Network


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

Hotspot


Dive into the research topics where Anne R. Crecelius is active.

Publication


Featured researches published by Anne R. Crecelius.


American Journal of Physiology-heart and Circulatory Physiology | 2010

Nitric Oxide, but not Vasodilating Prostaglandins, Contributes to the Improvement of Exercise Hyperemia via Ascorbic Acid in Healthy Older Adults

Anne R. Crecelius; Brett S. Kirby; Wyatt F. Voyles; Frank A. Dinenno

Acute ascorbic acid (AA) administration increases muscle blood flow during dynamic exercise in older adults, and this is associated with improved endothelium-dependent vasodilation. We directly tested the hypothesis that increase in muscle blood flow during AA administration is mediated via endothelium-derived vasodilators nitric oxide (NO) and prostaglandins (PGs). In 14 healthy older adults (64 ± 3 yr), we measured forearm blood flow (FBF; Doppler ultrasound) during rhythmic handgrip exercise at 10% maximum voluntary contraction. After 5-min steady-state exercise with saline, AA was infused via brachial artery catheter for 10 min during continued exercise, and this increased FBF ∼25% from 132 ± 16 to 165 ± 20 ml/min (P < 0.05). AA was infused for the remainder of the study. Next, subjects performed a 15-min exercise bout in which AA + saline was infused for 5 min, followed by 5 min of the nitric oxide synthase (NOS) inhibitor N(G)-monomethyl-l-arginine (l-NMMA) and then 5 min of the cyclooxygenase inhibitor ketorolac (group 1). The order of inhibition was reversed in eight subjects (group 2). In group 1, independent NOS inhibition reduced steady-state FBF by ∼20% (P < 0.05), and subsequent PG inhibition had no impact on FBF (Δ 3 ± 5%). Similarly, in group 2, independent PG inhibition had little effect on FBF (Δ -4 ± 4%), whereas subsequent NO inhibition significantly decreased FBF by ∼20% (P < 0.05). In a subgroup of five subjects, we inhibited NO and PG synthesis before AA administration. In these subjects, there was a minimal nonsignificant improvement in FBF with AA infusion (Δ 7 ± 3%; P = nonsignificant vs. zero). Together, our data indicate that the increase in muscle blood flow during dynamic exercise with acute AA administration in older adults is mediated primarily via an increase in the bioavailability of NO derived from the NOS pathway.


Circulation Research | 2012

Impaired skeletal muscle blood flow control with advancing age in humans: attenuated ATP release and local vasodilation during erythrocyte deoxygenation

Brett S. Kirby; Anne R. Crecelius; Wyatt F. Voyles; Frank A. Dinenno

Rationale: Skeletal muscle blood flow is coupled with the oxygenation state of hemoglobin in young adults, whereby the erythrocyte functions as an oxygen sensor and releases ATP during deoxygenation to evoke vasodilation. Whether this function is impaired in humans of advanced age is unknown. Objective: To test the hypothesis that older adults demonstrate impaired muscle blood flow and lower intravascular ATP during conditions of erythrocyte deoxygenation. Methods and Results: We showed impaired forearm blood flow responses during 2 conditions of erythrocyte deoxygenation (systemic hypoxia and graded handgrip exercise) with age, which was caused by reduced local vasodilation. In young adults, both hypoxia and exercise significantly increased venous [ATP] and ATP effluent (forearm blood flow×[ATP]) draining the skeletal muscle. In contrast, hypoxia and exercise did not increase venous [ATP] in older adults, and both venous [ATP] and ATP effluent were substantially reduced compared with young people despite similar levels of deoxygenation. Next, we demonstrated that this could not be explained by augmented extracellular ATP hydrolysis in whole blood with age. Finally, we found that deoxygenation-mediated ATP release from isolated erythrocytes was essentially nonexistent in older adults. Conclusions: Skeletal muscle blood flow during conditions of erythrocyte deoxygenation was markedly reduced in aging humans, and reductions in plasma ATP and erythrocyte-mediated ATP release may be a novel mechanism underlying impaired vasodilation and oxygen delivery during hypoxemia with advancing age. Because aging is associated with elevated risk for ischemic cardiovascular disease and exercise intolerance, interventions that target erythrocyte-mediated ATP release may offer therapeutic potential.Rationale: Skeletal muscle blood flow is coupled with the oxygenation state of hemoglobin in young adults, whereby the erythrocyte functions as an oxygen sensor and releases ATP during deoxygenation to evoke vasodilation. Whether this function is impaired in humans of advanced age is unknown. Objective: To test the hypothesis that older adults demonstrate impaired muscle blood flow and lower intravascular ATP during conditions of erythrocyte deoxygenation. Methods and Results: We showed impaired forearm blood flow responses during 2 conditions of erythrocyte deoxygenation (systemic hypoxia and graded handgrip exercise) with age, which was caused by reduced local vasodilation. In young adults, both hypoxia and exercise significantly increased venous [ATP] and ATP effluent (forearm blood flow×[ATP]) draining the skeletal muscle. In contrast, hypoxia and exercise did not increase venous [ATP] in older adults, and both venous [ATP] and ATP effluent were substantially reduced compared with young people despite similar levels of deoxygenation. Next, we demonstrated that this could not be explained by augmented extracellular ATP hydrolysis in whole blood with age. Finally, we found that deoxygenation-mediated ATP release from isolated erythrocytes was essentially nonexistent in older adults. Conclusions: Skeletal muscle blood flow during conditions of erythrocyte deoxygenation was markedly reduced in aging humans, and reductions in plasma ATP and erythrocyte-mediated ATP release may be a novel mechanism underlying impaired vasodilation and oxygen delivery during hypoxemia with advancing age. Because aging is associated with elevated risk for ischemic cardiovascular disease and exercise intolerance, interventions that target erythrocyte-mediated ATP release may offer therapeutic potential. # Novelty and Significance {#article-title-41}


