1
|
Sun L, Gao M, Yang GY, Lu FT, Liang ZJ, Guo KM, Lv XF, Du YH, Liang SJ, Tang YB, Zhou JG, Guan YY, Ma MM. ClC-5 knockout mitigates angiotensin II-induced hypertension and endothelial dysfunction. Life Sci 2025; 362:123342. [PMID: 39740756 DOI: 10.1016/j.lfs.2024.123342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 12/22/2024] [Accepted: 12/26/2024] [Indexed: 01/02/2025]
Abstract
AIMS Impairment of nitric oxide (NO) production is a major cause of endothelial dysfunction and hypertension. ClC-5 Cl- channel is abundantly expressed in the vascular endothelium. However, it remains unclear how it regulates endothelial function. MATERIALS AND METHODS In this study, we used mice with a knockout of the Clcn5 gene encoding ClC-5 protein globally or specifically in vascular endothelium. KEY FINDINGS ClC-5 knockout globally or specifically in vascular endothelium mitigates the elevation of mean blood pressure and impairment of endothelial dysfunction induced by Angiotensin II. This effect is mediated by the reversal of the impairment of NO production after the stimulation of the Akt/endothelial nitric oxide synthase (eNOS) signal pathway. Application of a low Cl- extracellular solution onto endothelial cells stimulates a ClC-5-dependent current and lowered intracellular Cl- concentration, which activates with-no-lysine (K)-1 (WNK1), a Cl--sensitive kinase. Silencing ClC-5 or WNK1 expression rescues the impairment of endothelial NO production induced by a low Cl- solution. In contrast, overexpression of ClC-5 or WNK1 led to the opposite results. WNK1, found to be associated with Rho-specific guanine nucleotide dissociation inhibitor (RhoGDI), increases RhoA activity, and thereby inhibits the endothelial Akt/eNOS signaling pathway. SIGNIFICANCE ClC-5 knockout mitigates Ang II-induced hypertension and endothelial dysfunction by promoting NO production via regulating WNK1/RhoA/Akt/eNOS signaling pathway. The results may be useful for developing novel treatments of endothelial dysfunction associated-diseases.
Collapse
Affiliation(s)
- Lu Sun
- Department of Pharmacology, Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China; Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Min Gao
- Department of Pharmacy, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, China
| | - Gui-Yong Yang
- Department of Pharmacology, Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Feng-Ting Lu
- Department of Pharmacology, Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China; Department of Molecular Medicine, School of Medicine, Sun Yat-Sen University, Shenzhen 518107, China
| | - Zhu-Jun Liang
- Department of Pharmacology, Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Kai-Min Guo
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Xiao-Fei Lv
- Department of Pharmacology, Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yan-Hua Du
- Department of Pharmacology, Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Si-Jia Liang
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yu-Bo Tang
- Department of Pharmacy, The First Affiliated Hospital of Sun Yat-sen University, 510080 Guangzhou, China
| | - Jia-Guo Zhou
- Department of Pharmacology, Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yong-Yuan Guan
- Department of Pharmacology, Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Ming-Ming Ma
- Department of Pharmacology, Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China.
| |
Collapse
|
2
|
Hassan D, Menges CW, Testa JR, Bellacosa A. AKT kinases as therapeutic targets. J Exp Clin Cancer Res 2024; 43:313. [PMID: 39614261 PMCID: PMC11606119 DOI: 10.1186/s13046-024-03207-4] [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: 07/24/2024] [Accepted: 10/03/2024] [Indexed: 12/01/2024] Open
Abstract
AKT, or protein kinase B, is a central node of the PI3K signaling pathway that is pivotal for a range of normal cellular physiologies that also underlie several pathological conditions, including inflammatory and autoimmune diseases, overgrowth syndromes, and neoplastic transformation. These pathologies, notably cancer, arise if either the activity of AKT or its positive or negative upstream or downstream regulators or effectors goes unchecked, superimposed on by its intersection with a slew of other pathways. Targeting the PI3K/AKT pathway is, therefore, a prudent countermeasure. AKT inhibitors have been tested in many clinical trials, primarily in combination with other drugs. While some have recently garnered attention for their favorable profile, concern over resistance and off-target effects have continued to hinder their widespread adoption in the clinic, mandating a discussion on alternative modes of targeting. In this review, we discuss isoform-centric targeting that may be more effective and less toxic than traditional pan-AKT inhibitors and its significance for disease prevention and treatment, including immunotherapy. We also touch on the emerging mutant- or allele-selective covalent allosteric AKT inhibitors (CAAIs), as well as indirect, novel AKT-targeting approaches, and end with a briefing on the ongoing quest for more reliable biomarkers predicting sensitivity and response to AKT inhibitors, and their current state of affairs.
Collapse
Affiliation(s)
- Dalal Hassan
- Nuclear Dynamics and Cancer Program, Cancer Epigenetics Institute, Institute for Cancer Research, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111, USA
- Thomas Jefferson University, 901 Walnut St, Philadelphia, PA, 19107, USA
| | - Craig W Menges
- Cancer Prevention and Control Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111, USA
| | - Joseph R Testa
- Cancer Prevention and Control Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111, USA
| | - Alfonso Bellacosa
- Nuclear Dynamics and Cancer Program, Cancer Epigenetics Institute, Institute for Cancer Research, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111, USA.
| |
Collapse
|
3
|
Zhou W, Ghersi JJ, Ristori E, Semanchik N, Prendergast A, Zhang R, Carneiro P, Baldissera G, Sessa WC, Nicoli S. Akt is a mediator of artery specification during zebrafish development. Development 2024; 151:dev202727. [PMID: 39101673 PMCID: PMC11441982 DOI: 10.1242/dev.202727] [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: 01/23/2024] [Accepted: 07/16/2024] [Indexed: 08/06/2024]
Abstract
The dorsal aorta (DA) is the first major blood vessel to develop in the embryonic cardiovascular system. Its formation is governed by a coordinated process involving the migration, specification, and arrangement of angioblasts into arterial and venous lineages, a process conserved across species. Although vascular endothelial growth factor a (VEGF-A) is known to drive DA specification and formation, the kinases involved in this process remain ambiguous. Thus, we investigated the role of protein kinase B (Akt) in zebrafish by generating a quadruple mutant (aktΔ/Δ), in which expression and activity of all Akt genes - akt1, -2, -3a and -3b - are strongly decreased. Live imaging of developing aktΔ/Δ DA uncovers early arteriovenous malformations. Single-cell RNA-sequencing analysis of aktΔ/Δ endothelial cells corroborates the impairment of arterial, yet not venous, cell specification. Notably, endothelial specific expression of ligand-independent activation of Notch or constitutively active Akt1 were sufficient to re-establish normal arterial specification in aktΔ/Δ. The Akt loss-of-function mutant unveils that Akt kinase can act upstream of Notch in arterial endothelial cells, and is involved in proper embryonic artery specification. This sheds light on cardiovascular development, revealing a mechanism behind congenital malformations.
Collapse
Affiliation(s)
- Wenping Zhou
- Vascular Biology & Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06511, USA
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Joey J Ghersi
- Vascular Biology & Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA
- Yale Cardiovascular Research Center, Department of Internal Medicine, Section of Cardiology, Yale University School of Medicine, New Haven, CT 06511, USA
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
- Pathologies Foetomaternelles et Néonatales, Centre Hospitalier Universitaire Sainte-Justine Research Center, Montréal, QC H3T 1C5, Canada
- Department of Pathology and Cell Biology, Faculty of Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Emma Ristori
- Vascular Biology & Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA
- Yale Cardiovascular Research Center, Department of Internal Medicine, Section of Cardiology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Nicole Semanchik
- Vascular Biology & Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA
- Yale Cardiovascular Research Center, Department of Internal Medicine, Section of Cardiology, Yale University School of Medicine, New Haven, CT 06511, USA
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Andrew Prendergast
- Department of Comparative Medicine, Yale zebrafish Research Core, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Rong Zhang
- Vascular Biology & Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Paola Carneiro
- Vascular Biology & Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA
- Yale Cardiovascular Research Center, Department of Internal Medicine, Section of Cardiology, Yale University School of Medicine, New Haven, CT 06511, USA
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Gabriel Baldissera
- Yale Cardiovascular Research Center, Department of Internal Medicine, Section of Cardiology, Yale University School of Medicine, New Haven, CT 06511, USA
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
| | - William C Sessa
- Vascular Biology & Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Stefania Nicoli
- Vascular Biology & Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Cardiovascular Research Center, Department of Internal Medicine, Section of Cardiology, Yale University School of Medicine, New Haven, CT 06511, USA
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
4
|
Clark GC, Lai A, Agarwal A, Liu Z, Wang XY. Biopterin metabolism and nitric oxide recoupling in cancer. Front Oncol 2024; 13:1321326. [PMID: 38469569 PMCID: PMC10925643 DOI: 10.3389/fonc.2023.1321326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 12/26/2023] [Indexed: 03/13/2024] Open
Abstract
Tetrahydrobiopterin is a cofactor necessary for the activity of several enzymes, the most studied of which is nitric oxide synthase. The role of this cofactor-enzyme relationship in vascular biology is well established. Recently, tetrahydrobiopterin metabolism has received increasing attention in the field of cancer immunology and immunotherapy due to its involvement in the cytotoxic T cell response. Past research has demonstrated that when the availability of BH4 is low, as it is in chronic inflammatory conditions and tumors, electron transfer in the active site of nitric oxide synthase becomes uncoupled from the oxidation of arginine. This results in the production of radical species that are capable of a direct attack on tetrahydrobiopterin, further depleting its local availability. This feedforward loop may act like a molecular switch, reinforcing low tetrahydrobiopterin levels leading to altered NO signaling, restrained immune effector activity, and perpetual vascular inflammation within the tumor microenvironment. In this review, we discuss the evidence for this underappreciated mechanism in different aspects of tumor progression and therapeutic responses. Furthermore, we discuss the preclinical evidence supporting a clinical role for tetrahydrobiopterin supplementation to enhance immunotherapy and radiotherapy for solid tumors and the potential safety concerns.
Collapse
Affiliation(s)
- Gene Chatman Clark
- Department of Biochemistry, Virginia Commonwealth University, Richmond, VA, United States
- School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Alan Lai
- School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | | | - Zheng Liu
- Department of Human Molecular Genetics, Virginia Commonwealth University, Richmond, VA, United States
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Xiang-Yang Wang
- Department of Human Molecular Genetics, Virginia Commonwealth University, Richmond, VA, United States
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
- Institute of Molecular Medicine, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
5
|
Yan BH, Xu QX, Ge X, Gao MT, Li Y, Guo L, Hu P, Pan Y. Molecular mechanisms of Chengshi Beixie Fenqing Decoction based on network pharmacology: pivotal roles of relaxin signaling pathway and its associated target proteins against Benign prostatic hyperplasia. J Biomol Struct Dyn 2024; 42:2075-2093. [PMID: 37102991 DOI: 10.1080/07391102.2023.2203237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 04/10/2023] [Indexed: 04/28/2023]
Abstract
Benign prostatic hyperplasia (BPH) is a common disease that affects the quality of life of middle-aged and older men. We investigated the therapeutical effects of Chengshi Beixie Fenqing Decoction (CBFD), a classic traditional Chinese medicine prescription, on BPH through in vivo model and network pharmacology. Bioactives in CBFD were detected through UPLC-Q-Tof-MS/MS and GC-MS, and filtered by the modified Lipinski's rule. Target proteins associated with the filtered compounds and BPH are selected from public databases. Venn diagram identified the overlapping target proteins between the bioactives-interacted target proteins and the BPH-targeted proteins. The bioactive-protein interactive networking of BPH was analyzed through the KEGG pathway on STRING to identify potential ligand-target and visualized the rich factors on the R packet. After that, the molecular docking test (MDT) was performed between bioactives and target proteins. It showed that the mechanism of CBFD against BPH was related to 104 signaling pathways of 42 compounds. AKT1, 6-demethyl-4'-methyl-N-methylcoclaurine and relaxin signaling pathways were selected as a hub target, key bioactivitie and hub signaling pathway, respectively. In addition, three major compounds, 6-demethyl-4'-methyl-N-methylcoclaurine, isoliensinine and liensinine, had the highest affinity on MDT for the three crucial target proteins, AKT1, JUN and MAPK1. These proteins were associated with the relaxin signaling pathway, which regulated the level of nitric oxide and is implicated in both BPH development and CBFD. We concluded that the three key bioactivities found in Plumula nelumbinis of CBFD may contribute to improving BPH condition by activating the relaxin signaling pathways.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Bing-Hui Yan
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Qi-Xuan Xu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Xiao Ge
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Ming-Tong Gao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yun Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Liang Guo
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Po Hu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yang Pan
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
6
|
Nguyen TD, Rahman NT, Sessa WC, Lee MY. Endothelial nitric oxide synthase (eNOS) S1176 phosphorylation status governs atherosclerotic lesion formation. Front Cardiovasc Med 2023; 10:1279868. [PMID: 38034389 PMCID: PMC10683645 DOI: 10.3389/fcvm.2023.1279868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 10/11/2023] [Indexed: 12/02/2023] Open
Abstract
Objective We have previously demonstrated the in vivo importance of the Akt-eNOS substrate-kinase relationship, as defective postnatal angiogenesis characteristic of global Akt1-null mice is rescued when bred to 'gain-of-function' eNOS S1176D mutant mice. While multiple studies support the vascular protective role of endothelial NO generation, the causal role of Akt1-dependent eNOS S1176 phosphorylation during atherosclerotic plaque formation is not yet clear. Approach and results We herein bred congenic 'loss-of-function' eNOS S1176A and 'gain-of-function' eNOS S1176D mutant mice to the exacerbated atherogenic Akt1-/-; ApoE-/- double knockout mice to definitively test the importance of Akt-mediated eNOS S1176 phosphorylation during atherogenesis. We find that a single amino acid substitution at the eNOS S1176 phosphorylation site yields divergent effects on atherosclerotic plaque formation, as an eNOS phospho-mimic aspartate (D) substitution at S1176 leads to favorable lipid profiles and decreased indices of atherosclerosis, even when on a proatherogenic Akt1 global deletion background. Conversely, mice harboring an unphosphorylatable mutation to alanine (S1176A) result in increased plasma lipids, increased lesion formation and cellular apoptosis, phenocopying the physiological consequence of eNOS deletion and/or impaired enzyme function. Furthermore, gene expression analyses of whole aortas indicate a combinatorial detriment from NO deficiency and Western Diet challenge, as 'loss-of-function' eNOS S1176A mice on a Western Diet present a unique expression pattern indicative of augmented T-cell activity when compared to eNOS S1176D mice. Conclusions By using genetic epistasis approaches, we conclusively demonstrate that Akt-mediated eNOS S1176 phosphorylation and subsequent eNOS activation remains to be the most physiologically relevant method of NO production to promote athero-protective effects.
Collapse
Affiliation(s)
- Tung D. Nguyen
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago School of Medicine, Chicago, IL, United States
| | - Nur-Taz Rahman
- Bioinformatics Support Group, Yale University Cushing/Whitney Medical Library, New Haven, CT, United States
| | - William C. Sessa
- Department of Pharmacology, Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, United States
| | - Monica Y. Lee
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago School of Medicine, Chicago, IL, United States
| |
Collapse
|
7
|
Karakuyu NF, Ertunc O, Bedir M, Dogan HK, Taner R, Sevuk MA, Imeci OB, Ergonul E. Protective role of nebivolol via AKT1/Hif-1α/eNOS signaling pathway: nephrotoxicity caused by methotrexate in a rat model. Can J Physiol Pharmacol 2023; 101:393-399. [PMID: 36947849 DOI: 10.1139/cjpp-2022-0433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
Methotrexate (MTX) is an antineoplastic and anti-inflammatory agent, which is used in severe diseases. Its use should be limited due to side effects such as nephrotoxicity, myelotoxicity, and hepatotoxicity. Nebivolol (NBV), which is a beta-blocker used in the treatment of hypertension, also contributes to vasodilation in tissues by activating the endothelial nitric oxide synthase (eNOS) enzyme. The purpose of this study is to research the effect of NBV on MTX-induced nephrotoxicity through the AKT1/hypoxia-inducible factor 1-alpha (Hif-1α)/eNOS signaling pathway. The rats were randomly divided into three groups of eight each. The groups were control, MTX, and MTX + NBV. A single dose of 20 mg/kg MTX was given intraperitoneally to the rats on the first day of the study and 10 mg/kg NBV was given orally to the treatment group for 7 days. At the end of the study, rats' blood and kidney tissues were taken for histopathological, immunohistochemical, and biochemical examinations. MTX administration significantly decreased the expression levels of AKT1, eNOS, and Hif-1α compared with the control group (p < 0.001 for all), and NBV treatment increased these values compared with the MTX group (p < 0.001 for all). In conclusion, NBV treatment ameliorated the MTX-induced nephrotoxicity via AKT1/Hif-1α/eNOS signaling pathway.
Collapse
Affiliation(s)
- N F Karakuyu
- Department of Pharmacology, Faculty of Pharmacy, Suleyman Demirel University, Isparta, Turkey
| | - O Ertunc
- Department of Pathology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - M Bedir
- Department of Medical Biochemistry, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - H K Dogan
- Department of Bioengineering, School of Engineering, Suleyman Demirel University, Isparta, Turkey
| | - R Taner
- Department of Bioengineering, School of Engineering, Suleyman Demirel University, Isparta, Turkey
| | - M A Sevuk
- Department of Pharmacology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - O B Imeci
- Department of Pharmacology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - E Ergonul
- Department of Medical Education, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| |
Collapse
|
8
|
Huang L, Cheng F, Zhang X, Zielonka J, Nystoriak MA, Xiang W, Raygor K, Wang S, Lakshmanan A, Jiang W, Yuan S, Hou KS, Zhang J, Wang X, Syed AU, Juric M, Takahashi T, Navedo MF, Wang RA. Nitric oxide synthase and reduced arterial tone contribute to arteriovenous malformation. SCIENCE ADVANCES 2023; 9:eade7280. [PMID: 37235659 PMCID: PMC10219588 DOI: 10.1126/sciadv.ade7280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 04/20/2023] [Indexed: 05/28/2023]
Abstract
Mechanisms underlying arteriovenous malformations (AVMs) are poorly understood. Using mice with endothelial cell (EC) expression of constitutively active Notch4 (Notch4*EC), we show decreased arteriolar tone in vivo during brain AVM initiation. Reduced vascular tone is a primary effect of Notch4*EC, as isolated pial arteries from asymptomatic mice exhibited reduced pressure-induced arterial tone ex vivo. The nitric oxide (NO) synthase (NOS) inhibitor NG-nitro-l-arginine (L-NNA) corrected vascular tone defects in both assays. L-NNA treatment or endothelial NOS (eNOS) gene deletion, either globally or specifically in ECs, attenuated AVM initiation, assessed by decreased AVM diameter and delayed time to moribund. Administering nitroxide antioxidant 4-hydroxy-2,2,6,6-tetramethylpiperidine-1-oxyl also attenuated AVM initiation. Increased NOS-dependent production of hydrogen peroxide, but not NO, superoxide, or peroxynitrite was detected in isolated Notch4*EC brain vessels during AVM initiation. Our data suggest that eNOS is involved in Notch4*EC-mediated AVM formation by up-regulating hydrogen peroxide and reducing vascular tone, thereby permitting AVM initiation and progression.
Collapse
Affiliation(s)
- Lawrence Huang
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Feng Cheng
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Xuetao Zhang
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Jacek Zielonka
- Free Radical Research Laboratory, Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Matthew A. Nystoriak
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, USA
| | - Weiwei Xiang
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Kunal Raygor
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Shaoxun Wang
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Aditya Lakshmanan
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Weiya Jiang
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Sai Yuan
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Kevin S. Hou
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Jiayi Zhang
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Xitao Wang
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Arsalan U. Syed
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, USA
| | - Matea Juric
- Free Radical Research Laboratory, Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Takamune Takahashi
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Manuel F. Navedo
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, USA
| | - Rong A. Wang
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
9
|
Apolinário PP, Zanchetta FC, Breder JSC, Adams G, Consonni SR, Gillis R, Saad MJA, Lima MHM. Anti-inflammatory, procollagen, and wound repair properties of topical insulin gel. Braz J Med Biol Res 2023; 56:e12640. [PMID: 37194835 DOI: 10.1590/1414-431x2023e12640] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 03/16/2023] [Indexed: 05/18/2023] Open
Abstract
Diabetes mellitus is associated with impaired wound healing. The topical use of insulin is a promising therapy because it may favor all phases of the wound healing process. This study aimed to investigate the therapeutic outcomes of insulin gel in wounds of hyperglycemic mice. After diabetes induction, a 1-cm2 full-thickness wound was created on each animal's dorsum. The lesions were treated daily for 14 days with insulin gel (insulin group) or vehicle gel without insulin (vehicle group). Tissue samples were extracted on days 4, 7, 10, and 14 after the creation of the lesion. The samples were analyzed with hematoxylin/eosin and Sirius red staining, immunohistochemistry, Bio-Plex immunoassays, and western blotting. Insulin gel favored re-epithelialization at day 10 and increased the organization and deposition of collagen. Additionally, it modulated the expression of cytokines (interleukin (IL)-4 and IL-10) and increased the expression of arginase I, VEGF receptor 1, and VEGF on day 10. Activation of the insulin signaling pathway occurred via IRβ, IRS1, and IKK on day 10 and activation of Akt and IRS1 on day 14. These results suggested that insulin gel improved wound healing in hyperglycemic mice by modulating the expression of inflammatory factors, growth factors, and proteins of the insulin signaling pathway.
Collapse
Affiliation(s)
- P P Apolinário
- Colégio Técnico de Campinas, Universidade Estadual de Campinas, Campinas, SP, Brasil
| | - F C Zanchetta
- Faculdade de Enfermagem, Universidade Estadual de Campinas, Campinas, SP, Brasil
| | - J S C Breder
- Faculdade de Enfermagem, Universidade Estadual de Campinas, Campinas, SP, Brasil
| | - G Adams
- Faculty of Medicine and Health Science, University of Nottingham, Nottingham, UK
| | - S R Consonni
- Insituto de Biologia, Universidade Estadual de Campinas, Campinas, SP, Brasil
| | - R Gillis
- Department of Service Sector Management, Sheffield Hallam University, Sheffield, UK
| | - M J A Saad
- Faculdade de Ciências Médicas, Universidade Estadual de Campinas, Campinas, SP, Brasil
| | - M H M Lima
- Faculdade de Enfermagem, Universidade Estadual de Campinas, Campinas, SP, Brasil
| |
Collapse
|
10
|
Salyers ZR, Coleman M, Le D, Ryan TE. AAV-mediated expression of PFKFB3 in myofibers, but not endothelial cells, improves ischemic muscle function in mice with critical limb ischemia. Am J Physiol Heart Circ Physiol 2022; 323:H424-H436. [PMID: 35867710 PMCID: PMC11834898 DOI: 10.1152/ajpheart.00121.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 07/15/2022] [Accepted: 07/19/2022] [Indexed: 01/18/2023]
Abstract
6-Phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3) is a powerful driver of angiogenesis through its modulation of glycolytic metabolism within endothelial cells. Recent work has demonstrated that PFKFB3 modulates the response to muscle ischemia, however the cell specificity of these effects is not fully understood. In this study, we tested the impact of viral mediated expression of PFKFB3, driven by gene promoters specific for myofibers or endothelial cells, on ischemic hindlimb revascularization and muscle function. We hypothesized that both endothelium- and muscle-specific expression of PFKFB3 would attenuate limb pathology following femoral artery ligation. Male and female BALB/cJ mice were injected with adeno-associated virus encoding the either a green fluorescent protein (GFP) or PFKFB3 driven by either the human skeletal actin (ACTA1) or cadherin-5 (Cdh5) promoters. Four weeks after AAV treatment, mice were subjected to unilateral femoral artery ligation and limb perfusion and muscle function were assessed. Both endothelium- and muscle-specific PFKFB3 expression resulted in significantly more perfused capillaries within the ischemic limb muscle, but neither changed myofiber size/area. Muscle-specific, but not endothelium-specific, PFKFB3 expression significantly improved maximal force production in ischemic muscle (P = 0.0005). Notably, there was a significant effect of sex on maximal force levels in both cohorts of mice (P = 0.0075 and P = 0.0481), indicating that female mice had higher ischemic muscle strength compared with male mice, regardless of treatment group. Taken together, these data demonstrate that although both muscle- and endothelium-specific expression of PFKFB3 enhanced ischemic revascularization, only muscle-specific PFKFB3 expression improved muscle function.NEW & NOTEWORTHY Critical limb ischemia (CLI) carries a significant risk for limb amputation, and treatment options remain limited. We tested the impact of expression of PFKFB3 in myofibers or endothelial cells on limb pathology in mice with CLI. Although both muscle and endothelium-specific PFKFB3 expression increased perfused capillary density, only muscle-specific PFKFB3 expression improve contractile function. Regardless of treatment, female mice demonstrated better recovery from limb ischemic compared with male mice.
Collapse
Affiliation(s)
- Zachary R. Salyers
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL
| | - Madeline Coleman
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL
| | - Dennis Le
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL
| | - Terence E. Ryan
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL
- Center for Exercise Science, University of Florida, Gainesville, FL
- Myology Institute, University of Florida, Gainesville, FL
| |
Collapse
|
11
|
Jin YJ, Chennupati R, Li R, Liang G, Wang S, Iring A, Graumann J, Wettschureck N, Offermanns S. Protein kinase N2 mediates flow-induced endothelial NOS activation and vascular tone regulation. J Clin Invest 2021; 131:e145734. [PMID: 34499618 DOI: 10.1172/jci145734] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 09/01/2021] [Indexed: 01/31/2023] Open
Abstract
Formation of NO by endothelial NOS (eNOS) is a central process in the homeostatic regulation of vascular functions including blood pressure regulation, and fluid shear stress exerted by the flowing blood is a main stimulus of eNOS activity. Previous work has identified several mechanosensing and -transducing processes in endothelial cells, which mediate this process and induce the stimulation of eNOS activity through phosphorylation of the enzyme via various kinases including AKT. How the initial mechanosensing and signaling processes are linked to eNOS phosphorylation is unclear. In human endothelial cells, we demonstrated that protein kinase N2 (PKN2), which is activated by flow through the mechanosensitive cation channel Piezo1 and Gq/G11-mediated signaling, as well as by Ca2+ and phosphoinositide-dependent protein kinase 1 (PDK1), plays a pivotal role in this process. Active PKN2 promoted the phosphorylation of human eNOS at serine 1177 and at a newly identified site, serine 1179. These phosphorylation events additively led to increased eNOS activity. PKN2-mediated eNOS phosphorylation at serine 1177 involved the phosphorylation of AKT synergistically with mTORC2-mediated AKT phosphorylation, whereas active PKN2 directly phosphorylated human eNOS at serine 1179. Mice with induced endothelium-specific deficiency of PKN2 showed strongly reduced flow-induced vasodilation and developed arterial hypertension accompanied by reduced eNOS activation. These results uncover a central mechanism that couples upstream mechanosignaling processes in endothelial cells to the regulation of eNOS-mediated NO formation, vascular tone, and blood pressure.
Collapse
Affiliation(s)
- Young-June Jin
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Ramesh Chennupati
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Rui Li
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Guozheng Liang
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - ShengPeng Wang
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Yanta District, Xi'an, China
| | - András Iring
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Laboratory of Molecular Medicine, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Johannes Graumann
- Scientific Service Group Biomolecular Mass Spectrometry, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Nina Wettschureck
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Centre for Molecular Medicine, Medical Faculty, JW Goethe University Frankfurt, Frankfurt, Germany.,Cardiopulmonary Institute (CPI), Frankfurt, Germany.,German Center for Cardiovascular Research (DZHK), Rhine-Main Site, Frankfurt and Bad Nauheim, Germany
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Centre for Molecular Medicine, Medical Faculty, JW Goethe University Frankfurt, Frankfurt, Germany.,Cardiopulmonary Institute (CPI), Frankfurt, Germany.,German Center for Cardiovascular Research (DZHK), Rhine-Main Site, Frankfurt and Bad Nauheim, Germany
| |
Collapse
|
12
|
Nguyen H, Koh JY, Li H, Islas-Robles A, Meda Venkata SP, Wang JM, Monks TJ. A novel imidazolinone metformin-methylglyoxal metabolite promotes endothelial cell angiogenesis via the eNOS/HIF-1α pathway. FASEB J 2021; 35:e21645. [PMID: 34105824 PMCID: PMC8237315 DOI: 10.1096/fj.202002674rr] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 04/13/2021] [Accepted: 04/20/2021] [Indexed: 12/28/2022]
Abstract
Peripheral arterial disease (PAD) is one of the major complications of diabetes due to an impairment in angiogenesis. Since there is currently no drug with satisfactory efficacy to enhance blood vessel formation, discovering therapies to improve angiogenesis is critical. An imidazolinone metabolite of the metformin‐methylglyoxal scavenging reaction, (E)‐1,1‐dimethyl‐2‐(5‐methyl‐4‐oxo‐4,5‐dihydro‐1H‐imidazol‐2‐yl) guanidine (IMZ), was recently characterized and identified in the urine of type‐2 diabetic patients. Here, we report the pro‐angiogenesis effect of IMZ (increased aortic sprouting, cell migration, network formation, and upregulated multiple pro‐angiogenic factors) in human umbilical vein endothelial cells. Using genetic and pharmacological approaches, we showed that IMZ augmented angiogenesis by activating the endothelial nitric oxide synthase (eNOS)/hypoxia‐inducible factor‐1 alpha (HIF‐1α) pathway. Furthermore, IMZ significantly promoted capillary density in the in vivo Matrigel plug angiogenesis model. Finally, the role of IMZ in post‐ischemic angiogenesis was examined in a chronic hyperglycemia mouse model subjected to hind limb ischemia. We observed improved blood perfusion, increased capillary density, and reduced tissue necrosis in mice receiving IMZ compared to control mice. Our data demonstrate the pro‐angiogenic effects of IMZ, its underlying mechanism, and provides a structural basis for the development of potential pro‐angiogenic agents for the treatment of PAD.
Collapse
Affiliation(s)
- Huong Nguyen
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
| | - Jia Yi Koh
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
| | - Hainan Li
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
| | | | - Sai Pranathi Meda Venkata
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
| | - Jie-Mei Wang
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA.,Centers for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
| | - Terrence J Monks
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
| |
Collapse
|
13
|
Lopez-Ramirez MA, Lai CC, Soliman SI, Hale P, Pham A, Estrada EJ, McCurdy S, Girard R, Verma R, Moore T, Lightle R, Hobson N, Shenkar R, Poulsen O, Haddad GG, Daneman R, Gongol B, Sun H, Lagarrigue F, Awad IA, Ginsberg MH. Astrocytes propel neurovascular dysfunction during cerebral cavernous malformation lesion formation. J Clin Invest 2021; 131:139570. [PMID: 34043589 PMCID: PMC8245174 DOI: 10.1172/jci139570] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 05/24/2021] [Indexed: 12/13/2022] Open
Abstract
Cerebral cavernous malformations (CCMs) are common neurovascular lesions caused by loss-of-function mutations in 1 of 3 genes, including KRIT1 (CCM1), CCM2, and PDCD10 (CCM3), and generally regarded as an endothelial cell-autonomous disease. Here we reported that proliferative astrocytes played a critical role in CCM pathogenesis by serving as a major source of VEGF during CCM lesion formation. An increase in astrocyte VEGF synthesis is driven by endothelial nitric oxide (NO) generated as a consequence of KLF2- and KLF4-dependent elevation of eNOS in CCM endothelium. The increased brain endothelial production of NO stabilized HIF-1α in astrocytes, resulting in increased VEGF production and expression of a "hypoxic" program under normoxic conditions. We showed that the upregulation of cyclooxygenase-2 (COX-2), a direct HIF-1α target gene and a known component of the hypoxic program, contributed to the development of CCM lesions because the administration of a COX-2 inhibitor significantly prevented the progression of CCM lesions. Thus, non-cell-autonomous crosstalk between CCM endothelium and astrocytes propels vascular lesion development, and components of the hypoxic program represent potential therapeutic targets for CCMs.
