Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Laurel A. Grisanti is active.

Publication


Featured researches published by Laurel A. Grisanti.


Neuron | 2009

GABAB Receptor Activation Inhibits Neuronal Excitability and Spatial Learning in the Entorhinal Cortex by Activating TREK-2 K+ Channels

Pan-Yue Deng; Zhaoyang Xiao; Chuanxiu Yang; Lalida Rojanathammanee; Laurel A. Grisanti; John A. Watt; Jonathan D. Geiger; Rugao Liu; James E. Porter; Saobo Lei

The entorhinal cortex (EC) is regarded as the gateway to the hippocampus and thus is essential for learning and memory. Whereas the EC expresses a high density of GABA(B) receptors, the functions of these receptors in this region remain unexplored. Here, we examined the effects of GABA(B) receptor activation on neuronal excitability in the EC and spatial learning. Application of baclofen, a specific GABA(B) receptor agonist, inhibited significantly neuronal excitability in the EC. GABA(B) receptor-mediated inhibition in the EC was mediated via activating TREK-2, a type of two-pore domain K(+) channels, and required the functions of inhibitory G proteins and protein kinase A pathway. Depression of neuronal excitability in the EC underlies GABA(B) receptor-mediated inhibition of spatial learning as assessed by Morris water maze. Our study indicates that GABA(B) receptors exert a tight control over spatial learning by modulating neuronal excitability in the EC.


Molecular Immunology | 2010

Pro-inflammatory responses in human monocytes are β1-adrenergic receptor subtype dependent☆☆☆

Laurel A. Grisanti; Janel Evanson; Erica Marchus; Heather Jorissen; Andrew P. Woster; Wanda DeKrey; Edward R. Sauter; Colin K. Combs; James E. Porter

Stress induced circulating catecholamines are hypothesized to selectively activate adrenergic receptors (ARs) on immunocompetent cells modulating their inflammatory response to trauma or environmental toxins. We characterized changes in expression of a pro-inflammatory cytokine modulated by beta-AR activation in human primary and immortalized monocytes that had been simultaneously stimulated with lipopolysaccharide (LPS). Results from cytokine antibody arrays demonstrated that half-maximal effective concentrations of the selective beta-AR agonist isoproterenol (Iso) qualitatively increased LPS-mediated expression of the soluble cytokine, interleukin-1beta (IL-1beta). Semi-quantitative immunoblot techniques confirmed a synergistic increase of IL-1beta production in both LPS stimulated THP-1 cells and primary human monocytes co-incubated with Iso. Immunoblot techniques as well as radioligand binding studies were also used to characterize the heterogeneous expression of beta(1)- and beta(2)-AR subtypes on THP-1 cells. beta-AR activation is classically associated with generation of cAMP in many tissues and cell types. Therefore, using the method of Schild, we generated Iso concentration-response curves in the presence of fixed subtype-selective beta-AR antagonist concentrations to demonstrate that beta(1)-AR activation was exclusively linked with the generation of cAMP in THP-1 cells. Furthermore, use of a selective kinase inhibitor demonstrated that Iso potentiated the expression of soluble IL-1beta through activation of cAMP-dependent protein kinase A. Finally, discriminating concentrations of subtype-selective beta-AR antagonists revealed that beta(1)-AR stimulation alone accounted for the synergistic production of IL-1beta in LPS stimulated monocytes co-incubated with Iso. These results demonstrate a unique synergistic pro-inflammatory response mediated through a beta(1)-AR cAMP-dependent mechanism in LPS-challenged monocytic cells.


Journal of Pharmacology and Experimental Therapeutics | 2011

α1-adrenergic receptors positively regulate Toll-like receptor cytokine production from human monocytes and macrophages.