The Journal of Physiology | 2011

Augmented skeletal muscle hyperaemia during hypoxic exercise in humans is blunted by combined inhibition of nitric oxide and vasodilating prostaglandins

Anne R. Crecelius; Brett S. Kirby; Wyatt F. Voyles; Frank A. Dinenno

Non‐technical summary  Blood flow to muscle increases during exercise in order to deliver more oxygen. When there is less oxygen in the blood, as in systemic hypoxia, blood flow also increases. If exercise occurs during hypoxia, the blood flow response is greater than during normal oxygen conditions, but the mechanisms by which this happens are not clear. We show that two substances that the body produces, nitric oxide and prostaglandins, contribute to this increased blood flow during hypoxic exercise. These results help us better understand how oxygen delivery is regulated and may be especially important for populations which are unable to produce these substances that help increase blood flow.


The Journal of Physiology | 2011

Combined inhibition of nitric oxide and vasodilating prostaglandins abolishes forearm vasodilatation to systemic hypoxia in healthy humans

Rachel R. Markwald; Brett S. Kirby; Anne R. Crecelius; Rick E. Carlson; Wyatt F. Voyles; Frank A. Dinenno

Non‐technical summary  During hypoxia, there is less oxygen in the air we breathe and also in the blood being pumped away from the heart. Our blood vessels must relax in order to deliver more blood to match the resting oxygen demand of the muscles. The way in which multiple systems in the body coordinate this response is not well known. We examined the local response of the blood vessels to a hypoxic stimulus and show that two substances that the body produces, nitric oxide and prostaglandins, are necessary to cause relaxation of the blood vessels and increases in blood flow. These results help us better understand how oxygen delivery is regulated and may be especially important for populations that are unable to produce these substances that help increase blood flow, such as people with sleep apnoea, heart failure and diabetes.


American Journal of Physiology-heart and Circulatory Physiology | 2011

Mechanisms of ATP-mediated vasodilation in humans: modest role for nitric oxide and vasodilating prostaglandins.

Anne R. Crecelius; Brett S. Kirby; Jennifer C. Richards; Leora J. Garcia; Wyatt F. Voyles; Dennis G. Larson; Gary J. Luckasen; Frank A. Dinenno

ATP is an endothelium-dependent vasodilator, and findings regarding the underlying signaling mechanisms are equivocal. We sought to determine the independent and interactive roles of nitric oxide (NO) and vasodilating prostaglandins (PGs) in ATP-mediated vasodilation in young, healthy humans and determine whether any potential role was dependent on ATP dose or the timing of inhibition. In protocol 1 (n = 18), a dose-response curve to intrabrachial infusion of ATP was performed before and after both single and combined inhibition of NO synthase [N(G)-monomethyl-L-arginine (L-NMMA)] and cyclooxygenase (ketorolac). Forearm blood flow (FBF) was measured via venous occlusion plethysmography and forearm vascular conductance (FVC) was calculated. In this protocol, neither individual nor combined NO/PG inhibition had any effect on the vasodilatory response (P = 0.22-0.99). In protocol 2 (n = 16), we determined whether any possible contribution of both NO and PGs to ATP vasodilation was greater at low vs. high doses of ATP and whether inhibition during steady-state infusion of the respective dose of ATP impacted the dilation. FBF in this protocol was measured via Doppler ultrasound. In protocol 2, infusion of low (n = 8)- and high-dose (n = 8) ATP for 5 min evoked a significant increase in FVC above baseline (low = 198 ± 24%; high = 706 ± 79%). Infusion of L-NMMA and ketorolac together reduced steady-state FVC during both low- and high-dose ATP (P < 0.05), and in a subsequent trial with continuous NO/PG blockade, the vasodilator response from baseline to 5 min of steady-state infusion was similarly reduced for both low (ΔFVC = -31 ± 11%)- and high-dose ATP (ΔFVC -25 ± 11%; P = 0.70 low vs. high dose). Collectively, our findings indicate a potential modest role for NO and PGs in the vasodilatory response to exogenous ATP in the human forearm that does not appear to be dose or timing dependent; however, this is dependent on the method for assessing forearm vascular responses. Importantly, the majority of ATP-mediated vasodilation is independent of these putative endothelium-dependent pathways in humans.


American Journal of Physiology-heart and Circulatory Physiology | 2013

Mechanisms of rapid vasodilation after a brief contraction in human skeletal muscle

Anne R. Crecelius; Brett S. Kirby; Gary J. Luckasen; Dennis G. Larson; Frank A. Dinenno

A monophasic increase in skeletal muscle blood flow is observed after a brief single forearm contraction in humans, yet the underlying vascular signaling pathways remain largely undetermined. Evidence from experimental animals indicates an obligatory role of vasodilation via K⁺-mediated smooth muscle hyperpolarization, and human data suggest little to no independent role for nitric oxide (NO) or vasodilating prostaglandins (PGs). We tested the hypothesis that K⁺-mediated vascular hyperpolarization underlies the rapid vasodilation in humans and that combined inhibition of NO and PGs would have a minimal effect on this response. We measured forearm blood flow (Doppler ultrasound) and calculated vascular conductance 10 s before and for 30 s after a single 1-s dynamic forearm contraction at 10%, 20%, and 40% maximum voluntary contraction in 16 young adults. To inhibit K⁺-mediated vasodilation, BaCl₂ and ouabain were infused intra-arterially to inhibit inwardly rectifying K⁺ channels and Na⁺-K⁺-ATPase, respectively. Combined enzymatic inhibition of NO and PG synthesis occurred via NG-monomethyl-L-arginine (L-NMMA; NO synthase) and ketorolac (cyclooxygenase), respectively. In protocol 1 (n = 8), BaCl₂ + ouabain reduced peak vasodilation (range: 30-45%, P < 0.05) and total postcontraction vasodilation (area under the curve, ~55-75% from control) at all intensities. Contrary to our hypothesis, L-NMMA + ketorolac had a further impact (peak: ~60% and area under the curve: ~80% from control). In protocol 2 (n = 8), the order of inhibitors was reversed, and the findings were remarkably similar. We conclude that K⁺-mediated hyperpolarization and NO and PGs, in combination, significantly contribute to contraction-induced rapid vasodilation and that inhibition of these signaling pathways nearly abolishes this phenomenon in humans.


The Journal of Physiology | 2012

ATP-mediated Vasodilatation Occurs via Activation of Inwardly-Rectifying Potassium Channels in Humans

Anne R. Crecelius; Brett S. Kirby; Gary J. Luckasen; Dennis G. Larson; Frank A. Dinenno

•  ATP is a substance in the blood vessels that can cause vasodilatation and increase blood flow and oxygen delivery in humans. •  The exact signalling pathways that ATP stimulates to cause vasodilatation are not well known. •  We show that a large portion of ATP‐mediated vasodilatation occurs through the activation of inwardly rectifying potassium channels (Kir). •  Our results lend insight into the vasodilator mechanisms of ATP, a substance that is important for vascular control. •  Further, our results may stimulate additional investigations in humans regarding the activation of Kir channels and subsequent vascular hyperpolarization during other physiologically relevant conditions.


Circulation Research | 2013

Reactive Hyperemia Occurs via Activation of Inwardly-Rectifying Potassium Channels and Na+/K+-ATPase in Humans

Anne R. Crecelius; Jennifer C. Richards; Gary J. Luckasen; Dennis Larson; Frank A. Dinenno

Rationale: Reactive hyperemia (RH) in the forearm circulation is an important marker of cardiovascular health, yet the underlying vasodilator signaling pathways are controversial and thus remain unclear. Objective: We hypothesized that RH occurs via activation of inwardly rectifying potassium (KIR) channels and Na+/K+-ATPase and is largely independent of the combined production of the endothelial autocoids nitric oxide (NO) and prostaglandins in young healthy humans. Methods and Results: In 24 (23±1 years) subjects, we performed RH trials by measuring forearm blood flow (FBF; venous occlusion plethysmography) after 5 minutes of arterial occlusion. In protocol 1, we studied 2 groups of 8 subjects and assessed RH in the following conditions. For group 1, we studied control (saline), KIR channel inhibition (BaCl2), combined inhibition of KIR channels and Na+/K+-ATPase (BaCl2 and ouabain, respectively), and combined inhibition of KIR channels, Na+/K+-ATPase, NO, and prostaglandins (BaCl2, ouabain, L-NMMA [NG-monomethyl-L-arginine] and ketorolac, respectively). Group 2 received ouabain rather than BaCl2 in the second trial. In protocol 2 (n=8), the following 3 RH trials were performed: control; L-NMMA plus ketorolac; and L-NMMA plus ketorolac plus BaCl2 plus ouabain. All infusions were intra-arterial (brachial). Compared with control, BaCl2 significantly reduced peak FBF (−50±6%; P<0.05), whereas ouabain and L-NMMA plus ketorolac did not. Total FBF (area under the curve) was attenuated by BaCl2 (−61±3%) and ouabain (−44±12%) alone, and this effect was enhanced when combined (−87±4%), nearly abolishing RH. L-NMMA plus ketorolac did not impact total RH FBF before or after administration of BaCl2 plus ouabain. Conclusions: Activation of KIR channels is the primary determinant of peak RH, whereas activation of both KIR channels and Na+/K+-ATPase explains nearly all of the total (AUC) RH in humans.


The Journal of Physiology | 2011

Modulation of postjunctional α-adrenergic vasoconstriction during exercise and exogenous ATP infusions in ageing humans

Brett S. Kirby; Anne R. Crecelius; Wyatt F. Voyles; Frank A. Dinenno

Non‐technical summary  When muscles contract, a variety of signals interact and ultimately increase blood flow and oxygen delivery to the active muscle. However, when there is simultaneous activation of the sympathetic nervous system, noradrenaline (norepinephrine) is released which tries to cause vasoconstriction in the same blood vessels that are trying to relax (vasodilate). In young adults, we have shown that there is a unique ability of the contracting muscle to limit this vasoconstriction, and also that a circulating factor called ATP mimics the exercise response. In the present study, we demonstrate that older healthy adults have an impaired ability to limit the sympathetic vasoconstrictor signal during exercise; however, this ability is preserved when we administer ATP. Thus, if this impairment in the ability of contracting muscle to limit sympathetic vasoconstriction in older adults is related to ATP, we speculate that circulating levels of ATP may be impaired during exercise.


American Journal of Physiology-heart and Circulatory Physiology | 2014

KIR channel activation contributes to onset and steady-state exercise hyperemia in humans

Anne R. Crecelius; Gary J. Luckasen; Dennis G. Larson; Frank A. Dinenno

We tested the hypothesis that activation of inwardly rectifying potassium (KIR) channels and Na(+)-K(+)-ATPase, two pathways that lead to hyperpolarization of vascular cells, contributes to both the onset and steady-state hyperemic response to exercise. We also determined whether after inhibiting these pathways nitric oxide (NO) and prostaglandins (PGs) are involved in the hyperemic response. Forearm blood flow (FBF; Doppler ultrasound) was determined during rhythmic handgrip exercise at 10% maximal voluntary contraction for 5 min in the following conditions: control [saline; trial 1 (T1)]; with combined inhibition of KIR channels and Na(+)-K(+)-ATPase alone [via barium chloride (BaCl2) and ouabain, respectively; trial 2 (T2)]; and with additional combined nitric oxide synthase (N(G)-monomethyl-l-arginine) and cyclooxygenase inhibition [ketorolac; trial 3 (T3)]. In T2, the total hyperemic responses were attenuated ~50% from control (P < 0.05) at exercise onset, and there was minimal further effect in T3 (protocol 1; n = 11). In protocol 2 (n = 8), steady-state FBF was significantly reduced during T2 vs. T1 (133 ± 15 vs. 167 ± 17 ml/min; Δ from control: -20 ± 3%; P < 0.05) and further reduced during T3 (120 ± 15 ml/min; -29 ± 3%; P < 0.05 vs. T2). In protocol 3 (n = 8), BaCl2 alone reduced FBF during onset (~50%) and steady-state exercise (~30%) as observed in protocols 1 and 2, respectively, and addition of ouabain had no further impact. Our data implicate activation of KIR channels as a novel contributing pathway to exercise hyperemia in humans.

Collaboration


Dive into the Anne R. Crecelius's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Brett S. Kirby

Colorado State University

View shared research outputs
Top Co-Authors

Avatar

Dennis G. Larson

University of Colorado Hospital

View shared research outputs
Top Co-Authors

Avatar

Gary J. Luckasen

University of Colorado Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Matthew Racine

Colorado State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge