1
|
Trinh VH, Nguyen Huu T, Sah DK, Choi JM, Yoon HJ, Park SC, Jung YS, Lee SR. Redox Regulation of PTEN by Reactive Oxygen Species: Its Role in Physiological Processes. Antioxidants (Basel) 2024; 13:199. [PMID: 38397797 PMCID: PMC10886030 DOI: 10.3390/antiox13020199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 01/25/2024] [Accepted: 01/27/2024] [Indexed: 02/25/2024] Open
Abstract
Phosphatase and tensin homolog (PTEN) is a tumor suppressor due to its ability to regulate cell survival, growth, and proliferation by downregulating the PI3K/AKT signaling pathway. In addition, PTEN plays an essential role in other physiological events associated with cell growth demands, such as ischemia-reperfusion, nerve injury, and immune responsiveness. Therefore, recently, PTEN inhibition has emerged as a potential therapeutic intervention in these situations. Increasing evidence demonstrates that reactive oxygen species (ROS), especially hydrogen peroxide (H2O2), are produced and required for the signaling in many important cellular processes under such physiological conditions. ROS have been shown to oxidize PTEN at the cysteine residue of its active site, consequently inhibiting its function. Herein, we provide an overview of studies that highlight the role of the oxidative inhibition of PTEN in physiological processes.
Collapse
Affiliation(s)
- Vu Hoang Trinh
- Department of Biochemistry, Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju 501190, Republic of Korea; (V.H.T.); (T.N.H.); (D.K.S.); (J.M.C.); (H.J.Y.)
- Department of Oncology, Department of Medical Sciences, Pham Ngoc Thach University of Medicine, Ho Chi Minh City 700000, Vietnam
| | - Thang Nguyen Huu
- Department of Biochemistry, Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju 501190, Republic of Korea; (V.H.T.); (T.N.H.); (D.K.S.); (J.M.C.); (H.J.Y.)
| | - Dhiraj Kumar Sah
- Department of Biochemistry, Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju 501190, Republic of Korea; (V.H.T.); (T.N.H.); (D.K.S.); (J.M.C.); (H.J.Y.)
| | - Jin Myung Choi
- Department of Biochemistry, Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju 501190, Republic of Korea; (V.H.T.); (T.N.H.); (D.K.S.); (J.M.C.); (H.J.Y.)
| | - Hyun Joong Yoon
- Department of Biochemistry, Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju 501190, Republic of Korea; (V.H.T.); (T.N.H.); (D.K.S.); (J.M.C.); (H.J.Y.)
| | - Sang Chul Park
- The Future Life & Society Research Center, Advanced Institute of Aging Science, Chonnam National University, Gwangju 61469, Republic of Korea;
| | - Yu Seok Jung
- Chonnam National University Medical School, Gwangju 501190, Republic of Korea;
| | - Seung-Rock Lee
- Department of Biochemistry, Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju 501190, Republic of Korea; (V.H.T.); (T.N.H.); (D.K.S.); (J.M.C.); (H.J.Y.)
| |
Collapse
|
2
|
Liu Q, Dong Y, Escames G, Wu X, Ren J, Yang W, Zhang S, Zhu Y, Tian Y, Acuña‐Castroviejo D, Yang Y. Identification of PIK3CG as a hub in septic myocardial injury using network pharmacology and weighted gene co-expression network analysis. Bioeng Transl Med 2023; 8:e10384. [PMID: 36684068 PMCID: PMC9842026 DOI: 10.1002/btm2.10384] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/13/2022] [Accepted: 07/16/2022] [Indexed: 01/25/2023] Open
Abstract
Sepsis causes multiple organ injuries, among which the heart is one most severely damaged organ. Melatonin (MEL) alleviates septic myocardial injury, although a systematic and comprehensive approach is still lacking to understand the precise protective machinery of MEL. This study aimed to examine the underlying mechanisms of MEL on improvement of septic myocardial injury at a systematic level. This study integrated three analytic modalities including database investigations, RNA-seq analysis, and weighted gene co-expression network analysis (WCGNA), in order to acquire a set of genes associated with the pathogenesis of sepsis. The Drugbank database was employed to predict genes that may serve as pharmacological targets for MEL-elicited benefits, if any. A pharmacological protein-protein interaction network was subsequently constructed, and 66 hub genes were captured which were enriched in a variety of immune response pathways. Notably, PIK3CG, one of the hub genes, displayed high topological characteristic values, strongly suggesting its promise as a novel target for MEL-evoked treatment of septic myocardial injury. Importantly, molecular docking simulation experiments as well as in vitro and in vivo studies supported an essential role for PIK3CG in MEL-elicited effect on septic myocardial injury. This study systematically clarified the mechanisms of MEL intervention in septic myocardial injury involved multiple targets and multiple pathways. Moreover, PIK3CG-governed signaling cascade plays an important role in the etiology of sepsis and septic myocardial injury. Findings from our study provide valuable information on novel intervention targets for the management of septic myocardial injury. More importantly, this study has indicated the utility of combining a series of techniques for disease target discovery and exploration of possible drug targets, which should shed some light on elucidation of experimental and clinical drug action mechanisms systematically.
Collapse
Affiliation(s)
- Qiong Liu
- Key Laboratory of Resource Biology and Biotechnology in Western ChinaMinistry of Education, Faculty of life Science and Medicine, Northwest UniversityXi'anChina
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular DiseasesXi'an No. 3 Hospital, The Affiliated Hospital of Northwest University, Faculty of life Science and Medicine, Northwest UniversityXi'anChina
| | - Yushu Dong
- Institute of Neuroscience, General Hospital of Northern Theater CommandShenyangChina
| | - Germaine Escames
- Biomedical Research Center, Health Sciences Technology ParkUniversity of GranadaGranadaSpain
- Ibs. Granada, CIBERfesGranadaSpain
- UGC of Clinical LaboratoriesUniversitu San Cecilio's HospitalGranadaSpain
| | - Xue Wu
- Key Laboratory of Resource Biology and Biotechnology in Western ChinaMinistry of Education, Faculty of life Science and Medicine, Northwest UniversityXi'anChina
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular DiseasesXi'an No. 3 Hospital, The Affiliated Hospital of Northwest University, Faculty of life Science and Medicine, Northwest UniversityXi'anChina
| | - Jun Ren
- Department of CardiologyZhongshan Hospital, Fudan UniversityShanghaiChina
- Shanghai Institute of Cardiovascular DiseasesShanghaiChina
- Department of Laboratory Medicine and PathologyUniversity of WashingtonSeattleWashingtonUSA
| | - Wenwen Yang
- Key Laboratory of Resource Biology and Biotechnology in Western ChinaMinistry of Education, Faculty of life Science and Medicine, Northwest UniversityXi'anChina
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular DiseasesXi'an No. 3 Hospital, The Affiliated Hospital of Northwest University, Faculty of life Science and Medicine, Northwest UniversityXi'anChina
| | - Shaofei Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western ChinaMinistry of Education, Faculty of life Science and Medicine, Northwest UniversityXi'anChina
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular DiseasesXi'an No. 3 Hospital, The Affiliated Hospital of Northwest University, Faculty of life Science and Medicine, Northwest UniversityXi'anChina
| | - Yanli Zhu
- Key Laboratory of Resource Biology and Biotechnology in Western ChinaMinistry of Education, Faculty of life Science and Medicine, Northwest UniversityXi'anChina
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular DiseasesXi'an No. 3 Hospital, The Affiliated Hospital of Northwest University, Faculty of life Science and Medicine, Northwest UniversityXi'anChina
| | - Ye Tian
- Key Laboratory of Resource Biology and Biotechnology in Western ChinaMinistry of Education, Faculty of life Science and Medicine, Northwest UniversityXi'anChina
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular DiseasesXi'an No. 3 Hospital, The Affiliated Hospital of Northwest University, Faculty of life Science and Medicine, Northwest UniversityXi'anChina
| | - Darío Acuña‐Castroviejo
- Biomedical Research Center, Health Sciences Technology ParkUniversity of GranadaGranadaSpain
- Ibs. Granada, CIBERfesGranadaSpain
- UGC of Clinical LaboratoriesUniversitu San Cecilio's HospitalGranadaSpain
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western ChinaMinistry of Education, Faculty of life Science and Medicine, Northwest UniversityXi'anChina
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular DiseasesXi'an No. 3 Hospital, The Affiliated Hospital of Northwest University, Faculty of life Science and Medicine, Northwest UniversityXi'anChina
| |
Collapse
|
3
|
Hu XQ, Zhang L. Oxidative Regulation of Vascular Ca v1.2 Channels Triggers Vascular Dysfunction in Hypertension-Related Disorders. Antioxidants (Basel) 2022; 11:antiox11122432. [PMID: 36552639 PMCID: PMC9774363 DOI: 10.3390/antiox11122432] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/28/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
Blood pressure is determined by cardiac output and peripheral vascular resistance. The L-type voltage-gated Ca2+ (Cav1.2) channel in small arteries and arterioles plays an essential role in regulating Ca2+ influx, vascular resistance, and blood pressure. Hypertension and preeclampsia are characterized by high blood pressure. In addition, diabetes has a high prevalence of hypertension. The etiology of these disorders remains elusive, involving the complex interplay of environmental and genetic factors. Common to these disorders are oxidative stress and vascular dysfunction. Reactive oxygen species (ROS) derived from NADPH oxidases (NOXs) and mitochondria are primary sources of vascular oxidative stress, whereas dysfunction of the Cav1.2 channel confers increased vascular resistance in hypertension. This review will discuss the importance of ROS derived from NOXs and mitochondria in regulating vascular Cav1.2 and potential roles of ROS-mediated Cav1.2 dysfunction in aberrant vascular function in hypertension, diabetes, and preeclampsia.
Collapse
|
4
|
Lanahan SM, Wymann MP, Lucas CL. The role of PI3Kγ in the immune system: new insights and translational implications. Nat Rev Immunol 2022; 22:687-700. [PMID: 35322259 PMCID: PMC9922156 DOI: 10.1038/s41577-022-00701-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2022] [Indexed: 12/27/2022]
Abstract
Over the past two decades, new insights have positioned phosphoinositide 3-kinase-γ (PI3Kγ) as a context-dependent modulator of immunity and inflammation. Recent advances in protein structure determination and drug development have allowed for generation of highly specific PI3Kγ inhibitors, with the first now in clinical trials for several oncology indications. Recently, a monogenic immune disorder caused by PI3Kγ deficiency was discovered in humans and modelled in mice. Human inactivated PI3Kγ syndrome confirms the immunomodulatory roles of PI3Kγ and strengthens newly defined roles of this molecule in modulating inflammatory cytokine release in macrophages. Here, we review the functions of PI3Kγ in the immune system and discuss how our understanding of its potential as a therapeutic target has evolved.
Collapse
Affiliation(s)
- Stephen M Lanahan
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | | | - Carrie L Lucas
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
5
|
PI3K Isoforms in Vascular Biology, A Focus on the Vascular System-Immune Response Connection. Curr Top Microbiol Immunol 2022; 436:289-309. [DOI: 10.1007/978-3-031-06566-8_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
6
|
Relevance of stromal interaction molecule 1 (STIM1) in experimental and human stroke. Pflugers Arch 2021; 474:141-153. [PMID: 34757454 DOI: 10.1007/s00424-021-02636-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 10/20/2021] [Accepted: 10/28/2021] [Indexed: 10/19/2022]
Abstract
Stroke represents a main cause of death and permanent disability worldwide. In the attempt to develop targeted preventive and therapeutic strategies, several efforts were performed over the last decades to identify the specific molecular abnormalities preceding cerebral ischemia and neuronal death. In this regard, mitochondrial dysfunction, autophagy, and intracellular calcium homeostasis appear important contributors to stroke development, as underscored by recent pre-clinical evidence. Intracellular calcium (Ca2+) homeostasis is regulated, among other mechanisms, by the calcium sensor stromal interaction molecule 1 (STIM1) and calcium release-activated calcium modulator (ORAI) members, which mediate the store-operated Ca2+ entry (SOCE). The activity of SOCE is deregulated in animal models of ischemic stroke, leading to ischemic injury exacerbation. We found a different pattern of expression of few SOCE components, dependent from a STIM1 mutation, in cerebral endothelial cells isolated from the stroke-prone spontaneously hypertensive rat (SHRSP), compared to the stroke-resistant (SHRSR) strain, suggesting a potential involvement of this mechanism into the stroke predisposition of SHRSP. In this article, we discuss the relevant role of STIM1 in experimental stroke, as highlighted by the current literature and by our recent experimental findings, and the available evidence in the human disease. We also provide a glance on future perspectives and clinical implications of STIM1.
Collapse
|
7
|
Forte M, Marchitti S, Cotugno M, Di Nonno F, Stanzione R, Bianchi F, Schirone L, Schiavon S, Vecchio D, Sarto G, Scioli M, Raffa S, Tocci G, Relucenti M, Torrisi MR, Valenti V, Versaci F, Vecchione C, Volpe M, Frati G, Rubattu S, Sciarretta S. Trehalose, a natural disaccharide, reduces stroke occurrence in the stroke-prone spontaneously hypertensive rat. Pharmacol Res 2021; 173:105875. [PMID: 34500062 DOI: 10.1016/j.phrs.2021.105875] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/02/2021] [Accepted: 09/03/2021] [Indexed: 01/18/2023]
Abstract
Cerebrovascular disease, a frequent complication of hypertension, is a major public health issue for which novel therapeutic and preventive approaches are needed. Autophagy activation is emerging as a potential therapeutic and preventive strategy toward stroke. Among usual activators of autophagy, the natural disaccharide trehalose (TRE) has been reported to be beneficial in preclinical models of neurodegenerative diseases, atherosclerosis and myocardial infarction. In this study, we tested for the first time the effects of TRE in the stroke-prone spontaneously hypertensive rat (SHRSP) fed with a high-salt stroke permissive diet (JD). We found that TRE reduced stroke occurrence and renal damage in high salt-fed SHRSP. TRE was also able to decrease systolic blood pressure. Through ex-vivo studies, we assessed the beneficial effect of TRE on the vascular function of high salt-fed SHRSP. At the molecular level, TRE restored brain autophagy and reduced mitochondrial mass, along with the improvement of mitochondrial function. The beneficial effects of TRE were associated with increased nuclear translocation of TFEB, a transcriptional activator of autophagy. Our results suggest that TRE may be considered as a natural compound efficacious for the prevention of hypertension-related target organ damage, with particular regard to stroke and renal damage.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Leonardo Schirone
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy; Department of Internal, Anesthetic and Cardiovascular Clinical Sciences, Sapienza" University of Rome, Roma, Italy
| | - Sonia Schiavon
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Daniele Vecchio
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Gianmarco Sarto
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | | | - Salvatore Raffa
- Department of Clinical and Molecular Medicine, School of Medicine and Psychology, Sapienza University of Rome, Roma, Italy
| | - Giuliano Tocci
- Department of Clinical and Molecular Medicine, School of Medicine and Psychology, Sapienza University of Rome, Roma, Italy
| | - Michela Relucenti
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Section of Human Anatomy, Sapienza University of Rome, Rome, Italy
| | - Maria Rosaria Torrisi
- Department of Clinical and Molecular Medicine, School of Medicine and Psychology, Sapienza University of Rome, Roma, Italy
| | | | | | - Carmine Vecchione
- IRCCS Neuromed, Pozzilli, Italy; Department of Medicine and Surgery, University of Salerno, 84081 Baronissi, Italy
| | - Massimo Volpe
- Department of Clinical and Molecular Medicine, School of Medicine and Psychology, Sapienza University of Rome, Roma, Italy
| | - Giacomo Frati
- IRCCS Neuromed, Pozzilli, Italy; Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Speranza Rubattu
- IRCCS Neuromed, Pozzilli, Italy; Department of Clinical and Molecular Medicine, School of Medicine and Psychology, Sapienza University of Rome, Roma, Italy.
| | - Sebastiano Sciarretta
- IRCCS Neuromed, Pozzilli, Italy; Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy.
| |
Collapse
|
8
|
Tracy EP, Hughes W, Beare JE, Rowe G, Beyer A, LeBlanc AJ. Aging-Induced Impairment of Vascular Function: Mitochondrial Redox Contributions and Physiological/Clinical Implications. Antioxid Redox Signal 2021; 35:974-1015. [PMID: 34314229 PMCID: PMC8905248 DOI: 10.1089/ars.2021.0031] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Significance: The vasculature responds to the respiratory needs of tissue by modulating luminal diameter through smooth muscle constriction or relaxation. Coronary perfusion, diastolic function, and coronary flow reserve are drastically reduced with aging. This loss of blood flow contributes to and exacerbates pathological processes such as angina pectoris, atherosclerosis, and coronary artery and microvascular disease. Recent Advances: Increased attention has recently been given to defining mechanisms behind aging-mediated loss of vascular function and development of therapeutic strategies to restore youthful vascular responsiveness. The ultimate goal aims at providing new avenues for symptom management, reversal of tissue damage, and preventing or delaying of aging-induced vascular damage and dysfunction in the first place. Critical Issues: Our major objective is to describe how aging-associated mitochondrial dysfunction contributes to endothelial and smooth muscle dysfunction via dysregulated reactive oxygen species production, the clinical impact of this phenomenon, and to discuss emerging therapeutic strategies. Pathological changes in regulation of mitochondrial oxidative and nitrosative balance (Section 1) and mitochondrial dynamics of fission/fusion (Section 2) have widespread effects on the mechanisms underlying the ability of the vasculature to relax, leading to hyperconstriction with aging. We will focus on flow-mediated dilation, endothelial hyperpolarizing factors (Sections 3 and 4), and adrenergic receptors (Section 5), as outlined in Figure 1. The clinical implications of these changes on major adverse cardiac events and mortality are described (Section 6). Future Directions: We discuss antioxidative therapeutic strategies currently in development to restore mitochondrial redox homeostasis and subsequently vascular function and evaluate their potential clinical impact (Section 7). Antioxid. Redox Signal. 35, 974-1015.
Collapse
Affiliation(s)
- Evan Paul Tracy
- Department of Physiology, University of Louisville, Louisville, Kentucky, USA
| | - William Hughes
- Department of Medicine and Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Jason E Beare
- Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, USA.,Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, Kentucky, USA
| | - Gabrielle Rowe
- Department of Physiology, University of Louisville, Louisville, Kentucky, USA
| | - Andreas Beyer
- Department of Medicine and Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Amanda Jo LeBlanc
- Department of Physiology, University of Louisville, Louisville, Kentucky, USA.,Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
9
|
Carnevale D, Carnevale L, Perrotta S, Pallante F, Migliaccio A, Iodice D, Perrotta M, Lembo G. Chronic 3D Vascular-Immune Interface Established by Coculturing Pressurized Resistance Arteries and Immune Cells. Hypertension 2021; 78:1648-1661. [PMID: 34565186 PMCID: PMC8516815 DOI: 10.1161/hypertensionaha.121.17447] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Daniela Carnevale
- Department of Molecular Medicine, "Sapienza University" of Rome, Italy (D.C., S.P., M.P., G.L.).,Unit of Neuro and Cardiovascular Pathophysiology, IRCCS Neuromed, Pozzilli (IS), Italy (D.C., L.C., F.P., A.M., D.I., G.L.)
| | - Lorenzo Carnevale
- Unit of Neuro and Cardiovascular Pathophysiology, IRCCS Neuromed, Pozzilli (IS), Italy (D.C., L.C., F.P., A.M., D.I., G.L.)
| | - Sara Perrotta
- Department of Molecular Medicine, "Sapienza University" of Rome, Italy (D.C., S.P., M.P., G.L.)
| | - Fabio Pallante
- Unit of Neuro and Cardiovascular Pathophysiology, IRCCS Neuromed, Pozzilli (IS), Italy (D.C., L.C., F.P., A.M., D.I., G.L.)
| | - Agnese Migliaccio
- Unit of Neuro and Cardiovascular Pathophysiology, IRCCS Neuromed, Pozzilli (IS), Italy (D.C., L.C., F.P., A.M., D.I., G.L.)
| | - Daniele Iodice
- Unit of Neuro and Cardiovascular Pathophysiology, IRCCS Neuromed, Pozzilli (IS), Italy (D.C., L.C., F.P., A.M., D.I., G.L.)
| | - Marialuisa Perrotta
- Department of Molecular Medicine, "Sapienza University" of Rome, Italy (D.C., S.P., M.P., G.L.)
| | - Giuseppe Lembo
- Department of Molecular Medicine, "Sapienza University" of Rome, Italy (D.C., S.P., M.P., G.L.).,Unit of Neuro and Cardiovascular Pathophysiology, IRCCS Neuromed, Pozzilli (IS), Italy (D.C., L.C., F.P., A.M., D.I., G.L.)
| |
Collapse
|
10
|
Proprotein Convertase Subtilisin Kexin Type 9 Inhibitors Reduce Platelet Activation Modulating ox-LDL Pathways. Int J Mol Sci 2021; 22:ijms22137193. [PMID: 34281247 PMCID: PMC8267646 DOI: 10.3390/ijms22137193] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/25/2021] [Accepted: 06/30/2021] [Indexed: 01/02/2023] Open
Abstract
Background: Proprotein convertase subtilisin kexin type 9 inhibitors (PCSK9i) lower LDL-cholesterol and slow atherosclerosis preventing cardiovascular events. While it is known that circulating PCSK9 enhances platelet activation (PA) and that PCSK9i reduce it, the underlying mechanism is not still clarified. Methods: In a multicenter before–after study in 80 heterozygous familial hypercholesterolemia (HeFH) patients on treatment with maximum tolerated statin dose ± ezetimibe, PA, soluble-NOX2-derived peptide (sNOX2-dp), and oxidized-LDL (ox-LDL) were measured before and after six months of PCSK9i treatment. In vitro study investigates the effects of plasma from HeFH patients before and after PCK9i on PA in washed platelets (wPLTs) from healthy subjects. Results: Compared to baseline, PCSK9i reduced the serum levels of LDL-c, ox-LDL, Thromboxane (Tx) B2, sNOX2-dp, and PCSK9 (p < 0.001). The decrease of TxB2 correlates with that of ox-LDL, while ox-LDL reduction correlated with PCSK9 and sNOX2-dp delta. In vitro study demonstrated that wPLTs resuspended in plasma from HeFH after PCSK9i treatment induced lower PA and sNOX2-dp release than those obtained using plasma before PCSK9i treatment. This reduction was vanished by adding ox-LDL. ox-LDL-induced PA was blunted by CD36, LOX1, and NOX2 inhibition. Conclusions: PCSK9i treatment reduces PA modulating NOX2 activity and in turn ox-LDL formation in HeFH patients.
Collapse
|
11
|
Cardiovascular toxicity of PI3Kα inhibitors. Clin Sci (Lond) 2021; 134:2595-2622. [PMID: 33063821 DOI: 10.1042/cs20200302] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/27/2020] [Accepted: 09/30/2020] [Indexed: 02/07/2023]
Abstract
The phosphoinositide 3-kinases (PI3Ks) are a family of intracellular lipid kinases that phosphorylate the 3'-hydroxyl group of inositol membrane lipids, resulting in the production of phosphatidylinositol 3,4,5-trisphosphate from phosphatidylinositol 4,5-bisphosphate. This results in downstream effects, including cell growth, proliferation, and migration. The heart expresses three PI3K class I enzyme isoforms (α, β, and γ), and these enzymes play a role in cardiac cellular survival, myocardial hypertrophy, myocardial contractility, excitation, and mechanotransduction. The PI3K pathway is associated with various disease processes but is particularly important to human cancers since many gain-of-function mutations in this pathway occur in various cancers. Despite the development, testing, and regulatory approval of PI3K inhibitors in recent years, there are still significant challenges when creating and utilizing these drugs, including concerns of adverse effects on the heart. There is a growing body of evidence from preclinical studies revealing that PI3Ks play a crucial cardioprotective role, and thus inhibition of this pathway could lead to cardiac dysfunction, electrical remodeling, vascular damage, and ultimately, cardiovascular disease. This review will focus on PI3Kα, including the mechanisms underlying the adverse cardiovascular effects resulting from PI3Kα inhibition and the potential clinical implications of treating patients with these drugs, such as increased arrhythmia burden, biventricular cardiac dysfunction, and impaired recovery from cardiotoxicity. Recommendations for future directions for preclinical and clinical work are made, highlighting the possible role of PI3Kα inhibition in the progression of cancer-related cachexia and female sex and pre-existing comorbidities as independent risk factors for cardiac abnormalities after cancer treatment.
Collapse
|
12
|
Durrant TN, Hers I. PI3K inhibitors in thrombosis and cardiovascular disease. Clin Transl Med 2020; 9:8. [PMID: 32002690 PMCID: PMC6992830 DOI: 10.1186/s40169-020-0261-6] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 01/13/2020] [Indexed: 12/15/2022] Open
Abstract
Phosphoinositide 3-kinases (PI3Ks) are lipid kinases that regulate important intracellular signalling and vesicle trafficking events via the generation of 3-phosphoinositides. Comprising eight core isoforms across three classes, the PI3K family displays broad expression and function throughout mammalian tissues, and the (patho)physiological roles of these enzymes in the cardiovascular system present the PI3Ks as potential therapeutic targets in settings such as thrombosis, atherosclerosis and heart failure. This review will discuss the PI3K enzymes and their roles in cardiovascular physiology and disease, with a particular focus on platelet function and thrombosis. The current progress and future potential of targeting the PI3K enzymes for therapeutic benefit in cardiovascular disease will be considered, while the challenges of developing drugs against these master cellular regulators will be discussed.
Collapse
Affiliation(s)
- Tom N Durrant
- Department of Chemistry, University of Oxford, Oxford, OX1 3QZ, UK.
| | - Ingeborg Hers
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK.
| |
Collapse
|
13
|
Function, Regulation and Biological Roles of PI3Kγ Variants. Biomolecules 2019; 9:biom9090427. [PMID: 31480354 PMCID: PMC6770443 DOI: 10.3390/biom9090427] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 08/14/2019] [Accepted: 08/15/2019] [Indexed: 12/19/2022] Open
Abstract
Phosphatidylinositide 3-kinase (PI3K) γ is the only class IB PI3K member playing significant roles in the G-protein-dependent regulation of cell signaling in health and disease. Originally found in the immune system, increasing evidence suggest a wide array of functions in the whole organism. PI3Kγ occur as two different heterodimeric variants: PI3Kγ (p87) and PI3Kγ (p101), which share the same p110γ catalytic subunit but differ in their associated non-catalytic subunit. Here we concentrate on specific PI3Kγ features including its regulation and biological functions. In particular, the roles of its non-catalytic subunits serving as the main regulators determining specificity of class IB PI3Kγ enzymes are highlighted.
Collapse
|
14
|
Maffei A, Lembo G, Carnevale D. PI3Kinases in Diabetes Mellitus and Its Related Complications. Int J Mol Sci 2018; 19:ijms19124098. [PMID: 30567315 PMCID: PMC6321267 DOI: 10.3390/ijms19124098] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 12/13/2018] [Accepted: 12/15/2018] [Indexed: 02/07/2023] Open
Abstract
Recent studies have shown that phosphoinositide 3-kinases (PI3Ks) have become the target of many pharmacological treatments, both in clinical trials and in clinical practice. PI3Ks play an important role in glucose regulation, and this suggests their possible involvement in the onset of diabetes mellitus. In this review, we gather our knowledge regarding the effects of PI3K isoforms on glucose regulation in several organs and on the most clinically-relevant complications of diabetes mellitus, such as cardiomyopathy, vasculopathy, nephropathy, and neurological disease. For instance, PI3K α has been proven to be protective against diabetes-induced heart failure, while PI3K γ inhibition is protective against the disease onset. In vessels, PI3K γ can generate oxidative stress, while PI3K β inhibition is anti-thrombotic. Finally, we describe the role of PI3Ks in Alzheimer’s disease and ADHD, discussing the relevance for diabetic patients. Given the high prevalence of diabetes mellitus, the multiple effects here described should be taken into account for the development and validation of drugs acting on PI3Ks.
Collapse
Affiliation(s)
- Angelo Maffei
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, 86077 Pozzilli, Italy.
| | - Giuseppe Lembo
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, 86077 Pozzilli, Italy.
- Department of Molecular Medicine, "Sapienza" University of Rome, 00161 Rome, Italy.
| | - Daniela Carnevale
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, 86077 Pozzilli, Italy.
- Department of Molecular Medicine, "Sapienza" University of Rome, 00161 Rome, Italy.
| |
Collapse
|
15
|
Li Z, Ding Q, Ling LP, Wu Y, Meng DX, Li X, Zhang CQ. Metformin attenuates motility, contraction, and fibrogenic response of hepatic stellate cells in vivo and in vitro by activating AMP-activated protein kinase. World J Gastroenterol 2018; 24:819-832. [PMID: 29467552 PMCID: PMC5807940 DOI: 10.3748/wjg.v24.i7.819] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 12/12/2017] [Accepted: 12/26/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the effect of metformin on activated hepatic stellate cells (HSCs) and the possible signaling pathways involved.
METHODS A fibrotic mouse model was generated by intraperitoneal injection of carbon tetrachloride (CCl4) and subsequent treatment with or without metformin. The level of fibrosis was detected by hematoxylin-eosin staining, Sirius Red staining, and immunohistochemistry. The HSC cell line LX-2 was used for in vitro studies. The effect of metformin on cell proliferation (CCK8 assay), motility (scratch test and Transwell assay), contraction (collagen gel contraction assay), extracellular matrix (ECM) secretion (Western blot), and angiogenesis (ELISA and tube formation assay) was investigated. We also analyzed the possible signaling pathways involved by Western blot analysis.
RESULTS Mice developed marked liver fibrosis after intraperitoneal injection with CCl4 for 6 wk. Metformin decreased the activation of HSCs, reduced the deposition of ECM, and inhibited angiogenesis in CCl4-treated mice. Platelet-derived growth factor (PDGF) promoted the fibrogenic response of HSCs in vitro, while metformin inhibited the activation, proliferation, migration, and contraction of HSCs, and reduced the secretion of ECM. Metformin decreased the expression of vascular endothelial growth factor (VEGF) in HSCs through inhibition of hypoxia inducible factor (HIF)-1α in both PDGF-BB treatment and hypoxic conditions, and it down-regulated VEGF secretion by HSCs and inhibited HSC-based angiogenesis in hypoxic conditions in vitro. The inhibitory effects of metformin on activated HSCs were mediated by inhibiting the Akt/mammalian target of rapamycin (mTOR) and extracellular signal-regulated kinase (ERK) pathways via the activation of adenosine monophosphate-activated protein kinase (AMPK).
CONCLUSION Metformin attenuates the fibrogenic response of HSCs in vivo and in vitro, and may therefore be useful for the treatment of chronic liver diseases.
Collapse
Affiliation(s)
- Zhen Li
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, Shandong Province, China
- Shandong Provincial Engineering and Technological Research Center for Liver Disease Prevention and Control, Jinan 250021, Shandong Province, China
| | - Qian Ding
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, Shandong Province, China
| | - Li-Ping Ling
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, Shandong Province, China
- Shandong Provincial Engineering and Technological Research Center for Liver Disease Prevention and Control, Jinan 250021, Shandong Province, China
| | - Ying Wu
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, Shandong Province, China
- Shandong Provincial Engineering and Technological Research Center for Liver Disease Prevention and Control, Jinan 250021, Shandong Province, China
| | - Dong-Xiao Meng
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, Shandong Province, China
- Shandong Provincial Engineering and Technological Research Center for Liver Disease Prevention and Control, Jinan 250021, Shandong Province, China
| | - Xiao Li
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, Shandong Province, China
- Shandong Provincial Engineering and Technological Research Center for Liver Disease Prevention and Control, Jinan 250021, Shandong Province, China
| | - Chun-Qing Zhang
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, Shandong Province, China
| |
Collapse
|
16
|
Long non-coding RNA MT1DP shunts the cellular defense to cytotoxicity through crosstalk with MT1H and RhoC in cadmium stress. Cell Discov 2018; 4:5. [PMID: 29507753 PMCID: PMC5824791 DOI: 10.1038/s41421-017-0005-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 12/12/2017] [Accepted: 12/12/2017] [Indexed: 12/18/2022] Open
Abstract
Metallothioneins (MTs) are known to protect cells against oxidative stress, especially providing protection against cadmium (Cd) toxicity in hepatocytes. There are various gene variants and pseudogenes for MTs; however, there is little understanding on the functions of those non-coding MT members that are known to be expressed as long non-coding RNAs (lncRNAs) nowadays. Different from most protein-coding MT members, MT1DP was here found that remarkably induced to provoke cytotoxicity in hepatocytes in response to Cd treatment. MT1DP exerted such a pro-apoptotic function in Cd-treated hepatocytes through interacting with two partners: RhoC and MT1H. On one hand, MT1DP interacted with RhoC protein to increase the latter’s stability by preventing lysosome-dependent protein degradation. Therefore, upon Cd stress, MT1DP/RhoC complex was quickly reinforced to activate RhoC-CCN1/2-AKT signaling and potentiate Ca2+ influx, leading to enhanced Cd uptake and elevated Cd toxicity. On the other hand, MT1H, a protein-coding member of the MT family with little known function, was found to quickly respond to Cd exposure along with MT1DP. Mechanistically, MT1H and MT1DP were uncovered to mutually protect each other through a reciprocal ceRNA mechanism, building up a positive feedback loop to enforce MT1DP-conducted signaling upon Cd exposure. Moreover, MT1DP was found to contribute much more to the activation of RhoC-CCN1/2-AKT signaling than MT1H. Considered together, we here unveiled a mystery whether a pseudogene within the MT family, MT1DP, has actual biological functions in regulating Cd-induced cellular defense. Our findings unearthed an important role of pseudogene MT1DP in calibrating the cellular machinery to switch the cellular defense to cytotoxicity through crosslinking an interplay between its two partners, namely MT1H and RhoC, under cadmium stress.
