1
|
Zhang S, Ma J, Wang X, Zhao D, Zhang J, Jiang L, Duan W, Wang X, Hong Z, Li Z, Liu J. GPR30 Alleviates Pressure Overload-Induced Myocardial Hypertrophy in Ovariectomized Mice by Regulating Autophagy. Int J Mol Sci 2023; 24:ijms24020904. [PMID: 36674423 PMCID: PMC9867279 DOI: 10.3390/ijms24020904] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/26/2022] [Accepted: 12/28/2022] [Indexed: 01/06/2023] Open
Abstract
The incidence of heart failure mainly resulting from cardiac hypertrophy and fibrosis increases sharply in post-menopausal women compared with men at the same age, which indicates a cardioprotective role of estrogen. Previous studies in our group have shown that the novel estrogen receptor G Protein Coupled Receptor 30 (GPR30) could attenuate myocardial fibrosis caused by ischemic heart disease. However, the role of GPR30 in myocardial hypertrophy in ovariectomized mice has not been investigated yet. In this study, female mice with bilateral ovariectomy or sham surgery underwent transverse aortic constriction (TAC) surgery. After 8 weeks, mice in the OVX + TAC group exhibited more severe myocardial hypertrophy and fibrosis than mice in the TAC group. G1, the specific agonist of GPR30, could attenuate myocardial hypertrophy and fibrosis of mice in the OVX + TAC group. Furthermore, the expression of LC3II was significantly higher in the OVX + TAC group than in the OVX + TAC + G1 group, which indicates that autophagy might play an important role in this process. An in vitro study showed that G1 alleviated AngiotensionII (AngII)-induced hypertrophy and reduced the autophagy level of H9c2 cells, as revealed by LC3II expression and tandem mRFP-GFP-LC3 fluorescence analysis. Additionally, Western blot results showed that the AKT/mTOR pathway was inhibited in the AngII group, whereas it was restored in the AngII + G1 group. To further verify the mechanism, PI3K inhibitor LY294002 or autophagy activator rapamycin was added in the AngII + G1 group, and the antihypertrophy effect of G1 on H9c2 cells was blocked by LY294002 or rapamycin. In summary, our results demonstrate that G1 can attenuate cardiac hypertrophy and fibrosis and improve the cardiac function of mice in the OVX + TAC group through AKT/mTOR mediated inhibition of autophagy. Thus, this study demonstrates a potential option for the drug treatment of pressure overload-induced cardiac hypertrophy in postmenopausal women.
Collapse
|
2
|
Borges JI, Ferraino KE, Cora N, Nagliya D, Suster MS, Carbone AM, Lymperopoulos A. Adrenal G Protein-Coupled Receptors and the Failing Heart: A Long-distance, Yet Intimate Affair. J Cardiovasc Pharmacol 2022; 80:386-392. [PMID: 34983911 PMCID: PMC9294064 DOI: 10.1097/fjc.0000000000001213] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 12/11/2021] [Indexed: 01/31/2023]
Abstract
Systolic heart failure (HF) is a chronic clinical syndrome characterized by the reduction in cardiac function and still remains the disease with the highest mortality worldwide. Despite considerable advances in pharmacological treatment, HF represents a severe clinical and social burden. Chronic human HF is characterized by several important neurohormonal perturbations, emanating from both the autonomic nervous system and the adrenal glands. Circulating catecholamines (norepinephrine and epinephrine) and aldosterone elevations are among the salient alterations that confer significant hormonal burden on the already compromised function of the failing heart. This is why sympatholytic treatments (such as β-blockers) and renin-angiotensin system inhibitors or mineralocorticoid receptor antagonists, which block the effects of angiotensin II (AngII) and aldosterone on the failing heart, are part of the mainstay HF pharmacotherapy presently. The adrenal gland plays an important role in the modulation of cardiac neurohormonal stress because it is the source of almost all aldosterone, of all epinephrine, and of a significant amount of norepinephrine reaching the failing myocardium from the blood circulation. Synthesis and release of these hormones in the adrenals is tightly regulated by adrenal G protein-coupled receptors (GPCRs), such as adrenergic receptors and AngII receptors. In this review, we discuss important aspects of adrenal GPCR signaling and regulation, as they pertain to modulation of cardiac function in the context of chronic HF, by focusing on the 2 best studied adrenal GPCR types in that context, adrenergic receptors and AngII receptors (AT 1 Rs). Particular emphasis is given to findings from the past decade and a half that highlight the emerging roles of the GPCR-kinases and the β-arrestins in the adrenals, 2 protein families that regulate the signaling and functioning of GPCRs in all tissues, including the myocardium and the adrenal gland.
Collapse
Affiliation(s)
- Jordana I. Borges
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University, Fort Lauderdale, FL 33328-2018, USA
| | - Krysten E. Ferraino
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University, Fort Lauderdale, FL 33328-2018, USA
| | - Natalie Cora
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University, Fort Lauderdale, FL 33328-2018, USA
| | - Deepika Nagliya
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University, Fort Lauderdale, FL 33328-2018, USA
| | - Malka S. Suster
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University, Fort Lauderdale, FL 33328-2018, USA
| | - Alexandra M. Carbone
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University, Fort Lauderdale, FL 33328-2018, USA
| | - Anastasios Lymperopoulos
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University, Fort Lauderdale, FL 33328-2018, USA
| |
Collapse
|
3
|
Mohamed BA, Elkenani M, Mobarak S, Marques Rodrigues D, Annamalai K, Schnelle M, Bader M, Hasenfuss G, Toischer K. Hemodynamic stress-induced cardiac remodelling is not modulated by ablation of phosphodiesterase 4D interacting protein. J Cell Mol Med 2022; 26:4440-4452. [PMID: 35860864 PMCID: PMC9357604 DOI: 10.1111/jcmm.17468] [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: 02/22/2022] [Revised: 06/13/2022] [Accepted: 06/19/2022] [Indexed: 11/28/2022] Open
Abstract
Adrenergic stimulation in the heart activates the protein kinase A (PKA), which phosphorylates key proteins involved in intracellular Ca2+ handling. PKA is held in proximity to its substrates by protein scaffolds, the A kinase anchoring proteins (AKAPs). We have previously identified the transcript of phosphodiesterase 4D interacting protein (Pde4dip; also known as myomegalin), one of the sarcomeric AKAPs, as being differentially expressed following hemodynamic overload, a condition inducing hyperadrenergic state in the heart. Here, we addressed whether PDE4DIP is involved in the adverse cardiac remodelling following hemodynamic stress. Homozygous Pde4dip knockout (KO) mice, generated by CRISPR-Cas9 technology, and wild-type (WT) littermates were exposed to aortocaval shunt (shunt) or transthoracic aortic constriction (TAC) to induce hemodynamic volume overload (VO) or pressure overload (PO), respectively. The mortality, cardiac structure, function and pathological cardiac remodelling were followed up after hemodynamic injuries. The PDE4DIP protein level was markedly downregulated in volume-overloaded- but upregulated in pressure-overloaded-WT hearts. Following shunt or TAC, mortality rates were comparably increased in both genotypes. Twelve weeks after shunt or TAC, Pde4dip-KO animals showed a similar degree of cardiac hypertrophy, dilatation and dysfunction as WT mice. Cardiomyocyte hypertrophy, myocardial fibrosis, reactivation of cardiac stress genes and downregulation of ATPase, Ca2+ transporting, cardiac muscle, slow twitch 2 transcript did not differ between WT and Pde4dip-KO hearts following shunt or TAC. In summary, despite a differential expression of PDE4DIP protein in remodelled WT hearts, Pde4dip deficiency does not modulate adverse cardiac remodelling after hemodynamic VO or PO.
Collapse
Affiliation(s)
- Belal A Mohamed
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany.,DZHK (German Centre for Cardiovascular Research), Göttingen, Germany
| | - Manar Elkenani
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany.,DZHK (German Centre for Cardiovascular Research), Göttingen, Germany
| | - Sherok Mobarak
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Daniel Marques Rodrigues
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany.,DZHK (German Centre for Cardiovascular Research), Göttingen, Germany
| | - Karthika Annamalai
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany.,DZHK (German Centre for Cardiovascular Research), Göttingen, Germany
| | - Moritz Schnelle
- DZHK (German Centre for Cardiovascular Research), Göttingen, Germany.,Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Michael Bader
- Max-Delbrück-Center for Molecular Medicine (MDC), Berlin-Buch, Germany.,DZHK (German Centre for Cardiovascular Research), Berlin, Germany.,Charité Universitätsmedizin, Berlin, Germany
| | - Gerd Hasenfuss
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany.,DZHK (German Centre for Cardiovascular Research), Göttingen, Germany
| | - Karl Toischer
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany.,DZHK (German Centre for Cardiovascular Research), Göttingen, Germany
| |
Collapse
|
4
|
Guitart-Mampel M, Urquiza P, Borges JI, Lymperopoulos A, Solesio ME. Impact of Aldosterone on the Failing Myocardium: Insights from Mitochondria and Adrenergic Receptors Signaling and Function. Cells 2021; 10:1552. [PMID: 34205363 PMCID: PMC8235589 DOI: 10.3390/cells10061552] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/08/2021] [Accepted: 06/16/2021] [Indexed: 02/06/2023] Open
Abstract
The mineralocorticoid aldosterone regulates electrolyte and blood volume homeostasis, but it also adversely modulates the structure and function of the chronically failing heart, through its elevated production in chronic human post-myocardial infarction (MI) heart failure (HF). By activating the mineralocorticoid receptor (MR), a ligand-regulated transcription factor, aldosterone promotes inflammation and fibrosis of the heart, while increasing oxidative stress, ultimately induding mitochondrial dysfunction in the failing myocardium. To reduce morbidity and mortality in advanced stage HF, MR antagonist drugs, such as spironolactone and eplerenone, are used. In addition to the MR, aldosterone can bind and stimulate other receptors, such as the plasma membrane-residing G protein-coupled estrogen receptor (GPER), further complicating it signaling properties in the myocardium. Given the salient role that adrenergic receptor (ARs)-particularly βARs-play in cardiac physiology and pathology, unsurprisingly, that part of the impact of aldosterone on the failing heart is mediated by its effects on the signaling and function of these receptors. Aldosterone can significantly precipitate the well-documented derangement of cardiac AR signaling and impairment of AR function, critically underlying chronic human HF. One of the main consequences of HF in mammalian models at the cellular level is the presence of mitochondrial dysfunction. As such, preventing mitochondrial dysfunction could be a valid pharmacological target in this condition. This review summarizes the current experimental evidence for this aldosterone/AR crosstalk in both the healthy and failing heart, and the impact of mitochondrial dysfunction in HF. Recent findings from signaling studies focusing on MR and AR crosstalk via non-conventional signaling of molecules that normally terminate the signaling of ARs in the heart, i.e., the G protein-coupled receptor-kinases (GRKs), are also highlighted.
Collapse
Affiliation(s)
- Mariona Guitart-Mampel
- Department of Biology, College of Arts and Sciences, Rutgers University, Camden, NJ 08103, USA; (M.G.-M.); (P.U.)
| | - Pedro Urquiza
- Department of Biology, College of Arts and Sciences, Rutgers University, Camden, NJ 08103, USA; (M.G.-M.); (P.U.)
| | - Jordana I. Borges
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA;
| | - Anastasios Lymperopoulos
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA;
| | - Maria E. Solesio
- Department of Biology, College of Arts and Sciences, Rutgers University, Camden, NJ 08103, USA; (M.G.-M.); (P.U.)
| |
Collapse
|
5
|
Lymperopoulos A, Cora N, Maning J, Brill AR, Sizova A. Signaling and function of cardiac autonomic nervous system receptors: Insights from the GPCR signalling universe. FEBS J 2021; 288:2645-2659. [PMID: 33599081 DOI: 10.1111/febs.15771] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 02/02/2021] [Accepted: 02/16/2021] [Indexed: 12/16/2022]
Abstract
The two branches of the autonomic nervous system (ANS), adrenergic and cholinergic, exert a multitude of effects on the human myocardium thanks to the activation of distinct G protein-coupled receptors (GPCRs) expressed on the plasma membranes of cardiac myocytes, cardiac fibroblasts, and coronary vascular endothelial cells. Norepinephrine (NE)/epinephrine (Epi) and acetylcholine (ACh) are released from cardiac ANS terminals and mediate the biological actions of the ANS on the heart via stimulation of cardiac adrenergic or muscarinic receptors, respectively. In addition, several other neurotransmitters/hormones act as facilitators of ANS neurotransmission in the heart, taking part in the so-called nonadrenergic noncholinergic (NANC) part of the ANS's control of cardiac function. These NANC mediators also use several different cell membrane-residing GPCRs to exert their effects in the myocardium. Cardiac ANS dysfunction and an imbalance between the activities of its two branches underlie a variety of cardiovascular diseases, from heart failure and hypertension to coronary artery disease, myocardial ischemia, and arrhythmias. In this review, we present the main well-established signaling modalities used by cardiac autonomic GPCRs, including receptors for salient NANC mediators, and we also highlight the latest developments pertaining to cardiac cell type-specific signal transduction, resulting in cell type-specific cardiac effects of each of these autonomic receptors.
Collapse
Affiliation(s)
- Anastasios Lymperopoulos
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Natalie Cora
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Jennifer Maning
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Ava R Brill
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Anastasiya Sizova
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University, Fort Lauderdale, FL, USA
| |
Collapse
|
6
|
Takano APC, Senger N, Barreto-Chaves MLM. The endocrinological component and signaling pathways associated to cardiac hypertrophy. Mol Cell Endocrinol 2020; 518:110972. [PMID: 32777452 DOI: 10.1016/j.mce.2020.110972] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 07/14/2020] [Accepted: 07/30/2020] [Indexed: 02/06/2023]
Abstract
Although myocardial growth corresponds to an adaptive response to maintain cardiac contractile function, the cardiac hypertrophy is a condition that occurs in many cardiovascular diseases and typically precedes the onset of heart failure. Different endocrine factors such as thyroid hormones, insulin, insulin-like growth factor 1 (IGF-1), angiotensin II (Ang II), endothelin (ET-1), catecholamines, estrogen, among others represent important stimuli to cardiomyocyte hypertrophy. Thus, numerous endocrine disorders manifested as changes in the local environment or multiple organ systems are especially important in the context of progression from cardiac hypertrophy to heart failure. Based on that information, this review summarizes experimental findings regarding the influence of such hormones upon signalling pathways associated with cardiac hypertrophy. Understanding mechanisms through which hormones differentially regulate cardiac hypertrophy could open ways to obtain therapeutic approaches that contribute to prevent or delay the onset of heart failure related to endocrine diseases.
Collapse
Affiliation(s)
| | - Nathalia Senger
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, Brazil
| | | |
Collapse
|
7
|
Sapouckey SA, Morselli LL, Deng G, Patil CN, Balapattabi K, Oliveira V, Claflin KE, Gomez J, Pearson NA, Potthoff MJ, Gibson-Corley KN, Sigmund CD, Grobe JL. Exploration of cardiometabolic and developmental significance of angiotensinogen expression by cells expressing the leptin receptor or agouti-related peptide. Am J Physiol Regul Integr Comp Physiol 2020; 318:R855-R869. [PMID: 32186897 DOI: 10.1152/ajpregu.00297.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Angiotensin II (ANG II) Agtr1a receptor (AT1A) is expressed in cells of the arcuate nucleus of the hypothalamus that express the leptin receptor (Lepr) and agouti-related peptide (Agrp). Agtr1a expression in these cells is required to stimulate resting energy expenditure in response to leptin and high-fat diets (HFDs), but the mechanism activating AT1A signaling by leptin remains unclear. To probe the role of local paracrine/autocrine ANG II generation and signaling in this mechanism, we bred mice harboring a conditional allele for angiotensinogen (Agt, encoding AGT) with mice expressing Cre-recombinase via the Lepr or Agrp promoters to cause cell-specific deletions of Agt (AgtLepr-KO and AgtAgrp-KO mice, respectively). AgtLepr-KO mice were phenotypically normal, arguing against a paracrine/autocrine AGT signaling mechanism for metabolic control. In contrast, AgtAgrp-KO mice exhibited reduced preweaning survival, and surviving adults exhibited altered renal structure and steroid flux, paralleling previous reports of animals with whole body Agt deficiency or Agt disruption in albumin (Alb)-expressing cells (thought to cause liver-specific disruption). Surprisingly, adult AgtAgrp-KO mice exhibited normal circulating AGT protein and hepatic Agt mRNA expression but reduced Agt mRNA expression in adrenal glands. Reanalysis of RNA-sequencing data sets describing transcriptomes of normal adrenal glands suggests that Agrp and Alb are both expressed in this tissue, and fluorescent reporter gene expression confirms Cre activity in adrenal gland of both Agrp-Cre and Alb-Cre mice. These findings lead to the iconoclastic conclusion that extrahepatic (i.e., adrenal) expression of Agt is critically required for normal renal development and survival.
Collapse
Affiliation(s)
- Sarah A Sapouckey
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, Iowa
| | - Lisa L Morselli
- Division of Endocrinology, Department of Internal Medicine, University of Iowa, Iowa City, Iowa
| | - Guorui Deng
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, Iowa
| | - Chetan N Patil
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | | | - Vanessa Oliveira
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Kristin E Claflin
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, Iowa
| | - Javier Gomez
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Nicole A Pearson
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, Iowa
| | - Matthew J Potthoff
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, Iowa.,Obesity Research & Education Initiative, University of Iowa, Iowa City, Iowa.,Fraternal Order of Eagles' Diabetes Research Center, University of Iowa, Iowa City, Iowa
| | - Katherine N Gibson-Corley
- Fraternal Order of Eagles' Diabetes Research Center, University of Iowa, Iowa City, Iowa.,Department of Pathology, University of Iowa, Iowa City, Iowa
| | - Curt D Sigmund
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin.,Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Justin L Grobe
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin.,Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, Wisconsin.,Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
8
|
de Montgolfier O, Pinçon A, Pouliot P, Gillis MA, Bishop J, Sled JG, Villeneuve L, Ferland G, Lévy BI, Lesage F, Thorin-Trescases N, Thorin É. High Systolic Blood Pressure Induces Cerebral Microvascular Endothelial Dysfunction, Neurovascular Unit Damage, and Cognitive Decline in Mice. Hypertension 2019; 73:217-228. [PMID: 30571552 DOI: 10.1161/hypertensionaha.118.12048] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A chronic and gradual increase in pulse pressure (PP) is associated with cognitive decline and dementia in older individuals, but the mechanisms remain ill-defined. We hypothesized that a chronic elevation of PP would cause brain microvascular endothelial mechanical stress, damage the neurovascular unit, and ultimately induce cognitive impairment in mice, potentially contributing to the progression of vascular dementia and Alzheimer disease. To test our hypothesis, male control wild-type mice and Alzheimer disease model APP/PS1 (amyloid precursor protein/presenilin 1) mice were exposed to a transverse aortic constriction for 6 weeks, creating a PP overload in the right carotid (ipsilateral). We show that the transverse aortic constriction procedure associated with high PP induces a cascade of vascular damages in the ipsilateral parenchymal microcirculation: in wild-type mice, it impairs endothelial dilatory and blood brain barrier functions and causes microbleeds, a reduction in microvascular density, microvascular cell death by apoptosis, leading to severe hypoperfusion and parenchymal cell senescence. These damages were associated with brain inflammation and a significant reduction in learning and spatial memories. In APP/PS1 mice, that endogenously display severe cerebral vascular dysfunctions, microbleeds, parenchymal inflammation and cognitive dysfunction, transverse aortic constriction-induced high PP further aggravates cerebrovascular damage, Aβ (beta-amyloid) accumulation, and prevents learning. Our study, therefore, demonstrates that brain microvessels are vulnerable to a high PP and mechanical stress associated with transverse aortic constriction, promoting severe vascular dysfunction, disruption of the neurovascular unit, and cognitive decline. Hence, chronic elevated amplitude of the PP could contribute to the development and progression of vascular dementia including Alzheimer disease.
Collapse
Affiliation(s)
- Olivia de Montgolfier
- From the Department of Pharmacology and Physiology (O.d.M., A.P.), Université de Montréal, Quebec, Canada.,Montreal Heart Institute, Research Center, Quebec, Canada (O.d.M., A.P., M.-A.G., L.V., G.F., F.L., N.T.-T., E.T.)
| | - Anthony Pinçon
- From the Department of Pharmacology and Physiology (O.d.M., A.P.), Université de Montréal, Quebec, Canada.,Montreal Heart Institute, Research Center, Quebec, Canada (O.d.M., A.P., M.-A.G., L.V., G.F., F.L., N.T.-T., E.T.)
| | | | - Marc-Antoine Gillis
- Montreal Heart Institute, Research Center, Quebec, Canada (O.d.M., A.P., M.-A.G., L.V., G.F., F.L., N.T.-T., E.T.)
| | - Jonathan Bishop
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada (J.B., J.G.S.)
| | - John G Sled
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada (J.B., J.G.S.).,Department of Medical Biophysics, University of Toronto, Ontario, Canada (J.G.S.)
| | - Louis Villeneuve
- Montreal Heart Institute, Research Center, Quebec, Canada (O.d.M., A.P., M.-A.G., L.V., G.F., F.L., N.T.-T., E.T.)
| | - Guylaine Ferland
- Department of Nutrition (G.F.), Université de Montréal, Quebec, Canada.,Montreal Heart Institute, Research Center, Quebec, Canada (O.d.M., A.P., M.-A.G., L.V., G.F., F.L., N.T.-T., E.T.)
| | - Bernard I Lévy
- Institut des Vaisseaux et du Sang, Hôpital Lariboisière, Paris, France (B.I.L.)
| | - Frédéric Lesage
- Montreal Heart Institute, Research Center, Quebec, Canada (O.d.M., A.P., M.-A.G., L.V., G.F., F.L., N.T.-T., E.T.).,Ecole Polytechnique de Montréal, Quebec, Canada (P.P., F.L.)
| | - Nathalie Thorin-Trescases
- Montreal Heart Institute, Research Center, Quebec, Canada (O.d.M., A.P., M.-A.G., L.V., G.F., F.L., N.T.-T., E.T.)
| | - Éric Thorin
- Department of Surgery (E.T.), Université de Montréal, Quebec, Canada.,Montreal Heart Institute, Research Center, Quebec, Canada (O.d.M., A.P., M.-A.G., L.V., G.F., F.L., N.T.-T., E.T.)
| |
Collapse
|
9
|
Arribas-Blázquez M, Olivos-Oré LA, Barahona MV, Sánchez de la Muela M, Solar V, Jiménez E, Gualix J, McIntosh JM, Ferrer-Montiel A, Miras-Portugal MT, Artalejo AR. Overexpression of P2X3 and P2X7 Receptors and TRPV1 Channels in Adrenomedullary Chromaffin Cells in a Rat Model of Neuropathic Pain. Int J Mol Sci 2019; 20:ijms20010155. [PMID: 30609840 PMCID: PMC6337219 DOI: 10.3390/ijms20010155] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 12/25/2018] [Accepted: 12/26/2018] [Indexed: 11/16/2022] Open
Abstract
We have tested the hypothesis that neuropathic pain acting as a stressor drives functional plasticity in the sympathoadrenal system. The relation between neuropathic pain and adrenal medulla function was studied with behavioral, immunohistochemical and electrophysiological techniques in rats subjected to chronic constriction injury of the sciatic nerve. In slices of the adrenal gland from neuropathic animals, we have evidenced increased cholinergic innervation and spontaneous synaptic activity at the splanchnic nerve–chromaffin cell junction. Likewise, adrenomedullary chromaffin cells displayed enlarged acetylcholine-evoked currents with greater sensitivity to α-conotoxin RgIA, a selective blocker of α9 subunit-containing nicotinic acetylcholine receptors, as well as increased exocytosis triggered by voltage-activated Ca2+ entry. Altogether, these adaptations are expected to facilitate catecholamine output into the bloodstream. Last, but most intriguing, functional and immunohistochemical data indicate that P2X3 and P2X7 purinergic receptors and transient receptor potential vanilloid-1 (TRPV1) channels are overexpressed in chromaffin cells from neuropathic animals. These latter observations are reminiscent of molecular changes characteristic of peripheral sensitization of nociceptors following the lesion of a peripheral nerve, and suggest that similar phenomena can occur in other tissues, potentially contributing to behavioral manifestations of neuropathic pain.
Collapse
Affiliation(s)
- Marina Arribas-Blázquez
- Department of Pharmacology and Toxicology, Veterinary Faculty, Universidad Complutense de Madrid, 28040 Madrid, Spain.
- Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, 28040 Madrid, Spain.
| | - Luis Alcides Olivos-Oré
- Department of Pharmacology and Toxicology, Veterinary Faculty, Universidad Complutense de Madrid, 28040 Madrid, Spain.
- Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, 28040 Madrid, Spain.
| | - María Victoria Barahona
- Department of Pharmacology and Toxicology, Veterinary Faculty, Universidad Complutense de Madrid, 28040 Madrid, Spain.
- Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, 28040 Madrid, Spain.
| | - Mercedes Sánchez de la Muela
- Department of Animal Medicine and Surgery, Veterinary Faculty, Universidad Complutense de Madrid, 28040 Madrid, Spain.
| | - Virginia Solar
- Department of Pharmacology and Toxicology, Veterinary Faculty, Universidad Complutense de Madrid, 28040 Madrid, Spain.
| | - Esperanza Jiménez
- Department of Pharmacology and Toxicology, Veterinary Faculty, Universidad Complutense de Madrid, 28040 Madrid, Spain.
| | - Javier Gualix
- Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, 28040 Madrid, Spain.
- Department of Biochemistry and Molecular Biology, Veterinary Faculty, Universidad Complutense de Madrid, 28040 Madrid, Spain.
| | - J Michael McIntosh
- George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, UT 84148, USA.
- Departments of Biology and Psychiatry, University of Utah, Salt Lake City, UT 84112, USA.
| | - Antonio Ferrer-Montiel
- Instituto de Biología Molecular y Celular (IBMC), Universitas Miguel Hernández, 03202 Elche, Spain.
| | - María Teresa Miras-Portugal
- Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, 28040 Madrid, Spain.
- Department of Biochemistry and Molecular Biology, Veterinary Faculty, Universidad Complutense de Madrid, 28040 Madrid, Spain.
| | - Antonio R Artalejo
- Department of Pharmacology and Toxicology, Veterinary Faculty, Universidad Complutense de Madrid, 28040 Madrid, Spain.
- Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, 28040 Madrid, Spain.
| |
Collapse
|
10
|
Pfleger J, Gross P, Johnson J, Carter RL, Gao E, Tilley DG, Houser SR, Koch WJ. G protein-coupled receptor kinase 2 contributes to impaired fatty acid metabolism in the failing heart. J Mol Cell Cardiol 2018; 123:108-117. [PMID: 30171848 DOI: 10.1016/j.yjmcc.2018.08.025] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 08/17/2018] [Accepted: 08/28/2018] [Indexed: 12/19/2022]
Abstract
Increased G protein-coupled receptor kinase (GRK)2 is central to heart failure (HF) pathogenesis, via desensitization of β-adrenergic receptors and loss of contractile reserve. Since GRK2 has been shown to compromise fatty acid (FA) oxidation, this kinase may link metabolic and contractile defects in HF. The aim of this study was to investigate the mechanistic role of GRK2 in FA metabolism and bioenergetics in the heart. For that purpose, we measured FA uptake and cluster of differentiation (CD)36 expression, phosphorylation, and ubiquitination in mice with cardiac-specific overexpression of GRK2 (TgGRK2) or expression of its c-terminus (GRK2 inhibitor- TgβARKct) or in global heterozygous GRK2 knockout (GRK2+/-) mice. Cellular bioenergetics were also measured in isolated cardiomyocytes following adenoviral delivery of exogenous GRK2, βARKct, or short hairpin GRK2 (shGRK2). Additionally, CD36 expression and phosphorylation were evaluated following transverse aortic constriction (TAC) in wild type (WT) and GRK2+/- mice. Our results show a 33% ± 0.81 reduction in FA uptake rate, accompanied by 51% ± 0.17 lower CD36 protein, and 70% ± 0.23 and 69% ± 0.18 increases in CD36 phosphorylation and ubiquitination, respectively, in the TgGRK2 mice. Moreover, an in vitro kinase assay suggests that GRK2 directly phosphorylates CD36. In isolated cardiomyocytes, GRK2 overexpression induced a 26% ± 2.21 decrease in maximal respiration, which was enhanced (20% ± 4.02-5.14) with inhibition of the kinase. Importantly, in hearts with systolic dysfunction, notable reductions in CD36 mRNA and protein, as well as a significant increase in CD36 phosphorylation were normalized in the GRK2+/- mice post-TAC. Thus, we propose that GRK2 up-regulation in HF is, at least partly, responsible for reduced FA uptake and oxidation and may be a nodal link between metabolic and contractile defects.
Collapse
Affiliation(s)
- Jessica Pfleger
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Polina Gross
- Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Jaslyn Johnson
- Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Rhonda L Carter
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Erhe Gao
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Douglas G Tilley
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Steven R Houser
- Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Walter J Koch
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA.
| |
Collapse
|
11
|
Allison BJ, Nguyen V, Yiallourou SR, Nitsos I, Black MJ, Polglase GR. The effect of sex and prematurity on the cardiovascular baroreflex response in sheep. Exp Physiol 2018; 103:9-18. [PMID: 29072344 DOI: 10.1113/ep086494] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 10/13/2017] [Indexed: 11/08/2022]
Abstract
NEW FINDINGS What is the central question of this study? Late preterm infants are often assumed to escape long-term morbidities known to impact earlier preterm offspring. Is this true for the cardiovascular system? What is the main finding and its importance? We show that late preterm birth is a risk factor for cardiovascular dysfunction in early adulthood and is influenced by sex. Early signs of cardiovascular dysfunction might predispose to heart disease in adulthood. Very preterm infants have an increased risk of cardiovascular disease; however, the effects of a late preterm birth on future cardiovascular function are not known. We hypothesized that after a late preterm birth, the well-described impairments in heart rate variability and baroreflex sensitivity would persist into adulthood. To test this hypothesis, sheep born preterm (0.9 gestation; nine male and seven female) or term (11 male and six female) underwent surgery at 14 months of age for insertion of femoral arterial and venous catheters and a femoral flow probe. After recovery, heart rate variability was assessed, followed by a baroreflex challenge (using the vasoactive agents phenylephrine and sodium nitroprusside) in conscious adult lambs. Our data demonstrate decreased low-frequency normalised units (LFnu) and low-frequency/high-frequency ratio in female but not male ex-preterm sheep at rest. When challenged, mature male ex-preterm sheep have an increased blood pressure response but dampened heart rate baroreflex response. We show that even a late preterm birth leads to cardiovascular dysfunction in adulthood. These early signs of cardiovascular dysfunction might underpin the later hypertension and increased risk of heart disease observed in adults born preterm. These findings are particularly important because late preterm infants are often assumed to escape the long-term morbidities known to impact on very preterm and extremely preterm offspring.
Collapse
Affiliation(s)
- Beth J Allison
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Department of Obstetrics and Gynecology, Monash University, Clayton, Victoria, Australia
| | - Vivian Nguyen
- Department of Anatomy and Developmental Biology and Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Stephanie R Yiallourou
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Department of Obstetrics and Gynecology, Monash University, Clayton, Victoria, Australia
| | - Ilias Nitsos
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Mary Jane Black
- Department of Anatomy and Developmental Biology and Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Graeme R Polglase
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Department of Obstetrics and Gynecology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
12
|
Mokhtar DM, Hussein MT, Hassan AHS. Melatonin Elicits Stimulatory Action on the Adrenal Gland of Soay Ram: Morphometrical, Immunohistochemical, and Ultrastructural Study. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2017; 23:1173-1188. [PMID: 29199635 DOI: 10.1017/s1431927617012727] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Endogenous melatonin is a hormone secreted by pineal gland; it has several roles in metabolism, reproduction, and remarkable antioxidant properties. Studies on the melatonin effect on the adrenal glands which are important endocrine organs, controlling essential physiological functions, are still deficient. In this study, we attempted to investigate the effect of exogenous melatonin treatment on the adrenal cortex and medulla using several approaches. Adrenal glands of 15 Soay ram were examined to detect the effect of melatonin treatment. Our results revealed that the cells of adrenal cortex of the treated animals were separated by wide and numerous blood sinusoids and showed signs of increase steroidogenic activity, which are evidenced by functional hypertrophy with increase profiles of mitochondria, smooth endoplasmic reticulum, and lipid droplets. The most striking ultrastructural features in the medulla of the treated group were the engorgement of chromaffin cells with enlarged secretory granules enclosed within a significantly increased diameter of these cells. The cytoplasm of these cells showed numerous mitochondria, rough endoplasmic reticulum (rER), Golgi apparatus, lysosomes, and glycogen granules. Exocytosis of secretory granules to the lumen of blood vessels was evident in the treated group. Piecemeal degranulation mode of secretion was recorded after melatonin treatment. Chromaffin cells in the control group expressed moderate immunoreactivity to Synaptophysin and tyrosine hydroxylase, compared with intensified expression after melatonin treatment. The ganglion cells of the melatonin-treated group showed a significant increase in diameter with numerous rER. The most interesting feature in this study is the presence of small granule chromaffin cells (SGC) and telocytes (TCs) for the first time in the adrenal glands of sheep. Moreover, these SGC cells, Schwann cells, fibroblasts, and progenitor stem cells showed a stimulatory response. The TCs were small branched cells scattered in the adrenal glands around cortical cells, chromaffin cells, nerve fibers, and blood vessels. These cells increased significantly in number, length of their telopodes, and secretory activity after melatonin treatment. In addition, multiple profiles of unmyelinated nerve fibers were demonstrated in all treated specimens. These results indicated that melatonin treatment caused a stimulatory action on all cellular and neuronal elements of the adrenal gland. This study may act as a new direction for treatment of adrenal insufficiency.
Collapse
Affiliation(s)
- Doaa M Mokhtar
- Department of Anatomy and Histology, Faculty of Veterinary Medicine,Assiut University,Assiut,Egypt
| | - Manal T Hussein
- Department of Anatomy and Histology, Faculty of Veterinary Medicine,Assiut University,Assiut,Egypt
| | - Ahmed H S Hassan
- Department of Anatomy and Histology, Faculty of Veterinary Medicine,Assiut University,Assiut,Egypt
| |
Collapse
|
13
|
Orlowska-Baranowska E, Gadomska vel Betka L, Gora J, Baranowski R, Pedzich-Placha E, Zakrzewski D, Dlugosz A, Kossowska H, Zebrowska A, Zakoscielna E, Janiszewska A, Hryniewiecki T, Gaciong Z, Placha G. Functional polymorphism of the renalase gene is associated with cardiac hypertrophy in female patients with aortic stenosis. PLoS One 2017; 12:e0186729. [PMID: 29065134 PMCID: PMC5655536 DOI: 10.1371/journal.pone.0186729] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Accepted: 10/08/2017] [Indexed: 12/18/2022] Open
Abstract
Renalase decreases circulating catecholamines concentration and is important in maintaining primary cellular metabolism. Renalase acts through the plasma membrane calcium ATPase 4b in the heart, which affects pressure overload but not exercise induced heart hypertrophy. The aim of this study was to test the association between a functional polymorphism Glu37Asp (rs2296545) of the renalase gene and left ventricular hypertrophy in a large cohort of patients with aortic stenosis. The study group consisted of 657 patients with aortic stenosis referred for aortic valve replacement. Preoperative echocardiographic assessment was performed to obtain cardiac phenotypes. Generalized-linear models were implemented to analyze data using crude or full model adjusted for selected clinical factors. In females, the Asp37 variant of the Glu37Asp polymorphism was associated with higher left ventricular mass (p = 0.0021 and p = 0.055 crude and full model respectively), intraventricular septal thickness (p = 0.0003 and p = 0.0143) and posterior wall thickness (p = 0.0005 and p = 0.0219) all indexed to body surface area, as well as relative wall thickness (p = 0.001 and p = 0.0097). No significant associations were found among the male patients. In conclusion, we have found the association of the renalase Glu37Asp polymorphism with left ventricle hypertrophy in large group of females with aortic stenosis. The Glu37Asp polymorphism causes not only amino-acid substitution in FAD binding domain but may also change binding affinity of the hypoxia- and hypertrophy-related transcription factors and influence renalase gene expression. Our data suggest that renalase might play a role in hypertrophic response to pressure overload, but the exact mechanism requires further investigation.
Collapse
Affiliation(s)
| | - Lucja Gadomska vel Betka
- Department of Internal Medicine, Hypertension, and Vascular Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Jaroslaw Gora
- Department of Internal Medicine, Hypertension, and Vascular Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Rafal Baranowski
- Department of Arrhythmia, Institute of Cardiology, Warsaw, Poland
| | - Ewa Pedzich-Placha
- Department of Internal Medicine, Hypertension, and Vascular Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Dariusz Zakrzewski
- Department of Acquired Cardiac Defects, Institute of Cardiology, Warsaw, Poland
| | - Angelika Dlugosz
- Department of Internal Medicine, Hypertension, and Vascular Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Helena Kossowska
- Department of Internal Medicine, Hypertension, and Vascular Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Agnieszka Zebrowska
- Department of Acquired Cardiac Defects, Institute of Cardiology, Warsaw, Poland
| | - Ewelina Zakoscielna
- Department of Internal Medicine, Hypertension, and Vascular Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Anna Janiszewska
- Department of Internal Medicine, Hypertension, and Vascular Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Tomasz Hryniewiecki
- Department of Acquired Cardiac Defects, Institute of Cardiology, Warsaw, Poland
| | - Zbigniew Gaciong
- Department of Internal Medicine, Hypertension, and Vascular Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Grzegorz Placha
- Department of Internal Medicine, Hypertension, and Vascular Diseases, Medical University of Warsaw, Warsaw, Poland
- * E-mail:
| |
Collapse
|
14
|
Artalejo AR, Olivos-Oré LA. Alpha2-adrenoceptors in adrenomedullary chromaffin cells: functional role and pathophysiological implications. Pflugers Arch 2017; 470:61-66. [PMID: 28836008 DOI: 10.1007/s00424-017-2059-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Revised: 08/12/2017] [Accepted: 08/15/2017] [Indexed: 12/20/2022]
Abstract
Chromaffin cells from the adrenal medulla participate in stress responses by releasing catecholamines into the bloodstream. Main control of adrenal catecholamine secretion is exerted both neurally (by the splanchnic nerve fibers) and humorally (by corticosteroids, circulating noradrenaline, etc.). It should be noted, however, that secretory products themselves (catecholamines, ATP, opioids, ascorbic acid, chromogranins) could also influence the secretory response in an autocrine/paracrine manner. This form of control is activity-dependent and can be either inhibitory or excitatory. Among the inhibitory influences, it stands out the one mediated by α2-adrenergic autoreceptors activated by released catecholamines. α2-adrenoceptors are G protein-coupled receptors capable to inhibit exocytotic secretion through a direct interaction of Gβγ subunits with voltage-gated Ca2+ channels. Interestingly, upon intense and/or prolonged stimulation, α2-adrenergic receptors become desensitized by the intervention of G protein-coupled receptor kinase 2 (GRK2). In several experimental models of heart failure, there has been reported the up-regulation of GRK2 and the loss of functioning of inhibitory α2-adrenoceptors resulting in enhanced release of adrenomedullary catecholamines. Given the importance of circulating catecholamines in the pathophysiology of heart failure, the recovery of α2-adrenergic modulation of the secretory response from chromaffin cells appears as a novel strategy for a better control of the patients with this cardiac disease.
Collapse
Affiliation(s)
- Antonio R Artalejo
- Institute for Research in Neurochemistry & Department of Toxicology and Pharmacology, Faculty of Veterinary, Universidad Complutense de Madrid, Avda. Puerta de Hierro s/n, 28029, Madrid, Spain.
| | - Luis Alcides Olivos-Oré
- Institute for Research in Neurochemistry & Department of Toxicology and Pharmacology, Faculty of Veterinary, Universidad Complutense de Madrid, Avda. Puerta de Hierro s/n, 28029, Madrid, Spain
| |
Collapse
|
15
|
Xiao L, Gu Y, Gao L, Shangguan J, Chen Y, Zhang Y, Li L. Sanggenon C protects against pressure overload‑induced cardiac hypertrophy via the calcineurin/NFAT2 pathway. Mol Med Rep 2017; 16:5338-5346. [PMID: 28849031 PMCID: PMC5647066 DOI: 10.3892/mmr.2017.7288] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 05/24/2017] [Indexed: 01/06/2023] Open
Abstract
The effects of Sanggenon C on oxidative stress and inflammation have previously been reported; however, little is currently known regarding the effects of Sanggenon C on cardiac hypertrophy and fibrosis. In the present study, aortic banding (AB) was performed on mice to induce cardiac hypertrophy. After 1 week AB surgery, mice were treated daily with 10 or 20 mg/kg Sanggenon C for 3 weeks. Subsequently, cardiac function was detected using echocardiography and catheter-based measurements of hemodynamic parameters. In addition, the extent of cardiac hypertrophy was evaluated by pathological staining and molecular analysis of heart tissue in each group. After 4 weeks of AB, vehicle-treated mice exhibited cardiac hypertrophy, fibrosis, and deteriorated systolic and diastolic function, whereas treatment with 10 and 20 mg/kg Sanggenon C treatment ameliorated these alterations, as evidenced by attenuated cardiac hypertrophy and fibrosis, and preserved cardiac function. Furthermore, AB-induced activation of calcineurin and nuclear factor of activated T cells 2 (NFAT2) was reduced following Sanggenon C treatment. These results suggest that Sanggenon C may exert protective effects against cardiac hypertrophy and fibrosis via suppression of the calcineurin/NFAT2 pathway.
Collapse
Affiliation(s)
- Lili Xiao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yulei Gu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Lu Gao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Jiahong Shangguan
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yang Chen
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yanzhou Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Ling Li
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
16
|
Hühner L, Rilka J, Gilsbach R, Zhou X, Machado V, Spittau B. Interleukin-4 Protects Dopaminergic Neurons In vitro but Is Dispensable for MPTP-Induced Neurodegeneration In vivo. Front Mol Neurosci 2017; 10:62. [PMID: 28337124 PMCID: PMC5343015 DOI: 10.3389/fnmol.2017.00062] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 02/24/2017] [Indexed: 01/10/2023] Open
Abstract
Microglia are involved in physiological as well as neuropathological processes in the central nervous system (CNS). Their functional states are often referred to as M1-like and M2-like activation, and are believed to contribute to neuroinflammation-mediated neurodegeneration or neuroprotection, respectively. Parkinson’s disease (PD) is one the most common neurodegenerative disease and is characterized by the progressive loss of midbrain dopaminergic (mDA) neurons in the substantia nigra resulting in bradykinesia, tremor, and rigidity. Interleukin 4 (IL4)-mediated M2-like activation of microglia, which is characterized by upregulation of alternative markers Arginase 1 (Arg1) and Chitinase 3 like 3 (Ym1) has been well studied in vitro but the role of endogenous IL4 during CNS pathologies in vivo is not well understood. Interestingly, microglia activation by IL4 has been described to promote neuroprotective and neurorestorative effects, which might be important to slow the progression of neurodegenerative diseases. In the present study, we addressed the role of endogenous and exogenous IL4 during MPP+-induced degeneration of mDA neurons in vitro and further addressed the impact of IL4-deficiency on neurodegeneration in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD in vivo. Our results clearly demonstrate that exogenous IL4 is important to protect mDA neurons in vitro, but endogenous IL4 seems to be dispensable for development and maintenance of the nigrostriatal system as well as MPTP-induced loss of TH+ neurons in vivo. These results underline the importance of IL4 in promoting a neuroprotective microglia activation state and strengthen the therapeutic potential of exogenous IL4 for protection of mDA neurons in PD models.
