induced fewer capillaries than those in control mice. The vessels and endothelial cells (ECs) derived from EGKO mice were defective in vascular sprouting and tube formation induced by VEGF. Biochemical analyses revealed a substantial reduction of endothelial NOS (eNOS) activation in Gab1-deficient vessels and ECs following VEGF stimulation. Interestingly, the phosphorylation of Akt, an enzyme known to promote VEGF-induced eNOS activation, was increased in Gab1-deficient vessels and ECs whereas protein kinase A (PKA) activity was significantly decreased. Introduction of an active form of PKA rescued VEGF-induced eNOS activation and tube formation in EGKO ECs. Reexpression of WT or mutant Gab1 molecules in EGKO ECs revealed requirement of Gab1/Shp2 association for the activation of PKA and eNOS. Taken together, these results identify Gab1 as a critical upstream signaling component in VEGF-induced eNOS activation and tube formation, which is dependent on PKA. Of note, this pathway is conserved in primary human ECs for VEGF-induced eNOS activation and tube formation, suggesting considerable potential in treatment of human ischemic diseases.
Collapse
|
40
|
Pattillo CB, Bir SC, Branch BG, Greber E, Shen X, Pardue S, Patel RP, Kevil CG. Dipyridamole reverses peripheral ischemia and induces angiogenesis in the Db/Db diabetic mouse hind-limb model by decreasing oxidative stress. Free Radic Biol Med 2011; 50:262-9. [PMID: 21070849 PMCID: PMC4413947 DOI: 10.1016/j.freeradbiomed.2010.10.714] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2010] [Revised: 10/23/2010] [Accepted: 10/28/2010] [Indexed: 11/28/2022]
Abstract
Dipyridamole anti-platelet therapy has previously been suggested to ameliorate chronic tissue ischemia in healthy animals. However, it is not known if dipyridamole therapy represents a viable approach to alleviating chronic peripheral tissue ischemia associated with type 2 diabetes. Here we examine the hypothesis that dipyridamole treatment restores reperfusion of chronic hind-limb ischemia in the murine B6.BKS-Lepr(db/db) diabetic model. Dipyridamole therapy quickly rectified ischemic hind-limb blood flow to near preligation levels within 3 days of the start of therapy. Restoration of ischemic tissue blood flow was associated with increased vascular density and endothelial cell proliferation observed only in ischemic limbs. Dipyridamole significantly increased total nitric oxide metabolite levels in tissue, which were not associated with changes in endothelial NO synthase expression or phosphorylation. Interestingly, dipyridamole therapy significantly decreased ischemic tissue superoxide and protein carbonyl levels, identifying a dominant antioxidant mechanistic response. Dipyridamole therapy also moderately reduced diabetic hyperglycemia and attenuated development of dyslipidemia over time. Together, these data reveal that dipyridamole therapy is an effective modality for the treatment of chronic tissue ischemia during diabetes and highlights the importance of dipyridamole antioxidant activity in restoring tissue NO bioavailability during diabetes.
Collapse
Affiliation(s)
| | - Shyamal C. Bir
- Department of Pathology, LSU Health Sciences Center-Shreveport, Shreveport, Louisiana
| | - Billy G. Branch
- Department of Pathology, LSU Health Sciences Center-Shreveport, Shreveport, Louisiana
| | - Eric Greber
- Department of Pathology, LSU Health Sciences Center-Shreveport, Shreveport, Louisiana
| | - Xinggui Shen
- Department of Pathology, LSU Health Sciences Center-Shreveport, Shreveport, Louisiana
| | - Sibile Pardue
- Department of Pathology, LSU Health Sciences Center-Shreveport, Shreveport, Louisiana
| | - Rakesh P. Patel
- Department of Pathology and Center for Free Radical Biology, University of Alabama-Birmingham, Birmingham, Alabama
| | - Christopher G. Kevil
- Department of Pathology, LSU Health Sciences Center-Shreveport, Shreveport, Louisiana
| |
Collapse
|
41
|
Uchiyama S, Ikeda Y, Urano Y, Horie Y, Yamaguchi T. The Japanese Aggrenox (Extended-Release Dipyridamole plus Aspirin) Stroke Prevention versus Aspirin Programme (JASAP) Study: A Randomized, Double-Blind, Controlled Trial. Cerebrovasc Dis 2011; 31:601-13. [DOI: 10.1159/000327035] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2010] [Accepted: 03/03/2011] [Indexed: 11/19/2022] Open
|
42
|
Pattillo CB, Fang K, Terracciano J, Kevil CG. Reperfusion of chronic tissue ischemia: nitrite and dipyridamole regulation of innate immune responses. Ann N Y Acad Sci 2010; 1207:83-8. [PMID: 20955430 DOI: 10.1111/j.1749-6632.2010.05737.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Chronic and intermittent ischemic vascular disorders represent a burgeoning clinical challenge. Previous studies have focused on the idea that therapeutic angiogenesis strategies could alleviate tissue ischemia; however, it is now appreciated that vascular disease is not simply limited to vascular wall cells but also influenced by simultaneously occurring inflammatory responses. Our laboratory has discovered that pharmacological treatment of permanent tissue ischemia with dipyridamole significantly augments ischemic tissue reperfusion, angiogenesis, and arteriogenesis over time. We have found that the beneficial effects of dipyridamole therapy are due to its ability to increase tissue nitric oxide bioavailability that corrects tissue redox imbalance. Importantly, we have also discovered that dipyridamole treatment invoking nitric oxide (NO) production significantly downregulates various innate immune response genes during chronic ischemic tissue injury. These findings demonstrate that dipyridamole-induced production of nitrite/NO significantly decreases inflammatory responses while increasing vascular growth in ischemic tissues.
Collapse
Affiliation(s)
- Christopher B Pattillo
- Department of Pathology, LSU Health Sciences Center-Shreveport, Shreveport, Louisiana, USA
| | | | | | | |
Collapse
|
43
|
Melani A, Cipriani S, Corti F, Pedata F. Effect of intravenous administration of dipyridamole in a rat model of chronic cerebral ischemia. Ann N Y Acad Sci 2010; 1207:89-96. [DOI: 10.1111/j.1749-6632.2010.05732.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
44
|
Pattillo CB, Bir S, Rajaram V, Kevil CG. Inorganic nitrite and chronic tissue ischaemia: a novel therapeutic modality for peripheral vascular diseases. Cardiovasc Res 2010; 89:533-41. [PMID: 20851809 DOI: 10.1093/cvr/cvq297] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Ischaemic tissue damage represents the ultimate form of tissue pathophysiology due to cardiovascular disease, which is the leading cause of morbidity and mortality across the globe. A significant amount of basic research and clinical investigation has been focused on identifying cellular and molecular pathways to alleviate tissue damage and dysfunction due to ischaemia and subsequent reperfusion. Over many years, the gaseous molecule nitric oxide (NO) has emerged as an important regulator of cardiovascular health as well as protector against tissue ischaemia and reperfusion injury. However, clinical translation of NO therapy for these pathophysiological conditions has not been realized for various reasons. Work from our laboratory and several others suggests that a new form of NO-associated therapy may be possible through the use of nitrite anion (sodium nitrite), a prodrug which can be reduced to NO in ischaemic tissues. In this manner, nitrite anion serves as a highly selective NO donor in ischaemic tissues without substantially altering otherwise normal tissue. This surprising and novel discovery has reinvigorated hopes for effectively restoring NO bioavailability in vulnerable tissues while continuing to reveal the complexity of NO biology and metabolism within the cardiovascular system. However, some concerns may exist regarding the effect of nitrite on carcinogenesis. This review highlights the emergence of nitrite anion as a selective NO prodrug for ischaemic tissue disorders and discusses the potential therapeutic utility of this agent for peripheral vascular disease.
Collapse
Affiliation(s)
- Christopher B Pattillo
- Department of Pathology and Cardiology, LSU Health Sciences Center-Shreveport, 1501 Kings Hwy, Shreveport, LA 71130, USA
| | | | | | | |
Collapse
|
45
|
Passer BJ, Cheema T, Zhou B, Wakimoto H, Zaupa C, Razmjoo M, Sarte J, Wu S, Wu CL, Noah JW, Li Q, Buolamwini JK, Yen Y, Rabkin SD, Martuza RL. Identification of the ENT1 antagonists dipyridamole and dilazep as amplifiers of oncolytic herpes simplex virus-1 replication. Cancer Res 2010; 70:3890-5. [PMID: 20424118 DOI: 10.1158/0008-5472.can-10-0155] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Oncolytic herpes simplex virus-1 (oHSV) vectors selectively replicate in tumor cells, where they kill through oncolysis while sparing normal cells. One of the drawbacks of oHSV vectors is their limited replication and spread to neighboring cancer cells. Here, we report the outcome of a high-throughput chemical library screen to identify small-molecule compounds that augment the replication of oHSV G47Delta. Of the 2,640-screened bioactives, 6 compounds were identified and subsequently validated for enhanced G47Delta replication. Two of these compounds, dipyridamole and dilazep, interfered with nucleotide metabolism by potently and directly inhibiting the equilibrative nucleoside transporter-1 (ENT1). Replicative amplification promoted by dipyridamole and dilazep were dependent on HSV mutations in ICP6, the large subunit of ribonucleotide reductase. Our results indicate that ENT1 antagonists augment oHSV replication in tumor cells by increasing cellular ribonucleoside activity.
Collapse
Affiliation(s)
- Brent J Passer
- Departments of Neurosurgery and Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|