local control of skeletal muscle blood flow during exercise: … · 2013-04-14 · exercise with...

13
doi: 10.1152/japplphysiol.00895.2011 111:1527-1538, 2011. First published 1 September 2011; J Appl Physiol Darren P. Casey and Michael J. Joyner exercise: influence of available oxygen Local control of skeletal muscle blood flow during You might find this additional info useful... 165 articles, 132 of which you can access for free at: This article cites http://jap.physiology.org/content/111/6/1527.full#ref-list-1 3 other HighWire-hosted articles: This article has been cited by http://jap.physiology.org/content/111/6/1527#cited-by including high resolution figures, can be found at: Updated information and services http://jap.physiology.org/content/111/6/1527.full can be found at: Journal of Applied Physiology about Additional material and information http://www.the-aps.org/publications/jappl This information is current as of April 7, 2013. http://www.the-aps.org/. © 2011 the American Physiological Society. ISSN: 8750-7587, ESSN: 1522-1601. Visit our website at year (monthly) by the American Physiological Society, 9650 Rockville Pike, Bethesda MD 20814-3991. Copyright physiology, especially those papers emphasizing adaptive and integrative mechanisms. It is published 12 times a publishes original papers that deal with diverse area of research in applied Journal of Applied Physiology by guest on April 7, 2013 http://jap.physiology.org/ Downloaded from

Upload: others

Post on 06-Jul-2020

3 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Local control of skeletal muscle blood flow during exercise: … · 2013-04-14 · exercise with normoxia, anemia, hypoxia, and anemia hyp-oxia were dependent on Ca O 2 not Pa O

doi: 10.1152/japplphysiol.00895.2011111:1527-1538, 2011. First published 1 September 2011;J Appl Physiol 

Darren P. Casey and Michael J. Joynerexercise: influence of available oxygenLocal control of skeletal muscle blood flow during

You might find this additional info useful...

 165 articles, 132 of which you can access for free at: This article citeshttp://jap.physiology.org/content/111/6/1527.full#ref-list-1

 3 other HighWire-hosted articles: This article has been cited by http://jap.physiology.org/content/111/6/1527#cited-by

including high resolution figures, can be found at: Updated information and serviceshttp://jap.physiology.org/content/111/6/1527.full

can be found at: Journal of Applied Physiology about Additional material and informationhttp://www.the-aps.org/publications/jappl

This information is current as of April 7, 2013.

http://www.the-aps.org/. © 2011 the American Physiological Society. ISSN: 8750-7587, ESSN: 1522-1601. Visit our website at year (monthly) by the American Physiological Society, 9650 Rockville Pike, Bethesda MD 20814-3991. Copyrightphysiology, especially those papers emphasizing adaptive and integrative mechanisms. It is published 12 times a

publishes original papers that deal with diverse area of research in appliedJournal of Applied Physiology

by guest on April 7, 2013

http://jap.physiology.org/D

ownloaded from

Page 2: Local control of skeletal muscle blood flow during exercise: … · 2013-04-14 · exercise with normoxia, anemia, hypoxia, and anemia hyp-oxia were dependent on Ca O 2 not Pa O

Local control of skeletal muscle blood flow during exercise: influenceof available oxygen

Darren P. Casey1,2 and Michael J. Joyner1

Departments of 1Anesthesiology and 2Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota

Submitted 18 July 2011; accepted in final form 31 August 2011

Casey DP, Joyner MJ. Local control of skeletal muscle blood flow duringexercise: influence of available oxygen. J Appl Physiol 111: 1527–1538, 2011. Firstpublished September 1, 2011; doi:10.1152/japplphysiol.00895.2011.—Reductionsin oxygen availability (O2) by either reduced arterial O2 content or reducedperfusion pressure can have profound influences on the circulation, includingvasodilation in skeletal muscle vascular beds. The purpose of this review is to putinto context the present evidence regarding mechanisms responsible for the localcontrol of blood flow during acute systemic hypoxia and/or local hypoperfusion incontracting muscle. The combination of submaximal exercise and hypoxia pro-duces a “compensatory” vasodilation and augmented blood flow in contractingmuscles relative to the same level of exercise under normoxic conditions. A similarcompensatory vasodilation is observed in response to local reductions in oxygenavailability (i.e., hypoperfusion) during normoxic exercise. Available evidence sug-gests that nitric oxide (NO) contributes to the compensatory dilator response undereach of these conditions, whereas adenosine appears to only play a role duringhypoperfusion. During systemic hypoxia the NO-mediated component of thecompensatory vasodilation is regulated through a �-adrenergic receptor mechanismat low-intensity exercise, while an additional (not yet identified) source of NO islikely to be engaged as exercise intensity increases during hypoxia. Potentialcandidates for stimulating and/or interacting with NO at higher exercise intensitiesinclude prostaglandins and/or ATP. Conversely, prostaglandins do not appear toplay a role in the compensatory vasodilation during exercise with hypoperfusion.Taken together, the data for both hypoxia and hypoperfusion suggest NO isimportant in the compensatory vasodilation seen when oxygen availability islimited. This is important from a basic biological perspective and also haspathophysiological implications for diseases associated with either hypoxia orhypoperfusion.

hypoxia; hypoperfusion; vasodilation

DURING DYNAMIC MUSCLE CONTRACTIONS there is an increasedmetabolic demand that is matched closely by increases inskeletal muscle blood flow (exercise hyperemia) and oxygendelivery. Blood flow increases rapidly at the onset of contrac-tions and can increase up to 100-fold over resting values duringintense exercise (2). The mechanisms responsible for increas-ing blood flow at the onset of exercise as well as maintainingit over time involve a complex interaction between mechanicalfactors and various local metabolic and endothelial derivedsubstances that influence vascular tone (132). Over the lastseveral decades exercise hyperemia in general and the potentialmechanisms responsible for the regulation of flow to contract-ing skeletal muscles have been reviewed extensively (13, 33,35, 68, 75, 80, 81, 131, 152). These reviews have focusedprimarily on blood flow responses during exercise under nor-moxic conditions. However, reductions in oxygen (O2) avail-ability (i.e., hypoxia) bring about several cardiovascular ad-

justments and can have profound influences on the circulation.Along these lines, the mechanisms responsible for the localregulation of skeletal muscle blood flow during exercise arelikely enhanced and possibly different than those during nor-moxic exercise.

Therefore, the purpose of this review is to discuss how O2

availability can impact blood flow and vasodilation in con-tracting skeletal muscles. Although cardiac pumping capac-ity (especially during large muscle mass and/or high inten-sity exercise) can be important in the hyperemic response toexercise, the emphasis in this review is on the local mech-anisms regulating active muscle blood flow under conditionsof reduced O2 availability during submaximal exercise.Specifically, this review will focus on 1) how acute hypoxiaand hypoperfusion interact with exercise to govern thehyperemic responses in contracting muscles of humans and2) the local vasodilator signals involved in the regulation ofmuscle blood flow during acute systemic (hypoxia) andlocal (hypoperfusion) reductions in O2 availability. We willalso discuss the potential role of any systemic pressorresponse under these conditions.

Address for reprint requests and other correspondence: D. P. Casey, Dept. ofAnesthesiology, Mayo Clinic, 200 First St. SW, Rochester, MN 55905 (e-mail:[email protected]).

J Appl Physiol 111: 1527–1538, 2011.First published September 1, 2011; doi:10.1152/japplphysiol.00895.2011. Review

8750-7587/11 Copyright © 2011 the American Physiological Societyhttp://www.jap.org 1527

by guest on April 7, 2013

http://jap.physiology.org/D

ownloaded from

Page 3: Local control of skeletal muscle blood flow during exercise: … · 2013-04-14 · exercise with normoxia, anemia, hypoxia, and anemia hyp-oxia were dependent on Ca O 2 not Pa O

SYSTEMIC HYPOXIA

Effect on Blood Flow to Resting Muscle

At rest acute exposure to moderate hypoxia elicits vasodi-lation and an augmented blood flow in skeletal muscle vascularbeds of young apparently healthy humans (9–11, 25, 26, 37,84, 124, 158, 160, 161). This vasodilation occurs despite theresting muscle being relatively overperfused, as evidenced bylimited O2 extraction (25, 26, 59). The augmented skeletalmuscle vasodilation in response to hypoxia also occurs despitean enhanced sympathetic vasoconstrictor activity (52, 83, 130,143). Evidence suggests that a reduced �-adrenergic respon-siveness associated with hypoxia is not responsible for thisdilation (37, 161). Local blockade of �-adrenergic receptors inthe forearm reveals a substantially greater vasodilator responseduring systemic hypoxia (158, 160). Thus activation of sym-pathetic vasoconstrictor nerves during hypoxia likely masksthe degree of vasodilation. It should be noted that other studieshave reported that hypoxia-mediated forearm vascular conduc-tance was unaffected by local blockade of sympathetic nerves(9) and �-adrenergic blockade (118). Taken together, vasodi-lation prevails over the vasoconstrictor response in determiningvasomotor tone during acute hypoxia in resting skeletal mus-cle.

A number of vasodilator substances or systems, includingbut not limited to �-adrenergic receptor activation, nitricoxide (NO), prostaglandins, and adenosine, have been im-plicated to be involved in hypoxic vasodilation at rest andhave been previously reviewed in detail (51). Briefly, weand others (10, 158, 160) have demonstrated that �-adren-ergic receptor activation is responsible for a substantialportion of the hypoxic vasodilation at rest. Moreover, alarge portion of the �-adrenergic receptor-mediated hypoxicvasodilation at rest occurs through a NO pathway (158).This is consistent with the NO-dependent nature of at leastsome of the vasodilator responses to �2-mediated vasodila-tion (31, 45). Along these lines, NO synthase (NOS) inhi-bition attenuates the hypoxic vasodilator response in thehuman forearm in some studies (11, 25) but not others (90).In the latter study, the investigators locally blocked �-ad-renergic receptors via intra-arterial infusions of propranololprior to the experimental trials with NOS inhibition andtherefore potentially masked the NO-mediated vasodilatorresponse that is observed in other studies without concurrent�-adrenergic blockade. Under similar conditions, singleinhibition of prostaglandin synthesis also did not attenuatethe vasodilator response to systemic hypoxia (90). However,the combined inhibition of NO and prostaglandins abolished thevasodilator response to hypoxia at rest, thus indicating asynergistic role between these two pathways in regulatingperipheral vascular tone and ultimately blood flow (90). Recentevidence suggests both interstitial and plasma adenosine stim-ulates NO and prostacyclin formation in the human leg undernormoxic conditions (103). Moreover, there is strong evidencein animals that adenosine plays a major role in hypoxia-induced skeletal muscle vasodilation (15, 100, 114, 115, 141).However, available data related to the contribution of adeno-sine in the hypoxic vasodilator response in human studies areconflicting (25, 26, 84).

Hypoxic Exercise

The combination of submaximal dynamic exercise and hyp-oxia produces a “compensatory” vasodilation and augmentedblood flow in contracting muscles (both quadriceps and fore-arm) relative to the same level of exercise under normoxicconditions (16, 26, 32, 122, 126, 160, 161). This augmentedvasodilation exceeds that predicted by a simple sum of theindividual dilator responses to hypoxia alone and normoxicexercise. Additionally, this enhanced hypoxic exercise hyper-emia is proportional to the hypoxia-induced fall in arterial O2

content, thus preserving muscle O2 delivery and ensuring it ismatched to demand (32, 49, 122, 126). Compensatory vaso-dilation is also observed when O2 availability is reduced bycarbon monoxide administration and/or experimentally in-duced anemia (48, 49, 121, 122). In this context, O2 deliv-ery, rather than blood flow, appears to be the regulatedvariable producing the augmented hyperemia during hy-poxic exercise (47).

Potential Causes of Compensatory Vasodilation

Acute exposure to systemic hypoxia results in substantialreductions in arterial O2 content (CaO2

) as well as arterial O2

tension (PaO2). Theoretically, reductions in each of these vari-

ables could serve as a signal for the compensatory vasodilationobserved during hypoxic exercise (77, 78, 122). Roach andcolleagues (122) reported that blood flow and O2 delivery tothe lower limbs during rhythmic two-legged knee extensionexercise with normoxia, anemia, hypoxia, and anemia � hyp-oxia were dependent on CaO2

not PaO2. Evidence that a low

PaO2alone does not cause vasodilation was based on the

observation of similar limb blood flows in the two conditions(hypoxia and anemia) with nearly identical CaO2

but widelydifferent PaO2

(�40 vs. �105 mmHg). In a subsequent study,leg blood flow and vascular conductance were shown to betightly coupled to the level of arterial oxyhemoglobin, regard-less of a normal or an 11-fold difference in PaO2

(47–538mmHg) (49). Taken together, these results strongly suggestthat factors sensitive to CaO2

serve as the signal for thecompensatory increases in skeletal muscle blood flow andvasodilation during hypoxic exercise.

Regulation of the Vascular Tone During Hypoxic Exercise

Vasodilator line-up: usual suspects? Several vasodilatorpathways have been proposed and examined as likely regula-tors of skeletal muscle blood flow in response to changes inCaO2

. Increases in arterial epinephrine are evident with acuteexposure to systemic hypoxia and are increased further duringsubmaximal cycling exercise (92). Additionally, an intensity-dependent rise in arterial epinephrine occurs during incremen-tal forearm exercise, suggesting that �-adrenergic receptor-mediated vasodilation might contribute to the augmented hy-poxic exercise hyperemia (161). Wolfel and colleagues (163)originally demonstrated that �-adrenergic blockade reduced O2

delivery to the contracting muscles during submaximal exer-cise at altitude, but was compensated for by an enhanced tissueO2 extraction. However, the use of systemic �-blockade re-duced cardiac output and likely engaged vasoconstricting car-diovascular reflexes, therefore making it difficult to determinewhether alterations in �-adrenergic receptor-mediated vasodi-

Review

1528 OXYGEN AVAILABILITY AND COMPENSATORY VASODILATION

J Appl Physiol • VOL 111 • DECEMBER 2011 • www.jap.org

by guest on April 7, 2013

http://jap.physiology.org/D

ownloaded from

Page 4: Local control of skeletal muscle blood flow during exercise: … · 2013-04-14 · exercise with normoxia, anemia, hypoxia, and anemia hyp-oxia were dependent on Ca O 2 not Pa O

lation within skeletal muscle vasculature contributed to thereduced O2 delivery. Along these lines, local inhibition of�-adrenergic receptors (via intra-arterial infusion of proprano-lol) has demonstrated that in the absence of overlying sympa-thetic vasoconstriction, �-adrenergic receptor activation con-tributes to the hypoxic compensatory vasodilation during mild(10% maximum voluntary contraction; MVC) forearm exercise(160). However, the �-adrenergic component decreases withincreased exercise intensity (20% MVC). Taken together, thissuggests that other local vasodilating factors are likely respon-sible for the compensatory vasodilation during hypoxic exer-cise, especially at higher exercise intensities.

