Network


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

Hotspot


Dive into the research topics where Suzanne E. Brett is active.

Publication


Featured researches published by Suzanne E. Brett.


American Journal of Physiology-heart and Circulatory Physiology | 1999

Codistribution of NOS and caveolin throughout peripheral vasculature and skeletal muscle of hamsters

Steven S. Segal; Suzanne E. Brett; William C. Sessa

In isolated cell systems, nitric oxide synthase (NOS) activity is regulated by caveolin (CAV), a resident caveolae coat protein. Because little is known of this interaction in vivo, we tested whether NOS and caveolin are distributed together in the intact organism. Using immunohistochemistry, we investigated the localization of constitutive neuronal (nNOS) and endothelial (eNOS) enzyme isoforms along with caveolin-1 (CAV-1) and caveolin-3 (CAV-3) throughout the systemic vasculature and peripheral tissues of the hamster. The carotid artery, abdominal aorta, vena cava, femoral artery and vein, feed artery and collecting vein of the cheek pouch retractor muscle, capillaries and muscle fibers of retractor and cremaster muscles, and arterioles and venules of the cheek pouch were studied. In endothelial cells, eNOS and CAV-1 were present throughout the vasculature, whereas nNOS and CAV-3 were absent except in capillaries, which reacted for nNOS. In smooth muscle cells, nNOS and CAV-1 were also expressed systemically, whereas eNOS was absent; CAV-3 was present in the arterial but not the venous vasculature. Both nNOS and CAV-3 were located at the sarcolemma of skeletal muscle fibers, which were devoid of eNOS and CAV-1. These immunolabeling patterns suggest functional interactions between eNOS and CAV-1 throughout the endothelium, regional differences in the modulation of nNOS by caveolin isoforms in vascular smooth muscle, and modulation of nNOS by CAV-3 in skeletal muscle.


The Journal of Physiology | 2008

KIR channels function as electrical amplifiers in rat vascular smooth muscle

Pamela D. Smith; Suzanne E. Brett; Kevin D. Luykenaar; Shaun L. Sandow; Sean P. Marrelli; Edward J. Vigmond; Donald G. Welsh

Strong inward rectifying K+ (KIR) channels have been observed in vascular smooth muscle and can display negative slope conductance. In principle, this biophysical characteristic could enable KIR channels to ‘amplify’ responses initiated by other K+ conductances. To test this, we have characterized the diversity of smooth muscle KIR properties in resistance arteries, confirmed the presence of negative slope conductance and then determined whether KIR inhibition alters the responsiveness of middle cerebral, coronary septal and third‐order mesenteric arteries to K+ channel activators. Our initial characterization revealed that smooth muscle KIR channels were highly expressed in cerebral and coronary, but not mesenteric arteries. These channels comprised KIR2.1 and 2.2 subunits and electrophysiological recordings demonstrated that they display negative slope conductance. Computational modelling predicted that a KIR‐like current could amplify the hyperpolarization and dilatation initiated by a vascular K+ conductance. This prediction was consistent with experimental observations which showed that 30 μm Ba2+ attenuated the ability of K+ channel activators to dilate cerebral and coronary arteries. This attenuation was absent in mesenteric arteries where smooth muscle KIR channels were poorly expressed. In summary, smooth muscle KIR expression varies among resistance arteries and when channel are expressed, their negative slope conductance amplifies responses initiated by smooth muscle and endothelial K+ conductances. These findings highlight the fact that the subtle biophysical properties of KIR have a substantive, albeit indirect, role in enabling agonists to alter the electrical state of a multilayered artery.


The Journal of Physiology | 2010

Intravascular pressure augments cerebral arterial constriction by inducing voltage-insensitive Ca2+ waves

Rania E. Mufti; Suzanne E. Brett; Cam Ha T. Tran; Rasha Abd El-Rahman; Yana Anfinogenova; Ahmed F. El-Yazbi; William C. Cole; Peter P. Jones; S. R. Wayne Chen; Donald G. Welsh