Laurel A. Grisanti; Andrew P. Woster; Julie Dahlman; Edward R. Sauter; Colin K. Combs; James E. Porter

Catecholamines released from the sympathetic nervous system in response to stress or injury affect expression of inflammatory cytokines generated by immune cells. α1-Adrenergic receptors (ARs) are expressed on innate immune cell populations, but their subtype expression patterns and signaling characteristics are not well characterized. Primary human monocytes, a human monocytic cell line, and monocyte-derived macrophage cells were used to measure expression of the proinflammatory mediator interleukin (IL)-1β responding to lipopolysaccharide (LPS) in the presence or absence of α1-AR activation. Based on our previous findings, we hypothesized that α1-AR stimulation on innate immune cells positively regulates LPS-initiated IL-1β production. IL-1β production in response to LPS was synergistically higher for both monocytes and macrophages in the presence of the selective α1-AR agonist (R)-(−)-phenylephrine hydrochloride (PE). This synergistic IL-1β response could be blocked with a selective α1-AR antagonist as well as inhibitors of protein kinase C (PKC). Radioligand binding studies characterized a homogenous α1B-AR subtype population on monocytes, which changed to a heterogeneous receptor subtype expression pattern when differentiated to macrophages. Furthermore, increased p38 mitogen-activated protein kinase (MAPK) activation was observed only with concurrent PE and LPS stimulation, peaking after 120 and 30 min in monocytes and macrophages, respectively. Blocking the PKC/p38 MAPK signaling pathway in both innate immune cell types inhibited the synergistic IL-1β increase observed with concurrent PE and LPS treatments. This study characterizes α1-AR subtype expression on both human monocyte and macrophage cells and illustrates a mechanism by which increased IL-1β production can be modulated by α1-AR input.


Journal of Biological Chemistry | 2009

Noradrenergic Depression of Neuronal Excitability in the Entorhinal Cortex via Activation of TREK-2 K+ Channels

Zhaoyang Xiao; Pan-Yue Deng; Lalida Rojanathammanee; Chuanxiu Yang; Laurel A. Grisanti; Kannika Permpoonputtana; David Weinshenker; Van Doze; James E. Porter; Saobo Lei

The entorhinal cortex is closely associated with the consolidation and recall of memories, Alzheimer disease, schizophrenia, and temporal lobe epilepsy. Norepinephrine is a neurotransmitter that plays a significant role in these physiological functions and neurological diseases. Whereas the entorhinal cortex receives profuse noradrenergic innervations from the locus coeruleus of the pons and expresses high densities of adrenergic receptors, the function of norepinephrine in the entorhinal cortex is still elusive. Accordingly, we examined the effects of norepinephrine on neuronal excitability in the entorhinal cortex and explored the underlying cellular and molecular mechanisms. Application of norepinephrine-generated hyperpolarization and decreased the excitability of the neurons in the superficial layers with no effects on neuronal excitability in the deep layers of the entorhinal cortex. Norepinephrine-induced hyperpolarization was mediated by α2A adrenergic receptors and required the functions of Gαi proteins, adenylyl cyclase, and protein kinase A. Norepinephrine-mediated depression on neuronal excitability was mediated by activation of TREK-2, a type of two-pore domain K+ channel, and mutation of the protein kinase A phosphorylation site on TREK-2 channels annulled the effects of norepinephrine. Our results indicate a novel action mode in which norepinephrine depresses neuronal excitability in the entorhinal cortex by disinhibiting protein kinase A-mediated tonic inhibition of TREK-2 channels.


Circulation Research | 2014

GRK5-Mediated Exacerbation of Pathological Cardiac Hypertrophy Involves Facilitation of Nuclear NFAT Activity

Jonathan Hullmann; Laurel A. Grisanti; Catherine A. Makarewich; Erhe Gao; Jessica I. Gold; J. Kurt Chuprun; Douglass G. Tilley; Steven R. Houser; Walter J. Koch

Rationale: G protein–coupled receptor kinases (GRKs) acting in the cardiomyocyte regulate important signaling events that control cardiac function. Both GRK2 and GRK5, the predominant GRKs expressed in the heart, have been shown to be upregulated in failing human myocardium. Although the canonical role of GRKs is to desensitize G protein–coupled receptors via phosphorylation, it has been demonstrated that GRK5, unlike GRK2, can reside in the nucleus of myocytes and exert G protein–coupled receptor–independent effects that promote maladaptive cardiac hypertrophy and heart failure. Objective: To explore novel mechanisms by which GRK5 acting in the nucleus of cardiomyocytes participates in pathological cardiac hypertrophy. Methods and Results: In this study, we have found that GRK5-mediated pathological cardiac hypertrophy involves the activation of the nuclear factor of activated T cells (NFAT) because GRK5 causes enhancement of NFAT-mediated hypertrophic gene transcription. Transgenic mice with cardiomyocyte-specific GRK5 overexpression activate an NFAT-reporter in mice basally and after hypertrophic stimulation, including transverse aortic constriction and phenylephrine treatment. Complimentary to this, GRK5 null mice exhibit less NFAT transcriptional activity after transverse aortic constriction. Furthermore, the loss of NFATc3 expression in the heart protected GRK5 overexpressing transgenic mice from the exaggerated hypertrophy and early progression to heart failure seen after transverse aortic constriction. Molecular studies suggest that GRK5 acts in concert with NFAT to increase hypertrophic gene transcription in the nucleus via GRK5’s ability to bind DNA directly without a phosphorylation event. Conclusions: GRK5, acting in a kinase independent manner, is a facilitator of NFAT activity and part of a DNA-binding complex responsible for pathological hypertrophic gene transcription.


Journal of Molecular and Cellular Cardiology | 2014

β-Adrenergic receptor-mediated transactivation of epidermal growth factor receptor decreases cardiomyocyte apoptosis through differential subcellular activation of ERK1/2 and Akt

Laurel A. Grisanti; Jennifer A. Talarico; Rhonda L. Carter; Justine E. Yu; Ashley A. Repas; Scott W. Radcliffe; Hoang-ai Tang; Catherine A. Makarewich; Steven R. Houser; Douglas G. Tilley

β-Adrenergic receptor (βAR)-mediated transactivation of epidermal growth factor receptor (EGFR) has been shown to relay pro-survival effects via unknown mechanisms. We hypothesized that acute βAR-mediated EGFR transactivation in the heart promotes differential subcellular activation of ERK1/2 and Akt, promoting cell survival through modulation of apoptosis. C57BL/6 mice underwent acute i.p. injection with isoproterenol (ISO)±AG 1478 (EGFR antagonist) to assess the impact of βAR-mediated EGFR transactivation on the phosphorylation of ERK1/2 (P-ERK1/2) and Akt (P-Akt) in distinct cardiac subcellular fractions. Increased P-ERK1/2 and P-Akt were observed in cytosolic, plasma membrane and nuclear fractions following ISO stimulation. Whereas the P-ERK1/2 response was EGFR-sensitive in all fractions, the P-Akt response was EGFR-sensitive only in the plasma membrane and nucleus, results confirmed in primary rat neonatal cardiomyocytes (RNCM). βAR-mediated EGFR-transactivation also decreased apoptosis in serum-depleted RNCM, as measured via TUNEL as well as caspase 3 activity/cleavage, which were sensitive to the inhibition of either ERK1/2 (PD184352) or Akt (LY-294002) signaling. Caspase 3 activity/cleavage was also sensitive to the inhibition of transcription, which, with an increase in nuclear P-ERK1/2 and P-Akt in response to ISO, suggested that βAR-mediated EGFR transactivation may regulate apoptotic gene transcription. An Apoptosis PCR Array identified tnfsf10 (TRAIL) to be altered by ISO in an EGFR-sensitive manner, results confirmed via RT-PCR and ELISA measurement of both membrane-bound and soluble cardiomyocyte TRAIL levels. βAR-mediated EGFR transactivation induces differential subcellular activation of ERK1/2 and Akt leading to increased cell survival through the modulation of caspase 3 activity and apoptotic gene expression in cardiomyocytes.


Circulation | 2014

β-Adrenergic Receptor-Mediated Cardiac Contractility is Inhibited via Vasopressin Type 1A-Receptor-Dependent Signaling

Douglas G. Tilley; Weizhong Zhu; Valerie D. Myers; Larry A. Barr; Erhe Gao; Xue Li; Jiangliang Song; Rhonda L. Carter; Catherine A. Makarewich; Daohai Yu; Constantine D. Troupes; Laurel A. Grisanti; Ryan C. Coleman; Walter J. Koch; Steven R. Houser; Joseph Y. Cheung; Arthur M. Feldman

Background— Enhanced arginine vasopressin levels are associated with increased mortality during end-stage human heart failure, and cardiac arginine vasopressin type 1A receptor (V1AR) expression becomes increased. Additionally, mice with cardiac-restricted V1AR overexpression develop cardiomyopathy and decreased &bgr;-adrenergic receptor (&bgr;AR) responsiveness. This led us to hypothesize that V1AR signaling regulates &bgr;AR responsiveness and in doing so contributes to development of heart failure. Methods and Results— Transaortic constriction resulted in decreased cardiac function and &bgr;AR density and increased cardiac V1AR expression, effects reversed by a V1AR-selective antagonist. Molecularly, V1AR stimulation led to decreased &bgr;AR ligand affinity, as well as &bgr;AR-induced Ca2+ mobilization and cAMP generation in isolated adult cardiomyocytes, effects recapitulated via ex vivo Langendorff analysis. V1AR-mediated regulation of &bgr;AR responsiveness was demonstrated to occur in a previously unrecognized Gq protein–independent/G protein receptor kinase–dependent manner. Conclusions— This newly discovered relationship between cardiac V1AR and &bgr;AR may be informative for the treatment of patients with acute decompensated heart failure and elevated arginine vasopressin.


Current Topics in Membranes | 2011

Modulation of Immune Cell Function by α1-Adrenergic Receptor Activation

Laurel A. Grisanti; Dianne M. Perez; James E. Porter

Publisher Summary This chapter discusses the modulation of immune cell function by α 1 -adrenergic receptor activation. The sympathetic nervous system regulates human immune system functions through the epinephrine (Epi) and norepinephrine (NE) activation of adrenergic receptors (ARs) expressed on immune competent cell populations. The anti-inflammatory effects that are most often attributed to increased sympathetic activity have been shown to occur through β 2 - and α 2 -AR stimulation. However, dichotomous AR effects on immune system responses are becoming increasingly apparent. The endogenous catecholamines, Epi and NE, are critical for initiating the fight or flight response of the sympathetic nervous system. Epi and NE are released from peripheral neurons and the adrenal medulla in response to physical as well as psychological stress to regulate a number of physiological functions, including energy metabolism, cardiovascular homeostasis, and thermal adaptation. The human innate immune system is a nonspecific means of defense against pathogenic challenges. This generic means of defense is thought to be a more evolutionary primitive design compared to the adaptive immune system.


Journal of Biological Chemistry | 2013

Differential Activation of Cultured Neonatal Cardiomyocytes by Plasmalemmal Versus Intracellular G Protein-coupled Receptor 55

Justine E. Yu; Elena Deliu; Xue-Quian Zhang; Nicholas E. Hoffman; Rhonda L. Carter; Laurel A. Grisanti; G. Cristina Brailoiu; Muniswamy Madesh; Joseph Y. Cheung; Thomas Force; Mary E. Abood; Walter J. Koch; Douglas G. Tilley; Eugen Brailoiu