NO release from an endothelial source and/or desaturation ofhemoglobin in erythrocytes (85, 109, 140, 147) has beenproposed during hypoxia. Along these lines, there is a signif-icant blunting of the compensatory vasodilation in the forearmduring mild to moderate (10–20% MVC) hypoxic exerciseafter NOS inhibition (25). Although NO-mediated mechanismscontribute to the compensatory vasodilation during hypoxicexercise, it appears that it is regulated through different path-ways with increasing exercise intensity. At lower-intensityforearm exercise (10% MVC) NO contributes to the compen-satory vasodilation via �-adrenergic receptor activation. Athigher intensity exercise the contribution of NO becomesindependent of the �-adrenergic receptors (19). Therefore, analternative and currently unknown stimulus for NO-mediatedvasodilation occurs during higher intensity hypoxic exercise.

Increases in plasma but not skeletal muscle interstitial NOduring hypoxia in humans suggest an endovascular or endo-thelial NO source (85). Therefore, it is possible that theaugmented vasodilation during hypoxic exercise is a result ofthe direct release of NO from endothelial cells and/or erythro-cytes. In this context, luminal hypoxia can elicit the directrelease of NO from the endothelium (109) and from erythro-cytes in the form S-nitrosohemoglobin (147). However, basedon the location for intraluminal NG-monomethyl-L-arginine(NOS inhibitor) administration and the fact that NOS inhibitionis effective in reducing forearm blood flow during hypoxicexercise (25), it is reasonable to suspect that the NO respon-sible for the compensatory vasodilatation is from endothelialsources vs. an erythrocyte source. Furthermore, the effective-ness of NOS inhibition in reducing the compensatory vasodi-latation during hypoxic exercise also argues against the ideathat nitrite and/or formation of erythrocyte nitroso species is akey vasodilator in this response (39, 46). Along these lines, ifnitrite were responsible, either directly or indirectly (via reduc-tion to NO), the compensatory vasodilation would have beenmaintained despite NOS inhibition.

In animals adenosine is thought to be a key mediator ofskeletal muscle blood flow during normoxic exercise (95, 107,110, 111). However, adenosine receptor blockade appears toonly modestly attenuate the exercise-induced skeletal musclevasodilation in human limbs, but these findings are not con-sistent (26, 91, 98, 112). The classic adenosine hypothesisproposes that adenosine mediates vasodilation and helps toensure adequate blood flow to metabolically active tissueduring periods of reduced O2 availability (i.e., hypoxia and/orischemia) (7). This concept would therefore suggest that aden-osine may contribute to the compensatory vasodilation duringhypoxic exercise. However, our group demonstrated that aden-osine is not obligatory for the compensatory vasodilation

during hypoxic exercise in humans (26). This was observedwith and without overlying sympathetic �-adrenergic vasocon-striction, suggesting that the enhanced sympathetic outflowassociated with systemic hypoxia did not mask any underlyingadenosine-mediated vasodilation. Moreover, adenosine doesnot contribute to the compensatory vasodilation after NOSinhibition, thus indicating that adenosine does not act throughan NO-independent pathway in this response (25). Usingpositron emission tomography (PET) imaging, Heinonen andcolleagues (59) recently demonstrated that adenosine receptorantagonism (via aminophylline) did not affect blood flow inexercising human quadriceps femoris muscle, suggesting nocontribution of adenosine to capillary blood flow during hy-poxic exercise. Additionally, adenosine receptor blockade didnot change blood flow heterogeneity within the active andinactive muscles during hypoxic exercise.

Deoxygenation (i.e., hypoxia) and mechanical deformation(i.e., exercise) of red blood cells can lead to ATP release fromerythrocytes (6, 64, 144, 145). Moreover, venous plasma levelsof ATP are elevated during hypoxia at rest and during exercisecompared with normoxic conditions (48, 99). Both in vitro andin vivo studies suggest that ATP-induced vasodilation is me-diated through a NO pathway (28, 94, 96), thus an ATP/NO-mediated component to the compensatory vasodilation duringhypoxic exercise is an attractive hypothesis. It is believed thatthe vasodilator actions of ATP are mediated through thepurinergic P2y receptors located on vascular endothelial cells(113). Unfortunately, specific pharmacological antagonists forP2 receptors to address the role of ATP in the hypoxia-inducedcompensatory vasodilation are currently unavailable for humanuse. However, it is important to note that experimentallyincreasing limb vasodilation during hypoxic exercise (via intra-arterial ATP infusion) does not improve leg and systemic O2

consumption due to a reduction in O2 extraction across theexercising leg (17). Another potential candidate for stimulatingand/or interacting with NO in the compensatory vasodilatorresponse might be prostaglandins. There is strong evidence thatsuggests an interaction between prostaglandins and NO in theregulation of skeletal muscle blood flow at rest and duringnormoxic exercise (97, 101, 133, 134). As indicated earlier,there is also evidence to suggest a synergistic role betweenthese two pathways in the compensatory vasodilation duringhypoxia at rest (90). Recent evidence suggests that combinedNO/PG inhibition substantially reduces forearm blood flow andvasodilation during hypoxic exercise in young healthy adults(29). It is important to note that the aforementioned study didnot include single inhibition trials of each vasodilator pathwayof interest (NO and prostaglandins). Therefore, it is difficult todiscern the relative contribution of PGs and its interaction withNO in the compensatory vasodilation during hypoxic exercise.

Sympathetic Restraint of Hypoxia-InducedCompensatory Vasodilation

In young healthy humans the sympathetic response to exer-cise is greater in hypoxia than it is in normoxia (54, 69, 136).The increased sympathetic vasoconstrictor activity has beenpostulated to be needed to match O2 delivery with O2 demand(88) and maintain arterial blood pressure during the hypoxicstress and in turn limit the degree of compensatory vasodilation(148). Along these lines, �-adrenergic receptor blockade in the

Review

1529OXYGEN AVAILABILITY AND COMPENSATORY VASODILATION

J Appl Physiol • VOL 111 • DECEMBER 2011 • www.jap.org

by guest on April 7, 2013

http://jap.physiology.org/D

ownloaded from

Page 5: Local control of skeletal muscle blood flow during exercise: … · 2013-04-14 · exercise with normoxia, anemia, hypoxia, and anemia hyp-oxia were dependent on Ca O 2 not Pa O

human forearm (160) and in the hindlimbs of dogs (148)reveals a greater vasodilation during hypoxic exercise com-pared with control hypoxic exercise conditions (i.e., duringsaline infusion). Despite the augmented sympathetic vasoconstric-tor activity a substantial compensatory vasodilation persistsduring hypoxic exercise. It is well documented that sympa-thetic vasoconstrictor responses are blunted in the vascularbeds of contracting skeletal muscle of animals (150, 151) andhumans (36, 56, 57, 117, 153), a phenomenon referred to as“functional sympatholysis.” Moreover, there is evidence tosuggest that hypoxia can attenuate vasoconstrictor responses tosympathetic nerve activation and exogenous norepinephrine inresting skeletal muscle of animals and humans (12, 60, 61).Therefore, it is possible that an augmented functional sympa-tholysis might partially explain the compensatory vasodilationduring hypoxic exercise. In this context, Hansen et al. (55)demonstrated a reduced forearm vasoconstrictor responsive-ness to lower body negative pressure during moderate hypoxicexercise compared with normoxic exercise. However, our lab-oratory (161) has demonstrated that the greater vasodilationobserved during hypoxic forearm exercise was not due to areduced postjunctional vasoconstrictor responsiveness (aug-mented functional sympatholysis) to endogenous norepineph-rine (via intra-arterial infusion of tyramine).

Compensatory Vasodilation: Is it Always Present?

It is clear that compensatory vasodilation occurs in responseto acute hypoxia during submaximal forearm (19, 25, 26, 29,160, 161) and single and/or two-legged knee extension (16, 32,77, 122, 126) exercise. That is, during submaximal exercise,alterations in O2 availability are compensated for by an aug-mented muscle blood flow to help maintain O2 delivery to theactive muscles. However, this compensatory vasodilation ap-pears to be absent during maximal exercise, especially whenperformed with a large muscle mass (74, 77, 78, 120). Themuscle blood flow and vasodilator response to maximal exer-cise during moderate and severe hypoxia are likely limited toa certain degree by a reduction in maximal cardiac output (16)and not fully capable of compensating when the maximalpumping capacity of the heart is taxed. Under these circum-stances it seems likely baroreflex-mediated vasoconstrictorresponses are engaged. However, it is important to note thateven in well trained cyclists who have a high cardiovascularreserve, leg blood flow and leg O2 consumption is reduced atpeak exercise using the one-legged knee extension model(119). The lack of a compensatory vasodilation is also ob-served during submaximal exercise after acclimatization (long-term exposure to hypoxia), which may be due to an increasein CaO2

.In summary, the studies discussed here clearly demonstrate

that reductions in available O2 via systemic hypoxia promotecompensatory vasodilation and an augmented blood flow inskeletal muscle vascular beds at rest and during submaximalexercise. The compensatory vasodilation during hypoxic exer-cise is essential to ensure the maintenance of oxygen deliveryto the active muscles. It appears that factors sensitive to CaO2

serve as the signal for the compensatory increases in skeletalmuscle blood flow and vasodilation during hypoxic exercise. Inhumans, NO-mediated vasodilation plays an obligatory role inthe augmented blood flow during incremental hypoxic exer-

cise. However, the NO-mediated component of the compensa-tory vasodilation during hypoxic exercise is regulated throughdifferent pathways with increasing exercise intensity. That is, a�-adrenergic receptor-stimulated NO component exists duringlow-intensity hypoxic exercise, whereas the source of NOcontributing to compensatory dilation is less dependent on�-adrenergic mechanisms as exercise intensity increases. It iscurrently unclear what the stimulus of NO release is withincreasing intensity of muscle contraction but does not appearto be adenosine. ATP released from erythrocytes and/or endo-thelial-derived prostaglandins remain attractive candidates forstimulating NO during higher intensity hypoxic exercise.

The vasodilator response to acute hypoxia detailed above inyoung healthy humans has been demonstrated to be attenuatedin older adults at rest and during exercise (27, 79). Addition-ally, limited reports suggest that compensatory vasodilation isimpaired in some populations with cardiovascular risk factors(5), whereas it is preserved in others (86, 157). From a clinicalperspective, if an attenuated vasodilator response occurs inother vascular beds (i.e., coronary circulation) with aging andcardiovascular risk factors, certain individuals may be at risk ofreduced perfusion of the myocardium and consequently myo-cardial injury during exercise with hypoxia.

HYPEROXIA: THE OPPOSITE SIDE OF THE COIN

It is apparent that compromises in O2 availability (i.e.,hypoxia) elicit a compensatory vasodilator response in skeletalmuscle vascular beds to ensure adequate O2 delivery. Con-versely, increased O2 availability (i.e., hyperoxia) reducesresting muscle blood flow in humans (8, 58, 116, 165). Addi-tionally, muscle blood flow is reduced during and after hyper-oxic exercise compared with normoxic conditions (24, 48, 116,159). The challenge of increasing O2 availability by increasingthe fraction of inspired O2 (i.e., FIO2

� 100% O2) undernormobaric environments is that CaO2

can only be elevated5–10%. Additional approaches using hyperoxia (FIO2

� 100%O2) superimposed onto experimentally induced polycythemiahave also been employed to study the effects of increased O2

availability on muscle blood flow during knee-extensor exer-cise (47). However, this approach only resulted in an �11%increase in CaO2

. These relatively small increases in CaO2are

near the signal-to-noise ratio for muscle blood flow measure-ments. To address this potential limitation, studies on the bloodflow response in the exercising forearm to larger increases inestimated CaO2

via hyperbaric hyperoxia have recently beenperformed (24). Large increases in estimated O2 content (�25–30%) resulted in a substantial reduction in exercising forearmblood flow despite significantly higher perfusion pressures,thus indicating a greater increase in vascular resistance com-pared with normoxic conditions (24). The blunted forearmvascular conductance during hyperoxic exercise (�25% lower)mirrored the estimated increase in arterial O2 content (�25–30%). These findings during hyperbaric hyperoxic exerciseparallel previous findings in which hypoxic-mediated increasesin forearm vascular conductance during exercise mirrors thefall in O2 content (25, 26).

The mechanisms responsible for the increased vascular re-sistance and reduced blood flow during hyperoxic exercise arecurrently unclear. In general, the potential mechanisms includebut are not limited to 1) greater sympathetic restraint and/or

Review

1530 OXYGEN AVAILABILITY AND COMPENSATORY VASODILATION

J Appl Physiol • VOL 111 • DECEMBER 2011 • www.jap.org

by guest on April 7, 2013

http://jap.physiology.org/D

ownloaded from

Page 6: Local control of skeletal muscle blood flow during exercise: … · 2013-04-14 · exercise with normoxia, anemia, hypoxia, and anemia hyp-oxia were dependent on Ca O 2 not Pa O

2) decreased release of or responsiveness to local vasodilatorswithin the contracting muscle. Although hyperoxia reducesmuscle sympathetic nerve activity at rest (62, 135, 165), itis unclear whether this holds true during exercise and contrib-utes to the reduced muscle blood flow and vasodilation. Lim-ited evidence suggests that hyperoxia does not appear toenhance the magnitude of change in sympathetic nerve activityto non-active skeletal muscle during dynamic exercise (135).Additionally, it is possible that increased oxygen levels mayattenuate functional sympatholysis and therefore enhance va-soconstriction in the contracting muscles (55). Finally, someevidence suggests that the vascular response (i.e., constriction)to hyperoxia in the hindlimbs of dogs is not mediated by theautonomic nervous system but is rather attributed to a directaction of high arterial O2 tension (4). Therefore, it is possiblethat the increase in vascular resistance under hyperoxic exer-cise conditions may be a direct vasoconstrictor action of O2 onthe arterial and arteriolar wall. As discussed earlier, erythro-cytes have the ability to sense changes in O2 during hypoxicconditions and modulate vascular tone via release of ATP, thusleading to appropriate changes in blood flow and matching O2

delivery with metabolic need (42). Therefore, large increases inO2 availability during hyperbaric hyperoxic exercise couldtheoretically attenuate the release of ATP from erythrocytes(Fig. 1) (48).

A shift in the vasodilator/vasoconstrictor balance due to anattenuated release of or responsiveness to local vasodilatorswithin the contracting muscle could also promote the reducedblood flow during hyperoxic exercise. In this context, prosta-glandin-mediated vasodilation following isometric exercise isreduced during hyperoxia (162). Additionally, vasoconstrictionvia a reduction in basal NO production has been observed in

porcine coronary arteries exposed to elevated levels of O2

(106). Along the same lines, an increase in free radical pro-duction (i.e., superoxide anions) during hyperbaric oxygen-ation causes vasoconstriction in the cerebral circulation of ratsvia inactivation of NO (167). However, recent evidence sug-gests that NO metabolites are unaffected in humans duringnormobaric hyperoxic exercise (39). Therefore it is unclearwhether alterations in the local vasodilator milieu are respon-sible for the blunted blood flow response to hyperoxic exercise.

LOCAL REDUCTIONS IN OXYGEN (VIA HYPOPERFUSION)

Over the past several years our laboratory has been inter-ested in addressing whether the compensatory vasodilator sig-nals that maintain oxygen delivery to active muscles duringexercise with systemic hypoxia are the same or different thanthose that cause compensatory vasodilation during normoxicexercise with hypoperfusion. During conditions of insufficientoxygen delivery to the active muscle (e.g., hypoperfusion orischemia), metabolites can accumulate that either stimulateblood pressure, raising sensory nerves within the muscle, orevoke vasodilation (1, 30, 67, 70, 137, 155, 156). Either ofthese mechanisms might act to restore blood flow to thecontracting muscle and relieve the flow/metabolism mismatch.

Reflex Pressor Response

Activation of chemosensitive afferent nerves in the activemuscle evokes reflex increases in sympathetic outflow andarterial pressure, termed the muscle chemoreflex or metabore-flex (1, 30, 67, 70, 155, 156). The increased pressure is thoughtto augment blood flow to the contracting muscle and partiallyrestore the flow/metabolism matching. Evidence for restorationof blood flow to active muscle via a reflex pressor responsecomes primarily from studies in conscious animals. In dogs,the hemodynamic consequences of muscle chemoreflex acti-vation, via graded reductions in flow and perfusion pressure,have been studied extensively using the “Seattle model” (53,104, 105, 138, 139, 164). In this model, a terminal aorticoccluder in combination with a flow probe in close proximityis used to suddenly reduce blood flow to the hindlimb musclesof dogs running on a treadmill. Using this model, imposedreductions in blood flow and oxygen delivery to activehindlimb muscles during dynamic exercise can evoke a pow-erful pressor response that restores �50% of the flow deficit(105). The reflex pressor response to acute hypoperfusionduring exercise in dogs is dependent on the intensity of theexercise (123) and apparently generated when O2 delivery fallsbelow some critical level causing accumulation of a pressorsubstance (Fig. 2) (139). The reflex pressor response in normaldogs is generated primarily by a rise in cardiac output (3, 53,72, 104, 164), whereas in dogs with congestive heart failure thesystemic pressor response is caused primarily by peripheralvasoconstriction (53). Taken together, the Seattle model clearlydemonstrates that the reflex pressor response contributes to therestoration of blood flow in the hindlimbs of dogs duringexercise. However, it should be noted that Britton et al. (14)demonstrated that autoregulation in the hindlimbs of dogs alsocontributes significantly to the regulation of blood flow inresponse to acute hypoperfusion during exercise.

Fig. 1. Thigh plasma venous ATP responses at rest and during incrementalknee-extensor exercise with exposure to normoxia, hypoxia, hyperoxia, andcarbon monoxide (CO) � normoxia. *Significantly different from restingvalues (P � 0.05). [Adapted with permission from Ref. 48].

Review

1531OXYGEN AVAILABILITY AND COMPENSATORY VASODILATION

J Appl Physiol • VOL 111 • DECEMBER 2011 • www.jap.org

by guest on April 7, 2013

http://jap.physiology.org/D

ownloaded from

Page 7: Local control of skeletal muscle blood flow during exercise: … · 2013-04-14 · exercise with normoxia, anemia, hypoxia, and anemia hyp-oxia were dependent on Ca O 2 not Pa O

Is the Pressor Response Apparent in Humans?

In humans, large increases in blood pressure during ischemicexercise were first described by Alam and Smirk (1). Theseinvestigators observed that the activity of even a small group ofmuscles may give rise to a substantial increase in systolic bloodpressure (up to 70 mmHg) provided that the circulation of theblood through the exercising muscles was arrested completely.Lorentsen (87) later demonstrated that the blood pressureresponse to plantar flexion in patients with unilateral intermit-tent claudication due to atherosclerosis obliterans was signifi-cantly greater during exercise with the diseased limbs com-pared with exercise with nondiseased limbs. Taken together,these studies suggest that the reflex pressor response in humansis engaged during exercise with ischemia and/or hypoperfu-sion. However, stimulation of the muscle chemoreflex evokesa large increase in vasoconstricting sympathetic nerve trafficdirected toward the skeletal muscle (30, 89, 154). While thereflex increase in arterial pressure has the potential to restoreblood flow and O2 delivery to the active muscles, the reflexincrease in vasoconstricting sympathetic outflow might alsorestrain blood flow to some degree. Whether the pressorresponse improves blood flow to the hypoperfused activehuman skeletal muscle has not been extensively studied (rela-tive to that in animals) and is not well understood.

To evaluate the blood flow response to hypoperfusion incontracting muscles the majority of studies in humans haverelied on the use of external pressure to reduce limb blood flowduring exercise, which have produced conflicting results (30,66, 127, 149). Using O2 saturation of venous blood (%SvO2

) asan indirect method of measuring blood flow, studies in theforearm reported that the reflex pressor response did not appearto restore blood flow to rhythmically contracting muscles (66).In contrast, Rowell et al. (127) concluded that the reflex pressorresponse partially restored the decrease in leg blood flow(%SvO2

) induced by external positive pressure. Using a more

direct measurement of flow (brachial artery mean blood veloc-ity), Daley et al. (30) demonstrated that flow is not restored bya reflex pressor response at higher levels of external pressure(�50 mmHg) due to sympathetic vasoconstriction limiting theability of the pressor response to restore the flow. Potentialproblems of these earlier studies mainly revolve around theindirect measures of blood flow (66, 127) and the method usedto reduce limb blood flow (positive pressure) during exercise(30, 66, 127, 149). While external positive pressure clearlyreduces perfusion pressure to the contracting muscles, it differsfundamentally with the Seattle model because it affects allelements of the vascular tree, including the microcirculationand veins, and might either limit expression of local vasodila-tation or limit the ability of changes in arterial pressure toimprove blood flow.

To address the limitations of external pressure, our labora-tory recently developed an experimental model in humans todirectly measure the blood flow response in hypoperfusedskeletal muscle produced by forearm exercise in the presenceof a brachial artery occlusion. For this purpose a small ballooncatheter is inserted into the brachial artery and inflated toreduce forearm blood flow and vascular conductance duringhandgrip exercise (Fig. 3) (23). With this experimental modelthere is a substantial restoration of flow to hypoperfusedexercising human muscle. Interestingly, the restoration of flowoccurs in the absence of a significant pressor response, thussuggesting local vasodilator and/or myogenic mechanisms arelikely responsible. The absence of a significant pressor re-sponse (above exercise alone) during hypoperfusion suggeststhat a reflex increase in pressure is not necessary to restoreblood flow to hypoperfused contracting human muscle. Thelack of a pressor response in our model may be explained bythe small muscle mass (forearm), moderate exercise intensity(20% MVC), magnitude of the reduction in forearm blood flow(�50%) induced by balloon inflation, or a combination ofthese being not sufficient enough to augment the pressorresponse beyond that of exercise alone. However, recent evi-dence suggests that using an upper arm cuff to reduce flow by�40% during forearm exercise at 15% MVC was sufficient toevoke a reflex pressor response (63). It should be noted that theuse of an upper arm cuff likely blocked venous outflow,leading to increased intramuscular pressure, which could acti-vate sympathoexcitatory mechanosensitive afferents (93, 125).Therefore, the reflex pressor response observed might havebeen due to an enhanced mechanoreflex, which is not likelyengaged using an intra-arterial balloon catheter to inducehypoperfusion.

Local Vasodilation

During systemic hypoxia there is a compensatory vasodila-tion that mirrors the decrease in O2 content (25, 26, 47, 160,161). However, during local reductions in O2 availability (viahypoperfusion) the compensatory response appears to be lessthan perfect in humans (20–23). That is, flow does not returncompletely (100% recovery) to preinflation levels. One poten-tial explanation for the incomplete compensatory vasodilationduring acute hypoperfusion may be related to an initial rise invascular resistance at the onset of balloon inflation. The initialrise in vascular resistance in these studies is in contrast to theclassic view of autoregulation (a decrease in perfusion pressure

Fig. 2. Effects of CO on the rise of systemic arterial pressure to gradedreductions in hindlimb perfusion during exercise (2 mph, 0% grade). Squaresand solid lines are control (no CO) exercise data, and circles and dashed linesare exercise with CO data. Filled symbols represent free-flow data; opensymbols represent data collected during reduced hindlimb perfusion. Thesedata demonstrate that when terminal aortic flow is decreased during mildexercise the reflex rise in systemic arterial pressure occurs at higher flows afterarterial oxygen content is reduced with CO. [Adapted with permission fromRef. 139].

Review

1532 OXYGEN AVAILABILITY AND COMPENSATORY VASODILATION

J Appl Physiol • VOL 111 • DECEMBER 2011 • www.jap.org

by guest on April 7, 2013

http://jap.physiology.org/D

ownloaded from

Page 8: Local control of skeletal muscle blood flow during exercise: … · 2013-04-14 · exercise with normoxia, anemia, hypoxia, and anemia hyp-oxia were dependent on Ca O 2 not Pa O

is followed by a reduction in resistance to blood flow throughthe muscle) (14, 50, 108, 146). However, the initial increase invascular resistance is not unprecedented in that an immediateincrease in vascular resistance has also been reported to occurin isolated perfused skeletal muscle of dogs (65). The reasonfor the acute increase in vascular resistance at the onset ofballoon inflation in our model of hypoperfusion is not com-pletely clear but may be related to dampening of pulsatile flowby balloon inflation. One possibility is that pulsatile flow is acritical component for the release of endothelium-derived va-sodilators (128). Another possibility is that a sudden drop inperfusion pressure causes the resistance vessels to recoil beforeautoregulatory vasodilation, a response that was also noted byKoch et al. (76) during mild exercise in dogs. Alterations invascular bed compliance at the onset of balloon inflation mayalso contribute to the acute increase in vascular resistanceobserved in our model of hypoperfusion (166). Since no majorpressor response is observed at the onset of balloon inflation(20–23) it is unlikely that a marked rise in sympathetic outflowexplains the acute increase in vascular resistance.

The increase in vascular resistance at the onset of ballooninflation is followed by gradual autoregulatory compensationsuch that the vascular resistance decreases over time towardand, in some cases, below preinflation values. In this contextthe magnitude of blood flow restoration following acute hypo-perfusion in contracting human skeletal muscle is predicted bythe reduction in downstream forearm vascular resistance(Fig. 4). Additionally, the recovery of flow is not associatedwith changes in balloon resistance and systemic arterial pres-sure (i.e., pressor response). Therefore, local vasodilator mech-anisms must be responsible for the reduction in vascularresistance and restoration of flow to hypoperfused contractingforearm muscles.

VASODILATOR MECHANISMS: USUAL SUSPECTS REVISITED?

The vasodilator mechanisms in the microcirculation thatcontribute to the regulation of flow distal to the occlusion likelyinclude a myogenic response of the microcirculation and all themechanisms that are involved in vasodilation in response toincreased metabolic needs, including endothelial factors andmetabolic messengers released from the muscle. As previouslymentioned, NO appears to contribute to the regulation ofskeletal muscle blood flow during systemic hypoxia (11, 19,

25, 29, 158). Similarly NOS inhibition during moderate inten-sity handgrip exercise blunts the magnitude of restoration offorearm blood flow and vascular conductance in hypoperfusedcontracting muscles (21). Moreover, the compensatory vaso-dilation to hypoperfusion in the contracting human forearmwas slowed with NOS inhibition. To our knowledge this is theonly study to examine the contribution of NO to the restorationof blood flow in hypoperfused contracting skeletal muscle.However, parallel findings have been reported in the coronarycirculation (40, 142). Duncker and Bache (40) assessed thecontribution of NO to the vasodilation of coronary resistancearteries during exercise in the presence of a flow-limitingcoronary stenosis. Inhibition of NO production during partialinflation of a hydraulic coronary occluder in dogs performingmoderate treadmill exercise resulted in a decreased meanmyocardial blood flow in the region perfused by the stenoticartery, but not in the normally perfused control region.

A substantial recovery of forearm blood flow still remainsafter NOS inhibition, thus suggesting that additional vasodila-

Fig. 3. Schematic of intra-arterial balloon catheter system.Forearm blood flow is reduced by partially occluding thebrachial artery via inflation of a Fogarty balloon catheterwith saline using a calibrated microsyringe for tight controlof balloon volume. Brachial artery blood velocity is mea-sured (via Doppler ultrasound) proximal to the balloon. Theconfiguration of the balloon upstream from the lumen of theintroducer allows measurement of the brachial arterial pres-sure (BAP) distal to the balloon that is perfusing thecontracting forearm muscles. Rhythmic forearm exercise isperformed with a hand grip device by lifting a weight 4–5cm over a pulley system at a duty cycle of 1 s contraction/and 2-s relaxation (20 contractions/min).

Fig. 4. Linear regression analysis of the relationship between % recovery offorearm blood flow (FBF) and reduction in vascular resistance (%) duringballoon inflation (n � 151) from all trials performed in references (20–23).The correlation demonstrates that the magnitude of flow restoration is pre-dicted by the reduction in vascular resistance during forearm exercise withhypoperfusion.

Review

1533OXYGEN AVAILABILITY AND COMPENSATORY VASODILATION

J Appl Physiol • VOL 111 • DECEMBER 2011 • www.jap.org

by guest on April 7, 2013

http://jap.physiology.org/D

ownloaded from

Page 9: Local control of skeletal muscle blood flow during exercise: … · 2013-04-14 · exercise with normoxia, anemia, hypoxia, and anemia hyp-oxia were dependent on Ca O 2 not Pa O

tor mechanisms are involved in the compensatory response.Previous evidence in the Seattle model suggests that adenosinereceptor antagonism blunts the recovery of hindlimb bloodflow in dogs during high-intensity exercise in response topartial vascular occlusion (76). Additionally, active hyperemiain dog skeletal muscle is potentiated during experimentallyrestricted flow in the presence of dipyridamole, thus suggestinga role of adenosine in the regulation of blood flow underconditions of arterial occlusion (73). Furthermore, adenosineproduction is positively correlated to the degree of hypoperfu-sion in the coronary circulation of dogs (34) and appears tocontribute to vasodilation of the coronary resistance vesselsduring exercise in the presence of a flow-limiting stenosis (82).In agreement with the aforementioned studies in dogs, data inthe human forearm suggest 1) adenosine contributes to theoverall magnitude of compensatory vasodilation during exer-cise with acute hypoperfusion in a manner that can be inde-pendent of NO-mediated mechanisms and 2) combined inhi-bition of adenosine receptors and NO formation results inadditive reduction in the compensatory vasodilation (20).

Lastly, prostaglandins have been reported to be involved inthe regulation of skeletal muscle blood flow following periodsof ischemia (18, 43, 71, 102) and during systemic hypoxia (29,90, 114). However, non-selective cyclooxygenase (COX) in-hibition with Ketorolac to block the production of prostaglan-dins failed to reduce the recovery of forearm blood flow andvascular conductance during exercise with acute hypoperfusion(22). These findings are in agreement with available data in thecoronary circulation of pigs where COX inhibition with indo-methacin did not alter coronary blood flow in response to anexperimentally induced flow-limiting stenosis (129). However,COX inhibition in patients with coronary artery disease causescoronary vasoconstriction (38, 44), thus suggesting that vaso-dilator prostaglandins may play greater role in the regulation ofcoronary vasomotor control in chronic vs. acute ischemia andin blood vessels subjected to atherosclerotic disease processes.Taken together, the mechanisms involved in the compensatoryvasodilator response to acute hypoperfusion in contractinghuman muscle suggests that adenosine and NO contribute,whereas prostaglandins are not obligatory (Fig. 5).

SUMMARY AND PERSPECTIVES

This review highlights the key vasodilator signals and/orpathways that contribute to the local control of skeletal muscleblood flow during exercise under conditions with reducedoxygen availability (Table 1). Both acute systemic (hypoxia)and local (hypoperfusion) mediated reductions in O2 availabil-ity elicit a compensatory vasodilator response. Although NOappears to be a common link between the hypoxia and hy-poperfusion-induced vasodilation (Table 1), its stimulus and/orinteractions with other vasodilator systems varies between thetwo conditions. Both NO and adenosine contribute to thecompensatory vasodilator responses to hypoperfusion (20, 21).By contrast, adenosine does not seem to play a major role inthe compensatory vasodilator responses to hypoxia (25, 26,59). Thus the story with adenosine is mixed; during hypoper-fusion it appears to contribute in a synergistic manner withnitric oxide to compensatory vasodilation. By contrast, duringhypoxia it does not contribute. Although combined NOS/COXinhibition has been demonstrated to reduce the compensatoryvasodilator response during hypoxic forearm exercise (29), it isunclear what the independent role of prostaglandins are in thisresponse. Available evidence suggests prostaglandins are notobligatory to the compensatory dilation observed during fore-arm exercise with hypoperfusion (22).

Despite the majority of the data presented in this reviewbeing from studies in young apparently healthy adults, it has

Fig. 5. Reduction in compensatory vasodilation [i.e., % recovery of forearmvascular conductance (FVC) compared with respective control (no drug) trial]during various pharmacological trials. Single inhibition of NOS with NG-monomethyl-L-arginine (white bar) and adenosine receptor blockade withaminophylline (black bar) substantially attenuates the compensatory vasodila-tion during exercise with acute hypoperfusion. Combined NOS/ADO inhibi-tion (dark gray bar) reveals an even greater reduction in %FVC compared withblockade of each factor alone. Single inhibition of COX with Ketorolac hasminimal effect on the compensatory vasodilation (light gray bar). CombinedNOS/COX inhibition (hatched bar) has minimal effect on the compensatoryvasodilation compared with NOS inhibition alone (white bar). Data werederived from Refs. 20–22. ADO, adenosine; COX, cyclooxygenase; NOS,nitric oxide synthase; PG, prostaglandin.

Table 1. Systemic vs. local reductions in oxygen availability: contribution of local vasodilators in the control of humanskeletal muscle blood flow during exercise

Systemic Hypoxia Local Hypoperfusion

(Low O2) (Low O2 � 2 perfusion)Perfect (proportional to the fall in arterial O2 content) Compensation Imperfect (�100% recovery)�� �-adrenergic receptor activation* ?�� Nitric oxide ��� Adenosine ��? Prostaglandins �

*Is exercise intensity dependent; contribution decreases with increased exercise intensity. ��Contributes to the compensatory vasodilation; � is notobligatory in the compensatory; vasodilation; ?lack of evidence for or against a role in the compensatory vasodilation.

Review

1534 OXYGEN AVAILABILITY AND COMPENSATORY VASODILATION

J Appl Physiol • VOL 111 • DECEMBER 2011 • www.jap.org

by guest on April 7, 2013

http://jap.physiology.org/D

ownloaded from

Page 10: Local control of skeletal muscle blood flow during exercise: … · 2013-04-14 · exercise with normoxia, anemia, hypoxia, and anemia hyp-oxia were dependent on Ca O 2 not Pa O

important implications for understanding conditions associatedwith impaired endothelial function or reduced NO bioavailabil-ity. In this context, data from aging humans clearly demon-strate an attenuated compensatory vasodilation during hypoxicexercise, which appears to be from reduced NO signaling (27).Additionally, the observation that NO contributes to the com-pensatory flow response in the microcirculation during an acuteproximal occlusion (21) raises concerns that the ability torestore flow might be impaired in conditions with endothelialdysfunction and/or reduced NO bioavailability (i.e., vasculardisease). Therefore, patients with impaired endothelium-de-pendent dilation are likely to be more susceptible to hypoper-fusion and/or ischemia during exercise, especially in vascularregions distal to a stenosis. Finally, there appears to be a greatdeal of similarity between the regulation of resistance vesseltone in human skeletal muscle (20–22) with those observed inthe coronary circulation of dogs and pigs during exercise withacute hypoperfusion (34, 40, 41, 82, 129). Thus an impairedcompensatory vasodilator response in the forearm might reflectimpairments in other vascular beds.

