1
|
Munteanu C, Popescu C, Vlădulescu-Trandafir AI, Onose G. Signaling Paradigms of H 2S-Induced Vasodilation: A Comprehensive Review. Antioxidants (Basel) 2024; 13:1158. [PMID: 39456412 PMCID: PMC11505308 DOI: 10.3390/antiox13101158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/21/2024] [Accepted: 09/23/2024] [Indexed: 10/28/2024] Open
Abstract
Hydrogen sulfide (H2S), a gas traditionally considered toxic, is now recognized as a vital endogenous signaling molecule with a complex physiology. This comprehensive study encompasses a systematic literature review that explores the intricate mechanisms underlying H2S-induced vasodilation. The vasodilatory effects of H2S are primarily mediated by activating ATP-sensitive potassium (K_ATP) channels, leading to membrane hyperpolarization and subsequent relaxation of vascular smooth muscle cells (VSMCs). Additionally, H2S inhibits L-type calcium channels, reducing calcium influx and diminishing VSMC contraction. Beyond ion channel modulation, H2S profoundly impacts cyclic nucleotide signaling pathways. It stimulates soluble guanylyl cyclase (sGC), increasing the production of cyclic guanosine monophosphate (cGMP). Elevated cGMP levels activate protein kinase G (PKG), which phosphorylates downstream targets like vasodilator-stimulated phosphoprotein (VASP) and promotes smooth muscle relaxation. The synergy between H2S and nitric oxide (NO) signaling further amplifies vasodilation. H2S enhances NO bioavailability by inhibiting its degradation and stimulating endothelial nitric oxide synthase (eNOS) activity, increasing cGMP levels and potent vasodilatory responses. Protein sulfhydration, a post-translational modification, plays a crucial role in cell signaling. H2S S-sulfurates oxidized cysteine residues, while polysulfides (H2Sn) are responsible for S-sulfurating reduced cysteine residues. Sulfhydration of key proteins like K_ATP channels and sGC enhances their activity, contributing to the overall vasodilatory effect. Furthermore, H2S interaction with endothelium-derived hyperpolarizing factor (EDHF) pathways adds another layer to its vasodilatory mechanism. By enhancing EDHF activity, H2S facilitates the hyperpolarization and relaxation of VSMCs through gap junctions between endothelial cells and VSMCs. Recent findings suggest that H2S can also modulate transient receptor potential (TRP) channels, particularly TRPV4 channels, in endothelial cells. Activating these channels by H2S promotes calcium entry, stimulating the production of vasodilatory agents like NO and prostacyclin, thereby regulating vascular tone. The comprehensive understanding of H2S-induced vasodilation mechanisms highlights its therapeutic potential. The multifaceted approach of H2S in modulating vascular tone presents a promising strategy for developing novel treatments for hypertension, ischemic conditions, and other vascular disorders. The interaction of H2S with ion channels, cyclic nucleotide signaling, NO pathways, ROS (Reactive Oxygen Species) scavenging, protein sulfhydration, and EDHF underscores its complexity and therapeutic relevance. In conclusion, the intricate signaling paradigms of H2S-induced vasodilation offer valuable insights into its physiological role and therapeutic potential, promising innovative approaches for managing various vascular diseases through the modulation of vascular tone.
Collapse
Affiliation(s)
- Constantin Munteanu
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa” Iași, 700454 Iași, Romania
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (A.-I.V.-T.); (G.O.)
| | - Cristina Popescu
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (A.-I.V.-T.); (G.O.)
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania
| | - Andreea-Iulia Vlădulescu-Trandafir
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (A.-I.V.-T.); (G.O.)
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania
| | - Gelu Onose
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (A.-I.V.-T.); (G.O.)
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania
| |
Collapse
|
2
|
Milewski K, Orzeł-Gajowik K, Zielińska M. Mitochondrial Changes in Rat Brain Endothelial Cells Associated with Hepatic Encephalopathy: Relation to the Blood-Brain Barrier Dysfunction. Neurochem Res 2024; 49:1489-1504. [PMID: 35917006 PMCID: PMC11106209 DOI: 10.1007/s11064-022-03698-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 02/17/2022] [Accepted: 07/14/2022] [Indexed: 12/06/2022]
Abstract
The mechanisms underlying cerebral vascular dysfunction and edema during hepatic encephalopathy (HE) are unclear. Blood-brain barrier (BBB) impairment, resulting from increased vascular permeability, has been reported in acute and chronic HE. Mitochondrial dysfunction is a well-documented result of HE mainly affecting astrocytes, but much less so in the BBB-forming endothelial cells. Here we review literature reports and own experimental data obtained in HE models emphasizing alterations in mitochondrial dynamics and function as a possible contributor to the status of brain endothelial cell mitochondria in HE. Own studies on the expression of the mitochondrial fusion-fission controlling genes rendered HE animal model-dependent effects: increase of mitochondrial fusion controlling genes opa1, mfn1 in cerebral vessels in ammonium acetate-induced hyperammonemia, but a decrease of the two former genes and increase of fis1 in vessels in thioacetamide-induced HE. In endothelial cell line (RBE4) after 24 h ammonia and/or TNFα treatment, conditions mimicking crucial aspects of HE in vivo, we observed altered expression of mitochondrial fission/fusion genes: a decrease of opa1, mfn1, and, increase of the fission related fis1 gene. The effect in vitro was paralleled by the generation of reactive oxygen species, decreased total antioxidant capacity, decreased mitochondrial membrane potential, as well as increased permeability of RBE4 cell monolayer to fluorescein isothiocyanate dextran. Electron microscopy documented enlarged mitochondria in the brain endothelial cells of rats in both in vivo models. Collectively, the here observed alterations of cerebral endothelial mitochondria are indicative of their fission, and decreased potential of endothelial mitochondria are likely to contribute to BBB dysfunction in HE.
Collapse
Affiliation(s)
- Krzysztof Milewski
- Department of Neurotoxicology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego St. 5, 02-106, Warsaw, Poland.
| | - Karolina Orzeł-Gajowik
- Department of Neurotoxicology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego St. 5, 02-106, Warsaw, Poland
| | - Magdalena Zielińska
- Department of Neurotoxicology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego St. 5, 02-106, Warsaw, Poland.
| |
Collapse
|
3
|
Morton KS, Wahl AK, Meyer JN. The effect of common paralytic agents used for fluorescence imaging on redox tone and ATP levels in Caenorhabditis elegans. PLoS One 2024; 19:e0292415. [PMID: 38669260 PMCID: PMC11051652 DOI: 10.1371/journal.pone.0292415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 03/30/2024] [Indexed: 04/28/2024] Open
Abstract
One aspect of Caenorhabditis elegans that makes it a highly valuable model organism is the ease of use of in vivo genetic reporters, facilitated by its transparent cuticle and highly tractable genetics. Despite the rapid advancement of these technologies, worms must be paralyzed for most imaging applications, and few investigations have characterized the impacts of common chemical anesthetic methods on the parameters measured, in particular biochemical measurements such as cellular energetics and redox tone. Using two dynamic reporters, QUEEN-2m for relative ATP levels and reduction-oxidation sensitive GFP (roGFP) for redox tone, we assess the impact of commonly used chemical paralytics. We report that no chemical anesthetic is entirely effective at doses required for full paralysis without altering redox tone or ATP levels, and that anesthetic use alters the detected outcome of rotenone exposure on relative ATP levels and redox tone. We also assess the use of cold shock, commonly used in combination with physical restraint methods, and find that cold shock does not alter either ATP levels or redox tone. In addition to informing which paralytics are most appropriate for research in these topics, we highlight the need for tailoring the use of anesthetics to different endpoints and experimental questions. Further, we reinforce the need for developing less disruptive paralytic methods for optimal imaging of dynamic in vivo reporters.
Collapse
Affiliation(s)
- Katherine S. Morton
- Nicholas School of Environment, Duke University, Durham, North Carolina, United States of America
| | - Ashlyn K. Wahl
- Nicholas School of Environment, Duke University, Durham, North Carolina, United States of America
| | - Joel N. Meyer
- Nicholas School of Environment, Duke University, Durham, North Carolina, United States of America
| |
Collapse
|
4
|
Guven C, Taskin E, Aydın Ö, Kaya ST, Sevgiler Y. Diazoxide attenuates DOX-induced cardiotoxicity in cultured rat myocytes. Biotech Histochem 2024; 99:113-124. [PMID: 38439686 DOI: 10.1080/10520295.2024.2324368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024] Open
Abstract
Doxorubicin (DOX)-induced cardiotoxicity is a well known clinical problem, and many investigations have been made of its possible amelioration. We have investigated whether diazoxide (DIA), an agonist at mitochondrial ATP-sensitive potassium channels (mitoKATP), could reverse DOX-induced apoptotic myocardial cell loss, in cultured rat cardiomyocytes. The role of certain proteins in this pathway was also studied. The rat cardiomyocyte cell line (H9c2) was treated with DOX, and also co-treated with DOX and DIA, for 24 h. Distribution of actin filaments, mitochondrial membrane potential, superoxide dismutase (SOD) activity, total oxidant and antioxidant status (TOS and TAS, respectively), and some protein expressions, were assessed. DOX significantly decreased SOD activity, increased ERK1/2 protein levels, and depolarised the mitochondrial membrane, while DIA co-treatment inhibited such changes. DIA co-treatment ameliorated DOX-induced cytoskeletal changes via F-actin distribution and mitoKATP structure. Co-treatment also decreased ERK1/2 and cytochrome c protein levels. Cardiomyocyte loss due to oxidative stress-mediated apoptosis is a key event in DOX-induced cytotoxicity. DIA had protective effects on DOX-induced cardiotoxicity, via mitoKATP integrity, especially with elevated SUR2A levels; but also by a cascade including SOD/AMPK/ERK1/2. Therefore, DIA may be considered a candidate agent for protecting cardiomyocytes against DOX chemotherapy.
Collapse
Affiliation(s)
- Celal Guven
- Department of Biophysics, Faculty of Medicine, Adıyaman University, Adıyaman, Turkey
| | - Eylem Taskin
- Department of Physiology, Faculty of Medicine, Adıyaman University, Adıyaman, Turkey
| | - Özgül Aydın
- Department of Biology, Institute of Natural and Applied Sciences, Adıyaman University, Adıyaman, Turkey
| | - Salih Tunç Kaya
- Department of Biology, Faculty of Science and Letters, Düzce University, Düzce, Turkey
| | - Yusuf Sevgiler
- Department of Biology, Faculty of Science and Letters, Adıyaman University, Adıyaman, Turkey
| |
Collapse
|
5
|
Rasool GS, Al-Awadi SJ, Hussien AA, Al-Attar MM. Genetic variation of CYP2C9 gene and its correlation with cardiovascular disease risk factors. Mol Biol Rep 2024; 51:105. [PMID: 38227154 DOI: 10.1007/s11033-023-09151-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 12/13/2023] [Indexed: 01/17/2024]
Abstract
BACKGROUND The major enzyme that is responsible for Sulfonylureas (SUs) metabolism is hepatic cytochrome P-450 2C9 (CYP2C9). It is encoded by the polymorphic gene CYP2C9, which has many allelic variants, among those the CYP2C9*2 and CYP2C9*3 are the most common and clinically significant allelic variations. People with diabetes mellitus type 2 (T2DM) are more likely to develop cardiovascular disease (CVD), and their risk of dying from it is more than two times higher than that of people without the condition. The purpose of this study was to evaluate the association of genetic variations in the CYP2C9 gene with cardiovascular risk factors by investigating CYP2C9*1, *2, *3, *5, *11, and *13 allelic variants. METHODS AND RESULTS A total of 226 participants were enrolled in the current case-control study. Allele-specific amplification- PCR (ASA-PCR) was used to determine the allele of different variations and the results were confirmed by sequencing. The findings of this study showed the presence of the CYP2C9*2 allele in the T2DM group does not differ from its percentage in the control group. Also, CYP2C9*3 allele frequencies identified by Hardy-Weinberg equilibrium (HWE) analysis law were not significant, p = 0.6593 and 0.5828 in T2DM and control groups. There is no statistically significant difference between the control and diabetes groups involving the distribution of CYP2C9 alleles and CYP2C9*5, *11, and *13 polymorphisms were absent in the Iraqi population. No carrier for the CYP2C9*3 homozygous state was found in both groups. CONCLUSIONS According to these results T2DM patients with the CYP2C9*2 and *3 variants have an increased risk of developing hypertension.
Collapse
Affiliation(s)
- Ghada S Rasool
- Department of Anatomy, Faculty of Medicine, Ninevah University, Mosul, Iraq
| | - Salwa J Al-Awadi
- Department of Molecular and Medical Genetics Technologies, College of Biotechnology, Al-Nahrain University, Baghdad, Iraq
| | - Asmaa A Hussien
- Department of Molecular and Medical Genetics Technologies, College of Biotechnology, Al-Nahrain University, Baghdad, Iraq
| | - Marwa M Al-Attar
- Department of Biology, College of Science, Mustansiriyah University, Baghdad, Iraq.
| |
Collapse
|
6
|
ElSheikh A, Driggers CM, Shyng SL. Non-radioactive Rb + Efflux Assay for Screening K ATP Channel Modulators. Methods Mol Biol 2024; 2796:191-210. [PMID: 38856903 DOI: 10.1007/978-1-0716-3818-7_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
ATP-sensitive potassium (KATP) channels function as metabolic sensors that link cell membrane excitability to the cellular energy status by controlling potassium ion (K+) flow across the cell membrane according to intracellular ATP and ADP concentrations. As such, KATP channels influence a broad spectrum of physiological processes, including insulin secretion and cardiovascular functions. KATP channels are hetero-octamers, consisting of four inward rectifier potassium channel subunits, Kir6.1 or Kir6.2, and four sulfonylurea receptors (SURs), SUR1, SUR2A, or SUR2B. Different Kir6 and SUR isoforms assemble into KATP channel subtypes with distinct tissue distributions and physiological functions. Mutations in the genes encoding KATP channel subunits underlie various human diseases. Targeted treatment for these diseases requires subtype-specific KATP channel modulators. Rubidium ions (Rb+) also pass through KATP channels, and Rb+ efflux assays can be used to assess KATP channel function and activity. Flame atomic absorption spectroscopy (Flame-AAS) combined with microsampling can measure Rb+ in small volume, which provides an efficient tool to screen for compounds that alter KATP channel activity in Rb+ efflux assays. In this chapter, we describe a detailed protocol for Rb+ efflux assays designed to identify new KATP channel modulators with potential therapeutic utilities.