Background: The LPI-sensitive receptor GPR55 signals through Ca2+. Results: Activation of sarcolemmal versus intracellular GPR55 mobilizes Ca2+ from distinct pools and associates with cardiomyocyte depolarization and hyperpolarization, respectively. Conclusion: GPR55 location critically affects LPI-induced modulation of cardiomyocyte function. Significance: We identify GPR55 as a new receptor regulating cardiac function at two cellular sites. The L-α-lysophosphatidylinositol (LPI)-sensitive receptor GPR55 is coupled to Ca2+ signaling. Low levels of GPR55 expression in the heart have been reported. Similar to other G protein-coupled receptors involved in cardiac function, GPR55 may be expressed both at the sarcolemma and intracellularly. Thus, to explore the role of GPR55 in cardiomyocytes, we used calcium and voltage imaging and extracellular administration or intracellular microinjection of GPR55 ligands. We provide the first evidence that, in cultured neonatal ventricular myocytes, LPI triggers distinct signaling pathways via GPR55, depending on receptor localization. GPR55 activation at the sarcolemma elicits, on one hand, Ca2+ entry via L-type Ca2+ channels and, on the other, inositol 1,4,5-trisphosphate-dependent Ca2+ release. The latter signal is further amplified by Ca2+-induced Ca2+ release via ryanodine receptors. Conversely, activation of GPR55 at the membrane of intracellular organelles promotes Ca2+ release from acidic-like Ca2+ stores via the endolysosomal NAADP-sensitive two-pore channels. This response is similarly enhanced by Ca2+-induced Ca2+ release via ryanodine receptors. Extracellularly applied LPI produces Ca2+-independent membrane depolarization, whereas the Ca2+ signal induced by intracellular microinjection of LPI converges to hyperpolarization of the sarcolemma. Collectively, our findings point to GPR55 as a novel G protein-coupled receptor regulating cardiac function at two cellular sites. This work may serve as a platform for future studies exploring the potential of GPR55 as a therapeutic target in cardiac disorders.


Molecular Pharmacology | 2009

The 27-kDa Heat Shock Protein Confers Cytoprotective Effects through a β2-Adrenergic Receptor Agonist-Initiated Complex with β-Arrestin

Lalida Rojanathammanee; Erin B. Harmon; Laurel A. Grisanti; Piyarat Govitrapong; Manuchair Ebadi; Bryon D. Grove; Masaru Miyagi; James E. Porter

Heat shock proteins represent an emerging model for the coordinated, multistep regulation of apoptotic signaling events. Although certain aspects of the biochemistry associated with heat shock protein cytoprotective effects are known, little information is found describing the regulation of heat shock protein responses to harmful stimuli. During screening for noncanonical β adrenergic receptor signaling pathways in human urothelial cells, using mass spectroscopy techniques, an agonist-dependent interaction with β-arrestin and the 27-kDa heat shock protein was observed in vitro. Formation of this β-arrestin/Hsp27 complex in response to the selective β adrenergic receptor agonist isoproterenol, was subsequently confirmed in situ by immunofluorescent colocalization studies. Radioligand binding techniques characterized a homogeneous population of the β2 adrenergic receptor subtype expressed on these cells. Using terminal deoxynucleotidyl transferase biotin-dUTP nick end labeling, immunoblot analysis and quantitation of caspase-3 activity to detect apoptosis, preincubation of these cells with isoproterenol was found to be sufficient for protection against programmed cell death initiated by staurosporine. RNA interference strategies confirmed the necessity for Hsp27 as well as both β-arrestin isoforms to confer this cytoprotective consequence of β adrenergic receptor activation in this cell model. As a result, these studies represent the first description of an agonist-dependent relationship between a small heat shock protein and β-arrestin to form a previously unknown antiapoptotic “signalosome.”

Collaboration


Dive into the Laurel A. Grisanti's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joseph Y. Cheung

Pennsylvania State University

View shared research outputs
Researchain Logo
Decentralizing Knowledge