This study examined whether elevated intravascular pressure stimulates asynchronous Ca2+ waves in cerebral arterial smooth muscle cells and if their generation contributes to myogenic tone development. The endothelium was removed from rat cerebral arteries, which were then mounted in an arteriograph, pressurized (20–100 mmHg) and examined under a variety of experimental conditions. Diameter and membrane potential (VM) were monitored using conventional techniques; Ca2+ wave generation and myosin light chain (MLC20)/MYPT1 (myosin phosphatase targeting subunit) phosphorylation were assessed by confocal microscopy and Western blot analysis, respectively. Elevating intravascular pressure increased the proportion of smooth muscle cells firing asynchronous Ca2+ waves as well as event frequency. Ca2+ wave augmentation occurred primarily at lower intravascular pressures (<60 mmHg) and ryanodine, a plant alkaloid that depletes the sarcoplasmic reticulum (SR) of Ca2+, eliminated these events. Ca2+ wave generation was voltage insensitive as Ca2+ channel blockade and perturbations in extracellular [K+] had little effect on measured parameters. Ryanodine‐induced inhibition of Ca2+ waves attenuated myogenic tone and MLC20 phosphorylation without altering arterial VM. Thapsigargin, an SR Ca2+‐ATPase inhibitor also attenuated Ca2+ waves, pressure‐induced constriction and MLC20 phosphorylation. The SR‐driven component of the myogenic response was proportionally greater at lower intravascular pressures and subsequent MYPT1 phosphorylation measures revealed that SR Ca2+ waves facilitated pressure‐induced MLC20 phosphorylation through mechanisms that include myosin light chain phosphatase inhibition. Cumulatively, our findings show that mechanical stimuli augment Ca2+ wave generation in arterial smooth muscle and that these transient events facilitate tone development particularly at lower intravascular pressures by providing a proportion of the Ca2+ required to directly control MLC20 phosphorylation.


American Journal of Physiology-heart and Circulatory Physiology | 2013

Identification of L- and T-type Ca2+ channels in rat cerebral arteries: role in myogenic tone development

Rasha Abd El-Rahman; Osama F. Harraz; Suzanne E. Brett; Yana Anfinogenova; Rania E. Mufti; Daniel Goldman; Donald G. Welsh

L-type Ca(2+) channels are broadly expressed in arterial smooth muscle cells, and their voltage-dependent properties are important in tone development. Recent studies have noted that these Ca(2+) channels are not singularly expressed in vascular tissue and that other subtypes are likely present. In this study, we ascertained which voltage-gated Ca(2+) channels are expressed in rat cerebral arterial smooth muscle and determined their contribution to the myogenic response. mRNA analysis revealed that the α(1)-subunit of L-type (Ca(v)1.2) and T-type (Ca(v)3.1 and Ca(v)3.2) Ca(2+) channels are present in isolated smooth muscle cells. Western blot analysis subsequently confirmed protein expression in whole arteries. With the use of patch clamp electrophysiology, nifedipine-sensitive and -insensitive Ba(2+) currents were isolated and each were shown to retain electrical characteristics consistent with L- and T-type Ca(2+) channels. The nifedipine-insensitive Ba(2+) current was blocked by mibefradil, kurtoxin, and efonidpine, T-type Ca(2+) channel inhibitors. Pressure myography revealed that L-type Ca(2+) channel inhibition reduced tone at 20 and 80 mmHg, with the greatest effect at high pressure when the vessel is depolarized. In comparison, the effect of T-type Ca(2+) channel blockade on myogenic tone was more limited, with their greatest effect at low pressure where vessels are hyperpolarized. Blood flow modeling revealed that the vasomotor responses induced by T-type Ca(2+) blockade could alter arterial flow by ∼20-50%. Overall, our findings indicate that L- and T-type Ca(2+) channels are expressed in cerebral arterial smooth muscle and can be electrically isolated from one another. Both conductances contribute to myogenic tone, although their overall contribution is unequal.


American Journal of Physiology-heart and Circulatory Physiology | 2014

Nitric Oxide Suppresses Vascular Voltage-Gated T-Type Ca2+ Channels Through cGMP/PKG Signaling

Osama F. Harraz; Suzanne E. Brett; Donald G. Welsh

Recent reports have noted that T-type Ca2+ channels (CaV3.x) are expressed in vascular smooth muscle and are potential targets of regulation. In this study, we examined whether and by what mechanism nitric oxide (NO), a key vasodilator, influences this conductance. Using patch-clamp electrophysiology and rat cerebral arterial smooth muscle cells, we monitored an inward Ba2+ current that was divisible into a nifedipine-sensitive and -insensitive component. The latter was abolished by T-type channel blocker and displayed classic T-type properties including faster activation and steady-state inactivation at hyperpolarized potentials. NO donors (sodium nitroprusside, S-nitroso-N-acetyl-dl-penicillamine), along with activators of protein kinase G (PKG) signaling, suppressed T-type currents. Inhibitors of guanylyl cyclase/PKG {1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ) and KT5823, respectively}, had no effect on basal currents; KT5823 did, however, mask T-type Ca2+ channel current inhibition by NO/PKG. Functional experiments confirmed an inhibitory effect for NO on the T-type contribution to cerebral arterial myogenic tone. Cumulatively, our findings support the view that T-type Ca2+ channels are a regulatory target of vasodilatory signaling pathways. This targeting will influence Ca2+ dynamics and consequent tone development in the cerebral circulation.