Collapse
Affiliation(s)
- Assmaa ElSheikh
- Department of Chemical Physiology and Biochemistry, School of Medicine, Oregon Health and Science University, Portland, OR, USA.
- Department of Medical Biochemistry, Tanta University, Tanta, Egypt.
| | - Camden M Driggers
- Department of Chemical Physiology and Biochemistry, School of Medicine, Oregon Health and Science University, Portland, OR, USA
| | - Show-Ling Shyng
- Department of Chemical Physiology and Biochemistry, School of Medicine, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
7
|
Kourampi I, Katsioupa M, Oikonomou E, Tsigkou V, Marinos G, Goliopoulou A, Katsarou O, Kalogeras K, Theofilis P, Tsatsaragkou A, Siasos G, Tousoulis D, Vavuranakis M. The Role of Ranolazine in Heart Failure-Current Concepts. Am J Cardiol 2023; 209:92-103. [PMID: 37844876 DOI: 10.1016/j.amjcard.2023.09.066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 09/09/2023] [Accepted: 09/15/2023] [Indexed: 10/18/2023]
Abstract
Heart failure is a complex clinical syndrome with a detrimental impact on mortality and morbidity. Energy substrate utilization and myocardial ion channel regulation have gained research interest especially after the introduction of sodium-glucose co-transporter 2 inhibitors in the treatment of heart failure. Ranolazine or N-(2,6-dimethylphenyl)-2-(4-[2-hydroxy-3-(2-methoxyphenoxy) propyl] piperazin-1-yl) acetamide hydrochloride is an active piperazine derivative which inhibits late sodium current thus minimizing calcium overload in the ischemic cardiomyocytes. Ranolazine also prevents fatty acid oxidation and favors glycose utilization ameliorating the "energy starvation" of the failing heart. Heart failure with preserved ejection fraction is characterized by diastolic impairment; according to the literature ranolazine could be beneficial in the management of increased left ventricular end-diastolic pressure, right ventricular systolic dysfunction and wall shear stress which is reflected by the high natriuretic peptides. Fewer data is evident regarding the effects of ranolazine in heart failure with reduced ejection fraction and mainly support the control of the sodium-calcium exchanger and function of sarcoendoplasmic reticulum calcium adenosine triphosphatase. Ranolazine's therapeutic mechanisms in myocardial ion channels and energy utilization are documented in patients with chronic coronary syndromes. Nevertheless, ranolazine might have a broader effect in the therapy of heart failure and further mechanistic research is required.
Collapse
Affiliation(s)
- Islam Kourampi
- 3rd Department of Cardiology, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Maria Katsioupa
- 3rd Department of Cardiology, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece.
| | - Vasiliki Tsigkou
- 3rd Department of Cardiology, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Georgios Marinos
- 3rd Department of Cardiology, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Athina Goliopoulou
- 3rd Department of Cardiology, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Ourania Katsarou
- 3rd Department of Cardiology, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Konstantinos Kalogeras
- 3rd Department of Cardiology, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Panagiotis Theofilis
- 1st Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Aikaterini Tsatsaragkou
- 3rd Department of Cardiology, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Gerasimos Siasos
- 3rd Department of Cardiology, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece; Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston Massachusetts
| | - Dimitris Tousoulis
- 1st Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Manolis Vavuranakis
- 3rd Department of Cardiology, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| |
Collapse
|
8
|
Dupuy M, Gueguinou M, Potier-Cartereau M, Lézot F, Papin M, Chantôme A, Rédini F, Vandier C, Verrecchia F. SK Ca- and Kv1-type potassium channels and cancer: Promising therapeutic targets? Biochem Pharmacol 2023; 216:115774. [PMID: 37678626 DOI: 10.1016/j.bcp.2023.115774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 09/09/2023]
Abstract
Ion channels are transmembrane structures that allow the passage of ions across cell membranes such as the plasma membrane or the membranes of various organelles like the nucleus, endoplasmic reticulum, Golgi apparatus or mitochondria. Aberrant expression of various ion channels has been demonstrated in several tumor cells, leading to the promotion of key functions in tumor development, such as cell proliferation, resistance to apoptosis, angiogenesis, invasion and metastasis. The link between ion channels and these key biological functions that promote tumor development has led to the classification of cancers as oncochannelopathies. Among all ion channels, the most varied and numerous, forming the largest family, are the potassium channels, with over 70 genes encoding them in humans. In this context, this review will provide a non-exhaustive overview of the role of plasma membrane potassium channels in cancer, describing 1) the nomenclature and structure of potassium channels, 2) the role of these channels in the control of biological functions that promotes tumor development such as proliferation, migration and cell death, and 3) the role of two particular classes of potassium channels, the SKCa- and Kv1- type potassium channels in cancer progression.
Collapse
Affiliation(s)
- Maryne Dupuy
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, F-44000 Nantes, France.
| | | | | | - Frédéric Lézot
- Sorbonne University, INSERM UMR933, Hôpital Trousseau (AP-HP), Paris F-75012, France
| | - Marion Papin
- N2C UMR 1069, University of Tours, INSERM, Tours, France
| | | | - Françoise Rédini
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, F-44000 Nantes, France
| | | | - Franck Verrecchia
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, F-44000 Nantes, France.
| |
Collapse
|
9
|
Goel H, Carey M, Elshaikh A, Krinock M, Goyal D, Nadar SK. Cardioprotective and Antianginal Efficacy of Nicorandil: A Comprehensive Review. J Cardiovasc Pharmacol 2023; 82:69-85. [PMID: 37256547 DOI: 10.1097/fjc.0000000000001436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 04/15/2023] [Indexed: 06/01/2023]
Abstract
ABSTRACT Angina pectoris remains a significant burden despite advances in medical therapy and coronary revascularization. Many patients (up to 30%) with angina have normal coronary arteries, with coronary microvascular disease and/or coronary artery vasospasm being major drivers of the myocardial demand-supply mismatch. Even among patients revascularized for symptomatic epicardial coronary stenosis, recurrent angina remains highly prevalent. Medical therapy for angina currently centers around 2 disparate goals, viz secondary prevention of hard clinical outcomes and symptom control. Vasodilators, such as nitrates, have been first-line antianginal agents for decades, along with beta-blockers and calcium channel blockers. However, efficacy in symptoms control is heterogenous, depending on underlying mechanism(s) of angina in an individual patient, often necessitating multiple agents. Nicorandil (NCO) is an antianginal agent first discovered in the late 1970s with a uniquely dual mechanism of action. Like a typical nitrate, it mediates medium-large vessel vasodilation through nitric oxide. In addition, NCO has adenosine triphosphate (ATP)-dependent potassium channel agonist activity (K ATP ), mediating microvascular dilatation. Hence, it has proven effective in both coronary artery vasospasm and coronary microvascular disease, typically challenging patient populations. Moreover, emerging evidence suggests that cardiomyocyte protection against ischemia through ischemic preconditioning may be mediated through K ATP agonism. Finally, there is now fairly firm evidence in favor of NCO in terms of hard event reduction among patients with stable coronary artery disease, following myocardial infarction, and perhaps even among patients with congestive heart failure. This review aims to summarize the mechanism of action of NCO, its efficacy as an antianginal, and current evidence behind its impact on hard outcomes. Finally, we review other cardiac and emerging noncardiac indications for NCO use.
Collapse
Affiliation(s)
- Harsh Goel
- Department of Medicine, St Luke's University Hospital, Bethlehem, PA
- Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Matthew Carey
- Department of Medicine, St Luke's University Hospital, Bethlehem, PA
| | | | - Matthew Krinock
- Department of Medicine, St Luke's University Hospital, Bethlehem, PA
- ‡Department of Cardiology, St Luke's University Hospital, Bethlehem, PA
| | - Deepak Goyal
- Department of Cardiology, Worcestershire Acute Hospitals NHS Trust, Worcester, UK; and
| | - Sunil K Nadar
- Department of Cardiology, Dudley Group of Hospitals NHS Trust, Dudley, UK
| |
Collapse
|
10
|
Behera R, Sharma V, Grewal AK, Kumar A, Arora B, Najda A, Albadrani GM, Altyar AE, Abdel-Daim MM, Singh TG. Mechanistic correlation between mitochondrial permeability transition pores and mitochondrial ATP dependent potassium channels in ischemia reperfusion. Biomed Pharmacother 2023; 162:114599. [PMID: 37004326 DOI: 10.1016/j.biopha.2023.114599] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 04/03/2023] Open
Abstract
Mitochondrial dysfunction is one of the fundamental causes of ischemia reperfusion (I/R) damage. I/R refers to the paradoxical progression of cellular dysfunction and death that occurs when blood flow is restored to previously ischemic tissues. I/R causes a significant rise in mitochondrial permeability resulting in the opening of mitochondrial permeability transition pores (MPTP). The MPTP are broad, nonspecific channels present in the inner mitochondrial membrane (IMM), and are known to mediate the deadly permeability alterations that trigger mitochondrial driven cell death. Protection from reperfusion injury occurs when long-term ischemia is accompanied by short-term ischemic episodes or inhibition of MPTP from opening via mitochondrial ATP dependent potassium (mitoKATP) channels. These channels located in the IMM, play an essential role in ischemia preconditioning (PC) and protect against cell death by blocking MPTP opening. This review primarily focuses on the interaction between the MPTP and mitoKATP along with their role in the I/R injury. This article also describes the molecular composition of the MPTP and mitoKATP in order to promote future knowledge and treatment of diverse I/R injuries in various organs.
Collapse
|
11
|
El-Meanawy SK, Dooge H, Wexler AC, Kosmach AC, Serban L, Santos EA, Alvarado FJ, Hacker TA, Ramratnam M. Overexpression of a Short Sulfonylurea Splice Variant Increases Cardiac Glucose Uptake and Uncouples Mitochondria by Regulating ROMK Activity. Life (Basel) 2023; 13:1015. [PMID: 37109544 PMCID: PMC10146620 DOI: 10.3390/life13041015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/04/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
The mitochondrial splice variant of the sulfonylurea receptor (SUR2A-55) is associated with protection from myocardial ischemia-reperfusion (IR) injury, increased mitochondrial ATP sensitive K+ channel activity (mitoKATP) and altered glucose metabolism. While mitoKATP channels composed of CCDC51 and ABCB8 exist, the mitochondrial K+ pore regulated by SUR2A-55 is unknown. We explored whether SUR2A-55 regulates ROMK to form an alternate mitoKATP. We assessed glucose uptake in mice overexpressing SUR2A-55 (TGSUR2A-55) compared with WT mice during IR injury. We then examined the expression level of ROMK and the effect of ROMK modulation on mitochondrial membrane potential (Δψm) in WT and TGSUR2A-55 mice. TGSUR2A-55 had increased glucose uptake compared to WT mice during IR injury. The expression of ROMK was similar in WT compared to TGSUR2A-55 mice. ROMK inhibition hyperpolarized resting cardiomyocyte Δψm from TGSUR2A-55 mice but not from WT mice. In addition, TGSUR2A-55 and ROMK inhibitor treated WT isolated cardiomyocytes had enhanced mitochondrial uncoupling. ROMK inhibition blocked diazoxide induced Δψm depolarization and prevented preservation of Δψm from FCCP perfusion in WT and to a lesser degree TGSUR2A-55 mice. In conclusion, cardio-protection from SUR2A-55 is associated with ROMK regulation, enhanced mitochondrial uncoupling and increased glucose uptake.
Collapse
Affiliation(s)
- Sarah K. El-Meanawy
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (S.K.E.-M.)
- Cardiology Section, Medical Service, William. S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Holly Dooge
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (S.K.E.-M.)
- Cardiology Section, Medical Service, William. S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Allison C. Wexler
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (S.K.E.-M.)
- Cardiology Section, Medical Service, William. S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Anna C. Kosmach
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (S.K.E.-M.)
| | - Lara Serban
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (S.K.E.-M.)
| | - Elizabeth A. Santos
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (S.K.E.-M.)
| | - Francisco J. Alvarado
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (S.K.E.-M.)
- Cardiovascular Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Timothy A. Hacker
- Cardiovascular Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Mohun Ramratnam
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (S.K.E.-M.)
- Cardiology Section, Medical Service, William. S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| |
Collapse
|
12
|
Semedi BP, Rehatta NM, Soetjipto S, Nugraha J, Mahyuddin MH, Arnindita JN, Wairooy NAP. How Effective is Angiotensin II in Decreasing Mortality of Vasodilatory Shock? A Systematic Review. Open Access Emerg Med 2023; 15:1-11. [PMID: 36636460 PMCID: PMC9830054 DOI: 10.2147/oaem.s391167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 12/20/2022] [Indexed: 01/05/2023] Open
Abstract
Background Patients with severe vasodilation accompanied by refractory hypotension despite high doses of vasopressors were associated with a high mortality rate. The Ang-2 for the Treatment of High-Output Shock (ATHOS) 3 trial demonstrated that angiotensin 2 (Ang-2) could effectively increase MAP and blood pressure in vasodilatory shock patients. This systematic review aims to summarize the impact of Ang-2 for the treatment of vasodilatory shock on clinical outcomes, including length of stay, MAP level (before and after), and mortality also Ang-2 dose needed. Methods A systematic search in PubMed, Sage, ScienceDirect, Scopus and Gray literature was conducted to obtain studies about the use of Ang-2 in vasodilatory shock patients. Results In all of the studies that we obtained, there were different results regarding mortality in patients with vasodilatory shock with Ang-2. Mortality was significantly lower when Ang-2 was administered to patients with elevated renin. The initial dose of Ang-2 can be started at 10-20 ng/kg/min, but there is no agreement on the maximum dose. Ang-2 may be considered a third-line vasopressor if the targeted MAP has not been achieved after administration of norepinephrine >200 ng/kg/min for more than 6 hours. Although not statistically significant, the use of Ang-2 can reduce the length of stay in the ICU and in the hospital when compared to patients without Ang-2 therapy, in addition to reducing the dose of vasopressor. Conclusion Overall, the use of Ang-2 has potential to be a regimen for patients with vasodilatory shock. Further study is needed to obtain more data.