Collapse
|
17
|
Li KC, Yu SH, Zhuge BZ. PIK3CG single nucleotide polymorphisms are associated with poor responsiveness to clopidogrel and increased risk of ischemia in patients with coronary heart disease. Medicine (Baltimore) 2017; 96:e7566. [PMID: 28885323 PMCID: PMC6392743 DOI: 10.1097/md.0000000000007566] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND This study explores the associations between PIK3CG single nucleotide polymorphisms (SNPs, rs1129293 and rs17398575) and patient responsiveness to clopidogrel to evaluate the risks of ischemia in patients with coronary heart disease (CHD). METHODS The study consisted of 513 CHD patients who received clopidogrel as part of antiplatelet therapy, after percutaneous coronary intervention. According to the patient responsiveness to clopidogrel, the subjects were assigned to either clopidogrel-resistant (CR) or clopidogrel-sensitive (CS) groups. CR group was determined by patients' platelet aggregation rate of ≥70% and poor responsiveness to clopidogrel, and CS group by patients' platelet aggregation rates of <70% and good responsiveness to clopidogrel. Polymerase chain reaction using TaqMan probe was employed to detect PIK3CG polymorphism. Haplotype and linkage disequilibrium analyses were performed. Prognosis analysis was performed using the Kaplan-Meier curve. RESULTS Significant difference was found in genotype and rs1129293 and rs17398575 allele frequency between the CR and CS groups. Haplotype analysis indicated that the frequency of TG allele was higher in the CR group compared with the CS group, and the frequency of CA allele was lower in the CR group compared with the CS group. Patients with rs1129293 CT + TT genotype and T allele, rs1129293 AG + GG genotype and G allele exhibited an increased CR risk. Logistic regression analysis determined hypertension history as an independent risk factor for CR. The Kaplan-Meier curve suggests that distribution curve of cumulative probability nonischemic events was different between patients with rs1129293 and rs17398575 alleles. Stable CHD patients with TT genotype of rs1129293 allele and GG genotype of rs17398575 allele showed poorer prognosis compared to those with other genotypes and patients with acute coronary syndromes. CONCLUSION A positive correlation may exist between PIK3CG SNPs (rs1129293 and rs17398575) and patients with poor responsiveness to clopidogrel. These findings show that this factor may contribute to an increased risk of ischemia in patients suffering from CHD.
Collapse
Affiliation(s)
- Ke-Cheng Li
- Department of Clinical Laboratory, People's Hospital of Rongcheng, Rongcheng
| | - Shu-Hong Yu
- Department of Blood Transfusion, Yantai Yuhuangding Hospital, Yantai
| | - Bao-Zhong Zhuge
- Department of Clinical Laboratory, Linyi People's Hospital, Linyi, P.R. China
| |
Collapse
|
18
|
Abstract
Receptor signaling relays on intracellular events amplified by secondary and tertiary messenger molecules. In cardiomyocytes and smooth muscle cells, cyclic AMP (cAMP) and subsequent calcium (Ca2+) fluxes are the best characterized receptor-regulated signaling events. However, most of receptors able to modify contractility and other intracellular responses signal through a variety of other messengers, and whether these signaling events are interconnected has long remained unclear. For example, the PI3K (phosphoinositide 3-kinase) pathway connected to the production of the lipid second messenger PIP3/PtdIns(3,4,5)P3 (phosphatidylinositol (3,4,5)-trisphosphate) is potentially involved in metabolic regulation, activation of hypertrophy, and survival pathways. Recent studies, highlighted in this review, started to interconnect PI3K pathway activation to Ca2+ signaling. This interdependency, by balancing contractility with metabolic control, is crucial for cells of the cardiovascular system and is emerging to play key roles in disease development. Better understanding of the interplay between Ca2+ and PI3K signaling is, thus, expected to provide new ground for therapeutic intervention. This review explores the emerging molecular mechanisms linking Ca2+ and PI3K signaling in health and disease.
Collapse
Affiliation(s)
- Alessandra Ghigo
- From the Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Italy (A.G., M. Li, E.H.); and INSERM U1048, I2MC and Université Toulouse III, France (M. Laffargue)
| | - Muriel Laffargue
- From the Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Italy (A.G., M. Li, E.H.); and INSERM U1048, I2MC and Université Toulouse III, France (M. Laffargue)
| | - Mingchuan Li
- From the Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Italy (A.G., M. Li, E.H.); and INSERM U1048, I2MC and Université Toulouse III, France (M. Laffargue)
| | - Emilio Hirsch
- From the Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Italy (A.G., M. Li, E.H.); and INSERM U1048, I2MC and Université Toulouse III, France (M. Laffargue).
| |
Collapse
|
19
|
Marcoux D, Qin LY, Ruan Z, Shi Q, Ruan Q, Weigelt C, Qiu H, Schieven G, Hynes J, Bhide R, Poss M, Tino J. Identification of highly potent and selective PI3Kδ inhibitors. Bioorg Med Chem Lett 2017; 27:2849-2853. [DOI: 10.1016/j.bmcl.2017.01.077] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 01/23/2017] [Accepted: 01/25/2017] [Indexed: 01/22/2023]
|
20
|
Liu Q, Shi Q, Marcoux D, Batt DG, Cornelius L, Qin LY, Ruan Z, Neels J, Beaudoin-Bertrand M, Srivastava AS, Li L, Cherney RJ, Gong H, Watterson SH, Weigelt C, Gillooly KM, McIntyre KW, Xie JH, Obermeier MT, Fura A, Sleczka B, Stefanski K, Fancher RM, Padmanabhan S, Rp T, Kundu I, Rajareddy K, Smith R, Hennan JK, Xing D, Fan J, Levesque PC, Ruan Q, Pitt S, Zhang R, Pedicord D, Pan J, Yarde M, Lu H, Lippy J, Goldstine C, Skala S, Rampulla RA, Mathur A, Gupta A, Arunachalam PN, Sack JS, Muckelbauer JK, Cvijic ME, Salter-Cid LM, Bhide RS, Poss MA, Hynes J, Carter PH, Macor JE, Ruepp S, Schieven GL, Tino JA. Identification of a Potent, Selective, and Efficacious Phosphatidylinositol 3-Kinase δ (PI3Kδ) Inhibitor for the Treatment of Immunological Disorders. J Med Chem 2017; 60:5193-5208. [PMID: 28541707 DOI: 10.1021/acs.jmedchem.7b00618] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PI3Kδ plays an important role controlling immune cell function and has therefore been identified as a potential target for the treatment of immunological disorders. This article highlights our work toward the identification of a potent, selective, and efficacious PI3Kδ inhibitor. Through careful SAR, the successful replacement of a polar pyrazole group by a simple chloro or trifluoromethyl group led to improved Caco-2 permeability, reduced Caco-2 efflux, reduced hERG PC activity, and increased selectivity profile while maintaining potency in the CD69 hWB assay. The optimization of the aryl substitution then identified a 4'-CN group that improved the human/rodent correlation in microsomal metabolic stability. Our lead molecule is very potent in PK/PD assays and highly efficacious in a mouse collagen-induced arthritis model.
Collapse
Affiliation(s)
- Qingjie Liu
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Qing Shi
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - David Marcoux
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Douglas G Batt
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Lyndon Cornelius
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Lan-Ying Qin
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Zheming Ruan
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - James Neels
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Myra Beaudoin-Bertrand
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Anurag S Srivastava
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Ling Li
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Robert J Cherney
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Hua Gong
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Scott H Watterson
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Carolyn Weigelt
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Kathleen M Gillooly
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Kim W McIntyre
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Jenny H Xie
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Mary T Obermeier
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Aberra Fura
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Bogdan Sleczka
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Kevin Stefanski
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - R M Fancher
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Shweta Padmanabhan
- Department of Discovery Synthesis, Biocon Bristol-Myers Squibb Research Centre , Biocon Park, Bommasandra IV Phase, Jigani Link Road, Bengaluru 560099, India
| | - Thatipamula Rp
- Department of Discovery Synthesis, Biocon Bristol-Myers Squibb Research Centre , Biocon Park, Bommasandra IV Phase, Jigani Link Road, Bengaluru 560099, India
| | - Ipsit Kundu
- Department of Discovery Synthesis, Biocon Bristol-Myers Squibb Research Centre , Biocon Park, Bommasandra IV Phase, Jigani Link Road, Bengaluru 560099, India
| | | | - Rodney Smith
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - James K Hennan
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Dezhi Xing
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Jingsong Fan
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Paul C Levesque
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Qian Ruan
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Sidney Pitt
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Rosemary Zhang
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Donna Pedicord
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Jie Pan
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Melissa Yarde
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Hao Lu
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Jonathan Lippy
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Christine Goldstine
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Stacey Skala
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Richard A Rampulla
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Arvind Mathur
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Anuradha Gupta
- Department of Discovery Synthesis, Biocon Bristol-Myers Squibb Research Centre , Biocon Park, Bommasandra IV Phase, Jigani Link Road, Bengaluru 560099, India
| | - Pirama Nayagam Arunachalam
- Department of Discovery Synthesis, Biocon Bristol-Myers Squibb Research Centre , Biocon Park, Bommasandra IV Phase, Jigani Link Road, Bengaluru 560099, India
| | - John S Sack
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Jodi K Muckelbauer
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Mary Ellen Cvijic
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Luisa M Salter-Cid
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Rajeev S Bhide
- Department of Discovery Synthesis, Biocon Bristol-Myers Squibb Research Centre , Biocon Park, Bommasandra IV Phase, Jigani Link Road, Bengaluru 560099, India
| | - Michael A Poss
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - John Hynes
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Percy H Carter
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | | | - Stefan Ruepp
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Gary L Schieven
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Joseph A Tino
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| |
Collapse
|
21
|
Tykocki NR, Boerman EM, Jackson WF. Smooth Muscle Ion Channels and Regulation of Vascular Tone in Resistance Arteries and Arterioles. Compr Physiol 2017; 7:485-581. [PMID: 28333380 DOI: 10.1002/cphy.c160011] [Citation(s) in RCA: 228] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Vascular tone of resistance arteries and arterioles determines peripheral vascular resistance, contributing to the regulation of blood pressure and blood flow to, and within the body's tissues and organs. Ion channels in the plasma membrane and endoplasmic reticulum of vascular smooth muscle cells (SMCs) in these blood vessels importantly contribute to the regulation of intracellular Ca2+ concentration, the primary determinant of SMC contractile activity and vascular tone. Ion channels provide the main source of activator Ca2+ that determines vascular tone, and strongly contribute to setting and regulating membrane potential, which, in turn, regulates the open-state-probability of voltage gated Ca2+ channels (VGCCs), the primary source of Ca2+ in resistance artery and arteriolar SMCs. Ion channel function is also modulated by vasoconstrictors and vasodilators, contributing to all aspects of the regulation of vascular tone. This review will focus on the physiology of VGCCs, voltage-gated K+ (KV) channels, large-conductance Ca2+-activated K+ (BKCa) channels, strong-inward-rectifier K+ (KIR) channels, ATP-sensitive K+ (KATP) channels, ryanodine receptors (RyRs), inositol 1,4,5-trisphosphate receptors (IP3Rs), and a variety of transient receptor potential (TRP) channels that contribute to pressure-induced myogenic tone in resistance arteries and arterioles, the modulation of the function of these ion channels by vasoconstrictors and vasodilators, their role in the functional regulation of tissue blood flow and their dysfunction in diseases such as hypertension, obesity, and diabetes. © 2017 American Physiological Society. Compr Physiol 7:485-581, 2017.
Collapse
Affiliation(s)
- Nathan R Tykocki
- Department of Pharmacology, University of Vermont, Burlington, Vermont, USA
| | - Erika M Boerman
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, USA
| | - William F Jackson
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
22
|
Maffei A, Cifelli G, Carnevale R, Iacobucci R, Pallante F, Fardella V, Fardella S, Hirsch E, Lembo G, Carnevale D. La inhibición de la PI3Kγ protege contra la miocardiopatía diabética en ratones. Rev Esp Cardiol 2017. [DOI: 10.1016/j.recesp.2016.04.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
23
|
The Multifaceted Roles of PI3Kγ in Hypertension, Vascular Biology, and Inflammation. Int J Mol Sci 2016; 17:ijms17111858. [PMID: 27834808 PMCID: PMC5133858 DOI: 10.3390/ijms17111858] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 10/22/2016] [Accepted: 11/03/2016] [Indexed: 12/12/2022] Open
Abstract
PI3Kγ is a multifaceted protein, crucially involved in cardiovascular and immune systems. Several studies described the biological and physiological functions of this enzyme in the regulation of cardiovascular system, while others stressed its role in the modulation of immunity. Although PI3Kγ has been historically investigated for its role in leukocytes, the last decade of research also dedicated efforts to explore its functions in the cardiovascular system. In this review, we report an overview recapitulating how PI3Kγ signaling participates in the regulation of vascular functions involved in blood pressure regulation. Moreover, we also summarize the main functions of PI3Kγ in immune responses that could be potentially important in the interaction with the cardiovascular system. Considering that vascular and immune mechanisms are increasingly emerging as intertwining players in hypertension, PI3Kγ could be an intriguing pathway acting on both sides. The availability of specific inhibitors introduces a perspective of further translational research and clinical approaches that could be exploited in hypertension.
Collapse
|
24
|
The prosurvival protein BAG3: a new participant in vascular homeostasis. Cell Death Dis 2016; 7:e2431. [PMID: 27763645 PMCID: PMC5133988 DOI: 10.1038/cddis.2016.321] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 05/30/2016] [Accepted: 05/31/2016] [Indexed: 02/03/2023]
Abstract
Bcl2-associated athanogene 3 (BAG3), is constitutively expressed in a few normal cell types, including myocytes, peripheral nerves and in the brain, and is also expressed in certain tumors. To date, the main studies about the role of BAG3 are focused on its pro-survival effect in tumors through various mechanisms that vary according to cellular type. Recently, elevated concentrations of a soluble form of BAG3 were described in patients affected by advanced stage of heart failure (HF), identifying BAG3 as a potentially useful biomarker in monitoring HF progression. Despite the finding of high levels of BAG3 in the sera of HF patients, there are no data on its possible role on the modulation of vascular tone and blood pressure levels. The aim of this study was to investigate the possible hemodynamic effects of BAG3 performing both in vitro and in vivo experiments. Through vascular reactivity studies, we demonstrate that BAG3 is capable of evoking dose-dependent vasorelaxation. Of note, BAG3 exerts its vasorelaxant effect on resistance vessels, typically involved in the blood pressure regulation. Our data further show that the molecular mechanism through which BAG3 exerts this effect is the activation of the PI3K/Akt signalling pathway leading to nitric oxide release by endothelial cells. Finally, we show that in vivo BAG3 administration is capable of regulating blood pressure and that this is dependent on eNOS regulation since this ability is lost in eNOS KO animals.
Collapse
|
25
|
Maffei A, Cifelli G, Carnevale R, Iacobucci R, Pallante F, Fardella V, Fardella S, Hirsch E, Lembo G, Carnevale D. PI3Kγ Inhibition Protects Against Diabetic Cardiomyopathy in Mice. ACTA ACUST UNITED AC 2016; 70:16-24. [PMID: 27422446 DOI: 10.1016/j.rec.2016.04.034] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 04/07/2016] [Indexed: 11/26/2022]
Abstract
INTRODUCTION AND OBJECTIVES Cardiovascular diseases, including cardiomyopathy, are the major complications in diabetes. A deeper understanding of the molecular mechanisms leading to cardiomyopathy is critical for developing novel therapies. We proposed phosphoinositide3-kinase gamma (PI3Kγ) as a molecular target against diabetic cardiomyopathy, given the role of PI3Kγ in cardiac remodeling to pressure overload. Given the availability of a pharmacological inhibitor of this molecular target GE21, we tested the validity of our hypothesis by inducing diabetes in mice with genetic ablation of PI3Kγ or knock-in for a catalytically inactive PI3Kγ. METHODS Mice were made diabetic by streptozotocin. Cardiac function was assessed by serial echocardiographic analyses, while fibrosis and inflammation were evaluated by histological analysis. RESULTS Diabetes induced cardiac dysfunction in wild-type mice. Systolic dysfunction was completely prevented, and diastolic dysfunction was partially blocked, in both PI3Kγ knock-out and kinase-dead mice. Cardiac dysfunction was similarly rescued by administration of the PI3Kγ inhibitor GE21 in a dose-dependent manner. These actions of genetic and pharmacological PI3Kγ inhibition were associated with a decrease in inflammation and fibrosis in diabetic hearts. CONCLUSIONS Our study demonstrates a fundamental role of PI3Kγ in diabetic cardiomyopathy in mice and the beneficial effect of pharmacological PI3Kγ inhibition, highlighting its potential as a promising strategy for clinical treatment of cardiac complications of diabetic patients.
Collapse
Affiliation(s)
- Angelo Maffei
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, Istituto Neurologico Mediterraneo, Pozzilli, Isernia, Italy
| | - Giuseppe Cifelli
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, Istituto Neurologico Mediterraneo, Pozzilli, Isernia, Italy
| | - Raimondo Carnevale
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, Istituto Neurologico Mediterraneo, Pozzilli, Isernia, Italy
| | - Roberta Iacobucci
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, Istituto Neurologico Mediterraneo, Pozzilli, Isernia, Italy
| | - Fabio Pallante
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, Istituto Neurologico Mediterraneo, Pozzilli, Isernia, Italy
| | - Valentina Fardella
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, Istituto Neurologico Mediterraneo, Pozzilli, Isernia, Italy
| | - Stefania Fardella
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, Istituto Neurologico Mediterraneo, Pozzilli, Isernia, Italy
| | - Emilio Hirsch
- Department of Molecular Biotechnologies and Health Sciences, University of Torino, Turin, Italy
| | - Giuseppe Lembo
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, Istituto Neurologico Mediterraneo, Pozzilli, Isernia, Italy; Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Daniela Carnevale
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, Istituto Neurologico Mediterraneo, Pozzilli, Isernia, Italy; Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
26
|
Carrizzo A, Ambrosio M, Damato A, Madonna M, Storto M, Capocci L, Campiglia P, Sommella E, Trimarco V, Rozza F, Izzo R, Puca AA, Vecchione C. Morus alba extract modulates blood pressure homeostasis through eNOS signaling. Mol Nutr Food Res 2016; 60:2304-2311. [PMID: 27234065 DOI: 10.1002/mnfr.201600233] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 05/12/2016] [Accepted: 05/17/2016] [Indexed: 11/12/2022]
Abstract
SCOPE Morus alba is a promising phytomedicine cultivated in oriental countries that is extensively used to prevent and treat various cardiovascular problems. To date, despite its beneficial effects, the molecular mechanisms involved remain unclear. Thus, we investigate the vascular and haemodynamic effects of Morus alba extract in an experimental model focusing our attention on the molecular mechanisms involved. METHODS AND RESULTS Through vascular reactivity studies, we demonstrate that Morus alba extract evokes endothelial vasorelaxation through a nitric oxide-dependent pathway. Our molecular analysis highlights an increase in endothelial nitric oxide synthase (eNOS) phosphorylation. In vivo administration of Morus alba extract reduces blood pressure levels exclusively in wild-type mice, whereas it fails to evoke any haemodynamic effects in eNOS-deficient mice. Molecular analyses revealed that its beneficial action on vasculature is mediated by the activation of two important proteins that act as stress sensors and chaperones: PERK and heat shock protein 90. Finally, Morus alba extract exerts antihypertensive action in an experimental model of arterial hypertension. CONCLUSION Through its action on eNOS signaling, Morus alba extract could act as a food supplement for the regulation of cardiovascular system, mainly in clinical conditions characterized by eNOS dysfunction, such as arterial hypertension.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Pietro Campiglia
- European Biomedical Research Institute of Salerno, Salerno, Italy.,Department of Pharmacy, School of Pharmacy, University of Salerno, Fisciano (SA), Italy
| | - Eduardo Sommella
- Department of Agriculture, Laboratory of Food Chemistry, University of Reggio Calabria, Reggio Calabria, Italy.,Department of Pharmacy, School of Pharmacy, University of Salerno, Fisciano (SA), Italy
| | - Valentina Trimarco
- Hypertension Research Center, Federico II University Hospital, Naples, Italy.,Department of Neurosciences, Federico II University, Naples, Italy
| | - Francesco Rozza
- Hypertension Research Center, Federico II University Hospital, Naples, Italy.,Department of Advanced Biomedical Sciences, University Federico II of Naples, Naples, Italy
| | - Raffaele Izzo
- Hypertension Research Center, Federico II University Hospital, Naples, Italy.,Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Annibale A Puca
- IRCCS Multimedica, Milan, Italy.,Department of Medicine and Surgery, University of Salerno, Baronissi (SA), Italy
| | - Carmine Vecchione
- IRCCS Neuromed, Pozzilli (IS), Italy. .,Department of Medicine and Surgery, University of Salerno, Baronissi (SA), Italy.
| |
Collapse
|
27
|
Kauffenstein G, Tamareille S, Prunier F, Roy C, Ayer A, Toutain B, Billaud M, Isakson BE, Grimaud L, Loufrani L, Rousseau P, Abraham P, Procaccio V, Monyer H, de Wit C, Boeynaems JM, Robaye B, Kwak BR, Henrion D. Central Role of P2Y6 UDP Receptor in Arteriolar Myogenic Tone. Arterioscler Thromb Vasc Biol 2016; 36:1598-606. [PMID: 27255725 DOI: 10.1161/atvbaha.116.307739] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 05/17/2016] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Myogenic tone (MT) of resistance arteries ensures autoregulation of blood flow in organs and relies on the intrinsic property of smooth muscle to contract in response to stretch. Nucleotides released by mechanical strain on cells are responsible for pleiotropic vascular effects, including vasoconstriction. Here, we evaluated the contribution of extracellular nucleotides to MT. APPROACH AND RESULTS We measured MT and the associated pathway in mouse mesenteric resistance arteries using arteriography for small arteries and molecular biology. Of the P2 receptors in mouse mesenteric resistance arteries, mRNA expression of P2X1 and P2Y6 was dominant. P2Y6 fully sustained UDP/UTP-induced contraction (abrogated in P2ry6(-/-) arteries). Preventing nucleotide hydrolysis with the ectonucleotidase inhibitor ARL67156 enhanced pressure-induced MT by 20%, whereas P2Y6 receptor blockade blunted MT in mouse mesenteric resistance arteries and human subcutaneous arteries. Despite normal hemodynamic parameters, P2ry6(-/-) mice were protected against MT elevation in myocardial infarction-induced heart failure. Although both P2Y6 and P2Y2 receptors contributed to calcium mobilization, P2Y6 activation was mandatory for RhoA-GTP binding, myosin light chain, P42-P44, and c-Jun N-terminal kinase phosphorylation in arterial smooth muscle cells. In accordance with the opening of a nucleotide conduit in pressurized arteries, MT was altered by hemichannel pharmacological inhibitors and impaired in Cx43(+/-) and P2rx7(-/-) mesenteric resistance arteries. CONCLUSIONS Signaling through P2 nucleotide receptors contributes to MT. This mechanism encompasses the release of nucleotides coupled to specific autocrine/paracrine activation of the uracil nucleotide P2Y6 receptor and may contribute to impaired tissue perfusion in cardiovascular diseases.
Collapse
Affiliation(s)
- Gilles Kauffenstein
- From the MITOVASC Institute, CNRS UMR 6214, INSERM U1083 (G.K., C.R., A.A., B.T., L.G., L.L., P.A., V.P., D.H.) and EA 3860 Cardioprotection Remodelage et Thrombose, University of Angers, Angers, France (S.T., F.P.); Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville (M.B., B.E.I.); University Hospital Angers, Angers, France (G.K., P.R., P.A., V.P.); Department of Clinical Neurobiology, University Hospital and German Cancer Research Center Heidelberg, Heidelberg, Germany (H.M.); Institut für Physiologie, Universität zu Lübeck and Deutsches Zentrum für Herz-Kreislauf-Forschung, Lübeck, Germany (C.d.W.); Institute of Interdisciplinary Research, IRIBHM, Université Libre de Bruxelles, Gosselies, Belgium (J.-M.B., B.R.); and Departments of Pathology and Immunology and Medical Specializations - Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.).
| | - Sophie Tamareille
- From the MITOVASC Institute, CNRS UMR 6214, INSERM U1083 (G.K., C.R., A.A., B.T., L.G., L.L., P.A., V.P., D.H.) and EA 3860 Cardioprotection Remodelage et Thrombose, University of Angers, Angers, France (S.T., F.P.); Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville (M.B., B.E.I.); University Hospital Angers, Angers, France (G.K., P.R., P.A., V.P.); Department of Clinical Neurobiology, University Hospital and German Cancer Research Center Heidelberg, Heidelberg, Germany (H.M.); Institut für Physiologie, Universität zu Lübeck and Deutsches Zentrum für Herz-Kreislauf-Forschung, Lübeck, Germany (C.d.W.); Institute of Interdisciplinary Research, IRIBHM, Université Libre de Bruxelles, Gosselies, Belgium (J.-M.B., B.R.); and Departments of Pathology and Immunology and Medical Specializations - Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.)
| | - Fabrice Prunier
- From the MITOVASC Institute, CNRS UMR 6214, INSERM U1083 (G.K., C.R., A.A., B.T., L.G., L.L., P.A., V.P., D.H.) and EA 3860 Cardioprotection Remodelage et Thrombose, University of Angers, Angers, France (S.T., F.P.); Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville (M.B., B.E.I.); University Hospital Angers, Angers, France (G.K., P.R., P.A., V.P.); Department of Clinical Neurobiology, University Hospital and German Cancer Research Center Heidelberg, Heidelberg, Germany (H.M.); Institut für Physiologie, Universität zu Lübeck and Deutsches Zentrum für Herz-Kreislauf-Forschung, Lübeck, Germany (C.d.W.); Institute of Interdisciplinary Research, IRIBHM, Université Libre de Bruxelles, Gosselies, Belgium (J.-M.B., B.R.); and Departments of Pathology and Immunology and Medical Specializations - Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.)
| | - Charlotte Roy
- From the MITOVASC Institute, CNRS UMR 6214, INSERM U1083 (G.K., C.R., A.A., B.T., L.G., L.L., P.A., V.P., D.H.) and EA 3860 Cardioprotection Remodelage et Thrombose, University of Angers, Angers, France (S.T., F.P.); Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville (M.B., B.E.I.); University Hospital Angers, Angers, France (G.K., P.R., P.A., V.P.); Department of Clinical Neurobiology, University Hospital and German Cancer Research Center Heidelberg, Heidelberg, Germany (H.M.); Institut für Physiologie, Universität zu Lübeck and Deutsches Zentrum für Herz-Kreislauf-Forschung, Lübeck, Germany (C.d.W.); Institute of Interdisciplinary Research, IRIBHM, Université Libre de Bruxelles, Gosselies, Belgium (J.-M.B., B.R.); and Departments of Pathology and Immunology and Medical Specializations - Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.)
| | - Audrey Ayer
- From the MITOVASC Institute, CNRS UMR 6214, INSERM U1083 (G.K., C.R., A.A., B.T., L.G., L.L., P.A., V.P., D.H.) and EA 3860 Cardioprotection Remodelage et Thrombose, University of Angers, Angers, France (S.T., F.P.); Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville (M.B., B.E.I.); University Hospital Angers, Angers, France (G.K., P.R., P.A., V.P.); Department of Clinical Neurobiology, University Hospital and German Cancer Research Center Heidelberg, Heidelberg, Germany (H.M.); Institut für Physiologie, Universität zu Lübeck and Deutsches Zentrum für Herz-Kreislauf-Forschung, Lübeck, Germany (C.d.W.); Institute of Interdisciplinary Research, IRIBHM, Université Libre de Bruxelles, Gosselies, Belgium (J.-M.B., B.R.); and Departments of Pathology and Immunology and Medical Specializations - Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.)
| | - Bertrand Toutain
- From the MITOVASC Institute, CNRS UMR 6214, INSERM U1083 (G.K., C.R., A.A., B.T., L.G., L.L., P.A., V.P., D.H.) and EA 3860 Cardioprotection Remodelage et Thrombose, University of Angers, Angers, France (S.T., F.P.); Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville (M.B., B.E.I.); University Hospital Angers, Angers, France (G.K., P.R., P.A., V.P.); Department of Clinical Neurobiology, University Hospital and German Cancer Research Center Heidelberg, Heidelberg, Germany (H.M.); Institut für Physiologie, Universität zu Lübeck and Deutsches Zentrum für Herz-Kreislauf-Forschung, Lübeck, Germany (C.d.W.); Institute of Interdisciplinary Research, IRIBHM, Université Libre de Bruxelles, Gosselies, Belgium (J.-M.B., B.R.); and Departments of Pathology and Immunology and Medical Specializations - Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.)
| | - Marie Billaud
- From the MITOVASC Institute, CNRS UMR 6214, INSERM U1083 (G.K., C.R., A.A., B.T., L.G., L.L., P.A., V.P., D.H.) and EA 3860 Cardioprotection Remodelage et Thrombose, University of Angers, Angers, France (S.T., F.P.); Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville (M.B., B.E.I.); University Hospital Angers, Angers, France (G.K., P.R., P.A., V.P.); Department of Clinical Neurobiology, University Hospital and German Cancer Research Center Heidelberg, Heidelberg, Germany (H.M.); Institut für Physiologie, Universität zu Lübeck and Deutsches Zentrum für Herz-Kreislauf-Forschung, Lübeck, Germany (C.d.W.); Institute of Interdisciplinary Research, IRIBHM, Université Libre de Bruxelles, Gosselies, Belgium (J.-M.B., B.R.); and Departments of Pathology and Immunology and Medical Specializations - Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.)
| | - Brant E Isakson
- From the MITOVASC Institute, CNRS UMR 6214, INSERM U1083 (G.K., C.R., A.A., B.T., L.G., L.L., P.A., V.P., D.H.) and EA 3860 Cardioprotection Remodelage et Thrombose, University of Angers, Angers, France (S.T., F.P.); Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville (M.B., B.E.I.); University Hospital Angers, Angers, France (G.K., P.R., P.A., V.P.); Department of Clinical Neurobiology, University Hospital and German Cancer Research Center Heidelberg, Heidelberg, Germany (H.M.); Institut für Physiologie, Universität zu Lübeck and Deutsches Zentrum für Herz-Kreislauf-Forschung, Lübeck, Germany (C.d.W.); Institute of Interdisciplinary Research, IRIBHM, Université Libre de Bruxelles, Gosselies, Belgium (J.-M.B., B.R.); and Departments of Pathology and Immunology and Medical Specializations - Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.)
| | - Linda Grimaud
- From the MITOVASC Institute, CNRS UMR 6214, INSERM U1083 (G.K., C.R., A.A., B.T., L.G., L.L., P.A., V.P., D.H.) and EA 3860 Cardioprotection Remodelage et Thrombose, University of Angers, Angers, France (S.T., F.P.); Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville (M.B., B.E.I.); University Hospital Angers, Angers, France (G.K., P.R., P.A., V.P.); Department of Clinical Neurobiology, University Hospital and German Cancer Research Center Heidelberg, Heidelberg, Germany (H.M.); Institut für Physiologie, Universität zu Lübeck and Deutsches Zentrum für Herz-Kreislauf-Forschung, Lübeck, Germany (C.d.W.); Institute of Interdisciplinary Research, IRIBHM, Université Libre de Bruxelles, Gosselies, Belgium (J.-M.B., B.R.); and Departments of Pathology and Immunology and Medical Specializations - Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.)
| | - Laurent Loufrani
- From the MITOVASC Institute, CNRS UMR 6214, INSERM U1083 (G.K., C.R., A.A., B.T., L.G., L.L., P.A., V.P., D.H.) and EA 3860 Cardioprotection Remodelage et Thrombose, University of Angers, Angers, France (S.T., F.P.); Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville (M.B., B.E.I.); University Hospital Angers, Angers, France (G.K., P.R., P.A., V.P.); Department of Clinical Neurobiology, University Hospital and German Cancer Research Center Heidelberg, Heidelberg, Germany (H.M.); Institut für Physiologie, Universität zu Lübeck and Deutsches Zentrum für Herz-Kreislauf-Forschung, Lübeck, Germany (C.d.W.); Institute of Interdisciplinary Research, IRIBHM, Université Libre de Bruxelles, Gosselies, Belgium (J.-M.B., B.R.); and Departments of Pathology and Immunology and Medical Specializations - Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.)
| | - Pascal Rousseau
- From the MITOVASC Institute, CNRS UMR 6214, INSERM U1083 (G.K., C.R., A.A., B.T., L.G., L.L., P.A., V.P., D.H.) and EA 3860 Cardioprotection Remodelage et Thrombose, University of Angers, Angers, France (S.T., F.P.); Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville (M.B., B.E.I.); University Hospital Angers, Angers, France (G.K., P.R., P.A., V.P.); Department of Clinical Neurobiology, University Hospital and German Cancer Research Center Heidelberg, Heidelberg, Germany (H.M.); Institut für Physiologie, Universität zu Lübeck and Deutsches Zentrum für Herz-Kreislauf-Forschung, Lübeck, Germany (C.d.W.); Institute of Interdisciplinary Research, IRIBHM, Université Libre de Bruxelles, Gosselies, Belgium (J.-M.B., B.R.); and Departments of Pathology and Immunology and Medical Specializations - Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.)
| | - Pierre Abraham
- From the MITOVASC Institute, CNRS UMR 6214, INSERM U1083 (G.K., C.R., A.A., B.T., L.G., L.L., P.A., V.P., D.H.) and EA 3860 Cardioprotection Remodelage et Thrombose, University of Angers, Angers, France (S.T., F.P.); Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville (M.B., B.E.I.); University Hospital Angers, Angers, France (G.K., P.R., P.A., V.P.); Department of Clinical Neurobiology, University Hospital and German Cancer Research Center Heidelberg, Heidelberg, Germany (H.M.); Institut für Physiologie, Universität zu Lübeck and Deutsches Zentrum für Herz-Kreislauf-Forschung, Lübeck, Germany (C.d.W.); Institute of Interdisciplinary Research, IRIBHM, Université Libre de Bruxelles, Gosselies, Belgium (J.-M.B., B.R.); and Departments of Pathology and Immunology and Medical Specializations - Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.)
| | - Vincent Procaccio
- From the MITOVASC Institute, CNRS UMR 6214, INSERM U1083 (G.K., C.R., A.A., B.T., L.G., L.L., P.A., V.P., D.H.) and EA 3860 Cardioprotection Remodelage et Thrombose, University of Angers, Angers, France (S.T., F.P.); Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville (M.B., B.E.I.); University Hospital Angers, Angers, France (G.K., P.R., P.A., V.P.); Department of Clinical Neurobiology, University Hospital and German Cancer Research Center Heidelberg, Heidelberg, Germany (H.M.); Institut für Physiologie, Universität zu Lübeck and Deutsches Zentrum für Herz-Kreislauf-Forschung, Lübeck, Germany (C.d.W.); Institute of Interdisciplinary Research, IRIBHM, Université Libre de Bruxelles, Gosselies, Belgium (J.-M.B., B.R.); and Departments of Pathology and Immunology and Medical Specializations - Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.)
| | - Hannah Monyer
- From the MITOVASC Institute, CNRS UMR 6214, INSERM U1083 (G.K., C.R., A.A., B.T., L.G., L.L., P.A., V.P., D.H.) and EA 3860 Cardioprotection Remodelage et Thrombose, University of Angers, Angers, France (S.T., F.P.); Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville (M.B., B.E.I.); University Hospital Angers, Angers, France (G.K., P.R., P.A., V.P.); Department of Clinical Neurobiology, University Hospital and German Cancer Research Center Heidelberg, Heidelberg, Germany (H.M.); Institut für Physiologie, Universität zu Lübeck and Deutsches Zentrum für Herz-Kreislauf-Forschung, Lübeck, Germany (C.d.W.); Institute of Interdisciplinary Research, IRIBHM, Université Libre de Bruxelles, Gosselies, Belgium (J.-M.B., B.R.); and Departments of Pathology and Immunology and Medical Specializations - Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.)
| | - Cor de Wit
- From the MITOVASC Institute, CNRS UMR 6214, INSERM U1083 (G.K., C.R., A.A., B.T., L.G., L.L., P.A., V.P., D.H.) and EA 3860 Cardioprotection Remodelage et Thrombose, University of Angers, Angers, France (S.T., F.P.); Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville (M.B., B.E.I.); University Hospital Angers, Angers, France (G.K., P.R., P.A., V.P.); Department of Clinical Neurobiology, University Hospital and German Cancer Research Center Heidelberg, Heidelberg, Germany (H.M.); Institut für Physiologie, Universität zu Lübeck and Deutsches Zentrum für Herz-Kreislauf-Forschung, Lübeck, Germany (C.d.W.); Institute of Interdisciplinary Research, IRIBHM, Université Libre de Bruxelles, Gosselies, Belgium (J.-M.B., B.R.); and Departments of Pathology and Immunology and Medical Specializations - Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.)
| | - Jean-Marie Boeynaems
- From the MITOVASC Institute, CNRS UMR 6214, INSERM U1083 (G.K., C.R., A.A., B.T., L.G., L.L., P.A., V.P., D.H.) and EA 3860 Cardioprotection Remodelage et Thrombose, University of Angers, Angers, France (S.T., F.P.); Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville (M.B., B.E.I.); University Hospital Angers, Angers, France (G.K., P.R., P.A., V.P.); Department of Clinical Neurobiology, University Hospital and German Cancer Research Center Heidelberg, Heidelberg, Germany (H.M.); Institut für Physiologie, Universität zu Lübeck and Deutsches Zentrum für Herz-Kreislauf-Forschung, Lübeck, Germany (C.d.W.); Institute of Interdisciplinary Research, IRIBHM, Université Libre de Bruxelles, Gosselies, Belgium (J.-M.B., B.R.); and Departments of Pathology and Immunology and Medical Specializations - Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.)
| | - Bernard Robaye
- From the MITOVASC Institute, CNRS UMR 6214, INSERM U1083 (G.K., C.R., A.A., B.T., L.G., L.L., P.A., V.P., D.H.) and EA 3860 Cardioprotection Remodelage et Thrombose, University of Angers, Angers, France (S.T., F.P.); Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville (M.B., B.E.I.); University Hospital Angers, Angers, France (G.K., P.R., P.A., V.P.); Department of Clinical Neurobiology, University Hospital and German Cancer Research Center Heidelberg, Heidelberg, Germany (H.M.); Institut für Physiologie, Universität zu Lübeck and Deutsches Zentrum für Herz-Kreislauf-Forschung, Lübeck, Germany (C.d.W.); Institute of Interdisciplinary Research, IRIBHM, Université Libre de Bruxelles, Gosselies, Belgium (J.-M.B., B.R.); and Departments of Pathology and Immunology and Medical Specializations - Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.)
| | - Brenda R Kwak
- From the MITOVASC Institute, CNRS UMR 6214, INSERM U1083 (G.K., C.R., A.A., B.T., L.G., L.L., P.A., V.P., D.H.) and EA 3860 Cardioprotection Remodelage et Thrombose, University of Angers, Angers, France (S.T., F.P.); Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville (M.B., B.E.I.); University Hospital Angers, Angers, France (G.K., P.R., P.A., V.P.); Department of Clinical Neurobiology, University Hospital and German Cancer Research Center Heidelberg, Heidelberg, Germany (H.M.); Institut für Physiologie, Universität zu Lübeck and Deutsches Zentrum für Herz-Kreislauf-Forschung, Lübeck, Germany (C.d.W.); Institute of Interdisciplinary Research, IRIBHM, Université Libre de Bruxelles, Gosselies, Belgium (J.-M.B., B.R.); and Departments of Pathology and Immunology and Medical Specializations - Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.)
| | - Daniel Henrion
- From the MITOVASC Institute, CNRS UMR 6214, INSERM U1083 (G.K., C.R., A.A., B.T., L.G., L.L., P.A., V.P., D.H.) and EA 3860 Cardioprotection Remodelage et Thrombose, University of Angers, Angers, France (S.T., F.P.); Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville (M.B., B.E.I.); University Hospital Angers, Angers, France (G.K., P.R., P.A., V.P.); Department of Clinical Neurobiology, University Hospital and German Cancer Research Center Heidelberg, Heidelberg, Germany (H.M.); Institut für Physiologie, Universität zu Lübeck and Deutsches Zentrum für Herz-Kreislauf-Forschung, Lübeck, Germany (C.d.W.); Institute of Interdisciplinary Research, IRIBHM, Université Libre de Bruxelles, Gosselies, Belgium (J.-M.B., B.R.); and Departments of Pathology and Immunology and Medical Specializations - Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.)
| |
Collapse
|
28
|
Villa F, Carrizzo A, Spinelli CC, Ferrario A, Malovini A, Maciąg A, Damato A, Auricchio A, Spinetti G, Sangalli E, Dang Z, Madonna M, Ambrosio M, Sitia L, Bigini P, Calì G, Schreiber S, Perls T, Fucile S, Mulas F, Nebel A, Bellazzi R, Madeddu P, Vecchione C, Puca AA. Genetic Analysis Reveals a Longevity-Associated Protein Modulating Endothelial Function and Angiogenesis. Circ Res 2015; 117:333-45. [PMID: 26034043 DOI: 10.1161/circresaha.117.305875] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 06/01/2015] [Indexed: 12/30/2022]
Abstract
RATIONALE Long living individuals show delay of aging, which is characterized by the progressive loss of cardiovascular homeostasis, along with reduced endothelial nitric oxide synthase activity, endothelial dysfunction, and impairment of tissue repair after ischemic injury. OBJECTIVE Exploit genetic analysis of long living individuals to reveal master molecular regulators of physiological aging and new targets for treatment of cardiovascular disease. METHODS AND RESULTS We show that the polymorphic variant rs2070325 (Ile229Val) in bactericidal/permeability-increasing fold-containing-family-B-member-4 (BPIFB4) associates with exceptional longevity, under a recessive genetic model, in 3 independent populations. Moreover, the expression of BPIFB4 is instrumental to maintenance of cellular and vascular homeostasis through regulation of protein synthesis. BPIFB4 phosphorylation/activation by protein-kinase-R-like endoplasmic reticulum kinase induces its complexing with 14-3-3 and heat shock protein 90, which is facilitated by the longevity-associated variant. In isolated vessels, BPIFB4 is upregulated by mechanical stress, and its knock-down inhibits endothelium-dependent vasorelaxation. In hypertensive rats and old mice, gene transfer of longevity-associated variant-BPIFB4 restores endothelial nitric oxide synthase signaling, rescues endothelial dysfunction, and reduces blood pressure levels. Furthermore, BPIFB4 is implicated in vascular repair. BPIFB4 is abundantly expressed in circulating CD34(+) cells of long living individuals, and its knock-down in endothelial progenitor cells precludes their capacity to migrate toward the chemoattractant SDF-1. In a murine model of peripheral ischemia, systemic gene therapy with longevity-associated variant-BPIFB4 promotes the recruitment of hematopoietic stem cells, reparative vascularization, and reperfusion of the ischemic muscle. CONCLUSIONS Longevity-associated variant-BPIFB4 may represent a novel therapeutic tool to fight endothelial dysfunction and promote vascular reparative processes.
Collapse
Affiliation(s)
- Francesco Villa
- From the National Research Council, Institute for Biomedical Technologies, Segrate (MI), Italy (F.V., C.C.S., A.F.); IRCCS Neuromed, Department of Vascular Physiopathology, Pozzilli (IS), Italy (A.C., A.D., M.M., M.A., S.F., C.V.); Department of Industrial and Information Engineering, University of Pavia, Pavia, Italy (A. Malovini, F.M., R.B.); IRCCS Multimedica, Cardiovascular Department, Milan, Italy (A. Maciąg, G.S., E.S., A.A.P.); Department of Translational Medicine, "Federico II" University, Naples, Italy (A.A.); TIGEM (Telethon Institute of Genetics and Medicine), Naples, Italy (A.A.); Department of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, Bristol, United Kingdom (Z.D., P.M.); Department of Biochemistry and Molecular Pharmacology IRCCS Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy (L.S., P.B.); National Research Council, Institute of Experimental Endocrinology and Oncology (IEOS), Naples, Italy (G.C.); Institute of Clinical Molecular Biology, Christian-Albrechts University and the Schleswig-Holstein University Hospital, Kiel, Germany (S.S., A.N.); Geriatrics Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA (T.P.); Dipartimento di Fisiologia e Farmacologia, Sapienza Università di Roma, Rome, Italy (S.F.); and Dipartimento di Medicina e Chirurgia, Università degli Studi di Salerno, 84081 Baronissi (SA), Italy (C.V., A.A.P.)
| | - Albino Carrizzo
- From the National Research Council, Institute for Biomedical Technologies, Segrate (MI), Italy (F.V., C.C.S., A.F.); IRCCS Neuromed, Department of Vascular Physiopathology, Pozzilli (IS), Italy (A.C., A.D., M.M., M.A., S.F., C.V.); Department of Industrial and Information Engineering, University of Pavia, Pavia, Italy (A. Malovini, F.M., R.B.); IRCCS Multimedica, Cardiovascular Department, Milan, Italy (A. Maciąg, G.S., E.S., A.A.P.); Department of Translational Medicine, "Federico II" University, Naples, Italy (A.A.); TIGEM (Telethon Institute of Genetics and Medicine), Naples, Italy (A.A.); Department of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, Bristol, United Kingdom (Z.D., P.M.); Department of Biochemistry and Molecular Pharmacology IRCCS Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy (L.S., P.B.); National Research Council, Institute of Experimental Endocrinology and Oncology (IEOS), Naples, Italy (G.C.); Institute of Clinical Molecular Biology, Christian-Albrechts University and the Schleswig-Holstein University Hospital, Kiel, Germany (S.S., A.N.); Geriatrics Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA (T.P.); Dipartimento di Fisiologia e Farmacologia, Sapienza Università di Roma, Rome, Italy (S.F.); and Dipartimento di Medicina e Chirurgia, Università degli Studi di Salerno, 84081 Baronissi (SA), Italy (C.V., A.A.P.)
| | - Chiara C Spinelli
- From the National Research Council, Institute for Biomedical Technologies, Segrate (MI), Italy (F.V., C.C.S., A.F.); IRCCS Neuromed, Department of Vascular Physiopathology, Pozzilli (IS), Italy (A.C., A.D., M.M., M.A., S.F., C.V.); Department of Industrial and Information Engineering, University of Pavia, Pavia, Italy (A. Malovini, F.M., R.B.); IRCCS Multimedica, Cardiovascular Department, Milan, Italy (A. Maciąg, G.S., E.S., A.A.P.); Department of Translational Medicine, "Federico II" University, Naples, Italy (A.A.); TIGEM (Telethon Institute of Genetics and Medicine), Naples, Italy (A.A.); Department of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, Bristol, United Kingdom (Z.D., P.M.); Department of Biochemistry and Molecular Pharmacology IRCCS Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy (L.S., P.B.); National Research Council, Institute of Experimental Endocrinology and Oncology (IEOS), Naples, Italy (G.C.); Institute of Clinical Molecular Biology, Christian-Albrechts University and the Schleswig-Holstein University Hospital, Kiel, Germany (S.S., A.N.); Geriatrics Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA (T.P.); Dipartimento di Fisiologia e Farmacologia, Sapienza Università di Roma, Rome, Italy (S.F.); and Dipartimento di Medicina e Chirurgia, Università degli Studi di Salerno, 84081 Baronissi (SA), Italy (C.V., A.A.P.)
| | - Anna Ferrario
- From the National Research Council, Institute for Biomedical Technologies, Segrate (MI), Italy (F.V., C.C.S., A.F.); IRCCS Neuromed, Department of Vascular Physiopathology, Pozzilli (IS), Italy (A.C., A.D., M.M., M.A., S.F., C.V.); Department of Industrial and Information Engineering, University of Pavia, Pavia, Italy (A. Malovini, F.M., R.B.); IRCCS Multimedica, Cardiovascular Department, Milan, Italy (A. Maciąg, G.S., E.S., A.A.P.); Department of Translational Medicine, "Federico II" University, Naples, Italy (A.A.); TIGEM (Telethon Institute of Genetics and Medicine), Naples, Italy (A.A.); Department of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, Bristol, United Kingdom (Z.D., P.M.); Department of Biochemistry and Molecular Pharmacology IRCCS Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy (L.S., P.B.); National Research Council, Institute of Experimental Endocrinology and Oncology (IEOS), Naples, Italy (G.C.); Institute of Clinical Molecular Biology, Christian-Albrechts University and the Schleswig-Holstein University Hospital, Kiel, Germany (S.S., A.N.); Geriatrics Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA (T.P.); Dipartimento di Fisiologia e Farmacologia, Sapienza Università di Roma, Rome, Italy (S.F.); and Dipartimento di Medicina e Chirurgia, Università degli Studi di Salerno, 84081 Baronissi (SA), Italy (C.V., A.A.P.)
| | - Alberto Malovini
- From the National Research Council, Institute for Biomedical Technologies, Segrate (MI), Italy (F.V., C.C.S., A.F.); IRCCS Neuromed, Department of Vascular Physiopathology, Pozzilli (IS), Italy (A.C., A.D., M.M., M.A., S.F., C.V.); Department of Industrial and Information Engineering, University of Pavia, Pavia, Italy (A. Malovini, F.M., R.B.); IRCCS Multimedica, Cardiovascular Department, Milan, Italy (A. Maciąg, G.S., E.S., A.A.P.); Department of Translational Medicine, "Federico II" University, Naples, Italy (A.A.); TIGEM (Telethon Institute of Genetics and Medicine), Naples, Italy (A.A.); Department of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, Bristol, United Kingdom (Z.D., P.M.); Department of Biochemistry and Molecular Pharmacology IRCCS Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy (L.S., P.B.); National Research Council, Institute of Experimental Endocrinology and Oncology (IEOS), Naples, Italy (G.C.); Institute of Clinical Molecular Biology, Christian-Albrechts University and the Schleswig-Holstein University Hospital, Kiel, Germany (S.S., A.N.); Geriatrics Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA (T.P.); Dipartimento di Fisiologia e Farmacologia, Sapienza Università di Roma, Rome, Italy (S.F.); and Dipartimento di Medicina e Chirurgia, Università degli Studi di Salerno, 84081 Baronissi (SA), Italy (C.V., A.A.P.)
| | - Anna Maciąg
- From the National Research Council, Institute for Biomedical Technologies, Segrate (MI), Italy (F.V., C.C.S., A.F.); IRCCS Neuromed, Department of Vascular Physiopathology, Pozzilli (IS), Italy (A.C., A.D., M.M., M.A., S.F., C.V.); Department of Industrial and Information Engineering, University of Pavia, Pavia, Italy (A. Malovini, F.M., R.B.); IRCCS Multimedica, Cardiovascular Department, Milan, Italy (A. Maciąg, G.S., E.S., A.A.P.); Department of Translational Medicine, "Federico II" University, Naples, Italy (A.A.); TIGEM (Telethon Institute of Genetics and Medicine), Naples, Italy (A.A.); Department of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, Bristol, United Kingdom (Z.D., P.M.); Department of Biochemistry and Molecular Pharmacology IRCCS Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy (L.S., P.B.); National Research Council, Institute of Experimental Endocrinology and Oncology (IEOS), Naples, Italy (G.C.); Institute of Clinical Molecular Biology, Christian-Albrechts University and the Schleswig-Holstein University Hospital, Kiel, Germany (S.S., A.N.); Geriatrics Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA (T.P.); Dipartimento di Fisiologia e Farmacologia, Sapienza Università di Roma, Rome, Italy (S.F.); and Dipartimento di Medicina e Chirurgia, Università degli Studi di Salerno, 84081 Baronissi (SA), Italy (C.V., A.A.P.)
| | - Antonio Damato
- From the National Research Council, Institute for Biomedical Technologies, Segrate (MI), Italy (F.V., C.C.S., A.F.); IRCCS Neuromed, Department of Vascular Physiopathology, Pozzilli (IS), Italy (A.C., A.D., M.M., M.A., S.F., C.V.); Department of Industrial and Information Engineering, University of Pavia, Pavia, Italy (A. Malovini, F.M., R.B.); IRCCS Multimedica, Cardiovascular Department, Milan, Italy (A. Maciąg, G.S., E.S., A.A.P.); Department of Translational Medicine, "Federico II" University, Naples, Italy (A.A.); TIGEM (Telethon Institute of Genetics and Medicine), Naples, Italy (A.A.); Department of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, Bristol, United Kingdom (Z.D., P.M.); Department of Biochemistry and Molecular Pharmacology IRCCS Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy (L.S., P.B.); National Research Council, Institute of Experimental Endocrinology and Oncology (IEOS), Naples, Italy (G.C.); Institute of Clinical Molecular Biology, Christian-Albrechts University and the Schleswig-Holstein University Hospital, Kiel, Germany (S.S., A.N.); Geriatrics Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA (T.P.); Dipartimento di Fisiologia e Farmacologia, Sapienza Università di Roma, Rome, Italy (S.F.); and Dipartimento di Medicina e Chirurgia, Università degli Studi di Salerno, 84081 Baronissi (SA), Italy (C.V., A.A.P.)
| | - Alberto Auricchio
- From the National Research Council, Institute for Biomedical Technologies, Segrate (MI), Italy (F.V., C.C.S., A.F.); IRCCS Neuromed, Department of Vascular Physiopathology, Pozzilli (IS), Italy (A.C., A.D., M.M., M.A., S.F., C.V.); Department of Industrial and Information Engineering, University of Pavia, Pavia, Italy (A. Malovini, F.M., R.B.); IRCCS Multimedica, Cardiovascular Department, Milan, Italy (A. Maciąg, G.S., E.S., A.A.P.); Department of Translational Medicine, "Federico II" University, Naples, Italy (A.A.); TIGEM (Telethon Institute of Genetics and Medicine), Naples, Italy (A.A.); Department of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, Bristol, United Kingdom (Z.D., P.M.); Department of Biochemistry and Molecular Pharmacology IRCCS Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy (L.S., P.B.); National Research Council, Institute of Experimental Endocrinology and Oncology (IEOS), Naples, Italy (G.C.); Institute of Clinical Molecular Biology, Christian-Albrechts University and the Schleswig-Holstein University Hospital, Kiel, Germany (S.S., A.N.); Geriatrics Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA (T.P.); Dipartimento di Fisiologia e Farmacologia, Sapienza Università di Roma, Rome, Italy (S.F.); and Dipartimento di Medicina e Chirurgia, Università degli Studi di Salerno, 84081 Baronissi (SA), Italy (C.V., A.A.P.)
| | - Gaia Spinetti
- From the National Research Council, Institute for Biomedical Technologies, Segrate (MI), Italy (F.V., C.C.S., A.F.); IRCCS Neuromed, Department of Vascular Physiopathology, Pozzilli (IS), Italy (A.C., A.D., M.M., M.A., S.F., C.V.); Department of Industrial and Information Engineering, University of Pavia, Pavia, Italy (A. Malovini, F.M., R.B.); IRCCS Multimedica, Cardiovascular Department, Milan, Italy (A. Maciąg, G.S., E.S., A.A.P.); Department of Translational Medicine, "Federico II" University, Naples, Italy (A.A.); TIGEM (Telethon Institute of Genetics and Medicine), Naples, Italy (A.A.); Department of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, Bristol, United Kingdom (Z.D., P.M.); Department of Biochemistry and Molecular Pharmacology IRCCS Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy (L.S., P.B.); National Research Council, Institute of Experimental Endocrinology and Oncology (IEOS), Naples, Italy (G.C.); Institute of Clinical Molecular Biology, Christian-Albrechts University and the Schleswig-Holstein University Hospital, Kiel, Germany (S.S., A.N.); Geriatrics Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA (T.P.); Dipartimento di Fisiologia e Farmacologia, Sapienza Università di Roma, Rome, Italy (S.F.); and Dipartimento di Medicina e Chirurgia, Università degli Studi di Salerno, 84081 Baronissi (SA), Italy (C.V., A.A.P.)
| | - Elena Sangalli
- From the National Research Council, Institute for Biomedical Technologies, Segrate (MI), Italy (F.V., C.C.S., A.F.); IRCCS Neuromed, Department of Vascular Physiopathology, Pozzilli (IS), Italy (A.C., A.D., M.M., M.A., S.F., C.V.); Department of Industrial and Information Engineering, University of Pavia, Pavia, Italy (A. Malovini, F.M., R.B.); IRCCS Multimedica, Cardiovascular Department, Milan, Italy (A. Maciąg, G.S., E.S., A.A.P.); Department of Translational Medicine, "Federico II" University, Naples, Italy (A.A.); TIGEM (Telethon Institute of Genetics and Medicine), Naples, Italy (A.A.); Department of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, Bristol, United Kingdom (Z.D., P.M.); Department of Biochemistry and Molecular Pharmacology IRCCS Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy (L.S., P.B.); National Research Council, Institute of Experimental Endocrinology and Oncology (IEOS), Naples, Italy (G.C.); Institute of Clinical Molecular Biology, Christian-Albrechts University and the Schleswig-Holstein University Hospital, Kiel, Germany (S.S., A.N.); Geriatrics Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA (T.P.); Dipartimento di Fisiologia e Farmacologia, Sapienza Università di Roma, Rome, Italy (S.F.); and Dipartimento di Medicina e Chirurgia, Università degli Studi di Salerno, 84081 Baronissi (SA), Italy (C.V., A.A.P.)
| | - Zexu Dang
- From the National Research Council, Institute for Biomedical Technologies, Segrate (MI), Italy (F.V., C.C.S., A.F.); IRCCS Neuromed, Department of Vascular Physiopathology, Pozzilli (IS), Italy (A.C., A.D., M.M., M.A., S.F., C.V.); Department of Industrial and Information Engineering, University of Pavia, Pavia, Italy (A. Malovini, F.M., R.B.); IRCCS Multimedica, Cardiovascular Department, Milan, Italy (A. Maciąg, G.S., E.S., A.A.P.); Department of Translational Medicine, "Federico II" University, Naples, Italy (A.A.); TIGEM (Telethon Institute of Genetics and Medicine), Naples, Italy (A.A.); Department of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, Bristol, United Kingdom (Z.D., P.M.); Department of Biochemistry and Molecular Pharmacology IRCCS Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy (L.S., P.B.); National Research Council, Institute of Experimental Endocrinology and Oncology (IEOS), Naples, Italy (G.C.); Institute of Clinical Molecular Biology, Christian-Albrechts University and the Schleswig-Holstein University Hospital, Kiel, Germany (S.S., A.N.); Geriatrics Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA (T.P.); Dipartimento di Fisiologia e Farmacologia, Sapienza Università di Roma, Rome, Italy (S.F.); and Dipartimento di Medicina e Chirurgia, Università degli Studi di Salerno, 84081 Baronissi (SA), Italy (C.V., A.A.P.)
| | - Michele Madonna
- From the National Research Council, Institute for Biomedical Technologies, Segrate (MI), Italy (F.V., C.C.S., A.F.); IRCCS Neuromed, Department of Vascular Physiopathology, Pozzilli (IS), Italy (A.C., A.D., M.M., M.A., S.F., C.V.); Department of Industrial and Information Engineering, University of Pavia, Pavia, Italy (A. Malovini, F.M., R.B.); IRCCS Multimedica, Cardiovascular Department, Milan, Italy (A. Maciąg, G.S., E.S., A.A.P.); Department of Translational Medicine, "Federico II" University, Naples, Italy (A.A.); TIGEM (Telethon Institute of Genetics and Medicine), Naples, Italy (A.A.); Department of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, Bristol, United Kingdom (Z.D., P.M.); Department of Biochemistry and Molecular Pharmacology IRCCS Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy (L.S., P.B.); National Research Council, Institute of Experimental Endocrinology and Oncology (IEOS), Naples, Italy (G.C.); Institute of Clinical Molecular Biology, Christian-Albrechts University and the Schleswig-Holstein University Hospital, Kiel, Germany (S.S., A.N.); Geriatrics Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA (T.P.); Dipartimento di Fisiologia e Farmacologia, Sapienza Università di Roma, Rome, Italy (S.F.); and Dipartimento di Medicina e Chirurgia, Università degli Studi di Salerno, 84081 Baronissi (SA), Italy (C.V., A.A.P.)
| | - Mariateresa Ambrosio
- From the National Research Council, Institute for Biomedical Technologies, Segrate (MI), Italy (F.V., C.C.S., A.F.); IRCCS Neuromed, Department of Vascular Physiopathology, Pozzilli (IS), Italy (A.C., A.D., M.M., M.A., S.F., C.V.); Department of Industrial and Information Engineering, University of Pavia, Pavia, Italy (A. Malovini, F.M., R.B.); IRCCS Multimedica, Cardiovascular Department, Milan, Italy (A. Maciąg, G.S., E.S., A.A.P.); Department of Translational Medicine, "Federico II" University, Naples, Italy (A.A.); TIGEM (Telethon Institute of Genetics and Medicine), Naples, Italy (A.A.); Department of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, Bristol, United Kingdom (Z.D., P.M.); Department of Biochemistry and Molecular Pharmacology IRCCS Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy (L.S., P.B.); National Research Council, Institute of Experimental Endocrinology and Oncology (IEOS), Naples, Italy (G.C.); Institute of Clinical Molecular Biology, Christian-Albrechts University and the Schleswig-Holstein University Hospital, Kiel, Germany (S.S., A.N.); Geriatrics Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA (T.P.); Dipartimento di Fisiologia e Farmacologia, Sapienza Università di Roma, Rome, Italy (S.F.); and Dipartimento di Medicina e Chirurgia, Università degli Studi di Salerno, 84081 Baronissi (SA), Italy (C.V., A.A.P.)
| | - Leopoldo Sitia
- From the National Research Council, Institute for Biomedical Technologies, Segrate (MI), Italy (F.V., C.C.S., A.F.); IRCCS Neuromed, Department of Vascular Physiopathology, Pozzilli (IS), Italy (A.C., A.D., M.M., M.A., S.F., C.V.); Department of Industrial and Information Engineering, University of Pavia, Pavia, Italy (A. Malovini, F.M., R.B.); IRCCS Multimedica, Cardiovascular Department, Milan, Italy (A. Maciąg, G.S., E.S., A.A.P.); Department of Translational Medicine, "Federico II" University, Naples, Italy (A.A.); TIGEM (Telethon Institute of Genetics and Medicine), Naples, Italy (A.A.); Department of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, Bristol, United Kingdom (Z.D., P.M.); Department of Biochemistry and Molecular Pharmacology IRCCS Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy (L.S., P.B.); National Research Council, Institute of Experimental Endocrinology and Oncology (IEOS), Naples, Italy (G.C.); Institute of Clinical Molecular Biology, Christian-Albrechts University and the Schleswig-Holstein University Hospital, Kiel, Germany (S.S., A.N.); Geriatrics Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA (T.P.); Dipartimento di Fisiologia e Farmacologia, Sapienza Università di Roma, Rome, Italy (S.F.); and Dipartimento di Medicina e Chirurgia, Università degli Studi di Salerno, 84081 Baronissi (SA), Italy (C.V., A.A.P.)
| | - Paolo Bigini
- From the National Research Council, Institute for Biomedical Technologies, Segrate (MI), Italy (F.V., C.C.S., A.F.); IRCCS Neuromed, Department of Vascular Physiopathology, Pozzilli (IS), Italy (A.C., A.D., M.M., M.A., S.F., C.V.); Department of Industrial and Information Engineering, University of Pavia, Pavia, Italy (A. Malovini, F.M., R.B.); IRCCS Multimedica, Cardiovascular Department, Milan, Italy (A. Maciąg, G.S., E.S., A.A.P.); Department of Translational Medicine, "Federico II" University, Naples, Italy (A.A.); TIGEM (Telethon Institute of Genetics and Medicine), Naples, Italy (A.A.); Department of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, Bristol, United Kingdom (Z.D., P.M.); Department of Biochemistry and Molecular Pharmacology IRCCS Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy (L.S., P.B.); National Research Council, Institute of Experimental Endocrinology and Oncology (IEOS), Naples, Italy (G.C.); Institute of Clinical Molecular Biology, Christian-Albrechts University and the Schleswig-Holstein University Hospital, Kiel, Germany (S.S., A.N.); Geriatrics Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA (T.P.); Dipartimento di Fisiologia e Farmacologia, Sapienza Università di Roma, Rome, Italy (S.F.); and Dipartimento di Medicina e Chirurgia, Università degli Studi di Salerno, 84081 Baronissi (SA), Italy (C.V., A.A.P.)
| | - Gaetano Calì
- From the National Research Council, Institute for Biomedical Technologies, Segrate (MI), Italy (F.V., C.C.S., A.F.); IRCCS Neuromed, Department of Vascular Physiopathology, Pozzilli (IS), Italy (A.C., A.D., M.M., M.A., S.F., C.V.); Department of Industrial and Information Engineering, University of Pavia, Pavia, Italy (A. Malovini, F.M., R.B.); IRCCS Multimedica, Cardiovascular Department, Milan, Italy (A. Maciąg, G.S., E.S., A.A.P.); Department of Translational Medicine, "Federico II" University, Naples, Italy (A.A.); TIGEM (Telethon Institute of Genetics and Medicine), Naples, Italy (A.A.); Department of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, Bristol, United Kingdom (Z.D., P.M.); Department of Biochemistry and Molecular Pharmacology IRCCS Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy (L.S., P.B.); National Research Council, Institute of Experimental Endocrinology and Oncology (IEOS), Naples, Italy (G.C.); Institute of Clinical Molecular Biology, Christian-Albrechts University and the Schleswig-Holstein University Hospital, Kiel, Germany (S.S., A.N.); Geriatrics Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA (T.P.); Dipartimento di Fisiologia e Farmacologia, Sapienza Università di Roma, Rome, Italy (S.F.); and Dipartimento di Medicina e Chirurgia, Università degli Studi di Salerno, 84081 Baronissi (SA), Italy (C.V., A.A.P.)
| | - Stefan Schreiber
- From the National Research Council, Institute for Biomedical Technologies, Segrate (MI), Italy (F.V., C.C.S., A.F.); IRCCS Neuromed, Department of Vascular Physiopathology, Pozzilli (IS), Italy (A.C., A.D., M.M., M.A., S.F., C.V.); Department of Industrial and Information Engineering, University of Pavia, Pavia, Italy (A. Malovini, F.M., R.B.); IRCCS Multimedica, Cardiovascular Department, Milan, Italy (A. Maciąg, G.S., E.S., A.A.P.); Department of Translational Medicine, "Federico II" University, Naples, Italy (A.A.); TIGEM (Telethon Institute of Genetics and Medicine), Naples, Italy (A.A.); Department of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, Bristol, United Kingdom (Z.D., P.M.); Department of Biochemistry and Molecular Pharmacology IRCCS Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy (L.S., P.B.); National Research Council, Institute of Experimental Endocrinology and Oncology (IEOS), Naples, Italy (G.C.); Institute of Clinical Molecular Biology, Christian-Albrechts University and the Schleswig-Holstein University Hospital, Kiel, Germany (S.S., A.N.); Geriatrics Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA (T.P.); Dipartimento di Fisiologia e Farmacologia, Sapienza Università di Roma, Rome, Italy (S.F.); and Dipartimento di Medicina e Chirurgia, Università degli Studi di Salerno, 84081 Baronissi (SA), Italy (C.V., A.A.P.)
| | - Thomas Perls
- From the National Research Council, Institute for Biomedical Technologies, Segrate (MI), Italy (F.V., C.C.S., A.F.); IRCCS Neuromed, Department of Vascular Physiopathology, Pozzilli (IS), Italy (A.C., A.D., M.M., M.A., S.F., C.V.); Department of Industrial and Information Engineering, University of Pavia, Pavia, Italy (A. Malovini, F.M., R.B.); IRCCS Multimedica, Cardiovascular Department, Milan, Italy (A. Maciąg, G.S., E.S., A.A.P.); Department of Translational Medicine, "Federico II" University, Naples, Italy (A.A.); TIGEM (Telethon Institute of Genetics and Medicine), Naples, Italy (A.A.); Department of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, Bristol, United Kingdom (Z.D., P.M.); Department of Biochemistry and Molecular Pharmacology IRCCS Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy (L.S., P.B.); National Research Council, Institute of Experimental Endocrinology and Oncology (IEOS), Naples, Italy (G.C.); Institute of Clinical Molecular Biology, Christian-Albrechts University and the Schleswig-Holstein University Hospital, Kiel, Germany (S.S., A.N.); Geriatrics Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA (T.P.); Dipartimento di Fisiologia e Farmacologia, Sapienza Università di Roma, Rome, Italy (S.F.); and Dipartimento di Medicina e Chirurgia, Università degli Studi di Salerno, 84081 Baronissi (SA), Italy (C.V., A.A.P.)
| | - Sergio Fucile
- From the National Research Council, Institute for Biomedical Technologies, Segrate (MI), Italy (F.V., C.C.S., A.F.); IRCCS Neuromed, Department of Vascular Physiopathology, Pozzilli (IS), Italy (A.C., A.D., M.M., M.A., S.F., C.V.); Department of Industrial and Information Engineering, University of Pavia, Pavia, Italy (A. Malovini, F.M., R.B.); IRCCS Multimedica, Cardiovascular Department, Milan, Italy (A. Maciąg, G.S., E.S., A.A.P.); Department of Translational Medicine, "Federico II" University, Naples, Italy (A.A.); TIGEM (Telethon Institute of Genetics and Medicine), Naples, Italy (A.A.); Department of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, Bristol, United Kingdom (Z.D., P.M.); Department of Biochemistry and Molecular Pharmacology IRCCS Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy (L.S., P.B.); National Research Council, Institute of Experimental Endocrinology and Oncology (IEOS), Naples, Italy (G.C.); Institute of Clinical Molecular Biology, Christian-Albrechts University and the Schleswig-Holstein University Hospital, Kiel, Germany (S.S., A.N.); Geriatrics Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA (T.P.); Dipartimento di Fisiologia e Farmacologia, Sapienza Università di Roma, Rome, Italy (S.F.); and Dipartimento di Medicina e Chirurgia, Università degli Studi di Salerno, 84081 Baronissi (SA), Italy (C.V., A.A.P.)
| | - Francesca Mulas
- From the National Research Council, Institute for Biomedical Technologies, Segrate (MI), Italy (F.V., C.C.S., A.F.); IRCCS Neuromed, Department of Vascular Physiopathology, Pozzilli (IS), Italy (A.C., A.D., M.M., M.A., S.F., C.V.); Department of Industrial and Information Engineering, University of Pavia, Pavia, Italy (A. Malovini, F.M., R.B.); IRCCS Multimedica, Cardiovascular Department, Milan, Italy (A. Maciąg, G.S., E.S., A.A.P.); Department of Translational Medicine, "Federico II" University, Naples, Italy (A.A.); TIGEM (Telethon Institute of Genetics and Medicine), Naples, Italy (A.A.); Department of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, Bristol, United Kingdom (Z.D., P.M.); Department of Biochemistry and Molecular Pharmacology IRCCS Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy (L.S., P.B.); National Research Council, Institute of Experimental Endocrinology and Oncology (IEOS), Naples, Italy (G.C.); Institute of Clinical Molecular Biology, Christian-Albrechts University and the Schleswig-Holstein University Hospital, Kiel, Germany (S.S., A.N.); Geriatrics Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA (T.P.); Dipartimento di Fisiologia e Farmacologia, Sapienza Università di Roma, Rome, Italy (S.F.); and Dipartimento di Medicina e Chirurgia, Università degli Studi di Salerno, 84081 Baronissi (SA), Italy (C.V., A.A.P.)
| | - Almut Nebel
- From the National Research Council, Institute for Biomedical Technologies, Segrate (MI), Italy (F.V., C.C.S., A.F.); IRCCS Neuromed, Department of Vascular Physiopathology, Pozzilli (IS), Italy (A.C., A.D., M.M., M.A., S.F., C.V.); Department of Industrial and Information Engineering, University of Pavia, Pavia, Italy (A. Malovini, F.M., R.B.); IRCCS Multimedica, Cardiovascular Department, Milan, Italy (A. Maciąg, G.S., E.S., A.A.P.); Department of Translational Medicine, "Federico II" University, Naples, Italy (A.A.); TIGEM (Telethon Institute of Genetics and Medicine), Naples, Italy (A.A.); Department of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, Bristol, United Kingdom (Z.D., P.M.); Department of Biochemistry and Molecular Pharmacology IRCCS Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy (L.S., P.B.); National Research Council, Institute of Experimental Endocrinology and Oncology (IEOS), Naples, Italy (G.C.); Institute of Clinical Molecular Biology, Christian-Albrechts University and the Schleswig-Holstein University Hospital, Kiel, Germany (S.S., A.N.); Geriatrics Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA (T.P.); Dipartimento di Fisiologia e Farmacologia, Sapienza Università di Roma, Rome, Italy (S.F.); and Dipartimento di Medicina e Chirurgia, Università degli Studi di Salerno, 84081 Baronissi (SA), Italy (C.V., A.A.P.)
| | - Riccardo Bellazzi
- From the National Research Council, Institute for Biomedical Technologies, Segrate (MI), Italy (F.V., C.C.S., A.F.); IRCCS Neuromed, Department of Vascular Physiopathology, Pozzilli (IS), Italy (A.C., A.D., M.M., M.A., S.F., C.V.); Department of Industrial and Information Engineering, University of Pavia, Pavia, Italy (A. Malovini, F.M., R.B.); IRCCS Multimedica, Cardiovascular Department, Milan, Italy (A. Maciąg, G.S., E.S., A.A.P.); Department of Translational Medicine, "Federico II" University, Naples, Italy (A.A.); TIGEM (Telethon Institute of Genetics and Medicine), Naples, Italy (A.A.); Department of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, Bristol, United Kingdom (Z.D., P.M.); Department of Biochemistry and Molecular Pharmacology IRCCS Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy (L.S., P.B.); National Research Council, Institute of Experimental Endocrinology and Oncology (IEOS), Naples, Italy (G.C.); Institute of Clinical Molecular Biology, Christian-Albrechts University and the Schleswig-Holstein University Hospital, Kiel, Germany (S.S., A.N.); Geriatrics Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA (T.P.); Dipartimento di Fisiologia e Farmacologia, Sapienza Università di Roma, Rome, Italy (S.F.); and Dipartimento di Medicina e Chirurgia, Università degli Studi di Salerno, 84081 Baronissi (SA), Italy (C.V., A.A.P.)
| | - Paolo Madeddu
- From the National Research Council, Institute for Biomedical Technologies, Segrate (MI), Italy (F.V., C.C.S., A.F.); IRCCS Neuromed, Department of Vascular Physiopathology, Pozzilli (IS), Italy (A.C., A.D., M.M., M.A., S.F., C.V.); Department of Industrial and Information Engineering, University of Pavia, Pavia, Italy (A. Malovini, F.M., R.B.); IRCCS Multimedica, Cardiovascular Department, Milan, Italy (A. Maciąg, G.S., E.S., A.A.P.); Department of Translational Medicine, "Federico II" University, Naples, Italy (A.A.); TIGEM (Telethon Institute of Genetics and Medicine), Naples, Italy (A.A.); Department of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, Bristol, United Kingdom (Z.D., P.M.); Department of Biochemistry and Molecular Pharmacology IRCCS Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy (L.S., P.B.); National Research Council, Institute of Experimental Endocrinology and Oncology (IEOS), Naples, Italy (G.C.); Institute of Clinical Molecular Biology, Christian-Albrechts University and the Schleswig-Holstein University Hospital, Kiel, Germany (S.S., A.N.); Geriatrics Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA (T.P.); Dipartimento di Fisiologia e Farmacologia, Sapienza Università di Roma, Rome, Italy (S.F.); and Dipartimento di Medicina e Chirurgia, Università degli Studi di Salerno, 84081 Baronissi (SA), Italy (C.V., A.A.P.)
| | - Carmine Vecchione
- From the National Research Council, Institute for Biomedical Technologies, Segrate (MI), Italy (F.V., C.C.S., A.F.); IRCCS Neuromed, Department of Vascular Physiopathology, Pozzilli (IS), Italy (A.C., A.D., M.M., M.A., S.F., C.V.); Department of Industrial and Information Engineering, University of Pavia, Pavia, Italy (A. Malovini, F.M., R.B.); IRCCS Multimedica, Cardiovascular Department, Milan, Italy (A. Maciąg, G.S., E.S., A.A.P.); Department of Translational Medicine, "Federico II" University, Naples, Italy (A.A.); TIGEM (Telethon Institute of Genetics and Medicine), Naples, Italy (A.A.); Department of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, Bristol, United Kingdom (Z.D., P.M.); Department of Biochemistry and Molecular Pharmacology IRCCS Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy (L.S., P.B.); National Research Council, Institute of Experimental Endocrinology and Oncology (IEOS), Naples, Italy (G.C.); Institute of Clinical Molecular Biology, Christian-Albrechts University and the Schleswig-Holstein University Hospital, Kiel, Germany (S.S., A.N.); Geriatrics Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA (T.P.); Dipartimento di Fisiologia e Farmacologia, Sapienza Università di Roma, Rome, Italy (S.F.); and Dipartimento di Medicina e Chirurgia, Università degli Studi di Salerno, 84081 Baronissi (SA), Italy (C.V., A.A.P.)
| | - Annibale A Puca
- From the National Research Council, Institute for Biomedical Technologies, Segrate (MI), Italy (F.V., C.C.S., A.F.); IRCCS Neuromed, Department of Vascular Physiopathology, Pozzilli (IS), Italy (A.C., A.D., M.M., M.A., S.F., C.V.); Department of Industrial and Information Engineering, University of Pavia, Pavia, Italy (A. Malovini, F.M., R.B.); IRCCS Multimedica, Cardiovascular Department, Milan, Italy (A. Maciąg, G.S., E.S., A.A.P.); Department of Translational Medicine, "Federico II" University, Naples, Italy (A.A.); TIGEM (Telethon Institute of Genetics and Medicine), Naples, Italy (A.A.); Department of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, Bristol, United Kingdom (Z.D., P.M.); Department of Biochemistry and Molecular Pharmacology IRCCS Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy (L.S., P.B.); National Research Council, Institute of Experimental Endocrinology and Oncology (IEOS), Naples, Italy (G.C.); Institute of Clinical Molecular Biology, Christian-Albrechts University and the Schleswig-Holstein University Hospital, Kiel, Germany (S.S., A.N.); Geriatrics Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA (T.P.); Dipartimento di Fisiologia e Farmacologia, Sapienza Università di Roma, Rome, Italy (S.F.); and Dipartimento di Medicina e Chirurgia, Università degli Studi di Salerno, 84081 Baronissi (SA), Italy (C.V., A.A.P.).
| |
Collapse
|
29
|
Siti HN, Kamisah Y, Kamsiah J. The role of oxidative stress, antioxidants and vascular inflammation in cardiovascular disease (a review). Vascul Pharmacol 2015; 71:40-56. [PMID: 25869516 DOI: 10.1016/j.vph.2015.03.005] [Citation(s) in RCA: 587] [Impact Index Per Article: 65.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 02/28/2015] [Accepted: 03/09/2015] [Indexed: 12/31/2022]
Abstract
The concept of mild chronic vascular inflammation as part of the pathophysiology of cardiovascular disease, most importantly hypertension and atherosclerosis, has been well accepted. Indeed there are links between vascular inflammation, endothelial dysfunction and oxidative stress. However, there are still gaps in our understanding regarding this matter that might be the cause behind disappointing results of antioxidant therapy for cardiovascular risk factors in large-scale long-term randomised controlled trials. Apart from the limitations of our knowledge, limitations in methodology and assessment of the body's endogenous and exogenous oxidant-antioxidant status are a serious handicap. The pleiotropic effects of antioxidant and anti-inflammation that are shown by some well-established antihypertensive agents and statins partly support the idea of using antioxidants in vascular diseases as still relevant. This review aims to provide an overview of the links between oxidative stress, vascular inflammation, endothelial dysfunction and cardiovascular risk factors, importantly focusing on blood pressure regulation and atherosclerosis. In view of the potential benefits of antioxidants, this review will also examine the proposed role of vitamin C, vitamin E and polyphenols in cardiovascular diseases as well as the success or failure of antioxidant therapy for cardiovascular diseases in clinical trials.
Collapse
Affiliation(s)
- Hawa N Siti
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia; Department of Basic Medical Sciences, Faculty of Medicine, Universiti Sultan Zainal Abidin, Terengganu, Malaysia
| | - Y Kamisah
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - J Kamsiah
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia.
| |
Collapse
|
30
|
Abstract
Intrarenal autoregulatory mechanisms maintain renal blood flow (RBF) and glomerular filtration rate (GFR) independent of renal perfusion pressure (RPP) over a defined range (80-180 mmHg). Such autoregulation is mediated largely by the myogenic and the macula densa-tubuloglomerular feedback (MD-TGF) responses that regulate preglomerular vasomotor tone primarily of the afferent arteriole. Differences in response times allow separation of these mechanisms in the time and frequency domains. Mechanotransduction initiating the myogenic response requires a sensing mechanism activated by stretch of vascular smooth muscle cells (VSMCs) and coupled to intracellular signaling pathways eliciting plasma membrane depolarization and a rise in cytosolic free calcium concentration ([Ca(2+)]i). Proposed mechanosensors include epithelial sodium channels (ENaC), integrins, and/or transient receptor potential (TRP) channels. Increased [Ca(2+)]i occurs predominantly by Ca(2+) influx through L-type voltage-operated Ca(2+) channels (VOCC). Increased [Ca(2+)]i activates inositol trisphosphate receptors (IP3R) and ryanodine receptors (RyR) to mobilize Ca(2+) from sarcoplasmic reticular stores. Myogenic vasoconstriction is sustained by increased Ca(2+) sensitivity, mediated by protein kinase C and Rho/Rho-kinase that favors a positive balance between myosin light-chain kinase and phosphatase. Increased RPP activates MD-TGF by transducing a signal of epithelial MD salt reabsorption to adjust afferent arteriolar vasoconstriction. A combination of vascular and tubular mechanisms, novel to the kidney, provides for high autoregulatory efficiency that maintains RBF and GFR, stabilizes sodium excretion, and buffers transmission of RPP to sensitive glomerular capillaries, thereby protecting against hypertensive barotrauma. A unique aspect of the myogenic response in the renal vasculature is modulation of its strength and speed by the MD-TGF and by a connecting tubule glomerular feedback (CT-GF) mechanism. Reactive oxygen species and nitric oxide are modulators of myogenic and MD-TGF mechanisms. Attenuated renal autoregulation contributes to renal damage in many, but not all, models of renal, diabetic, and hypertensive diseases. This review provides a summary of our current knowledge regarding underlying mechanisms enabling renal autoregulation in health and disease and methods used for its study.
Collapse
Affiliation(s)
- Mattias Carlström
- Department of Medicine, Division of Nephrology and Hypertension and Hypertension, Kidney and Vascular Research Center, Georgetown University, Washington, District of Columbia; Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; and Department of Cell Biology and Physiology, UNC Kidney Center, and McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Christopher S Wilcox
- Department of Medicine, Division of Nephrology and Hypertension and Hypertension, Kidney and Vascular Research Center, Georgetown University, Washington, District of Columbia; Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; and Department of Cell Biology and Physiology, UNC Kidney Center, and McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - William J Arendshorst
- Department of Medicine, Division of Nephrology and Hypertension and Hypertension, Kidney and Vascular Research Center, Georgetown University, Washington, District of Columbia; Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; and Department of Cell Biology and Physiology, UNC Kidney Center, and McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
31
|
The angiogenic factor PlGF mediates a neuroimmune interaction in the spleen to allow the onset of hypertension. Immunity 2014; 41:737-52. [PMID: 25517614 DOI: 10.1016/j.immuni.2014.11.002] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 09/23/2014] [Indexed: 12/12/2022]
Abstract
Hypertension is a health problem affecting over 1 billion people worldwide. How the immune system gets activated under hypertensive stimuli to contribute to blood pressure elevation is a fascinating enigma. Here we showed a splenic role for placental growth factor (PlGF), which accounts for the onset of hypertension, through immune system modulation. PlGF repressed the expression of the protein Timp3 (tissue inhibitor of metalloproteinases 3), through the transcriptional Sirt1-p53 axis. Timp3 repression allowed costimulation of T cells and their deployment toward classical organs involved in hypertension. We showed that the spleen is an essential organ for the development of hypertension through a noradrenergic drive mediated by the celiac ganglion efferent. Overall, we demonstrate that PlGF mediates the neuroimmune interaction in the spleen, organizing a unique and nonredundant response that allows the onset of hypertension.
Collapse
|
32
|
|
33
|
Bhattachariya A, Dahan D, Turczyńska KM, Swärd K, Hellstrand P, Albinsson S. Expression of microRNAs is essential for arterial myogenic tone and pressure-induced activation of the PI3-kinase/Akt pathway. Cardiovasc Res 2013; 101:288-96. [PMID: 24233972 DOI: 10.1093/cvr/cvt253] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AIMS The myogenic response is the intrinsic ability of small arteries to constrict in response to increased intraluminal pressure. Although microRNAs have been shown to play a role in vascular smooth muscle function, their importance in the regulation of the myogenic response is not known. In this study, we investigate the role of microRNAs in the regulation of myogenic tone by using smooth muscle-specific and tamoxifen-inducible deletion of the endonuclease Dicer in mice. METHODS AND RESULTS In order to avoid effects of Dicer deletion on smooth muscle differentiation and growth, we used an early time point (5 weeks) after the tamoxifen-induction of Dicer knockout (KO). At this time point, we found that myogenic tone was completely absent in the mesenteric arteries of Dicer KO mice. This was associated with a reduced pressure-induced Akt-phosphorylation, possibly via increased phosphatase and tensin homologue (PTEN) expression, which was found to be a target of miR-26a. Furthermore, loss of myogenic tone was associated with a decreased depolarization-induced calcium influx, and was restored by the L-type channel agonist Bay K 8644 or by transient stimulation with angiotensin II (Ang II). The effect of Ang II was dependent on AT1-receptors and activation of the PI3-kinase/Akt pathway. CONCLUSION In this study we have identified novel mechanisms that regulate myogenic tone in resistance arteries, which involves microRNA-dependent control of PI3-kinase/Akt signalling and L-type calcium influx. Furthermore, we have demonstrated that transient stimulation by Ang II can have long-lasting effects by potentiating myogenic tone.
Collapse
Affiliation(s)
- Anirban Bhattachariya
- Department of Experimental Medical Sciences, Lund University, BMC D12, SE-221 84 Lund, Sweden
| | | | | | | | | | | |
Collapse
|
34
|
Gebremedhin D, Terashvili M, Wickramasekera N, Zhang DX, Rau N, Miura H, Harder DR. Redox signaling via oxidative inactivation of PTEN modulates pressure-dependent myogenic tone in rat middle cerebral arteries. PLoS One 2013; 8:e68498. [PMID: 23861911 PMCID: PMC3702596 DOI: 10.1371/journal.pone.0068498] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Accepted: 05/30/2013] [Indexed: 11/18/2022] Open
Abstract
The present study examined the level of generation of reactive oxygen species (ROS) and roles of inactivation of the phosphatase PTEN and the PI3K/Akt signaling pathway in response to an increase in intramural pressure-induced myogenic cerebral arterial constriction. Step increases in intraluminal pressure of cannulated cerebral arteries induced myogenic constriction and concomitant formation of superoxide (O2 (.-)) and its dismutation product hydrogen peroxide (H2O2) as determined by fluorescent HPLC analysis, microscopic analysis of intensity of dihydroethidium fluorescence and attenuation of pressure-induced myogenic constriction by pretreatment with the ROS scavenger 4,hydroxyl-2,2,6,6-tetramethylpiperidine1-oxyl (tempol) or Mito-tempol or MitoQ in the presence or absence of PEG-catalase. An increase in intraluminal pressure induced oxidation of PTEN and activation of Akt. Pharmacological inhibition of endogenous PTEN activity potentiated pressure-dependent myogenic constriction and caused a reduction in NPo of a 238 pS arterial KCa channel current and an increase in [Ca(2+)]i level in freshly isolated cerebral arterial muscle cells (CAMCs), responses that were attenuated by Inhibition of the PI3K/Akt pathway. These findings demonstrate an increase in intraluminal pressure induced increase in ROS production triggered redox-sensitive signaling mechanism emanating from the cross-talk between oxidative inactivation of PTEN and activation of the PI3K/Akt signaling pathway that involves in the regulation of pressure-dependent myogenic cerebral arterial constriction.
Collapse
Affiliation(s)
- Debebe Gebremedhin
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
| | | | | | | | | | | | | |
Collapse
|
35
|
Zhang Z, Payne K, Cao C, Pallone TL. Mural propagation of descending vasa recta responses to mechanical stimulation. Am J Physiol Renal Physiol 2013; 305:F286-94. [PMID: 23698119 DOI: 10.1152/ajprenal.00220.2013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
To investigate the responses of descending vasa recta (DVR) to deformation of the abluminal surface, we devised an automated method that controls duration and frequency of stimulation by utilizing a stream of buffer from a micropipette. During stimulation at one end of the vessel, fluorescent responses from fluo4 or bis[1,3-dibutylbarbituric acid-(5)] trimethineoxonol [DiBAC₄(3)], indicating cytoplasmic calcium ([Ca²⁺]CYT) or membrane potential, respectively, were recorded from distant cells. Alternately, membrane potential was recorded from DVR pericytes by nystatin whole cell patch-clamp. Mechanical stimulation elicited reversible [Ca²⁺)]CYT responses that increased with frequency. Individual pericyte responses along the vessel were initiated within a fraction of a second of one another. Those responses were inhibited by gap junction blockade with 18 β-glycyrrhetinic acid (100 μM) or phosphoinositide 3 kinase inhibition with 2-morpholin-4-yl-8-phenylchromen-4-one (50 μM). [Ca²⁺]CYT responses were blocked by removal of extracellular Ca²⁺ or L-type voltage-gated channel blockade with nifedipine (10 μM). At concentrations selective for the T-type channel blockade, mibefradil (100 nM) was ineffective. During mechanostimulation, pericytes rapidly depolarized, as documented with either DiBAC4(3) fluorescence or patch-clamp recording. Single stimuli yielded depolarizations of 22.5 ± 2.2 mV while repetitive stimuli at 0.1 Hz depolarized pericytes by 44.2 ± 4.0 mV. We conclude that DVR are mechanosensitive and that rapid transmission of signals along the vessel axis requires participation of gap junctions, L-type Ca²⁺ channels, and pericyte depolarization.
Collapse
Affiliation(s)
- Zhong Zhang
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | | | | |
Collapse
|
36
|
Turczyńska KM, Bhattachariya A, Säll J, Göransson O, Swärd K, Hellstrand P, Albinsson S. Stretch-sensitive down-regulation of the miR-144/451 cluster in vascular smooth muscle and its role in AMP-activated protein kinase signaling. PLoS One 2013; 8:e65135. [PMID: 23705032 PMCID: PMC3660603 DOI: 10.1371/journal.pone.0065135] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Accepted: 04/22/2013] [Indexed: 11/25/2022] Open
Abstract
Vascular smooth muscle cells are constantly exposed to mechanical force by the blood pressure, which is thought to regulate smooth muscle growth, differentiation and contractile function. We have previously shown that the expression of microRNAs (miRNAs), small non-coding RNAs, is essential for regulation of smooth muscle phenotype including stretch-dependent contractile differentiation. In this study, we have investigated the effect of mechanical stretch on miRNA expression and the role of stretch-sensitive miRNAs for intracellular signaling in smooth muscle. MiRNA array analysis, comparing miRNA levels in stretched versus non-stretched portal veins, revealed a dramatic decrease in the miR-144/451 cluster level. Because this miRNA cluster is predicted to target AMPK pathway components, we next examined activation of this pathway. Diminished miR-144/451 expression was inversely correlated with increased phosphorylation of AMPKα at Thr172 in stretched portal vein. Similar to the effect of stretch, contractile differentiation could be induced in non-stretched portal veins by the AMPK activator, AICAR. Transfection with miR-144/451 mimics reduced the protein expression level of mediators in the AMPK pathway including MO25α, AMPK and ACC. This effect also decreased AICAR-induced activation of the AMPK signaling pathway. In conclusion, our results suggest that stretch-induced activation of AMPK in vascular smooth muscle is in part regulated by reduced levels of miR-144/451 and that this effect may play a role in promoting contractile differentiation of smooth muscle cells.
Collapse
|
37
|
Abstract
PI3Ks are signaling enzymes engaged by different types of membrane receptors and activated in cardiovascular diseases such as hypertension, atherosclerosis, thrombosis and heart failure. Studies performed on genetically modified animals have provided proof-of-concept that general or isoform-specific blockade of these enzymes can modify disease development and progression. Hence, therapeutic inhibition of PI3Ks with novel pharmacological compounds constitutes a promising area of drug development. In particular, inhibitors of PI3Ks have the potential to reduce blood pressure, restrain the development of atherosclerosis and/or stabilize atherosclerotic plaques, blunt platelet aggregation, prevent left ventricular remodeling and preserve myocardial contractility in heart failure. This review summarizes the rationale of PI3K inhibition in the most prevalent cardiovascular diseases, and the available data on the therapeutic effects of PI3K inhibitors in their preclinical models. Implications for future drug development and human therapy are also discussed.
Collapse
|
38
|
Reversible inhibition of vasoconstriction by thiazolidinediones related to PI3K/Akt inhibition in vascular smooth muscle cells. Biochem Pharmacol 2013. [DOI: 10.1016/j.bcp.2012.11.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
39
|
Tyagi S, Sharma S, Budhiraja RD. Effect of phosphatidylinositol 3-kinase-γ inhibitor CAY10505 in hypertension, and its associated vascular endothelium dysfunction in rats. Can J Physiol Pharmacol 2012; 90:881-5. [DOI: 10.1139/y2012-089] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This study has been designed to investigate the role of phosphatidyl-inositol 3-kinase-γ (PI3Kγ) in deoxycorticosterone acetate salt (DOCA) hypertension induced vascular endothelium dysfunction. Wistar rats were uninephrectomised and DOCA (40 mg·(kg body mass)−1, subcutaneous injection) was administered twice weekly for 6 weeks to produce hypertension. Rats with mean arterial blood pressure ≥ 140 mm Hg (1 mm Hg = 133.322 Pa) were selected as hypertensive. Vascular endothelium dysfunction was assessed in terms of attenuation of acetylcholine-induced endothelium-dependent relaxation (isolated aortic ring preparation), decrease in serum nitrate and (or) nitrite level, as well as reduced level of glutathione and disruption of integrity of vascular endothelium (histopathology). Five weeks of DOCA administration were followed by 7 days of daily administration of PI3Kγ inhibitor (5-[[5-(4-fluorophenyl)-2-furanyl]methylene]-2,4-thiazolidinedione (CAY10505), 0.6 mg·kg−1, per os (p.o.)), atorvastatin (30 mg·kg−1, p.o.), and losartan (25 mg·kg−1, p.o.) (positive control of hypertension), which significantly improved acetylcholine-induced endothelium dependent relaxation, serum nitrate and (or) nitrite level, glutathione level, and the vascular endothelial lining in hypertensive rats.Therefore, it may be concluded that CAY10505, a specific inhibitor of PI3Kγ, improves hypertension-associated vascular endothelial dysfunction. Thus, inhibition of PI3Kγ might be a useful approach in the therapeutics of vascular endothelium dysfunction.
Collapse
Affiliation(s)
- Sandeep Tyagi
- Department of Pharmacology, I.S.F. College of Pharmacy, G.T. Road, Moga-142 001 (Punjab), India
| | - Saurabh Sharma
- Department of Pharmacology, I.S.F. College of Pharmacy, G.T. Road, Moga-142 001 (Punjab), India
| | - Ramji Dass Budhiraja
- Department of Pharmacology, I.S.F. College of Pharmacy, G.T. Road, Moga-142 001 (Punjab), India
| |
Collapse
|
40
|
Carnevale D, Lembo G. PI3K in hypertension: a novel therapeutic target controlling vascular myogenic tone and target organ damage. Cardiovasc Res 2012; 95:403-8. [DOI: 10.1093/cvr/cvs166] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
|