1
|
Boulares A, Jdidi H, Bragazzi NL. Impact of Mouthwash-Induced Oral Microbiome Disruption on Alzheimer's Disease Risk: A Perspective Review. Int Dent J 2024:S0020-6539(24)00197-7. [PMID: 39379282 DOI: 10.1016/j.identj.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 10/10/2024] Open
Abstract
The widespread use of mouthwashes, particularly those containing chlorhexidine (CHX), has raised concerns about their impact on the oral microbiome and potential systemic health effects. This perspective review examines the current evidence linking CHX mouthwash use to disruptions in the oral microbiome and explores the potential indirect implications for Alzheimer's disease (AD) risk. CHX mouthwash is effective in reducing dental plaque and gingival inflammation, but it also significantly alters the composition of the oral microbiome, decreasing the abundance of nitrate-reducing bacteria critical for nitric oxide (NO) production. This disruption can lead to increased blood pressure, a major risk factor for AD. Given the established connection between hypertension and AD, the long-term use of CHX mouthwash may indirectly contribute to the onset of AD. However, the relationship between CHX mouthwash use and AD remains largely indirect, necessitating further longitudinal and cohort studies to investigate whether a direct causal link exists. The review aims to highlight the importance of maintaining a balanced oral microbiome for both oral and systemic health and calls for more research into safer oral hygiene practices and their potential impacts on neurodegenerative disease risk.
Collapse
Affiliation(s)
- Ayoub Boulares
- Laboratory Mobility, Faculty of Sport Sciences-STAPS, Aging & Exercise-ER20296, University of Poitiers, Poitiers, France
| | - Hela Jdidi
- Laboratory Mobility, Faculty of Sport Sciences-STAPS, Aging & Exercise-ER20296, University of Poitiers, Poitiers, France
| | - Nicola Luigi Bragazzi
- Department of Food and Drugs, Human Nutrition Unit (HNU), Medical School, University of Parma, Parma, Italy; Laboratory for Industrial and Applied Mathematics (LIAM), Department of Mathematics and Statistics, York University, Toronto, Ontario, Canada.
| |
Collapse
|
2
|
Ejikeme C, Safdar Z. Exploring the pathogenesis of pulmonary vascular disease. Front Med (Lausanne) 2024; 11:1402639. [PMID: 39050536 PMCID: PMC11267418 DOI: 10.3389/fmed.2024.1402639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 06/26/2024] [Indexed: 07/27/2024] Open
Abstract
Pulmonary hypertension (PH) is a complex cardiopulmonary disorder impacting the lung vasculature, resulting in increased pulmonary vascular resistance that leads to right ventricular dysfunction. Pulmonary hypertension comprises of 5 groups (PH group 1 to 5) where group 1 pulmonary arterial hypertension (PAH), results from alterations that directly affect the pulmonary arteries. Although PAH has a complex pathophysiology that is not completely understood, it is known to be a multifactorial disease that results from a combination of genetic, epigenetic and environmental factors, leading to a varied range of symptoms in PAH patients. PAH does not have a cure, its incidence and prevalence continue to increase every year, resulting in higher morbidity and mortality rates. In this review, we discuss the different pathologic mechanisms with a focus on epigenetic modifications and their roles in the development and progression of PAH. These modifications include DNA methylation, histone modifications, and microRNA dysregulation. Understanding these epigenetic modifications will improve our understanding of PAH and unveil novel therapeutic targets, thus steering research toward innovative treatment strategies.
Collapse
Affiliation(s)
| | - Zeenat Safdar
- Department of Pulmonary-Critical Care Medicine, Houston Methodist Lung Center, Houston Methodist Hospital, Houston, TX, United States
| |
Collapse
|
3
|
Krishtopaytis E, Ampnti SA, Obeidat M, Ramahi N, Lane J, Toth D, Paul D, Tonelli AR. Can Inhaled Nitric Oxide Response Predict Tolerance to Therapies and Survival in Patients With Combined Precapillary and Postcapillary Pulmonary Hypertension? Am J Cardiol 2023; 207:363-369. [PMID: 37778225 DOI: 10.1016/j.amjcard.2023.09.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/04/2023] [Accepted: 09/06/2023] [Indexed: 10/03/2023]
Abstract
Inhaled nitric oxide (iNO) relaxes the pulmonary circulation and variably increases the left ventricular preload and pulmonary artery wedge pressure (PAWP)-hemodynamic information that may help guide treatment decisions and assess prognosis in patients with combined precapillary and postcapillary pulmonary hypertension (PH). We included consecutive patients with combined precapillary and postcapillary PH (mean pulmonary artery pressure >20 mm Hg, PAWP >15 mm Hg, and pulmonary vascular resistance [PVR] >2 Woods unit [WU]) who underwent right-sided cardiac catheterization with iNO at the Cleveland Clinic Pulmonary Vascular Disease program between 2017 and 2022. We included 104 patients with baseline PAWP and PVR of 22.2 ± 4.2 mm Hg and 6.1 ± 3.2 WU, respectively. Pulmonary arterial hypertension (PAH) with postcapillary component and PH left heart disease with precapillary component were identified in 27 (26%) and 77 patients (74%), respectively. No side effects were noted during the administration of iNO. During iNO, the PVR decreased 1.1 ± 1.4 WU and the PAWP increased 1.3 ± 3.7 mm Hg. A more pronounced increase in PAWP with iNO was associated with a decrease in PVR (R -0.35, p <0.001) and increase in stroke volume (R 0.20, p = 0.046). Tolerance to PAH-specific medications, overall survival, and heart failure hospitalizations were not significantly associated with the change in PAWP or PVR with iNO. In conclusion, in patients with combined precapillary and postcapillary PH, iNO challenge is safe and caused a significant decrease in PVR, with an increase in PAWP. The changes in PAWP and PVR during iNO administration were not associated with tolerance to PAH-specific medications, heart failure-related hospitalization, or survival.
Collapse
Affiliation(s)
| | | | | | - Noor Ramahi
- Departments of Hospital Medicine, Fairview Hospital
| | - James Lane
- Pulmonary, Allergy and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, Cleveland, Ohio
| | - David Toth
- Pulmonary, Allergy and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, Cleveland, Ohio
| | - Deborah Paul
- Pulmonary, Allergy and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, Cleveland, Ohio
| | - Adriano Roberto Tonelli
- Pulmonary, Allergy and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, Cleveland, Ohio.
| |
Collapse
|
4
|
Alqarni AA, Aldhahir AM, Alghamdi SA, Alqahtani JS, Siraj RA, Alwafi H, AlGarni AA, Majrshi MS, Alshehri SM, Pang L. Role of prostanoids, nitric oxide and endothelin pathways in pulmonary hypertension due to COPD. Front Med (Lausanne) 2023; 10:1275684. [PMID: 37881627 PMCID: PMC10597708 DOI: 10.3389/fmed.2023.1275684] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 09/19/2023] [Indexed: 10/27/2023] Open
Abstract
Pulmonary hypertension (PH) due to chronic obstructive pulmonary disease (COPD) is classified as Group 3 PH, with no current proven targeted therapies. Studies suggest that cigarette smoke, the most risk factor for COPD can cause vascular remodelling and eventually PH as a result of dysfunction and proliferation of pulmonary artery smooth muscle cells (PASMCs) and pulmonary artery endothelial cells (PAECs). In addition, hypoxia is a known driver of pulmonary vascular remodelling in COPD, and it is also thought that the presence of hypoxia in patients with COPD may further exaggerate cigarette smoke-induced vascular remodelling; however, the underlying cause is not fully understood. Three main pathways (prostanoids, nitric oxide and endothelin) are currently used as a therapeutic target for the treatment of patients with different groups of PH. However, drugs targeting these three pathways are not approved for patients with COPD-associated PH due to lack of evidence. Thus, this review aims to shed light on the role of impaired prostanoids, nitric oxide and endothelin pathways in cigarette smoke- and hypoxia-induced pulmonary vascular remodelling and also discusses the potential of using these pathways as therapeutic target for patients with PH secondary to COPD.
Collapse
Affiliation(s)
- Abdullah A. Alqarni
- Department of Respiratory Therapy, Faculty of Medical Rehabilitation Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Respiratory Therapy Unit, King Abdulaziz University Hospital, Jeddah, Saudi Arabia
| | - Abdulelah M. Aldhahir
- Respiratory Therapy Department, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Sara A. Alghamdi
- Respiratory Care Department, Al Murjan Hospital, Jeddah, Saudi Arabia
| | - Jaber S. Alqahtani
- Department of Respiratory Care, Prince Sultan Military College of Health Sciences, Dammam, Saudi Arabia
| | - Rayan A. Siraj
- Department of Respiratory Care, College of Applied Medical Sciences, King Faisal University, Al Ahsa, Saudi Arabia
| | - Hassan Alwafi
- Faculty of Medicine, Umm Al-Qura University, Mecca, Saudi Arabia
| | - Abdulkareem A. AlGarni
- King Abdulaziz Hospital, The Ministry of National Guard Health Affairs, Al Ahsa, Saudi Arabia
- King Saud bin Abdulaziz University for Health Sciences, College of Applied Medical Sciences, Al Ahsa, Saudi Arabia
| | - Mansour S. Majrshi
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Respiratory Medicine, Royal Brompton Hospital, London, United Kingdom
| | - Saad M. Alshehri
- Department of Respiratory Therapy, King Fahad General Hospital, Jeddah, Saudi Arabia
| | - Linhua Pang
- Respiratory Medicine Research Group, Academic Unit for Translational Medical Sciences, University of Nottingham School of Medicine, Nottingham, United Kingdom
| |
Collapse
|
5
|
Douglass MS, Kaplowitz MR, Zhang Y, Fike CD. Impact of l-citrulline on nitric oxide signaling and arginase activity in hypoxic human pulmonary artery endothelial cells. Pulm Circ 2023; 13:e12221. [PMID: 37063746 PMCID: PMC10091859 DOI: 10.1002/pul2.12221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 03/28/2023] [Accepted: 03/30/2023] [Indexed: 04/18/2023] Open
Abstract
Impaired nitric oxide (NO) signaling contributes to the development of pulmonary hypertension (PH). The l-arginine precursor, l-citrulline, improves NO signaling and has therapeutic potential in PH. However, there is evidence that l-citrulline might increase arginase activity, which in turn, has been shown to contribute to PH. Our major purpose was to determine if l-citrulline increases arginase activity in hypoxic human pulmonary artery endothelial cells (PAECs). In addition, to avoid potential adverse effects from high dose l-citrulline monotherapy, we evaluated whether the effect on NO signaling is greater using co-treatment with l-citrulline and another agent that improves NO signaling, folic acid, than either alone. Arginase activity was measured in human PAECs cultured under hypoxic conditions in the presence of l-citrulline (0-1 mM). NO production and endothelial nitric oxide synthase (eNOS) coupling, as assessed by eNOS dimer-to-monomer ratios, were measured in PAECs treated with l-citrulline and/or folic acid (0.2 μM). Arginase activity increased in hypoxic PAECs treated with 1 mM but not with either 0.05 or 0.1 mM l-citrulline. Co-treatment with folic acid and 0.1 mM l-citrulline increased NO production and eNOS dimer-to-monomer ratios more than treatment with either alone. The potential to increase arginase activity suggests that there might be plasma l-citrulline concentrations that should not be exceeded when using l-citrulline to treat PH. Rather than progressively increasing the dose of l-citrulline as a monotherapy, co-therapy with l-citrulline and folic acid merits consideration, due to the possibility of achieving efficacy at lower doses and minimizing side effects.
Collapse
Affiliation(s)
| | | | - Yongmei Zhang
- Department of PediatricsUniversity of UtahSalt Lake CityUtahUSA
| | - Candice D. Fike
- Department of PediatricsUniversity of UtahSalt Lake CityUtahUSA
| |
Collapse
|
6
|
Körbelin J, Klein J, Matuszcak C, Runge J, Harbaum L, Klose H, Hennigs JK. Transcription factors in the pathogenesis of pulmonary arterial hypertension-Current knowledge and therapeutic potential. Front Cardiovasc Med 2023; 9:1036096. [PMID: 36684555 PMCID: PMC9853303 DOI: 10.3389/fcvm.2022.1036096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 11/21/2022] [Indexed: 01/09/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a disease characterized by elevated pulmonary vascular resistance and pulmonary artery pressure. Mortality remains high in severe cases despite significant advances in management and pharmacotherapy. Since currently approved PAH therapies are unable to significantly reverse pathological vessel remodeling, novel disease-modifying, targeted therapeutics are needed. Pathogenetically, PAH is characterized by vessel wall cell dysfunction with consecutive remodeling of the pulmonary vasculature and the right heart. Transcription factors (TFs) regulate the process of transcribing DNA into RNA and, in the pulmonary circulation, control the response of pulmonary vascular cells to macro- and microenvironmental stimuli. Often, TFs form complex protein interaction networks with other TFs or co-factors to allow for fine-tuning of gene expression. Therefore, identification of the underlying molecular mechanisms of TF (dys-)function is essential to develop tailored modulation strategies in PAH. This current review provides a compendium-style overview of TFs and TF complexes associated with PAH pathogenesis and highlights their potential as targets for vasculoregenerative or reverse remodeling therapies.
Collapse
Affiliation(s)
- Jakob Körbelin
- ENDomics Lab, Department of Medicine, Center of Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,*Correspondence: Jakob Körbelin,
| | - Julius Klein
- ENDomics Lab, Department of Medicine, Center of Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,Division of Pneumology and Center for Pulmonary Arterial Hypertension Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christiane Matuszcak
- ENDomics Lab, Department of Medicine, Center of Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,Division of Pneumology and Center for Pulmonary Arterial Hypertension Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Johannes Runge
- ENDomics Lab, Department of Medicine, Center of Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,Division of Pneumology and Center for Pulmonary Arterial Hypertension Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lars Harbaum
- Division of Pneumology and Center for Pulmonary Arterial Hypertension Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hans Klose
- Division of Pneumology and Center for Pulmonary Arterial Hypertension Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan K. Hennigs
- ENDomics Lab, Department of Medicine, Center of Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,Division of Pneumology and Center for Pulmonary Arterial Hypertension Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,Jan K. Hennigs,
| |
Collapse
|
7
|
Fike CD, Avachat C, Birnbaum AK, Aschner JL, Sherwin CM. Pharmacokinetics of L-Citrulline in Neonates at Risk of Developing Bronchopulmonary Dysplasia-Associated Pulmonary Hypertension. Paediatr Drugs 2023; 25:87-96. [PMID: 36316628 PMCID: PMC10039462 DOI: 10.1007/s40272-022-00542-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/02/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND Options to treat pulmonary hypertension (PH) in neonates with bronchopulmonary dysplasia (BPD) are few and largely ineffective. Improving the bioavailability of nitric oxide (NO) might be an efficacious treatment for BPD-PH. When administered orally, the NO-L-arginine precursor, L-citrulline, increases NO production in children and adults, however, pharmacokinetic (PK) studies of oral L-citrulline have not been performed in infants and children. OBJECTIVES This study characterized the PK of enterally administered L-citrulline in neonates at risk of developing BPD-PH to devise a model-informed dosing strategy. METHODS AND RESULTS Ten premature neonates (≤ 28 weeks gestation) were administered a single dose of 150 mg/kg (powder form solubilized in sterile water) oral L-citrulline at 32 ± 1 weeks postmenstrual age. Due to the need to limit blood draws, time windows were used to maximize the sampling over the dosing interval by assigning neonates to one of two groups (ii) samples collected pre-dose and at 1- and 2.5-h post-dose, and (ii) pre-dose and 0.25- and 3-h post-dose. The L-arginine concentrations (µmol/L) and the L-citrulline (µmol/L) plasma concentration-time data were evaluated using non-compartmental analysis (Phoenix WinNonlin version 8.1). Optimal dosage strategies were derived using a simulation-based methodology. Simulated doses of 51.5 mg or 37.5 mg/kg given four times a day produced steady-state concentrations close to a target of 50 µmol/L. The volume of distribution (V/F) and clearance (CL/F) were 302.89 ml and 774.96 ml/h, respectively, with the drug exhibiting a half-life of 16 minutes. The AUC from the time of dosing to the time of last concentration was 1473.3 h*μmol/L, with Cmax and Tmax of 799 μmol/L and 1.55 h, respectively. CONCLUSION This is the first PK study in neonates presenting data that can be used to inform dosing strategies in future randomized controlled trials evaluating enteral L-citrulline as a potential treatment to reduce PH associated with BPD in premature neonates. REGISTRATION Clinical trials.gov Identifier: NCT03542812.
Collapse
Affiliation(s)
- Candice D Fike
- Department of Pediatrics, The University of Utah Health, Salt Lake City, UT, USA
| | - Charul Avachat
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Angela K Birnbaum
- Department of Pediatrics, The University of Utah Health, Salt Lake City, UT, USA
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Judy L Aschner
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Pediatrics, Hackensack Meridian School of Medicine, Nutley, NJ, USA
| | - Catherine M Sherwin
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA.
- Department of Pediatrics, Wright State University Boonshoft School of Medicine, Dayton, OH, USA.
- Dayton Children's Hospital, Dayton, OH, USA.
| |
Collapse
|
8
|
Bioinformatic Exploration of Hub Genes and Potential Therapeutic Drugs for Endothelial Dysfunction in Hypoxic Pulmonary Hypertension. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:3677532. [PMID: 36483920 PMCID: PMC9723419 DOI: 10.1155/2022/3677532] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 11/02/2022] [Accepted: 11/15/2022] [Indexed: 11/29/2022]
Abstract
Hypoxic pulmonary hypertension (HPH) is a fatal chronic pulmonary circulatory disease, characterized by hypoxic pulmonary vascular constriction and remodeling. Studies performed to date have confirmed that endothelial dysfunction plays crucial roles in HPH, while the underlying mechanisms have not been fully revealed. The microarray dataset GSE11341 was downloaded from the Gene Expression Omnibus (GEO) database to identify differentially expressed genes (DEGs) between hypoxic and normoxic microvascular endothelial cell, followed by Gene Ontology (GO) annotation/Kyoto Encyclopedia of Genes and Genomes (KEGG), Gene Set Enrichment Analysis (GSEA) pathway enrichment analysis, and protein-protein interaction (PPI) network construction. Next, GSE160255 and RT-qPCR were used to validate hub genes. Meanwhile, GO/KEGG and GSEA were performed for each hub gene to uncover the potential mechanism. A nomogram based on hub genes was established. Furthermore, mRNA-miRNA network was predicted by miRNet, and the Connectivity Map (CMAP) database was in use to identify similarly acting therapeutic candidates. A total of 148 DEGs were screened in GSE11341, and three hub genes (VEGFA, CDC25A, and LOX) were determined and validated via GSE160255 and RT-qPCR. Abnormalities in the pathway of vascular smooth muscle contraction, lysosome, and glycolysis might play important roles in HPH pathogenesis. The hub gene-miRNA network showed that hsa-mir-24-3p, hsa-mir-124-3p, hsa-mir-195-5p, hsa-mir-146a-5p, hsa-mir-155-5p, and hsa-mir-23b-3p were associated with HPH. And on the basis of the identified hub genes, a practical nomogram is developed. To repurpose known and therapeutic drugs, three candidate compounds (procaterol, avanafil, and lestaurtinib) with a high level of confidence were obtained from the CMAP database. Taken together, the identification of these three hub genes, enrichment pathways, and potential therapeutic drugs might have important clinical implications for HPH diagnosis and treatment.
Collapse
|
9
|
Molecular Pathways in Pulmonary Arterial Hypertension. Int J Mol Sci 2022; 23:ijms231710001. [PMID: 36077398 PMCID: PMC9456336 DOI: 10.3390/ijms231710001] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/20/2022] [Accepted: 08/23/2022] [Indexed: 11/16/2022] Open
Abstract
Pulmonary arterial hypertension is a multifactorial, chronic disease process that leads to pulmonary arterial endothelial dysfunction and smooth muscular hypertrophy, resulting in impaired pliability and hemodynamics of the pulmonary vascular system, and consequent right ventricular dysfunction. Existing treatments target limited pathways with only modest improvement in disease morbidity, and little or no improvement in mortality. Ongoing research has focused on the molecular basis of pulmonary arterial hypertension and is going to be important in the discovery of new treatments and genetic pathways involved. This review focuses on the molecular pathogenesis of pulmonary arterial hypertension.
Collapse
|
10
|
Parajuli N, Kosanovic D. Editorial: Oxidative Stress in Cardiovascular Diseases and Pulmonary Hypertension. Front Cardiovasc Med 2022; 9:868988. [PMID: 35402568 PMCID: PMC8983953 DOI: 10.3389/fcvm.2022.868988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 02/28/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Nirmal Parajuli
- Immunology Research Program, Henry Ford Health System, Detroit, MI, United States
- *Correspondence: Nirmal Parajuli
| | - Djuro Kosanovic
- Department of Pulmonology, I. M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
- Djuro Kosanovic
| |
Collapse
|
11
|
Torbic H, Hohlfelder B, Krishnan S, Tonelli AR. A Review of Pulmonary Arterial Hypertension Treatment in Extracorporeal Membrane Oxygenation: A Case Series of Adult Patients. J Cardiovasc Pharmacol Ther 2022; 27:10742484211069005. [PMID: 35006031 DOI: 10.1177/10742484211069005] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Little data is published describing the use of medications prescribed for pulmonary arterial hypertension (PAH) in patients receiving extracorporeal membrane oxygenation (ECMO). Even though many patients with PAH may require ECMO as a bridge to transplant or recovery, little is reported regarding the use of PAH medications in this setting. METHODS This retrospective case series summarizes the clinical experience of 8 patients with PAH receiving ECMO and reviews medication management in the setting of ECMO. RESULTS Eight PAH patients, 5 of whom were female, ranging in age from 21 to 61 years old, were initiated on ECMO. Veno-arterial (VA) ECMO was used in 4 patients, veno-venous (VV) ECMO and hybrid ECMO configurations in 2 patients respectively. Common indications for ECMO included cardiogenic shock, bridge to transplant, and cardiac arrest. All patients were on intravenous (IV) prostacyclin therapy at baseline. Refractory hypotension was noted in 7 patients of whom 5 patients required downtitration or discontinuation of baseline PAH therapies. Three patients had continuous inhaled epoprostenol added during their time on ECMO. In patients who were decannulated from ECMO, PAH therapies were typically resumed or titrated back to baseline dosages. One patient required no adjustment in PAH therapy while on ECMO. Two patients were not able to be decannulated from ECMO. CONCLUSION The treatment of critically ill PAH patients is challenging given a variety of factors that could affect PAH drug concentrations. In particular, PAH patients on prostacyclin analogues placed on VA ECMO appear to have pronounced systemic vasodilation requiring vasopressors which is alleviated by temporarily reducing the intravenous prostacyclin dose. Patients should be closely monitored for potential need for rapid titrations in prostacyclin therapy to maintain hemodynamic stability.
Collapse
Affiliation(s)
- Heather Torbic
- 2569Department of Pharmacy, Cleveland Clinic, Cleveland, OH, USA
| | | | - Sudhir Krishnan
- Department of Critical Care Medicine, 2569Respiratory Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Adriano R Tonelli
- Department of Pulmonary and Critical Care Medicine, 2569Respiratory Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
12
|
Condon DF, Agarwal S, Chakraborty A, Auer N, Vazquez R, Patel H, Zamanian RT, de Jesus Perez VA, Condon DF. "NOVEL MECHANISMS TARGETED BY DRUG TRIALS IN PULMONARY ARTERIAL HYPERTENSION". Chest 2021; 161:1060-1072. [PMID: 34655569 PMCID: PMC9005865 DOI: 10.1016/j.chest.2021.10.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 09/21/2021] [Accepted: 10/04/2021] [Indexed: 10/20/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare disease associated with abnormally elevated pulmonary pressures and right heart failure resulting in high morbidity and mortality. While PAH prognosis has improved with the introduction of pulmonary vasodilators, disease progression remains a major problem. Given that available therapies are inadequate for preventing small vessel loss and obstruction, there is an active interest in identifying drugs capable of targeting angiogenesis and mechanisms involved in regulation of cell growth and fibrosis. Among the mechanisms linked to PAH pathogenesis, recent preclinical studies have identified promising compounds that are currently being tested in clinical trials. These drugs target seven of the major mechanisms associated with PAH pathogenesis: BMP signaling, tyrosine kinase receptors, estrogen metabolism, extracellular matrix, angiogenesis, epigenetics, and serotonin metabolism. In this review, we will discuss the preclinical studies that led to prioritization of these mechanisms and will discuss recently completed and ongoing phase 2/3 trials using novel interventions such as sotatercept, anastrozole, rodatristat ethyl, tyrosine kinase inhibitors, and endothelial progenitor cells among others. We anticipate that the next generation of compounds will build upon the success of the current standard of care and improve clinical outcomes and quality of life of patients afflicted with PAH.
Collapse
Affiliation(s)
- David F Condon
- Division of Pulmonary Allergy and Critical Care Medicine, Wall Center for Cardiopulmonary Research, Stanford University, Stanford, CA
| | - Stuti Agarwal
- Division of Pulmonary Allergy and Critical Care Medicine, Wall Center for Cardiopulmonary Research, Stanford University, Stanford, CA
| | - Ananya Chakraborty
- Division of Pulmonary Allergy and Critical Care Medicine, Wall Center for Cardiopulmonary Research, Stanford University, Stanford, CA
| | - Natasha Auer
- Division of Pulmonary Allergy and Critical Care Medicine, Wall Center for Cardiopulmonary Research, Stanford University, Stanford, CA
| | - Rocio Vazquez
- Division of Pulmonary Allergy and Critical Care Medicine, Wall Center for Cardiopulmonary Research, Stanford University, Stanford, CA
| | - Hiral Patel
- Division of Pulmonary Allergy and Critical Care Medicine, Wall Center for Cardiopulmonary Research, Stanford University, Stanford, CA
| | - Roham T Zamanian
- Division of Pulmonary Allergy and Critical Care Medicine, Wall Center for Cardiopulmonary Research, Stanford University, Stanford, CA
| | - Vinicio A de Jesus Perez
- Division of Pulmonary Allergy and Critical Care Medicine, Wall Center for Cardiopulmonary Research, Stanford University, Stanford, CA.
| | | |
Collapse
|
13
|
Janaszak-Jasiecka A, Siekierzycka A, Płoska A, Dobrucki IT, Kalinowski L. Endothelial Dysfunction Driven by Hypoxia-The Influence of Oxygen Deficiency on NO Bioavailability. Biomolecules 2021; 11:biom11070982. [PMID: 34356605 PMCID: PMC8301841 DOI: 10.3390/biom11070982] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/25/2021] [Accepted: 07/02/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death worldwide. The initial stage of CVDs is characterized by endothelial dysfunction, defined as the limited bioavailability of nitric oxide (NO). Thus, any factors that interfere with the synthesis or metabolism of NO in endothelial cells are involved in CVD pathogenesis. It is well established that hypoxia is both the triggering factor as well as the accompanying factor in cardiovascular disease, and diminished tissue oxygen levels have been reported to influence endothelial NO bioavailability. In endothelial cells, NO is produced by endothelial nitric oxide synthase (eNOS) from L-Arg, with tetrahydrobiopterin (BH4) as an essential cofactor. Here, we discuss the mechanisms by which hypoxia affects NO bioavailability, including regulation of eNOS expression and activity. What is particularly important is the fact that hypoxia contributes to the depletion of cofactor BH4 and deficiency of substrate L-Arg, and thus elicits eNOS uncoupling-a state in which the enzyme produces superoxide instead of NO. eNOS uncoupling and the resulting oxidative stress is the major driver of endothelial dysfunction and atherogenesis. Moreover, hypoxia induces impairment in mitochondrial respiration and endothelial cell activation; thus, oxidative stress and inflammation, along with the hypoxic response, contribute to the development of endothelial dysfunction.
Collapse
Affiliation(s)
- Anna Janaszak-Jasiecka
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, 80-211 Gdansk, Poland; (A.J.-J.); (A.S.); (A.P.)
- Biobanking and Biomolecular Resources Research Infrastructure Poland (BBMRI.pl), 80-211 Gdansk, Poland
| | - Anna Siekierzycka
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, 80-211 Gdansk, Poland; (A.J.-J.); (A.S.); (A.P.)
- Laboratory of Trace Elements Neurobiology, Institute of Pharmacology, Polish Academy of Sciences, 31-343 Krakow, Poland
| | - Agata Płoska
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, 80-211 Gdansk, Poland; (A.J.-J.); (A.S.); (A.P.)
- Biobanking and Biomolecular Resources Research Infrastructure Poland (BBMRI.pl), 80-211 Gdansk, Poland
| | - Iwona T. Dobrucki
- University of Illinois at Urbana-Champaign Beckman Institute for Advanced Science and Technology, 405 N Mathews Ave, MC-251, Urbana, IL 61801, USA;
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, 80-211 Gdansk, Poland; (A.J.-J.); (A.S.); (A.P.)
- Biobanking and Biomolecular Resources Research Infrastructure Poland (BBMRI.pl), 80-211 Gdansk, Poland
- BioTechMed Centre, Department of Mechanics of Materials and Structures, Gdansk University of Technology, 80-233 Gdansk, Poland
- Correspondence:
| |
Collapse
|
14
|
Qaiser KN, Tonelli AR. Novel Treatment Pathways in Pulmonary Arterial Hypertension. Methodist Debakey Cardiovasc J 2021; 17:106-114. [PMID: 34326930 PMCID: PMC8298123 DOI: 10.14797/cbhs2234] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/18/2020] [Indexed: 12/21/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe and progressive vascular disease characterized by pulmonary vascular remodeling, proliferation, and inflammation. Despite the availability of effective treatments, PAH may culminate in right ventricular failure and death. Currently approved medications act through three well-characterized pathways: the nitric oxide, endothelin, and prostacyclin pathways. Ongoing research efforts continue to expand our understanding of the molecular pathogenesis of this complex and multifactorial disease. Based on recent discoveries in the pathobiology of PAH, several new treatments are being developed and tested with the goal of modifying the disease process and ultimately improving the long-term prognosis.
Collapse
|
15
|
Sleep apnea and pulmonary hypertension: A riddle waiting to be solved. Pharmacol Ther 2021; 227:107935. [PMID: 34171327 DOI: 10.1016/j.pharmthera.2021.107935] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 06/14/2021] [Accepted: 06/17/2021] [Indexed: 02/02/2023]
Abstract
Obstructive sleep apnea (OSA) is an under-recognized yet highly prevalent disease that has major implications to cardiovascular health. Pulmonary hypertension (pH) is less common but none the less a fatal condition. The association of OSA and PH is a known but not well understood phenomenon. Furthermore, the relationship appears to be bi-directional with limited understanding of the mechanism(s) driving the processes. PH in OSA has real time consequences as it has been shown to increase mortality. Limited data suggests that treatment with continuous positive pressure therapy may be beneficial and reduce pulmonary pressure. In this review, we discuss current data on prevalence of PH in OSA and vice versa. We also explore the pathophysiology of this relationship and a proposed mechanism for their connection. Finally, we address the treatment of OSA with CPAP and its impact on pulmonary pressures. Gaps in knowledge and future research potential are illustrated and discoursed.
Collapse
|
16
|
Yao K, Wang S, Gaowa N, Huang S, Li S, Shao W. Identification of the molecular mechanisms underlying brisket disease in Holstein heifers via microbiota and metabolome analyses. AMB Express 2021; 11:86. [PMID: 34185184 PMCID: PMC8241945 DOI: 10.1186/s13568-021-01246-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 06/01/2021] [Indexed: 11/10/2022] Open
Abstract
Brisket disease (BD) is common among Holstein heifers in high-altitude environments, and this disease may result in serious economic loss. At present, no effective treatment is available for brisket disease. In this study, liver and cecum samples were collected from five heifers with BD and five healthy heifers (HH) for analyses of the metabolome and microbiota. The mean pulmonary arterial pressure and systolic blood pressure were significantly higher in BD heifers, whereas the average breathing rate, blood oxygen saturation, and glucose level were significantly lower in BD group than in the HH group. Further, 16S rDNA data showed that the abundance of Firmicutes was significantly lower and that of Bacteroidetes was significantly higher in BD group than in the HH group. At the genus level, the BD group heifers harbored fewer Ruminococcaceae and Lachnospiraceae than the HH group. Several metabolites, including beta-D-fructose, D-ribose, 1,4-beta-D-glucan, sucrose, and glucose-6-phosphate were present at low levels in BD heifers. Moreover, the mean pulmonary arterial pressure was negatively correlated with beta-D-fructose (r = - 0.74; P = 0.013), D-ribose (r = - 0.72; P = 0.018), and acetyl-tyrosine-ethyl-ester (r = - 0.71; P = 0.022). We also found that mean pulmonary arterial pressure was negatively correlated with most of the genera, including those in the families of Lachnospiraceae and Ruminococcaceae. In summary, the decreased levels of metabolites and microbial genera might affect BD by limiting the energy supply. This study may help us better understand the role of the microbiota in BD and provide new insights into the management of feeding to decrease the rate of BD in Holstein dairy cows in the Qinghai-Tibetan plateau.
Collapse
|
17
|
Aulak KS, Al Abdi S, Li L, Crabb JS, Ghosh A, Willard B, Stuehr DJ, Crabb JW, Dweik RA, Tonelli AR. Disease-specific platelet signaling defects in idiopathic pulmonary arterial hypertension. Am J Physiol Lung Cell Mol Physiol 2021; 320:L739-L749. [PMID: 33596129 PMCID: PMC8174825 DOI: 10.1152/ajplung.00500.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 01/29/2021] [Accepted: 02/15/2021] [Indexed: 12/13/2022] Open
Abstract
Idiopathic pulmonary arterial hypertension (IPAH) is a rapidly progressive disease with several treatment options. Long-term mortality remains high with great heterogeneity in treatment response. Even though most of the pathology of IPAH is observed in the lung, there is systemic involvement. Platelets from patients with IPAH have characteristic metabolic shifts and defects in activation; therefore, we investigated whether they could be used to identify other disease-specific abnormalities. We used proteomics to investigate protein expression changes in platelets from patients with IPAH compared with healthy controls. Key abnormalities of nitric oxide pathway were tested in platelets from a larger cohort of unique patients with IPAH. Platelets showed abnormalities in the prostacyclin and nitric oxide pathways, which are dysregulated in IPAH and hence targets of therapy. We detected reduced expression of G protein αs and increased expression of the regulatory subunits of the cAMP-dependent protein kinase (PKA) type II isoforms, supporting an overall decrease in the activation of the prostacyclin pathway. We noted reduced levels of the soluble guanylate cyclase (sGC) subunits and increased expression of the phosphodiesterase type 5 A (PDE5A), conditions that affect the response to nitric oxide. Ensuing analysis of 38 unique patients with IPAH demonstrated considerable variation in the levels and specific activity of sGC, a finding with novel implications for personalized therapy. Platelets have some of the characteristic vasoactive signal abnormalities seen in IPAH and may provide comprehensive ex vivo mechanistic information to direct therapeutic decisions.
Collapse
Affiliation(s)
- Kulwant S Aulak
- Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Sami Al Abdi
- Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Ling Li
- Proteomics Shared Laboratory Resource, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Jack S Crabb
- Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio
| | - Arnab Ghosh
- Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Belinda Willard
- Proteomics Shared Laboratory Resource, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Dennis J Stuehr
- Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - John W Crabb
- Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio
| | - Raed A Dweik
- Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
- Department of Pulmonary, Allergy and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, Cleveland, Ohio
| | - Adriano R Tonelli
- Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
- Department of Pulmonary, Allergy and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
18
|
Rai N, Shihan M, Seeger W, Schermuly RT, Novoyatleva T. Genetic Delivery and Gene Therapy in Pulmonary Hypertension. Int J Mol Sci 2021; 22:ijms22031179. [PMID: 33503992 PMCID: PMC7865388 DOI: 10.3390/ijms22031179] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 02/06/2023] Open
Abstract
Pulmonary hypertension (PH) is a progressive complex fatal disease of multiple etiologies. Hyperproliferation and resistance to apoptosis of vascular cells of intimal, medial, and adventitial layers of pulmonary vessels trigger excessive pulmonary vascular remodeling and vasoconstriction in the course of pulmonary arterial hypertension (PAH), a subgroup of PH. Multiple gene mutation/s or dysregulated gene expression contribute to the pathogenesis of PAH by endorsing the proliferation and promoting the resistance to apoptosis of pulmonary vascular cells. Given the vital role of these cells in PAH progression, the development of safe and efficient-gene therapeutic approaches that lead to restoration or down-regulation of gene expression, generally involved in the etiology of the disease is the need of the hour. Currently, none of the FDA-approved drugs provides a cure against PH, hence innovative tools may offer a novel treatment paradigm for this progressive and lethal disorder by silencing pathological genes, expressing therapeutic proteins, or through gene-editing applications. Here, we review the effectiveness and limitations of the presently available gene therapy approaches for PH. We provide a brief survey of commonly existing and currently applicable gene transfer methods for pulmonary vascular cells in vitro and describe some more recent developments for gene delivery existing in the field of PH in vivo.
Collapse
Affiliation(s)
- Nabham Rai
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
| | - Mazen Shihan
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
| | - Werner Seeger
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Institute for Lung Health (ILH), 35392 Giessen, Germany
| | - Ralph T. Schermuly
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
| | - Tatyana Novoyatleva
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
- Correspondence:
| |
Collapse
|
19
|
Abstract
Cyclic guanosine 3',5'-monophosphate (cGMP) is the key second messenger molecule in nitric oxide signaling. Its rapid generation and fate, but also its role in mediating acute cellular functions has been extensively studied. In the past years, genetic studies suggested an important role for cGMP in affecting the risk of chronic cardiovascular diseases, for example, coronary artery disease and myocardial infarction. Here, we review the role of cGMP in atherosclerosis and other cardiovascular diseases and discuss recent genetic findings and identified mechanisms. Finally, we highlight open questions and promising research topics.
Collapse
|
20
|
Shurbaji S, El-Sherbiny IM, Alser M, Ali IH, Kordi H, Al-Sadi A, Popelka A, Benslimane F, Yacoub M, Yalcin HC. Nitric Oxide Releasing Hydrogel Nanoparticles Decreases Epithelial Cell Injuries Associated With Airway Reopening. Front Bioeng Biotechnol 2021; 8:579788. [PMID: 33469529 PMCID: PMC7813943 DOI: 10.3389/fbioe.2020.579788] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 11/16/2020] [Indexed: 01/08/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is an acute inflammatory lung condition. It is characterized by disruption of gas exchange inside the alveoli, accumulation of protein edema, and an increase in lung stiffness. One major cause of ARDS is a lung infection, such as SARS-COV-2 infection. Lungs of ARDS patients need to be mechanically ventilated for airway reopening. Consequently, ventilation might damage delicate lung tissue leading to excess edema, known as ventilator-induced lung injury (VILI). Mortality of COVID-19 patients under VILI seems to be higher than non-COVID patients, necessitating effective preventative therapies. VILI occurs when small air bubbles form in the alveoli, injuring epithelial cells (EPC) due to shear stress. Nitric oxide (NO) inhalation was suggested as a therapy for ARDS, however, it was shown that it is not effective because of the extremely short half-life of NO. In this study, NO-releasing nanoparticles were produced and tested in an in vitro model, representing airways in the deep lung. Cellular injuries were quantified via fluorescent live/dead assay. Atomic force microscopy (AFM) was used to assess cell morphology. qRT-PCR was performed to assess the expression of inflammatory markers, specifically IL6 and CCL2. ELISA was performed to assess IL6 and confirm qRT-PCR results at the protein level. Finally, ROS levels were assessed in all groups. Here, we show that NO delivery via nanoparticles enhanced EPC survival and recovery, AFM measurements revealed that NO exposure affect cell morphology, while qRT-PCR demonstrated a significant downregulation in IL6 and CCL2 expression when treating the cells to NO both before and after shear exposure. ELISA results for IL6 confirmed qRT-PCR data. ROS experiment results support our findings from previous experiments. These findings demonstrate that NO-releasing nanoparticles can be used as an effective delivery approach of NO to deep lung to prevent/reduce ARDS associated inflammation and cell injuries. This information is particularly useful to treat severe ARDS due to COVID-19 infection. These nanoparticles will be useful when clinically administrated to COVID-19 patients to reduce the symptoms originating from lung distress.
Collapse
Affiliation(s)
- Samar Shurbaji
- Biomedical Research Center, Qatar University, Doha, Qatar
| | - Ibrahim M. El-Sherbiny
- Nanomedicine Lab, Center of Materials Sciences, Zewail City of Science and Technology, Giza, Egypt
| | - Maha Alser
- Biomedical Research Center, Qatar University, Doha, Qatar
| | - Isra H. Ali
- Nanomedicine Lab, Center of Materials Sciences, Zewail City of Science and Technology, Giza, Egypt
| | - Haya Kordi
- Biomedical Research Center, Qatar University, Doha, Qatar
- Department of Biomedical Sciences, College of Health Science-QU Health, Qatar University, Doha, Qatar
| | - Ameena Al-Sadi
- Biomedical Research Center, Qatar University, Doha, Qatar
- Department of Biomedical Sciences, College of Health Science-QU Health, Qatar University, Doha, Qatar
| | - Anton Popelka
- Center for Advanced Materials, Qatar University, Doha, Qatar
| | | | - Magdi Yacoub
- Heart Science Centre, Imperial College, National Heart and Lung Institute, London, United Kingdom
| | - Huseyin C. Yalcin
- Biomedical Research Center, Qatar University, Doha, Qatar
- Department of Biomedical Sciences, College of Health Science-QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
21
|
Treatment of Obstructive Sleep Apnea-Hypopnea Syndrome With a Mandible Advanced Device Increases Nitric Oxide Release and Ameliorates Pulmonary Hypertension in Rabbits. J Oral Maxillofac Surg 2020; 79:694.e1-694.e12. [PMID: 33259783 DOI: 10.1016/j.joms.2020.09.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 09/22/2020] [Accepted: 09/25/2020] [Indexed: 11/21/2022]
Abstract
PURPOSE To investigate the effects of mandible advanced device (MAD) therapy for obstructive sleep apnea-hypopnea syndrome (OSAHS) on nitric oxide (NO) release and changes in pulmonary artery pressure and structure. METHODS Thirty male New Zealand white rabbits were randomly divided into OSAHS, MAD, and control groups (n = 10 per group). The soft palate of rabbits in the OSAHS and MAD groups was injected with hydrophilic polyacrylamide gel to induce OSAHS. The MAD group wore a MAD, and the control group was not treated. Cone-beam computed tomography scans and polysomnography recordings were performed to confirm successful model establishment. All rabbits slept in a supine position for 4 to 6 hours daily and were observed for 8 consecutive weeks. The pulmonary artery pressure was measured by right heart catheterization. Pulmonary artery morphometry was analyzed by hematoxylin and eosin staining. NO levels in plasma and lung homogenate supernatants were detected by Griess reaction assay kits. RESULTS The OSAHS group exhibited higher pulmonary artery pressure (57.74 ± 1.79 mm Hg) than the MAD (19.99 ± 2.04 mm Hg) and control (14.49 ± 0.54 mm Hg) groups. The media thickness percentage of the pulmonary artery was higher in the OSAHS group (46.89 ± 2.72%) than the control group (15.87 ± 1.18%) and was markedly reduced by MAD (21.64 ± 1.45%). Blood oxygen saturation was positively correlated with the NO concentration in both the lung and plasma, and the NO concentration was negatively correlated with the media thickness percentage and media section percentage. CONCLUSIONS OSAHS induced a decrease in NO and pulmonary hypertension, which was relieved by MAD therapy.
Collapse
|
22
|
McClellan EB, Wang Z, Albertine KH, Kaplowitz MR, Zhang Y, Fike CD. l-Citrulline treatment alters the structure of the pulmonary circulation in hypoxic newborn pigs. Pediatr Pulmonol 2020; 55:2762-2772. [PMID: 32662946 PMCID: PMC7719608 DOI: 10.1002/ppul.24960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 07/12/2020] [Indexed: 11/07/2022]
Abstract
BACKGROUND Dysregulated nitric oxide (NO) signaling contributes to chronic hypoxia (CH)-induced pulmonary hypertension (PH). NO signaling is improved and pulmonary vascular resistance (PVR) is reduced in CH piglets treated with the l-arginine-NO precursor, l-citrulline. We hypothesized that l-citrulline might cause structural changes in the pulmonary circulation that would contribute to the reduction in PVR and that the l-citrulline-induced structural changes would be accompanied by alterations in vascular endothelial growth factor (VEGF) signaling. METHODS We evaluated small pulmonary arterial (PA) wall thickness, lung capillary density, and protein abundances of VEGF, VEGFR2, and phospho (p)-VEGFR2 in PA and peripheral lung samples of piglets raised in the lab in CH (10%-12% O2 ) from the day of life (DOL) 2 until DOL 11 to 12 or raised in room air (normoxia) by the vendor and studied on arrival to the lab on DOL 11 to 12. Some CH piglets were treated with oral l-citrulline (1-1.5 g/kg/d) starting on the third day of hypoxia. RESULTS PA wall thickness was 32% less and lung capillary formation was nearly doubled in l-citrulline treated than untreated CH piglets. Both of these l-citrulline-induced structural changes in the pulmonary circulation were accompanied by altered amounts of VEGF protein but not by altered amounts of either VEGFR2 or p-VEGFR2 proteins. CONCLUSIONS Alterations in the structure of the pulmonary circulation in CH piglets by l-citrulline are unlikely to be mediated by overall VEGF signaling. Nonetheless, l-citrulline- induced structural changes should reduce PVR and thereby contribute to the amelioration of CH-induced PH.
Collapse
Affiliation(s)
- Eric B McClellan
- Department of Pediatrics, University of Utah Health, Salt Lake City, Utah
| | - Zhengming Wang
- Department of Pediatrics, University of Utah Health, Salt Lake City, Utah
| | - Kurt H Albertine
- Department of Pediatrics, University of Utah Health, Salt Lake City, Utah
| | - Mark R Kaplowitz
- Department of Pediatrics, University of Utah Health, Salt Lake City, Utah
| | - Yongmei Zhang
- Department of Pediatrics, University of Utah Health, Salt Lake City, Utah
| | - Candice D Fike
- Department of Pediatrics, University of Utah Health, Salt Lake City, Utah
| |
Collapse
|
23
|
Brisket Disease Is Associated with Lower Volatile Fatty Acid Production and Altered Rumen Microbiome in Holstein Heifers. Animals (Basel) 2020; 10:ani10091712. [PMID: 32971776 PMCID: PMC7552702 DOI: 10.3390/ani10091712] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 09/17/2020] [Accepted: 09/19/2020] [Indexed: 01/01/2023] Open
Abstract
Simple Summary Development of the dairy industry in the high-altitude plateau environment through incorporation of Holstein cows is complicated by the risk of brisket disease. While the physiological effects of brisket disease are well-studied, its effects on rumen function and microbial community composition are not. There are clear shifts in volatile fatty acids production and rumen microbial community composition in Holstein heifers suffering from brisket disease. Observed shifts reveal key genera associated with healthy and disease states and suggest that bovine brisket disease is associated with impaired rumen functioning. This work supports further understanding of the roles of key rumen taxa in bovine brisket disease, with particular focus on candidate rumen biomarkers in healthy animals that may be able to reduce economic losses for farmers. Abstract Brisket disease is heritable but is also associated with non-genetic risk factors and effects of the disease on the rumen microbiome are unknown. Ten Holstein heifers were exposed to the plateau environment for three months and divided into two groups according to the index of brisket disease, the mean pulmonary arterial pressure (mPAP): brisket disease group (BD, n = 5, mPAP > 63 mmHg) and healthy heifer group (HH, n = 5, mPAP < 41 mmHg). Rumen fluid was collected for analysis of the concentrations of volatile fatty acids (VFAs). Extracted DNA from rumen contents was analyzed using Illumina MiSeq 16S rRNA sequencing technology. The concentration of total VFA and alpha-diversity metrics were significantly lower in BD group (p < 0.05). Ruminococcus and Treponema were significantly decreased in BD heifers (p < 0.05). Correlation analysis indicated that 10 genera were related to the mPAP (p < 0.05). Genera of Anaerofustis, Campylobacter, and Catonella were negatively correlated with total VFA and acetic acid (R < −0.7, p < 0.05), while genera of Blautia, YRC22, Ruminococcus, and Treponema were positively related to total VFA and acetic acid (R > 0.7; p < 0.05). Our findings may be a useful biomarker in future brisket disease work.
Collapse
|
24
|
Zhou Y, Gaucher C, Fries I, Hobekkaya MA, Martin C, Leonard C, Deschamps F, Sapin-Minet A, Parent M. Challenging development of storable particles for oral delivery of a physiological nitric oxide donor. Nitric Oxide 2020; 104-105:1-10. [PMID: 32771473 DOI: 10.1016/j.niox.2020.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/28/2020] [Accepted: 08/03/2020] [Indexed: 12/31/2022]
Abstract
Nitric oxide (NO) deficiency is often associated with several acute and chronic diseases. NO donors and especially S-nitrosothiols such as S-nitrosoglutathione (GSNO) have been identified as promising therapeutic agents. Although their permeability through the intestinal barrier have recently be proved, suitable drug delivery systems have to be designed for their oral administration. This is especially challenging due to the physico-chemical features of these drugs: high hydrophilicity and high lability. In this paper, three types of particles were prepared with an Eudragit® polymer: nanoparticles and microparticles obtained with a water-in-oil-in-water emulsion/evaporation process versus microparticles obtained with a solid-in-oil-in-water emulsion/evaporation process. They had a similar encapsulation efficiency (around 30%), and could be freeze-dried then be stored at least one month without modification of their critical attributes (size and GSNO content). However, microparticles had a slightly slower in vitro release of GSNO than nanoparticles, and were able to boost by a factor of two the drug intestinal permeability (Caco-2 model). Altogether, this study brings new data about GSNO intestinal permeability and three ready-to-use formulations suitable for further preclinical studies with oral administration.
Collapse
Affiliation(s)
- Yi Zhou
- Université de Lorraine, CITHEFOR, F-54000, Nancy, France
| | | | - Isabelle Fries
- Université de Lorraine, CITHEFOR, F-54000, Nancy, France
| | | | | | - Clément Leonard
- StaniPharm, 5 Rue Jacques Monod, BP 10, 54250, Champigneulles, France
| | - Frantz Deschamps
- StaniPharm, 5 Rue Jacques Monod, BP 10, 54250, Champigneulles, France
| | | | | |
Collapse
|
25
|
Hossain E, Sarkar O, Li Y, Anand-Srivastava MB. Sodium nitroprusside attenuates hyperproliferation of vascular smooth muscle cells from spontaneously hypertensive rats through the inhibition of overexpression of AT1 receptor, cell cycle proteins, and c-Src/growth factor receptor signaling pathways. Can J Physiol Pharmacol 2020; 98:35-43. [DOI: 10.1139/cjpp-2019-0338] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We recently showed that sodium nitroprusside (SNP), a NO donor, attenuated hypertension in spontaneously hypertensive rats (SHR). Since hypertension is associated with enhanced proliferation and hypertrophy of vascular smooth muscle cells (VSMC), the present study examines whether in vivo treatment of SHR with SNP could also inhibit the augmented proliferation of VSMC and explore the signaling mechanisms. Treatment of 8 week old SHR and Wistar Kyoto rats with SNP twice a week for 2 weeks inhibited the enhanced proliferation of VSMC from SHR, the enhanced expression of angiotensin II type 1 (AT1) receptor, and enhanced activation of c-Src and growth factor receptors and ERK1/2 signaling pathways. In addition, SNP also inhibited the overexpression of cell cycle proteins including cyclins D1, Cdk4, and phosphorylated pRB and restored the downregulated Cdk inhibitors p21Cip1 and p27Kip1 expression towards control levels. Furthermore, SNP-induced inhibition of enhanced levels of the AT1 receptor and enhanced proliferation was reversed by L-NAME, an inhibitor of nitric oxide synthase. These results suggest that the SNP-induced antiproliferative effect may be mediated through the inhibition of enhanced expression of the AT1 receptor, cell cycle proteins and activation of c-Src, growth factor receptors, and MAP kinase signaling.
Collapse
Affiliation(s)
- Ekhtear Hossain
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Oli Sarkar
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Yuan Li
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Madhu B. Anand-Srivastava
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| |
Collapse
|
26
|
Gai Z, Wang Z, Zhang L, Ma J, Zhu Q. Paeonol protects against hypertension in spontaneously hypertensive rats by restoring vascular endothelium. Biosci Biotechnol Biochem 2019; 83:1992-1999. [PMID: 31362597 DOI: 10.1080/09168451.2019.1648203] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
ABSTRACT
The present study focused on the effect of paeonol, one of the main components of Guizhi Fuling Pill, on blood pressure, cerebral blood flow, and vascular endothelium injury in spontaneously hypertensive rats to provide theoretical basis for the treatment of hypertension. After treatment with paeonol, the mean arterial pressure (MAP) of LSHRT and HSHRT rats decreased gradually with the prolongation of treatment time. The systolic blood flow velocity (Vs), diastolic blood flow velocity (Vd) and mean blood flow velocity (Vm) were significantly increased after paeonol treatment (p < 0.05). Paeonol effectively improved the blood pressure and increased the cerebral blood flow velocity in spontaneously hypertensive rats. This may be related to the fact that paeonol reduced the blood viscosity and the oxidative stress and improved the antioxidant capacity. Moreover, paeonol protected vascular endothelial cells and reduced vascular endothelial injury in spontaneously hypertensive rats.
Collapse
Affiliation(s)
- Zhonghui Gai
- Department of Heart Disease, Yantai Hospital of Traditional Chinese Medicine, Yantai, Shandong, China
| | - Zhenxing Wang
- Department of Encephalopathy, Yantai Hospital of Traditional Chinese Medicine, Yantai, Shandong, China
| | - Lei Zhang
- Department of Heart Disease, Yantai Hospital of Traditional Chinese Medicine, Yantai, Shandong, China
| | - Jun Ma
- Department of Heart Disease, Yantai Hospital of Traditional Chinese Medicine, Yantai, Shandong, China
| | - Qiao Zhu
- Department of Heart Disease, Yantai Hospital of Traditional Chinese Medicine, Yantai, Shandong, China
| |
Collapse
|
27
|
Beghetti M, Gorenflo M, Ivy DD, Moledina S, Bonnet D. Treatment of pediatric pulmonary arterial hypertension: A focus on the NO-sGC-cGMP pathway. Pediatr Pulmonol 2019; 54:1516-1526. [PMID: 31313530 PMCID: PMC6771736 DOI: 10.1002/ppul.24442] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 05/28/2019] [Indexed: 11/20/2022]
Abstract
OBJECTIVE While pulmonary arterial hypertension (PAH) is rare in infants and children, it results in substantial morbidity and mortality. In recent years, prognosis has improved, coinciding with the introduction of new PAH-targeted therapies, although much of their use in children is off-label. Evidence to guide the treatment of children with PAH is less extensive than for adults. The goal of this review is to discuss the treatment recommendations for children with PAH, as well as the evidence supporting the use of prostanoids, endothelin receptor antagonists (ERAs), and phosphodiesterase type 5 inhibitors (PDE5i) in this setting. DATA SOURCES Nonsystematic PubMed literature search and authors' expertise. STUDY SELECTION Articles were selected concentrating on the nitric oxide (NO)-soluble guanylate cyclase (sGC)-cyclic guanosine monophosphate (cGMP) pathway in PAH. The methodology of an ongoing study evaluating the sGC stimulator riociguat in children with PAH is also described. RESULTS Despite recent medical advances, improved therapeutic strategies for pediatric PAH are needed. The efficacy and tolerability of riociguat in adults with PAH have been well trialed. CONCLUSION The pooling of data across trials, supplemented by registry data, will help to confirm the safety and tolerability of prostanoids, ERAs, and PDE5i in children. Ongoing studies will clarify the place of sGC stimulators in the treatment strategy for pediatric PAH.
Collapse
Affiliation(s)
- Maurice Beghetti
- Pediatric Cardiology Unit and Centre Universitaire de Cardiologie et Chirurgie Cardiaque PédiatriqueChildren's University HospitalGenevaSwitzerland
| | - Matthias Gorenflo
- Department of Pediatrics II, Pediatric Cardiology and Congenital Heart Defects, Center for PediatricsUniversity Hospital HeidelbergGermany
| | - D. Dunbar Ivy
- Children's Hospital Colorado, Heart InstituteUniversity of Colorado School of MedicineDenverColorado
| | - Shahin Moledina
- Cardiology DepartmentGreat Ormond Street Hospital for Children NHS Foundation TrustLondonUK
| | - Damien Bonnet
- M3C‐Paediatric Cardiology, Necker Enfants Malades, AP‐HPUniversité Paris DescartesParisFrance
| |
Collapse
|
28
|
Tonelli AR, Aulak KS, Ahmed MK, Hausladen A, Abuhalimeh B, Casa CJ, Rogers SC, Timm D, Doctor A, Gaston B, Dweik RA. A pilot study on the kinetics of metabolites and microvascular cutaneous effects of nitric oxide inhalation in healthy volunteers. PLoS One 2019; 14:e0221777. [PMID: 31469867 PMCID: PMC6716644 DOI: 10.1371/journal.pone.0221777] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 08/14/2019] [Indexed: 11/19/2022] Open
Abstract
RATIONALE Inhaled nitric oxide (NO) exerts a variety of effects through metabolites and these play an important role in regulation of hemodynamics in the body. A detailed investigation into the generation of these metabolites has been overlooked. OBJECTIVES We investigated the kinetics of nitrite and S-nitrosothiol-hemoglobin (SNO-Hb) in plasma derived from inhaled NO subjects and how this modifies the cutaneous microvascular response. FINDINGS We enrolled 15 healthy volunteers. Plasma nitrite levels at baseline and during NO inhalation (15 minutes at 40 ppm) were 102 (86-118) and 114 (87-129) nM, respectively. The nitrite peak occurred at 5 minutes of discontinuing NO (131 (104-170) nM). Plasma nitrate levels were not significantly different during the study. SNO-Hb molar ratio levels at baseline and during NO inhalation were 4.7E-3 (2.5E-3-5.8E-3) and 7.8E-3 (4.1E-3-13.0E-3), respectively. Levels of SNO-Hb continued to climb up to the last study time point (30 min: 10.6E-3 (5.3E-3-15.5E-3)). The response to acetylcholine iontophoresis both before and during NO inhalation was inversely associated with the SNO-Hb level (r: -0.57, p = 0.03, and r: -0.54, p = 0.04, respectively). CONCLUSIONS Both nitrite and SNO-Hb increase during NO inhalation. Nitrite increases first, followed by a more sustained increase in Hb-SNO. Nitrite and Hb-SNO could be a mobile reservoir of NO with potential implications on the systemic microvasculature.
Collapse
Affiliation(s)
- Adriano R. Tonelli
- Department of Pulmonary, Allergy and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, Cleveland, OH, United States of America
- Pathobiology Division, Lerner Research Institute, Cleveland Clinic, OH, United States of America
| | - Kulwant S. Aulak
- Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, OH, United States of America
| | - Mostafa K. Ahmed
- Department of Chest Diseases, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Alfred Hausladen
- Institute for Transformative Molecular Medicine and Department of Medicine, Case Western Reserve University School of Medicine and University Hospitals Cleveland Medical Center, Cleveland, OH, United States of America
| | - Batool Abuhalimeh
- Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, OH, United States of America
| | - Charlie J. Casa
- Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, OH, United States of America
| | - Stephen C. Rogers
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - David Timm
- Department of Psychiatry, School of Medicine, Washington University, St. Louis, MO, United States of America
| | - Allan Doctor
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Benjamin Gaston
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Raed A. Dweik
- Department of Pulmonary, Allergy and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, Cleveland, OH, United States of America
- Pathobiology Division, Lerner Research Institute, Cleveland Clinic, OH, United States of America
| |
Collapse
|
29
|
Hossain E, Sarkar O, Li Y, Anand-Srivastava MB. Inhibition of overexpression of Giα proteins and nitroxidative stress contribute to sodium nitroprusside-induced attenuation of high blood pressure in SHR. Physiol Rep 2019; 6:e13658. [PMID: 29595917 PMCID: PMC5875540 DOI: 10.14814/phy2.13658] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 12/13/2017] [Accepted: 01/15/2018] [Indexed: 01/08/2023] Open
Abstract
We earlier showed that vascular smooth muscle cells (VSMC) from spontaneously hypertensive rats (SHR) exhibit enhanced expression of Giα proteins which was attributed to the decreased levels of nitric oxide (NO), because elevation of the intracellular levels of NO by NO donors; sodium nitroprusside (SNP) and S-Nitroso-N-acetyl-DL-penicillamine (SNAP), attenuated the enhanced expression of Giα proteins. Since the enhanced expression of Giα proteins is implicated in the pathogenesis of hypertension, the present study was undertaken to investigate if treatment of SHR with SNP could also attenuate the development of high blood pressure (BP) and explore the underlying molecular mechanisms. Intraperitoneal injection of SNP at a concentration of 0.5 mg/kg body weight twice a week for 2 weeks into SHR attenuated the high blood pressure by about 80 mmHg without affecting the BP in WKY rats. SNP treatment also attenuated the enhanced levels of superoxide anion (O2- ), hydrogen peroxide (H2 O2 ), peroxynitrite (ONOO- ), and NADPH oxidase activity in VSMC from SHR to control levels. In addition, the overexpression of different subunits of NADPH oxidase; Nox-1, Nox-2, Nox-4, P22phox , and P47phox , and Giα proteins in VSMC from SHR were also attenuated by SNP treatment. On the other hand, SNP treatment augmented the decreased levels of intracellular NO, eNOS, and cGMP in VSMC from SHR. These results suggest that SNP treatment attenuates the development of high BP in SHR through the elevation of intracellular levels of cGMP and inhibition of the enhanced levels of Giα proteins and nitroxidative stress.
Collapse
Affiliation(s)
- Ekhtear Hossain
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, Canada
| | - Oli Sarkar
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, Canada
| | - Yuan Li
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, Canada
| | - Madhu B Anand-Srivastava
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, Canada
| |
Collapse
|
30
|
Shekhawat P, Bagul M, Edwankar D, Pokharkar V. Enhanced dissolution/caco-2 permeability, pharmacokinetic and pharmacodynamic performance of re-dispersible eprosartan mesylate nanopowder. Eur J Pharm Sci 2019; 132:72-85. [PMID: 30797937 DOI: 10.1016/j.ejps.2019.02.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 02/11/2019] [Accepted: 02/15/2019] [Indexed: 02/06/2023]
Abstract
Eprosartan mesylate is an angiotensin receptor blocker which suffers from extremely poor bioavailability owing to its poor solubility and poor permeability. The rationale of the present work was to design the drug delivery system capable of overcoming these constraints. Nanoformulation of eprosartan mesylate was developed using ultrasonic wave-assisted liquid-antisolvent technique. Nanoformulation was further freeze dried with the addition of 1% of mannitol resulting in formation of re-dispersible EPM nanopowder. To prove our proof of principle, the re-dispersed nanopowder with z-average particle size 165.2 ± 1.8 nm was evaluated enormously for in-vitro dissolution behaviour and permeability assay through Caco-2 cell model. In-vitro dissolution study was performed at pH 1.2, pH 4.5 and pH 6.8. Result demonstrates enhanced dissolution from EPM nanopowder with negligible pH dependence. Transport studies accomplished using validated Caco-2 based cell model showed 11-fold enhanced apparent permeability of redispersed nanopowder when compared to pure EPM and corresponding physical mixture (p < 0.0001). In-vivo study reveals, exceptionally strong variations in plasma concentration of EPM through nanopowder (62 mg/kg) formulation when compared with physical mixture and pure EPM (62 mg/kg) group. Moreover, study manifests that 5-fold lower dose (12.4 mg/kg) of developed formulation yields higher exposure (4600 ± 36 ng·mL-1·h) than pure EPM (2349 ± 34 ng·mL-1·h) and corresponding physical mixture (2456 ± 49 ng·mL-1·h) at therapeutic dose (62 mg/kg). Further, L-NAME induced hypertensive model was undertaken to investigate effect of reduced dose of EPM nanopowder on systolic blood pressure, biochemical analysis and histopathology of heart. Results revealed pronounced antihypertensive potential of re-dispersed EPM nanopowder at 5-fold lower dose (12.4 mg/kg). In conclusion, our study indicates that nanopowder delivery might be the promising approach for providing enhanced oral bioavailability at lower dose.
Collapse
Affiliation(s)
- Prachi Shekhawat
- Department of Pharmaceutics, Poona College of Pharmacy, Bharati Vidyapeeth University, Erandwane, Pune, India
| | - Milind Bagul
- Raptim Research Limited, Mahape, Navi Mumbai, Maharashtra, India
| | - Diptee Edwankar
- Raptim Research Limited, Mahape, Navi Mumbai, Maharashtra, India
| | - Varsha Pokharkar
- Department of Pharmaceutics, Poona College of Pharmacy, Bharati Vidyapeeth University, Erandwane, Pune, India.
| |
Collapse
|
31
|
Ben Bouallègue F, Vauchot F, Bourdon A, Benkiran M, Boissin C, Charriot J, Bourdin A, Mariano-Goulart D. RV function improvement following nitric oxide inhalation demonstrated by gated blood pool SPECT in a patient with primary pulmonary hypertension. J Nucl Cardiol 2018; 25:2174-2176. [PMID: 29322380 DOI: 10.1007/s12350-017-1171-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 12/04/2017] [Indexed: 10/18/2022]
Affiliation(s)
- Fayçal Ben Bouallègue
- Nuclear Medicine Department, Montpellier University Hospital, Montpellier, France.
- PhyMedExp, INSERM, CNRS, Montpellier University, Montpellier, France.
| | - Fabien Vauchot
- Nuclear Medicine Department, Montpellier University Hospital, Montpellier, France
| | - Aurélie Bourdon
- Nuclear Medicine Department, Montpellier University Hospital, Montpellier, France
| | - Meriem Benkiran
- Nuclear Medicine Department, Montpellier University Hospital, Montpellier, France
| | - Clément Boissin
- Pneumology Department, Montpellier University Hospital, Montpellier, France
| | - Jérémy Charriot
- Pneumology Department, Montpellier University Hospital, Montpellier, France
| | - Arnaud Bourdin
- Pneumology Department, Montpellier University Hospital, Montpellier, France
- PhyMedExp, INSERM, CNRS, Montpellier University, Montpellier, France
| | - Denis Mariano-Goulart
- Nuclear Medicine Department, Montpellier University Hospital, Montpellier, France
- PhyMedExp, INSERM, CNRS, Montpellier University, Montpellier, France
| |
Collapse
|
32
|
Zhang B, Paffett ML, Naik JS, Jernigan NL, Walker BR, Resta TC. Cholesterol Regulation of Pulmonary Endothelial Calcium Homeostasis. CURRENT TOPICS IN MEMBRANES 2018; 82:53-91. [PMID: 30360783 DOI: 10.1016/bs.ctm.2018.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Cholesterol is a key structural component and regulator of lipid raft signaling platforms critical for cell function. Such regulation may involve changes in the biophysical properties of lipid microdomains or direct protein-sterol interactions that alter the function of ion channels, receptors, enzymes, and membrane structural proteins. Recent studies have implicated abnormal membrane cholesterol levels in mediating endothelial dysfunction that is characteristic of pulmonary hypertensive disorders, including that resulting from long-term exposure to hypoxia. Endothelial dysfunction in this setting is characterized by impaired pulmonary endothelial calcium entry and an associated imbalance that favors production vasoconstrictor and mitogenic factors that contribute to pulmonary hypertension. Here we review current knowledge of cholesterol regulation of pulmonary endothelial Ca2+ homeostasis, focusing on the role of membrane cholesterol in mediating agonist-induced Ca2+ entry and its components in the normal and hypertensive pulmonary circulation.
Collapse
Affiliation(s)
- Bojun Zhang
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, NM, United States
| | - Michael L Paffett
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, NM, United States
| | - Jay S Naik
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, NM, United States
| | - Nikki L Jernigan
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, NM, United States
| | - Benjimen R Walker
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, NM, United States
| | - Thomas C Resta
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, NM, United States.
| |
Collapse
|
33
|
Wang S, Azarfar A, Wang Y, Cao Z, Li S. N-carbamylglutamate restores nitric oxide synthesis and attenuates high altitude-induced pulmonary hypertension in Holstein heifers ascended to high altitude. J Anim Sci Biotechnol 2018; 9:63. [PMID: 30186602 PMCID: PMC6120069 DOI: 10.1186/s40104-018-0277-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 07/03/2018] [Indexed: 02/04/2023] Open
Abstract
Background High-altitude pulmonary hypertension (HAPH) is a life-threating condition for animals in high altitude, and disturbance of endothelial nitric oxide (NO) synthesis contributes to its pathogenesis. N-carbamylglutamate (NCG), which enhances arginine synthesis, promotes endogenous synthesis of NO. In this study, we determined the effects of NCG on alleviating HAPH in Holstein heifers that ascended to Tibet (Lhasa, 3,658 m). Methods Exp. 1, 2,000 Holstein heifers were transported from low elevation (1,027 m) to Lhasa. After being exposed to hypoxia for 1 yr, Holstein heifers were assigned to a healthy group (Control, n = 6) with mean pulmonary hypertension (mPAP) < 41 mmHg, and an HAPH affected group (HAPH, n = 6) with mPAP > 49 mmHg. Lung tissues were collected to evaluate histopathological changes and the expression of endothelial nitric oxide synthase (eNOS). Exp. 2, ten healthy heifers and 10 HAPH affected heifers were supplemented with NCG (20 g/d per heifer) for 4 wk. Physiological parameters were determined and blood samples were collected on d − 1 and d 28 of the feeding trial. Results Expression of eNOS in small pulmonary arteriole intima was higher in the healthy than HAPH group (P = 0.006), whereas HAPH group had significantly thicker media and adventitia than healthy group (all P < 0.05). The mRNA of eNOS and protein level of eNOS were higher in the lungs of heifers in the healthy group than in the HAPH group (both P < 0.001), whereas endothelin-1 protein levels were higher in HAPH group than in the healthy group (P = 0.025). NCG supplementation decreased mPAP and ammonia (both P = 0.001), whereas it increased the expression of eNOS, arginine, and plasma NO (all P < 0.05). Conclusions The expression of eNOS was decreased in Holstein heifers with HAPH. NCG supplementation decreased mPAP through the restoration of eNOS and endogenous NO synthesis. Electronic supplementary material The online version of this article (10.1186/s40104-018-0277-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shuxiang Wang
- 1State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 People's Republic of China
| | - Arash Azarfar
- 2Faculty of Agriculture, Department of Animal Science, Lorestan University, PO Box 465, Khorramabad, Iran
| | - Yajing Wang
- 1State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 People's Republic of China
| | - Zhijun Cao
- 1State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 People's Republic of China
| | - Shengli Li
- 1State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 People's Republic of China
| |
Collapse
|
34
|
Carpagnano GE, Radaeli A, Lacedonia D, Correale M, Carpagnano G, Palmiotti A, Barbaro MPF, Di Biase M, Brunetti N, Scioscia G, Malerba M. Exhaled Nitric Oxide and Exhaled Breath Temperature as Potential Biomarkers in Patients with Pulmonary Hypertension. BIOMED RESEARCH INTERNATIONAL 2018; 2018:7292045. [PMID: 30225263 PMCID: PMC6129334 DOI: 10.1155/2018/7292045] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 07/06/2018] [Accepted: 08/13/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND Pulmonary hypertension (PH) is a progressive fatal disease thus, noninvasive prognostic tools are needed to follow these patients. The aim of our study was to evaluate fractional exhaled nitric oxide (FeNO) and exhaled breath temperature (EBT) values in patients with PH from different causes and to correlate them with respiratory functional data. METHODS Twenty-four PH patients underwent spirometry, carbon monoxide diffusion (DLCO) test, transthoracic echocardiography, right-heart catheterization, and FeNO and EBT measurements. RESULTS We studied 3 groups according to the type of PH: 10 patients with pulmonary arterial hypertension (PAH) (group A), 11 patients with PH due to chronic obstructive pulmonary disease (COPD) (group B), and 3 patients with PH associated with left heart disease (group C). Mean FeNO values tend to be higher in group B (15.0 ± 9.3ppb) compared with other groups (respectively, 9.9 ± 5.7 and 8.5 ± 5.2 ppb in groups A and C; p = 0.271) but no statistical significance has been reached. Mean values of alveolar NO concentration (CANO) were higher in groups A and B compared to group C (respectively, 16.9 ± 12.6; 13.9 ± 6.8; and 6.7 ± 2.0 ppb) (p = 0.045). EBT mean values were significantly lower in group C when compared with other groups (group C: 29.0 +- 1.3°C, groups A and B: 30.9 ± 1.3 and 31.2 ± 1.2°C, respectively: p = 0.041). EBT levels were inversely correlated to mean pulmonary artery pressure (PAPm) levels (Spearman coefficient -0.481; p = 0.017). CONCLUSIONS eNO, CANO, and EBT have been evaluated in three groups of PH patients. Interestingly EBT reduction was correlated with PAPm increase, whereas FeNO was higher in COPD patients and CANO in PAH and COPD groups. Further studies are needed to clarify EBT, FeNO, and CANO roles as biomarkers in the monitoring of patients with PH.
Collapse
Affiliation(s)
- Giovanna Elisiana Carpagnano
- Sezione di Malattie dell'Apparato Respiratorio, Dipartimento di Scienze Mediche e Chirurgiche, Universita' di Foggia, Foggia, Italy
| | - Alessandro Radaeli
- Dipartimento di Emergenza Urgenza, Spedali Civili di Brescia, Brescia, Italy
| | - Donato Lacedonia
- Sezione di Malattie dell'Apparato Respiratorio, Dipartimento di Scienze Mediche e Chirurgiche, Universita' di Foggia, Foggia, Italy
| | - Michele Correale
- Cardiologia Universitaria, Dipartimento di Scienze Mediche e Chirurgiche, Universita' di Foggia, Foggia, Italy
| | - Giuseppe Carpagnano
- Cardiologia Universitaria, Dipartimento di Scienze Mediche e Chirurgiche, Universita' di Foggia, Foggia, Italy
| | - Antonio Palmiotti
- Cardiologia Universitaria, Dipartimento di Scienze Mediche e Chirurgiche, Universita' di Foggia, Foggia, Italy
| | - Maria Pia Foschino Barbaro
- Sezione di Malattie dell'Apparato Respiratorio, Dipartimento di Scienze Mediche e Chirurgiche, Universita' di Foggia, Foggia, Italy
| | - Matteo Di Biase
- Cardiologia Universitaria, Dipartimento di Scienze Mediche e Chirurgiche, Universita' di Foggia, Foggia, Italy
| | - Natale Brunetti
- Cardiologia Universitaria, Dipartimento di Scienze Mediche e Chirurgiche, Universita' di Foggia, Foggia, Italy
| | - Giulia Scioscia
- Sezione di Malattie dell'Apparato Respiratorio, Dipartimento di Scienze Mediche e Chirurgiche, Universita' di Foggia, Foggia, Italy
| | - Mario Malerba
- Malattie dell'Apparato Respiratorio, Dipartimento di Medicina Traslazionale, Università del Piemonte Orientale, Novara, Italy
| |
Collapse
|
35
|
Segura-Ibarra V, Wu S, Hassan N, Moran-Guerrero JA, Ferrari M, Guha A, Karmouty-Quintana H, Blanco E. Nanotherapeutics for Treatment of Pulmonary Arterial Hypertension. Front Physiol 2018; 9:890. [PMID: 30061840 PMCID: PMC6055049 DOI: 10.3389/fphys.2018.00890] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 06/20/2018] [Indexed: 12/21/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a devastating and fatal chronic lung disease. While current pharmacotherapies have improved patient quality of life, PAH drugs suffer from limitations in the form of short-term pharmacokinetics, instability, and poor organ specificity. Traditionally, nanotechnology-based delivery strategies have proven advantageous at increasing both circulation lifetimes of chemotherapeutics and accumulation in tumors due to enhanced permeability through fenestrated vasculature. Importantly, increased nanoparticle (NP) accumulation in diseased tissues has been observed pre-clinically in pathologies characterized by endothelial dysfunction and remodeled vasculature, including myocardial infarction and heart failure. Recently, this phenomenon has also been observed in preclinical models of PAH, leading to the exploration of NP-based drug delivery as a therapeutic modality in PAH. Herein, we discussed the advantages of NPs for efficacious treatment of PAH, including heightened therapeutic delivery to diseased lungs for increased drug bioavailability, as well as highlighted innovative nanotherapeutic approaches for PAH.
Collapse
Affiliation(s)
- Victor Segura-Ibarra
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, United States.,Escuela de Ingeniería y Ciencias, Tecnológico de Monterrey, Monterrey, Mexico
| | - Suhong Wu
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, United States
| | - Nida Hassan
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, United States.,McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Jose A Moran-Guerrero
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, United States.,Escuela de Ingeniería y Ciencias, Tecnológico de Monterrey, Monterrey, Mexico
| | - Mauro Ferrari
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, United States.,Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Ashrith Guha
- Department of Cardiology, Houston Methodist DeBakey Heart and Vascular Center, Houston Methodist Hospital, Houston, TX, United States.,Houston Methodist J.C. Walter Jr. Transplant Center, Houston Methodist Hospital, Houston, TX, United States
| | - Harry Karmouty-Quintana
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Elvin Blanco
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, United States.,Department of Cardiology, Houston Methodist DeBakey Heart and Vascular Center, Houston Methodist Hospital, Houston, TX, United States
| |
Collapse
|
36
|
Rashid J, Alobaida A, Al-Hilal TA, Hammouda S, McMurtry IF, Nozik-Grayck E, Stenmark KR, Ahsan F. Repurposing rosiglitazone, a PPAR-γ agonist and oral antidiabetic, as an inhaled formulation, for the treatment of PAH. J Control Release 2018; 280:113-123. [PMID: 29723610 DOI: 10.1016/j.jconrel.2018.04.049] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 04/28/2018] [Indexed: 12/20/2022]
Abstract
Peroxisome-proliferator-activated-receptor-gamma (PPAR-γ) is implicated, in some capacity, in the pathogenesis of pulmonary arterial hypertension (PAH). Rosiglitazone, an oral antidiabetic and PPAR-γ agonist, has the potential to dilate pulmonary arteries and to attenuate arterial remodeling in PAH. Here, we sought to test the hypothesis that rosiglitazone can be repurposed as inhaled formulation for the treatment of PAH. We have tested this conjecture by preparing and optimizing poly(lactic-co-glycolic) acid (PLGA) based particles of rosiglitazone, assessing the drug particles for pulmonary absorption, investigating the efficacy of the plain versus particulate drug formulation in improving the respiratory hemodynamics in PAH animals, and finally studying the effect of the drug in regulating the molecular markers associated with PAH pathogenesis. The optimized particles were slightly porous and spherical, and released 87.9% ± 6.7% of the drug in 24 h. The elimination half-life of the drug formulated in PLGA particles was 2.5-fold greater than that of the plain drug administered via the same route at the same dose. The optimized formulation, given via the pulmonary route, produced pulmonary selective vasodilation in PAH animals, but oral rosiglitazone had no effect in pulmonary hemodynamics. Rosiglitazone ameliorates the pathogenesis of PAH by balancing the molecular regulators involved in the vasoconstriction and vasodilation of human pulmonary arterial smooth muscle cells. All in all, data generated using intact animal and cellular models point to the conclusion that PLGA particles of an antidiabetic drug can be used for the treatment of a different disease, PAH.
Collapse
Affiliation(s)
- Jahidur Rashid
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center School of Pharmacy, Amarillo, TX 79106, USA
| | - Ahmad Alobaida
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center School of Pharmacy, Amarillo, TX 79106, USA
| | - Taslim A Al-Hilal
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center School of Pharmacy, Amarillo, TX 79106, USA
| | - Samia Hammouda
- The School of Sciences and Engineering, The American University in Cairo, Cairo, Egypt
| | - Ivan F McMurtry
- Department of Pharmacology, The Center for Lung Biology, University of South Alabama, Mobile, AL 36688, USA
| | - Eva Nozik-Grayck
- Department of Pediatrics, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kurt R Stenmark
- Department of Pediatrics, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Fakhrul Ahsan
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center School of Pharmacy, Amarillo, TX 79106, USA.
| |
Collapse
|
37
|
Abstract
Chronic thromboembolic pulmonary hypertension (CTEPH) is an uncommon and late complication of pulmonary embolism resulting from misguided remodelling of residual pulmonary thromboembolic material and small-vessel arteriopathy. CTEPH is the only form of pulmonary hypertension (PH) potentially curable by pulmonary endarterectomy (PEA). Unfortunately, several patients have either an unacceptable risk-benefit ratio for undergoing the surgical intervention or develop persistent PH after PEA. Novel medical and endovascular therapies can be considered for them. The soluble guanylate cyclase stimulator riociguat is recommended for the treatment of patients with inoperable disease or with recurrent/persistent PH after PEA. Other drugs developed for the treatment of other forms of PH, as prostanoids, phosphodiesterase-5 inhibitors and endothelin receptor antagonists have been used in the treatment of CTEPH, with limited benefit. Balloon pulmonary angioplasty is a novel and promising technique and is progressively emerging from the pioneering phase. Highly specialized training level and complex protocols of postoperative care are mandatory to consolidate the technical success of the surgical and endovascular intervention.
Collapse
Affiliation(s)
| | - Paolo Prandoni
- Department of Cardiac Thoracic and Vascular Sciences, University of Padua, Padua, Italy
| |
Collapse
|
38
|
Haikal RR, Hua C, Perry JJ, O'Nolan D, Syed I, Kumar A, Chester AH, Zaworotko MJ, Yacoub MH, Alkordi MH. Controlling the Uptake and Regulating the Release of Nitric Oxide in Microporous Solids. ACS APPLIED MATERIALS & INTERFACES 2017; 9:43520-43528. [PMID: 29182298 DOI: 10.1021/acsami.7b15095] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Representative compounds from three classes of microporous solids, namely, metal-organic frameworks (MOFs), hybrid ultra-microporous materials (HUMs), and porous-organic polymers (POPs), were investigated for their nitric oxide gas uptake and release behavior. Low-pressure sorption studies indicated strong chemisorption of NO on the free amine groups decorating the MOF UiO-66-NH2 when compared to its non-amine-functionalized parent. The HUMs demonstrated reversible physisorption within the low-pressure regime, but interestingly in one case there was evidence for chemisorption following pressurization with NO at 10 bar. Significant release of chemisorbed NO from the UiO-66-NH2 and one of the HUMs was triggered by addition of acid to the medium, a pH change from 7.4 to 5.4 being sufficient to trigger NO release. An imidazole-based POP exhibited chemisorption of NO at high pressure wherein the ring basicity facilitated both NO uptake and spontaneous release upon contact with the aqueous release medium.
Collapse
Affiliation(s)
- Rana R Haikal
- Center for Materials Science, Zewail City of Science and Technology , Sheikh Zayed Dist., 12588 Giza, Egypt
| | - Carol Hua
- Bernal Institute, Department of Chemical Sciences, University of Limerick , Limerick V94 T9PX, Ireland
| | - John J Perry
- Bernal Institute, Department of Chemical Sciences, University of Limerick , Limerick V94 T9PX, Ireland
| | - Daniel O'Nolan
- Bernal Institute, Department of Chemical Sciences, University of Limerick , Limerick V94 T9PX, Ireland
| | - Imran Syed
- Bernal Institute, Department of Chemical Sciences, University of Limerick , Limerick V94 T9PX, Ireland
| | - Amrit Kumar
- Bernal Institute, Department of Chemical Sciences, University of Limerick , Limerick V94 T9PX, Ireland
| | - Adrian H Chester
- Heart Science Centre, Imperial College , Harefield, Uxbridge UB9 6JH, United Kingdom
| | - Michael J Zaworotko
- Bernal Institute, Department of Chemical Sciences, University of Limerick , Limerick V94 T9PX, Ireland
| | - Magdi H Yacoub
- Heart Science Centre, Imperial College , Harefield, Uxbridge UB9 6JH, United Kingdom
| | - Mohamed H Alkordi
- Center for Materials Science, Zewail City of Science and Technology , Sheikh Zayed Dist., 12588 Giza, Egypt
| |
Collapse
|
39
|
Ahmed M, VanPatten S, Lakshminrusimha S, Patel H, Coleman TR, Al-Abed Y. Effects of novel muscarinic M3 receptor ligand C1213 in pulmonary arterial hypertension models. Physiol Rep 2017; 4:4/24/e13069. [PMID: 28039410 PMCID: PMC5210380 DOI: 10.14814/phy2.13069] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 11/07/2016] [Accepted: 11/13/2016] [Indexed: 11/24/2022] Open
Abstract
Pulmonary hypertension (PH) is a complex disease comprising a pathologic remodeling and thickening of the pulmonary vessels causing an after load on the right heart ventricle that can result in ventricular failure. Triggered by oxidative stress, episodes of hypoxia, and other undetermined causes, PH is associated with poor outcomes and a high rate of morbidity. In the neonate, this disease has a similar etiology but is further complicated by the transition to breathing after birth, which requires a reduction in vascular resistance. Persistent pulmonary hypertension of the newborn (PPHN) is one form of PH that is frequently unresponsive to current therapies including inhaled nitric oxide (due to lack of proper absorption and diffusion), and other therapeutics targeting signaling mediators in vascular endothelium and smooth muscle. The need for novel agents, which target distinct pathways in pulmonary hypertension, remains. Herein, we investigated the therapeutic effects of novel muscarinic receptor ligand C1213 in models of PH We demonstrated that via M3 muscarinic receptors, C1213 induced activating- eNOS phosphorylation (serine-1177), which is known to lead to nitric oxide (NO) production in endothelial cells. Using signaling pathway inhibitors, we discovered that AKT and calcium signaling contributed to eNOS phosphorylation induced by C1213. As expected for an eNOS-stimulating agent, in ex vivo and in vivo models, C1213 triggered pulmonary vasodilation and induced both pulmonary artery and systemic blood pressure reductions demonstrating its potential value in PH and PPHN In brief, this proof-of-concept study provides evidence that an M3 muscarinic receptor functionally selective ligand stimulates downstream pathways leading to antihypertensive effects using in vitro, ex vivo, and in vivo models of PH.
Collapse
Affiliation(s)
- Mohamed Ahmed
- Northwell Health, Pediatrics Department - Neonatology Division, Cohens Children's Medical Center, Manhasset, New York
| | - Sonya VanPatten
- Department of Medicinal Chemistry, Center for Molecular Innovation, Manhasset, New York
| | - Satyan Lakshminrusimha
- Departments of Pediatrics, Physiology and Biophysics, State University of New York at Buffalo, Buffalo, New York
| | - Hardik Patel
- Northwell Health, Pediatrics Department - Neonatology Division, Cohens Children's Medical Center, Manhasset, New York
| | - Thomas R Coleman
- The Feinstein Institute for Medical Research, Manhasset, New York
| | - Yousef Al-Abed
- Department of Medicinal Chemistry, Center for Molecular Innovation, Manhasset, New York
| |
Collapse
|
40
|
Telo S, Kırkıl G, Kuluöztürk M, Balin M, Deveci F. Can ADMA play a role in determining pulmonary hypertension related to chronic obstructive pulmonary disease? CLINICAL RESPIRATORY JOURNAL 2017; 12:1433-1438. [DOI: 10.1111/crj.12675] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 06/22/2017] [Accepted: 07/24/2017] [Indexed: 12/17/2022]
Affiliation(s)
- Selda Telo
- Department of Biochemistry and Clinical BiochemistryFirat University, School of Medicine (Firat Medical Center)Elazig 23119 Turkey
| | - Gamze Kırkıl
- Department of Chest DiseaseFirat University, School of Medicine, (Firat Medical Center)Elazig 23119 Turkey
| | - Mutlu Kuluöztürk
- Department of Chest DiseaseFirat University, School of Medicine, (Firat Medical Center)Elazig 23119 Turkey
| | - Mehmet Balin
- Department of CardiologyFirat University, School of Medicine (Firat Medical Center)Elazig 23119 Turkey
| | - Figen Deveci
- Department of Chest DiseaseFirat University, School of Medicine, (Firat Medical Center)Elazig 23119 Turkey
| |
Collapse
|
41
|
Sarkar O, Li Y, Anand-Srivastava MB. Nitric oxide attenuates overexpression of Giα proteins in vascular smooth muscle cells from SHR: Role of ROS and ROS-mediated signaling. PLoS One 2017; 12:e0179301. [PMID: 28692698 PMCID: PMC5503203 DOI: 10.1371/journal.pone.0179301] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 05/26/2017] [Indexed: 11/18/2022] Open
Abstract
Vascular smooth muscle cells (VSMC) from spontaneously hypertensive rats (SHR) exhibit decreased levels of nitric oxide (NO) that may be responsible for the overexpression of Giα proteins that has been shown as a contributing factor for the pathogenesis of hypertension in SHR. The present study was undertaken to investigate if increasing the intracellular levels of NO by NO donor S-Nitroso-N-acetyl-DL-penicillamine (SNAP) could attenuate the enhanced expression of Giα proteins in VSMC from SHR and explore the underlying mechanisms responsible for this response. The expression of Giα proteins and phosphorylation of ERK1/2, growth factor receptors and c-Src was determined by Western blotting using specific antibodies. Treatment of VSMC from SHR with SNAP for 24 hrs decreased the enhanced expression of Giα-2 and Giα-3 proteins and hyperproliferation that was not reversed by 1H (1, 2, 4) oxadiazole (4, 3-a) quinoxalin-1-one (ODQ), an inhibitor of soluble guanylyl cyclase, however, PD98059, a MEK inhibitor restored the SNAP-induced decreased expression of Giα proteins towards control levels. In addition, the increased production of superoxide anion, NAD(P)H oxidase activity, overexpression of AT1 receptor, Nox4, p22phox and p47phox proteins, enhanced levels of TBARS and protein carbonyl, increased phosphorylation of PDGF-R, EGF-R, c-Src and ERK1/2 in VSMC from SHR were all decreased to control levels by SNAP treatment. These results suggest that NO decreased the enhanced expression of Giα-2/3 proteins and hyperproliferation of VSMC from SHR by cGMP-independent mechanism and involves ROS and ROS-mediated transactivation of EGF-R/PDGF-R and MAP kinase signaling pathways.
Collapse
MESH Headings
- Animals
- Cell Proliferation/drug effects
- Cyclic GMP/analogs & derivatives
- Cyclic GMP/pharmacology
- DNA/biosynthesis
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Flavonoids/pharmacology
- GTP-Binding Protein alpha Subunits, Gi-Go/metabolism
- Male
- Muscle, Smooth, Vascular/cytology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/metabolism
- NADPH Oxidases/metabolism
- Nitric Oxide/pharmacology
- Nitric Oxide Donors/pharmacology
- Nitroprusside/pharmacology
- Oxadiazoles/pharmacology
- Oxidative Stress/drug effects
- Phosphorylation/drug effects
- Protein Carbonylation/drug effects
- Rats, Inbred SHR
- Rats, Inbred WKY
- Reactive Oxygen Species/metabolism
- Receptor, Angiotensin, Type 1/metabolism
- S-Nitroso-N-Acetylpenicillamine/pharmacology
- Signal Transduction/drug effects
- Superoxides/metabolism
- Thiobarbituric Acid Reactive Substances/metabolism
- src-Family Kinases/metabolism
Collapse
Affiliation(s)
- Oli Sarkar
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montréal, Montréal, Canada
| | - Yuan Li
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montréal, Montréal, Canada
| | - Madhu B. Anand-Srivastava
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montréal, Montréal, Canada
- * E-mail:
| |
Collapse
|
42
|
Foderaro AE, Baird GL, Bazargan-Lari A, Morrissey PE, Gohh RY, Poppas A, Klinger JR, Ventetuolo CE. Echocardiographic Pulmonary Hypertension Predicts Post-transplantation Renal Allograft Failure. Transplant Proc 2017; 49:1256-1261. [PMID: 28735990 PMCID: PMC5567835 DOI: 10.1016/j.transproceed.2017.01.085] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 01/13/2017] [Accepted: 01/24/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND Pulmonary hypertension in the setting of renal transplantation has been associated with early allograft dysfunction and increased mortality, but this relationship has not been extensively studied. METHODS We performed a retrospective cohort study of adult patients who underwent their first renal transplantation in the years 2003-2009 and had pre-transplantation echocardiograms. Pulmonary hypertension was defined as right ventricular systolic pressure ≥40 mm Hg in the absence of left-sided valvular disease and/or left ventricular ejection fraction ≤50%. Eighty-two of 205 patients (40%) met the inclusion criteria. The relationship between pulmonary hypertension and death-censored allograft failure (hemodialysis dependence or retransplantation) and serum creatinine was assessed with the use of Cox hazard regression and generalized mixed models. RESULTS The presence of pulmonary hypertension was associated with a 3-fold increase in the risk of death-censored allograft failure (95% confidence interval, 1.20-7.32; P = .02). Failure rates were 19% at 24 months and 51% at 96 months for those with pulmonary hypertension versus 7% at 24 months and 20% at 86 months for those without pulmonary hypertension (P = .01). Among those without graft failure, there was an increase in creatinine levels after transplantation (P = .01). Effect estimates were unchanged by adjustment for multiple covariates and when pulmonary hypertension was defined as right ventricular systolic pressure ≥36 mm Hg. CONCLUSIONS Pulmonary hypertension before renal transplantation carries a 3-fold increased risk of death-censored allograft failure. The relationship between the pulmonary circulation and renal allograft failure warrants further study.
Collapse
Affiliation(s)
- A E Foderaro
- Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - G L Baird
- Department of Surgery, Brown University School of Public Health, Providence, Rhode Island
| | - A Bazargan-Lari
- Department of Health Services, Policy, and Practice, Alpert Medical School of Brown University, Providence, Rhode Island
| | - P E Morrissey
- Lifespan Biostatistics Core, Lifespan Hospital System, Providence, Rhode Island
| | - R Y Gohh
- Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - A Poppas
- Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - J R Klinger
- Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - C E Ventetuolo
- Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island; Department of Internal Medicine, Kaiser Permanente, West Los Angeles, California.
| |
Collapse
|
43
|
Abstract
The pathogenesis of pulmonary arterial hypertension remains undefined. Changes in the expression and effects mediated by a number of vasoactive factors have been implicated to play a role in the onset and progression of the disease. The source of many of these mediators, such as nitric oxide (NO), prostacyclin and endothelin-1 (ET-1), is the pulmonary endothelium. This article focus in the role of nitric oxide in PAH, reviewing the evidence for its involvement in regulation of pulmonary a vascular tone under physiological conditions, the mechanisms by which it can contribute to the pathological changes seen in PAH and strategies for the use of NO as a therapy for treatment of the disease.
Collapse
Affiliation(s)
- Adrian H Chester
- National Heart & Lung Institute, Imperial College London, Heart Science Centre, Harefield, Middlesex, UB9 6JH, United Kingdom
| | - Magdi H Yacoub
- National Heart & Lung Institute, Imperial College London, Heart Science Centre, Harefield, Middlesex, UB9 6JH, United Kingdom
| | - Salvador Moncada
- School of Medical Sciences, Manchester Cancer Research Centre, University of Manchester, Wilmslow Road, Manchester, M20 4QL, United Kingdom
| |
Collapse
|
44
|
Koch CD, Gladwin MT, Freeman BA, Lundberg JO, Weitzberg E, Morris A. Enterosalivary nitrate metabolism and the microbiome: Intersection of microbial metabolism, nitric oxide and diet in cardiac and pulmonary vascular health. Free Radic Biol Med 2017; 105:48-67. [PMID: 27989792 PMCID: PMC5401802 DOI: 10.1016/j.freeradbiomed.2016.12.015] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/18/2016] [Accepted: 12/12/2016] [Indexed: 02/07/2023]
Abstract
Recent insights into the bioactivation and signaling actions of inorganic, dietary nitrate and nitrite now suggest a critical role for the microbiome in the development of cardiac and pulmonary vascular diseases. Once thought to be the inert, end-products of endothelial-derived nitric oxide (NO) heme-oxidation, nitrate and nitrite are now considered major sources of exogenous NO that exhibit enhanced vasoactive signaling activity under conditions of hypoxia and stress. The bioavailability of nitrate and nitrite depend on the enzymatic reduction of nitrate to nitrite by a unique set of bacterial nitrate reductase enzymes possessed by specific bacterial populations in the mammalian mouth and gut. The pathogenesis of pulmonary hypertension (PH), obesity, hypertension and CVD are linked to defects in NO signaling, suggesting a role for commensal oral bacteria to shape the development of PH through the formation of nitrite, NO and other bioactive nitrogen oxides. Oral supplementation with inorganic nitrate or nitrate-containing foods exert pleiotropic, beneficial vascular effects in the setting of inflammation, endothelial dysfunction, ischemia-reperfusion injury and in pre-clinical models of PH, while traditional high-nitrate dietary patterns are associated with beneficial outcomes in hypertension, obesity and CVD. These observations highlight the potential of the microbiome in the development of novel nitrate- and nitrite-based therapeutics for PH, CVD and their risk factors.
Collapse
Affiliation(s)
- Carl D Koch
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15261, USA.
| | - Mark T Gladwin
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15261, USA; Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh PA 15261, USA
| | - Bruce A Freeman
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh PA 15261, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Jon O Lundberg
- Department of Physiology and Pharmacology, Karolinska Institutet, S-17177 Stockholm, Sweden
| | - Eddie Weitzberg
- Department of Physiology and Pharmacology, Karolinska Institutet, S-17177 Stockholm, Sweden
| | - Alison Morris
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15261, USA; Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh PA 15261, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Physiology and Pharmacology, Karolinska Institutet, S-17177 Stockholm, Sweden
| |
Collapse
|
45
|
Simonneau G, Torbicki A, Dorfmüller P, Kim N. The pathophysiology of chronic thromboembolic pulmonary hypertension. Eur Respir Rev 2017; 26:26/143/160112. [DOI: 10.1183/16000617.0112-2016] [Citation(s) in RCA: 210] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 02/17/2017] [Indexed: 12/22/2022] Open
Abstract
Chronic thromboembolic pulmonary hypertension (CTEPH) is a rare, progressive pulmonary vascular disease that is usually a consequence of prior acute pulmonary embolism. CTEPH usually begins with persistent obstruction of large and/or middle-sized pulmonary arteries by organised thrombi. Failure of thrombi to resolve may be related to abnormal fibrinolysis or underlying haematological or autoimmune disorders. It is now known that small-vessel abnormalities also contribute to haemodynamic compromise, functional impairment and disease progression in CTEPH. Small-vessel disease can occur in obstructed areas, possibly triggered by unresolved thrombotic material, and downstream from occlusions, possibly because of excessive collateral blood supply from high-pressure bronchial and systemic arteries. The molecular processes underlying small-vessel disease are not completely understood and further research is needed in this area. The degree of small-vessel disease has a substantial impact on the severity of CTEPH and postsurgical outcomes. Interventional and medical treatment of CTEPH should aim to restore normal flow distribution within the pulmonary vasculature, unload the right ventricle and prevent or treat small-vessel disease. It requires early, reliable identification of patients with CTEPH and use of optimal treatment modalities in expert centres.
Collapse
|
46
|
Pepke-Zaba J, Ghofrani HA, Hoeper MM. Medical management of chronic thromboembolic pulmonary hypertension. Eur Respir Rev 2017; 26:26/143/160107. [DOI: 10.1183/16000617.0107-2016] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 02/14/2017] [Indexed: 02/07/2023] Open
Abstract
Chronic thromboembolic pulmonary hypertension (CTEPH) results from incomplete resolution of acute pulmonary emboli, organised into fibrotic material that obstructs large pulmonary arteries, and distal small-vessel arteriopathy. Pulmonary endarterectomy (PEA) is the treatment of choice for eligible patients with CTEPH; in expert centres, PEA has low in-hospital mortality rates and excellent long-term survival. Supportive medical therapy consists of lifelong anticoagulation plus diuretics and oxygen, as needed.An important recent advance in medical therapy for CTEPH is the arrival of medical therapies for patients with inoperable disease or persistent/recurrent pulmonary hypertension after PEA. The soluble guanylate cyclase stimulator riociguat is licensed for the treatment of CTEPH in patients with inoperable disease or with recurrent/persistent pulmonary hypertension after PEA. Clinical trials of this agent have shown improvements in patients' haemodynamics and exercise capacity. Phosphodiesterase-5 inhibitors, endothelin receptor antagonists and prostanoids have been used in the treatment of CTEPH, but evidence of benefit is limited. Challenges in the future development of medical therapy for CTEPH include better understanding of the underlying pathology, end-points to monitor the condition's progress, and the optimisation of pulmonary arterial hypertension therapies in relation to diverse patient characteristics and emerging options such as balloon pulmonary angioplasty.
Collapse
|
47
|
Mohamed NA, Ahmetaj-Shala B, Duluc L, Mackenzie LS, Kirkby NS, Reed DM, Lickiss PD, Davies RP, Freeman GR, Wojciak-Stothard B, Chester AH, El-Sherbiny IM, Mitchell JA, Yacoub MH. A New NO-Releasing Nanoformulation for the Treatment of Pulmonary Arterial Hypertension. J Cardiovasc Transl Res 2016; 9:162-4. [PMID: 26960567 PMCID: PMC4830862 DOI: 10.1007/s12265-016-9684-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 02/17/2016] [Indexed: 01/25/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a chronic and progressive disease which continues to carry an unacceptably high mortality and morbidity. The nitric oxide (NO) pathway has been implicated in the pathophysiology and progression of the disease. Its extremely short half-life and systemic effects have hampered the clinical use of NO in PAH. In an attempt to circumvent these major limitations, we have developed a new NO-nanomedicine formulation. The formulation was based on hydrogel-like polymeric composite NO-releasing nanoparticles (NO-RP). The kinetics of NO release from the NO-RP showed a peak at about 120 min followed by a sustained release for over 8 h. The NO-RP did not affect the viability or inflammation responses of endothelial cells. The NO-RP produced concentration-dependent relaxations of pulmonary arteries in mice with PAH induced by hypoxia. In conclusion, NO-RP drugs could considerably enhance the therapeutic potential of NO therapy for PAH.
Collapse
Affiliation(s)
- Nura A Mohamed
- Department of Cardiothoracic Pharmacology, National Heart and Lung Institute, Imperial College, Dovehouse Street, London, SW3 6LY, UK
- Heart Science Centre, Imperial College, Harefield, Uxbridge, UB9 6JH, UK
- Qatar Foundation Research and Development Division, Doha, Qatar
| | - Blerina Ahmetaj-Shala
- Department of Cardiothoracic Pharmacology, National Heart and Lung Institute, Imperial College, Dovehouse Street, London, SW3 6LY, UK
| | - Lucie Duluc
- Department of Experimental Medicine and Toxicology, Hammersmith Campus, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Louise S Mackenzie
- School of Life and Medical Sciences, University of Hertfordshire, Hatfield, AL10 9AB, UK
| | - Nicholas S Kirkby
- Department of Cardiothoracic Pharmacology, National Heart and Lung Institute, Imperial College, Dovehouse Street, London, SW3 6LY, UK
| | - Daniel M Reed
- Department of Cardiothoracic Pharmacology, National Heart and Lung Institute, Imperial College, Dovehouse Street, London, SW3 6LY, UK
| | - Paul D Lickiss
- Synthesis Section, Department of Chemistry, Imperial College London, South Kensington, London, SW7 2AZ, UK
| | - Robert P Davies
- Synthesis Section, Department of Chemistry, Imperial College London, South Kensington, London, SW7 2AZ, UK
| | - Gemma R Freeman
- Synthesis Section, Department of Chemistry, Imperial College London, South Kensington, London, SW7 2AZ, UK
| | - Beata Wojciak-Stothard
- Department of Experimental Medicine and Toxicology, Hammersmith Campus, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Adrian H Chester
- Heart Science Centre, Imperial College, Harefield, Uxbridge, UB9 6JH, UK
| | - Ibrahim M El-Sherbiny
- Center for Materials Science, Zewail City, Sheikh, Zayed District, 12588, 6th of October City, Giza, Egypt
| | - Jane A Mitchell
- Department of Cardiothoracic Pharmacology, National Heart and Lung Institute, Imperial College, Dovehouse Street, London, SW3 6LY, UK
| | - Magdi H Yacoub
- Heart Science Centre, Imperial College, Harefield, Uxbridge, UB9 6JH, UK.
| |
Collapse
|
48
|
Frey R, Becker C, Unger S, Schmidt A, Wensing G, Mück W. Assessment of the effects of renal impairment and smoking on the pharmacokinetics of a single oral dose of the soluble guanylate cyclase stimulator riociguat (BAY 63-2521). Pulm Circ 2016; 6:S15-26. [PMID: 27162624 PMCID: PMC4860531 DOI: 10.1086/685017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 06/28/2015] [Indexed: 12/26/2022] Open
Abstract
Renal impairment is a common comborbidity in patients with pulmonary hypertension. The breakdown of riociguat, an oral soluble guanylate cyclase stimulator used to treat pulmonary hypertension, may be affected by smoking because polycyclic aromatic hydrocarbons in tobacco smoke induce expression of one of the metabolizing enzymes, CYP1A1. Two nonrandomized, nonblinded studies were therefore performed to investigate the pharmacokinetics and safety of a single oral dose of riociguat 1.0 mg in individuals with mild, moderate, or severe renal impairment compared with age-, weight-, and sex-matched healthy controls, including either smokers and nonsmokers (study I) or nonsmokers alone (study II). Pharmacokinetic analyses focused on the integrated per-protocol data set of both studies (N = 63). In patients with renal impairment, the renal clearance of riociguat was reduced and its terminal half-life prolonged compared with those in healthy controls. There was a monotonic relationship between creatinine clearance on treatment day and riociguat renal clearance (R (2) = 0.62). However, increased riociguat exposure with decreasing renal function was not strictly proportional. Riociguat exposure appeared to be greater in nonsmokers than in the combined population of smokers and nonsmokers, irrespective of renal function. Adverse events were mild to moderate and in line with the mode of action of riociguat. No serious adverse events occurred. In conclusion, renal impairment was associated with reduced riociguat clearance compared with that in controls; however, riociguat exposure in patients with renal impairment was highly variable, and ranges overlapped with those observed in healthy controls.
Collapse
Affiliation(s)
- Reiner Frey
- Clinical Pharmacology, Pharma Research Center, Bayer Pharma, Wuppertal, Germany
| | - Corina Becker
- Clinical Pharmacology, Pharma Research Center, Bayer Pharma, Wuppertal, Germany
| | - Sigrun Unger
- Global Biostatistics, Pharma Research Center, Bayer Pharma, Wuppertal, Germany
| | - Anja Schmidt
- Clinical Pharmacology, Pharma Research Center, Bayer Pharma, Wuppertal, Germany
| | - Georg Wensing
- Clinical Pharmacology, Pharma Research Center, Bayer Pharma, Wuppertal, Germany
| | - Wolfgang Mück
- Clinical Pharmacology, Pharma Research Center, Bayer Pharma, Wuppertal, Germany
| |
Collapse
|
49
|
Fike CD, Dikalova A, Kaplowitz MR, Cunningham G, Summar M, Aschner JL. Rescue Treatment with L-Citrulline Inhibits Hypoxia-Induced Pulmonary Hypertension in Newborn Pigs. Am J Respir Cell Mol Biol 2015; 53:255-64. [PMID: 25536367 DOI: 10.1165/rcmb.2014-0351oc] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Infants with cardiopulmonary disorders associated with hypoxia develop pulmonary hypertension. We previously showed that initiation of oral L-citrulline before and continued throughout hypoxic exposure improves nitric oxide (NO) production and ameliorates pulmonary hypertension in newborn piglets. Rescue treatments, initiated after the onset of pulmonary hypertension, better approximate clinical strategies. Mechanisms by which L-citrulline improves NO production merit elucidation. The objective of this study was to determine whether starting L-citrulline after the onset of pulmonary hypertension inhibits disease progression and improves NO production by recoupling endothelial NO synthase (eNOS). Hypoxic and normoxic (control) piglets were studied. Some hypoxic piglets received oral L-citrulline starting on Day 3 of hypoxia and continuing throughout the remaining 7 days of hypoxic exposure. Catheters were placed for hemodynamic measurements, and pulmonary arteries were dissected to assess NO production and eNOS dimer-to-monomer ratios (a measure of eNOS coupling). Pulmonary vascular resistance was lower in L-citrulline-treated hypoxic piglets than in untreated hypoxic piglets but was higher than in normoxic controls. NO production and eNOS dimer-to-monomer ratios were greater in pulmonary arteries from L-citrulline-treated than from untreated hypoxic animals but were lower than in normoxic controls. When started after disease onset, oral L-citrulline treatment improves NO production by recoupling eNOS and inhibits the further development of chronic hypoxia-induced pulmonary hypertension in newborn piglets. Oral L-citrulline may be a novel strategy to halt or reverse pulmonary hypertension in infants suffering from cardiopulmonary conditions associated with hypoxia.
Collapse
Affiliation(s)
- Candice D Fike
- 1 Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee.,2 Monroe Carell Jr. Children's Hospital at Vanderbilt, Nashville, Tennessee
| | - Anna Dikalova
- 1 Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Mark R Kaplowitz
- 1 Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Gary Cunningham
- 3 Division of Genetics and Metabolism, Children's National Medical Center, Washington, District of Columbia; and
| | - Marshall Summar
- 3 Division of Genetics and Metabolism, Children's National Medical Center, Washington, District of Columbia; and
| | - Judy L Aschner
- 4 Department of Pediatrics, Albert Einstein College of Medicine and the Children's Hospital at Montefiore, New York, New York
| |
Collapse
|
50
|
Vascular nitric oxide: Beyond eNOS. J Pharmacol Sci 2015; 129:83-94. [PMID: 26499181 DOI: 10.1016/j.jphs.2015.09.002] [Citation(s) in RCA: 494] [Impact Index Per Article: 54.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 09/11/2015] [Accepted: 09/16/2015] [Indexed: 02/06/2023] Open
Abstract
As the first discovered gaseous signaling molecule, nitric oxide (NO) affects a number of cellular processes, including those involving vascular cells. This brief review summarizes the contribution of NO to the regulation of vascular tone and its sources in the blood vessel wall. NO regulates the degree of contraction of vascular smooth muscle cells mainly by stimulating soluble guanylyl cyclase (sGC) to produce cyclic guanosine monophosphate (cGMP), although cGMP-independent signaling [S-nitrosylation of target proteins, activation of sarco/endoplasmic reticulum calcium ATPase (SERCA) or production of cyclic inosine monophosphate (cIMP)] also can be involved. In the blood vessel wall, NO is produced mainly from l-arginine by the enzyme endothelial nitric oxide synthase (eNOS) but it can also be released non-enzymatically from S-nitrosothiols or from nitrate/nitrite. Dysfunction in the production and/or the bioavailability of NO characterizes endothelial dysfunction, which is associated with cardiovascular diseases such as hypertension and atherosclerosis.
Collapse
|