Circulation Research | 2014

CaV3.2 Channels and the Induction of Negative Feedback in Cerebral Arteries

Osama F. Harraz; Rasha Abd El-Rahman; Kamran Bigdely-Shamloo; Sean M. Wilson; Suzanne E. Brett; Monica Romero; Albert L. Gonzales; Scott Earley; Edward J. Vigmond; Anders Nygren; Bijoy K. Menon; Rania E. Mufti; Timothy Watson; Yves Starreveld; Tobias Fürstenhaupt; Philip R. Muellerleile; David T. Kurjiaka; Barry D. Kyle; Andrew P. Braun; Donald G. Welsh

Rationale: T-type (CaV3.1/CaV3.2) Ca2+ channels are expressed in rat cerebral arterial smooth muscle. Although present, their functional significance remains uncertain with findings pointing to a variety of roles. Objective: This study tested whether CaV3.2 channels mediate a negative feedback response by triggering Ca2+ sparks, discrete events that initiate arterial hyperpolarization by activating large-conductance Ca2+-activated K+ channels. Methods and Results: Micromolar Ni2+, an agent that selectively blocks CaV3.2 but not CaV1.2/CaV3.1, was first shown to depolarize/constrict pressurized rat cerebral arteries; no effect was observed in CaV3.2−/− arteries. Structural analysis using 3-dimensional tomography, immunolabeling, and a proximity ligation assay next revealed the existence of microdomains in cerebral arterial smooth muscle which comprised sarcoplasmic reticulum and caveolae. Within these discrete structures, CaV3.2 and ryanodine receptor resided in close apposition to one another. Computational modeling revealed that Ca2+ influx through CaV3.2 could repetitively activate ryanodine receptor, inducing discrete Ca2+-induced Ca2+ release events in a voltage-dependent manner. In keeping with theoretical observations, rapid Ca2+ imaging and perforated patch clamp electrophysiology demonstrated that Ni2+ suppressed Ca2+ sparks and consequently spontaneous transient outward K+ currents, large-conductance Ca2+-activated K+ channel mediated events. Additional functional work on pressurized arteries noted that paxilline, a large-conductance Ca2+-activated K+ channel inhibitor, elicited arterial constriction equivalent, and not additive, to Ni2+. Key experiments on human cerebral arteries indicate that CaV3.2 is present and drives a comparable response to moderate constriction. Conclusions: These findings indicate for the first time that CaV3.2 channels localize to discrete microdomains and drive ryanodine receptor–mediated Ca2+ sparks, enabling large-conductance Ca2+-activated K+ channel activation, hyperpolarization, and attenuation of cerebral arterial constriction.


The Journal of General Physiology | 2015

CaV1.2/CaV3.x channels mediate divergent vasomotor responses in human cerebral arteries.

Osama F. Harraz; Frank Visser; Suzanne E. Brett; Daniel Goldman; Anil Zechariah; Ahmed M. Hashad; Bijoy K. Menon; Timothy Watson; Yves Starreveld; Donald G. Welsh

Human cerebral arteries contain three Ca2+ channel subtypes with distinct physiological roles.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2015

Genetic Ablation of CaV3.2 Channels Enhances the Arterial Myogenic Response by Modulating the RyR-BKCa Axis

Osama F. Harraz; Suzanne E. Brett; Anil Zechariah; Monica Romero; Jose L. Puglisi; Sean M. Wilson; Donald G. Welsh

Objective—In resistance arteries, there is an emerging view that smooth muscle CaV3.2 channels restrain arterial constriction through a feedback response involving the large-conductance Ca2+-activated K+ channel (BKCa). Here, we used wild-type and CaV3.2 knockout (CaV3.2−/−) mice to definitively test whether CaV3.2 moderates myogenic tone in mesenteric arteries via the CaV3.2-ryanodine receptor-BKCa axis and whether this regulatory mechanism influences blood pressure regulation. Approach and Results—Using pressurized vessel myography, CaV3.2−/− mesenteric arteries displayed enhanced myogenic constriction to pressure but similar K+-induced vasoconstriction compared with wild-type C57BL/6 arteries. Electrophysiological and myography experiments subsequently confirmed the inability of micromolar Ni2+, a CaV3.2 blocker, to either constrict arteries or suppress T-type currents in CaV3.2−/− smooth muscle cells. The frequency of BKCa-induced spontaneous transient outward K+ currents dropped in wild-type but not in knockout arterial smooth muscle cells upon the pharmacological suppression of CaV3.2 channel. Line scan analysis performed on en face arteries loaded with Fluo-4 revealed the presence of Ca2+ sparks in all arteries, with the subsequent application of Ni2+ only affecting wild-type arteries. Although CaV3.2 channel moderated myogenic constriction of resistance arteries, the blood pressure measurements of CaV3.2−/− and wild-type animals were similar. Conclusions—Overall, our findings establish a negative feedback mechanism of the myogenic response in which CaV3.2 channel modulates downstream ryanodine receptor-BKCa to hyperpolarize and relax arteries.


Journal of Cerebral Blood Flow and Metabolism | 2017

KIR channels tune electrical communication in cerebral arteries

Maria Sancho; Nina C Samson; Bjørn Olav Hald; Ahmed M. Hashad; Sean P. Marrelli; Suzanne E. Brett; Donald G. Welsh

The conducted vasomotor response reflects electrical communication in the arterial wall and the distance signals spread is regulated by three factors including resident ion channels. This study defined the role of inward-rectifying K+ channels (KIR) in governing electrical communication along hamster cerebral arteries. Focal KCl application induced a vasoconstriction that conducted robustly, indicative of electrical communication among cells. Inhibiting dominant K+ conductances had no attenuating effect, the exception being Ba2+ blockade of KIR. Electrophysiology and Q-PCR analysis of smooth muscle cells revealed a Ba2+-sensitive KIR current comprised of KIR2.1/2.2 subunits. This current was surprisingly small and when incorporated into a model, failed to account for the observed changes in conduction. We theorized a second population of KIR channels exist and consistent with this idea, a robust Ba2+-sensitive KIR2.1/2.2 current was observed in endothelial cells. When both KIR currents were incorporated into, and then inhibited in our model, conduction decay was substantive, aligning with experiments. Enhanced decay was ascribed to the rightward shift in membrane potential and the increased feedback arising from voltage-dependent-K+ channels. In summary, this study shows that two KIR populations work collaboratively to govern electrical communication and the spread of vasomotor responses along cerebral arteries.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2015

Implications of αvβ3 Integrin Signaling in the Regulation of Ca2+ Waves and Myogenic Tone in Cerebral Arteries

Rania E. Mufti; Anil Zechariah; Maria Sancho; Neil Mazumdar; Suzanne E. Brett; Donald G. Welsh

Objective—The myogenic response is central to blood flow regulation in the brain. Its induction is tied to elevated cytosolic [Ca2+], a response primarily driven by voltage-gated Ca2+ channels and secondarily by Ca2+ wave production. Although the signaling events leading to the former are well studied, those driving Ca2+ waves remain uncertain. Approach and Results—We postulated that &agr;v&bgr;3 integrin signaling is integral to the generation of pressure-induced Ca2+ waves and cerebral arterial tone. This hypothesis was tested in rat cerebral arteries using the synergistic strengths of pressure myography, rapid Ca2+ imaging, and Western blot analysis. GRGDSP, a peptide that preferentially blocks &agr;v&bgr;3 integrin, attenuated myogenic tone, indicating the modest role for sarcoplasmic reticulum Ca2+ release in myogenic tone generation. The RGD peptide was subsequently shown to impair Ca2+ wave generation and myosin light chain 20 (MLC20) phosphorylation, the latter of which was attributed to the modulation of MLC kinase and MLC phosphatase via MYPT1-T855 phosphorylation. Subsequent experiments revealed that elevated pressure enhanced phospholipase C&ggr;1 phosphorylation in an RGD-dependent manner and that phospholipase C inhibition attenuated Ca2+ wave generation. Direct inhibition of inositol 1, 4, 5-triphosphate receptors also impaired Ca2+ wave generation, myogenic tone, and MLC20 phosphorylation, partly through the T-855 phosphorylation site of MYPT1. Conclusions—Our investigation reveals a hitherto unknown role for &agr;v&bgr;3 integrin as a cerebral arterial pressure sensor. The membrane receptor facilitates Ca2+ wave generation through a signaling cascade, involving phospholipase C&ggr;1, inositol 1,3,4 triphosphate production, and inositol 1, 4, 5-triphosphate receptor activation. These discrete asynchronous Ca2+ events facilitate MLC20 phosphorylation and, in part, myogenic tone by influencing both MLC kinase and MLC phosphatase activity.

Collaboration


Dive into the Suzanne E. Brett's collaboration.

Top Co-Authors

Avatar

Donald G. Welsh

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anil Zechariah

University of Duisburg-Essen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Maria Sancho

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge