1
|
Sun X, Moreno Caceres S, Yegambaram M, Lu Q, Pokharel MD, Boehme JT, Datar SA, Aggarwal S, Wang T, Fineman JR, Black SM. The mitochondrial redistribution of ENOS is regulated by AKT1 and dimer status. Nitric Oxide 2024; 152:90-100. [PMID: 39332480 DOI: 10.1016/j.niox.2024.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 09/21/2024] [Accepted: 09/24/2024] [Indexed: 09/29/2024]
Abstract
Previously, we have shown that endothelial nitric-oxide synthase (eNOS) dimer levels directly correlate with the interaction of eNOS with hsp90 (heat shock protein 90). Further, the disruption of eNOS dimerization correlates with its redistribution to the mitochondria. However, the causal link between these events has yet to be investigated and was the focus of this study. Our data demonstrates that simvastatin, which decreases the mitochondrial redistribution of eNOS, increased eNOS-hsp90 interactions and enhanced eNOS dimerization in cultured pulmonary arterial endothelial cells (PAEC) from a lamb model of pulmonary hypertension (PH). Our data also show that the dimerization of a monomeric fraction of human recombinant eNOS was stimulated in the presence of hsp90 and ATP. The over-expression of a dominant negative mutant of hsp90 (DNHsp90) decreased eNOS dimer levels and enhanced its mitochondrial redistribution. We also found that the peroxynitrite donor3-morpholinosydnonimine (SIN-1) increased the mitochondrial redistribution of eNOS in PAEC and this was again associated with decreased eNOS dimer levels. Our data also show in COS-7 cells, the SIN-1 mediated mitochondrial redistribution of wildtype eNOS (WT-eNOS) is significantly higher than a dimer stable eNOS mutant protein (C94R/C99R-eNOS). Conversely, the mitochondrial redistribution of a monomeric eNOS mutant protein (C96A-eNOS) was enhanced. Finally, we linked the SIN-1-mediated mitochondrial redistribution of eNOS to the Akt1-mediated phosphorylation of eNOS at Serine(S)617 and showed that the accessibility of this residue to phosphorylation is regulated by dimerization status. Thus, our data reveal a novel mechanism of pulmonary endothelial dysfunction mediated by mitochondrial redistribution of eNOS, regulated by dimerization status and the phosphorylation of S617.
Collapse
Affiliation(s)
- Xutong Sun
- Center for Translational Science, Florida International University, Port St. Lucie, FL, 34987, USA; Departments of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33174, USA
| | - Santiago Moreno Caceres
- Center for Translational Science, Florida International University, Port St. Lucie, FL, 34987, USA; Departments of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33174, USA
| | - Manivannan Yegambaram
- Center for Translational Science, Florida International University, Port St. Lucie, FL, 34987, USA; Departments of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33174, USA
| | - Qing Lu
- Center for Translational Science, Florida International University, Port St. Lucie, FL, 34987, USA; Departments of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33174, USA
| | - Marissa D Pokharel
- Center for Translational Science, Florida International University, Port St. Lucie, FL, 34987, USA; Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33174, USA
| | - Jason T Boehme
- The Department of Pediatrics, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Sanjeev A Datar
- The Department of Pediatrics, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Saurabh Aggarwal
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33174, USA
| | - Ting Wang
- Center for Translational Science, Florida International University, Port St. Lucie, FL, 34987, USA; Departments of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33174, USA
| | - Jeffrey R Fineman
- The Department of Pediatrics, University of California San Francisco, San Francisco, CA, 94143, USA; The Department of Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Stephen M Black
- Center for Translational Science, Florida International University, Port St. Lucie, FL, 34987, USA; Departments of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33174, USA; Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33174, USA.
| |
Collapse
|
2
|
Xie J, Zhang X, Cheng L, Deng Y, Ren H, Mu M, Zhao L, Mu C, Chen J, Liu K, Ma R. Integrated multi-omics analysis of the microbial profile characteristics associated with pulmonary arterial hypertension in congenital heart disease. Microbiol Spectr 2024:e0180824. [PMID: 39470277 DOI: 10.1128/spectrum.01808-24] [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: 07/22/2024] [Accepted: 10/09/2024] [Indexed: 10/30/2024] Open
Abstract
Dysregulation of immune and inflammatory cells around blood vessels and metabolic dysfunction are key mechanisms in the development of pulmonary arterial hypertension (PAH). The homeostasis of the human microbiome plays a crucial role in regulating immune responses and the progression of diseases. For pulmonary arterial hypertension associated with congenital heart disease involving body-lung shunt (PAH-CHD), the potential impact of the microbiome on the "gut-lung axis" remains underexplored. This study recruited 15 healthy individuals and 15 patients with pulmonary arterial hypertension due to congenital heart disease from Fuwai Yunnan Hospital, Chinese Academy of Medical Sciences, and Kunming Children's Hospital. We performed differential analyses of metabolites and microbiota from both the gut and lower respiratory tract for these two groups. The goal was to investigate the "gut-lung axis" microbiome and metabolome profiles in children with PAH-CHD and to analyze the interrelationships between these profiles. Ultimately, we aim to propose the potential value of these profiles in aiding diagnosis. The results indicated that the gut and pulmonary microbiota of children with PAH-CHD are characterized by an increased abundance of beneficial symbionts, which are closely linked to changes in the metabolome. Metabolite functional enrichment analysis revealed energy metabolism reprogramming in the PAH-CHD group, with active metabolic pathways associated with bile acid secretion and carnitine homeostasis. Moreover, the differential expression of metabolites was correlated with right heart function and growth development.IMPORTANCEPrevious studies have primarily focused on the relationship between the gut microbiome and PAH. However, the impact of microbial homeostasis on the progression of PAH-CHD from the perspective of the gut-lung axis has not been adequately elucidated. Our study utilizes an integrated multi-omics approach to report on the differential characteristics of gut and lung microbiota between children with PAH-CHD and reference subjects. We found that microbiota influence the pathological changes and disease manifestations of PAH-CHD through their metabolic activity. Additionally, alterations in metabolites impact the microbial ecological structure. Our findings suggest that modulating the microbiome composition may have positive implications for maintaining and regulating the immune environment and pathological progression of PAH-CHD.
Collapse
Affiliation(s)
- Jiahui Xie
- Department of Cardiovascular Surgery, Fuwai Yunnan Hospital, Chinese Academy of Medical Sciences/Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Xiaoyu Zhang
- Department of Cardiothoracic Surgery, The first hospital of Kunming, Kunming, Yunnan Province, China
| | - Liming Cheng
- Department of Anesthesiology and Surgical Intensive Care Unit, Kunming Children's Hospital, Kunming, Yunnan Province, China
| | - Yao Deng
- Department of Cardiovascular Surgery, Fuwai Yunnan Hospital, Chinese Academy of Medical Sciences/Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Haobo Ren
- Department of Cardiovascular Surgery, Fuwai Yunnan Hospital, Chinese Academy of Medical Sciences/Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Minghua Mu
- Department of Cardiovascular Surgery, Fuwai Yunnan Hospital, Chinese Academy of Medical Sciences/Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Liang Zhao
- Department of Cardiovascular Surgery, Fuwai Yunnan Hospital, Chinese Academy of Medical Sciences/Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Chunjie Mu
- Department of Cardiovascular Surgery, Fuwai Yunnan Hospital, Chinese Academy of Medical Sciences/Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Jiaxiang Chen
- Department of Cardiovascular Surgery, Fuwai Yunnan Hospital, Chinese Academy of Medical Sciences/Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Kai Liu
- Comprehensive Pediatrics, Kunming Children's Hospital, Kunming, Yunnan Province, China
| | - Runwei Ma
- Department of Cardiovascular Surgery, Fuwai Yunnan Hospital, Chinese Academy of Medical Sciences/Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| |
Collapse
|
3
|
Yang C, Liu YH, Zheng HK. Identification of metabolic biomarkers in idiopathic pulmonary arterial hypertension using targeted metabolomics and bioinformatics analysis. Sci Rep 2024; 14:25283. [PMID: 39455660 PMCID: PMC11511845 DOI: 10.1038/s41598-024-76514-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
Pulmonary arterial hypertension (PAH) is a life-threatening disease with a poor prognosis, and metabolic abnormalities play a critical role in its development. This study used metabolomics, machine learning algorithms and bioinformatics to screen for potential metabolic biomarkers associated with the diagnosis of PAH. In this study, plasma samples were collected from 17 patients diagnosed with idiopathic pulmonary arterial hypertension (IPAH) and 20 healthy controls. Plasma metabolomic profiling was performed by high-performance liquid chromatography-mass spectrometry. Gene profiles of PAH patients were obtained from the GEO database. Key differentially expressed metabolites (DEMs) and metabolism-related genes were subsequently identified using machine learning algorithms. Twenty differential plasma metabolites associated with IPAH were identified (VIP score > 1 and p < 0 0.05), and enrichment analysis revealed the arginine biosynthesis pathway as the most altered pathway. Using machine learning models, including least absolute shrinkage and selection operator (LASSO), random forest (RF) and support vector machine (SVM), we extracted key metabolites that correlated with clinical phenotypes. Our results suggested that five metabolites, kynurenine, homoserine, tryptophan, AMP, and spermine, are potential biomarkers for IPAH. Bioinformatics analysis also identified 3 metabolism-related genes, MAPK6, SLC7A11 and CDC42BPA, that are strongly correlated with pulmonary hypertension, demonstrating strong predictive power and clinical relevance. Our findings revealed some key genes associated with metabolism in PH, and provided crucial information about complex metabolic reprogramming signals and may lead to the identification of useful metabolic biomarkers for the diagnosis of PAH.
Collapse
Affiliation(s)
- Chuang Yang
- Department of cardiology, The Second Hospital of Jilin University, No.218 Ziqiang Street, Changchun, 130000, China
| | - Yi-Hang Liu
- Department of cardiology, The Second Hospital of Jilin University, No.218 Ziqiang Street, Changchun, 130000, China
| | - Hai-Kuo Zheng
- Department of cardiology, China-Japan Union Hospital of Jilin University, No.126, Xiantan Street, Changchun, 130033, China.
| |
Collapse
|
4
|
Prajapat SK, Maharana KC, Singh S. Mitochondrial dysfunction in the pathogenesis of endothelial dysfunction. Mol Cell Biochem 2024; 479:1999-2016. [PMID: 37642880 DOI: 10.1007/s11010-023-04835-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 08/14/2023] [Indexed: 08/31/2023]
Abstract
Cardiovascular diseases (CVDs) are a matter of concern worldwide, and mitochondrial dysfunction is one of the major contributing factors. Vascular endothelial dysfunction has a major role in the development of atherosclerosis because of the abnormal chemokine secretion, inflammatory mediators, enhancement of LDL oxidation, cytokine elevation, and smooth muscle cell proliferation. Endothelial cells transfer oxygen from the pulmonary circulatory system to the tissue surrounding the blood vessels, and a majority of oxygen is transferred to the myocardium by endothelial cells, which utilise a small amount of oxygen to generate ATP. Free radicals of oxide are produced by mitochondria, which are responsible for cellular oxygen uptake. Increased mitochondrial ROS generation and reduction in agonist-stimulated eNOS activation and nitric oxide bioavailability were directly linked to the observed change in mitochondrial dynamics, resulting in various CVDs and endothelial dysfunction. Presently, the manuscript mainly focuses on endothelial dysfunction, providing a deep understanding of the various features of mitochondrial mechanisms that are used to modulate endothelial dysfunction. We talk about recent findings and approaches that may make it possible to detect mitochondrial dysfunction as a potential biomarker for risk assessment and diagnosis of endothelial dysfunction. In the end, we cover several targets that may reduce mitochondrial dysfunction through both direct and indirect processes and assess the impact of several different classes of drugs in the context of endothelial dysfunction.
Collapse
Affiliation(s)
- Suresh Kumar Prajapat
- National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park (EPIP) Zandaha Road, Hajipur, Bihar, India
| | - Krushna Ch Maharana
- National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park (EPIP) Zandaha Road, Hajipur, Bihar, India
| | - Sanjiv Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Export Promotions Industrial Park (EPIP), Industrial Area, Dist: Vaishali, Hajipur, Bihar, 844102, India.
| |
Collapse
|
5
|
Zhang H, Chen L, Li J, Sun J, Zhao Q, Wang S, Li G. STAT3 phosphorylation at Tyr705 affects DRP1 (dynamin-related protein 1) controlled-mitochondrial fission during the development of apoptotic-resistance in pulmonary arterial endothelial cells. Genes Genomics 2024; 46:751-762. [PMID: 38733520 PMCID: PMC11208226 DOI: 10.1007/s13258-024-01522-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 05/01/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND The apoptosis-resistant pulmonary arterial endothelial cells (PAECs) are known to be major players in the pulmonary remodeling of pulmonary arterial hypertension (PAH) and exhibit an abnormal metabolic profile with mitochondrial dysfunction. Mitochondrial fission has been shown to regulate the apoptosis of several cell types, but this is largely unexplored in the PAECs. OBJECTIVE The roles of mitochondrial fission control by Dynamin related protein-1 (DRP1) in the development of PAECs apoptosis suppression were investigated in present study and the potential mechanisms behind this were furtherly explored. METHODS The mitochondrial morphology was investigated in PAECs from PAH rats with the pulmonary plexiform lesions, and the relations of it with DRP1 expression and apoptosis were furtherly identified in apoptosis-resistant PAECs induced by hypoxia. PAECs were isolated from rats with severe PAH and from normal subjects, the apoptotic-resistant PAECs were induced by hypoxia. DRP1 gene knockdown was achieved via DRP1-siRNA, DRP1 and STAT3 phosphorylation were blocked using its inhibitors, respectively. Apoptosis was analyzed by flow cytometry, and mitochondrial morphology was investigated by transmission electron microscope and confocal microscopy. RESULTS The PAECs isolated from PAH rats with the pulmonary plexiform-like lesions and displayed lower apoptotic rate with increased DRP1 expression and mitochondrial fragmentation. In addition, similar observations were achieved in apoptosis-resistant PAECs induced by hypoxia. Targeting DRP1 using siRNA and pharmacologic blockade prevented the mitochondrial fission and subsequent apoptotic resistance in PAECs under hypoxia. Mechanistically, STAT3 phosphorylation at Tyr705 was shown to be activated in both PAH and hypoxia-treated PAECs, leading to the regulation of DRP1 expression. Of importance, targeting STAT3Tyr705 phosphorylation prevented DRP1 disruption on apoptosis in PAECs under hypoxia. CONCLUSIONS These data indicated that STAT3 phosphorylation at Tyr705 impacted DRP1-controlled mitochondrial fission during the development of apoptosis-resistance in PAECs, suggesting mitochondrial dynamics may represent a therapeutic target for PAH.
Collapse
Affiliation(s)
- Han Zhang
- Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Beijing, 100029, China
| | - Li Chen
- Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Beijing, 100029, China
| | - Jiachen Li
- Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Beijing, 100029, China
| | - Jiashu Sun
- Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Beijing, 100029, China
| | - Qixu Zhao
- Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Beijing, 100029, China
| | - Sheng Wang
- Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Beijing, 100029, China
| | - Gang Li
- Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Beijing, 100029, China.
| |
Collapse
|
6
|
Yegambaram M, Sun X, Lu Q, Jin Y, Ornatowski W, Soto J, Aggarwal S, Wang T, Tieu K, Gu H, Fineman JR, Black SM. Mitochondrial hyperfusion induces metabolic remodeling in lung endothelial cells by modifying the activities of electron transport chain complexes I and III. Free Radic Biol Med 2024; 210:183-194. [PMID: 37979892 DOI: 10.1016/j.freeradbiomed.2023.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/02/2023] [Accepted: 11/11/2023] [Indexed: 11/20/2023]
Abstract
OBJECTIVE Pulmonary hypertension (PH) is a progressive disease with vascular remodeling as a critical structural alteration. We have previously shown that metabolic reprogramming is an early initiating mechanism in animal models of PH. This metabolic dysregulation has been linked to remodeling the mitochondrial network to favor fission. However, whether the mitochondrial fission/fusion balance underlies the metabolic reprogramming found early in PH development is unknown. METHODS Utilizing a rat early model of PH, in conjunction with cultured pulmonary endothelial cells (PECs), we utilized metabolic flux assays, Seahorse Bioassays, measurements of electron transport chain (ETC) complex activity, fluorescent microscopy, and molecular approaches to investigate the link between the disruption of mitochondrial dynamics and the early metabolic changes that occur in PH. RESULTS We observed increased fusion mediators, including Mfn1, Mfn2, and Opa1, and unchanged fission mediators, including Drp1 and Fis1, in a two-week monocrotaline-induced PH animal model (early-stage PH). We were able to establish a connection between increases in fusion mediator Mfn1 and metabolic reprogramming. Using an adenoviral expression system to enhance Mfn1 levels in pulmonary endothelial cells and utilizing 13C-glucose labeled substrate, we found increased production of 13C lactate and decreased TCA cycle metabolites, revealing a Warburg phenotype. The use of a 13C5-glutamine substrate showed evidence that hyperfusion also induces oxidative carboxylation. The increase in glycolysis was linked to increased hypoxia-inducible factor 1α (HIF-1α) protein levels secondary to the disruption of cellular bioenergetics and higher levels of mitochondrial reactive oxygen species (mt-ROS). The elevation in mt-ROS correlated with attenuated ETC complexes I and III activities. Utilizing a mitochondrial-targeted antioxidant to suppress mt-ROS, limited HIF-1α protein levels, which reduced cellular glycolysis and reestablished mitochondrial membrane potential. CONCLUSIONS Our data connects mitochondrial fusion-mediated mt-ROS to the Warburg phenotype in early-stage PH development.
Collapse
Affiliation(s)
- Manivannan Yegambaram
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33199, USA
| | - Xutong Sun
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33199, USA
| | - Qing Lu
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33199, USA
| | - Yan Jin
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA
| | | | - Jamie Soto
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA
| | - Saurabh Aggarwal
- Department of Cellular Biology & Pharmacology, Howard Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Ting Wang
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33199, USA; Department of Cellular Biology & Pharmacology, Howard Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Kim Tieu
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33199, USA
| | - Haiwei Gu
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33199, USA
| | - Jeffrey R Fineman
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, 94143, USA; Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Stephen M Black
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33199, USA; Department of Cellular Biology & Pharmacology, Howard Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA.
| |
Collapse
|
7
|
Yegambaram M, Sun X, Flores AG, Lu Q, Soto J, Richards J, Aggarwal S, Wang T, Gu H, Fineman JR, Black SM. Novel Relationship between Mitofusin 2-Mediated Mitochondrial Hyperfusion, Metabolic Remodeling, and Glycolysis in Pulmonary Arterial Endothelial Cells. Int J Mol Sci 2023; 24:17533. [PMID: 38139362 PMCID: PMC10744129 DOI: 10.3390/ijms242417533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
The disruption of mitochondrial dynamics has been identified in cardiovascular diseases, including pulmonary hypertension (PH), ischemia-reperfusion injury, heart failure, and cardiomyopathy. Mitofusin 2 (Mfn2) is abundantly expressed in heart and pulmonary vasculature cells at the outer mitochondrial membrane to modulate fusion. Previously, we have reported reduced levels of Mfn2 and fragmented mitochondria in pulmonary arterial endothelial cells (PAECs) isolated from a sheep model of PH induced by pulmonary over-circulation and restoring Mfn2 normalized mitochondrial function. In this study, we assessed the effect of increased expression of Mfn2 on mitochondrial metabolism, bioenergetics, reactive oxygen species production, and mitochondrial membrane potential in control PAECs. Using an adenoviral expression system to overexpress Mfn2 in PAECs and utilizing 13C labeled substrates, we assessed the levels of TCA cycle metabolites. We identified increased pyruvate and lactate production in cells, revealing a glycolytic phenotype (Warburg phenotype). Mfn2 overexpression decreased the mitochondrial ATP production rate, increased the rate of glycolytic ATP production, and disrupted mitochondrial bioenergetics. The increase in glycolysis was linked to increased hypoxia-inducible factor 1α (HIF-1α) protein levels, elevated mitochondrial reactive oxygen species (mt-ROS), and decreased mitochondrial membrane potential. Our data suggest that disrupting the mitochondrial fusion/fission balance to favor hyperfusion leads to a metabolic shift that promotes aerobic glycolysis. Thus, therapies designed to increase mitochondrial fusion should be approached with caution.
Collapse
Affiliation(s)
- Manivannan Yegambaram
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL 34987-2352, USA; (M.Y.); (X.S.); (A.G.F.); (Q.L.); (J.S.); (J.R.); (T.W.); (H.G.)
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL 33199, USA
| | - Xutong Sun
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL 34987-2352, USA; (M.Y.); (X.S.); (A.G.F.); (Q.L.); (J.S.); (J.R.); (T.W.); (H.G.)
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL 33199, USA
| | - Alejandro Garcia Flores
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL 34987-2352, USA; (M.Y.); (X.S.); (A.G.F.); (Q.L.); (J.S.); (J.R.); (T.W.); (H.G.)
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL 33199, USA
| | - Qing Lu
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL 34987-2352, USA; (M.Y.); (X.S.); (A.G.F.); (Q.L.); (J.S.); (J.R.); (T.W.); (H.G.)
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL 33199, USA
| | - Jamie Soto
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL 34987-2352, USA; (M.Y.); (X.S.); (A.G.F.); (Q.L.); (J.S.); (J.R.); (T.W.); (H.G.)
| | - Jaime Richards
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL 34987-2352, USA; (M.Y.); (X.S.); (A.G.F.); (Q.L.); (J.S.); (J.R.); (T.W.); (H.G.)
| | - Saurabh Aggarwal
- Department of Cellular Biology & Pharmacology, Howard Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA;
| | - Ting Wang
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL 34987-2352, USA; (M.Y.); (X.S.); (A.G.F.); (Q.L.); (J.S.); (J.R.); (T.W.); (H.G.)
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL 33199, USA
| | - Haiwei Gu
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL 34987-2352, USA; (M.Y.); (X.S.); (A.G.F.); (Q.L.); (J.S.); (J.R.); (T.W.); (H.G.)
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL 33199, USA
| | - Jeffrey R. Fineman
- Department of Pediatrics, University of California San Francisco, San Francisco, CA 94143, USA;
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94143, USA
| | - Stephen M. Black
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL 34987-2352, USA; (M.Y.); (X.S.); (A.G.F.); (Q.L.); (J.S.); (J.R.); (T.W.); (H.G.)
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL 33199, USA
- Department of Cellular Biology & Pharmacology, Howard Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA;
| |
Collapse
|
8
|
Renaud D, Scholl-Bürgi S, Karall D, Michel M. Comparative Metabolomics in Single Ventricle Patients after Fontan Palliation: A Strong Case for a Targeted Metabolic Therapy. Metabolites 2023; 13:932. [PMID: 37623876 PMCID: PMC10456471 DOI: 10.3390/metabo13080932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 07/28/2023] [Accepted: 08/03/2023] [Indexed: 08/26/2023] Open
Abstract
Most studies on single ventricle (SV) circulation take a physiological or anatomical approach. Although there is a tight coupling between cardiac contractility and metabolism, the metabolic perspective on this patient population is very recent. Early findings point to major metabolic disturbances, with both impaired glucose and fatty acid oxidation in the cardiomyocytes. Additionally, Fontan patients have systemic metabolic derangements such as abnormal glucose metabolism and hypocholesterolemia. Our literature review compares the metabolism of patients with a SV circulation after Fontan palliation with that of patients with a healthy biventricular (BV) heart, or different subtypes of a failing BV heart, by Pubmed review of the literature on cardiac metabolism, Fontan failure, heart failure (HF), ketosis, metabolism published in English from 1939 to 2023. Early evidence demonstrates that SV circulation is not only a hemodynamic burden requiring staged palliation, but also a metabolic issue with alterations similar to what is known for HF in a BV circulation. Alterations of fatty acid and glucose oxidation were found, resulting in metabolic instability and impaired energy production. As reported for patients with BV HF, stimulating ketone oxidation may be an effective treatment strategy for HF in these patients. Few but promising clinical trials have been conducted thus far to evaluate therapeutic ketosis with HF using a variety of instruments, including ketogenic diet, ketone esters, and sodium-glucose co-transporter-2 (SGLT2) inhibitors. An initial trial on a small cohort demonstrated favorable outcomes for Fontan patients treated with SGLT2 inhibitors. Therapeutic ketosis is worth considering in the treatment of Fontan patients, as ketones positively affect not only the myocardial energy metabolism, but also the global Fontan physiopathology. Induced ketosis seems promising as a concerted therapeutic strategy.
Collapse
Affiliation(s)
- David Renaud
- Fundamental and Biomedical Sciences, Paris-Cité University, 75006 Paris, France
- Health Sciences Faculty, Universidad Europea Miguel de Cervantes, 47012 Valladolid, Spain
- Fundacja Recover, 05-124 Skrzeszew, Poland
| | - Sabine Scholl-Bürgi
- Department of Child and Adolescent Health, Division of Pediatrics I—Inherited Metabolic Disorders, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Daniela Karall
- Department of Child and Adolescent Health, Division of Pediatrics I—Inherited Metabolic Disorders, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Miriam Michel
- Department of Child and Adolescent Health, Division of Pediatrics III—Cardiology, Pulmonology, Allergology and Cystic Fibrosis, Medical University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
9
|
Pokharel MD, Marciano DP, Fu P, Franco MC, Unwalla H, Tieu K, Fineman JR, Wang T, Black SM. Metabolic reprogramming, oxidative stress, and pulmonary hypertension. Redox Biol 2023; 64:102797. [PMID: 37392518 PMCID: PMC10363484 DOI: 10.1016/j.redox.2023.102797] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/15/2023] [Accepted: 06/23/2023] [Indexed: 07/03/2023] Open
Abstract
Mitochondria are highly dynamic organelles essential for cell metabolism, growth, and function. It is becoming increasingly clear that endothelial cell dysfunction significantly contributes to the pathogenesis and vascular remodeling of various lung diseases, including pulmonary arterial hypertension (PAH), and that mitochondria are at the center of this dysfunction. The more we uncover the role mitochondria play in pulmonary vascular disease, the more apparent it becomes that multiple pathways are involved. To achieve effective treatments, we must understand how these pathways are dysregulated to be able to intervene therapeutically. We know that nitric oxide signaling, glucose metabolism, fatty acid oxidation, and the TCA cycle are abnormal in PAH, along with alterations in the mitochondrial membrane potential, proliferation, and apoptosis. However, these pathways are incompletely characterized in PAH, especially in endothelial cells, highlighting the urgent need for further research. This review summarizes what is currently known about how mitochondrial metabolism facilitates a metabolic shift in endothelial cells that induces vascular remodeling during PAH.
Collapse
Affiliation(s)
- Marissa D Pokharel
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA; Department of Cellular Biology & Pharmacology, Howard Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - David P Marciano
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA; Department of Cellular Biology & Pharmacology, Howard Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Panfeng Fu
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33199, USA
| | - Maria Clara Franco
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA; Department of Cellular Biology & Pharmacology, Howard Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Hoshang Unwalla
- Department of Immunology and Nano-Medicine, Howard Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Kim Tieu
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33199, USA
| | - Jeffrey R Fineman
- Department of Pediatrics, The University of California San Francisco, San Francisco, CA, 94143, USA; Cardiovascular Research Institute, The University of California San Francisco, San Francisco, CA, 94143, USA
| | - Ting Wang
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33199, USA
| | - Stephen M Black
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA; Department of Cellular Biology & Pharmacology, Howard Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33199, USA.
| |
Collapse
|
10
|
Lu Q, Sun X, Yegambaram M, Ornatowski W, Wu X, Wang H, Garcia-Flores A, Da Silva V, Zemskov EA, Tang H, Fineman JR, Tieu K, Wang T, Black SM. Nitration-mediated activation of the small GTPase RhoA stimulates cellular glycolysis through enhanced mitochondrial fission. J Biol Chem 2023; 299:103067. [PMID: 36841483 PMCID: PMC10060112 DOI: 10.1016/j.jbc.2023.103067] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 02/27/2023] Open
Abstract
Mitochondrial fission and a Warburg phenotype of increased cellular glycolysis are involved in the pathogenesis of pulmonary hypertension (PH). The purpose of this study was to determine whether increases in mitochondrial fission are involved in a glycolytic switch in pulmonary arterial endothelial cells (PAECs). Mitochondrial fission is increased in PAEC isolated from a sheep model of PH induced by pulmonary overcirculation (Shunt PAEC). In Shunt PAEC we identified increases in the S616 phosphorylation responsible for dynamin-related protein 1 (Drp1) activation, the mitochondrial redistribution of Drp1, and increased cellular glycolysis. Reducing mitochondrial fission attenuated cellular glycolysis in Shunt PAEC. In addition, we observed nitration-mediated activation of the small GTPase RhoA in Shunt PAEC, and utilizing a nitration-shielding peptide, NipR1 attenuated RhoA nitration and reversed the Warburg phenotype. Thus, our data identify a novel link between RhoA, mitochondrial fission, and cellular glycolysis and suggest that targeting RhoA nitration could have therapeutic benefits for treating PH.