Collapse
Affiliation(s)
- Laura Hühner
- Department of Molecular Embryology, Institute for Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg Freiburg, Germany
| | - Jennifer Rilka
- Department of Molecular Embryology, Institute for Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg Freiburg, Germany
| | - Ralf Gilsbach
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Freiburg Freiburg, Germany
| | - Xiaolai Zhou
- Department of Molecular Embryology, Institute for Anatomy and Cell Biology, Faculty of Medicine, University of FreiburgFreiburg, Germany; Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, IthacaNY, USA
| | - Venissa Machado
- Department of Molecular Embryology, Institute for Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg Freiburg, Germany
| | - Björn Spittau
- Department of Molecular Embryology, Institute for Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg Freiburg, Germany
| |
Collapse
|
17
|
Mahata SK, Zheng H, Mahata S, Liu X, Patel KP. Effect of heart failure on catecholamine granule morphology and storage in chromaffin cells. J Endocrinol 2016; 230:309-23. [PMID: 27402067 PMCID: PMC4980258 DOI: 10.1530/joe-16-0146] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 06/24/2016] [Indexed: 12/16/2022]
Abstract
One of the key mechanisms involved in sympathoexcitation in chronic heart failure (HF) is the activation of the adrenal glands. Impact of the elevated catecholamines on the hemodynamic parameters has been previously demonstrated. However, studies linking the structural effects of such overactivation with secretory performance and cell metabolism in the adrenomedullary chromaffin cells in vivo have not been previously reported. In this study, HF was induced in male Sprague-Dawley rats by ligation of the left coronary artery. Five weeks after surgery, cardiac function was assessed by ventricular hemodynamics. HF rats showed increased adrenal weight and adrenal catecholamine levels (norepinephrine, epinephrine and dopamine) compared with sham-operated rats. Rats with HF demonstrated increased small synaptic and dense core vesicle in splanchnic-adrenal synapses indicating trans-synaptic activation of catecholamine biosynthetic enzymes, increased endoplasmic reticulum and Golgi lumen width to meet the demand of increased catecholamine synthesis and release, and more mitochondria with dilated cristae and glycogen to accommodate for the increased energy demand for the increased biogenesis and exocytosis of catecholamines from the adrenal medulla. These findings suggest that increased trans-synaptic activation of the chromaffin cells within the adrenal medulla may lead to increased catecholamines in the circulation which in turn contributes to the enhanced neurohumoral drive, providing a unique mechanistic insight for enhanced catecholamine levels in plasma commonly observed in chronic HF condition.
Collapse
Affiliation(s)
- Sushil K Mahata
- VA San Diego Healthcare System Metabolic Physiology & Ultrastructural Biology Lab.Department of Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Hong Zheng
- Department of Cellular and Integrative PhysiologyUniversity of Nebraska Medical Center, Omaha, NE, USA
| | - Sumana Mahata
- Caltech Division of BiologyCalifornia Institute of Technology, Pasadena, CA, USA
| | - Xuefei Liu
- Department of Cellular and Integrative PhysiologyUniversity of Nebraska Medical Center, Omaha, NE, USA
| | - Kaushik P Patel
- Department of Cellular and Integrative PhysiologyUniversity of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
18
|
Lymperopoulos A, Brill A, McCrink KA. GPCRs of adrenal chromaffin cells & catecholamines: The plot thickens. Int J Biochem Cell Biol 2016; 77:213-219. [PMID: 26851510 DOI: 10.1016/j.biocel.2016.02.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2015] [Revised: 02/01/2016] [Accepted: 02/02/2016] [Indexed: 12/14/2022]
Abstract
The circulating catecholamines (CAs) epinephrine (Epi) and norepinephrine (NE) derive from two major sources in the whole organism: the sympathetic nerve endings, which release NE on effector organs, and the chromaffin cells of the adrenal medulla, which are cells that synthesize, store and release Epi (mainly) and NE. All of the Epi in the body and a significant amount of circulating NE derive from the adrenal medulla. The secretion of CAs from adrenal chromaffin cells is regulated in a complex way by a variety of membrane receptors, the vast majority of which are G protein-coupled receptors (GPCRs), including adrenergic receptors (ARs), which act as "presynaptic autoreceptors" in this regard. There is a plethora of CA-secretagogue signals acting on these receptors but some of them, most notably the α2ARs, inhibit CA secretion. Over the past few years, however, a few new proteins present in chromaffin cells have been uncovered to participate in CA secretion regulation. Most prominent among these are GRK2 and β-arrestin1, which are known to interact with GPCRs regulating receptor signaling and function. The present review will discuss the molecular and signaling mechanisms by which adrenal chromaffin cell-residing GPCRs and their regulatory proteins modulate CA synthesis and secretion. Particular emphasis will be given to the newly discovered roles of GRK2 and β-arrestins in these processes and particular points of focus for future research will be highlighted, as well.
Collapse
Affiliation(s)
- Anastasios Lymperopoulos
- From the Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University, College of Pharmacy, 3200 S. University Dr., Fort Lauderdale, FL 33328-2018, USA.
| | - Ava Brill
- From the Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University, College of Pharmacy, 3200 S. University Dr., Fort Lauderdale, FL 33328-2018, USA
| | - Katie A McCrink
- From the Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University, College of Pharmacy, 3200 S. University Dr., Fort Lauderdale, FL 33328-2018, USA
| |
Collapse
|
19
|
Machado V, Gilsbach R, Das R, Schober A, Bogatyreva L, Hauschke D, Krieglstein K, Unsicker K, Spittau B. Gdf-15 deficiency does not alter vulnerability of nigrostriatal dopaminergic system in MPTP-intoxicated mice. Cell Tissue Res 2016; 365:209-23. [PMID: 27115420 DOI: 10.1007/s00441-016-2406-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 04/06/2016] [Indexed: 12/25/2022]
Abstract
Growth/differentiation factor-15 (Gdf-15) is a member of the transforming growth factor-β (Tgf-β) superfamily and has been shown to be a potent neurotrophic factor for midbrain dopaminergic (DAergic) neurons both in vitro and in vivo. Gdf-15 has also been shown to be involved in inflammatory processes. The aim of this study was to identify the role of endogenous Gdf-15 in the MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) mouse model of Parkinson's disease (PD) by comparing Gdf-15 (+/+) and Gdf-15 (-/-) mice. At 4 days and 14 days post-MPTP administration, both Gdf-15 (+/+) and Gdf-15 (-/-) mice showed a similar decline in DAergic neuron numbers and in striatal dopamine (DA) levels. This was followed by a comparable restorative phase at 90 days and 120 days, indicating that the absence of Gdf-15 does not affect the susceptibility or the recovery capacity of the nigrostriatal system after MPTP administration. The MPTP-induced microglial and astrocytic response was not significantly altered between the two genotypes. However, pro-inflammatory and anti-inflammatory cytokine profiling revealed the differential expression of markers in Gdf-15 (+/+) and Gdf-15 (-/-) mice after MPTP administration. Thus, the MPTP mouse model fails to uncover a major role of endogenous Gdf-15 in the protection of MPTP-lesioned nigrostriatal DAergic neurons, in contrast to its capacity to protect the 6-hydroxydopamine-intoxicated nigrostriatal system.
Collapse
Affiliation(s)
- Venissa Machado
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, University of Freiburg, 79104, Freiburg, Germany.,Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104, Freiburg, Germany.,Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
| | - Ralf Gilsbach
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Freiburg, 79104, Freiburg, Germany
| | - Richa Das
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, University of Freiburg, 79104, Freiburg, Germany.,German Center for Neurodegenerative Diseases, 53115, Bonn, Germany
| | - Andreas Schober
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, University of Freiburg, 79104, Freiburg, Germany
| | - Lioudmila Bogatyreva
- Institute of Medical Biometry and Medical Informatics, University of Freiburg, 79104, Freiburg, Germany
| | - Dieter Hauschke
- German Center for Neurodegenerative Diseases, 53115, Bonn, Germany
| | - Kerstin Krieglstein
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, University of Freiburg, 79104, Freiburg, Germany
| | - Klaus Unsicker
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, University of Freiburg, 79104, Freiburg, Germany.
| | - Björn Spittau
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, University of Freiburg, 79104, Freiburg, Germany.
| |
Collapse
|
20
|
Jafferjee M, Reyes Valero T, Marrero C, McCrink KA, Brill A, Lymperopoulos A. GRK2 Up-Regulation Creates a Positive Feedback Loop for Catecholamine Production in Chromaffin Cells. Mol Endocrinol 2016; 30:372-381. [PMID: 26849467 PMCID: PMC5414648 DOI: 10.1210/me.2015-1305] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 02/03/2016] [Indexed: 12/15/2022] Open
Abstract
Elevated sympathetic nervous system (SNS) activity aggravates several diseases, including heart failure. The molecular cause(s) underlying this SNS hyperactivity are not known. We have previously uncovered a neurohormonal mechanism, operating in adrenomedullary chromaffin cells, by which circulating catecholamine (CA) levels increase in heart failure: severe dysfunction of the adrenal α2-adrenergic receptors (ARs) due to the up-regulation of G protein-coupled receptor-kinase (GRK)-2, the kinase that desensitizes them. Herein we looked at the potential signaling mechanisms that bring about this GRK2 elevation in chromaffin cells. We found that chronic CA treatment of either PC12 or rat primary chromaffin cells can in itself result in GRK2 transcriptional up-regulation through α2ARs-Gi/o proteins-Src-ERK1/2. The resultant GRK2 increase severely enhances the α2AR desensitization/down-regulation elevating not only CA release but also CA biosynthesis, as evidenced by tyrosine hydroxylase up-regulation. Finally, GRK2 knockdown leads to enhanced apoptosis of PC12 cells, indicating an essential role for GRK2 in chromaffin cell homeostasis/survival. In conclusion, chromaffin cell GRK2 mediates a positive feedback loop that feeds into CA secretion, thereby enabling the adrenomedullary component of the SNS to turn itself on.
Collapse
Affiliation(s)
- Malika Jafferjee
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Ft Lauderdale, Florida 33328-2018
| | - Thairy Reyes Valero
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Ft Lauderdale, Florida 33328-2018
| | - Christine Marrero
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Ft Lauderdale, Florida 33328-2018
| | - Katie A McCrink
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Ft Lauderdale, Florida 33328-2018
| | - Ava Brill
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Ft Lauderdale, Florida 33328-2018
| | - Anastasios Lymperopoulos
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Ft Lauderdale, Florida 33328-2018
| |
Collapse
|
21
|
Andrade EF, Lobato RV, de Araújo TV, Orlando DR, Vicente da Costa D, de Oliveira Silva V, Rogatto GP, Zangeronimo MG, Rosa PV, Pereira LJ. Adaptation to physical training in rats orally supplemented with glycerol. Can J Physiol Pharmacol 2016; 93:63-9. [PMID: 25474597 DOI: 10.1139/cjpp-2014-0312] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We evaluated training adaptation and physical performance parameters in rats orally supplemented with glycerol, glucose, or saline, and submitted to moderate aerobic exercise. Thirty male rats were trained for 6 weeks and administered the supplements during the last 4 weeks of the experiment. Animals were distributed in a completely randomized factorial 2 × 3 design (with or without exercise and 3 substrates). Data were subjected to analysis of variance (ANOVA) and means were compared using the Student-Newmann-Keuls test at 5%. Among the trained animals, none of the substances caused differences in the percentages of protein, fat, or water content in the carcass. Compared with the sedentary animals, the trained animals supplemented with saline and glucose showed a higher protein percentage in the carcass. The relative mass of the heart and adrenal glands was higher in the trained animals. Glycerol improved the protein content in non-trained animals and increased the relative adrenal mass in both groups. Glycerol reduced the variation in levels of lactate and aspartate aminotransferase (AST) during the last exercise session. There was no difference between groups regarding the relative mass of the thymus and gastrocnemius or with the diameter of muscle fibers or the neutrophil-lymphocyte ratio. Supplementation with glycerol was efficient at attenuating variation in AST and lactate levels during exercise.
Collapse
|
22
|
Karabowicz P, Schlicker E, Pędzińska-Betiuk A, Kloza M, Malinowska B. The adrenal medulla, not CB1 receptors, mediates the inhibitory effect of acute transverse aortic constriction on the neurogenic vasopressor response. Life Sci 2015; 138:86-93. [PMID: 25498898 DOI: 10.1016/j.lfs.2014.11.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 11/12/2014] [Accepted: 11/21/2014] [Indexed: 11/26/2022]
Affiliation(s)
- Piotr Karabowicz
- Department of Experimental Physiology and Pathophysiology, Medical University of Białystok, Mickiewicz str. 2A, 15-222 Białystok, Poland
| | - Eberhard Schlicker
- Department of Pharmacology and Toxicology, University of Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - Anna Pędzińska-Betiuk
- Department of Experimental Physiology and Pathophysiology, Medical University of Białystok, Mickiewicz str. 2A, 15-222 Białystok, Poland
| | - Monika Kloza
- Department of Experimental Physiology and Pathophysiology, Medical University of Białystok, Mickiewicz str. 2A, 15-222 Białystok, Poland
| | - Barbara Malinowska
- Department of Experimental Physiology and Pathophysiology, Medical University of Białystok, Mickiewicz str. 2A, 15-222 Białystok, Poland.
| |
Collapse
|
23
|
Weise M, Vettel C, Spiger K, Gilsbach R, Hein L, Lorenz K, Wieland T, Aktories K, Orth JHC. A systemic Pasteurella multocida toxin aggravates cardiac hypertrophy and fibrosis in mice. Cell Microbiol 2015; 17:1320-31. [PMID: 25759205 DOI: 10.1111/cmi.12436] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 02/20/2015] [Accepted: 03/06/2015] [Indexed: 11/30/2022]
Abstract
Pasteurella multocida toxin (PMT) persistently activates heterotrimeric G proteins of the Gαq/11 , Gα12/13 and Gαi family without interaction with G protein-coupled receptors (GPCRs). We show that PMT acts on heart tissue in vivo and on cardiomyocytes and cardiac fibroblasts in vitro by deamidation of heterotrimeric G proteins. Increased normalized ventricle weights and fibrosis were detected after intraperitoneal administration of PMT in combination with the GPCR agonist phenylephrine. In neonatal rat cardiomyocytes, PMT stimulated the mitogen-activated protein kinase pathway, which is crucial for the development of cellular hypertrophy. The toxin induced phosphorylation of the canonical phosphorylation sites of the extracellular-regulated kinase 1/2 and, additionally, caused phosphorylation of the recently recognized autophosphorylation site, which appears to be important for the development of cellular hypertrophy. Moreover, PMT stimulated the small GTPases Rac1 and RhoA. Both switch proteins are involved in cardiomyocyte hypertrophy. In addition, PMT stimulated RhoA and Rac1 in neonatal rat cardiac fibroblasts. RhoA and Rac1 have been implicated in the regulation of connective tissue growth factor (CTGF) secretion and expression. Accordingly, we show that PMT treatment increased secretion and expression of CTGF in cardiac fibroblasts. Altogether, the data indicate that PMT is an inducer of pathological remodelling of cardiac cells and identifies the toxin as a promising tool for studying heterotrimeric G protein-dependent signalling in cardiac cells.
Collapse
Affiliation(s)
- Markus Weise
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Dept. I, Albert-Ludwigs-Universität Freiburg, Albertstr. 25, Freiburg, 79104, Germany
| | - Christiane Vettel
- Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Katharina Spiger
- Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Ralf Gilsbach
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Dept. II, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Lutz Hein
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Dept. II, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Kristina Lorenz
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany.,Comprehensive Heart Failure Center, University of Würzburg, Würzburg, Germany
| | - Thomas Wieland
- Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Klaus Aktories
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Dept. I, Albert-Ludwigs-Universität Freiburg, Albertstr. 25, Freiburg, 79104, Germany.,BIOSS Centre for Biological Signalling Studies, Universität Freiburg, Freiburg, Germany
| | - Joachim H C Orth
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Dept. I, Albert-Ludwigs-Universität Freiburg, Albertstr. 25, Freiburg, 79104, Germany
| |
Collapse
|
24
|
de Lucia C, Femminella GD, Gambino G, Pagano G, Allocca E, Rengo C, Silvestri C, Leosco D, Ferrara N, Rengo G. Adrenal adrenoceptors in heart failure. Front Physiol 2014; 5:246. [PMID: 25071591 PMCID: PMC4084669 DOI: 10.3389/fphys.2014.00246] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 06/13/2014] [Indexed: 01/08/2023] Open
Abstract
Heart failure (HF) is a chronic clinical syndrome characterized by the reduction in left ventricular (LV) function and it represents one of the most important causes of morbidity and mortality worldwide. Despite considerable advances in pharmacological treatment, HF represents a severe clinical and social burden. Sympathetic outflow, characterized by increased circulating catecholamines (CA) biosynthesis and secretion, is peculiar in HF and sympatholytic treatments (as β-blockers) are presently being used for the treatment of this disease. Adrenal gland secretes Epinephrine (80%) and Norepinephrine (20%) in response to acetylcholine stimulation of nicotinic cholinergic receptors on the chromaffin cell membranes. This process is regulated by adrenergic receptors (ARs): α2ARs inhibit CA release through coupling to inhibitory Gi-proteins, and β ARs (mainly β2ARs) stimulate CA release through coupling to stimulatory Gs-proteins. All ARs are G-protein-coupled receptors (GPCRs) and GPCR kinases (GRKs) regulate their signaling and function. Adrenal GRK2-mediated α2AR desensitization and downregulation are increased in HF and seem to be a fundamental regulator of CA secretion from the adrenal gland. Consequently, restoration of adrenal α2AR signaling through the inhibition of GRK2 is a fascinating sympatholytic therapeutic strategy for chronic HF. This strategy could have several significant advantages over existing HF pharmacotherapies minimizing side-effects on extra-cardiac tissues and reducing the chronic activation of the renin–angiotensin–aldosterone and endothelin systems. The role of adrenal ARs in regulation of sympathetic hyperactivity opens interesting perspectives in understanding HF pathophysiology and in the identification of new therapeutic targets.
Collapse
Affiliation(s)
- Claudio de Lucia
- Department of Medical Translational Sciences, University of Naples Federico II Naples, Italy
| | - Grazia D Femminella
- Department of Medical Translational Sciences, University of Naples Federico II Naples, Italy
| | - Giuseppina Gambino
- Department of Medical Translational Sciences, University of Naples Federico II Naples, Italy
| | - Gennaro Pagano
- Department of Medical Translational Sciences, University of Naples Federico II Naples, Italy
| | - Elena Allocca
- Department of Medical Translational Sciences, University of Naples Federico II Naples, Italy
| | - Carlo Rengo
- Department of Medical Translational Sciences, University of Naples Federico II Naples, Italy ; Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme Telese Terme, Italy
| | - Candida Silvestri
- Department of Medical Translational Sciences, University of Naples Federico II Naples, Italy
| | - Dario Leosco
- Department of Medical Translational Sciences, University of Naples Federico II Naples, Italy
| | - Nicola Ferrara
- Department of Medical Translational Sciences, University of Naples Federico II Naples, Italy ; Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme Telese Terme, Italy
| | - Giuseppe Rengo
- Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme Telese Terme, Italy
| |
Collapse
|
25
|
Kamal FA, Mickelsen DM, Wegman KM, Travers JG, Moalem J, Hammes SR, Smrcka AV, Blaxall BC. Simultaneous adrenal and cardiac g-protein-coupled receptor-gβγ inhibition halts heart failure progression. J Am Coll Cardiol 2014; 63:2549-2557. [PMID: 24703913 DOI: 10.1016/j.jacc.2014.02.587] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 02/10/2014] [Accepted: 02/25/2014] [Indexed: 12/29/2022]
Abstract
OBJECTIVES The authors propose simultaneous inhibition of Gβγ signaling in the heart and the adrenal gland as a novel therapeutic approach for heart failure (HF). BACKGROUND Elevated sympathetic nervous system activity is a salient characteristic of HF progression. It causes pathologic desensitization of β-adrenergic receptors (β-AR), facilitated predominantly through Gβγ-mediated signaling. The adrenal glands are key contributors to the chronically elevated plasma catecholamine levels observed in HF, where adrenal α2-AR feedback inhibitory function is impaired also through Gβγ-mediated signaling. METHODS We investigated the efficacy of a small molecule Gβγ inhibitor, gallein, in a clinically relevant, pressure-overload model of HF. RESULTS Daily gallein treatment (10 mg/kg/day), initiated 4 weeks after transverse aortic constriction, improved survival and cardiac function and attenuated cardiac remodeling. Mechanistically, gallein restored β-AR membrane density in cardiomyocytes, attenuated Gβγ-mediated G-protein-coupled receptor kinase 2-phosphoinositide 3-kinase γ membrane recruitment, and reduced Akt (protein kinase B) and glycogen synthase kinase 3β phosphorylation. Gallein also reduced circulating plasma catecholamine levels and catecholamine production in isolated mouse adrenal glands by restoring adrenal α2-AR feedback inhibition. In human adrenal endocrine tumors (pheochromocytoma), gallein attenuated catecholamine secretion, as well as G-protein-coupled receptor kinase 2 expression and membrane translocation. CONCLUSIONS These data suggest small molecule Gβγ inhibition as a systemic pharmacologic therapy for HF by simultaneously normalizing pathologic adrenergic/Gβγ signaling in both the heart and the adrenal gland. Our data also suggest important endocrine/cardiovascular interactions and a possible role for small molecule Gβγ inhibition in treating endocrine tumors such as pheochromocytoma, in addition to HF.
Collapse
Affiliation(s)
- Fadia A Kamal
- The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Deanne M Mickelsen
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester Medical Center, Rochester, New York
| | - Katherine M Wegman
- The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Joshua G Travers
- The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Jacob Moalem
- Department of Surgery, University of Rochester Medical Center, Rochester, New York; Department of Medicine, University of Rochester Medical Center, Rochester, New York
| | - Stephen R Hammes
- Department of Medicine, University of Rochester Medical Center, Rochester, New York
| | - Alan V Smrcka
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, New York
| | - Burns C Blaxall
- The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.
| |
Collapse
|
26
|
Ceylan-Isik AF, Dong M, Zhang Y, Dong F, Turdi S, Nair S, Yanagisawa M, Ren J. Cardiomyocyte-specific deletion of endothelin receptor A rescues aging-associated cardiac hypertrophy and contractile dysfunction: role of autophagy. Basic Res Cardiol 2013; 108:335. [PMID: 23381122 DOI: 10.1007/s00395-013-0335-3] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Revised: 12/27/2012] [Accepted: 01/24/2013] [Indexed: 11/25/2022]
Abstract
Cardiac aging is manifested as cardiac remodeling and contractile dysfunction although precise mechanisms remain elusive. This study was designed to examine the role of endothelin-1 (ET-1) in aging-associated myocardial morphological and contractile defects. Echocardiographic and cardiomyocyte contractile properties were evaluated in young (5-6 months) and old (26-28 months) C57BL/6 wild-type and cardiomyocyte-specific ET(A) receptor knockout (ETAKO) mice. Cardiac ROS production and histology were examined. Our data revealed that ETAKO mice displayed an improved survival. Aging increased plasma levels of ET-1 and Ang II, compromised cardiac function (fractional shortening, cardiomyocyte peak shortening, maximal velocity of shortening/relengthening and prolonged relengthening) and intracellular Ca(2+) handling (reduced intracellular Ca(2+) release and decay), the effects of which with the exception of ET-1 and Ang II levels was improved by ETAKO. Histological examination displayed cardiomyocyte hypertrophy and interstitial fibrosis associated with cardiac remodeling in aged C57 mice, which were alleviated in ETAKO mice. Aging promoted ROS generation, protein damage, ER stress, upregulated GATA4, ANP, NFATc3 and the autophagosome cargo protein p62, downregulated intracellular Ca(2+) regulatory proteins SERCA2a and phospholamban as well as the autophagic markers Beclin-1, Atg7, Atg5 and LC3BII, which were ablated by ETAKO. ET-1 triggered a decrease in autophagy and increased hypertrophic markers in vitro, the effect of which were reversed by the ET(A) receptor antagonist BQ123 and the autophagy inducer rapamycin. Antagonism of ET(A), but not ET(B) receptor, rescued cardiac aging, which was negated by autophagy inhibition. Taken together, our data suggest that cardiac ET(A) receptor ablation protects against aging-associated myocardial remodeling and contractile dysfunction possibly through autophagy regulation.
Collapse
Affiliation(s)
- Asli F Ceylan-Isik
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Gilsbach R, Hein L. Are the pharmacology and physiology of α₂ adrenoceptors determined by α₂-heteroreceptors and autoreceptors respectively? Br J Pharmacol 2012; 165:90-102. [PMID: 21658028 DOI: 10.1111/j.1476-5381.2011.01533.x] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
α(2)-Adrenoceptors are important mediators of physiological responses to the endogenous catecholamines noradrenaline and adrenaline. In addition, α(2)-adrenoceptors are pharmacological targets for the treatment of hypertension, sympathetic overactivity and glaucoma. α(2)-Adrenoceptors are also targeted to induce sedation and analgesia in anaesthesia and intensive care. α(2)-Adrenoceptors were first described as presynaptic receptors inhibiting the release of various transmitters from neurons in the central and peripheral nervous systems. In addition to these presynaptic neuronal receptors, α(2)-adrenoceptors were also identified in many non-neuronal cell types of the body. Gene-targeting in mice provided a comprehensive assignment of the physiological and pharmacological functions of these receptors to specific α(2A)-, α(2B) - and α(2C)-adrenoceptor subtypes. However, the specific cell types and signalling pathways involved in these subtype-specific α(2)-adrenoceptor functions were largely unexplored until recently. This review summarizes recent findings from transgenic mouse models, which were generated to define the role of α(2)-adrenoceptors in adrenergic neurons, that is, α(2)-autoreceptors, versus α(2)-adrenoceptors in non-adrenergic neurons, termed α(2)-heteroreceptors. α(2)-Autoreceptors are primarily required to limit release of noradrenaline from sympathetic nerves and adrenaline from adrenal chromaffin cells at rest. These receptors are desensitized upon chronic activation as it may for instance occur due to enhanced sympathetic activity during chronic heart failure. In contrast, pharmacological effects of acutely administered α(2)-adrenoceptor agonist drugs essentially require α(2)-heteroreceptors in non-adrenergic neurons, including analgesia, sedation, hypothermia and anaesthetic-sparing as well as bradycardia and hypotension. Thus a clear picture has emerged of the significance of auto- versus heteroreceptors in mediating the physiological functions of α(2)-adrenoceptors and the pharmacological functions of α(2)-adrenoceptor agonist drugs respectively.
Collapse
Affiliation(s)
- Ralf Gilsbach
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Freiburg, Freiburg, Germany.
| | | |
Collapse
|