Collapse
MESH Headings
- Animals
- Apoptosis Regulatory Proteins/deficiency
- Apoptosis Regulatory Proteins/genetics
- Astrocytes/pathology
- Astrocytes/physiology
- Cyclooxygenase 2/genetics
- Cyclooxygenase 2/metabolism
- Disease Models, Animal
- Disease Progression
- Endothelial Cells/metabolism
- Hemangioma, Cavernous, Central Nervous System/etiology
- Hemangioma, Cavernous, Central Nervous System/pathology
- Hemangioma, Cavernous, Central Nervous System/physiopathology
- Human Umbilical Vein Endothelial Cells
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Kruppel-Like Factor 4
- Kruppel-Like Transcription Factors/genetics
- Kruppel-Like Transcription Factors/metabolism
- Mice
- Mice, Knockout
- Models, Neurological
- Mutation
- Nitric Oxide/biosynthesis
- Nitric Oxide Synthase Type III/genetics
- Nitric Oxide Synthase Type III/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Vascular Endothelial Growth Factor A/biosynthesis
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Romuald Girard
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | | | - Thomas Moore
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Rhonda Lightle
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Nicholas Hobson
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Robert Shenkar
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | | | - Gabriel G. Haddad
- Department of Pediatrics, and
- Department of Neuroscience, Division of Respiratory Medicine, University of California, San Diego, La Jolla, California, USA
- Rady Children’s Hospital, San Diego, California, USA
| | - Richard Daneman
- Department of Pharmacology, University of California, San Diego, La Jolla, California, USA
| | | | | | | | - Issam A. Awad
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | | |
Collapse
|
14
|
Abstract
The prevalence of peripheral arterial disease (PAD) in the United States exceeds 10 million people, and PAD is a significant cause of morbidity and mortality across the globe. PAD is typically caused by atherosclerotic obstructions in the large arteries to the leg(s). The most common clinical consequences of PAD include pain on walking (claudication), impaired functional capacity, pain at rest, and loss of tissue integrity in the distal limbs that may lead to lower extremity amputation. Patients with PAD also have higher than expected rates of myocardial infarction, stroke, and cardiovascular death. Despite advances in surgical and endovascular procedures, revascularization procedures may be suboptimal in relieving symptoms, and some patients with PAD cannot be treated because of comorbid conditions. In some cases, relieving obstructive disease in the large conduit arteries does not assure complete limb salvage because of severe microvascular disease. Despite several decades of investigational efforts, medical therapies to improve perfusion to the distal limb are of limited benefit. Whereas recent studies of anticoagulant (eg, rivaroxaban) and intensive lipid lowering (such as PCSK9 [proprotein convertase subtilisin/kexin type 9] inhibitors) have reduced major cardiovascular and limb events in PAD populations, chronic ischemia of the limb remains largely resistant to medical therapy. Experimental approaches to improve limb outcomes have included the administration of angiogenic cytokines (either as recombinant protein or as gene therapy) as well as cell therapy. Although early angiogenesis and cell therapy studies were promising, these studies lacked sufficient control groups and larger randomized clinical trials have yet to achieve significant benefit. This review will focus on what has been learned to advance medical revascularization for PAD and how that information might lead to novel approaches for therapeutic angiogenesis and arteriogenesis for PAD.
Collapse
Affiliation(s)
- Brian H Annex
- Vascular Biology Center, Department of Medicine, Medical College of Georgia, Augusta University (B.H.A.)
| | - John P Cooke
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, TX (J.P.C.)
| |
Collapse
|
15
|
Wang T, Liu J, Liu H, Lee SR, Gonzalez L, Gorecka J, Shu C, Dardik A. Activation of EphrinB2 Signaling Promotes Adaptive Venous Remodeling in Murine Arteriovenous Fistulae. J Surg Res 2021; 262:224-239. [PMID: 33039109 PMCID: PMC8024410 DOI: 10.1016/j.jss.2020.08.071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 08/26/2020] [Accepted: 08/30/2020] [Indexed: 12/25/2022]
Abstract
BACKGROUND Arteriovenous fistulae (AVF) are the preferred mode of vascular access for hemodialysis. Before use, AVF remodel by thickening and dilating to achieve a functional conduit via an adaptive process characterized by expression of molecular markers characteristic of both venous and arterial identity. Although signaling via EphB4, a determinant of venous identity, mediates AVF maturation, the role of its counterpart EphrinB2, a determinant of arterial identity, remains unclear. We hypothesize that EphrinB2 signaling is active during AVF maturation and may be a mechanism of venous remodeling. METHODS Aortocaval fistulae were created or sham laparotomy was performed in C57Bl/6 mice, and specimens were examined on Days 7 or 21. EphrinB2 reverse signaling was activated with EphB4-Fc applied periadventitially in vivo and in endothelial cell culture medium in vitro. Downstream signaling was assessed using immunoblotting and immunofluorescence. RESULTS Venous remodeling during AVF maturation was characterized by increased expression of EphrinB2 as well as Akt1, extracellular signal-regulated kinases 1/2 (ERK1/2), and p38. Activation of EphrinB2 with EphB4-Fc increased phosphorylation of EphrinB2, endothelial nitric oxide synthase, Akt1, ERK1/2, and p38 and was associated with increased diameter and wall thickness in the AVF. Both mouse and human endothelial cells treated with EphB4-Fc increased phosphorylation of EphrinB2, endothelial nitric oxide synthase, Akt1, ERK1/2, and p38 and increased endothelial cell tube formation and migration. CONCLUSIONS Activation of EphrinB2 signaling by EphB4-Fc was associated with adaptive venous remodeling in vivo while activating endothelial cell function in vitro. Regulation of EphrinB2 signaling may be a new strategy to improve AVF maturation and patency.
Collapse
Affiliation(s)
- Tun Wang
- Department of Vascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China; The Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut; Department of Surgery, Yale School of Medicine, New Haven, Connecticut; Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut
| | - Jia Liu
- Department of Vascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China; The Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut; Department of Surgery, Yale School of Medicine, New Haven, Connecticut; Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut
| | - Haiyang Liu
- The Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut; Department of Surgery, Yale School of Medicine, New Haven, Connecticut; Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut
| | - Shin-Rong Lee
- The Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut; Department of Surgery, Yale School of Medicine, New Haven, Connecticut; Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut
| | - Luis Gonzalez
- The Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut; Department of Surgery, Yale School of Medicine, New Haven, Connecticut; Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut
| | - Jolanta Gorecka
- The Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut; Department of Surgery, Yale School of Medicine, New Haven, Connecticut; Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut
| | - Chang Shu
- Department of Vascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China; State Key Laboratory of Cardiovascular Disease, Center of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Alan Dardik
- The Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut; Department of Surgery, Yale School of Medicine, New Haven, Connecticut; Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut; Department of Surgery, VA Connecticut Healthcare System, West Haven, Connecticut.
| |
Collapse
|
16
|
Ninchoji T, Love DT, Smith RO, Hedlund M, Vestweber D, Sessa WC, Claesson-Welsh L. eNOS-induced vascular barrier disruption in retinopathy by c-Src activation and tyrosine phosphorylation of VE-cadherin. eLife 2021; 10:e64944. [PMID: 33908348 PMCID: PMC8087444 DOI: 10.7554/elife.64944] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 04/22/2021] [Indexed: 12/30/2022] Open
Abstract
Background Hypoxia and consequent production of vascular endothelial growth factor A (VEGFA) promote blood vessel leakiness and edema in ocular diseases. Anti-VEGFA therapeutics may aggravate hypoxia; therefore, therapy development is needed. Methods Oxygen-induced retinopathy was used as a model to test the role of nitric oxide (NO) in pathological neovascularization and vessel permeability. Suppression of NO formation was achieved chemically using L-NMMA, or genetically, in endothelial NO synthase serine to alanine (S1176A) mutant mice. Results Suppression of NO formation resulted in reduced retinal neoangiogenesis. Remaining vascular tufts exhibited reduced vascular leakage through stabilized endothelial adherens junctions, manifested as reduced phosphorylation of vascular endothelial (VE)-cadherin Y685 in a c-Src-dependent manner. Treatment with a single dose of L-NMMA in established retinopathy restored the vascular barrier and prevented leakage. Conclusions We conclude that NO destabilizes adheren junctions, resulting in vascular hyperpermeability, by converging with the VEGFA/VEGFR2/c-Src/VE-cadherin pathway. Funding This study was supported by the Swedish Cancer foundation (19 0119 Pj ), the Swedish Research Council (2020-01349), the Knut and Alice Wallenberg foundation (KAW 2020.0057) and a Fondation Leducq Transatlantic Network of Excellence Grant in Neurovascular Disease (17 CVD 03). KAW also supported LCW with a Wallenberg Scholar grant (2015.0275). WCS was supported by Grants R35 HL139945, P01 HL1070205, AHA MERIT Award. DV was supported by grants from the Deutsche Forschungsgemeinschaft, SFB1450, B03, and CRU342, P2.
Collapse
Affiliation(s)
- Takeshi Ninchoji
- Uppsala University, Rudbeck Laboratory, Department of Immunology, Genetics and PathologyUppsalaSweden
| | - Dominic T Love
- Uppsala University, Rudbeck Laboratory, Department of Immunology, Genetics and PathologyUppsalaSweden
| | - Ross O Smith
- Uppsala University, Rudbeck Laboratory, Department of Immunology, Genetics and PathologyUppsalaSweden
| | - Marie Hedlund
- Uppsala University, Rudbeck Laboratory, Department of Immunology, Genetics and PathologyUppsalaSweden
| | | | - William C Sessa
- Yale University School of Medicine, Department of Pharmacology and Vascular Biology and Therapeutics ProgramNew HavenUnited States
| | - Lena Claesson-Welsh
- Uppsala University, Rudbeck Laboratory, Department of Immunology, Genetics and PathologyUppsalaSweden
| |
Collapse
|
17
|
Dao VTV, Elbatreek MH, Fuchß T, Grädler U, Schmidt HHHW, Shah AM, Wallace A, Knowles R. Nitric Oxide Synthase Inhibitors into the Clinic at Last. Handb Exp Pharmacol 2021; 264:169-204. [PMID: 32797331 DOI: 10.1007/164_2020_382] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The 1998 Nobel Prize in Medicine and Physiology for the discovery of nitric oxide, a nitrogen containing reactive oxygen species (also termed reactive nitrogen or reactive nitrogen/oxygen species) stirred great hopes. Clinical applications, however, have so far pertained exclusively to the downstream signaling of cGMP enhancing drugs such as phosphodiesterase inhibitors and soluble guanylate cyclase stimulators. All clinical attempts, so far, to inhibit NOS have failed even though preclinical models were strikingly positive and clinical biomarkers correlated perfectly. This rather casts doubt on our current way of target identification in drug discovery in general and our way of patient stratification based on correlating but not causal biomarkers or symptoms. The opposite, NO donors, nitrite and enhancing NO synthesis by eNOS/NOS3 recoupling in situations of NO deficiency, are rapidly declining in clinical relevance or hold promise but need yet to enter formal therapeutic guidelines, respectively. Nevertheless, NOS inhibition in situations of NO overproduction often jointly with enhanced superoxide (or hydrogen peroxide production) still holds promise, but most likely only in acute conditions such as neurotrauma (Stover et al., J Neurotrauma 31(19):1599-1606, 2014) and stroke (Kleinschnitz et al., J Cereb Blood Flow Metab 1508-1512, 2016; Casas et al., Proc Natl Acad Sci U S A 116(14):7129-7136, 2019). Conversely, in chronic conditions, long-term inhibition of NOS might be too risky because of off-target effects on eNOS/NOS3 in particular for patients with cardiovascular risks or metabolic and renal diseases. Nitric oxide synthases (NOS) and their role in health (green) and disease (red). Only neuronal/type 1 NOS (NOS1) has a high degree of clinical validation and is in late stage development for traumatic brain injury, followed by a phase II safety/efficacy trial in ischemic stroke. The pathophysiology of NOS1 (Kleinschnitz et al., J Cereb Blood Flow Metab 1508-1512, 2016) is likely to be related to parallel superoxide or hydrogen peroxide formation (Kleinschnitz et al., J Cereb Blood Flow Metab 1508-1512, 2016; Casas et al., Proc Natl Acad Sci U S A 114(46):12315-12320, 2017; Casas et al., Proc Natl Acad Sci U S A 116(14):7129-7136, 2019) leading to peroxynitrite and protein nitration, etc. Endothelial/type 3 NOS (NOS3) is considered protective only and its inhibition should be avoided. The preclinical evidence for a role of high-output inducible/type 2 NOS (NOS2) isoform in sepsis, asthma, rheumatic arthritis, etc. was high, but all clinical development trials in these indications were neutral despite target engagement being validated. This casts doubt on the role of NOS2 in humans in health and disease (hence the neutral, black coloring).
Collapse
Affiliation(s)
- Vu Thao-Vi Dao
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Mahmoud H Elbatreek
- Department of Pharmacology and Personalised Medicine, MeHNS, FHML, Maastricht, The Netherlands.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Thomas Fuchß
- Takeda GmbH (former Nycomed/Altana Pharma), Konstanz, Germany
| | - Ulrich Grädler
- Takeda GmbH (former Nycomed/Altana Pharma), Konstanz, Germany
| | - Harald H H W Schmidt
- Department of Pharmacology and Personalised Medicine, MeHNS, FHML, Maastricht, The Netherlands
| | - Ajay M Shah
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, London, UK
| | - Alan Wallace
- Health and Life Sciences, Coventry University, Coventry, UK
| | - Richard Knowles
- Knowles Consulting Ltd., The Stevenage Bioscience Catalyst, Stevenage, UK.
| |
Collapse
|
18
|
Zhou Q, Tu T, Tai S, Tang L, Yang H, Zhu Z. Endothelial specific deletion of HMGB1 increases blood pressure and retards ischemia recovery through eNOS and ROS pathway in mice. Redox Biol 2021; 41:101890. [PMID: 33582562 PMCID: PMC7887649 DOI: 10.1016/j.redox.2021.101890] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/31/2021] [Accepted: 02/01/2021] [Indexed: 12/13/2022] Open
Abstract
Recent studies demonstrated HMGB1, an extracellular inflammation molecule, played an important role on endothelial cells. This study aimed to define the role and related mechanism of HMGB1 in endothelial cells. Endothelial-specific deletion of HMGB1(HMGB1ECKO) was generated and Akt/eNOS signaling, reactive oxygen species (ROS) production, endothelium dependent relaxation (EDR), and angiogenesis were determined in vitro and in vivo. Decreased activation of Akt/eNOS signaling, sprouting, and proliferation, and increased ROS production were evidenced in endothelial cells derived from HMGB1ECKO mice as compared with wild type controls. Decreased EDR and retarded blood flow recovery after hind limb ischemia were also demonstrated in HMGB1ECKO mice. Both impaired EDR and angiogenesis could be partly rescued by superoxide dismutase in HMGB1ECKO mice. In conclusion, intracellular HMGB1 might be a key regulator of endothelial Akt/eNOS pathway and ROS production, thus plays an important role in EDR regulation and angiogenesis.
Collapse
Affiliation(s)
- Qin Zhou
- Cardiovascular Department, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Tao Tu
- Cardiovascular Department, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shi Tai
- Cardiovascular Department, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Liang Tang
- Cardiovascular Department, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hui Yang
- Cardiovascular Department, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhaowei Zhu
- Cardiovascular Department, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
19
|
Kumar G, Dey SK, Kundu S. Functional implications of vascular endothelium in regulation of endothelial nitric oxide synthesis to control blood pressure and cardiac functions. Life Sci 2020; 259:118377. [PMID: 32898526 DOI: 10.1016/j.lfs.2020.118377] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 08/24/2020] [Accepted: 08/31/2020] [Indexed: 11/29/2022]
Abstract
The endothelium is the innermost vascular lining performing significant roles all over the human body while maintaining the blood pressure at physiological levels. Malfunction of endothelium is thus recognized as a biomarker linked with many vascular diseases including but not limited to atherosclerosis, hypertension and thrombosis. Alternatively, prevention of endothelial malfunctioning or regulating the functions of its associated physiological partners like endothelial nitric oxide synthase can prevent the associated vascular disorders which account for the highest death toll worldwide. While many anti-hypertensive drugs are available commercially, a comprehensive description of the key physiological roles of the endothelium and its regulation by endothelial nitric oxide synthase or vice versa is the need of the hour to understand its contribution in vascular homeostasis. This, in turn, will help in designing new therapeutics targeting endothelial nitric oxide synthase or its interacting partners present in the cellular pool. This review describes the central role of vascular endothelium in the regulation of endothelial nitric oxide synthase while outlining the emerging drug targets present in the vasculature with potential to treat vascular disorders including hypertension.
Collapse
Affiliation(s)
- Gaurav Kumar
- Department of Biochemistry, University of Delhi, South Campus, New Delhi 110021, India
| | - Sanjay Kumar Dey
- Department of Biochemistry, University of Delhi, South Campus, New Delhi 110021, India; Center for Advanced Biotechnology and Medicine, Rutgers University, NJ 08854, USA
| | - Suman Kundu
- Department of Biochemistry, University of Delhi, South Campus, New Delhi 110021, India.
| |
Collapse
|
20
|
Gopallawa I, Lee RJ. Targeting the phosphoinositide-3-kinase/protein kinase B pathway in airway innate immunity. World J Biol Chem 2020; 11:30-51. [PMID: 33024516 PMCID: PMC7520643 DOI: 10.4331/wjbc.v11.i2.30] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/24/2020] [Accepted: 08/26/2020] [Indexed: 02/06/2023] Open
Abstract
The airway innate immune system maintains the first line of defense against respiratory infections. The airway epithelium and associated immune cells protect the respiratory system from inhaled foreign organisms. These cells sense pathogens via activation of receptors like toll-like receptors and taste family 2 receptors (T2Rs) and respond by producing antimicrobials, inflammatory cytokines, and chemokines. Coordinated regulation of fluid secretion and ciliary beating facilitates clearance of pathogens via mucociliary transport. Airway cells also secrete antimicrobial peptides and radicals to directly kill microorganisms and inactivate viruses. The phosphoinositide-3-kinase/protein kinase B (Akt) kinase pathway regulates multiple cellular targets that modulate cell survival and proliferation. Akt also regulates proteins involved in innate immune pathways. Akt phosphorylates endothelial nitric oxide synthase (eNOS) enzymes expressed in airway epithelial cells. Activation of eNOS can have anti-inflammatory, anti-bacterial, and anti-viral roles. Moreover, Akt can increase the activity of the transcription factor nuclear factor erythroid 2 related factor-2 that protects cells from oxidative stress and may limit inflammation. In this review, we summarize the recent findings of non-cancerous functions of Akt signaling in airway innate host defense mechanisms, including an overview of several known downstream targets of Akt involved in innate immunity.
Collapse
Affiliation(s)
- Indiwari Gopallawa
- Department of Otorhinolaryngology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Robert J Lee
- Department of Otorhinolaryngology and Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| |
Collapse
|
21
|
Hsu MF, Koike S, Mello A, Nagy LE, Haj FG. Hepatic protein-tyrosine phosphatase 1B disruption and pharmacological inhibition attenuate ethanol-induced oxidative stress and ameliorate alcoholic liver disease in mice. Redox Biol 2020; 36:101658. [PMID: 32769011 PMCID: PMC7408361 DOI: 10.1016/j.redox.2020.101658] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/29/2020] [Accepted: 07/21/2020] [Indexed: 12/19/2022] Open
Abstract
Alcoholic liver disease (ALD) is a major health problem and a significant cause of liver-related death. Currently, the mainstay for ALD therapy is alcohol abstinence highlighting the need to develop pharmacotherapeutic approaches. Protein-tyrosine phosphatase 1B (PTP1B) is an established regulator of hepatic functions, but its role in ALD is mostly unexplored. In this study, we used mice with liver-specific PTP1B disruption as well as pharmacological inhibition to investigate the in vivo function of this phosphatase in ALD. We report upregulation of hepatic PTP1B in the chronic plus binge mouse model and, importantly, in liver biopsies of alcoholic hepatitis patients. Also, mice with hepatic PTP1B disruption attenuated ethanol-induced injury, inflammation, and steatosis compared with ethanol-fed control animals. Moreover, PTP1B deficiency was associated with decreased ethanol-induced oxidative stress in vivo and ex vivo. Further, pharmacological modulation of oxidative balance in hepatocytes identified diminished oxidative stress as a contributor to the salutary effects of PTP1B deficiency. Notably, PTP1B pharmacological inhibition elicited beneficial effects and mitigated hepatic injury, inflammation, and steatosis caused by ethanol feeding. In summary, these findings causally link hepatic PTP1B and ALD and define a potential therapeutic target for the management of this disease.
Collapse
Affiliation(s)
- Ming-Fo Hsu
- Department of Nutrition, University of California Davis, One Shields Ave, Davis, CA, 95616, USA.
| | - Shinichiro Koike
- Department of Nutrition, University of California Davis, One Shields Ave, Davis, CA, 95616, USA
| | - Aline Mello
- Department of Nutrition, University of California Davis, One Shields Ave, Davis, CA, 95616, USA
| | - Laura E Nagy
- Department of Inflammation and Immunity, Cleveland Clinic Foundation, Cleveland, OH, 44195, USA
| | - Fawaz G Haj
- Department of Nutrition, University of California Davis, One Shields Ave, Davis, CA, 95616, USA; Comprehensive Cancer Center, University of California Davis, Sacramento, CA, 95817, USA; Division of Endocrinology, Diabetes, and Metabolism, Department of Internal Medicine, University of California Davis, Sacramento, CA, 95817, USA.
| |
Collapse
|
22
|
Cheriyan VT, Alfaidi M, Jorgensen AN, Alam MA, Abdullah CS, Kolluru GK, Bhuiyan MS, Kevil CG, Orr AW, Nam HW. Neurogranin regulates eNOS function and endothelial activation. Redox Biol 2020; 34:101487. [PMID: 32173345 PMCID: PMC7327963 DOI: 10.1016/j.redox.2020.101487] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 02/27/2020] [Accepted: 02/29/2020] [Indexed: 02/07/2023] Open
Abstract
Endothelial nitric oxide (NO) is a critical mediator of vascular function and vascular remodeling. NO is produced by endothelial nitric oxide synthase (eNOS), which is activated by calcium (Ca2+)-dependent and Ca2+-independent pathways. Here, we report that neurogranin (Ng), which regulates Ca2+-calmodulin (CaM) signaling in the brain, is uniquely expressed in endothelial cells (EC) of human and mouse vasculature, and is also required for eNOS regulation. To test the role of Ng in eNOS activation, Ng knockdown in human aortic endothelial cells (HAEC) was performed using Ng SiRNA along with Ng knockout (Ng −/−) in mice. Depletion of Ng expression decreased eNOS activity in HAEC and NO production in mice. We show that Ng expression was decreased by short-term laminar flow and long-them oscillating flow shear stress, and that Ng siRNA with shear stress decreased eNOS expression as well as eNOS phosphorylation at S1177. We further reveled that lack of Ng expression decreases both AKT-dependent eNOS phosphorylation, NF-κB-mediated eNOS expression, and promotes endothelial activation. Our findings also indicate that Ng modulates Ca2+-dependent calcineurin (CaN) activity, which suppresses Ca2+-independent AKT-dependent eNOS signaling. Moreover, deletion of Ng in mice also reduced eNOS activity and caused endothelial dysfunction in flow-mediated dilation experiments. Our results demonstrate that Ng plays a crucial role in Ca2+-CaM-dependent eNOS regulation and contributes to vascular remodeling, which is important for the pathophysiology of cardiovascular disease. Neurogranin is expressed in endothelial cell and is required for eNOS regulation. Short-term laminar flow and long-them oscillating flow decrease Neurogranin expression in endothelial cells. Neurogranin knockdown decreases both AKT-dependent eNOS phosphorylation and eNOS expression. Deletion of Ng in mice reduces eNOS activity and caused endothelial dysfunction in flow-mediated dilation.
Collapse
Affiliation(s)
- Vino T Cheriyan
- Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University Health Sciences Center, Shreveport, LA, 71130, USA
| | - Mabruka Alfaidi
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, LA, 71130, USA
| | - Ashton N Jorgensen
- Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University Health Sciences Center, Shreveport, LA, 71130, USA
| | - Md Ashiqul Alam
- Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University Health Sciences Center, Shreveport, LA, 71130, USA
| | - Chowdhury S Abdullah
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, LA, 71130, USA
| | - Gopi K Kolluru
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, LA, 71130, USA
| | - Md Shenuarin Bhuiyan
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, LA, 71130, USA
| | - Christopher G Kevil
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, LA, 71130, USA
| | - A Wayne Orr
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, LA, 71130, USA
| | - Hyung W Nam
- Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University Health Sciences Center, Shreveport, LA, 71130, USA.
| |
Collapse
|
23
|
Casin KM, Kohr MJ. An emerging perspective on sex differences: Intersecting S-nitrosothiol and aldehyde signaling in the heart. Redox Biol 2020; 31:101441. [PMID: 32007450 PMCID: PMC7212482 DOI: 10.1016/j.redox.2020.101441] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 01/20/2020] [Accepted: 01/22/2020] [Indexed: 12/17/2022] Open
Abstract
Cardiovascular disease is the leading cause of the death for both men and women. Although baseline heart physiology and the response to disease are known to differ by sex, little is known about sex differences in baseline molecular signaling, especially with regard to redox biology. In this review, we describe current research on sex differences in cardiac redox biology with a focus on the regulation of nitric oxide and aldehyde signaling. Furthermore, we argue for a new perspective on cardiovascular sex differences research, one that focuses on baseline redox biology without the elimination or disruption of sex hormones.
Collapse
Affiliation(s)
- Kevin M Casin
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Mark J Kohr
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA.
| |
Collapse
|
24
|
Xu W, Comhair SAA, Chen R, Hu B, Hou Y, Zhou Y, Mavrakis LA, Janocha AJ, Li L, Zhang D, Willard BB, Asosingh K, Cheng F, Erzurum SC. Integrative proteomics and phosphoproteomics in pulmonary arterial hypertension. Sci Rep 2019; 9:18623. [PMID: 31819116 PMCID: PMC6901481 DOI: 10.1038/s41598-019-55053-6] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 11/21/2019] [Indexed: 02/06/2023] Open
Abstract
Pulmonary arterial endothelial cells (PAEC) are mechanistically linked to origins of pulmonary arterial hypertension (PAH). Here, global proteomics and phosphoproteomics of PAEC from PAH (n = 4) and healthy lungs (n = 5) were performed using LC-MS/MS to confirm known pathways and identify new areas of investigation in PAH. Among PAH and control cells, 170 proteins and 240 phosphopeptides were differentially expressed; of these, 45 proteins and 18 phosphopeptides were located in the mitochondria. Pathologic pathways were identified with integrative bioinformatics and human protein-protein interactome network analyses, then confirmed with targeted proteomics in PAH PAEC and non-targeted metabolomics and targeted high-performance liquid chromatography of metabolites in plasma from PAH patients (n = 30) and healthy controls (n = 12). Dysregulated pathways in PAH include accelerated one carbon metabolism, abnormal tricarboxylic acid (TCA) cycle flux and glutamate metabolism, dysfunctional arginine and nitric oxide pathways, and increased oxidative stress. Functional studies in cells confirmed abnormalities in glucose metabolism, mitochondrial oxygen consumption, and production of reactive oxygen species in PAH. Altogether, the findings indicate that PAH is typified by changes in metabolic pathways that are primarily found in mitochondria.
Collapse
Affiliation(s)
- Weiling Xu
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America.
| | - Suzy A A Comhair
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Ruoying Chen
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Bo Hu
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Yuan Hou
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Yadi Zhou
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Lori A Mavrakis
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Allison J Janocha
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Ling Li
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Dongmei Zhang
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Belinda B Willard
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Kewal Asosingh
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Feixiong Cheng
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Serpil C Erzurum
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America. .,Respiratory Institute, Cleveland Clinic, Cleveland, Ohio, United States of America.
| |
Collapse
|
25
|
Akt1-mediated CPR cooling protection targets regulators of metabolism, inflammation and contractile function in mouse cardiac arrest. PLoS One 2019; 14:e0220604. [PMID: 31398213 PMCID: PMC6688812 DOI: 10.1371/journal.pone.0220604] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 07/21/2019] [Indexed: 12/31/2022] Open
Abstract
Therapeutic hypothermia initiated during cardiopulmonary resuscitation (CPR) in pre-clinical studies appears to be highly protective against sudden cardiac arrest injury. Given the challenges to implementing CPR cooling clinically, insights into its critical mechanisms of protection could guide development of new CPR drugs that mimic hypothermia effects without the need for physical cooling. Here, we used Akt1-deficient mice that lose CPR hypothermia protection to identify hypothermia targets. Adult female C57BL/6 mice (Akt1+/+ and Akt1+/-) underwent 8 min of KCl-induced asystolic arrest and were randomized to receive hypothermia (30 ± 0.5°C) or normothermia. Hypothermia was initiated during CPR and extended for 1 h after resuscitation. Neurologically scored survival was measured at 72 h. Other outcomes included mean arterial pressure and target measures in heart and brain related to contractile function, glucose utilization and inflammation. Compared to northothermia, hypothermia improved both 2h mean arterial pressure and 72h neurologically intact survival in Akt1+/+ mice but not in Akt1+/- mice. In Akt1+/+ mice, hypothermia increased Akt and GSK3β phosphorylation, pyruvate dehydrogenase activation, and NAD+ and ATP production while decreasing IκBα degradation and NF-κB activity in both heart and brain at 30 min after CPR. It also increased phospholamban phosphorylation in heart tissue. Further, hypothermia reduced metabolic and inflammatory blood markers lactate and Pre-B cell Colony Enhancing Factor. Despite hypothermia treatment, all these effects were reversed in Akt1+/- mice. Taken together, drugs that target Akt1 and its effectors may have the potential to mimic hypothermia-like protection to improve sudden cardiac arrest survival when administered during CPR.
Collapse
|
26
|
Alwhaibi A, Verma A, Adil MS, Somanath PR. The unconventional role of Akt1 in the advanced cancers and in diabetes-promoted carcinogenesis. Pharmacol Res 2019; 145:104270. [PMID: 31078742 PMCID: PMC6659399 DOI: 10.1016/j.phrs.2019.104270] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 04/03/2019] [Accepted: 05/08/2019] [Indexed: 12/15/2022]
Abstract
Decades of research have elucidated the critical role of Akt isoforms in cancer as pro-tumorigenic and metastatic regulators through their specific effects on the cancer cells, tumor endothelial cells and the stromal cells. The pro-cancerous role of Akt isoforms through enhanced cell proliferation and suppression of apoptosis in cancer cells and the cells in the tumor microenvironment is considered a dogma. Intriguingly, studies also indicate that the Akt pathway is essential to protect the endothelial-barrier and prevent aberrant vascular permeability, which is also integral to tumor perfusion and metastasis. To complicate this further, a flurry of recent reports strongly indicates the metastasis suppressive role of Akt, Akt1 in particular in various cancer types. These reports emanated from different laboratories have elegantly demonstrated the paradoxical effect of Akt1 on cancer cell epithelial-to-mesenchymal transition, invasion, tumor endothelial-barrier disruption, and cancer metastasis. Here, we emphasize on the specific role of Akt1 in mediating tumor cell-vasculature reciprocity during the advanced stages of cancers and discuss how Akt1 differentially regulates cancer metastasis through mechanisms distinct from its pro-tumorigenic effects. Since Akt is integral for insulin signaling, endothelial function, and metabolic regulation, we also attempt to shed some light on the specific effects of diabetes in modulating Akt pathway in the promotion of tumor growth and metastasis.
Collapse
Affiliation(s)
- Abdulrahman Alwhaibi
- Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and the Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Arti Verma
- Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and the Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Mir S Adil
- Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and the Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Payaningal R Somanath
- Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and the Charlie Norwood VA Medical Center, Augusta, GA, USA; Department of Medicine, Vascular Biology Center and Cancer Center, Augusta University, USA.
| |
Collapse
|
27
|
Garcia V, Sessa WC. Endothelial NOS: perspective and recent developments. Br J Pharmacol 2019; 176:189-196. [PMID: 30341769 PMCID: PMC6295413 DOI: 10.1111/bph.14522] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 10/04/2018] [Indexed: 02/06/2023] Open
Abstract
Endothelial NOS (eNOS), and its product NO, are vital components of the control of vasomotor function and cardiovascular homeostasis. In the present review, we will take a deep dive into eNOS enzymology, function and mechanisms regulating endothelial NO. The mechanisms regulating eNOS and NO synthesis discussed here include alterations to transcriptional, post-translational modifications and protein-protein regulations. Also, we will discuss the phenotypes associated with various eNOS mutants and the consequences of a disrupted eNOS/NO cascade, highlighting the importance of eNOS function and vascular homeostasis. LINKED ARTICLES: This article is part of a themed section on Nitric Oxide 20 Years from the 1998 Nobel Prize. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.2/issuetoc.
Collapse
Affiliation(s)
- Victor Garcia
- Vascular Biology and Therapeutics Program, Department of PharmacologyYale University School of MedicineNew HavenCTUSA
| | - William C Sessa
- Vascular Biology and Therapeutics Program, Department of PharmacologyYale University School of MedicineNew HavenCTUSA
| |
Collapse
|
28
|
Fu J, Zou J, Chen C, Li H, Wang L, Zhou Y. Hydrogen molecules (H2) improve perfusion recovery via antioxidant effects in experimental peripheral arterial disease. Mol Med Rep 2018; 18:5009-5015. [PMID: 30320393 PMCID: PMC6236306 DOI: 10.3892/mmr.2018.9546] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Accepted: 09/03/2018] [Indexed: 11/24/2022] Open
Abstract
Reactive oxygen species (ROS) impair neovascularization and perfusion recovery following limb ischemia in patients with peripheral arterial disease (PAD). Hydrogen molecules (H2) comprise an antioxidant gas that has been reported to neutralize cytotoxic ROS. The present study investigated whether H2 may serve as a novel therapeutic strategy for PAD. H2-saturated water or dehydrogenized water was supplied to mice with experimental PAD. Laser Doppler perfusion imaging demonstrated that H2-saturated water improved perfusion recovery, decreased the rate of necrosis, increased the capillary density in the gastrocnemius muscle and increased the artery density in the abductor muscle in the ischemic limbs, at 14 and 21 days post-hindlimb ischemia. Ischemic muscle tissue was harvested 7 days after experimental PAD for biochemical testing and H2 was observed to reduce the levels of malondialdehyde and increase the levels of cyclic guanine monophosphate (cGMP). In cultured endothelial cells, H2-saturated culture medium resulted in reduced ROS levels, increased tube formation and increased cGMP levels. In macrophages, H2 decreased cellular ROS levels and promoted M2 polarization. H2-saturated water increases angiogenesis and arteriogenesis and subsequently improves perfusion recovery in a mouse PAD model via reduction of ROS levels.
Collapse
Affiliation(s)
- Jinrong Fu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jinjing Zou
- Department of Respiratory Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Cheng Chen
- Department of Respiratory Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Hongying Li
- Department of Gynecology, Hubei Maternal and Child Hospital, Wuhan, Hubei 430070, P.R. China
| | - Lei Wang
- Department of Cardiology, Hubei University of Chinese Medicine, Wuhan, Hubei 430060, P.R. China
| | - Yanli Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
29
|
Rabender CS, Bruno N, Alam A, Sundaresan G, Zweit J, Mikkelsen RB. Sepiapterin Enhances Tumor Radio- and Chemosensitivities by Promoting Vascular Normalization. J Pharmacol Exp Ther 2018; 365:536-543. [PMID: 29581154 PMCID: PMC11046730 DOI: 10.1124/jpet.117.245258] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 02/26/2018] [Indexed: 12/22/2022] Open
Abstract
Previously, we demonstrated that nitric oxide (NO) synthase (NOS) is uncoupled in a wide range of solid tumors and that restoring NOS coupling with the tetrahydrobiopterin precursor sepiapterin (SP) inhibits tumor progression. Endothelial dysfunction characterizes the poorly functional vasculature of solid tumors, and since NO is critical for regulation of endothelial function we asked whether SP, by recoupling NOS, improves tumor vasculature structure and function-enhancing chemotherapeutic delivery and response to radiotherapy. MMTV-neu mice with spontaneous breast tumors were treated with SP by oral gavage and evaluated by multispectral optoacoustic tomographic analysis of tumor HbO2 and by tissue staining for markers of hypoxia, blood perfusion, and markers of endothelial and smooth muscle proteins. Recoupling tumor NOS activity results in vascular normalization observed as reduced tumor hypoxia, improved tumor percentage of HbO2 and perfusion, as well as increased pericyte coverage of tumor blood vessels. The normalized vasculature and improved tumor oxygenation led to a greater than 2-fold increase in radiation-induced apoptosis compared with radiation or SP alone. High-performance liquid chromatography analysis of tumor doxorubicin levels showed a greater than 50% increase in doxorubicin uptake and a synergistic effect on tumor cell apoptosis. This study highlights for the first time the importance of NOS uncoupling and endothelial dysfunction in the development of tumor vasculature and presents a new approach for improving the tumoricidal efficacies of chemotherapy and radiotherapy.
Collapse
Affiliation(s)
- Christopher S Rabender
- Department of Radiation Oncology (C.S.R., N.B., A.A., R.B.M.) and Center for Molecular Imaging (G.S., J.Z.), Virginia Commonwealth University, Richmond, Virginia
| | - Ninu Bruno
- Department of Radiation Oncology (C.S.R., N.B., A.A., R.B.M.) and Center for Molecular Imaging (G.S., J.Z.), Virginia Commonwealth University, Richmond, Virginia
| | - Asim Alam
- Department of Radiation Oncology (C.S.R., N.B., A.A., R.B.M.) and Center for Molecular Imaging (G.S., J.Z.), Virginia Commonwealth University, Richmond, Virginia
| | - Gobalakrishnan Sundaresan
- Department of Radiation Oncology (C.S.R., N.B., A.A., R.B.M.) and Center for Molecular Imaging (G.S., J.Z.), Virginia Commonwealth University, Richmond, Virginia
| | - Jamal Zweit
- Department of Radiation Oncology (C.S.R., N.B., A.A., R.B.M.) and Center for Molecular Imaging (G.S., J.Z.), Virginia Commonwealth University, Richmond, Virginia
| | - Ross B Mikkelsen
- Department of Radiation Oncology (C.S.R., N.B., A.A., R.B.M.) and Center for Molecular Imaging (G.S., J.Z.), Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
30
|
Lee MY, Gamez-Mendez A, Zhang J, Zhuang Z, Vinyard DJ, Kraehling J, Velazquez H, Brudvig GW, Kyriakides TR, Simons M, Sessa WC. Endothelial Cell Autonomous Role of Akt1: Regulation of Vascular Tone and Ischemia-Induced Arteriogenesis. Arterioscler Thromb Vasc Biol 2018; 38:870-879. [PMID: 29449333 DOI: 10.1161/atvbaha.118.310748] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 01/25/2018] [Indexed: 12/30/2022]
Abstract
OBJECTIVE The importance of PI3K/Akt signaling in the vasculature has been demonstrated in several models, as global loss of Akt1 results in impaired postnatal ischemia- and VEGF-induced angiogenesis. The ubiquitous expression of Akt1, however, raises the possibility of cell-type-dependent Akt1-driven actions, thereby necessitating tissue-specific characterization. APPROACH AND RESULTS Herein, we used an inducible, endothelial-specific Akt1-deleted adult mouse model (Akt1iECKO) to characterize the endothelial cell autonomous functions of Akt1 in the vascular system. Endothelial-targeted ablation of Akt1 reduces eNOS (endothelial nitric oxide synthase) phosphorylation and promotes both increased vascular contractility in isolated vessels and elevated diastolic blood pressures throughout the diurnal cycle in vivo. Furthermore, Akt1iECKO mice subject to the hindlimb ischemia model display impaired blood flow and decreased arteriogenesis. CONCLUSIONS Endothelial Akt1 signaling is necessary for ischemic resolution post-injury and likely reflects the consequence of NO insufficiency critical for vascular repair.
Collapse
Affiliation(s)
- Monica Y Lee
- From the Vascular Biology and Therapeutics Program, Department of Pharmacology (M.Y.L., A.G.-M., J.K., W.C.S.), Vascular Biology and Therapeutics Program, Department of Pathology (T.R.K.), and Department of Cell Biology (M.S.), Yale University School of Medicine, New Haven, CT; Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, New Haven, CT (J.Z., Z.Z., M.S.); Department of Chemistry, Yale University, New Haven, CT (D.J.V., G.W.B.); and Department of Internal Medicine, VA Connecticut Healthcare System, West Haven, CT (H.V.)
| | - Ana Gamez-Mendez
- From the Vascular Biology and Therapeutics Program, Department of Pharmacology (M.Y.L., A.G.-M., J.K., W.C.S.), Vascular Biology and Therapeutics Program, Department of Pathology (T.R.K.), and Department of Cell Biology (M.S.), Yale University School of Medicine, New Haven, CT; Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, New Haven, CT (J.Z., Z.Z., M.S.); Department of Chemistry, Yale University, New Haven, CT (D.J.V., G.W.B.); and Department of Internal Medicine, VA Connecticut Healthcare System, West Haven, CT (H.V.)
| | - Jiasheng Zhang
- From the Vascular Biology and Therapeutics Program, Department of Pharmacology (M.Y.L., A.G.-M., J.K., W.C.S.), Vascular Biology and Therapeutics Program, Department of Pathology (T.R.K.), and Department of Cell Biology (M.S.), Yale University School of Medicine, New Haven, CT; Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, New Haven, CT (J.Z., Z.Z., M.S.); Department of Chemistry, Yale University, New Haven, CT (D.J.V., G.W.B.); and Department of Internal Medicine, VA Connecticut Healthcare System, West Haven, CT (H.V.)
| | - Zhenwu Zhuang
- From the Vascular Biology and Therapeutics Program, Department of Pharmacology (M.Y.L., A.G.-M., J.K., W.C.S.), Vascular Biology and Therapeutics Program, Department of Pathology (T.R.K.), and Department of Cell Biology (M.S.), Yale University School of Medicine, New Haven, CT; Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, New Haven, CT (J.Z., Z.Z., M.S.); Department of Chemistry, Yale University, New Haven, CT (D.J.V., G.W.B.); and Department of Internal Medicine, VA Connecticut Healthcare System, West Haven, CT (H.V.)
| | - David J Vinyard
- From the Vascular Biology and Therapeutics Program, Department of Pharmacology (M.Y.L., A.G.-M., J.K., W.C.S.), Vascular Biology and Therapeutics Program, Department of Pathology (T.R.K.), and Department of Cell Biology (M.S.), Yale University School of Medicine, New Haven, CT; Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, New Haven, CT (J.Z., Z.Z., M.S.); Department of Chemistry, Yale University, New Haven, CT (D.J.V., G.W.B.); and Department of Internal Medicine, VA Connecticut Healthcare System, West Haven, CT (H.V.)
| | - Jan Kraehling
- From the Vascular Biology and Therapeutics Program, Department of Pharmacology (M.Y.L., A.G.-M., J.K., W.C.S.), Vascular Biology and Therapeutics Program, Department of Pathology (T.R.K.), and Department of Cell Biology (M.S.), Yale University School of Medicine, New Haven, CT; Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, New Haven, CT (J.Z., Z.Z., M.S.); Department of Chemistry, Yale University, New Haven, CT (D.J.V., G.W.B.); and Department of Internal Medicine, VA Connecticut Healthcare System, West Haven, CT (H.V.)
| | - Heino Velazquez
- From the Vascular Biology and Therapeutics Program, Department of Pharmacology (M.Y.L., A.G.-M., J.K., W.C.S.), Vascular Biology and Therapeutics Program, Department of Pathology (T.R.K.), and Department of Cell Biology (M.S.), Yale University School of Medicine, New Haven, CT; Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, New Haven, CT (J.Z., Z.Z., M.S.); Department of Chemistry, Yale University, New Haven, CT (D.J.V., G.W.B.); and Department of Internal Medicine, VA Connecticut Healthcare System, West Haven, CT (H.V.)
| | - Gary W Brudvig
- From the Vascular Biology and Therapeutics Program, Department of Pharmacology (M.Y.L., A.G.-M., J.K., W.C.S.), Vascular Biology and Therapeutics Program, Department of Pathology (T.R.K.), and Department of Cell Biology (M.S.), Yale University School of Medicine, New Haven, CT; Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, New Haven, CT (J.Z., Z.Z., M.S.); Department of Chemistry, Yale University, New Haven, CT (D.J.V., G.W.B.); and Department of Internal Medicine, VA Connecticut Healthcare System, West Haven, CT (H.V.)
| | - Themis R Kyriakides
- From the Vascular Biology and Therapeutics Program, Department of Pharmacology (M.Y.L., A.G.-M., J.K., W.C.S.), Vascular Biology and Therapeutics Program, Department of Pathology (T.R.K.), and Department of Cell Biology (M.S.), Yale University School of Medicine, New Haven, CT; Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, New Haven, CT (J.Z., Z.Z., M.S.); Department of Chemistry, Yale University, New Haven, CT (D.J.V., G.W.B.); and Department of Internal Medicine, VA Connecticut Healthcare System, West Haven, CT (H.V.)
| | - Michael Simons
- From the Vascular Biology and Therapeutics Program, Department of Pharmacology (M.Y.L., A.G.-M., J.K., W.C.S.), Vascular Biology and Therapeutics Program, Department of Pathology (T.R.K.), and Department of Cell Biology (M.S.), Yale University School of Medicine, New Haven, CT; Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, New Haven, CT (J.Z., Z.Z., M.S.); Department of Chemistry, Yale University, New Haven, CT (D.J.V., G.W.B.); and Department of Internal Medicine, VA Connecticut Healthcare System, West Haven, CT (H.V.)
| | - William C Sessa
- From the Vascular Biology and Therapeutics Program, Department of Pharmacology (M.Y.L., A.G.-M., J.K., W.C.S.), Vascular Biology and Therapeutics Program, Department of Pathology (T.R.K.), and Department of Cell Biology (M.S.), Yale University School of Medicine, New Haven, CT; Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, New Haven, CT (J.Z., Z.Z., M.S.); Department of Chemistry, Yale University, New Haven, CT (D.J.V., G.W.B.); and Department of Internal Medicine, VA Connecticut Healthcare System, West Haven, CT (H.V.).
| |
Collapse
|
31
|
Labrousse-Arias D, Martínez-Ruiz A, Calzada MJ. Hypoxia and Redox Signaling on Extracellular Matrix Remodeling: From Mechanisms to Pathological Implications. Antioxid Redox Signal 2017; 27:802-822. [PMID: 28715969 DOI: 10.1089/ars.2017.7275] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
SIGNIFICANCE The extracellular matrix (ECM) is an essential modulator of cell behavior that influences tissue organization. It has a strong relevance in homeostasis and translational implications for human disease. In addition to ECM structural proteins, matricellular proteins are important regulators of the ECM that are involved in a myriad of different pathologies. Recent Advances: Biochemical studies, animal models, and study of human diseases have contributed to the knowledge of molecular mechanisms involved in remodeling of the ECM, both in homeostasis and disease. Some of them might help in the development of new therapeutic strategies. This review aims to review what is known about some of the most studied matricellular proteins and their regulation by hypoxia and redox signaling, as well as the pathological implications of such regulation. CRITICAL ISSUES Matricellular proteins have complex regulatory functions and are modulated by hypoxia and redox signaling through diverse mechanisms, in some cases with controversial effects that can be cell or tissue specific and context dependent. Therefore, a better understanding of these regulatory processes would be of great benefit and will open new avenues of considerable therapeutic potential. FUTURE DIRECTIONS Characterizing the specific molecular mechanisms that modulate matricellular proteins in pathological processes that involve hypoxia and redox signaling warrants additional consideration to harness the potential therapeutic value of these regulatory proteins. Antioxid. Redox Signal. 27, 802-822.
Collapse
Affiliation(s)
- David Labrousse-Arias
- 1 Servicio de Inmunología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP) , Madrid, Spain
| | - Antonio Martínez-Ruiz
- 1 Servicio de Inmunología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP) , Madrid, Spain .,2 Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV) , Madrid, Spain
| | - María J Calzada
- 1 Servicio de Inmunología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP) , Madrid, Spain .,3 Departmento de Medicina, Universidad Autónoma de Madrid , Madrid, Spain
| |
Collapse
|
32
|
Atochin D, Litvak M, Huang S, Kim YR, Huang P. Role of eNOS in water exchange index maintenance-MRI studies. ACTA ACUST UNITED AC 2017. [DOI: 10.1088/1742-6596/886/1/012002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
33
|
Ha JM, Jin SY, Lee HS, Shin HK, Lee DH, Song SH, Kim CD, Bae SS. Regulation of retinal angiogenesis by endothelial nitric oxide synthase signaling pathway. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2016; 20:533-8. [PMID: 27610040 PMCID: PMC5015000 DOI: 10.4196/kjpp.2016.20.5.533] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 06/27/2016] [Accepted: 07/14/2016] [Indexed: 11/27/2022]
Abstract
Angiogenesis plays an essential role in embryo development, tissue repair, inflammatory diseases, and tumor growth. In the present study, we showed that endothelial nitric oxide synthase (eNOS) regulates retinal angiogenesis. Mice that lack eNOS showed growth retardation, and retinal vessel development was significantly delayed. In addition, the number of tip cells and filopodia length were significantly reduced in mice lacking eNOS. Retinal endothelial cell proliferation was significantly blocked in mice lacking eNOS, and EMG-2-induced endothelial cell sprouting was significantly reduced in aortic vessels isolated from eNOS-deficient mice. Finally, pericyte recruitment to endothelial cells and vascular smooth muscle cell coverage to blood vessels were attenuated in mice lacking eNOS. Taken together, we suggest that the endothelial cell function and blood vessel maturation are regulated by eNOS during retinal angiogenesis.
Collapse
Affiliation(s)
- Jung Min Ha
- Gene and Cell Therapy Center for Vessel-Associated Disease, Medical Research Institute, and Department of Pharmacology, Pusan National University School of Medicine, Yangsan 50612, Korea
| | - Seo Yeon Jin
- Gene and Cell Therapy Center for Vessel-Associated Disease, Medical Research Institute, and Department of Pharmacology, Pusan National University School of Medicine, Yangsan 50612, Korea
| | - Hye Sun Lee
- Gene and Cell Therapy Center for Vessel-Associated Disease, Medical Research Institute, and Department of Pharmacology, Pusan National University School of Medicine, Yangsan 50612, Korea
| | - Hwa Kyoung Shin
- Department of Anatomy, Pusan National University of Korean Medicine, Yangsan 50612, Korea
| | - Dong Hyung Lee
- Department of Obstetrics and Gynecology, Pusan National University Hospital, Yangsan 50612, Korea
| | - Sang Heon Song
- Department of Internal Medicine, Pusan National University Hospital, Busan 49241, Korea
| | - Chi Dae Kim
- Gene and Cell Therapy Center for Vessel-Associated Disease, Medical Research Institute, and Department of Pharmacology, Pusan National University School of Medicine, Yangsan 50612, Korea
| | - Sun Sik Bae
- Gene and Cell Therapy Center for Vessel-Associated Disease, Medical Research Institute, and Department of Pharmacology, Pusan National University School of Medicine, Yangsan 50612, Korea
| |
Collapse
|
34
|
Li X, Padhan N, Sjöström EO, Roche FP, Testini C, Honkura N, Sáinz-Jaspeado M, Gordon E, Bentley K, Philippides A, Tolmachev V, Dejana E, Stan RV, Vestweber D, Ballmer-Hofer K, Betsholtz C, Pietras K, Jansson L, Claesson-Welsh L. VEGFR2 pY949 signalling regulates adherens junction integrity and metastatic spread. Nat Commun 2016; 7:11017. [PMID: 27005951 PMCID: PMC4814575 DOI: 10.1038/ncomms11017] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 02/09/2016] [Indexed: 01/11/2023] Open
Abstract
The specific role of VEGFA-induced permeability and vascular leakage in physiology and pathology has remained unclear. Here we show that VEGFA-induced vascular leakage depends on signalling initiated via the VEGFR2 phosphosite Y949, regulating dynamic c-Src and VE-cadherin phosphorylation. Abolished Y949 signalling in the mouse mutant Vegfr2Y949F/Y949F leads to VEGFA-resistant endothelial adherens junctions and a block in molecular extravasation. Vessels in Vegfr2Y949F/Y949F mice remain sensitive to inflammatory cytokines, and vascular morphology, blood pressure and flow parameters are normal. Tumour-bearing Vegfr2Y949F/Y949F mice display reduced vascular leakage and oedema, improved response to chemotherapy and, importantly, reduced metastatic spread. The inflammatory infiltration in the tumour micro-environment is unaffected. Blocking VEGFA-induced disassembly of endothelial junctions, thereby suppressing tumour oedema and metastatic spread, may be preferable to full vascular suppression in the treatment of certain cancer forms. Signals through VEGF receptor 2 (VEGFR2) increase vascular permeability, promoting cancer progression. Here the authors show that a point mutation in VEGFR2 preventing its auto-phosphorylation leads to reduced metastatic spread and improved response to chemotherapy in tumor-bearing mice, without affecting tumor inflammation.
Collapse
Affiliation(s)
- Xiujuan Li
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Narendra Padhan
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Elisabet O Sjöström
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Francis P Roche
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Chiara Testini
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Naoki Honkura
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Miguel Sáinz-Jaspeado
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Emma Gordon
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Katie Bentley
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden.,Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, Massachusetts 02215, USA
| | - Andrew Philippides
- Centre for Computational Neuroscience and Robotics, University of Sussex, Chichester 1 CI 104, Brighton BN1 9RH, UK
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Elisabetta Dejana
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden.,c/o IFOM-IEO Campus, Via Adamello, 16, 20139 Milan, Italy
| | - Radu V Stan
- Department of Pathology, Dartmouth College, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire 03756, USA
| | - Dietmar Vestweber
- Department of Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
| | - Kurt Ballmer-Hofer
- Biomolecular Research, Molecular Cell Biology, Paul-Scherrer Institute, 5232 Villigen-PSI, Switzerland
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden.,Karolinska Institutet, Dept. Medical Biochemistry and Biophysics, Div. Vascular Biology, 17177 Stockholm, Sweden
| | - Kristian Pietras
- Translational Cancer Research, Medicon Village, Lund University, Building 404:A3, 22381 Lund, Sweden
| | - Leif Jansson
- Department of Medical Cell Biology, Biomedical Center, Uppsala University, Box 571, 751 23 Uppsala, Sweden
| | - Lena Claesson-Welsh
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| |
Collapse
|
35
|
Kraehling JR, Hao Z, Lee MY, Vinyard DJ, Velazquez H, Liu X, Stan RV, Brudvig GW, Sessa WC. Uncoupling Caveolae From Intracellular Signaling In Vivo. Circ Res 2015; 118:48-55. [PMID: 26602865 DOI: 10.1161/circresaha.115.307767] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 11/24/2015] [Indexed: 11/16/2022]
Abstract
RATIONALE Caveolin-1 (Cav-1) negatively regulates endothelial nitric oxide (NO) synthase-derived NO production, and this has been mapped to several residues on Cav-1, including F92. Herein, we reasoned that endothelial expression of an F92ACav-1 transgene would let us decipher the mechanisms and relationships between caveolae structure and intracellular signaling. OBJECTIVE This study was designed to separate caveolae formation from its downstream signaling effects. METHODS AND RESULTS An endothelial-specific doxycycline-regulated mouse model for the expression of Cav-1-F92A was developed. Blood pressure by telemetry and nitric oxide bioavailability by electron paramagnetic resonance and phosphorylation of vasodilator-stimulated phosphoprotein were determined. Caveolae integrity in the presence of Cav-1-F92A was measured by stabilization of caveolin-2, sucrose gradient, and electron microscopy. Histological analysis of heart and lung, echocardiography, and signaling were performed. CONCLUSIONS This study shows that mutant Cav-1-F92A forms caveolae structures similar to WT but leads to increases in NO bioavailability in vivo, thereby demonstrating that caveolae formation and downstream signaling events occur through independent mechanisms.
Collapse
Affiliation(s)
- Jan R Kraehling
- From the Vascular Biology and Therapeutics Program (J.R.K., Z.H., M.Y.L., W.C.S.) and Department of Pharmacology (J.R.K., Z.H., M.Y.L., W.C.S.), Yale University School of Medicine, New Haven, CT; Department of Chemistry, Yale University, New Haven, CT (D.J.V., G.W.B.); Department of Internal Medicine, VA Connecticut Healthcare System, West Haven, CT (H.V.); Department of Cell Biology, Yale University, School of Medicine, New Haven, CT (X.L.); and Department of Pathology, Dartmouth Medical School, Lebanon, NH (R.V.S.)
| | - Zhengrong Hao
- From the Vascular Biology and Therapeutics Program (J.R.K., Z.H., M.Y.L., W.C.S.) and Department of Pharmacology (J.R.K., Z.H., M.Y.L., W.C.S.), Yale University School of Medicine, New Haven, CT; Department of Chemistry, Yale University, New Haven, CT (D.J.V., G.W.B.); Department of Internal Medicine, VA Connecticut Healthcare System, West Haven, CT (H.V.); Department of Cell Biology, Yale University, School of Medicine, New Haven, CT (X.L.); and Department of Pathology, Dartmouth Medical School, Lebanon, NH (R.V.S.)
| | - Monica Y Lee
- From the Vascular Biology and Therapeutics Program (J.R.K., Z.H., M.Y.L., W.C.S.) and Department of Pharmacology (J.R.K., Z.H., M.Y.L., W.C.S.), Yale University School of Medicine, New Haven, CT; Department of Chemistry, Yale University, New Haven, CT (D.J.V., G.W.B.); Department of Internal Medicine, VA Connecticut Healthcare System, West Haven, CT (H.V.); Department of Cell Biology, Yale University, School of Medicine, New Haven, CT (X.L.); and Department of Pathology, Dartmouth Medical School, Lebanon, NH (R.V.S.)
| | - David J Vinyard
- From the Vascular Biology and Therapeutics Program (J.R.K., Z.H., M.Y.L., W.C.S.) and Department of Pharmacology (J.R.K., Z.H., M.Y.L., W.C.S.), Yale University School of Medicine, New Haven, CT; Department of Chemistry, Yale University, New Haven, CT (D.J.V., G.W.B.); Department of Internal Medicine, VA Connecticut Healthcare System, West Haven, CT (H.V.); Department of Cell Biology, Yale University, School of Medicine, New Haven, CT (X.L.); and Department of Pathology, Dartmouth Medical School, Lebanon, NH (R.V.S.)
| | - Heino Velazquez
- From the Vascular Biology and Therapeutics Program (J.R.K., Z.H., M.Y.L., W.C.S.) and Department of Pharmacology (J.R.K., Z.H., M.Y.L., W.C.S.), Yale University School of Medicine, New Haven, CT; Department of Chemistry, Yale University, New Haven, CT (D.J.V., G.W.B.); Department of Internal Medicine, VA Connecticut Healthcare System, West Haven, CT (H.V.); Department of Cell Biology, Yale University, School of Medicine, New Haven, CT (X.L.); and Department of Pathology, Dartmouth Medical School, Lebanon, NH (R.V.S.)
| | - Xinran Liu
- From the Vascular Biology and Therapeutics Program (J.R.K., Z.H., M.Y.L., W.C.S.) and Department of Pharmacology (J.R.K., Z.H., M.Y.L., W.C.S.), Yale University School of Medicine, New Haven, CT; Department of Chemistry, Yale University, New Haven, CT (D.J.V., G.W.B.); Department of Internal Medicine, VA Connecticut Healthcare System, West Haven, CT (H.V.); Department of Cell Biology, Yale University, School of Medicine, New Haven, CT (X.L.); and Department of Pathology, Dartmouth Medical School, Lebanon, NH (R.V.S.)
| | - Radu V Stan
- From the Vascular Biology and Therapeutics Program (J.R.K., Z.H., M.Y.L., W.C.S.) and Department of Pharmacology (J.R.K., Z.H., M.Y.L., W.C.S.), Yale University School of Medicine, New Haven, CT; Department of Chemistry, Yale University, New Haven, CT (D.J.V., G.W.B.); Department of Internal Medicine, VA Connecticut Healthcare System, West Haven, CT (H.V.); Department of Cell Biology, Yale University, School of Medicine, New Haven, CT (X.L.); and Department of Pathology, Dartmouth Medical School, Lebanon, NH (R.V.S.)
| | - Gary W Brudvig
- From the Vascular Biology and Therapeutics Program (J.R.K., Z.H., M.Y.L., W.C.S.) and Department of Pharmacology (J.R.K., Z.H., M.Y.L., W.C.S.), Yale University School of Medicine, New Haven, CT; Department of Chemistry, Yale University, New Haven, CT (D.J.V., G.W.B.); Department of Internal Medicine, VA Connecticut Healthcare System, West Haven, CT (H.V.); Department of Cell Biology, Yale University, School of Medicine, New Haven, CT (X.L.); and Department of Pathology, Dartmouth Medical School, Lebanon, NH (R.V.S.)
| | - William C Sessa
- From the Vascular Biology and Therapeutics Program (J.R.K., Z.H., M.Y.L., W.C.S.) and Department of Pharmacology (J.R.K., Z.H., M.Y.L., W.C.S.), Yale University School of Medicine, New Haven, CT; Department of Chemistry, Yale University, New Haven, CT (D.J.V., G.W.B.); Department of Internal Medicine, VA Connecticut Healthcare System, West Haven, CT (H.V.); Department of Cell Biology, Yale University, School of Medicine, New Haven, CT (X.L.); and Department of Pathology, Dartmouth Medical School, Lebanon, NH (R.V.S.)
| |
Collapse
|
36
|
Freitas M, Rodrigues AR, Tomada N, Fonseca J, Magalhães A, Gouveia AM, Neves D. Effects of Aging and Cardiovascular Disease Risk Factors on the Expression of Sirtuins in the Human Corpus Cavernosum. J Sex Med 2015; 12:2141-52. [PMID: 26556180 DOI: 10.1111/jsm.13035] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Sirtuin (SIRT)1 was recently identified in human corpus cavernosum (CC). We hypothesized that other sirtuins could also be expressed in the CC. Expression of these enzymes in tissues is affected by aging, the main independent risk factor for erectile dysfunction besides other cardiovascular disease risk factors (CVDRF), such as diabetes or obesity. AIM The aim of this study was to characterize the expression of SIRT1-3 and SIRT5-7 in human CC relatively to age and CVDRF. METHODS Samples of CC collected from patients submitted to programmed surgeries or organ donors were divided in three groups according to age and presence of CVDRF. Expression of SIRT1-3 and SIRT5-7 mRNAs was assessed by real-time polymerase chain reaction. Cellular localization and semi-quantification of sirtuins proteins were performed by immunofluorescence and Western blotting (WB), respectively. Nuclear factor kappa B (NFkB)-p65, inducible (iNOS) and endothelial nitric oxide synthase (eNOS) levels were also assayed by WB. MAIN OUTCOME MEASURES The main outcome measure was to characterize the expression of SIRT1-3 and SIRT5-7 in human CC. RESULTS SIRT1-3 and SIRT5-7 mRNAs were detected in all individuals, without statistical differences among groups, excepting SIRT7 that decreased four times in aged groups relatively to young (P = 0.013). WB analysis demonstrated that aged individuals with CVDRF presented higher levels of SIRT7 protein relatively to young (P = 0.0495) and lower levels of SIRT3 protein relatively to healthy aged (P = 0.0077). Expression of NFkB-p65 and iNOS were higher in aged than in young individuals (P = 0.0185; P = 0.004, respectively). No differences in other sirtuins or total eNOS were seen among groups although phospho eNOS Ser(1177) levels decreased in groups of aged men relatively to young (P = 0.0043; P = 0.0099). CONCLUSIONS Our results demonstrate for the first time expression of SIRT2-3 and SIRT5-7 in the human CC. Aged individuals with CVDRF presented an increase in SIRT7 protein levels and a decrease in mitochondrial SIRT3. This finding suggests that CVDRF induces the loss of antioxidant defense mechanisms leading to endothelial injury.
Collapse
Affiliation(s)
- Margarida Freitas
- Department of Experimental Biology, Faculty of Medicine of Universidade do Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Institute for Molecular and Cell Biology (IBMC) of Universidade do Porto, Porto, Portugal
| | - Adriana R Rodrigues
- Department of Experimental Biology, Faculty of Medicine of Universidade do Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Institute for Molecular and Cell Biology (IBMC) of Universidade do Porto, Porto, Portugal
| | - Nuno Tomada
- Department of Urology, Central Hospital of S. João, Porto, Portugal
| | - João Fonseca
- Department of Experimental Biology, Faculty of Medicine of Universidade do Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Institute for Molecular and Cell Biology (IBMC) of Universidade do Porto, Porto, Portugal
| | - Alexandre Magalhães
- REQUIMTE/Department of Chemistry & Biochemistry, Faculty of Sciences of Universidade do Porto, Porto, Portugal
| | - Alexandra M Gouveia
- Department of Experimental Biology, Faculty of Medicine of Universidade do Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Institute for Molecular and Cell Biology (IBMC) of Universidade do Porto, Porto, Portugal.,Faculty of Nutrition and Food Sciences, Universidade do Porto, Porto, Portugal
| | - Delminda Neves
- Department of Experimental Biology, Faculty of Medicine of Universidade do Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Institute for Molecular and Cell Biology (IBMC) of Universidade do Porto, Porto, Portugal
| |
Collapse
|
37
|
Siragusa M, Fröhlich F, Park EJ, Schleicher M, Walther TC, Sessa WC. Stromal cell-derived factor 2 is critical for Hsp90-dependent eNOS activation. Sci Signal 2015; 8:ra81. [PMID: 26286023 DOI: 10.1126/scisignal.aaa2819] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Endothelial nitric oxide synthase (eNOS) catalyzes the conversion of l-arginine and molecular oxygen into l-citrulline and nitric oxide (NO), a gaseous second messenger that influences cardiovascular physiology and disease. Several mechanisms regulate eNOS activity and function, including phosphorylation at Ser and Thr residues and protein-protein interactions. Combining a tandem affinity purification approach and mass spectrometry, we identified stromal cell-derived factor 2 (SDF2) as a component of the eNOS macromolecular complex in endothelial cells. SDF2 knockdown impaired agonist-stimulated NO synthesis and decreased the phosphorylation of eNOS at Ser(1177), a key event required for maximal activation of eNOS. Conversely, SDF2 overexpression dose-dependently increased NO synthesis through a mechanism involving Akt and calcium (induced with ionomycin), which increased the phosphorylation of Ser(1177) in eNOS. NO synthesis by iNOS (inducible NOS) and nNOS (neuronal NOS) was also enhanced upon SDF2 overexpression. We found that SDF2 was a client protein of the chaperone protein Hsp90, interacting preferentially with the M domain of Hsp90, which is the same domain that binds to eNOS. In endothelial cells exposed to vascular endothelial growth factor (VEGF), SDF2 was required for the binding of Hsp90 and calmodulin to eNOS, resulting in eNOS phosphorylation and activation. Thus, our data describe a function for SDF2 as a component of the Hsp90-eNOS complex that is critical for signal transduction in endothelial cells.
Collapse
Affiliation(s)
- Mauro Siragusa
- Vascular Biology and Therapeutics Program, Department of Pharmacology, Yale University School of Medicine, 10 Amistad Street, New Haven, CT 06520, USA
| | - Florian Fröhlich
- Department of Genetics and Complex Diseases, Harvard School of Public Health, 677 Huntington Avenue, Boston, MA 02115, USA
| | - Eon Joo Park
- Vascular Biology and Therapeutics Program, Department of Pharmacology, Yale University School of Medicine, 10 Amistad Street, New Haven, CT 06520, USA
| | - Michael Schleicher
- Vascular Biology and Therapeutics Program, Department of Pharmacology, Yale University School of Medicine, 10 Amistad Street, New Haven, CT 06520, USA
| | - Tobias C Walther
- Department of Genetics and Complex Diseases, Harvard School of Public Health, 677 Huntington Avenue, Boston, MA 02115, USA
| | - William C Sessa
- Vascular Biology and Therapeutics Program, Department of Pharmacology, Yale University School of Medicine, 10 Amistad Street, New Haven, CT 06520, USA.
| |
Collapse
|
38
|
Lin Y, Jiang W, Ng J, Jina A, Wang RA. Endothelial ephrin-B2 is essential for arterial vasodilation in mice. Microcirculation 2015; 21:578-86. [PMID: 24673722 DOI: 10.1111/micc.12135] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2014] [Accepted: 03/24/2014] [Indexed: 12/22/2022]
Abstract
OBJECTIVE The cell surface protein ephrin-B2 is expressed in arterial and not venous ECs throughout development and adulthood. Endothelial ephrin-B2 is required for vascular development and angiogenesis, but its role in established arteries is currently unknown. We investigated the physiological role of ephrin-B2 signaling in adult endothelium. METHODS We generated adult conditional knockout mice lacking the Efnb2 gene specifically in ECs and evaluated the vasodilation responses to blood flow increase and ACh in the cremaster muscle preparation by intravital microscope and in carotid artery by in vivo ultrasound. RESULTS We found that the Efnb2 conditional knockout mice were defective in acute arterial dilation. Vasodilation was impaired in cremaster arterioles in response to either increased flow or ACh, and in the carotid arteries in response to increased flow. Levels of cGMP, an effector of NO, were diminished in mutant arteries following ACh stimulation. GSNO, a donor for the vasodilator NO, alleviated the vasodilatory defects in the mutants. Immunostaining showed that a subset of ephrin-B2 proteins colocalized with caveolin-1, a negative regulator of eNOS. CONCLUSIONS Our data suggest that endothelial ephrin-B2 is required for endothelial-dependent arterial dilation and NO signaling in adult endothelium.
Collapse
Affiliation(s)
- Yuankai Lin
- Laboratory for Accelerated Vascular Research, Division of Vascular Surgery, Department of Surgery, University of California, San Francisco, California, USA
| | | | | | | | | |
Collapse
|
39
|
Mutchler SM, Straub AC. Compartmentalized nitric oxide signaling in the resistance vasculature. Nitric Oxide 2015; 49:8-15. [PMID: 26028569 DOI: 10.1016/j.niox.2015.05.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 05/15/2015] [Accepted: 05/20/2015] [Indexed: 01/23/2023]
Abstract
Nitric oxide (NO) was first described as a bioactive molecule through its ability to stimulate soluble guanylate cyclase, but the revelation that NO was the endothelium derived relaxation factor drove the field to its modern state. The wealth of research conducted over the past 30 years has provided us with a picture of how diverse NO signaling can be within the vascular wall, going beyond simple vasodilation to include such roles as signaling through protein S-nitrosation. This expanded view of NO's actions requires highly regulated and compartmentalized production. Importantly, resistance arteries house multiple proteins involved in the production and transduction of NO allowing for efficient movement of the molecule to regulate vascular tone and reactivity. In this review, we focus on the many mechanisms regulating NO production and signaling action in the vascular wall, with a focus on the control of endothelial nitric oxide synthase (eNOS), the enzyme responsible for synthesizing most of the NO within these confines. We also explore how cross talk between the endothelium and smooth muscle in the microcirculation can modulate NO signaling, illustrating that this one small molecule has the capability to produce a plethora of responses.
Collapse
Affiliation(s)
- Stephanie M Mutchler
- Heart, Lung, Blood and Vascular Medicine Institute, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15216, USA
| | - Adam C Straub
- Heart, Lung, Blood and Vascular Medicine Institute, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15216, USA.
| |
Collapse
|
40
|
Konradi J, Mollenhauer M, Baldus S, Klinke A. Redox-sensitive mechanisms underlying vascular dysfunction in heart failure. Free Radic Res 2015; 49:721-42. [DOI: 10.3109/10715762.2015.1027200] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
41
|
Craps J, Wilvers C, Joris V, De Jongh B, Vanderstraeten J, Lobysheva I, Balligand JL, Sonveaux P, Gilon P, Many MC, Gérard AC, Colin IM. Involvement of nitric oxide in iodine deficiency-induced microvascular remodeling in the thyroid gland: role of nitric oxide synthase 3 and ryanodine receptors. Endocrinology 2015; 156:707-20. [PMID: 25406019 DOI: 10.1210/en.2014-1729] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Iodine deficiency (ID) induces microvascular changes in the thyroid gland via a TSH-independent reactive oxygen species-hypoxia inducible factor (HIF)-1α-vascular endothelial growth factor (VEGF) pathway. The involvement of nitric oxide (NO) in this pathway and the role of calcium (Ca(2+)) and of ryanodine receptors (RYRs) in NO synthase 3 (NOS3) activation were investigated in a murine model of goitrogenesis and in 3 in vitro models of ID, including primary cultures of human thyrocytes. ID activated NOS3 and the production of NO in thyrocytes in vitro and increased the thyroid blood flow in vivo. Using bevacizumab (a blocking antibody against VEGF-A) in mice, it appeared that NOS3 is activated upstream of VEGF-A. L-nitroarginine methyl ester (a NOS inhibitor) blocked the ID-induced increase in thyroid blood flow in vivo and NO production in vitro, as well as ID-induced VEGF-A mRNA and HIF-1α expression in vitro, whereas S-nitroso-acetyl-penicillamine (a NO donor) did the opposite. Ca(2+) is involved in this pathway as intracellular Ca(2+) flux increased after ID, and thapsigargin activated NOS3 and increased VEGF-A mRNA expression. Two of the 3 known mammalian RYR isoforms (RYR1 and RYR2) were shown to be expressed in thyrocytes. RYR inhibition using ryanodine at 10μM decreased ID-induced NOS3 activation, HIF-1α, and VEGF-A expression, whereas RYR activation with ryanodine at 1nM increased NOS3 activation and VEGF-A mRNA expression. In conclusion, during the early phase of TSH-independent ID-induced microvascular activation, ID sequentially activates RYRs and NOS3, thereby supporting ID-induced activation of the NO/HIF-1α/VEGF-A pathway in thyrocytes.
Collapse
Affiliation(s)
- J Craps
- Pôle de Morphologie (J.C., C.W., B.D.J., J.V., M.-C.M., I.M.C.), de Pharmacologie et Thérapeutique (V.J., I.L., J.-L.B., P.S.), et d'Endocrinologie, Diabète et Nutrition (P.G.), Institut de Recherche Expérimentale et Clinique, Secteur des Sciences de la Santé, Faculté de Médecine, Université catholique de Louvain, 1200, Brussels, Belgium; and Service d'Endocrino-Diabétologie (A.-C.G., I.M.C.), Centre Hospitalier Régional, 7000, Mons-Hainaut, Belgium
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Bancroft T, Bouaouina M, Roberts S, Lee M, Calderwood DA, Schwartz M, Simons M, Sessa WC, Kyriakides TR. Up-regulation of thrombospondin-2 in Akt1-null mice contributes to compromised tissue repair due to abnormalities in fibroblast function. J Biol Chem 2014; 290:409-22. [PMID: 25389299 DOI: 10.1074/jbc.m114.618421] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Vascular remodeling is essential for tissue repair and is regulated by multiple factors, including thrombospondin-2 (TSP2) and hypoxia/VEGF-induced activation of Akt. In contrast to TSP2 knock-out (KO) mice, Akt1 KO mice have elevated TSP2 expression and delayed tissue repair. To investigate the contribution of increased TSP2 to Akt1 KO mice phenotypes, we generated Akt1/TSP2 double KO (DKO) mice. Full-thickness excisional wounds in DKO mice healed at an accelerated rate when compared with Akt1 KO mice. Isolated dermal Akt1 KO fibroblasts expressed increased TSP2 and displayed altered morphology and defects in migration and adhesion. These defects were rescued in DKO fibroblasts or after TSP2 knockdown. Conversely, the addition of exogenous TSP2 to WT cells induced cell morphology and migration rates that were similar to those of Akt1 KO cells. Akt1 KO fibroblasts displayed reduced adhesion to fibronectin with manganese stimulation when compared with WT and DKO cells, revealing an Akt1-dependent role for TSP2 in regulating integrin-mediated adhesions; however, this effect was not due to changes in β1 integrin surface expression or activation. Consistent with these results, Akt1 KO fibroblasts displayed reduced Rac1 activation that was dependent upon expression of TSP2 and could be rescued by a constitutively active Rac mutant. Our observations show that repression of TSP2 expression is a critical aspect of Akt1 function in tissue repair.
Collapse
Affiliation(s)
- Tara Bancroft
- From the Departments of Pathology, the Program of Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, Connecticut 06520
| | | | - Sophia Roberts
- the Program of Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Monica Lee
- the Program of Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, Connecticut 06520 Pharmacology
| | - David A Calderwood
- the Program of Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, Connecticut 06520 Cell Biology, Pharmacology
| | - Martin Schwartz
- the Program of Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, Connecticut 06520 Cardiology, and
| | - Michael Simons
- the Program of Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, Connecticut 06520 Cardiology, and
| | - William C Sessa
- the Program of Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, Connecticut 06520 Pharmacology
| | - Themis R Kyriakides
- From the Departments of Pathology, the Program of Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, Connecticut 06520 Biomedical Engineering and
| |
Collapse
|
43
|
Endothelial Akt1 mediates angiogenesis by phosphorylating multiple angiogenic substrates. Proc Natl Acad Sci U S A 2014; 111:12865-70. [PMID: 25136137 DOI: 10.1073/pnas.1408472111] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The PI3K/Akt pathway is necessary for several key endothelial cell (EC) functions, including cell growth, migration, survival, and vascular tone. However, existing literature supports the idea that Akt can be either pro- or antiangiogenic, possibly due to compensation by multiple isoforms in the EC when a single isoform is deleted. Thus, biochemical, genetic, and proteomic studies were conducted to examine isoform-substrate specificity for Akt1 vs. Akt2. In vitro, Akt1 preferentially phosphorylates endothelial nitric oxide synthase (eNOS) and promotes NO release, whereas nonphysiological overexpression of Akt2 can bypass the loss of Akt1. Conditional deletion of Akt1 in the EC, in the absence or presence of Akt2, retards retinal angiogenesis, implying that Akt1 exerts a nonredundant function during physiological angiogenesis. Finally, proteomic analysis of Akt substrates isolated from Akt1- or Akt2-deficient ECs documents that phosphorylation of multiple Akt substrates regulating angiogenic signaling is reduced in Akt1-deficient, but not Akt2-deficient, ECs, including eNOS and Forkhead box proteins. Therefore, Akt1 promotes angiogenesis largely due to phosphorylation and regulation of important downstream effectors that promote aspects of angiogenic signaling.
Collapse
|
44
|
Aicart-Ramos C, Sánchez-Ruiloba L, Gómez-Parrizas M, Zaragoza C, Iglesias T, Rodríguez-Crespo I. Protein kinase D activity controls endothelial nitric oxide synthesis. J Cell Sci 2014; 127:3360-72. [PMID: 24928905 DOI: 10.1242/jcs.148601] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) regulates key functions of the endothelium, such as angiogenesis or vessel repair in processes involving endothelial nitric oxide synthase (eNOS) activation. One of the effector kinases that become activated in endothelial cells upon VEGF treatment is protein kinase D (PKD). Here, we show that PKD phosphorylates eNOS, leading to its activation and a concomitant increase in NO synthesis. Using mass spectrometry, we show that the purified active kinase specifically phosphorylates recombinant eNOS on Ser1179. Treatment of endothelial cells with VEGF or phorbol 12,13-dibutyrate (PDBu) activates PKD and increases eNOS Ser1179 phosphorylation. In addition, pharmacological inhibition of PKD and gene silencing of both PKD1 and PKD2 abrogate VEGF signaling, resulting in a clear diminished migration of endothelial cells in a wound healing assay. Finally, inhibition of PKD in mice results in an almost complete disappearance of the VEGF-induced vasodilatation, as monitored through determination of the diameter of the carotid artery. Hence, our data indicate that PKD is a new regulatory kinase of eNOS in endothelial cells whose activity orchestrates mammalian vascular tone.
Collapse
Affiliation(s)
- Clara Aicart-Ramos
- Departamento de Bioquímica y Biología Molecular I, Universidad Complutense de Madrid, Madrid 28040, Spain
| | - Lucía Sánchez-Ruiloba
- Instituto de Investigaciones Biomédicas "Alberto Sols". CSIC-UAM, C/Arturo Duperier, Madrid 28029, Spain CIBERNED, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | | | - Carlos Zaragoza
- Cardiovascular Research Unit University Francisco de Vitoria/Hospital Ramón y Cajal, Ctra Colmenar Viejo Km 9,100, Madrid 28034, Spain
| | - Teresa Iglesias
- Instituto de Investigaciones Biomédicas "Alberto Sols". CSIC-UAM, C/Arturo Duperier, Madrid 28029, Spain CIBERNED, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Ignacio Rodríguez-Crespo
- Departamento de Bioquímica y Biología Molecular I, Universidad Complutense de Madrid, Madrid 28040, Spain
| |
Collapse
|
45
|
Kalyanaraman H, Schwappacher R, Joshua J, Zhuang S, Scott BT, Klos M, Casteel DE, Frangos JA, Dillmann W, Boss GR, Pilz RB. Nongenomic thyroid hormone signaling occurs through a plasma membrane-localized receptor. Sci Signal 2014; 7:ra48. [PMID: 24847117 PMCID: PMC6504257 DOI: 10.1126/scisignal.2004911] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Thyroid hormone (TH) is essential for vertebrate development and the homeostasis of most adult tissues, including bone. TH stimulates target gene expression through the nuclear thyroid receptors TRα and TRβ; however, TH also has rapid, transcription-independent (nongenomic) effects. We found a previously uncharacterized plasma membrane-bound receptor that was necessary and sufficient for nongenomic TH signaling in several cell types. We determined that this receptor is generated by translation initiation from an internal methionine of TRα, which produces a transcriptionally incompetent protein that is palmitoylated and associates with caveolin-containing plasma membrane domains. TH signaling through this receptor stimulated a pro-proliferative and pro-survival program by increasing the intracellular concentrations of calcium, nitric oxide (NO), and cyclic guanosine monophosphate (cGMP), which led to the sequential activation of protein kinase G II (PKGII), the tyrosine kinase Src, and extracellular signal-regulated kinase (ERK) and Akt signaling. Hypothyroid mice exhibited a cGMP-deficient state with impaired bone formation and increased apoptosis of osteocytes, which was rescued by a direct stimulator of guanylate cyclase. Our results link nongenomic TH signaling to a previously uncharacterized membrane-bound receptor, and identify NO synthase, guanylate cyclase, and PKGII as TH effectors that activate kinase cascades to regulate cell survival and proliferation.
Collapse
Affiliation(s)
- Hema Kalyanaraman
- Department of Medicine, University of California, San Diego, San Diego, CA 92093, USA
| | - Raphaela Schwappacher
- Department of Medicine, University of California, San Diego, San Diego, CA 92093, USA
| | - Jisha Joshua
- Department of Medicine, University of California, San Diego, San Diego, CA 92093, USA
| | - Shunhui Zhuang
- Department of Medicine, University of California, San Diego, San Diego, CA 92093, USA
| | - Brian T Scott
- Department of Medicine, University of California, San Diego, San Diego, CA 92093, USA
| | - Matthew Klos
- Department of Surgery, University of California, San Diego, San Diego, CA 92093, USA
| | - Darren E Casteel
- Department of Medicine, University of California, San Diego, San Diego, CA 92093, USA
| | - John A Frangos
- La Jolla Bioengineering Institute, La Jolla, CA 92121, USA
| | - Wolfgang Dillmann
- Department of Medicine, University of California, San Diego, San Diego, CA 92093, USA
| | - Gerry R Boss
- Department of Medicine, University of California, San Diego, San Diego, CA 92093, USA
| | - Renate B Pilz
- Department of Medicine, University of California, San Diego, San Diego, CA 92093, USA.
| |
Collapse
|
46
|
Tomada I, Negrão R, Almeida H, Neves D. Long-term high-fat consumption leads to downregulation of Akt phosphorylation of eNOS at Ser1177 and upregulation of Sirtuin-1 expression in rat cavernous tissue. AGE (DORDRECHT, NETHERLANDS) 2014; 36:597-611. [PMID: 24105250 PMCID: PMC4039277 DOI: 10.1007/s11357-013-9591-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2013] [Accepted: 09/26/2013] [Indexed: 06/02/2023]
Abstract
Long-term consumption of high-fat diets negatively interferes with metabolic status and promotes endothelial dysfunction and inflammation. In the cavernous tissue, these outcomes become conspicuous in the elderly and strongly affect penile erection, a vascular process highly dependent on local nitric oxide bioavailability. Although epidemiological data links erectile dysfunction to nutritional patterns, the underlying molecular mechanisms remain unclear. Therefore, we investigated the effects of long-term high-fat diet on endothelial nitric oxide synthase (eNOS)-Sirtuin-1 axis and Akt/eNOS phosphorylation in the cavernous tissue of Sprague-Dawley rats, and compared with energy-restricted animals. We demonstrated that high-fat diet intake led to a noteworthy decrease in eNOS phosphorylation at Ser1177 residue through the Akt pathway, which seems to be compensated by upregulation of phosphorylation at Ser615, but without an increment in nitric oxide production. These results are accompanied by an increase of systemic inflammatory markers and upregulation of the inducible NOS and of the deacetylase Sirtuin-1 in the cavernous tissue to levels apparently detrimental to cells and to metabolic homeostasis. Conversely, in long-term energy-restricted animals, the rate of phosphorylation of eNOS at Ser1177 diminished, but the activation of the enzyme increased through phosphorylation of eNOS at Ser615, resulting in an enhancement in nitric oxide bioavailability. Taken together, our results demonstrate that long-term nutritional conditions override the influence of age on the eNOS expression and activation in rat cavernous tissue.
Collapse
Affiliation(s)
- I Tomada
- Department of Experimental Biology of Faculty of Medicine, Universidade do Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal,
| | | | | | | |
Collapse
|
47
|
García C, Nuñez-Anita RE, Thebault S, Arredondo Zamarripa D, Jeziorsky MC, Martínez de la Escalera G, Clapp C. Requirement of phosphorylatable endothelial nitric oxide synthase at Ser-1177 for vasoinhibin-mediated inhibition of endothelial cell migration and proliferation in vitro. Endocrine 2014; 45:263-70. [PMID: 23640371 DOI: 10.1007/s12020-013-9964-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 04/16/2013] [Indexed: 12/23/2022]
Abstract
Endothelial nitric oxide synthase (eNOS)-derived nitric oxide is a major vasorelaxing factor and a mediator of vasopermeability and angiogenesis. Vasoinhibins, a family of antiangiogenic prolactin fragments that include 16 K prolactin, block most eNOS-mediated vascular effects. Vasoinhibins activate protein phosphatase 2A, causing eNOS inactivation through dephosphorylation of eNOS at serine residue 1179 in bovine endothelial cells and thereby blocking vascular permeability. In this study, we examined whether human eNOS phosphorylation at S1177 (analogous to bovine S1179) influences other actions of vasoinhibins. Bovine umbilical vein endothelial cells were stably transfected with human wild-type eNOS (WT) or with phospho-mimetic (S1177D) or non-phosphorylatable (S1177A) eNOS mutants. Vasoinhibins inhibited the increases in eNOS activity, migration, and proliferation following the overexpression of WT eNOS but did not affect these responses in cells expressing S1177D and S1177A eNOS mutants. We conclude that eNOS inhibition by dephosphorylation of S1177 is fundamental for the inhibition of endothelial cell migration and proliferation by vasoinhibins.
Collapse
Affiliation(s)
- Celina García
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus UNAM-Juriquilla, 76230, Querétaro, QRO, Mexico
| | | | | | | | | | | | | |
Collapse
|
48
|
Pong T, Scherrer-Crosbie M, Atochin DN, Bloch KD, Huang PL. Phosphomimetic modulation of eNOS improves myocardial reperfusion and mimics cardiac postconditioning in mice. PLoS One 2014; 9:e85946. [PMID: 24465805 PMCID: PMC3897570 DOI: 10.1371/journal.pone.0085946] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 12/04/2013] [Indexed: 12/16/2022] Open
Abstract
Objective Myocardial infarction resulting from ischemia-reperfusion injury can be reduced by cardiac postconditioning, in which blood flow is restored intermittently prior to full reperfusion. Although key molecular mechanisms and prosurvival pathways involved in postconditioning have been identified, a direct role for eNOS-derived NO in improving regional myocardial perfusion has not been shown. The objective of this study is to measure, with high temporal and spatial resolution, regional myocardial perfusion during ischemia-reperfusion and postconditioning, in order to determine the contribution of regional blood flow effects of NO to infarct size and protection. Methods and Results We used myocardial contrast echocardiography to measure regional myocardial blood flow in mice over time. Reperfusion after myocardial ischemia-reperfusion injury is improved by postconditioning, as well as by phosphomimetic eNOS modulation. Knock-in mice expressing a phosphomimetic S1176D form of eNOS showed improved myocardial reperfusion and significantly reduced infarct size. eNOS knock-out mice failed to show cardioprotection from postconditioning. The size of the no-reflow zone following ischemia-reperfusion is substantially reduced by postconditioning and by the phosphomimetic eNOS mutation. Conclusions and Significance Using myocardial contrast echocardiography, we show that temporal dynamics of regional myocardial perfusion restoration contribute to reduced infarct size after postconditioning. eNOS has direct effects on myocardial blood flow following ischemia-reperfusion, with reduction in the size of the no-reflow zone. These results have important implications for ongoing clinical trials on cardioprotection, because the degree of protective benefit may be significantly influenced by the regional hemodynamic effects of eNOS-derived NO.
Collapse
Affiliation(s)
- Terrence Pong
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, United States of America
- Harvard-MIT Division of Health Sciences & Technology, Cambridge, Massachusetts, United States of America
| | - Marielle Scherrer-Crosbie
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Cardiac Ultrasound Laboratory, Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Dmitriy N. Atochin
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Kenneth D. Bloch
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Paul L. Huang
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, United States of America
- Harvard-MIT Division of Health Sciences & Technology, Cambridge, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
49
|
Qin C, Zhou S, Xiao Y, Chen L. Erythropoietin enhances mitochondrial biogenesis in cardiomyocytes exposed to chronic hypoxia through Akt/eNOS signalling pathway. Cell Biol Int 2014; 38:335-42. [PMID: 24436050 DOI: 10.1002/cbin.10205] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 10/27/2013] [Indexed: 12/15/2022]
Abstract
Adaptation of cardiomyocytes to chronic hypoxia in cyanotic patients remains unclear. Mitochondrial biogenesis is enhanced in myocardium from cyanotic patients, which is possibly an adaptive response. Erythropoietin (EPO) in blood and its receptor (EPOR) on cardiomyocytes are upregulated by chronic hypoxia, suggesting that EPO-EPOR interaction is increased, which is inferred to positively regulate mitochondrial biogenesis through protein kinase B (Akt)/endothelial nitric oxide synthase (eNOS) signalling pathway. H9c2 cardiomyocytes were exposed to hypoxia (1% O(2)) for 1 week and treated with different doses of recombinant human erythropoietin (rhEPO). Mitochondrial number, mitochondrial DNA (mtDNA) copy number and peroxisome proliferator activated receptor gamma coactivator alpha (PGC-1α) mRNA expression increased in a dose-dependent manner induced by rhEPO. Akt and eNOS were significantly phosphorylated by rhEPO. Both blocking Akt with Wortmannin and silencing eNOS expression with shRNA plasmid decreased the mtDNA copy number and PGC-1α mRNA expression induced by rhEPO. Blocking Akt was associated with the decreased phosphorylation of Akt and eNOS. RNA interference led to a reduction in the total and phosphorylated proteins of eNOS. Thus EPO enhances mitochondrial biogenesis in cardiomyocytes exposed to chronic hypoxia, at least partly through Akt/eNOS signalling, which might be an adaptive mechanism of cardiomyocytes associated with the increased EPO-EPOR interaction in patients with cyanotic congenital heart disease (CCHD).
Collapse
Affiliation(s)
- Chuan Qin
- Department of Cardiovascular Surgery, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | | | | | | |
Collapse
|
50
|
Ju R, Zhuang ZW, Zhang J, Lanahan AA, Kyriakides T, Sessa WC, Simons M. Angiopoietin-2 secretion by endothelial cell exosomes: regulation by the phosphatidylinositol 3-kinase (PI3K)/Akt/endothelial nitric oxide synthase (eNOS) and syndecan-4/syntenin pathways. J Biol Chem 2013; 289:510-9. [PMID: 24235146 DOI: 10.1074/jbc.m113.506899] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Angiopoietin-2 (Ang2) is an extracellular protein and one of the principal ligands of Tie2 receptor that is involved in the regulation of vascular integrity, quiescence, and inflammation. The mode of secretion of Ang2 has never been established, however. Here, we provide evidence that Ang2 is secreted from endothelial cells via exosomes and that this process is inhibited by the PI3K/Akt/endothelial nitric oxide synthase (eNOS) signaling pathway, whereas it is positively regulated by the syndecan-4/syntenin pathway. Vascular defects in Akt1 null mice arise, in part, because of excessive Ang2 secretion and can be rescued by the syndecan-4 knock-out that reduces extracellular Ang2 levels. This novel mechanism connects three critical signaling pathways: angiopoietin/Tie2, PI3K/Akt/eNOS, and syndecan/syntenin, which play important roles in vascular growth and stabilization.
Collapse
Affiliation(s)
- Rong Ju
- From the Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine
| | | | | | | | | | | | | |
Collapse
|