Collapse
Affiliation(s)
- Qing Lu
- Center of Translational Science, Florida International University, Port St Lucie, Florida, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, Florida, USA
| | - Xutong Sun
- Center of Translational Science, Florida International University, Port St Lucie, Florida, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, Florida, USA
| | | | - Wojciech Ornatowski
- Department of Medicine, University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Xiaomin Wu
- Department of Medicine, University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Hui Wang
- Department of Medicine, University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Alejandro Garcia-Flores
- Center of Translational Science, Florida International University, Port St Lucie, Florida, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, Florida, USA
| | - Victoria Da Silva
- Center of Translational Science, Florida International University, Port St Lucie, Florida, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, Florida, USA
| | - Evgeny A Zemskov
- Center of Translational Science, Florida International University, Port St Lucie, Florida, USA; Department of Cellular Biology & Pharmacology, Howard Wertheim College of Medicine, Florida International University, Miami, Florida, USA
| | - Haiyang Tang
- Center of Translational Science, Florida International University, Port St Lucie, Florida, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, Florida, USA
| | - Jeffrey R Fineman
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA; Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, USA
| | - Kim Tieu
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, Florida, USA
| | - Ting Wang
- Center of Translational Science, Florida International University, Port St Lucie, Florida, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, Florida, USA
| | - Stephen M Black
- Center of Translational Science, Florida International University, Port St Lucie, Florida, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, Florida, USA; Department of Cellular Biology & Pharmacology, Howard Wertheim College of Medicine, Florida International University, Miami, Florida, USA.
| |
Collapse
|
11
|
Zvyagina VI, Belskikh ES. Comparative Assessment of the Functional Activity of Rat Epididymal Mitochondria in Oxidative Stress Induced by Hyperhomocysteinemia and L-NAME Administration. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022020065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
12
|
El-Sherbiny HR, El-Shalofy AS, Samir H. Exogenous L-carnitine Administration Ameliorates the Adverse Effects of Heat Stress on Testicular Hemodynamics, Echotexture, and Total Antioxidant Capacity in Rams. Front Vet Sci 2022; 9:860771. [PMID: 35464382 PMCID: PMC9019560 DOI: 10.3389/fvets.2022.860771] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 03/03/2022] [Indexed: 12/23/2022] Open
Abstract
Heat stress (HS) diminishes the testicular antioxidant defense systems, which adversely affect the testicular blood perfusion. Improving the testicular hemodynamics during HS conditions is of a great impact on the whole reproductive performance in rams. This study aimed to evaluate the ameliorative effects of L-carnitine (LC) on the testicular blood flow and echotextures and also on the total antioxidants (TAC) and nitric oxide (NO) concentrations in the serum during HS conditions in rams. Testicular blood flow was evaluated through scanning of the supra-testicular artery (STA) spectral patterns through pulsed Doppler ultrasonography [peak systolic velocity (PSV), end-diastolic velocity (EDV), time average maximum velocity (TAMAX), resistive index (RI), and pulsatility index (PI)], while the echotexture assessment of testicular parenchyma was performed by a computerized software program. Moreover, TAC and NO concentrations were assayed colorimetrically using the spectrophotometer. There were significant decreases (P < 0.05) in values of PSV at 48 and 168 h (23.45 ± 0.39 and 23.37 ± 1.41 cm/s, respectively), and TAMAX at 1, 48, and 168 h (17.65 ± 0.95, 17.5 ± 0.13, and 16.9 ± 1.05 cm/s, respectively) after LC administration compared to just before administration (31.92 ± 1.13 and 21.58 ± 0.92 cm/s, respectively). Values of RI and PI of the examined STA significantly decreased, especially at 1 h for RI (0.45 ± 0.02) and 1 and 48 h for PI (0.66 ± 0.06 and 0.65 ± 0.05, respectively) after LC treatment to 0 h (0.55 ± 0.03 and 0.84 ± 0.06, respectively). The EDV values did not show any significant (P < 0.05) changes in all the experimental time points. There were significant (P < 0.05) increases in the values of pixel intensity of the testicular parenchyma, especially at 1 and 168 h (78.71 ± 2.50 and 88.56 ± 4.10, respectively) after LC administration, compared to just before administration (69.40 ± 4.75). Serum NO levels tend to increase after LC administration (P = 0.07) concerning just before administration. While TAC values showed significant gradual increase and reached the highest values at 168 h (2.75 ± 0.58 mM/l) after LC administration, compared to 0 h (1.12 ± 0.05 mM/l). In conclusion, exogenous LC administration ameliorates testicular hemodynamic disruptions, as measured by spectral Doppler ultrasonography, via augmentation of the rams' total antioxidant capacity under HS conditions.
Collapse
|
13
|
Metabolism, Mitochondrial Dysfunction, and Redox Homeostasis in Pulmonary Hypertension. Antioxidants (Basel) 2022; 11:antiox11020428. [PMID: 35204311 PMCID: PMC8869288 DOI: 10.3390/antiox11020428] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 02/05/2023] Open
Abstract
Pulmonary hypertension (PH) represents a group of disorders characterized by elevated mean pulmonary artery (PA) pressure, progressive right ventricular failure, and often death. Some of the hallmarks of pulmonary hypertension include endothelial dysfunction, intimal and medial proliferation, vasoconstriction, inflammatory infiltration, and in situ thrombosis. The vascular remodeling seen in pulmonary hypertension has been previously linked to the hyperproliferation of PA smooth muscle cells. This excess proliferation of PA smooth muscle cells has recently been associated with changes in metabolism and mitochondrial biology, including changes in glycolysis, redox homeostasis, and mitochondrial quality control. In this review, we summarize the molecular mechanisms that have been reported to contribute to mitochondrial dysfunction, metabolic changes, and redox biology in PH.
Collapse
|
14
|
Liang S, Yegambaram M, Wang T, Wang J, Black SM, Tang H. Mitochondrial Metabolism, Redox, and Calcium Homeostasis in Pulmonary Arterial Hypertension. Biomedicines 2022; 10:biomedicines10020341. [PMID: 35203550 PMCID: PMC8961787 DOI: 10.3390/biomedicines10020341] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/25/2022] [Accepted: 01/26/2022] [Indexed: 02/06/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease characterized by elevated pulmonary arterial pressure due to increased pulmonary vascular resistance, secondary to sustained pulmonary vasoconstriction and excessive obliterative pulmonary vascular remodeling. Work over the last decade has led to the identification of a critical role for metabolic reprogramming in the PAH pathogenesis. It is becoming clear that in addition to its role in ATP generation, the mitochondrion is an important organelle that regulates complex and integrative metabolic- and signal transduction pathways. This review focuses on mitochondrial metabolism alterations that occur in deranged pulmonary vessels and the right ventricle, including abnormalities in glycolysis and glucose oxidation, fatty acid oxidation, glutaminolysis, redox homeostasis, as well as iron and calcium metabolism. Further understanding of these mitochondrial metabolic mechanisms could provide viable therapeutic approaches for PAH patients.
Collapse
Affiliation(s)
- Shuxin Liang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China; (S.L.); (J.W.)
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Manivannan Yegambaram
- Center for Translational Science, 11350 SW Village Pkwy, Port St. Lucie, FL 34987, USA; (M.Y.); (T.W.)
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Port St. Lucie, FL 34987, USA
| | - Ting Wang
- Center for Translational Science, 11350 SW Village Pkwy, Port St. Lucie, FL 34987, USA; (M.Y.); (T.W.)
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Port St. Lucie, FL 34987, USA
| | - Jian Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China; (S.L.); (J.W.)
| | - Stephen M. Black
- Center for Translational Science, 11350 SW Village Pkwy, Port St. Lucie, FL 34987, USA; (M.Y.); (T.W.)
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Port St. Lucie, FL 34987, USA
- Department of Cellular Biology & Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Port St. Lucie, FL 34987, USA
- Correspondence: (S.M.B.); (H.T.)
| | - Haiyang Tang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China; (S.L.); (J.W.)
- Correspondence: (S.M.B.); (H.T.)
| |
Collapse
|
15
|
Zemskov EA, Wu X, Aggarwal S, Yegambaram M, Gross C, Lu Q, Wang H, Tang H, Wang T, Black SM. Nitration of protein kinase G-Iα modulates cyclic nucleotide crosstalk via phosphodiesterase 3A: Implications for acute lung injury. J Biol Chem 2021; 297:100946. [PMID: 34252457 PMCID: PMC8342797 DOI: 10.1016/j.jbc.2021.100946] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 06/22/2021] [Accepted: 07/08/2021] [Indexed: 12/05/2022] Open
Abstract
Phosphodiesterase 3A (PDE3A) selectively cleaves the phosphodiester bond of cAMP and is inhibited by cGMP, making it an important regulator of cAMP-cGMP signaling crosstalk in the pulmonary vasculature. In addition, the nitric oxide-cGMP axis is known to play an important role in maintaining endothelial barrier function. However, the potential role of protein kinase G-Iα (PKG-Iα) in this protective process is unresolved and was the focus of our study. We describe here a novel mechanism regulating PDE3A activity, which involves a PKG-Iα-dependent inhibitory phosphorylation of PDE3A at serine 654. We also show that this phosphorylation is critical for maintaining intracellular cAMP levels in the pulmonary endothelium and endothelial barrier integrity. In an animal model of acute lung injury (ALI) induced by challenging mice with lipopolysaccharide (LPS), an increase in PDE3 activity and a decrease in cAMP levels in lung tissue was associated with reduced PKG activity upon PKG-Iα nitration at tyrosine 247. The peroxynitrite scavenger manganese (III) tetrakis(1-methyl-4-pyridyl)porphyrin prevented this increase in PDE3 activity in LPS-exposed lungs. In addition, site-directed mutagenesis of PDE3A to replace serine 654 with alanine yielded a mutant protein that was insensitive to PKG-dependent regulation. Taken together, our data demonstrate a novel functional link between nitrosative stress induced by LPS during ALI and the downregulation of barrier-protective intracellular cAMP levels. Our data also provide new evidence that PKG-Iα is critical for endothelial barrier maintenance and that preservation of its catalytic activity may be efficacious in ALI therapy.
Collapse
Affiliation(s)
- Evgeny A Zemskov
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Xiaomin Wu
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Saurabh Aggarwal
- Vascular Biology Center, Augusta University, Augusta, Georgia, USA
| | | | - Christine Gross
- Vascular Biology Center, Augusta University, Augusta, Georgia, USA
| | - Qing Lu
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Hui Wang
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona, USA; College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Haiyang Tang
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona, USA; College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China; Center for Translational Science, Florida International University, Port Saint Lucie, Florida, USA
| | - Ting Wang
- Center for Translational Science, Florida International University, Port Saint Lucie, Florida, USA; Department of Internal Medicine, The University of Arizona, Phoenix, Arizona, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, Florida, USA
| | - Stephen M Black
- Center for Translational Science, Florida International University, Port Saint Lucie, Florida, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, Florida, USA; Cellular Biology & Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA.
| |
Collapse
|
16
|
Abstract
Pulmonary arterial hypertension (PAH) is characterized by impaired regulation of pulmonary hemodynamics and vascular growth. Alterations of metabolism and bioenergetics are increasingly recognized as universal hallmarks of PAH, as metabolic abnormalities are identified in lungs and hearts of patients, animal models of the disease, and cells derived from lungs of patients. Mitochondria are the primary organelle critically mediating the complex and integrative metabolic pathways in bioenergetics, biosynthetic pathways, and cell signaling. Here, we review the alterations in metabolic pathways that are linked to the pathologic vascular phenotype of PAH, including abnormalities in glycolysis and glucose oxidation, fatty acid oxidation, glutaminolysis, arginine metabolism, one-carbon metabolism, the reducing and oxidizing cell environment, and the tricarboxylic acid cycle, as well as the effects of PAH-associated nuclear and mitochondrial mutations on metabolism. Understanding of the metabolic mechanisms underlying PAH provides important knowledge for the design of new therapeutics for treatment of patients.
Collapse
Affiliation(s)
- Weiling Xu
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA;
| | - Allison J Janocha
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA;
| | - Serpil C Erzurum
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA; .,Respiratory Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| |
Collapse
|
17
|
Wang H, Sun X, Lu Q, Zemskov EA, Yegambaram M, Wu X, Wang T, Tang H, Black SM. The mitochondrial redistribution of eNOS is involved in lipopolysaccharide induced inflammasome activation during acute lung injury. Redox Biol 2021; 41:101878. [PMID: 33578126 PMCID: PMC7879038 DOI: 10.1016/j.redox.2021.101878] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/17/2021] [Accepted: 01/21/2021] [Indexed: 01/03/2023] Open
Abstract
Acute lung injury (ALI) is a devastating clinical syndrome with no effective therapies. Inflammasome activation has been reported to play a critical role in the initiation and progression of ALI. The molecular mechanisms involved in regulating the activation of inflammasome in ALI remains unresolved, although increases in mitochondrial derived reactive oxygen species (mito-ROS) are involved. Our previous work has shown that the mitochondrial redistribution of uncoupled eNOS impairs mitochondrial bioenergetics and increases mito-ROS generation. Thus, the focus of our study was to determine if lipopolysaccharide (LPS)-mediated inflammasome activation involves the mitochondrial redistribution of uncoupled eNOS. Our data show that the increase in mito-ROS involved in LPS-mediated inflammasome activation is associated with the disruption of mitochondrial bioenergetics in human lung microvascular endothelial cells (HLMVEC) and the mitochondrial redistribution of eNOS. These effects are dependent on RhoA-ROCK signaling and are mediated via increased phosphorylation of eNOS at Threonine (T)-495. A derivative of the mitochondrial targeted Szeto-Schiller peptide (SSP) attached to the antioxidant Tiron (T-SSP), significantly attenuated LPS-mediated mito-ROS generation and inflammasome activation in HLMVEC. Further, T-SSP attenuated mitochondrial superoxide production in a mouse model of sepsis induced ALI. This in turn significantly reduced the inflammatory response and attenuated lung injury. Thus, our findings show that the mitochondrial redistribution of uncoupled eNOS is intimately involved in the activation of the inflammatory response in ALI and implicate attenuating mito-ROS as a therapeutic strategy in humans.
Collapse
Affiliation(s)
- Hui Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China; Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA
| | - Xutong Sun
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA
| | - Qing Lu
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA
| | - Evgeny A Zemskov
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA
| | - Manivannan Yegambaram
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA
| | - Xiaomin Wu
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA
| | - Ting Wang
- Department of Internal Medicine, The University of Arizona Health Sciences, Phoenix, AZ, USA
| | - Haiyang Tang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China; Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA.
| | - Stephen M Black
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
18
|
Lu Q, Zemskov EA, Sun X, Wang H, Yegambaram M, Wu X, Garcia-Flores A, Song S, Tang H, Kangath A, Cabanillas GZ, Yuan JXJ, Wang T, Fineman JR, Black SM. Activation of the mechanosensitive Ca 2+ channel TRPV4 induces endothelial barrier permeability via the disruption of mitochondrial bioenergetics. Redox Biol 2021; 38:101785. [PMID: 33221570 PMCID: PMC7691184 DOI: 10.1016/j.redox.2020.101785] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/29/2020] [Accepted: 11/01/2020] [Indexed: 12/20/2022] Open
Abstract
Mechanical ventilation is a life-saving intervention in critically ill patients with respiratory failure due to acute respiratory distress syndrome (ARDS), a refractory lung disease with an unacceptable high mortality rate. Paradoxically, mechanical ventilation also creates excessive mechanical stress that directly augments lung injury, a syndrome known as ventilator-induced lung injury (VILI). The specific mechanisms involved in VILI-induced pulmonary capillary leakage, a key pathologic feature of VILI are still far from resolved. The mechanoreceptor, transient receptor potential cation channel subfamily V member 4, TRPV4 plays a key role in the development of VILI through unresolved mechanism. Endothelial nitric oxide synthase (eNOS) uncoupling plays an important role in sepsis-mediated ARDS so in this study we investigated whether there is a role for eNOS uncoupling in the barrier disruption associated with TRPV4 activation during VILI. Our data indicate that the TRPV4 agonist, 4α-Phorbol 12,13-didecanoate (4αPDD) induces pulmonary arterial endothelial cell (EC) barrier disruption through the disruption of mitochondrial bioenergetics. Mechanistically, this occurs via the mitochondrial redistribution of uncoupled eNOS secondary to a PKC-dependent phosphorylation of eNOS at Threonine 495 (T495). A specific decoy peptide to prevent T495 phosphorylation reduced eNOS uncoupling and mitochondrial redistribution and preserved PAEC barrier function under 4αPDD challenge. Further, our eNOS decoy peptide was able to preserve lung vascular integrity in a mouse model of VILI. Thus, we have revealed a functional link between TRPV4 activation, PKC-dependent eNOS phosphorylation at T495, and EC barrier permeability. Reducing pT495-eNOS could be a new therapeutic approach for the prevention of VILI.
Collapse
Affiliation(s)
- Qing Lu
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA
| | - Evgeny A Zemskov
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA
| | - Xutong Sun
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA
| | - Hui Wang
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA; College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Manivannan Yegambaram
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA
| | - Xiaomin Wu
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA
| | - Alejandro Garcia-Flores
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA
| | - Shanshan Song
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA
| | - Haiyang Tang
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA; College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Archana Kangath
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA
| | - Gabriela Zubiate Cabanillas
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA; Department of Chemist-Biological Sciences, Universidad de Sonora, Hermosillo, SON, Mexico
| | - Jason X-J Yuan
- Department of Medicine, University of California, San Diego, CA, USA
| | - Ting Wang
- Department of Internal Medicine, The University of Arizona Health Sciences, Phoenix, AZ, USA
| | - Jeffrey R Fineman
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA; Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Stephen M Black
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
19
|
Mamazhakypov A, Weiß A, Zukunft S, Sydykov A, Kojonazarov B, Wilhelm J, Vroom C, Petrovic A, Kosanovic D, Weissmann N, Seeger W, Fleming I, Iglarz M, Grimminger F, Ghofrani HA, Pullamsetti SS, Schermuly RT. Effects of macitentan and tadalafil monotherapy or their combination on the right ventricle and plasma metabolites in pulmonary hypertensive rats. Pulm Circ 2020; 10:2045894020947283. [PMID: 33240483 PMCID: PMC7672745 DOI: 10.1177/2045894020947283] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 07/10/2020] [Indexed: 12/19/2022] Open
Abstract
Pulmonary arterial hypertension is a severe respiratory disease characterized by pulmonary artery remodeling. RV dysfunction and dysregulated circulating metabolomics are associated with adverse outcomes in pulmonary arterial hypertension. We investigated effects of tadalafil and macitentan alone or in combination on the RV and plasma metabolomics in SuHx and PAB models. For SuHx model, rats were injected with SU5416 and exposed to hypoxia for three weeks and then were returned to normoxia and treated with either tadalafil (10 mg/kg in chow) or macitentan (10 mg/kg in chow) or their combination (both 10 mg/kg in chow) for two weeks. For PAB model, rats were subjected to either sham or PAB surgery for three weeks and treated with above-mentioned drugs from week 1 to week 3. Following terminal echocardiographic and hemodynamic measurements, tissue samples were collected for metabolomic, histological and gene expression analysis. Both SuHx and PAB rats developed RV remodeling/dysfunction with severe and mild plasma metabolomic alterations, respectively. In SuHx rats, tadalafil and macitentan alone or in combination improved RV remodeling/function with the effects of macitentan and combination therapy being superior to tadalafil. All therapies similarly attenuated SuHx-induced changes in plasma metabolomics. In PAB rats, only macitentan improved RV remodeling/function, while only tadalafil attenuated PAB-induced changes in plasma metabolomics.
Collapse
Affiliation(s)
- Argen Mamazhakypov
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center, Member of the German Lung Center (DZL), Justus-Liebig-University Giessen, Giessen, Germany
| | - Astrid Weiß
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center, Member of the German Lung Center (DZL), Justus-Liebig-University Giessen, Giessen, Germany
| | - Sven Zukunft
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany & German Center of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt am Main, Germany
| | - Akylbek Sydykov
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center, Member of the German Lung Center (DZL), Justus-Liebig-University Giessen, Giessen, Germany
| | - Baktybek Kojonazarov
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center, Member of the German Lung Center (DZL), Justus-Liebig-University Giessen, Giessen, Germany
| | - Jochen Wilhelm
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center, Member of the German Lung Center (DZL), Justus-Liebig-University Giessen, Giessen, Germany
| | - Christina Vroom
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center, Member of the German Lung Center (DZL), Justus-Liebig-University Giessen, Giessen, Germany
| | - Aleksandar Petrovic
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center, Member of the German Lung Center (DZL), Justus-Liebig-University Giessen, Giessen, Germany
| | - Djuro Kosanovic
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center, Member of the German Lung Center (DZL), Justus-Liebig-University Giessen, Giessen, Germany.,Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Norbert Weissmann
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center, Member of the German Lung Center (DZL), Justus-Liebig-University Giessen, Giessen, Germany
| | - Werner Seeger
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center, Member of the German Lung Center (DZL), Justus-Liebig-University Giessen, Giessen, Germany.,Department of Lung Development and Remodelling, Max-Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany & German Center of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt am Main, Germany
| | - Marc Iglarz
- Actelion Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Friedrich Grimminger
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center, Member of the German Lung Center (DZL), Justus-Liebig-University Giessen, Giessen, Germany
| | - Hossein A Ghofrani
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center, Member of the German Lung Center (DZL), Justus-Liebig-University Giessen, Giessen, Germany
| | - Soni S Pullamsetti
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center, Member of the German Lung Center (DZL), Justus-Liebig-University Giessen, Giessen, Germany.,Department of Lung Development and Remodelling, Max-Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Ralph T Schermuly
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center, Member of the German Lung Center (DZL), Justus-Liebig-University Giessen, Giessen, Germany
| |
Collapse
|
20
|
RAC1 nitration at Y 32 IS involved in the endothelial barrier disruption associated with lipopolysaccharide-mediated acute lung injury. Redox Biol 2020; 38:101794. [PMID: 33248422 PMCID: PMC7664366 DOI: 10.1016/j.redox.2020.101794] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 11/06/2020] [Accepted: 11/07/2020] [Indexed: 02/06/2023] Open
Abstract
Acute lung injury (ALI), a devastating illness induced by systemic inflammation e.g., sepsis or local lung inflammation e.g., COVID-19 mediated severe pneumonia, has an unacceptably high mortality and has no effective therapy. ALI is associated with increased pulmonary microvascular hyperpermeability and alveolar flooding. The small Rho GTPases, RhoA and Rac1 are central regulators of vascular permeability through cytoskeleton rearrangements. RhoA and Rac1 have opposing functional outcome: RhoA induces an endothelial contractile phenotype and barrier disruption, while Rac1 stabilizes endothelial junctions and increases barrier integrity. In ALI, RhoA activity is increased while Rac1 activity is reduced. We have shown that the activation of RhoA in lipopolysaccharide (LPS)-mediated ALI, is dependent, at least in part, on a single nitration event at tyrosine (Y)34. Thus, the purpose of this study was to determine if the inhibition of Rac1 is also dependent on its nitration. Our data show that Rac1 inhibition by LPS is associated with its nitration that mass spectrometry identified as Y32, within the switch I region adjacent to the nucleotide-binding site. Using a molecular modeling approach, we designed a nitration shielding peptide for Rac1, designated NipR2 (nitration inhibitor peptide for the Rho GTPases 2), which attenuated the LPS-induced nitration of Rac1 at Y32, preserves Rac1 activity and attenuates the LPS-mediated disruption of the endothelial barrier in human lung microvascular endothelial cells (HLMVEC). Using a murine model of ALI induced by intratracheal installation of LPS we found that NipR2 successfully prevented Rac1 nitration and Rac1 inhibition, and more importantly attenuated pulmonary inflammation, reduced lung injury and prevented the loss of lung function. Together, our data identify a new post-translational mechanism of Rac1 inhibition through its nitration at Y32. As NipR2 also reduces sepsis induced ALI in the mouse lung, we conclude that Rac1 nitration is a therapeutic target in ALI. Endotoxin exposure induces site specific nitration of Rac1 at Y32 via peroxynitrite stress. Rac1 nitration at Y32 leads to persistent Rac GTPase inhibition and endothelial barrier disruption. Novel Rac1 nitration shielding peptide, NipR2 blocks Rac1 nitration and rescues endotoxin induced lung inflammation. NipR2 is potentially an effective therapy for sepsis induced lung injury by targeting Rac1 Y32 nitration.
Collapse
|
21
|
Sun X, Lu Q, Yegambaram M, Kumar S, Qu N, Srivastava A, Wang T, Fineman JR, Black SM. TGF-β1 attenuates mitochondrial bioenergetics in pulmonary arterial endothelial cells via the disruption of carnitine homeostasis. Redox Biol 2020; 36:101593. [PMID: 32554303 PMCID: PMC7303661 DOI: 10.1016/j.redox.2020.101593] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/19/2020] [Accepted: 05/21/2020] [Indexed: 12/23/2022] Open
Abstract
Transforming growth factor beta-1 (TGF-β1) signaling is increased and mitochondrial function is decreased in multiple models of pulmonary hypertension (PH) including lambs with increased pulmonary blood flow (PBF) and pressure (Shunt). However, the potential link between TGF-β1 and the loss of mitochondrial function has not been investigated and was the focus of our investigations. Our data indicate that exposure of pulmonary arterial endothelial cells (PAEC) to TGF-β1 disrupted mitochondrial function as determined by enhanced mitochondrial ROS generation, decreased mitochondrial membrane potential, and disrupted mitochondrial bioenergetics. These events resulted in a decrease in cellular ATP levels, decreased hsp90/eNOS interactions and attenuated shear-mediated NO release. TGF-β1 induced mitochondrial dysfunction was linked to a nitration-mediated activation of Akt1 and the subsequent mitochondrial translocation of endothelial NO synthase (eNOS) resulting in the nitration of carnitine acetyl transferase (CrAT) and the disruption of carnitine homeostasis. The increase in Akt1 nitration correlated with increased NADPH oxidase activity associated with increased levels of p47phox, p67phox, and Rac1. The increase in NADPH oxidase was associated with a decrease in peroxisome proliferator-activated receptor type gamma (PPARγ) and the PPARγ antagonist, GW9662, was able to mimic the disruptive effect of TGF-β1 on mitochondrial bioenergetics. Together, our studies reveal for the first time, that TGF-β1 can disrupt mitochondrial function through the disruption of cellular carnitine homeostasis and suggest that stimulating carinitine homeostasis may be an avenue to treat pulmonary vascular disease.
Collapse
Affiliation(s)
- Xutong Sun
- Department of Medicine, Arizona Health Sciences Center, University of Arizona, Tucson, AZ, 85721, USA
| | - Qing Lu
- Department of Medicine, Arizona Health Sciences Center, University of Arizona, Tucson, AZ, 85721, USA
| | - Manivannan Yegambaram
- Department of Medicine, Arizona Health Sciences Center, University of Arizona, Tucson, AZ, 85721, USA
| | - Sanjiv Kumar
- Center for Blood Disorders, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Ning Qu
- Department of Medicine, Arizona Health Sciences Center, University of Arizona, Tucson, AZ, 85721, USA
| | - Anup Srivastava
- Department of Medicine, Arizona Health Sciences Center, University of Arizona, Tucson, AZ, 85721, USA
| | - Ting Wang
- Department of Internal Medicine University of Arizona, Phoenix, AZ, 85004, The Department of Pediatrics and the Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Jeffrey R Fineman
- Department of Internal Medicine University of Arizona, Phoenix, AZ, 85004, The Department of Pediatrics and the Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Stephen M Black
- Department of Medicine, Arizona Health Sciences Center, University of Arizona, Tucson, AZ, 85721, USA.
| |
Collapse
|
22
|
Chen C, Luo F, Wu P, Huang Y, Das A, Chen S, Chen J, Hu X, Li F, Fang Z, Zhou S. Metabolomics reveals metabolite changes of patients with pulmonary arterial hypertension in China. J Cell Mol Med 2020; 24:2484-2496. [PMID: 31945804 PMCID: PMC7028857 DOI: 10.1111/jcmm.14937] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 12/01/2019] [Accepted: 12/16/2019] [Indexed: 12/12/2022] Open
Abstract
The specific mechanism of pulmonary arterial hypertension (PAH) remains elusive. The present study aimed to explore the underlying mechanism of PAH through the identity of novel biomarkers for PAH using metabolomics approach. Serum samples from 40 patients with idiopathic PAH (IPAH), 20 patients with congenital heart disease-associated PAH (CHD-PAH) and 20 healthy controls were collected and analysed by ultra-high-performance liquid chromatography coupled with high-resolution mass spectrometry (UPLC-HRMS). Orthogonal partial least square-discriminate analysis (OPLS-DA) was applied to screen potential biomarkers. These results were validated in monocrotaline (MCT)-induced PAH rat model. The OPLS-DA model was successful in screening distinct metabolite signatures which distinguished IPAH and CHD-PAH patients from healthy controls, respectively (26 and 15 metabolites). Unbiased analysis from OPLS-DA identified 31 metabolites from PAH patients which were differentially regulated compared to the healthy controls. Our analysis showed dysregulation of the different metabolic pathways, including lipid metabolism, glucose metabolism, amino acid metabolism and phospholipid metabolism pathways in PAH patients compared to their healthy counterpart. Among these metabolites from dysregulated metabolic pathways, a panel of metabolites from lipid metabolism and fatty acid oxidation (lysophosphatidylcholine, phosphatidylcholine, perillic acid, palmitoleic acid, N-acetylcholine-d-sphingomyelin, oleic acid, palmitic acid and 2-Octenoylcarnitine metabolites) were found to have a close association with PAH. The results from the analysis of both real-time quantitative PCR and Western blot showed that expression of LDHA, CD36, FASN, PDK1 GLUT1 and CPT-1 in right heart/lung were significantly up-regulated in MCT group than the control group.
Collapse
Affiliation(s)
- Chenyang Chen
- Department of Cardiovascular MedicineThe Second Xiangya HospitalCentral South UniversityChangshaChina
- Department of Cardiovascular MedicineThe Third Xiangya HospitalCentral South UniversityChangshaChina
| | - Fei Luo
- Department of Cardiovascular MedicineThe Second Xiangya HospitalCentral South UniversityChangshaChina
| | - Panyun Wu
- Department of Cardiovascular MedicineThe Second Xiangya HospitalCentral South UniversityChangshaChina
| | - Yiyuan Huang
- Department of Cardiovascular MedicineThe Second Xiangya HospitalCentral South UniversityChangshaChina
| | - Avash Das
- Department of Molecular GeneticsUniversity of Texas Southwestern Medical CenterDallasTXUSA
| | - Shenglan Chen
- Department of Cardiovascular MedicineThe Second Xiangya HospitalCentral South UniversityChangshaChina
| | - Jingyuan Chen
- Department of Cardiovascular MedicineThe Second Xiangya HospitalCentral South UniversityChangshaChina
| | - Xinqun Hu
- Department of Cardiovascular MedicineThe Second Xiangya HospitalCentral South UniversityChangshaChina
| | - Fei Li
- Kunming Institute of BotanyChinese Academy of SciencesKunmingChina
| | - Zhenfei Fang
- Department of Cardiovascular MedicineThe Second Xiangya HospitalCentral South UniversityChangshaChina
| | - Shenhua Zhou
- Department of Cardiovascular MedicineThe Second Xiangya HospitalCentral South UniversityChangshaChina
| |
Collapse
|
23
|
Suliman HB, Nozik-Grayck E. Mitochondrial Dysfunction: Metabolic Drivers of Pulmonary Hypertension. Antioxid Redox Signal 2019; 31:843-857. [PMID: 30604624 PMCID: PMC6751393 DOI: 10.1089/ars.2018.7705] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significance: Pulmonary hypertension (PH) is a progressive disease characterized by pulmonary vascular remodeling and lung vasculopathy. The disease displays progressive dyspnea, pulmonary artery uncoupling and right ventricular (RV) dysfunction. The overall survival rate is ranging from 28-72%. Recent Advances: The molecular events that promote the development of PH are complex and incompletely understood. Metabolic impairment has been proposed to contribute to the pathophysiology of PH with evidence for mitochondrial dysfunction involving the electron transport chain proteins, antioxidant enzymes, apoptosis regulators, and mitochondrial quality control. Critical Issues: It is vital to characterize the mechanisms by which mitochondrial dysfunction contribute to PH pathogenesis. This review focuses on the currently available publications that supports mitochondrial mechanisms in PH pathophysiology. Future Directions: Further studies of these metabolic mitochondrial alterations in PH could be viable targets of diagnostic and therapeutic intervention.
Collapse
Affiliation(s)
- Hagir B Suliman
- Department of Anesthesiology, Duke University Medical Centers, Durham, North Carolina
| | - Eva Nozik-Grayck
- Department of Pediatrics, Cardiovascular Pulmonary Research Labs and Pediatric Critical Care Medicine, University of Colorado Denver, Aurora, Colorado
| |
Collapse
|
24
|
Zemskov EA, Lu Q, Ornatowski W, Klinger CN, Desai AA, Maltepe E, Yuan JXJ, Wang T, Fineman JR, Black SM. Biomechanical Forces and Oxidative Stress: Implications for Pulmonary Vascular Disease. Antioxid Redox Signal 2019; 31:819-842. [PMID: 30623676 PMCID: PMC6751394 DOI: 10.1089/ars.2018.7720] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significance: Oxidative stress in the cell is characterized by excessive generation of reactive oxygen species (ROS). Superoxide (O2-) and hydrogen peroxide (H2O2) are the main ROS involved in the regulation of cellular metabolism. As our fundamental understanding of the underlying causes of lung disease has increased it has become evident that oxidative stress plays a critical role. Recent Advances: A number of cells in the lung both produce, and respond to, ROS. These include vascular endothelial and smooth muscle cells, fibroblasts, and epithelial cells as well as the cells involved in the inflammatory response, including macrophages, neutrophils, eosinophils. The redox system is involved in multiple aspects of cell metabolism and cell homeostasis. Critical Issues: Dysregulation of the cellular redox system has consequential effects on cell signaling pathways that are intimately involved in disease progression. The lung is exposed to biomechanical forces (fluid shear stress, cyclic stretch, and pressure) due to the passage of blood through the pulmonary vessels and the distension of the lungs during the breathing cycle. Cells within the lung respond to these forces by activating signal transduction pathways that alter their redox state with both physiologic and pathologic consequences. Future Directions: Here, we will discuss the intimate relationship between biomechanical forces and redox signaling and its role in the development of pulmonary disease. An understanding of the molecular mechanisms induced by biomechanical forces in the pulmonary vasculature is necessary for the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Evgeny A Zemskov
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Qing Lu
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Wojciech Ornatowski
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Christina N Klinger
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Ankit A Desai
- Department of Medicine, Indiana University, Indianapolis, Indiana
| | - Emin Maltepe
- Department of Pediatrics, University of California, San Francisco, San Francisco, California
| | - Jason X-J Yuan
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Ting Wang
- Department of Internal Medicine, The University of Arizona Health Sciences, Phoenix, Arizona
| | - Jeffrey R Fineman
- Department of Pediatrics, University of California, San Francisco, San Francisco, California
| | - Stephen M Black
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| |
Collapse
|
25
|
Valle ML, Dworshak J, Sharma A, Ibrahim AS, Al-Shabrawey M, Sharma S. Inhibition of interleukin-6 trans-signaling prevents inflammation and endothelial barrier disruption in retinal endothelial cells. Exp Eye Res 2018; 178:27-36. [PMID: 30240585 DOI: 10.1016/j.exer.2018.09.009] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 07/20/2018] [Accepted: 09/17/2018] [Indexed: 12/27/2022]
Abstract
Vascular inflammation plays a critical role in the pathogenesis of diabetic retinopathy. Recently, Interleukin-6 (IL-6) trans-signaling via soluble IL-6 receptor (sIL-6R) has emerged as a prominent regulator of inflammation in endothelial cells. This study was designed to test the hypothesis that selective inhibition of the IL-6 trans-signaling pathway will attenuate inflammation and subsequent barrier disruption in retinal endothelial cells. Human retinal endothelial cells (HRECs) were exposed to IL-6 and sIL-6R to induce IL-6 trans-signaling and the commercially available compound sgp130Fc (soluble gp-130 fused chimera) was used to selectively inhibit IL-6 trans-signaling. IL-6 trans-signaling activation caused a significant increase in STAT3 phosphorylation, expression of adhesion molecules, ROS production and apoptosis in HRECs whereas a significant decrease in mitochondrial membrane potential and NO production was observed in IL-6 trans-signaling activated cells. These changes were not observed in cells pre-treated with sgp130Fc. IL-6 trans-signaling activation was sufficient to cause barrier disruption in endothelial monolayers and pre-treatment of HRECs with sgp130Fc, maintained endothelial barrier function similar to that of untreated cells. Thus, in conclusion, these results indicate that IL-6 trans-signaling is an important mediator of inflammation, apoptosis and barrier disruptive effects in the retinal endothelial cells and inhibition of the IL-6 trans-signaling pathway using sgp130-Fc attenuates vascular inflammation and endothelial barrier disruption.
Collapse
Affiliation(s)
- Maria L Valle
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Janine Dworshak
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Ashok Sharma
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA; Department of Population Health Sciences, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Ahmed S Ibrahim
- James & Jean Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA; Department of Oral Biology and Anatomy, Dental College of Georgia, Augusta University, Augusta, GA, USA; Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Mohamed Al-Shabrawey
- James & Jean Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA; Department of Oral Biology and Anatomy, Dental College of Georgia, Augusta University, Augusta, GA, USA
| | - Shruti Sharma
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA; Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA; James & Jean Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
26
|
Lee JH, Mand MR, Kao CH, Zhou Y, Ryu SW, Richards AL, Coon JJ, Paull TT. ATM directs DNA damage responses and proteostasis via genetically separable pathways. Sci Signal 2018; 11:eaan5598. [PMID: 29317520 PMCID: PMC5898228 DOI: 10.1126/scisignal.aan5598] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The protein kinase ATM is a master regulator of the DNA damage response but also responds directly to oxidative stress. Loss of ATM causes ataxia telangiectasia, a neurodegenerative disorder with pleiotropic symptoms that include cerebellar dysfunction, cancer, diabetes, and premature aging. We genetically separated the activation of ATM by DNA damage from that by oxidative stress using separation-of-function mutations. We found that deficient activation of ATM by the Mre11-Rad50-Nbs1 complex and DNA double-strand breaks resulted in loss of cell viability, checkpoint activation, and DNA end resection in response to DNA damage. In contrast, loss of oxidative activation of ATM had minimal effects on DNA damage-related outcomes but blocked ATM-mediated initiation of checkpoint responses after oxidative stress and resulted in deficiencies in mitochondrial function and autophagy. In addition, expression of a variant ATM incapable of activation by oxidative stress resulted in widespread protein aggregation. These results indicate a direct relationship between the mechanism of ATM activation and its effects on cellular metabolism and DNA damage responses in human cells and implicate ATM in the control of protein homeostasis.
Collapse
Affiliation(s)
- Ji-Hoon Lee
- Howard Hughes Medical Institute, Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Michael R Mand
- Howard Hughes Medical Institute, Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Chung-Hsuan Kao
- Howard Hughes Medical Institute, Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Yi Zhou
- Howard Hughes Medical Institute, Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Seung W Ryu
- Howard Hughes Medical Institute, Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Alicia L Richards
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Joshua J Coon
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Tanya T Paull
- Howard Hughes Medical Institute, Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
27
|
Steurer MA, Oltman S, Baer RJ, Feuer S, Liang L, Paynter RA, Rand L, Ryckman KK, Keller RL, Pawlowski LLJ. Altered metabolites in newborns with persistent pulmonary hypertension. Pediatr Res 2018; 84:272-278. [PMID: 29895840 PMCID: PMC7691760 DOI: 10.1038/s41390-018-0023-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 03/06/2018] [Accepted: 04/03/2018] [Indexed: 11/09/2022]
Abstract
BACKGROUND There is an emerging evidence that pulmonary hypertension is associated with amino acid, carnitine, and thyroid hormone aberrations. We aimed to characterize metabolic profiles measured by the newborn screen (NBS) in infants with persistent pulmonary hypertension of the newborn (PPHN) METHODS: Nested case-control study from population-based database. Cases were infants with ICD-9 code for PPHN receiving mechanical ventilation. Controls receiving mechanical ventilation were matched 2:1 for gestational age, sex, birth weight, parenteral nutrition administration, and age at NBS collection. Infants were divided into derivation and validation datasets. A multivariable logistic regression model was derived from candidate metabolites, and the area under the receiver operator characteristic curve (AUROC) was generated from the validation dataset. RESULTS We identified 1076 cases and 2152 controls. Four metabolites remained in the final model. Ornithine (OR 0.32, CI 0.26-0.41), tyrosine (OR 0.48, CI 0.40-0.58), and TSH 0.50 (0.45-0.55) were associated with decreased odds of PPHN; phenylalanine was associated with increased odds of PPHN (OR 4.74, CI 3.25-6.90). The AUROC was 0.772 (CI 0.737-0.807). CONCLUSIONS In a large, population-based dataset, infants with PPHN have distinct, early metabolic profiles. These data provide insight into the pathophysiology of PPHN, identifying potential therapeutic targets and novel biomarkers to assess the response.
Collapse
Affiliation(s)
- Martina A. Steurer
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA,Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA,California Preterm Birth Initiative, University of California San Francisco, San Francisco, CA, USA
| | - Scott Oltman
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Rebecca J. Baer
- California Preterm Birth Initiative, University of California San Francisco, San Francisco, CA, USA,Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Sky Feuer
- California Preterm Birth Initiative, University of California San Francisco, San Francisco, CA, USA
| | - Liang Liang
- Department of Genetics, Stanford University, Palo Alto, CA, USA
| | - Randi A. Paynter
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA,California Preterm Birth Initiative, University of California San Francisco, San Francisco, CA, USA
| | - Larry Rand
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA and
| | - Kelli K. Ryckman
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA, USA
| | - Roberta L. Keller
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Laura L. Jelliffe Pawlowski
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA,California Preterm Birth Initiative, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
28
|
l -carnitine supplementation during vitrification or warming of in vivo -produced ovine embryos does not affect embryonic survival rates, but alters CrAT and PRDX1 expression. Theriogenology 2018; 105:150-157. [DOI: 10.1016/j.theriogenology.2017.09.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 09/12/2017] [Accepted: 09/17/2017] [Indexed: 01/01/2023]
|
29
|
Altered Carnitine Homeostasis in Children With Increased Pulmonary Blood Flow Due to Ventricular Septal Defects. Pediatr Crit Care Med 2017; 18:931-934. [PMID: 28723882 PMCID: PMC5628126 DOI: 10.1097/pcc.0000000000001275] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVES Congenital heart disease with increased pulmonary blood flow results in progressive pulmonary vascular endothelial dysfunction and associated increased perioperative morbidity. Using our ovine model of congenital heart disease with increased pulmonary blood flow, we have previously demonstrated progressive endothelial dysfunction associated with disruption in carnitine homeostasis, mitochondrial dysfunction, decreased nitric oxide signaling, and enhanced reactive oxygen species generation. However, potential alterations in these parameters in patients with congenital heart disease have not been investigated. The objective of this study was to test the hypothesis that children with increased pulmonary blood flow will have evidence of altered carnitine homeostasis, mitochondrial dysfunction, decreased nitric oxide levels, and increased reactive oxygen species generation. DESIGN A prospective single-center cohort study. SETTING A tertiary care cardiac ICU/PICU. PATIENTS Arterial blood samples from 18 patients with congenital heart disease associated with increased pulmonary blood flow (ventricular septal defect), 20 with congenital heart disease without increased pulmonary blood flow (tetralogy of Fallot), and 10 without heart disease (controls) were obtained. INTERVENTIONS Plasma levels of total carnitine, free carnitine, acylcarnitine, and lactate-to-pyruvate ratios, an indicator of mitochondrial function, were determined and compared. In addition, levels of superoxide and hydrogen peroxide were determined and compared in patients with ventricular septal defect and controls. Statistical analysis was performed using an unpaired t test and analysis of variance. MEASUREMENTS AND MAIN RESULTS Baseline acylcarnitine levels (25.7 ± 13 vs 12.7 ± 8.3; p < 0.05), the acylcarnitine-to-free carnitine ratio (0.8 ± 0.1 vs 0.3 ± 0.05; p < 0.05), and the lactate-to-pyruvate ratio were higher in ventricular septal defect (27.5 ± 3.8 vs 11.1 ± 4.1, p < 0.05) than tetralogy of Fallot; there were no differences between tetralogy of Fallot and control. Superoxide and H2O2 levels were also higher in ventricular septal defect compared with controls, and NOx levels were lower in ventricular septal defect patients compared with tetralogy of Fallot and controls (p < 0.05). CONCLUSIONS These data suggest that increased pulmonary blood flow from ventricular septal defect results in altered carnitine and mitochondrial homeostasis, decreased nitric oxide signaling, and increased reactive oxygen species production. These data are consistent with our animal data demonstrating that altered carnitine homeostasis results in mitochondrial dysfunction, increased reactive oxygen species production, and decreased bioavailable nitric oxide. Since disruption of carnitine metabolism may contribute to endothelial dysfunction, carnitine supplementation may attenuate endothelial dysfunction associated with increased pulmonary blood flow and warrants further investigation.
Collapse
|
30
|
Clark EL, Bush SJ, McCulloch MEB, Farquhar IL, Young R, Lefevre L, Pridans C, Tsang HG, Wu C, Afrasiabi C, Watson M, Whitelaw CB, Freeman TC, Summers KM, Archibald AL, Hume DA. A high resolution atlas of gene expression in the domestic sheep (Ovis aries). PLoS Genet 2017; 13:e1006997. [PMID: 28915238 PMCID: PMC5626511 DOI: 10.1371/journal.pgen.1006997] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 10/03/2017] [Accepted: 08/24/2017] [Indexed: 02/08/2023] Open
Abstract
Sheep are a key source of meat, milk and fibre for the global livestock sector, and an important biomedical model. Global analysis of gene expression across multiple tissues has aided genome annotation and supported functional annotation of mammalian genes. We present a large-scale RNA-Seq dataset representing all the major organ systems from adult sheep and from several juvenile, neonatal and prenatal developmental time points. The Ovis aries reference genome (Oar v3.1) includes 27,504 genes (20,921 protein coding), of which 25,350 (19,921 protein coding) had detectable expression in at least one tissue in the sheep gene expression atlas dataset. Network-based cluster analysis of this dataset grouped genes according to their expression pattern. The principle of 'guilt by association' was used to infer the function of uncharacterised genes from their co-expression with genes of known function. We describe the overall transcriptional signatures present in the sheep gene expression atlas and assign those signatures, where possible, to specific cell populations or pathways. The findings are related to innate immunity by focusing on clusters with an immune signature, and to the advantages of cross-breeding by examining the patterns of genes exhibiting the greatest expression differences between purebred and crossbred animals. This high-resolution gene expression atlas for sheep is, to our knowledge, the largest transcriptomic dataset from any livestock species to date. It provides a resource to improve the annotation of the current reference genome for sheep, presenting a model transcriptome for ruminants and insight into gene, cell and tissue function at multiple developmental stages.
Collapse
Affiliation(s)
- Emily L. Clark
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Stephen J. Bush
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Mary E. B. McCulloch
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Iseabail L. Farquhar
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Rachel Young
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Lucas Lefevre
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Clare Pridans
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Hiu G. Tsang
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Chunlei Wu
- Department of Integrative and Computational Biology, The Scripps Research Institute, La Jolla, CA, United States of America
| | - Cyrus Afrasiabi
- Department of Integrative and Computational Biology, The Scripps Research Institute, La Jolla, CA, United States of America
| | - Mick Watson
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - C. Bruce Whitelaw
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Tom C. Freeman
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Kim M. Summers
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Mater Research Institute and University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Alan L. Archibald
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - David A. Hume
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Mater Research Institute and University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| |
Collapse
|
31
|
Sun X, Kellner M, Desai AA, Wang T, Lu Q, Kangath A, Qu N, Klinger C, Fratz S, Yuan JXJ, Jacobson JR, Garcia JGN, Rafikov R, Fineman JR, Black SM. Asymmetric Dimethylarginine Stimulates Akt1 Phosphorylation via Heat Shock Protein 70-Facilitated Carboxyl-Terminal Modulator Protein Degradation in Pulmonary Arterial Endothelial Cells. Am J Respir Cell Mol Biol 2017; 55:275-87. [PMID: 26959555 DOI: 10.1165/rcmb.2015-0185oc] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Asymmetric dimethylarginine (ADMA) induces the mitochondrial translocation of endothelial nitric oxide synthase (eNOS) through the nitration-mediated activation of Akt1. However, it is recognized that the activation of Akt1 requires phosphorylation events at threonine (T) 308 and serine (S) 473. Thus, the current study was performed to elucidate the potential effect of ADMA on Akt1 phosphorylation and the mechanisms that are involved. Exposure of pulmonary arterial endothelial cells to ADMA enhanced Akt1 phosphorylation at both threonine 308 and Ser473 without altering Akt1 protein levels, phosphatase and tensin homolog activity, or membrane Akt1 levels. Heat shock protein (Hsp) 90 plays a pivotal role in maintaining Akt1 activity, and our results demonstrate that ADMA decreased Hsp90-Akt1 interactions, but, surprisingly, overexpression of a dominant-negative Hsp90 mutant increased Akt1 phosphorylation. ADMA exposure or overexpression of dominant-negative Hsp90 increased Hsp70 levels, and depletion of Hsp70 abolished ADMA-induced Akt1 phosphorylation. ADMA decreased the interaction of Akt1 with its endogenous inhibitor, carboxyl-terminal modulator protein (CTMP). This was mediated by the proteasomal-dependent degradation of CTMP. The overexpression of CTMP attenuated ADMA-induced Akt1 phosphorylation at Ser473, eNOS phosphorylation at Ser617, and eNOS mitochondrial translocation. Finally, we found that the mitochondrial translocation of eNOS in our lamb model of pulmonary hypertension is associated with increased Akt1 and eNOS phosphorylation and reduced Akt1-CTMP protein interactions. In conclusion, our data suggest that CTMP is directly involved in ADMA-induced Akt1 phosphorylation in vitro and in vivo, and that increasing CTMP levels may be an avenue to treat pulmonary hypertension.
Collapse
Affiliation(s)
- Xutong Sun
- 1 Department of Medicine, Division of Translational and Regenerative Medicine, The University of Arizona, Tucson, Arizona
| | - Manuela Kellner
- 1 Department of Medicine, Division of Translational and Regenerative Medicine, The University of Arizona, Tucson, Arizona
| | - Ankit A Desai
- 1 Department of Medicine, Division of Translational and Regenerative Medicine, The University of Arizona, Tucson, Arizona
| | - Ting Wang
- 1 Department of Medicine, Division of Translational and Regenerative Medicine, The University of Arizona, Tucson, Arizona
| | - Qing Lu
- 2 Department of Neuroscience and Regenerative Medicine, Georgia Regents University, Augusta, Georgia
| | - Archana Kangath
- 1 Department of Medicine, Division of Translational and Regenerative Medicine, The University of Arizona, Tucson, Arizona
| | - Ning Qu
- 1 Department of Medicine, Division of Translational and Regenerative Medicine, The University of Arizona, Tucson, Arizona
| | - Christina Klinger
- 1 Department of Medicine, Division of Translational and Regenerative Medicine, The University of Arizona, Tucson, Arizona
| | - Sohrab Fratz
- 3 Pediatric Cardiology and Congenital Heart Disease, German Heart Center at the Technical University of Munich, Munich, Germany
| | - Jason X-J Yuan
- 1 Department of Medicine, Division of Translational and Regenerative Medicine, The University of Arizona, Tucson, Arizona
| | - Jeffrey R Jacobson
- 4 Department of Medicine, University of Illinois Chicago, Chicago, Illinois; and
| | - Joe G N Garcia
- 1 Department of Medicine, Division of Translational and Regenerative Medicine, The University of Arizona, Tucson, Arizona
| | - Ruslan Rafikov
- 1 Department of Medicine, Division of Translational and Regenerative Medicine, The University of Arizona, Tucson, Arizona
| | - Jeffrey R Fineman
- 5 Department of Pediatrics and.,6 Cardiovascular Research Institute, University of California San Francisco, San Francisco, California
| | - Stephen M Black
- 1 Department of Medicine, Division of Translational and Regenerative Medicine, The University of Arizona, Tucson, Arizona
| |
Collapse
|
32
|
Mishra C, Kumar S, Panigrahi M, Yathish HM, Chaudhary R, Chauhan A, Kumar A, Sonawane AA. Single Nucleotide Polymorphisms in 5' Upstream Region of Bovine TLR4 Gene Affecting Expression Profile and Transcription Factor Binding Sites. Anim Biotechnol 2017; 29:119-128. [PMID: 28594279 DOI: 10.1080/10495398.2017.1326929] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The present study in the 5' upstream region of TLR4 gene revealed four Single Nucleotide Polymorphisms (SNPs) in Vrindavani and Tharparkar cattle. The polymorphic information content (PIC), heterozygosity and allelic diversity values were low to moderate for these SNPs. In Vrindavani cattle, one SNP was found to be in Hardy-Weinberg Equilibrium (HWE) and the remaining three were found to be in linkage disequilibrium (LD) as indicated statistically (P > 0.05). In Tharparkar cattle, two SNPs were found to be in HWE and were not in LD as indicated statistically (P > 0.05). These SNPs were used for construction of haplotypes. In-silico analysis of these SNPs predicted abolition of eight transcription factor binding sites and creation of eight new sites. The quantitative real time PCR analysis did not show any significant variation of gene expression among haplotypes. However, gene expression between breed was found to be significant (P < 0.05) which suggested that upstream region of bovine TLR4 gene has a crucial role in its expression. These findings in TLR4 gene offer essential evidence that can be useful in future research exploring its role in immunity. TLR4 can be used as a marker for selection for disease resistance in bovines.
Collapse
Affiliation(s)
- Chinmoy Mishra
- a Department of Animal Genetics , Indian Veterinary Research Institute , Uttar Pradesh , India
| | - Subodh Kumar
- a Department of Animal Genetics , Indian Veterinary Research Institute , Uttar Pradesh , India
| | - Manjit Panigrahi
- a Department of Animal Genetics , Indian Veterinary Research Institute , Uttar Pradesh , India
| | - H M Yathish
- a Department of Animal Genetics , Indian Veterinary Research Institute , Uttar Pradesh , India
| | - Rajni Chaudhary
- a Department of Animal Genetics , Indian Veterinary Research Institute , Uttar Pradesh , India
| | - Anuj Chauhan
- a Department of Animal Genetics , Indian Veterinary Research Institute , Uttar Pradesh , India
| | - Amit Kumar
- a Department of Animal Genetics , Indian Veterinary Research Institute , Uttar Pradesh , India
| | - Arvind A Sonawane
- a Department of Animal Genetics , Indian Veterinary Research Institute , Uttar Pradesh , India
| |
Collapse
|
33
|
Evans CE, Zhao YY. Molecular Basis of Nitrative Stress in the Pathogenesis of Pulmonary Hypertension. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 967:33-45. [PMID: 29047079 DOI: 10.1007/978-3-319-63245-2_3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Pulmonary hypertension (PH) is a lung vascular disease with marked increases in pulmonary vascular resistance and pulmonary artery pressure (>25 mmHg at rest). In PH patients, increases in pulmonary vascular resistance lead to impaired cardiac output and reduced exercise tolerance. If untreated, PH progresses to right heart failure and premature lethality. The mechanisms that control the pathogenesis of PH are incompletely understood, but evidence from human and animal studies implicate nitrative stress in the development of PH. Increased levels of reactive oxygen species (ROS) and reactive nitrogen species (RNS) result in nitrative stress, which in turn induces posttranslational modification of key proteins important for maintaining pulmonary vascular homeostasis. This affects their functions and thereby contributes to the pathogenesis of PH. In this chapter, molecular mechanisms underlying nitrative stress-induced PH are reviewed, molecular sources of ROS and RNS are delineated, and evidence of nitrative stress in PH patients is described. A better understanding of such mechanisms could lead to the development of novel treatments for PH.
Collapse
Affiliation(s)
- Colin E Evans
- Department of Pharmacology, University of Illinois College of Medicine, 835 South Wolcott Avenue, E403-MSB, M/C 868, Chicago, IL, 60612, USA.,Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL, USA.,British Heart Foundation Center of Research Excellence, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - You-Yang Zhao
- Department of Pharmacology, University of Illinois College of Medicine, 835 South Wolcott Avenue, E403-MSB, M/C 868, Chicago, IL, 60612, USA. .,Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL, USA.
| |
Collapse
|
34
|
Yu Q, Chan SY. Mitochondrial and Metabolic Drivers of Pulmonary Vascular Endothelial Dysfunction in Pulmonary Hypertension. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 967:373-383. [PMID: 29047100 DOI: 10.1007/978-3-319-63245-2_24] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Pulmonary hypertension (PH) is a deadly and increasingly prevalent vascular disease characterized by excessive pulmonary vascular remodeling and right ventricular dysfunction which leads to right heart failure, multiorgan dysfunction, and premature death. The cause of the vascular remodeling in PH remains elusive, and thus current treatments are marginally effective and prognosis of PH remains poor. Increasing evidence indicates the pathogenic importance of endothelial dysfunction in PH. However, the underlying mechanisms of such dysfunction are not well described. Endothelial apoptosis and hyperproliferation have been identified in patients with PH. Both are linked with the increased oxidative stress and inflammatory responses, and are influenced by various genetic and exogenous stresses. Importantly, contrary to historic dogma that suggested a negligible role for mitochondria and energy balance in endothelial pathology, recent findings have implicated the role of endothelial metabolism directly in PH. This chapter addresses the emerging role of mitochondria in pulmonary vascular endothelial dysfunction in PH. A more sophisticated understanding of the biochemical, metabolic, molecular, and physiologic underpinnings of this emerging paradigm should enable the development of a new generation of targeted therapies that will stunt or reverse pulmonary vascular remodeling.
Collapse
Affiliation(s)
- Qiujun Yu
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh Medical Center, 200 Lothrop Street BST1704.2, Pittsburgh, PA, 15261, USA.,Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, 200 Lothrop Street BST1704.2, Pittsburgh, PA, 15261, USA
| | - Stephen Y Chan
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh Medical Center, 200 Lothrop Street BST1704.2, Pittsburgh, PA, 15261, USA. .,Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, 200 Lothrop Street BST1704.2, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
35
|
Rafikova O, Meadows ML, Kinchen JM, Mohney RP, Maltepe E, Desai AA, Yuan JXJ, Garcia JGN, Fineman JR, Rafikov R, Black SM. Metabolic Changes Precede the Development of Pulmonary Hypertension in the Monocrotaline Exposed Rat Lung. PLoS One 2016; 11:e0150480. [PMID: 26937637 PMCID: PMC4777490 DOI: 10.1371/journal.pone.0150480] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 02/14/2016] [Indexed: 12/13/2022] Open
Abstract
There is increasing interest in the potential for metabolic profiling to evaluate the progression of pulmonary hypertension (PH). However, a detailed analysis of the metabolic changes in lungs at the early stage of PH, characterized by increased pulmonary artery pressure but prior to the development of right ventricle hypertrophy and failure, is lacking in a preclinical animal model of PH. Thus, we undertook a study using rats 14 days after exposure to monocrotaline (MCT), to determine whether we could identify early stage metabolic changes prior to the manifestation of developed PH. We observed changes in multiple pathways associated with the development of PH, including activated glycolysis, increased markers of proliferation, disruptions in carnitine homeostasis, increased inflammatory and fibrosis biomarkers, and a reduction in glutathione biosynthesis. Further, our global metabolic profile data compare favorably with prior work carried out in humans with PH. We conclude that despite the MCT-model not recapitulating all the structural changes associated with humans with advanced PH, including endothelial cell proliferation and the formation of plexiform lesions, it is very similar at a metabolic level. Thus, we suggest that despite its limitations it can still serve as a useful preclinical model for the study of PH.
Collapse
Affiliation(s)
- Olga Rafikova
- Division of Translational and Regenerative Medicine, The University of Arizona, Tucson, Arizona, United States of America
- Department of Medicine, The University of Arizona, Tucson, Arizona, United States of America
| | - Mary L. Meadows
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia, United States of America
| | | | | | - Emin Maltepe
- Division of Neonatology, University of California San Francisco, San Francisco, California, United States of America
| | - Ankit A. Desai
- Department of Medicine, The University of Arizona, Tucson, Arizona, United States of America
| | - Jason X.-J. Yuan
- Division of Translational and Regenerative Medicine, The University of Arizona, Tucson, Arizona, United States of America
- Department of Medicine, The University of Arizona, Tucson, Arizona, United States of America
| | - Joe G. N. Garcia
- Department of Medicine, The University of Arizona, Tucson, Arizona, United States of America
| | - Jeffrey R. Fineman
- Department of Pediatrics, University of California San Francisco, San Francisco, California, United States of America
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, United States of America
| | - Ruslan Rafikov
- Division of Translational and Regenerative Medicine, The University of Arizona, Tucson, Arizona, United States of America
- Department of Medicine, The University of Arizona, Tucson, Arizona, United States of America
- * E-mail:
| | - Stephen M. Black
- Division of Translational and Regenerative Medicine, The University of Arizona, Tucson, Arizona, United States of America
- Department of Medicine, The University of Arizona, Tucson, Arizona, United States of America
| |
Collapse
|
36
|
Reconciling paradigms of abnormal pulmonary blood flow and quasi-malignant cellular alterations in pulmonary arterial hypertension. Vascul Pharmacol 2016; 83:17-25. [PMID: 26804008 DOI: 10.1016/j.vph.2016.01.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 01/19/2016] [Indexed: 01/29/2023]
Abstract
In pulmonary arterial hypertension (PAH) structural and functional abnormalities of the small lung vessels interact and lead to a progressive increase in pulmonary vascular resistance and right heart failure. A current pathobiological concept characterizes PAH as a 'quasi-malignant' disease focusing on cancer-like alterations in endothelial cells (EC) and the importance of their acquired apoptosis-resistant, hyper-proliferative phenotype in the process of vascular remodeling. While changes in pulmonary blood flow (PBF) have been long-since recognized and linked to the development of PAH, little is known about a possible relationship between an altered PBF and the quasi-malignant cell phenotype in the pulmonary vascular wall. This review summarizes recognized and hypothetical effects of an abnormal PBF on the pulmonary vascular bed and links these to quasi-malignant changes found in the pulmonary endothelium. Here we describe that abnormal PBF does not only trigger a pulmonary vascular cell growth program, but may also maintain the cancer-like phenotype of the endothelium. Consequently, normalization of PBF and EC response to abnormal PBF may represent a treatment strategy in patients with established PAH.
Collapse
|
37
|
Zhou L, Zhang J, Jiang XM, Xie DJ, Wang JS, Li L, Li B, Wang ZM, Rothman AM, Lawrie A, Chen SL. Pulmonary Artery Denervation Attenuates Pulmonary Arterial Remodeling in Dogs With Pulmonary Arterial Hypertension Induced by Dehydrogenized Monocrotaline. JACC Cardiovasc Interv 2015; 8:2013-2023. [DOI: 10.1016/j.jcin.2015.09.015] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 09/21/2015] [Accepted: 09/21/2015] [Indexed: 12/24/2022]
|
38
|
Rafikova O, Rafikov R, Meadows ML, Kangath A, Jonigk D, Black SM. The sexual dimorphism associated with pulmonary hypertension corresponds to a fibrotic phenotype. Pulm Circ 2015; 5:184-97. [PMID: 25992281 DOI: 10.1086/679724] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 08/18/2014] [Indexed: 01/23/2023] Open
Abstract
Although female predominance in the development of all types of pulmonary hypertension (PH) is well established, many clinical studies have confirmed that females have better prognosis and higher survival rate than males. There is no clear explanation of why sex influences the pathogenesis and progression of PH. Using a rat angioproliferative model of PH, which closely resembles the primary pathological changes observed in humans, we evaluated the role of sex in the development and progression of PH. Female rats had a more pronounced increase in medial thickness in the small pulmonary arteries. However, the infiltration of small pulmonary arteries by inflammatory cells was found only in male rats, and this corresponded to increased myeloperoxidase activity and abundant adventitial and medial fibrosis that were not present in female rats. Although the level of right ventricle (RV) peak systolic pressure was similar in both groups, the survival rate in male rats was significantly lower. Moreover, male rats presented with a more pronounced increase in RV thickness that correlated with diffuse RV fibrosis and significantly impaired right cardiac function. The reduction in fibrosis in female rats correlated with increased expression of caveolin-1 and reduced endothelial nitric oxide synthase-derived superoxide. We conclude that, in the pathogenesis of PH, female sex is associated with greater remodeling of the pulmonary arteries but greater survival. Conversely, in males, the development of pulmonary and cardiac fibrosis leads to early and severe RV failure, and this may be an important reason for the lower survival rate among males.
Collapse
Affiliation(s)
- Olga Rafikova
- Pulmonary Vascular Disease Program, Vascular Biology Center, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, USA ; These authors contributed equally to this study
| | - Ruslan Rafikov
- Pulmonary Vascular Disease Program, Vascular Biology Center, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, USA ; These authors contributed equally to this study
| | - Mary Louise Meadows
- Pulmonary Vascular Disease Program, Vascular Biology Center, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, USA
| | - Archana Kangath
- Pulmonary Vascular Disease Program, Vascular Biology Center, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, USA
| | - Danny Jonigk
- Institute of Pathology, Hannover Medical School, Hanover, Germany
| | - Stephen M Black
- Pulmonary Vascular Disease Program, Vascular Biology Center, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, USA
| |
Collapse
|
39
|
Sun X, Kumar S, Sharma S, Aggarwal S, Lu Q, Gross C, Rafikova O, Lee SG, Dasarathy S, Hou Y, Meadows ML, Han W, Su Y, Fineman JR, Black SM. Endothelin-1 induces a glycolytic switch in pulmonary arterial endothelial cells via the mitochondrial translocation of endothelial nitric oxide synthase. Am J Respir Cell Mol Biol 2014; 50:1084-95. [PMID: 24392990 DOI: 10.1165/rcmb.2013-0187oc] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Recent studies have indicated that, during the development of pulmonary hypertension (PH), there is a switch from oxidative phosphorylation to glycolysis in the pulmonary endothelium. However, the mechanisms underlying this phenomenon have not been elucidated. Endothelin (ET)-1, an endothelial-derived vasoconstrictor peptide, is increased in PH, and has been shown to play an important role in the oxidative stress associated with PH. Thus, in this study, we investigated whether there was a potential link between increases in ET-1 and mitochondrial remodeling. Our data indicate that ET-1 induces the redistribution of endothelial nitric oxide synthase (eNOS) from the plasma membrane to the mitochondria in pulmonary arterial endothelial cells, and that this was dependent on eNOS uncoupling. We also found that ET-1 disturbed carnitine metabolism, resulting in the attenuation of mitochondrial bioenergetics. However, ATP levels were unchanged due to a compensatory increase in glycolysis. Further mechanistic investigations demonstrated that ET-1 mediated the redistribution of eNOS via the phosphorylation of eNOS at Thr495 by protein kinase C δ. In addition, the glycolytic switch appeared to be dependent on mitochondrial-derived reactive oxygen species that led to the activation of hypoxia-inducible factor signaling. Finally, the cell culture data were confirmed in vivo using the monocrotaline rat model of PH. Thus, we conclude that ET-1 induces a glycolytic switch in pulmonary arterial endothelial cells via the redistribution of uncoupled eNOS to the mitochondria, and that preventing this event may be an approach for the treatment of PH.
Collapse
Affiliation(s)
- Xutong Sun
- 1 Pulmonary Disease Program, Vascular Biology Center, and
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Sharp J, Farha S, Park MM, Comhair SA, Lundgrin EL, Tang WHW, Bongard RD, Merker MP, Erzurum SC. Coenzyme Q supplementation in pulmonary arterial hypertension. Redox Biol 2014; 2:884-91. [PMID: 25180165 PMCID: PMC4143816 DOI: 10.1016/j.redox.2014.06.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 06/15/2014] [Accepted: 06/17/2014] [Indexed: 12/18/2022] Open
Abstract
Mitochondrial dysfunction is a fundamental abnormality in the vascular endothelium and smooth muscle of patients with pulmonary arterial hypertension (PAH). Because coenzyme Q (CoQ) is essential for mitochondrial function and efficient oxygen utilization as the electron carrier in the inner mitochondrial membrane, we hypothesized that CoQ would improve mitochondrial function and benefit PAH patients. To test this, oxidized and reduced levels of CoQ, cardiac function by echocardiogram, mitochondrial functions of heme synthesis and cellular metabolism were evaluated in PAH patients (N=8) in comparison to healthy controls (N=7), at baseline and after 12 weeks oral CoQ supplementation. CoQ levels were similar among PAH and control individuals, and increased in all subjects with CoQ supplementation. PAH patients had higher CoQ levels than controls with supplementation, and a tendency to a higher reduced-to-oxidized CoQ ratio. Cardiac parameters improved with CoQ supplementation, although 6-minute walk distances and BNP levels did not significantly change. Consistent with improved mitochondrial synthetic function, hemoglobin increased and red cell distribution width (RDW) decreased in PAH patients with CoQ, while hemoglobin declined slightly and RDW did not change in healthy controls. In contrast, metabolic and redox parameters, including lactate, pyruvate and reduced or oxidized gluthathione, did not change in PAH patients with CoQ. In summary, CoQ improved hemoglobin and red cell maturation in PAH, but longer studies and/or higher doses with a randomized placebo-controlled controlled design are necessary to evaluate the clinical benefit of this simple nutritional supplement.
Collapse
Affiliation(s)
- Jacqueline Sharp
- Respiratory Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States ; Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States ; Heart and Vascular Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Samar Farha
- Respiratory Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States ; Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Margaret M Park
- Respiratory Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States ; Heart and Vascular Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Suzy A Comhair
- Respiratory Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States ; Cleveland Clinic Lerner College of Medicine, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Erika L Lundgrin
- Cleveland Clinic Lerner College of Medicine, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - W H Wilson Tang
- Heart and Vascular Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States ; Cleveland Clinic Lerner College of Medicine, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Robert D Bongard
- Department of Medicine, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States
| | - Marilyn P Merker
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States ; Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, 53226, United States ; Zablocki VAMC, 5000 W National Ave., Milwaukee, WI 53295, United States
| | - Serpil C Erzurum
- Respiratory Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States ; Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States ; Cleveland Clinic Lerner College of Medicine, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| |
Collapse
|
41
|
Yu ZR, Ning Y, Yu H, Tang NJ. A HPLC-Q-TOF-MS-based urinary metabolomic approach to identification of potential biomarkers of metabolic syndrome. ACTA ACUST UNITED AC 2014; 34:276-283. [DOI: 10.1007/s11596-014-1271-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Revised: 03/01/2014] [Indexed: 01/03/2023]
|
42
|
Rafikov R, Kumar S, Aggarwal S, Hou Y, Kangath A, Pardo D, Fineman JR, Black SM. Endothelin-1 stimulates catalase activity through the PKCδ-mediated phosphorylation of serine 167. Free Radic Biol Med 2014; 67:255-64. [PMID: 24211614 PMCID: PMC3945115 DOI: 10.1016/j.freeradbiomed.2013.10.814] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 10/15/2013] [Accepted: 10/17/2013] [Indexed: 01/03/2023]
Abstract
Our previous studies have shown that endothelin-1 (ET-1) stimulates catalase activity in endothelial cells and in lambs with acute increases in pulmonary blood flow (PBF), without altering gene expression. The purpose of this study was to investigate the molecular mechanism by which this occurs. Exposing pulmonary arterial endothelial cells to ET-1 increased catalase activity and decreased cellular hydrogen peroxide (H2O2) levels. These changes correlated with an increase in serine-phosphorylated catalase. Using the inhibitory peptide δV1.1, this phosphorylation was shown to be protein kinase Cδ (PKCδ) dependent. Mass spectrometry identified serine 167 as the phosphorylation site. Site-directed mutagenesis was used to generate a phospho-mimic (S167D) catalase. Activity assays using recombinant protein purified from Escherichia coli or transiently transfected COS-7 cells demonstrated that S167D catalase had an increased ability to degrade H2O2 compared to the wild-type enzyme. Using a phospho-specific antibody, we were able to verify that pS167 catalase levels are modulated in lambs with acute increases in PBF in the presence and absence of the ET receptor antagonist tezosentan. S167 is located on the dimeric interface, suggesting it could be involved in regulating the formation of catalase tetramers. To evaluate this possibility we utilized analytical gel filtration to examine the multimeric structure of recombinant wild-type and S167D catalase. We found that recombinant wild-type catalase was present as a mixture of monomers and dimers, whereas S167D catalase was primarily tetrameric. Further, the incubation of wild-type catalase with PKCδ was sufficient to convert wild-type catalase into a tetrameric structure. In conclusion, this is the first report indicating that the phosphorylation of catalase regulates its multimeric structure and activity.
Collapse
Affiliation(s)
- Ruslan Rafikov
- Pulmonary Disease Program, Vascular Biology Center, Georgia Regents University, Augusta GA 30912
- Please address correspondence and proofs to: Stephen M. Black, Ph.D., Vascular Biology Center, Georgia Regents University, 1459 Laney Walker Blvd, CB 3211-B, Augusta, GA-30912, Tel: 706-721-7860,
| | - Sanjiv Kumar
- Pulmonary Disease Program, Vascular Biology Center, Georgia Regents University, Augusta GA 30912
- Please address correspondence and proofs to: Stephen M. Black, Ph.D., Vascular Biology Center, Georgia Regents University, 1459 Laney Walker Blvd, CB 3211-B, Augusta, GA-30912, Tel: 706-721-7860,
| | - Saurabh Aggarwal
- Pulmonary Disease Program, Vascular Biology Center, Georgia Regents University, Augusta GA 30912
| | - Yali Hou
- Pulmonary Disease Program, Vascular Biology Center, Georgia Regents University, Augusta GA 30912
| | - Archana Kangath
- Pulmonary Disease Program, Vascular Biology Center, Georgia Regents University, Augusta GA 30912
| | - Daniel Pardo
- Pulmonary Disease Program, Vascular Biology Center, Georgia Regents University, Augusta GA 30912
| | - Jeffrey R. Fineman
- Department of Pediatrics University of California, San Francisco, CA, 94143
- Cardiovascular Research Institute, University of California, San Francisco, CA, 94143
| | - Stephen M. Black
- Pulmonary Disease Program, Vascular Biology Center, Georgia Regents University, Augusta GA 30912
| |
Collapse
|
43
|
Aggarwal S, Gross CM, Rafikov R, Kumar S, Fineman JR, Ludewig B, Jonigk D, Black SM. Nitration of tyrosine 247 inhibits protein kinase G-1α activity by attenuating cyclic guanosine monophosphate binding. J Biol Chem 2014; 289:7948-61. [PMID: 24469460 DOI: 10.1074/jbc.m113.534313] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The cGMP-dependent protein kinase G-1α (PKG-1α) is a downstream mediator of nitric oxide and natriuretic peptide signaling. Alterations in this pathway play a key role in the pathogenesis and progression of vascular diseases associated with increased vascular tone and thickness, such as pulmonary hypertension. Previous studies have shown that tyrosine nitration attenuates PKG-1α activity. However, little is known about the mechanisms involved in this event. Utilizing mass spectrometry, we found that PKG-1α is susceptible to nitration at tyrosine 247 and 425. Tyrosine to phenylalanine mutants, Y247F- and Y425F-PKG-1α, were both less susceptible to nitration than WT PKG-1α, but only Y247F-PKG-1α exhibited preserved activity, suggesting that the nitration of Tyr(247) is critical in attenuating PKG-1α activity. The overexpression of WT- or Y247F-PKG-1α decreased the proliferation of pulmonary artery smooth muscle cells (SMC), increased the expression of SMC contractile markers, and decreased the expression of proliferative markers. Nitrosative stress induced a switch from a contractile to a synthetic phenotype in cells expressing WT- but not Y247F-PKG-1α. An antibody generated against 3-NT-Y247 identified increased levels of nitrated PKG-1α in humans with pulmonary hypertension. Finally, to gain a more mechanistic understanding of how nitration attenuates PKG activity, we developed a homology model of PKG-1α. This model predicted that the nitration of Tyr(247) would decrease the affinity of PKG-1α for cGMP, which we confirmed using a [(3)H]cGMP binding assay. Our study shows that the nitration of Tyr(247) and the attenuation of cGMP binding is an important mechanism regulating in PKG-1α activity and SMC proliferation/differentiation.
Collapse
Affiliation(s)
- Saurabh Aggarwal
- From the Pulmonary Disease Program, Vascular Biology Center, Georgia Regents University, Augusta, Georgia 30912
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Rafikov R, Dimitropoulou C, Aggarwal S, Kangath A, Gross C, Pardo D, Sharma S, Jezierska-Drutel A, Patel V, Snead C, Lucas R, Verin A, Fulton D, Catravas JD, Black SM. Lipopolysaccharide-induced lung injury involves the nitration-mediated activation of RhoA. J Biol Chem 2014; 289:4710-22. [PMID: 24398689 DOI: 10.1074/jbc.m114.547596] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Acute lung injury (ALI) is characterized by increased endothelial hyperpermeability. Protein nitration is involved in the endothelial barrier dysfunction in LPS-exposed mice. However, the nitrated proteins involved in this process have not been identified. The activation of the small GTPase RhoA is a critical event in the barrier disruption associated with LPS. Thus, in this study we evaluated the possible role of RhoA nitration in this process. Mass spectroscopy identified a single nitration site, located at Tyr(34) in RhoA. Tyr(34) is located within the switch I region adjacent to the nucleotide-binding site. Utilizing this structure, we developed a peptide designated NipR1 (nitration inhibitory peptide for RhoA 1) to shield Tyr(34) against nitration. TAT-fused NipR1 attenuated RhoA nitration and barrier disruption in LPS-challenged human lung microvascular endothelial cells. Further, treatment of mice with NipR1 attenuated vessel leakage and inflammatory cell infiltration and preserved lung function in a mouse model of ALI. Molecular dynamics simulations suggested that the mechanism by which Tyr(34) nitration stimulates RhoA activity was through a decrease in GDP binding to the protein caused by a conformational change within a region of Switch I, mimicking the conformational shift observed when RhoA is bound to a guanine nucleotide exchange factor. Stopped flow kinetic analysis was used to confirm this prediction. Thus, we have identified a new mechanism of nitration-mediated RhoA activation involved in LPS-mediated endothelial barrier dysfunction and show the potential utility of "shielding" peptides to prevent RhoA nitration in the management of ALI.
Collapse
Affiliation(s)
- Ruslan Rafikov
- From the Program in Pulmonary Vascular Disease, Vascular Biology Center and
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
El-Ashmawy NE, Khalil RM. A review on the role of L-carnitine in the management of tamoxifen side effects in treated women with breast cancer. Tumour Biol 2013; 35:2845-55. [PMID: 24338689 DOI: 10.1007/s13277-013-1477-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 11/26/2013] [Indexed: 12/20/2022] Open
Abstract
L-carnitine is an antioxidant and is found to be a protective agent against many diseases including cancer. This review illustrates the possible role of L-carnitine as an add-on therapy to breast cancer patients maintained on tamoxifen. The objectives of carnitine treatment are diverse: improving tamoxifen-related side effects, offering better cancer prognosis by reducing the risk of developing cancer recurrence or metastasis, and modulating the growth factors which may be, in part, a prospective illustration to overcome tamoxifen resistance. So, it could be recommended to supplement L-carnitine to breast cancer patients starting tamoxifen treatment.
Collapse
Affiliation(s)
- Nahla E El-Ashmawy
- Biochemistry Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | | |
Collapse
|
46
|
Aggarwal S, Gross CM, Sharma S, Fineman JR, Black SM. Reactive oxygen species in pulmonary vascular remodeling. Compr Physiol 2013; 3:1011-34. [PMID: 23897679 DOI: 10.1002/cphy.c120024] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The pathogenesis of pulmonary hypertension is a complex multifactorial process that involves the remodeling of pulmonary arteries. This remodeling process encompasses concentric medial thickening of small arterioles, neomuscularization of previously nonmuscular capillary-like vessels, and structural wall changes in larger pulmonary arteries. The pulmonary arterial muscularization is characterized by vascular smooth muscle cell hyperplasia and hypertrophy. In addition, in uncontrolled pulmonary hypertension, the clonal expansion of apoptosis-resistant endothelial cells leads to the formation of plexiform lesions. Based upon a large number of studies in animal models, the three major stimuli that drive the vascular remodeling process are inflammation, shear stress, and hypoxia. Although, the precise mechanisms by which these stimuli impair pulmonary vascular function and structure are unknown, reactive oxygen species (ROS)-mediated oxidative damage appears to play an important role. ROS are highly reactive due to their unpaired valence shell electron. Oxidative damage occurs when the production of ROS exceeds the quenching capacity of the antioxidant mechanisms of the cell. ROS can be produced from complexes in the cell membrane (nicotinamide adenine dinucleotide phosphate-oxidase), cellular organelles (peroxisomes and mitochondria), and in the cytoplasm (xanthine oxidase). Furthermore, low levels of tetrahydrobiopterin (BH4) and L-arginine the rate limiting cofactor and substrate for endothelial nitric oxide synthase (eNOS), can cause the uncoupling of eNOS, resulting in decreased NO production and increased ROS production. This review will focus on the ROS generation systems, scavenger antioxidants, and oxidative stress associated alterations in vascular remodeling in pulmonary hypertension.
Collapse
Affiliation(s)
- Saurabh Aggarwal
- Pulmonary Disease Program, Vascular Biology Center, Georgia Health Sciences University, Augusta, Georgia, USA
| | | | | | | | | |
Collapse
|
47
|
Xu W, Erzurum SC. Endothelial cell energy metabolism, proliferation, and apoptosis in pulmonary hypertension. Compr Physiol 2013; 1:357-72. [PMID: 23737177 DOI: 10.1002/cphy.c090005] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a fatal disease characterized by impaired regulation of pulmonary hemodynamics and excessive growth and dysfunction of the endothelial cells that line the arteries in PAH lungs. Establishment of methods for culture of pulmonary artery endothelial cells from PAH lungs has provided the groundwork for mechanistic translational studies that confirm and extend findings from model systems and spontaneous pulmonary hypertension in animals. Endothelial cell hyperproliferation, survival, and alterations of biochemical-metabolic pathways are the unifying endothelial pathobiology of the disease. The hyperproliferative and apoptosis-resistant phenotype of PAH endothelial cells is dependent upon the activation of signal transducer and activator of transcription (STAT) 3, a fundamental regulator of cell survival and angiogenesis. Animal models of PAH, patients with PAH, and human PAH endothelial cells produce low nitric oxide (NO). In association with the low level of NO, endothelial cells have reduced mitochondrial numbers and cellular respiration, which is associated with more than a threefold increase in glycolysis for energy production. The shift to glycolysis is related to low levels of NO and likely to the pathologic expression of the prosurvival and proangiogenic signal transducer, hypoxia-inducible factor (HIF)-1, and the reduced mitochondrial antioxidant manganese superoxide dismutase (MnSOD). In this article, we review the phenotypic changes of the endothelium in PAH and the biochemical mechanisms accounting for the proliferative, glycolytic, and strongly proangiogenic phenotype of these dysfunctional cells, which consequently foster the panvascular progressive pulmonary remodeling in PAH.
Collapse
Affiliation(s)
- Weiling Xu
- Departments of Pathobiology, Lerner Research Institute, Respiratory Institute, Cleveland Clinic, Cleveland, OH, USA.
| | | |
Collapse
|
48
|
Sharma S, Aramburo A, Rafikov R, Sun X, Kumar S, Oishi PE, Datar SA, Raff G, Xoinis K, Kalkan G, Fratz S, Fineman JR, Black SM. L-carnitine preserves endothelial function in a lamb model of increased pulmonary blood flow. Pediatr Res 2013; 74:39-47. [PMID: 23628882 PMCID: PMC3709010 DOI: 10.1038/pr.2013.71] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 02/01/2013] [Indexed: 12/30/2022]
Abstract
BACKGROUND In our model of a congenital heart defect (CHD) with increased pulmonary blood flow (PBF; shunt), we have recently shown a disruption in carnitine homeostasis, associated with mitochondrial dysfunction and decreased endothelial nitric oxide synthase (eNOS)/heat shock protein (Hsp)90 interactions that contribute to eNOS uncoupling, increased superoxide levels, and decreased bioavailable nitric oxide (NO). Therefore, we undertook this study to test the hypothesis that L-carnitine therapy would maintain mitochondrial function and NO signaling. METHODS Thirteen fetal lambs underwent in utero placement of an aortopulmonary graft. Immediately after delivery, lambs received daily treatment with oral L-carnitine or its vehicle. RESULTS L-Carnitine-treated lambs had decreased levels of acylcarnitine and a reduced acylcarnitine:free carnitine ratio as compared with vehicle-treated shunt lambs. These changes correlated with increased carnitine acetyl transferase (CrAT) protein and enzyme activity and decreased levels of nitrated CrAT. The lactate:pyruvate ratio was also decreased in L-carnitine-treated lambs. Hsp70 protein levels were significantly decreased, and this correlated with increases in eNOS/Hsp90 interactions, NOS activity, and NOx levels, and a significant decrease in eNOS-derived superoxide. Furthermore, acetylcholine significantly decreased left pulmonary vascular resistance only in L-carnitine-treated lambs. CONCLUSION L-Carnitine therapy may improve the endothelial dysfunction noted in children with CHDs and has important clinical implications that warrant further investigation.
Collapse
Affiliation(s)
- Shruti Sharma
- Pulmonary Vascular Disease Program, Vascular Biology Center, Georgia Health Sciences University, Augusta GA 30912
| | - Angela Aramburo
- Department of Pediatrics, University of California, San Francisco CA,Department of Pediatrics, University Autonomous Barcelona, Spain
| | - Ruslan Rafikov
- Pulmonary Vascular Disease Program, Vascular Biology Center, Georgia Health Sciences University, Augusta GA 30912
| | - Xutong Sun
- Pulmonary Vascular Disease Program, Vascular Biology Center, Georgia Health Sciences University, Augusta GA 30912
| | - Sanjiv Kumar
- Pulmonary Vascular Disease Program, Vascular Biology Center, Georgia Health Sciences University, Augusta GA 30912
| | - Peter E. Oishi
- Department of Pediatrics, University of California, San Francisco CA,Cardiovascular Research Institute, University of California, San Francisco CA
| | - Sanjeev A. Datar
- Department of Pediatrics, University of California, San Francisco CA
| | - Gary Raff
- Department of Cardiothoracic Surgery, University of California, Davis CA
| | - Kon Xoinis
- Department of Pediatrics, University of California, San Francisco CA
| | - Gohkan Kalkan
- Department of Pediatrics, University of California, San Francisco CA
| | - Sohrab Fratz
- Department of Pediatric Cardiology and Congenital Heart Disease, Deutsches Herzzentrum München, Klinik an der Technischen Universität München, Lazarettstrasse 36, 80636 Munich, Germany
| | - Jeffrey R. Fineman
- Department of Pediatrics, University of California, San Francisco CA,Cardiovascular Research Institute, University of California, San Francisco CA
| | - Stephen M. Black
- Pulmonary Vascular Disease Program, Vascular Biology Center, Georgia Health Sciences University, Augusta GA 30912
| |
Collapse
|
49
|
Sun X, Sharma S, Fratz S, Kumar S, Rafikov R, Aggarwal S, Rafikova O, Lu Q, Burns T, Dasarathy S, Wright J, Schreiber C, Radman M, Fineman JR, Black SM. Disruption of endothelial cell mitochondrial bioenergetics in lambs with increased pulmonary blood flow. Antioxid Redox Signal 2013; 18:1739-52. [PMID: 23244702 PMCID: PMC3619212 DOI: 10.1089/ars.2012.4806] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
AIMS The mitochondrial dysfunction in our lamb model of congenital heart disease with increased pulmonary blood flow (PBF) (Shunt) is associated with disrupted carnitine metabolism. Our recent studies have also shown that asymmetric dimethylarginine (ADMA) levels are increased in Shunt lambs and ADMA increases the nitration of mitochondrial proteins in lamb pulmonary arterial endothelial cells (PAEC) in a nitric oxide synthase (NOS)-dependent manner. Thus, we determined whether there was a mechanistic link between endothelial nitric oxide synthase (eNOS), ADMA, and the disruption of carnitine homeostasis in PAEC. RESULTS Exposure of PAEC to ADMA induced the redistribution of eNOS to the mitochondria, resulting in an increase in carnitine acetyl transferase (CrAT) nitration and decreased CrAT activity. The resulting increase in acyl-carnitine levels resulted in mitochondrial dysfunction and the disruption of mitochondrial bioenergetics. Since the addition of L-arginine prevented these pathologic changes, we examined the effect of L-arginine supplementation on carnitine homeostasis, mitochondrial function, and nitric oxide (NO) signaling in Shunt lambs. We found that the treatment of Shunt lambs with L-arginine prevented the ADMA-mediated mitochondrial redistribution of eNOS, the nitration-mediated inhibition of CrAT, and maintained carnitine homeostasis. In turn, adenosine-5'-triphosphate levels and eNOS/heat shock protein 90 interactions were preserved, and this decreased NOS uncoupling and enhanced NO generation. INNOVATION Our data link alterations in cellular L-arginine metabolism with the disruption of mitochondrial bioenergetics and implicate altered carnitine homeostasis as a key player in this process. CONCLUSION L-arginine supplementation may be a useful therapy to prevent the mitochondrial dysfunction involved in the pulmonary vascular alterations secondary to increased PBF.
Collapse
Affiliation(s)
- Xutong Sun
- Pulmonary Disease Program, Vascular Biology Center, Georgia Health Sciences University, Augusta, GA 30912, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Role of carnitine acetyl transferase in regulation of nitric oxide signaling in pulmonary arterial endothelial cells. Int J Mol Sci 2012; 14:255-72. [PMID: 23344032 PMCID: PMC3565262 DOI: 10.3390/ijms14010255] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Revised: 11/26/2012] [Accepted: 11/30/2012] [Indexed: 12/16/2022] Open
Abstract
Congenital heart defects with increased pulmonary blood flow (PBF) result in pulmonary endothelial dysfunction that is dependent, at least in part, on decreases in nitric oxide (NO) signaling. Utilizing a lamb model with left-to-right shunting of blood and increased PBF that mimics the human disease, we have recently shown that a disruption in carnitine homeostasis, due to a decreased carnitine acetyl transferase (CrAT) activity, correlates with decreased bioavailable NO. Thus, we undertook this study to test the hypothesis that the CrAT enzyme plays a major role in regulating NO signaling through its effect on mitochondrial function. We utilized the siRNA gene knockdown approach to mimic the effect of decreased CrAT activity in pulmonary arterial endothelial cells (PAEC). Our data indicate that silencing the CrAT gene disrupted cellular carnitine homeostasis, reduced the expression of mitochondrial superoxide dismutase-and resulted in an increase in oxidative stress within the mitochondrion. CrAT gene silencing also disrupted mitochondrial bioenergetics resulting in reduced ATP generation and decreased NO signaling secondary to a reduction in eNOS/Hsp90 interactions. Thus, this study links the disruption of carnitine homeostasis to the loss of NO signaling observed in children with CHD. Preserving carnitine homeostasis may have important clinical implications that warrant further investigation.
Collapse
|