Collapse
Affiliation(s)
- Bambang Pujo Semedi
- Doctoral Program of Medical Science, Faculty of Medicine, Universitas Airlangga, Surabaya, East Java, 60132, Indonesia,Department of Anesthesiology and Reanimation, Medical Faculty of Medicine, Universitas Airlangga—Dr Soetomo General Hospital, Surabaya, East Java, 60132, Indonesia
| | - Nancy Margarita Rehatta
- Department of Anesthesiology and Reanimation, Medical Faculty of Medicine, Universitas Airlangga—Dr Soetomo General Hospital, Surabaya, East Java, 60132, Indonesia,Correspondence: Nancy Margarita Rehatta, Email
| | - Soetjipto Soetjipto
- Department of Medical Biochemistry, Medical Faculty of Universitas Airlangga, Surabaya, East Java, 60132, Indonesia
| | - Jusak Nugraha
- Department of Clinical Pathology, Medical Faculty of Universitas Airlangga, Surabaya, East Java, 60132, Indonesia
| | | | | | - Nabilah A P Wairooy
- Medical Faculty Universitas Airlangga, Surabaya, East Java, 60132, Indonesia
| |
Collapse
|
13
|
Suwara J, Radzikowska-Cieciura E, Chworos A, Pawlowska R. The ATP-dependent Pathways and Human Diseases. Curr Med Chem 2023; 30:1232-1255. [PMID: 35319356 DOI: 10.2174/0929867329666220322104552] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 12/31/2021] [Accepted: 01/04/2022] [Indexed: 11/22/2022]
Abstract
Adenosine triphosphate (ATP) is one of the most important molecules of life, present both inside the cells and extracellularly. It is an essential building block for nucleic acids biosynthesis and crucial intracellular energy storage. However, one of the most interesting functions of ATP is the role of a signaling molecule. Numerous studies indicate the involvement of ATP-dependent pathways in maintaining the proper functioning of individual tissues and organs. Herein, the latest data indicating the ATP function in the network of intra- and extracellular signaling pathways including purinergic signaling, MAP kinase pathway, mTOR and calcium signaling are collected. The main ATP-dependent processes maintaining the proper functioning of the nervous, cardiovascular and immune systems, as well as skin and bones, are summarized. The disturbances in the ATP amount, its cellular localization, or interaction with target elements may induce pathological changes in signaling pathways leading to the development of serious diseases. The impact of an ATP imbalance on the development of dangerous health dysfunctions such as neurodegeneration diseases, cardiovascular diseases (CVDs), diabetes mellitus, obesity, cancers and immune pathogenesis are discussed here.
Collapse
Affiliation(s)
- Justyna Suwara
- Department of Bioorganic Chemistry, Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Sienkiewicza 112, 90-363, Lodz, Poland
| | - Ewa Radzikowska-Cieciura
- Department of Bioorganic Chemistry, Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Sienkiewicza 112, 90-363, Lodz, Poland
| | - Arkadiusz Chworos
- Department of Bioorganic Chemistry, Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Sienkiewicza 112, 90-363, Lodz, Poland
| | - Roza Pawlowska
- Department of Bioorganic Chemistry, Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Sienkiewicza 112, 90-363, Lodz, Poland
| |
Collapse
|
14
|
Balakina-Vikulova NA, Katsnelson LB. Integrative Mathematical Model of Electrical, Metabolic and Mechanical Processes in Human Cardiomyocytes. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022070122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
15
|
Kumar A, Kumari S, Singh D. Insights into the Cellular Interactions and Molecular Mechanisms of Ketogenic Diet for Comprehensive Management of Epilepsy. Curr Neuropharmacol 2022; 20:2034-2049. [PMID: 35450526 PMCID: PMC9886834 DOI: 10.2174/1570159x20666220420130109] [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: 06/24/2022] [Revised: 01/27/2022] [Accepted: 03/25/2022] [Indexed: 11/22/2022] Open
Abstract
A high-fat diet with appropriate protein and low carbohydrate content, widely known as the ketogenic diet (KD), is considered as an effective non-pharmacotherapeutic treatment option for certain types of epilepsies. Several preclinical and clinical studies have been carried out to elucidate its mechanism of antiepileptic action. Ketone bodies produced after KD's breakdown interact with cellular excito-inhibitory processes and inhibit abnormal neuronal firing. The generated ketone bodies decrease glutamate release by inhibiting the vesicular glutamate transporter 1 and alter the transmembrane potential by hyperpolarization. Apart from their effect on the well-known pathogenic mechanisms of epilepsy, some recent studies have shown the interaction of KD metabolites with novel neuronal targets, particularly adenosine receptors, adenosine triphosphate-sensitive potassium channel, mammalian target of rapamycin, histone deacetylase, hydroxycarboxylic acid receptors, and the NLR family pyrin domain containing 3 inflammasomes to suppress seizures. The role of KD in augmenting gut microbiota as a potential mechanism for epileptic seizure suppression has been established. Furthermore, some recent findings also support the beneficial effect of KD against epilepsy- associated comorbidities. Despite several advantages of the KD in epilepsy management, its use is also associated with a wide range of side effects. Hypoglycemia, excessive ketosis, acidosis, renal stones, cardiomyopathies, and other metabolic disturbances are the primary adverse effects observed with the use of KD. However, in some recent studies, modified KD has been tested with lesser side effects and better tolerability. The present review discusses the molecular mechanism of KD and its role in managing epilepsy and its associated comorbidities.
Collapse
Affiliation(s)
- Amit Kumar
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR- Institute of Himalayan Bioresource Technology, Palampur 176061, Himachal Pradesh, India; ,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Savita Kumari
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR- Institute of Himalayan Bioresource Technology, Palampur 176061, Himachal Pradesh, India; ,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Damanpreet Singh
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR- Institute of Himalayan Bioresource Technology, Palampur 176061, Himachal Pradesh, India; ,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India,Address correspondence to this author at the Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur-176061, Himachal Pradesh, India; Tel: +91-9417923132; E-mails: ;
| |
Collapse
|
16
|
McClenaghan C, Nichols CG. Kir6.1 and SUR2B in Cantú syndrome. Am J Physiol Cell Physiol 2022; 323:C920-C935. [PMID: 35876283 PMCID: PMC9467476 DOI: 10.1152/ajpcell.00154.2022] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/07/2022] [Accepted: 07/07/2022] [Indexed: 12/25/2022]
Abstract
Kir6.1 and SUR2 are subunits of ATP-sensitive potassium (KATP) channels expressed in a wide range of tissues. Extensive study has implicated roles of these channel subunits in diverse physiological functions. Together they generate the predominant KATP conductance in vascular smooth muscle and are the target of vasodilatory drugs. Roles for Kir6.1/SUR2 dysfunction in disease have been suggested based on studies of animal models and human genetic discoveries. In recent years, it has become clear that gain-of-function (GoF) mutations in both genes result in Cantú syndrome (CS)-a complex, multisystem disorder. There is currently no targeted therapy for CS, but studies of mouse models of the disease reveal that pharmacological reversibility of cardiovascular and gastrointestinal pathologies can be achieved by administration of the KATP channel inhibitor, glibenclamide. Here we review the function, structure, and physiological and pathological roles of Kir6.1/SUR2B channels, with a focus on CS. Recent studies have led to much improved understanding of the underlying pathologies and the potential for treatment, but important questions remain: Can the study of genetically defined CS reveal new insights into Kir6.1/SUR2 function? Do these reveal new pathophysiological mechanisms that may be important in more common diseases? And is our pharmacological armory adequately stocked?
Collapse
Affiliation(s)
- Conor McClenaghan
- Department of Cell Biology and Physiology, Center for the Investigation of Membrane Excitability Diseases (CIMED), Washington University, St. Louis, Missouri
| | - Colin G Nichols
- Department of Cell Biology and Physiology, Center for the Investigation of Membrane Excitability Diseases (CIMED), Washington University, St. Louis, Missouri
| |
Collapse
|
17
|
Mustapha S, Azemi AK, Wan Ahmad WAN, Rasool AHG, Mustafa MR, Mokhtar SS. Inhibition of Endoplasmic Reticulum Stress Improves Acetylcholine-Mediated Relaxation in the Aorta of Type-2 Diabetic Rats. Molecules 2022; 27:5107. [PMID: 36014347 PMCID: PMC9413505 DOI: 10.3390/molecules27165107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/05/2022] [Accepted: 08/08/2022] [Indexed: 12/02/2022] Open
Abstract
Endoplasmic reticulum (ER) stress contributes to insulin resistance and macro- and microvascular complications associated with diabetes. This study aimed to evaluate the effect of ER stress inhibition on endothelial function in the aorta of type-2 diabetic rats. Type-2 diabetes was developed in male Sprague-Dawley rats using a high-fat diet and low-dose streptozotocin. Rat aortic tissues were harvested to study endothelial-dependent relaxation. The mechanisms for acetylcholine-mediated relaxation were investigated using pharmacological blockers, Western blotting, oxidative stress, and inflammatory markers. Acetylcholine-mediated relaxation was diminished in the aorta of diabetic rats compared to control rats; supplementation with TUDCA improved relaxation. In the aortas of control and diabetic rats receiving TUDCA, the relaxation was mediated via eNOS/PI3K/Akt, NAD(P)H, and the KATP channel. In diabetic rats, acetylcholine-mediated relaxation involved eNOS/PI3K/Akt and NAD(P)H, but not the KATP channel. The expression of ER stress markers was upregulated in the aorta of diabetic rats and reduced with TUDCA supplementation. The expression of eNOS and Akt were lower in diabetic rats but were upregulated after supplementation with TUDCA. The levels of MDA, IL-6, and SOD activity were higher in the aorta of the diabetic rats compared to control rats. This study demonstrated that endothelial function was impaired in diabetes, however, supplementation with TUDCA improved the function via eNOS/Akt/PI3K, NAD(P)H, and the KATP channel. The improvement of endothelial function was associated with increased expressions of eNOS and Akt. Thus, ER stress plays a crucial role in the impairment of endothelial-dependent relaxation. Mitigating ER stress could be a potential strategy for improving endothelial dysfunction in type-2 diabetes.
Collapse
Affiliation(s)
- Sagir Mustapha
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu 16150, Kelantan, Malaysia
- Department of Pharmacology and Therapeutics, Ahmadu Bello University, Zaria 810107, Kaduna, Nigeria
| | - Ahmad Khusairi Azemi
- Institute of Marine Biotechnology, Universiti Malaysia Terengganu, Kuala Terengganu 21030, Terengganu, Malaysia
| | - Wan Amir Nizam Wan Ahmad
- Biomedicine Programme, School of Health Sciences, Universiti Sains Malaysia, Kota Bharu 16150, Kelantan, Malaysia
| | - Aida Hanum Ghulam Rasool
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu 16150, Kelantan, Malaysia
| | - Mohd Rais Mustafa
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Selangor, Malaysia
| | - Siti Safiah Mokhtar
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu 16150, Kelantan, Malaysia
| |
Collapse
|
18
|
ATP-Sensitive Potassium Channels in Migraine: Translational Findings and Therapeutic Potential. Cells 2022; 11:cells11152406. [PMID: 35954249 PMCID: PMC9367966 DOI: 10.3390/cells11152406] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 07/29/2022] [Accepted: 07/30/2022] [Indexed: 12/10/2022] Open
Abstract
Globally, migraine is a leading cause of disability with a huge impact on both the work and private life of affected persons. To overcome the societal migraine burden, better treatment options are needed. Increasing evidence suggests that ATP-sensitive potassium (KATP) channels are involved in migraine pathophysiology. These channels are essential both in blood glucose regulation and cardiovascular homeostasis. Experimental infusion of the KATP channel opener levcromakalim to healthy volunteers and migraine patients induced headache and migraine attacks in 82-100% of participants. Thus, this is the most potent trigger of headache and migraine identified to date. Levcromakalim likely induces migraine via dilation of cranial arteries. However, other neuronal mechanisms are also proposed. Here, basic KATP channel distribution, physiology, and pharmacology are reviewed followed by thorough review of clinical and preclinical research on KATP channel involvement in migraine. KATP channel opening and blocking have been studied in a range of preclinical migraine models and, within recent years, strong evidence on the importance of their opening in migraine has been provided from human studies. Despite major advances, translational difficulties exist regarding the possible anti-migraine efficacy of KATP channel blockage. These are due to significant species differences in the potency and specificity of pharmacological tools targeting the various KATP channel subtypes.
Collapse
|
19
|
de Sousa Melo SR, Dos Santos LR, da Cunha Soares T, Cardoso BEP, da Silva Dias TM, Morais JBS, de Paiva Sousa M, de Sousa TGV, da Silva NC, da Silva LD, Cruz KJC, do Nascimento Marreiro D. Participation of Magnesium in the Secretion and Signaling Pathways of Insulin: an Updated Review. Biol Trace Elem Res 2022; 200:3545-3553. [PMID: 35666386 DOI: 10.1007/s12011-021-02966-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 10/11/2021] [Indexed: 11/02/2022]
Abstract
Several studies have demonstrated the participation of various minerals in mechanisms involving insulin. Magnesium, in particular, plays an important role in the secretion and action of this hormone. Therefore, this review aimed to examine the latest insights into the biochemical and molecular aspects of the participation of magnesium in insulin sensitivity. Magnesium plays a vital role in the activity of intracellular proteins involved in insulin secretion in β-pancreatic cells, such as glucokinase, ATPase, and protein kinase C. In addition, evidence suggests that this mineral participates directly in insulin sensitivity and signaling in peripheral tissues, acting in the phosphorylation of the receptor tyrosine kinase and the insulin receptor substrates 1, insulin receptor substrates 2, phosphatidylinositol 3-kinase, and protein kinase B, and indirectly by reducing oxidative stress and chronic low-grade inflammation, which also lead to insulin resistance. Thus, magnesium deficiency is associated with glucose intolerance, while magnesium supplementation stimulates insulin secretion in pancreatic cells and improves insulin sensitivity in peripheral tissues. However, studies must consider assess short- and long-term nutritional status of mineral before performing intervention, the relevance of the balance of other nutrients that influence hormone secretion and sensibility, and health status of the assessed population.
Collapse
Affiliation(s)
| | - Loanne Rocha Dos Santos
- Graduate Program in Food and Nutrition, Federal University of Piauí, Teresina (Piauí), Brasil
| | - Tamires da Cunha Soares
- Graduate Program in Food and Nutrition, Federal University of Piauí, Teresina (Piauí), Brasil
| | | | | | | | - Mickael de Paiva Sousa
- Graduate Program in Food and Nutrition, Federal University of Piauí, Teresina (Piauí), Brasil
| | | | | | | | - Kyria Jayanne Clímaco Cruz
- Department of Nutrition, Health Sciences Center, Federal University of Piauí, Rua Hugo Napoleão, 665, Ed. Palazzo Reale, Apto 2001, Jockey, CEP 64048-320, Teresina, Piauí, Brasil
| | - Dilina do Nascimento Marreiro
- Department of Nutrition, Health Sciences Center, Federal University of Piauí, Rua Hugo Napoleão, 665, Ed. Palazzo Reale, Apto 2001, Jockey, CEP 64048-320, Teresina, Piauí, Brasil.
| |
Collapse
|
20
|
Manavi MA. Neuroprotective effects of glucagon-like peptide-1 (GLP-1) analogues in epilepsy and associated comorbidities. Neuropeptides 2022; 94:102250. [PMID: 35561568 DOI: 10.1016/j.npep.2022.102250] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/04/2022] [Accepted: 05/04/2022] [Indexed: 10/18/2022]
Abstract
Epilepsy is a common neurological condition induced by losing equilibrium of different pathway as well as neurotransmitters that affects over 50 million people globally. Furthermore, long-term administration of anti-seizure medications has been associated with psychological adverse effects. Also, epilepsy has been related to an increased prevalence of obesity and called type 2 diabetes mellitus. On the other hand, GLP-1 receptors are located throughout the brain, including the hippocampus, which have been associated to majority of neurological conditions, such as epilepsy and psychiatric disorders. Moreover, the impact of different GLP-1 analogues on diverse neurotransmitter systems and associated cellular and molecular pathways as a potential therapeutic target for epilepsy and associated comorbidities has piqued curiosity. In this regard, the anticonvulsant effects of GLP-1 analogues have been investigated in various animal models and promising results such as anticonvulsants as well as cognitive improvements have been observed. For instance, GLP-1 analogues like liraglutide in addition to their possible anticonvulsant benefits, could be utilized to alleviate mental cognitive problems caused by both epilepsy and anti-seizure medication side effects. In this review and growing protective function of GLP-1 in epilepsy induced by disturbed neurotransmitter pathways and the probable mechanisms of action of GLP-1 analogues as well as the GLP-1 receptor in these effects have been discussed.
Collapse
Affiliation(s)
- Mohammad Amin Manavi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
21
|
Anti-invasive effects of minoxidil on human breast cancer cells: combination with ranolazine. Clin Exp Metastasis 2022; 39:679-689. [PMID: 35643818 PMCID: PMC9338910 DOI: 10.1007/s10585-022-10166-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 04/11/2022] [Indexed: 12/11/2022]
Abstract
A plethora of ion channels have been shown to be involved systemically in the pathophysiology of cancer and ion channel blockers can produce anti-metastatic effects. However, although ion channels are known to frequently function in concerted action, little is known about possible combined effects of ion channel modulators on metastatic cell behaviour. Here, we investigated functional consequences of pharmacologically modulating ATP-gated potassium (KATP) channel and voltage-gated sodium channel (VGSC) activities individually and in combination. Two triple-negative human breast cancer cell lines were used: MDA-MB-231 and MDA-MB-468, the latter mainly for comparison. Most experiments were carried out on hypoxic cells. Electrophysiological effects were studied by whole-cell patch clamp recording. Minoxidil (a KATP channel opener) and ranolazine (a blocker of the VGSC persistent current) had no effect on cell viability and proliferation, alone or in combination. In contrast, invasion was significantly reduced in a dose-dependent manner by clinical concentrations of minoxidil and ranolazine. Combining the two drugs produced significant additive effects at concentrations as low as 0.625 μM ranolazine and 2.5 μM minoxidil. Electrophysiologically, acute application of minoxidil shifted VGSC steady-state inactivation to more hyperpolarised potentials and slowed recovery from inactivation, consistent with inhibition of VGSC activation. We concluded (i) that clinically relevant doses of minoxidil and ranolazine individually could inhibit cellular invasiveness dose dependently and (ii) that their combination was additionally effective. Accordingly, ranolazine, minoxidil and their combination may be repurposed as novel anti-metastatic agents.
Collapse
|
22
|
Arrell DK, Park S, Yamada S, Alekseev AE, Garmany A, Jeon R, Vuckovic I, Lindor JZ, Terzic A. K ATP channel dependent heart multiome atlas. Sci Rep 2022; 12:7314. [PMID: 35513538 PMCID: PMC9072320 DOI: 10.1038/s41598-022-11323-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 04/21/2022] [Indexed: 11/09/2022] Open
Abstract
Plasmalemmal ATP sensitive potassium (KATP) channels are recognized metabolic sensors, yet their cellular reach is less well understood. Here, transgenic Kir6.2 null hearts devoid of the KATP channel pore underwent multiomics surveillance and systems interrogation versus wildtype counterparts. Despite maintained organ performance, the knockout proteome deviated beyond a discrete loss of constitutive KATP channel subunits. Multidimensional nano-flow liquid chromatography tandem mass spectrometry resolved 111 differentially expressed proteins and their expanded network neighborhood, dominated by metabolic process engagement. Independent multimodal chemometric gas and liquid chromatography mass spectrometry unveiled differential expression of over one quarter of measured metabolites discriminating the Kir6.2 deficient heart metabolome. Supervised class analogy ranking and unsupervised enrichment analysis prioritized nicotinamide adenine dinucleotide (NAD+), affirmed by extensive overrepresentation of NAD+ associated circuitry. The remodeled metabolome and proteome revealed functional convergence and an integrated signature of disease susceptibility. Deciphered cardiac patterns were traceable in the corresponding plasma metabolome, with tissue concordant plasma changes offering surrogate metabolite markers of myocardial latent vulnerability. Thus, Kir6.2 deficit precipitates multiome reorganization, mapping a comprehensive atlas of the KATP channel dependent landscape.
Collapse
Affiliation(s)
- D Kent Arrell
- Marriott Heart Disease Research Program, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA.,Marriott Family Comprehensive Cardiac Regenerative Medicine, Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA.,Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Sungjo Park
- Marriott Heart Disease Research Program, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA.,Marriott Family Comprehensive Cardiac Regenerative Medicine, Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA.,Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA.,Department of Biochemistry & Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Satsuki Yamada
- Marriott Heart Disease Research Program, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA.,Marriott Family Comprehensive Cardiac Regenerative Medicine, Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA.,Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA.,Division of Geriatric Medicine & Gerontology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Alexey E Alekseev
- Marriott Heart Disease Research Program, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA.,Marriott Family Comprehensive Cardiac Regenerative Medicine, Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA.,Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA.,Institute of Theoretical and Experimental Biophysics, Russian Academy of Science, Pushchino, Moscow Region, Russia
| | - Armin Garmany
- Marriott Heart Disease Research Program, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA.,Marriott Family Comprehensive Cardiac Regenerative Medicine, Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA.,Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA.,Mayo Clinic Alix School of Medicine, Regenerative Sciences Track, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, USA
| | - Ryounghoon Jeon
- Marriott Heart Disease Research Program, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA.,Marriott Family Comprehensive Cardiac Regenerative Medicine, Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA.,Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Ivan Vuckovic
- Department of Biochemistry & Molecular Biology, Mayo Clinic, Rochester, MN, USA.,Metabolomics Core, Mayo Clinic, Rochester, MN, USA
| | - Jelena Zlatkovic Lindor
- Marriott Heart Disease Research Program, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA.,Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Andre Terzic
- Marriott Heart Disease Research Program, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA. .,Marriott Family Comprehensive Cardiac Regenerative Medicine, Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA. .,Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA. .,Department of Clinical Genomics, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
23
|
Pirotte B, Florence X, Goffin E, Leleux F, Lebrun P. Research Advancements on Fluorinated and Non-Fluorinated 4-Phenyl(thio)ureido-Substituted 2,2-Dimethylchromans Acting as Inhibitors of Insulin Release and Smooth Muscle Relaxants. Med Chem 2022; 18:884-894. [PMID: 35189799 DOI: 10.2174/1573406418666220221145500] [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: 10/22/2021] [Revised: 11/29/2021] [Accepted: 12/21/2021] [Indexed: 11/22/2022]
Abstract
AIMS The present study aimed at characterizing the impact of the presence or absence of fluorine atoms on the phenyl and benzopyran rings of 4-phenyl(thio)ureido-substituted 2,2-dimethylchromans on their ability to inhibit insulin release from pancreatic -cells or to relax vascular smooth muscle cells. METHODS Most compounds were found to inhibit insulin secretion and to provoke a marked myorelaxant activity. RESULTS The lack of a fluorine or a chlorine atom at the 6-position of the 2,2-dimethylchroman core structure reduced the inhibitory activity on the pancreatic endocrine tissue. One of the most active compounds on both tissues, compound 11h (BPDZ 678), was selected for further pharmacological investigations. CONCLUSION The biological data suggested that 11h mainly expressed the profile of a KATP channel opener on pancreatic -cells, although a calcium entry blockade effect was also observed. On vascular smooth muscle cells, 11h behaved as a calcium entry blocker.
Collapse
Affiliation(s)
- Bernard Pirotte
- Laboratoire de Chimie Pharmaceutique, Center for Interdisciplinary Research on Medicines (CIRM), Université de Liège, Quartier Hôpital, Avenue Hippocrate 15, B-4000 Liège, Belgium
| | - Xavier Florence
- Laboratoire de Chimie Pharmaceutique, Center for Interdisciplinary Research on Medicines (CIRM), Université de Liège, Quartier Hôpital, Avenue Hippocrate 15, B-4000 Liège, Belgium
- Laboratoire de Physiologie et Pharmacologie, Université Libre de Bruxelles, Faculté de Médecine, 808 Route de Lennik, B-1070 Bruxelles, Belgium
| | - Eric Goffin
- Laboratoire de Chimie Pharmaceutique, Center for Interdisciplinary Research on Medicines (CIRM), Université de Liège, Quartier Hôpital, Avenue Hippocrate 15, B-4000 Liège, Belgium
| | - Fabienne Leleux
- Laboratoire de Physiologie et Pharmacologie, Université Libre de Bruxelles, Faculté de Médecine, 808 Route de Lennik, B-1070 Bruxelles, Belgium
| | - Philippe Lebrun
- Laboratoire de Physiologie et Pharmacologie, Université Libre de Bruxelles, Faculté de Médecine, 808 Route de Lennik, B-1070 Bruxelles, Belgium
| |
Collapse
|
24
|
Zhang X, Li G. Effects of a mitochondrial-acting drug on ischemia/reperfusion-induced ventricular arrhythmias, cardiac mitochondrial function and inflammatory cytokines in rat: Role of adenosine triphosphate-sensitive potassium channels. EUR J INFLAMM 2022. [DOI: 10.1177/1721727x221115041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Objective Ischemia/reperfusion (IR)-induced myocardial arrhythmias are a common clinical manifestation in patients with myocardial infarction after reperfusion therapy. Mitochondria play a critical role in cardioprotection. Here, we investigated the effects of KH176 as a new mitochondrial-acting drug on IR-induced ventricular arrhythmias, mitochondrial function, pro-inflammatory cytokines production, and the role of mitochondrial ATP-dependent K (mK-ATP) channels in rats’ hearts. Methods The hearts of Sprague Dawley rats (250 ± 30 g; 36 rats) underwent 35 min of ischemia followed by 120 min of reperfusion. Myocardial in vivo ischemia was induced by ligation of the left anterior descending coronary artery. KH176 at concentrations of 10 and 50 μM was intraperitoneally injected to rats 10 min before reperfusion onset. Ventricular arrhythmias were quantified during reperfusion, and cardiac mitochondrial function, nitric oxide, and pro-inflammatory cytokines levels were measured by fluorometric, spectrophotometric, and ELISA techniques. Results Administration of KH176 significantly reduced lactate-dehydrogenase release and the number, duration, incidence, and severity of ventricular arrhythmias induced by reperfusion injury. IR-induced elevation of mitochondrial reactive oxygen species production, and cardiac pro-inflammatory cytokines TNF-α, IL-6, and IL-1β, as well as reduction of mitochondrial membrane potential, ATP and nitric oxide levels were significantly restored by KH176 at 50 μM. However, the blockade of mK-ATP channels by 5-hydroxydecanoate considerably inhibited the effects of KH176 on all parameters except nitric oxide. Conclusion KH176 showed strong cardiac antiarrhythmic effects on IR-induced injury through improving mitochondrial function and reducing inflammatory and oxidative responses, and these protective effects are mediated by cardiac mK-ATP channels.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Pharmacy, Jiangsu Taizhou People’s Hospital, China
| | - Geng Li
- Department of Cardiology, Hubei Third People's Hospital Affiliated to Jianghan University, Wuhan, Hubei, China
| |
Collapse
|
25
|
Modulation of the Blood-Brain Barrier for Drug Delivery to Brain. Pharmaceutics 2021; 13:pharmaceutics13122024. [PMID: 34959306 PMCID: PMC8708282 DOI: 10.3390/pharmaceutics13122024] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 11/15/2021] [Accepted: 11/25/2021] [Indexed: 12/26/2022] Open
Abstract
The blood-brain barrier (BBB) precisely controls brain microenvironment and neural activity by regulating substance transport into and out of the brain. However, it severely hinders drug entry into the brain, and the efficiency of various systemic therapies against brain diseases. Modulation of the BBB via opening tight junctions, inhibiting active efflux and/or enhancing transcytosis, possesses the potential to increase BBB permeability and improve intracranial drug concentrations and systemic therapeutic efficiency. Various strategies of BBB modulation have been reported and investigated preclinically and/or clinically. This review describes conventional and emerging BBB modulation strategies and related mechanisms, and safety issues according to BBB structures and functions, to try to give more promising directions for designing more reasonable preclinical and clinical studies.
Collapse
|
26
|
Singareddy SS, Roessler HI, McClenaghan C, Ikle JM, Tryon RC, van Haaften G, Nichols CG. ATP-sensitive potassium channels in zebrafish cardiac and vascular smooth muscle. J Physiol 2021; 600:299-312. [PMID: 34820842 DOI: 10.1113/jp282157] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 11/18/2021] [Indexed: 12/12/2022] Open
Abstract
ATP-sensitive potassium channels (KATP channels) are hetero-octameric nucleotide-gated ion channels that couple cellular metabolism to excitability in various tissues. In the heart, KATP channels are activated during ischaemia and potentially during adrenergic stimulation. In the vasculature, they are normally active at a low level, reducing vascular tone, but the ubiquitous nature of these channels leads to complex and poorly understood channelopathies as a result of gain- or loss-of-function mutations. Zebrafish (ZF) models of these channelopathies may provide insights to the link between molecular dysfunction and complex pathophysiology, but this requires understanding the tissue dependence of channel activity and subunit specificity. Thus far, direct analysis of ZF KATP expression and functional properties has only been performed in pancreatic β-cells. Using a comprehensive combination of genetically modified fish, electrophysiology and gene expression analysis, we demonstrate that ZF cardiac myocytes (CM) and vascular smooth muscle (VSM) express functional KATP channels of similar subunit composition, structure and metabolic sensitivity to their mammalian counterparts. However, in contrast to mammalian cardiovascular KATP channels, ZF channels are insensitive to potassium channel opener drugs (pinacidil, minoxidil) in both chambers of the heart and in VSM. The results provide a first characterization of the molecular properties of fish KATP channels and validate the use of such genetically modified fish as models of human Cantú syndrome and ABCC9-related Intellectual Disability and Myopathy syndrome. KEY POINTS: Zebrafish cardiac myocytes (CM) and vascular smooth muscle (VSM) express functional KATP channels of similar subunit composition, structure and metabolic sensitivity to their mammalian counterparts. In contrast to mammalian cardiovascular KATP channels, zebrafish channels are insensitive to potassium channel opener drugs (pinacidil, minoxidil) in both chambers of the heart and in VSM. We provide a first characterization of the molecular properties of fish KATP channels and validate the use of such genetically modified fish as models of human Cantú syndrome and ABCC9-related Intellectual Disability and Myopathy syndrome.
Collapse
Affiliation(s)
- Soma S Singareddy
- Department of Cell Biology and Physiology and Center for the Investigation of Membrane Excitability Diseases, Washington University in St Louis, St Louis, MO, USA
| | - Helen I Roessler
- Department of Genetics, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Conor McClenaghan
- Department of Cell Biology and Physiology and Center for the Investigation of Membrane Excitability Diseases, Washington University in St Louis, St Louis, MO, USA
| | - Jennifer M Ikle
- Department of Cell Biology and Physiology and Center for the Investigation of Membrane Excitability Diseases, Washington University in St Louis, St Louis, MO, USA
| | - Robert C Tryon
- Department of Cell Biology and Physiology and Center for the Investigation of Membrane Excitability Diseases, Washington University in St Louis, St Louis, MO, USA
| | - Gijs van Haaften
- Department of Genetics, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Colin G Nichols
- Department of Cell Biology and Physiology and Center for the Investigation of Membrane Excitability Diseases, Washington University in St Louis, St Louis, MO, USA
| |
Collapse
|
27
|
Gasior K, Korshunov K, Trombley PQ, Bertram R. Fast-slow analysis as a technique for understanding the neuronal response to current ramps. J Comput Neurosci 2021; 50:145-159. [PMID: 34665376 DOI: 10.1007/s10827-021-00799-0] [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: 03/18/2021] [Revised: 08/19/2021] [Accepted: 09/22/2021] [Indexed: 11/28/2022]
Abstract
The standard protocol for studying the spiking properties of single neurons is the application of current steps while monitoring the voltage response. Although this is informative, the jump in applied current is artificial. A more physiological input is where the applied current is ramped up, reflecting chemosensory input. Unsurprisingly, neurons can respond differently to the two protocols, since ion channel activation and inactivation are affected differently. Understanding the effects of current ramps, and changes in their slopes, is facilitated by mathematical models. However, techniques for analyzing current ramps are under-developed. In this article, we demonstrate how current ramps can be analyzed in single neuron models. The primary issue is the presence of gating variables that activate on slow time scales and are therefore far from equilibrium throughout the ramp. The use of an appropriate fast-slow analysis technique allows one to fully understand the neural response to ramps of different slopes. This study is motivated by data from olfactory bulb dopamine neurons, where both fast ramp (tens of milliseconds) and slow ramp (tens of seconds) protocols are used to understand the spiking profiles of the cells. The slow ramps generate experimental bifurcation diagrams with the applied current as a bifurcation parameter, thereby establishing asymptotic spiking activity patterns. The faster ramps elicit purely transient behavior that is of relevance to most physiological inputs, which are short in duration. The two protocols together provide a broader understanding of the neuron's spiking profile and the role that slowly activating ion channels can play.
Collapse
Affiliation(s)
- Kelsey Gasior
- Department of Mathematics, University of Ottawa, Ottawa, ON, K1N 6N5, Canada
| | - Kirill Korshunov
- Department of Biological Science and Program in Neuroscience, Florida State University, Tallahassee, FL, 32306, USA
| | - Paul Q Trombley
- Department of Biological Science and Program in Neuroscience, Florida State University, Tallahassee, FL, 32306, USA
| | - Richard Bertram
- Department of Mathematics and Programs in Neuroscience and Molecular Biophysics, Florida State University, Tallahassee, FL, 32306, USA.
| |
Collapse
|
28
|
Abstract
ATP-sensitive K+ channels (KATP) are inwardly-rectifying potassium channels, broadly expressed throughout the body. KATP is regulated by adenine nucleotides, characteristically being activated by falling ATP and rising ADP levels thus playing an important physiological role by coupling cellular metabolism with membrane excitability. The hetero-octameric channel complex is formed of 4 pore-forming inward rectifier Kir6.x subunits (Kir6.1 or Kir6.2) and 4 regulatory sulfonylurea receptor subunits (SUR1, SUR2A, or SUR2B). These subunits can associate in various tissue-specific combinations to form functional KATP channels with distinct electrophysiological and pharmacological properties. KATP channels play many important physiological roles and mutations in channel subunits can result in diseases such as disorders of insulin handling, cardiac arrhythmia, cardiomyopathy, and neurological abnormalities. The tissue-specific expression of KATP channel subunits coupled with their rich and diverse pharmacology makes KATP channels attractive therapeutic targets in the treatment of endocrine and cardiovascular diseases.
Collapse
|
29
|
Sullivan RT, Lam NT, Haberman M, Beatka MJ, Afzal MZ, Lawlor MW, Strande JL. Cardioprotective effect of nicorandil on isoproterenol induced cardiomyopathy in the Mdx mouse model. BMC Cardiovasc Disord 2021; 21:302. [PMID: 34130633 PMCID: PMC8207777 DOI: 10.1186/s12872-021-02112-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 06/07/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) associated cardiomyopathy is a major cause of morbidity and mortality. In an in vitro DMD cardiomyocyte model, nicorandil reversed stress-induced cell injury through multiple pathways implicated in DMD. We aimed to test the efficacy of nicorandil on the progression of cardiomyopathy in mdx mice following a 10-day treatment protocol. METHODS A subset of mdx mice was subjected to low-dose isoproterenol injections over 5 days to induce a cardiac phenotype and treated with vehicle or nicorandil for 10 days. Baseline and day 10 echocardiograms were obtained to assess cardiac function. At 10 days, cardiac tissue was harvested for further analysis, which included histologic analysis and assessment of oxidative stress. Paired student's t test was used for in group comparison, and ANOVA was used for multiple group comparisons. RESULTS Compared to vehicle treated mice, isoproterenol decreased ejection fraction and fractional shortening on echocardiogram. Nicorandil prevented isoproterenol induced cardiac dysfunction. Isoproterenol increased cardiac fibrosis, which nicorandil prevented. Isoproterenol increased gene expression of NADPH oxidase, which decreased to baseline with nicorandil treatment. Superoxide dismutase 2 protein expression increased in those treated with nicorandil, and xanthine oxidase activity decreased in mice treated with nicorandil during isoproterenol stress compared to all other groups. CONCLUSIONS In conclusion, nicorandil is cardioprotective in mdx mice and warrants continued investigation as a therapy for DMD associated cardiomyopathy.
Collapse
Affiliation(s)
- Rachel T Sullivan
- Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI, 53226, USA.
| | - Ngoc T Lam
- Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI, 53226, USA
| | - Margaret Haberman
- Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI, 53226, USA
| | - Margaret J Beatka
- Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI, 53226, USA
| | - Muhammad Z Afzal
- Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI, 53226, USA
| | - Michael W Lawlor
- Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI, 53226, USA
| | - Jennifer L Strande
- Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI, 53226, USA
| |
Collapse
|
30
|
Kirkman DL, Robinson AT, Rossman MJ, Seals DR, Edwards DG. Mitochondrial contributions to vascular endothelial dysfunction, arterial stiffness, and cardiovascular diseases. Am J Physiol Heart Circ Physiol 2021; 320:H2080-H2100. [PMID: 33834868 PMCID: PMC8163660 DOI: 10.1152/ajpheart.00917.2020] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 03/12/2021] [Accepted: 04/05/2021] [Indexed: 12/11/2022]
Abstract
Cardiovascular disease (CVD) affects one in three adults and remains the leading cause of death in America. Advancing age is a major risk factor for CVD. Recent plateaus in CVD-related mortality rates in high-income countries after decades of decline highlight a critical need to identify novel therapeutic targets and strategies to mitigate and manage the risk of CVD development and progression. Vascular dysfunction, characterized by endothelial dysfunction and large elastic artery stiffening, is independently associated with an increased CVD risk and incidence and is therefore an attractive target for CVD prevention and management. Vascular mitochondria have emerged as an important player in maintaining vascular homeostasis. As such, age- and disease-related impairments in mitochondrial function contribute to vascular dysfunction and consequent increases in CVD risk. This review outlines the role of mitochondria in vascular function and discusses the ramifications of mitochondrial dysfunction on vascular health in the setting of age and disease. The adverse vascular consequences of increased mitochondrial-derived reactive oxygen species, impaired mitochondrial quality control, and defective mitochondrial calcium cycling are emphasized, in particular. Current evidence for both lifestyle and pharmaceutical mitochondrial-targeted strategies to improve vascular function is also presented.
Collapse
Affiliation(s)
- Danielle L Kirkman
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, Richmond, Virginia
| | | | - Matthew J Rossman
- Department of Integrative Physiology, University of Colorado, Boulder, Colorado
| | - Douglas R Seals
- Department of Integrative Physiology, University of Colorado, Boulder, Colorado
| | - David G Edwards
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, Delaware
| |
Collapse
|
31
|
Ozyel B, Le Gall G, Needs PW, Kroon PA. Anti-Inflammatory Effects of Quercetin on High-Glucose and Pro-Inflammatory Cytokine Challenged Vascular Endothelial Cell Metabolism. Mol Nutr Food Res 2021; 65:e2000777. [PMID: 33481349 PMCID: PMC8614122 DOI: 10.1002/mnfr.202000777] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 12/09/2020] [Indexed: 12/15/2022]
Abstract
SCOPE Pro-inflammatory stimuli such as hyperglycemia and cytokines have been shown to negatively affect endothelial cell functions. The aim of this study is to assess the potential of quercetin and its human metabolites to overcome the deleterious effects of hyperglycemic or inflammatory conditions on the vascular endothelium by modulating endothelial cell metabolism. METHODS AND RESULTS A metabolomics approach enabled identification and quantification of 27 human umbilical vein endothelial cell (HUVEC) metabolites. Treatment of HUVECs with high-glucose concentrations causes significant increases in lactate and glutamate concentrations. Quercetin inhibits glucose-induced increases in lactate and adenosine 5'-triphosphate (ATP) and also increased inosine concentrations. Tumor necrosis factor α-treatment (TNFα) of HUVECs causes increases in asparagine and decreases in aspartate concentrations. Co-treatment with quercetin reduces pyruvate concentrations compared to TNFα-only treated controls. Subsequently, it was shown that quercetin and its HUVEC phase-2 conjugates inhibit adenosine deaminase, xanthine oxidase and 5'nucleotidase (CD73) but not ectonucleoside triphosphate diphosphohydrolase-1 (CD39) or purine nucleoside phosphorylase activities. CONCLUSION Quercetin was shown to alter the balance of HUVEC metabolites towards a less inflamed phenotype, both alone and in the presence of pro-inflammatory stimuli. These changes are consistent with the inhibition of particular enzymes involved in purine metabolism by quercetin and its HUVEC metabolites.
Collapse
Affiliation(s)
- Besim Ozyel
- Nutrition and Dietetics DepartmentEuropean University of LefkeLefke, Northern Cyprus, TR‐10Mersin9910Turkey
| | - Gwénaëlle Le Gall
- Norwich Medical SchoolUniversity of East AngliaBob Champion Research and Education Building, James Watson Road, Norwich Research ParkNorwichNR4 7UQUK
| | - Paul W. Needs
- Quadram Institute BioscienceNorwich Research ParkNorwichNR4 7UQUK
| | - Paul A. Kroon
- Quadram Institute BioscienceNorwich Research ParkNorwichNR4 7UQUK
| |
Collapse
|
32
|
Wang J, Bai J, Duan P, Wang H, Li Y, Zhu Q. Kir6.1 improves cardiac dysfunction in diabetic cardiomyopathy via the AKT-FoxO1 signalling pathway. J Cell Mol Med 2021; 25:3935-3949. [PMID: 33547878 PMCID: PMC8051713 DOI: 10.1111/jcmm.16346] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 12/07/2020] [Accepted: 01/11/2021] [Indexed: 12/13/2022] Open
Abstract
Previous studies have shown that the expression of inwardly rectifying potassium channel 6.1 (Kir6.1) in heart mitochondria is significantly reduced in type 1 diabetes. However, whether its expression and function are changed and what role it plays in type 2 diabetic cardiomyopathy (DCM) have not been reported. This study investigated the role and mechanism of Kir6.1 in DCM. We found that the cardiac function and the Kir6.1 expression in DCM mice were decreased. We generated mice overexpressing or lacking Kir6.1 gene specifically in the heart. Kir6.1 overexpression improved cardiac dysfunction in DCM. Cardiac‐specific Kir6.1 knockout aggravated cardiac dysfunction. Kir6.1 regulated the phosphorylation of AKT and Foxo1 in DCM. We further found that Kir6.1 overexpression also improved cardiomyocyte dysfunction and up‐regulated the phosphorylation of AKT and FoxO1 in neonatal rat ventricular cardiomyocytes with insulin resistance. Furthermore, FoxO1 activation down‐regulated the expression of Kir6.1 and decreased the mitochondrial membrane potential (ΔΨm) in cardiomyocytes. FoxO1 inactivation up‐regulated the expression of Kir6.1 and increased the ΔΨm in cardiomyocytes. Chromatin immunoprecipitation assay demonstrated that the Kir6.1 promoter region contains a functional FoxO1‐binding site. In conclusion, Kir6.1 improves cardiac dysfunction in DCM, probably through the AKT‐FoxO1 signalling pathway.
Collapse
Affiliation(s)
- Jinxin Wang
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China.,Department of Geriatric Cardiology, Chinese PLA General Hospital, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Jing Bai
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Peng Duan
- Department of Cardiology, Chinese PLA No. 371 Hospital, Henan, China
| | - Hao Wang
- Department of Geriatric Cardiology, Chinese PLA General Hospital, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Yang Li
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Qinglei Zhu
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
33
|
Abstract
The accumulation of triglycerides (TGs) in macrophages induces cell death, a risk factor in the pathogenesis of atherosclerosis. We had previously reported that TG-induced macrophage death is triggered by caspase-1 and -2, therefore we investigated the mechanism underlying this phenomenon. We found that potassium efflux is increased in TG-treated THP-1 macrophages and that the inhibition of potassium efflux blocks TG-induced cell death as well as caspase-1 and -2 activation. Furthermore, reducing ATP concentration (known to induce potassium efflux), restored cell viability and caspase-1 and -2 activity. The activation of pannexin-1 (a channel that releases ATP), was increased after TG treatment in THP-1 macrophages. Inhibition of pannexin-1 activity using its inhibitor, probenecid, recovered cell viability and blocked the activation of caspase-1 and -2 in TG-treated macrophages. These results suggest that TG-induced THP-1 macrophage cell death is induced via pannexin-1 activation, which increases extracellular ATP, leading to an increase in potassium efflux.
Collapse
Affiliation(s)
- Byung Chul Jung
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 94720, United States
- Department of Biomedical Laboratory Science, College of Health Sciences, Yonsei University, Wonju 26493, Korea
| | - Sung Hoon Kim
- Department of Biomedical Laboratory Science, College of Health Sciences, Yonsei University, Wonju 26493, Korea
- Department of Biomedical Laboratory Science, Korea Nazarene University, Cheonan 31172, Korea
| | - Jaewon Lim
- Department of Biomedical Laboratory Science, College of Health Sciences, Yonsei University, Wonju 26493, Korea
- Department of Biomedical Laboratory Science, College of Medical Sciences, Daegu Haany University, Gyeongsan 38610, Korea
| | - Yoon Suk Kim
- Department of Biomedical Laboratory Science, College of Health Sciences, Yonsei University, Wonju 26493, Korea
| |
Collapse
|
34
|
The multifaceted roles of sulfane sulfur species in cancer-associated processes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2020; 1862:148338. [PMID: 33212042 DOI: 10.1016/j.bbabio.2020.148338] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/30/2020] [Accepted: 11/13/2020] [Indexed: 02/07/2023]
Abstract
Sulfane sulfur species comprise a variety of biologically relevant hydrogen sulfide (H2S)-derived species, including per- and poly-sulfidated low molecular weight compounds and proteins. A growing body of evidence suggests that H2S, currently recognized as a key signaling molecule in human physiology and pathophysiology, plays an important role in cancer biology by modulating cell bioenergetics and contributing to metabolic reprogramming. This is accomplished through functional modulation of target proteins via H2S binding to heme iron centers or H2S-mediated reversible per- or poly-sulfidation of specific cysteine residues. Since sulfane sulfur species are increasingly viewed not only as a major source of H2S but also as key mediators of some of the biological effects commonly attributed to H2S, the multifaceted role of these species in cancer biology is reviewed here with reference to H2S, focusing on their metabolism, signaling function, impact on cell bioenergetics and anti-tumoral properties.
Collapse
|
35
|
Abramavičius S, Volkevičiūtė A, Tunaitytė A, Venslauskas M, Bubulis A, Bajoriūnas V, Stankevičius E. Low-Frequency (20 kHz) Ultrasonic Modulation of Drug Action. ULTRASOUND IN MEDICINE & BIOLOGY 2020; 46:3017-3031. [PMID: 32768323 DOI: 10.1016/j.ultrasmedbio.2020.07.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 04/27/2020] [Accepted: 07/07/2020] [Indexed: 06/11/2023]
Abstract
We tested the effect of low-frequency ultrasound (LUS, 20 kHz, 4 W/cm2) on the function of rat mesentery and human pulmonary arteries with wire myography. The vessels were induced to contract with either noradrenaline or physiologic saline solution (PSS) with a high potassium concentration (KPSS) and then incubated with capsaicin (2.1 × 10-7 M, TRPV1 [transient receptor potential vanilloid 1] activator), dopamine (1 × 10-4 M, dopamine and α2-receptor activator), or fenoldopam (dopamineA1 receptor agonist, 1 × 10-4 M) with and without glibenclamide (1 μM, KATP [adenosine triphosphate {sensitive potassium channel (ATP)}-sensitive potassium channel] inhibitor and α2-receptor modulator), and insonated. Vessels were incubated in Ca2+-free PSS and induced to contract with added extracellular Ca2+ and noradrenaline. Pulmonary arteries were induced to contract with KPSS and dopamine. Then the vessels were insonated. LUS inhibited the influx of external Ca2+, inhibited the dopamine-induced vasoconstriction in the KPSS (glibenclamide reversible), reduced the capsaicin-induced vasorelaxation, increased the gentamicin-induced vasorelaxation and increased the dopamine-induced contraction in the KPSS in human pulmonary arteries.
Collapse
Affiliation(s)
- Silvijus Abramavičius
- Institute of Physiology and Pharmacology, Lithuanian University of Health Sciences, Kaunas, Lithuania; Laboratory of Preclinical Drug Investigation, Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania.
| | - Augusta Volkevičiūtė
- Institute of Physiology and Pharmacology, Lithuanian University of Health Sciences, Kaunas, Lithuania; Laboratory of Preclinical Drug Investigation, Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Agilė Tunaitytė
- Institute of Physiology and Pharmacology, Lithuanian University of Health Sciences, Kaunas, Lithuania; Laboratory of Preclinical Drug Investigation, Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Mantas Venslauskas
- Institute of Mechatronics, Kaunas University of Technology, Kaunas, Lithuania
| | - Algimantas Bubulis
- Institute of Mechatronics, Kaunas University of Technology, Kaunas, Lithuania
| | - Vytis Bajoriūnas
- Institute of Physiology and Pharmacology, Lithuanian University of Health Sciences, Kaunas, Lithuania; Laboratory of Preclinical Drug Investigation, Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Edgaras Stankevičius
- Institute of Physiology and Pharmacology, Lithuanian University of Health Sciences, Kaunas, Lithuania; Laboratory of Preclinical Drug Investigation, Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| |
Collapse
|
36
|
Coburn RF. Coronary and cerebral metabolism-blood flow coupling and pulmonary alveolar ventilation-blood flow coupling may be disabled during acute carbon monoxide poisoning. J Appl Physiol (1985) 2020; 129:1039-1050. [PMID: 32853110 DOI: 10.1152/japplphysiol.00172.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Current evidence indicates that the toxicity of carbon monoxide (CO) poisoning results from increases in reactive oxygen species (ROS) generation plus tissue hypoxia resulting from decreases in capillary Po2 evoked by effects of increases in blood [carboxyhemoglobin] on the oxyhemoglobin dissociation curve. There has not been consideration of how increases in Pco could influence metabolism-blood flow coupling, a physiological mechanism that regulates the uniformity of tissue Po2, and alveolar ventilation-blood flow coupling, a mechanism that increases the efficiency of pulmonary O2 uptake. Using published data, I consider hypotheses that these coupling mechanisms, triggered by O2 and CO sensors located in arterial and arteriolar vessels in the coronary and cerebral circulations and in lung intralobar arteries, are disrupted during acute CO poisoning. These hypotheses are supported by calculations that show that the Pco in these vessels can reach levels during CO poisoning that would exert effects on signal transduction molecules involved in these coupling mechanisms.NEW & NOTEWORTHY This article introduces and supports a postulate that the tissue hypoxia component of carbon monoxide poisoning results in part from impairment of physiological adaptation mechanisms whereby tissues can match regional blood flow to O2 uptake, and the lung can match regional blood flow to alveolar ventilation.
Collapse
Affiliation(s)
- Ronald F Coburn
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
37
|
Glucagon-Like Peptide-1 Analog Liraglutide Attenuates Pressure-Overload Induced Cardiac Hypertrophy and Apoptosis through Activating ATP Sensitive Potassium Channels. Cardiovasc Drugs Ther 2020; 35:87-101. [PMID: 33057968 DOI: 10.1007/s10557-020-07088-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/22/2020] [Indexed: 12/16/2022]
Abstract
PURPOSE This study aimed to investigate whether inhibition of glucagon-like peptide-1 (GLP-1) on pressure overload induced cardiac hypertrophy and apoptosis is related to activation of ATP sensitive potassium (KATP) channels. METHODS Male SD rats were randomly divided into five groups: sham, control (abdominal aortic constriction), GLP-1 analog liraglutide (0.3 mg/kg/twice day), KATP channel blocker glibenclamide (5 mg/kg/day), and liraglutide plus glibenclamide. RESULTS Relative to the control on week 16, liraglutide upregulated protein and mRNA levels of KATP channel subunits Kir6.2/SUR2 and their expression in the myocardium, vascular smooth muscle, aortic endothelium, and cardiac microvasculature. Consistent with a reduction in aortic wall thickness (61.4 ± 7.6 vs. 75.0 ± 7.6 μm, p < 0.05), liraglutide enhanced maximal aortic endothelium-dependent relaxation in response to acetylcholine (71.9 ± 8.7 vs. 38.6 ± 4.8%, p < 0.05). Along with a reduction in heart to body weight ratio (2.6 ± 0.1 vs. 3.4 ± 0.4, mg/g, p < 0.05) by liraglutide, hypertrophied cardiomyocytes (371.0 ± 34.4 vs. 933.6 ± 156.6 μm2, p < 0.05) and apoptotic cells (17.5 ± 8.2 vs. 44.7 ± 7.9%, p < 0.05) were reduced. Expression of anti-apoptotic protein BCL-2 and contents of myocardial ATP were augmented, and expression of cleaved-caspase 3 and levels of serum Tn-I/-T were reduced. Echocardiography and hemodynamic measurement showed that cardiac systolic function was enhanced as evidenced by increased ejection fraction (88.4 ± 4.8 vs. 73.8 ± 5.1%, p < 0.05) and left ventricular systolic pressure (105.2 ± 10.8 vs. 82.7 ± 7.9 mmHg, p < 0.05), and diastolic function was preserved as shown by a reduction of ventricular end-diastolic pressure (-3.1 ± 2.9 vs. 6.7 ± 2.8 mmHg, p < 0.05). Furthermore, left ventricular internal diameter at end-diastole (5.8 ± 0.5 vs. 7.7 ± 0.6 mm, p < 0.05) and left ventricular internal diameter at end-systole (3.0 ± 0.6 vs. 4.7 ± 0.4 mm, p < 0.05) were improved. Dietary administration of glibenclamide alone did not alter all the parameters measured but significantly blocked liraglutide-exerted cardioprotection. CONCLUSION Liraglutide ameliorates cardiac hypertrophy and apoptosis, potentially via activating KATP channel-mediated signaling pathway. These data suggest that liraglutide might be considered as an adjuvant therapy to treat patients with heart failure.
Collapse
|
38
|
Li Y, Aziz Q, Anderson N, Ojake L, Tinker A. Endothelial ATP-Sensitive Potassium Channel Protects Against the Development of Hypertension and Atherosclerosis. Hypertension 2020; 76:776-784. [PMID: 32654556 PMCID: PMC7418932 DOI: 10.1161/hypertensionaha.120.15355] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 05/18/2020] [Accepted: 06/19/2020] [Indexed: 12/28/2022]
Abstract
In the endothelium, ATP-sensitive potassium (KATP) channels are thought to couple cellular metabolism with membrane excitability, calcium entry, and endothelial mediator release. We hypothesized that endothelial KATP channels have a broad role protecting against high blood pressure and atherosclerosis. Endothelial-specific Kir6.1 KO mice (eKO) and eKO mice on an apolipoprotein E KO background were generated (A-eKO) to investigate the role of KATP channels in the endothelium. Basal blood pressure was not elevated in eKO mice. However, when challenged with a high-salt diet and the eNOS inhibitor L-NAME, eKO mice became more hypertensive than their littermate controls. In aorta, NO release at least partly contributes to the endothelium-dependent vasorelaxation induced by pinacidil. In A-eKO mice atherosclerotic plaque density was significantly greater than in their littermate controls when challenged with a high-fat diet, particularly in the aortic arch region. Levels of endothelial dysfunction markers were higher in eKO compared with WT mice; however, these were not significant for A-eKO mice compared with their littermate controls. Furthermore, decreased vascular reactivity was observed in the mesenteric arteries of A-eKO mice, but not in aorta when on a high-fat diet. Our data support a role for endothelial Kir6.1-containing KATP channels in the endothelial protection against environmental stressors: the maintenance of blood pressure homeostasis in response to high salt and endothelial integrity when challenged with a high-fat diet.
Collapse
Affiliation(s)
- Yiwen Li
- The Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom
| | - Qadeer Aziz
- The Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom
| | - Naomi Anderson
- The Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom
| | - Leona Ojake
- The Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom
| | - Andrew Tinker
- The Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom
| |
Collapse
|
39
|
Zhang DM, Lin YF. Functional modulation of sarcolemmal K ATP channels by atrial natriuretic peptide-elicited intracellular signaling in adult rabbit ventricular cardiomyocytes. Am J Physiol Cell Physiol 2020; 319:C194-C207. [PMID: 32432931 DOI: 10.1152/ajpcell.00409.2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
ATP-sensitive potassium (KATP) channels couple cell metabolic status to membrane excitability and are crucial for stress adaptation and cytoprotection in the heart. Atrial natriuretic peptide (ANP), a cardiac peptide important for cardiovascular homeostasis, also exhibits cytoprotective features including protection against myocardial ischemia-reperfusion injuries. However, how ANP modulates cardiac KATP channels is largely unknown. In the present study we sought to address this issue by investigating the role of ANP signaling in functional modulation of sarcolemmal KATP (sarcKATP) channels in ventricular myocytes freshly isolated from adult rabbit hearts. Single-channel recordings were performed in combination with pharmacological approaches in the cell-attached patch configuration. Bath application of ANP markedly potentiated sarcKATP channel activities induced by metabolic inhibition with sodium azide, whereas the KATP-stimulating effect of ANP was abrogated by selective inhibition of the natriuretic peptide receptor type A (NPR-A), cGMP-dependent protein kinase (PKG), reactive oxygen species (ROS), extracellular signal-regulated protein kinase (ERK)1/2, Ca2+/calmodulin-dependent protein kinase II (CaMKII), or the ryanodine receptor (RyR). Blockade of RyRs also nullified hydrogen peroxide (H2O2)-induced stimulation of sarcKATP channels in intact cells. Furthermore, single-channel kinetic analyses revealed that ANP enhanced the function of ventricular sarcKATP channels through destabilizing the long closures and facilitating the opening transitions, without affecting the single-channel conductance. In conclusion, here we report that ANP positively modulates the activity of ventricular sarcKATP channels via an intracellular signaling mechanism consisting of NPR-A, PKG, ROS, ERK1/2, CaMKII, and RyR2. This novel mechanism may regulate cardiac excitability and contribute to cytoprotection, in part, by opening myocardial KATP channels.
Collapse
Affiliation(s)
- Dai-Min Zhang
- Department of Physiology and Membrane Biology, University of California, Davis, California
| | - Yu-Fung Lin
- Department of Physiology and Membrane Biology, University of California, Davis, California.,Department of Anesthesiology and Pain Medicine, University of California, Davis, California
| |
Collapse
|
40
|
Huang Y, Hu D, Huang C, Nichols CG. Genetic Discovery of ATP-Sensitive K + Channels in Cardiovascular Diseases. Circ Arrhythm Electrophysiol 2020; 12:e007322. [PMID: 31030551 DOI: 10.1161/circep.119.007322] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The ATP-sensitive K+ (KATP) channels are hetero-octameric protein complexes comprising 4 pore-forming (Kir6.x) subunits and 4 regulatory sulfonylurea receptor (SURx) subunits. They are prominent in myocytes, pancreatic β cells, and neurons and link cellular metabolism with membrane excitability. Using genetically modified animals and genomic analysis in patients, recent studies have implicated certain ATP-sensitive K+ channel subtypes in physiological and pathological processes in a variety of cardiovascular diseases. In this review, we focus on the causal relationship between ATP-sensitive K+ channel activity and pathophysiology in the cardiovascular system, particularly from the perspective of genetic changes in human and animal models.
Collapse
Affiliation(s)
- Yan Huang
- Department of Cardiology, Renmin Hospital of Wuhan University; Cardiovascular Research Institute, Wuhan University, PR China (Y.H., D.H., C.H.).,Hubei Key Laboratory of Cardiology, Wuhan, PR China (Y.H., D.H., C.H.)
| | - Dan Hu
- Department of Cardiology, Renmin Hospital of Wuhan University; Cardiovascular Research Institute, Wuhan University, PR China (Y.H., D.H., C.H.).,Hubei Key Laboratory of Cardiology, Wuhan, PR China (Y.H., D.H., C.H.)
| | - Congxin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University; Cardiovascular Research Institute, Wuhan University, PR China (Y.H., D.H., C.H.).,Hubei Key Laboratory of Cardiology, Wuhan, PR China (Y.H., D.H., C.H.)
| | - Colin G Nichols
- Center for the Investigation of Membrane Excitability Diseases and Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, MO (C.G.N.)
| |
Collapse
|
41
|
Breen MS, Dobbyn A, Li Q, Roussos P, Hoffman GE, Stahl E, Chess A, Sklar P, Li JB, Devlin B, Buxbaum JD. Global landscape and genetic regulation of RNA editing in cortical samples from individuals with schizophrenia. Nat Neurosci 2019; 22:1402-1412. [PMID: 31455887 PMCID: PMC6791127 DOI: 10.1038/s41593-019-0463-7] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 07/09/2019] [Indexed: 12/28/2022]
Abstract
RNA editing critically regulates neurodevelopment and normal neuronal function. The global landscape of RNA editing was surveyed across 364 schizophrenia cases and 383 control postmortem brain samples from the CommonMind Consortium, comprising two regions: dorsolateral prefrontal cortex and anterior cingulate cortex. In schizophrenia, RNA editing sites in genes encoding AMPA-type glutamate receptors and postsynaptic density proteins were less edited, whereas those encoding translation initiation machinery were edited more. These sites replicate between brain regions, map to 3'-untranslated regions and intronic regions, share common sequence motifs and overlap with binding sites for RNA-binding proteins crucial for neurodevelopment. These findings cross-validate in hundreds of non-overlapping dorsolateral prefrontal cortex samples. Furthermore, ~30% of RNA editing sites associate with cis-regulatory variants (editing quantitative trait loci or edQTLs). Fine-mapping edQTLs with schizophrenia risk loci revealed co-localization of eleven edQTLs with six loci. The findings demonstrate widespread altered RNA editing in schizophrenia and its genetic regulation, and suggest a causal and mechanistic role of RNA editing in schizophrenia neuropathology.
Collapse
Affiliation(s)
- Michael S Breen
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Amanda Dobbyn
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Qin Li
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Panos Roussos
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research, Education, and Clinical Center (VISN 2 South), James J. Peters VA Medical Center, Bronx, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gabriel E Hoffman
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technologies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eli Stahl
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technologies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Andrew Chess
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technologies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Cell Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Pamela Sklar
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technologies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jin Billy Li
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Bernie Devlin
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Joseph D Buxbaum
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
42
|
Shi R, Xu JW, Xiao ZT, Chen RF, Zhang YL, Lin JB, Cheng KL, Wei GY, Li PB, Zhou WL, Su WW. Naringin and Naringenin Relax Rat Tracheal Smooth by Regulating BK Ca Activation. J Med Food 2019; 22:963-970. [PMID: 31259654 DOI: 10.1089/jmf.2018.4364] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Naringin and its aglycone, naringenin, occur naturally in our regular diet and traditional Chinese medicines. This study aimed to detect an effective therapeutic approach for cough variant asthma (CVA) through evaluating the relaxant effect of these two bioactive herbal monomers as antitussive and antiasthmatic on rat tracheal smooth muscle. The relaxant effect was determined by measuring muscular tension with a mechanical recording system in rat tracheal rings. Cytosolic Ca2+ concentration was measured using a confocal imaging system in primary cultured tracheal smooth muscle cells. In rat tracheal rings, addition of both naringin and naringenin could concentration dependently relax carbachol (CCh)-evoked tonic contraction. This epithelium-independent relaxation could be suppressed by BaCl2, tetraethylammonium, and iberiotoxin (IbTX), but not by glibenclamide. After stimulating primary cultured tracheal smooth muscle cells by CCh or high KCl, the intracellular Ca2+ increase could be inhibited by both naringin and naringenin, respectively. This reaction was also suppressed by IbTX. These results demonstrate that both naringin and naringenin can relax tracheal smooth muscle through opening big conductance Ca2+-activated K+ channel, which mediates plasma membrane hyperpolarization and reduces Ca2+ influx. Our data indicate a potentially effective therapeutic approach of naringin and naringenin for CVA.
Collapse
Affiliation(s)
- Rui Shi
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Reevaluation of Postmarket Traditional Chinese Medicine, School of Life Sciences, Sun Yat-sen University, Guangzhou, China.,State Key Laboratory of Biocontrol and Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, China.,School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Jia-Wen Xu
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Zi-Ting Xiao
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Reevaluation of Postmarket Traditional Chinese Medicine, School of Life Sciences, Sun Yat-sen University, Guangzhou, China.,State Key Laboratory of Biocontrol and Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, China.,School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Ruo-Fei Chen
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yi-Lin Zhang
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Jia-Bi Lin
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Reevaluation of Postmarket Traditional Chinese Medicine, School of Life Sciences, Sun Yat-sen University, Guangzhou, China.,State Key Laboratory of Biocontrol and Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, China.,School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Ke-Ling Cheng
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Reevaluation of Postmarket Traditional Chinese Medicine, School of Life Sciences, Sun Yat-sen University, Guangzhou, China.,State Key Laboratory of Biocontrol and Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, China.,School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Gu-Yi Wei
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Reevaluation of Postmarket Traditional Chinese Medicine, School of Life Sciences, Sun Yat-sen University, Guangzhou, China.,State Key Laboratory of Biocontrol and Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, China.,School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Pei-Bo Li
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Reevaluation of Postmarket Traditional Chinese Medicine, School of Life Sciences, Sun Yat-sen University, Guangzhou, China.,State Key Laboratory of Biocontrol and Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, China.,School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Wen-Liang Zhou
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Wei-Wei Su
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Reevaluation of Postmarket Traditional Chinese Medicine, School of Life Sciences, Sun Yat-sen University, Guangzhou, China.,State Key Laboratory of Biocontrol and Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, China.,School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
43
|
Saponara S, Fusi F, Spiga O, Trezza A, Hopkins B, Brimble MA, Rennison D, Bova S. The Selective Rat Toxicant Norbormide Blocks K ATP Channels in Smooth Muscle Cells But Not in Insulin-Secreting Cells. Front Pharmacol 2019; 10:598. [PMID: 31191321 PMCID: PMC6540933 DOI: 10.3389/fphar.2019.00598] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 05/09/2019] [Indexed: 12/02/2022] Open
Abstract
Norbormide is a toxicant selective for rats to which it induces a widespread vasoconstriction. In a recent paper, we hypothesized a role of ATP-sensitive potassium (KATP) channels in norbormide-induced vasoconstriction. The current study was undertaken to verify this hypothesis by comparing the effects of norbormide with those of glibenclamide, a known KATP channel blocker. The whole-cell patch-clamp method was used to record KATP currents in myocytes freshly isolated from the rat and mouse caudal artery and from the rat gastric fundus, as well as in insulin-secreting pancreatic beta cells (INS-1 cells). Smooth muscle contractile function was assessed on either rat caudal artery rings or gastric fundus strips. Molecular modeling and docking simulation to KATP channel proteins were investigated in silico. Both norbormide (a racemic mixture of endo and exo isomers) and glibenclamide inhibited KATP currents in rat and mouse caudal artery myocytes, as well as in gastric fundus smooth muscle cells. In rat INS-1 cells, only glibenclamide blocked KATP channels, whereas norbormide was ineffective. The inhibitory effect of norbormide in rat caudal artery myocytes was not stereo-specific as both the endo isomers (active as vasoconstrictor) and the exo isomers (inactive as vasoconstrictor) had similar inhibitory activity. In rat caudal artery rings, norbormide-induced contraction was partially reverted by the KATP channel opener pinacidil. Computational approaches indicated the SUR subunit of KATP channels as the binding site for norbormide. KATP channel inhibition may play a role in norbormide-induced vasoconstriction, but does not explain the species selectivity, tissue selectivity, and stereoselectivity of its constricting activity. The lack of effect in INS-1 cells suggests a potential selectivity of norbormide for smooth muscle KATP channels.
Collapse
Affiliation(s)
- Simona Saponara
- Department of Life Sciences, University of Siena, Siena, Italy
| | - Fabio Fusi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Ottavia Spiga
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Alfonso Trezza
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Brian Hopkins
- Landcare Research, Lincoln, New Zealand.,School of Chemical Sciences, University of Auckland, Auckland, New Zealand
| | - Margaret A Brimble
- School of Chemical Sciences, University of Auckland, Auckland, New Zealand
| | - David Rennison
- School of Chemical Sciences, University of Auckland, Auckland, New Zealand
| | - Sergio Bova
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| |
Collapse
|
44
|
Wang Y, Hall LM, Kujawa M, Li H, Zhang X, O'Meara M, Ichinose T, Wang JM. Methylglyoxal triggers human aortic endothelial cell dysfunction via modulation of the K ATP/MAPK pathway. Am J Physiol Cell Physiol 2019; 317:C68-C81. [PMID: 30995106 DOI: 10.1152/ajpcell.00117.2018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Endothelial dysfunction is a key risk factor in diabetes-related multiorgan damage. Methylglyoxal (MGO), a highly reactive dicarbonyl generated primarily as a by-product of glycolysis, is increased in both type 1 and type 2 diabetic patients. MGO can rapidly bind with proteins, nucleic acids, and lipids, resulting in structural and functional changes. MGO can also form advanced glycation end products (AGEs). How MGO causes endothelial cell dysfunction, however, is not clear. Human aortic endothelial cells (HAECs) from healthy (H-HAECs) and type 2 diabetic (D-HAECs) donors were cultured in endothelial growth medium (EGM-2). D-HAECs demonstrated impaired network formation (on Matrigel) and proliferation (MTT assay), as well as increased apoptosis (caspase-3/7 activity and TUNEL staining), compared with H-HAECs. High glucose (25 mM) or AGEs (200 ng/ml) did not induce such immediate, detrimental effects as MGO (10 µM). H-HAECs were treated with MGO (10 µM) for 24 h with or without the ATP-sensitive potassium (KATP) channel antagonist glibenclamide (1 µM). MGO significantly impaired H-HAEC network formation and proliferation and induced cell apoptosis, which was reversed by glibenclamide. Furthermore, siRNA against the KATP channel protein Kir6.1 significantly inhibited endothelial cell function at basal status but rescued impaired endothelial cell function upon MGO exposure. Meanwhile, activation of MAPK pathways p38 kinase, c-Jun NH2-terminal kinase (JNK), and extracellular signal-regulated kinase (ERK) (determined by Western blot analyses of their phosphorylated forms, p-JNK, p-p38, and p-ERK) in D-HAECs were significantly enhanced compared with those in H-HAECs. MGO exposure enhanced the activation of all three MAPK pathways in H-HAECs, whereas glibenclamide reversed the activation of p-stress-activated protein kinase/JNK induced by MGO. Glyoxalase-1 (GLO1) is the endogenous MGO-detoxifying enzyme. In healthy mice that received an inhibitor of GLO1, MGO deposition in aortic wall was enhanced and endothelial cell sprouting from isolated aortic segment was significantly inhibited. Our data suggest that MGO triggers endothelial cell dysfunction by activating the JNK/p38 MAPK pathway. This effect arises partly through activation of KATP channels. By understanding how MGO induces endothelial dysfunction, our study may provide useful information for developing MGO-targeted interventions to treat vascular disorders in diabetes.
Collapse
Affiliation(s)
- Yihan Wang
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University , Detroit, Michigan
| | - Leo M Hall
- Department of Ophthalmology, Visual and Anatomical Sciences, School of Medicine, Wayne State University , Detroit, Michigan
| | - Marisa Kujawa
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University , Detroit, Michigan
| | - Hainan Li
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University , Detroit, Michigan
| | - Xiang Zhang
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University , Detroit, Michigan
| | - Megan O'Meara
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University , Detroit, Michigan
| | - Tomomi Ichinose
- Department of Ophthalmology, Visual and Anatomical Sciences, School of Medicine, Wayne State University , Detroit, Michigan
| | - Jie-Mei Wang
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University , Detroit, Michigan.,Centers for Molecular Medicine and Genetics, Wayne State University , Detroit, Michigan.,Cardiovascular Research Institute, Wayne State University, Detroit, Michigan
| |
Collapse
|
45
|
Pirotte B, Florence X, Goffin E, Lebrun P. 2,2-Dimethyl-3,4-dihydro-2 H-1,4-benzoxazines as isosteres of 2,2-dimethylchromans acting as inhibitors of insulin release and vascular smooth muscle relaxants. MEDCHEMCOMM 2019; 10:431-438. [PMID: 31015906 DOI: 10.1039/c8md00593a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 02/01/2019] [Indexed: 11/21/2022]
Abstract
The present study describes the synthesis and biological evaluation of 4-phenylureido/thioureido-substituted 2,2-dimethyl-3,4-dihydro-2H-1,4-benzoxazines as isosteres of corresponding 2,2-dimethylchromans reported to be pancreatic β-cell KATP channel openers. The benzoxazines were found to be less active as inhibitors of the glucose-induced insulin release than their corresponding chromans, while the myorelaxant activity of some 4-arylureido-substituted benzoxazines was more pronounced than that exhibited by their chroman counterparts. The myorelaxant activity of the most potent benzoxazine 8e was further characterized on rat aortic rings precontracted by 30 mM KCl in the presence of glibenclamide (10 μM) or precontracted by 80 mM extracellular KCl. Our findings indicate that, on vascular smooth muscle cells, the benzoxazine 8e mainly behaved as a calcium entry blocker.
Collapse
Affiliation(s)
- Bernard Pirotte
- Laboratoire de Chimie Pharmaceutique , Center for Interdisciplinary Research on Medicines (CIRM) , Université de Liège , Quartier Hôpital , Avenue Hippocrate 15 , B-4000 Liège , Belgium .
| | - Xavier Florence
- Laboratoire de Chimie Pharmaceutique , Center for Interdisciplinary Research on Medicines (CIRM) , Université de Liège , Quartier Hôpital , Avenue Hippocrate 15 , B-4000 Liège , Belgium . .,Laboratoire de Physiologie et Pharmacologie , Université Libre de Bruxelles , Faculté de Médecine , 808 Route de Lennik , B-1070 Bruxelles , Belgium
| | - Eric Goffin
- Laboratoire de Chimie Pharmaceutique , Center for Interdisciplinary Research on Medicines (CIRM) , Université de Liège , Quartier Hôpital , Avenue Hippocrate 15 , B-4000 Liège , Belgium .
| | - Philippe Lebrun
- Laboratoire de Physiologie et Pharmacologie , Université Libre de Bruxelles , Faculté de Médecine , 808 Route de Lennik , B-1070 Bruxelles , Belgium
| |
Collapse
|
46
|
Ye P, Zhu Y, Gu Y, Zhang D, Chen S. Functional protection against cardiac diseases depends on ATP-sensitive potassium channels. J Cell Mol Med 2018; 22:5801-5806. [PMID: 30596400 PMCID: PMC6237599 DOI: 10.1111/jcmm.13893] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 08/12/2018] [Indexed: 12/20/2022] Open
Abstract
ATP-sensitive potassium channels (KATP) channels are widely distributed in various tissues, including pancreatic beta cells, muscle tissue and brain tissue. KATP channels play an important role in cardioprotection in physiological/pathological situations. KATP channels are inhibited by an increase in the intracellular ATP concentration and are stimulated by an increase in the intracellular MgADP concentration. Activation of KATP channels decreases ischaemia/reperfusion injury, protects cardiomyocytes from heart failure, and reduces the occurrence of arrhythmias. KATP channels are involved in various signalling pathways, and their participation in protective processes is regulated by endogenous signalling molecules, such as nitric oxide and hydrogen sulphide. KATP channels may act as a new drug target to fight against cardiovascular disease in the development of related drugs in the future. This review highlights the potential mechanisms correlated with the protective role of KATP channels and their therapeutic value in cardiovascular diseases.
Collapse
Affiliation(s)
- Peng Ye
- Department of CardiologyNanjing First HospitalNanjing Medical UniversityJiangsuChina
| | - Yan‐Rong Zhu
- Department of CardiologyNanjing First HospitalNanjing Medical UniversityJiangsuChina
| | - Yue Gu
- Department of CardiologyNanjing First HospitalNanjing Medical UniversityJiangsuChina
| | - Dai‐Min Zhang
- Department of CardiologyNanjing First HospitalNanjing Medical UniversityJiangsuChina
| | - Shao‐Liang Chen
- Department of CardiologyNanjing First HospitalNanjing Medical UniversityJiangsuChina
| |
Collapse
|
47
|
Zhong CJ, Chen MM, Lu M, Ding JH, Du RH, Hu G. Astrocyte-specific deletion of Kir6.1/K-ATP channel aggravates cerebral ischemia/reperfusion injury through endoplasmic reticulum stress in mice. Exp Neurol 2018; 311:225-233. [PMID: 30315808 DOI: 10.1016/j.expneurol.2018.10.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 09/27/2018] [Accepted: 10/09/2018] [Indexed: 12/16/2022]
Abstract
ATP-sensitive potassium (K-ATP) channels, coupling cell metabolism to cell membrane potential, are involved in brain diseases including stroke. Emerging evidence shows that astrocytes play important roles in the pathophysiology of cerebral ischemia. Kir6.1, a pore-forming subunit of K-ATP channel, is prominently expressed in astrocytes and participates in regulating its function. However, the exact role of astrocytic Kir6.1-containg K-ATP channel (Kir6.1/K-ATP) in ischemic stroke remains unclear. Here, we found that astrocytic Kir6.1 knockout (KO) mice exhibited larger infarct areas and more severe brain edema and neurological deficits in middle cerebral artery occlusion stroke model. Both activated gliosis and neuronal loss were aggravated in astrocytic Kir6.1 KO mice. Furthermore, the protein levels of pro-apoptotic protein Bcl-2 associated X (Bax) and active caspase-3 were up-regulated and the expression of anti-apoptotic protein Bcl-2 was down-regulated in astrocytic Kir6.1 KO mice. This is accompanied by enhanced endoplasmic reticulum stress (ER stress) responses in brain tissues and in astrocytes during ischemia/reperfusion (I/R) injury. Finally, inhibition of ER stress rescued astrocyte apoptosis induced by Kir6.1 deletion during I/R injury. Collectively, our findings reveal that astrocytic Kir6.1/K-ATP channel protects brain from cerebral ischemia/reperfusion injury through inhibiting ER stress and suggest that astrocytic Kir6.1/K-ATP channel is a promising therapeutic target for ischemic stroke.
Collapse
Affiliation(s)
- Chong-Jin Zhong
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Nongmian Avenue, Nanjing, Jiangsu 211166, PR China
| | - Miao-Miao Chen
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Nongmian Avenue, Nanjing, Jiangsu 211166, PR China
| | - Ming Lu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Nongmian Avenue, Nanjing, Jiangsu 211166, PR China
| | - Jian-Hua Ding
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Nongmian Avenue, Nanjing, Jiangsu 211166, PR China
| | - Ren-Hong Du
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Nongmian Avenue, Nanjing, Jiangsu 211166, PR China.
| | - Gang Hu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Nongmian Avenue, Nanjing, Jiangsu 211166, PR China; Department of Pharmacology, Nanjing University of Chinese Medicine, 138 Xianlin, PR China.
| |
Collapse
|
48
|
Bernardo BC, Ooi JYY, Weeks KL, Patterson NL, McMullen JR. Understanding Key Mechanisms of Exercise-Induced Cardiac Protection to Mitigate Disease: Current Knowledge and Emerging Concepts. Physiol Rev 2018; 98:419-475. [PMID: 29351515 DOI: 10.1152/physrev.00043.2016] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The benefits of exercise on the heart are well recognized, and clinical studies have demonstrated that exercise is an intervention that can improve cardiac function in heart failure patients. This has led to significant research into understanding the key mechanisms responsible for exercise-induced cardiac protection. Here, we summarize molecular mechanisms that regulate exercise-induced cardiac myocyte growth and proliferation. We discuss in detail the effects of exercise on other cardiac cells, organelles, and systems that have received less or little attention and require further investigation. This includes cardiac excitation and contraction, mitochondrial adaptations, cellular stress responses to promote survival (heat shock response, ubiquitin-proteasome system, autophagy-lysosomal system, endoplasmic reticulum unfolded protein response, DNA damage response), extracellular matrix, inflammatory response, and organ-to-organ crosstalk. We summarize therapeutic strategies targeting known regulators of exercise-induced protection and the challenges translating findings from bench to bedside. We conclude that technological advancements that allow for in-depth profiling of the genome, transcriptome, proteome and metabolome, combined with animal and human studies, provide new opportunities for comprehensively defining the signaling and regulatory aspects of cell/organelle functions that underpin the protective properties of exercise. This is likely to lead to the identification of novel biomarkers and therapeutic targets for heart disease.
Collapse
Affiliation(s)
- Bianca C Bernardo
- Baker Heart and Diabetes Institute , Melbourne , Australia ; Department of Paediatrics, University of Melbourne , Victoria , Australia ; Department of Diabetes, Central Clinical School, Monash University , Victoria , Australia ; Department of Medicine, Central Clinical School, Monash University , Victoria , Australia ; and Department of Physiology, School of Biomedical Sciences , Victoria , Australia
| | - Jenny Y Y Ooi
- Baker Heart and Diabetes Institute , Melbourne , Australia ; Department of Paediatrics, University of Melbourne , Victoria , Australia ; Department of Diabetes, Central Clinical School, Monash University , Victoria , Australia ; Department of Medicine, Central Clinical School, Monash University , Victoria , Australia ; and Department of Physiology, School of Biomedical Sciences , Victoria , Australia
| | - Kate L Weeks
- Baker Heart and Diabetes Institute , Melbourne , Australia ; Department of Paediatrics, University of Melbourne , Victoria , Australia ; Department of Diabetes, Central Clinical School, Monash University , Victoria , Australia ; Department of Medicine, Central Clinical School, Monash University , Victoria , Australia ; and Department of Physiology, School of Biomedical Sciences , Victoria , Australia
| | - Natalie L Patterson
- Baker Heart and Diabetes Institute , Melbourne , Australia ; Department of Paediatrics, University of Melbourne , Victoria , Australia ; Department of Diabetes, Central Clinical School, Monash University , Victoria , Australia ; Department of Medicine, Central Clinical School, Monash University , Victoria , Australia ; and Department of Physiology, School of Biomedical Sciences , Victoria , Australia
| | - Julie R McMullen
- Baker Heart and Diabetes Institute , Melbourne , Australia ; Department of Paediatrics, University of Melbourne , Victoria , Australia ; Department of Diabetes, Central Clinical School, Monash University , Victoria , Australia ; Department of Medicine, Central Clinical School, Monash University , Victoria , Australia ; and Department of Physiology, School of Biomedical Sciences , Victoria , Australia
| |
Collapse
|
49
|
Purinergic Vasotoxicity: Role of the Pore/Oxidant/K ATP Channel/Ca 2+ Pathway in P2X 7-Induced Cell Death in Retinal Capillaries. Vision (Basel) 2018; 2. [PMID: 30288454 PMCID: PMC6166475 DOI: 10.3390/vision2030025] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
P2X7 receptor/channels in the retinal microvasculature not only regulate vasomotor activity, but can also trigger cells in the capillaries to die. While it is known that this purinergic vasotoxicity is dependent on the transmembrane pores that form during P2X7 activation, events linking pore formation with cell death remain uncertain. To better understand this pathophysiological process, we used YO-PRO-1 uptake, dichlorofluorescein fluorescence, perforated-patch recordings, fura-2 imaging and trypan blue dye exclusion to assess the effects of the P2X7 agonist, benzoylbenzoyl-ATP (BzATP), on pore formation, oxidant production, ion channel activation, [Ca2+]i and cell viability. Experiments demonstrated that exposure of retinal microvessels to BzATP increases capillary cell oxidants via a mechanism dependent on pore formation and the enzyme, NADPH oxidase. Indicative that oxidation plays a key role in purinergic vasotoxicity, an inhibitor of this enzyme completely prevented BzATP-induced death. We further discovered that vasotoxicity was boosted 4-fold by a pathway involving the oxidation-driven activation of hyperpolarizing KATP channels and the resulting increase in calcium influx. Our findings revealed that the previously unappreciated pore/oxidant/KATP channel/Ca2+ pathway accounts for 75% of the capillary cell death triggered by sustained activation of P2X7 receptor/channels. Elucidation of this pathway is of potential therapeutic importance since purinergic vasotoxicity may play a role in sight-threatening disorders such as diabetic retinopathy.
Collapse
|
50
|
Trezza A, Cicaloni V, Porciatti P, Langella A, Fusi F, Saponara S, Spiga O. From in silico to in vitro: a trip to reveal flavonoid binding on the Rattus norvegicus Kir6.1 ATP-sensitive inward rectifier potassium channel. PeerJ 2018; 6:e4680. [PMID: 29736333 PMCID: PMC5936070 DOI: 10.7717/peerj.4680] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 04/09/2018] [Indexed: 12/18/2022] Open
Abstract
Background ATP-sensitive inward rectifier potassium channels (Kir), are a potassium channel family involved in many physiological processes. KATP dysfunctions are observed in several diseases such as hypoglycaemia, hyperinsulinemia, Prinzmetal angina–like symptoms, cardiovascular diseases. Methods A broader view of the KATP mechanism is needed in order to operate on their regulation, and in this work we clarify the structure of the Rattus norvegicus ATP-sensitive inward rectifier potassium channel 8 (Kir6.1), which has been obtained through a homology modelling procedure. Due to the medical use of flavonoids, a considerable increase in studies on their influence on human health has recently been observed, therefore our aim is to study, through computational methods, the three-dimensional (3D) conformation together with mechanism of action of Kir6.1 with three flavonoids. Results Computational analysis by performing molecular dynamics (MD) and docking simulation on rat 3D modelled structure have been completed, in its closed and open conformation state and in complex with Quercetin, 5-Hydroxyflavone and Rutin flavonoids. Our study showed that only Quercetin and 5-Hydroxyflavone were responsible for a significant down-regulation of the Kir6.1 activity, stabilising it in a closed conformation. This hypothesis was supported by in vitro experiments demonstrating that Quercetin and 5-Hydroxyflavone were capable to inhibit KATP currents of rat tail main artery myocytes recorded by the patch-clamp technique. Conclusion Combined methodological approaches, such as molecular modelling, docking and MD simulations of Kir6.1 channel, used to elucidate flavonoids intrinsic mechanism of action, are introduced, revealing a new potential druggable protein site.
Collapse
Affiliation(s)
- Alfonso Trezza
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Vittoria Cicaloni
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy.,Toscana Life Sciences Foundation, Siena, Italy
| | - Piera Porciatti
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Andrea Langella
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Fabio Fusi
- Department of Life Sciences, University of Siena, Siena, Italy
| | - Simona Saponara
- Department of Life Sciences, University of Siena, Siena, Italy
| | - Ottavia Spiga
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| |
Collapse
|