In conclusion, the available literature reviewed here showsthat under most circumstances skeletal muscle vasodilation istightly coupled to changes in available O2. Indeed, the com-pensatory vasodilation observed during hypoxic exercise gen-erally mirrors the fall in CaO2

. Conversely, under conditions ofincreased O2 availability (i.e., hyperoxia), vascular conduc-tance is reduced and mirrors the increase in CaO2

. Takentogether, these findings suggest that skeletal muscle blood flowis perfectly matched to changes in systemic O2 availability. Incontrast, the compensatory vasodilation in response to localreductions in O2 availability via hypoperfusion appears to beless than perfect in human skeletal muscle.

GRANTS

This work was supported by the National Institutes of Health researchGrants HL-46493 (to M. J. Joyner) and AR-55819 (to D.P. Casey) and byCTSA RR-024150. The Caywood Professorship via the Mayo Foundation alsosupported this research.

DISCLOSURES

No conflicts of interest, financial or otherwise, are declared by the authors.

AUTHOR CONTRIBUTIONS

Author contributions: D.P.C. prepared figures; D.P.C. and M.J.J. draftedmanuscript; D.P.C. and M.J.J. edited and revised manuscript; D.P.C. andM.J.J. approved final version of manuscript.

REFERENCES

1. Alam M, Smirk FH. Observations in man upon a blood pressure raisingreflex arising from the voluntary muscles. J Physiol 89: 372–383, 1937.

2. Andersen P, Saltin B. Maximal perfusion of skeletal muscle in man. JPhysiol 366: 233–249, 1985.

3. Augustyniak RA, Collins HL, Ansorge EJ, Rossi NF, O’Leary DS.Severe exercise alters the strength and mechanisms of the musclemetaboreflex. Am J Physiol Heart Circ Physiol 280: H1645–H1652,2001.

4. Bachofen M, Gage A, Bachofen H. Vascular response to changes inblood oxygen tension under various blood flow rates. Am J Physiol 220:1786–1792, 1971.

5. Barreto-Filho JA, Consolim-Colombo FM, Guerra-Riccio GM, San-tos RD, Chacra AP, Lopes HF, Teixeira SH, Martinez T, Krieger JE,Krieger EM. Hypercholesterolemia blunts forearm vasorelaxation andenhances the pressor response during acute systemic hypoxia. Arterio-scler Thromb Vasc Biol 23: 1660–1666, 2003.

6. Bergfeld GR, Forrester T. Release of ATP from human erythrocytes inresponse to a brief period of hypoxia and hypercapnia. Cardiovasc Res26: 40–47, 1992.

7. Berne RM. Cardiac nucleotides in hypoxia: possible role in regulation ofcoronary blood flow. Am J Physiol 204: 317–322, 1963.

8. Bird AD, Telfer AB. The effect of oxygen at 1 and 2 atmospheres onresting forearm blood flow. Surg Gynecol Obstet 123: 260–268, 1966.

9. Black JE, Roddie IC. The mechanism of the changes in forearmvascular resistance during hypoxia. J Physiol 143: 226–235, 1958.

10. Blauw GJ, Westendorp RG, Simons M, Chang PC, Frolich M,Meinders AE. beta-Adrenergic receptors contribute to hypoxaemia in-duced vasodilation in man. Br J Clin Pharmacol 40: 453–458, 1995.

11. Blitzer ML, Lee SD, Creager MA. Endothelium-derived nitric oxidemediates hypoxic vasodilation of resistance vessels in humans. Am JPhysiol Heart Circ Physiol 271: H1182–H1185, 1996.

12. Boegehold MA, Johnson PC. Periarteriolar and tissue PO2 duringsympathetic escape in skeletal muscle. Am J Physiol Heart Circ Physiol254: H929–H936, 1988.

13. Boushel R, Langberg H, Risum N, Kjaer M. Regulation of blood flowby prostaglandins. Current Vascular Pharmacol 2: 191–197, 2004.

14. Britton SL, Metting PJ, Ronau TF, Strader JR, Weldy DL. Autoreg-ulation of hind-limb blood flow in conscious dogs. J Physiol 368:409–422, 1985.

15. Bryan PT, Marshall JM. Adenosine receptor subtypes and vasodilata-tion in rat skeletal muscle during systemic hypoxia: a role for A1receptors. J Physiol 514: 151–162, 1999.

16. Calbet JA, Boushel R, Radegran G, Sondergaard H, Wagner PD,Saltin B. Determinants of maximal oxygen uptake in severe acutehypoxia. Am J Physiol Regul Integr Comp Physiol 284: R291–R303,2003.

17. Calbet JA, Lundby C, Sander M, Robach P, Saltin B, Boushel R.Effects of ATP-induced leg vasodilation on VO2 peak and leg O2

extraction during maximal exercise in humans. Am J Physiol RegulIntegr Comp Physiol 291: R447–R453, 2006.

18. Carlsson I, Wennmalm A. Effect of different prostaglandin synthesisinhibitors on post-occlusive blood flow in human forearm. Prostaglan-dins 26: 241–252, 1983.

19. Casey DP, Curry TB, Wilkins BW, Joyner MJ. Nitric oxide mediatedvasodilation becomes independent of �-adrenergic receptor activationwith increased intensity of hypoxic exercise. J Appl Physiol 110: 687–694, 2011.

20. Casey DP, Joyner MJ. Contribution of adenosine to the compensatorydilation in hypoperfused contracting human muscles is independent ofnitric oxide. J Appl Physiol 110: 1181–1189, 2011.

21. Casey DP, Joyner MJ. NOS inhibition blunts and delays the compen-satory dilation in hypoperfused contracting human muscles. J ApplPhysiol 107: 1685–1692, 2009.

22. Casey DP, Joyner MJ. Prostaglandins do not contribute to the nitricoxide-mediated compensatory vasodilation in hypoperfused exercisingmuscle. Am J Physiol Heart Circ Physiol 301: H261–H268, 2011.

23. Casey DP, Joyner MJ. Skeletal muscle blood flow responses to hypo-perfusion at rest and during rhythmic exercise in humans. J Appl Physiol107: 429–437, 2009.

24. Casey DP, Joyner MJ, Claus PL, Curry TB. Hyperbaric hyperoxiareduces exercising forearm blood flow in humans. Am J Physiol HeartCirc Physiol 300: H1892–H1897, 2011.

25. Casey DP, Madery BD, Curry TB, Eisenach JH, Wilkins BW, JoynerMJ. Nitric oxide contributes to the augmented vasodilatation duringhypoxic exercise. J Physiol 588: 373–385, 2010.

26. Casey DP, Madery BD, Pike TL, Eisenach JH, Dietz NM, Joyner MJ,Wilkins BW. Adenosine receptor antagonist and augmented vasodilationduring hypoxic exercise. J Appl Physiol 107: 1128–1137, 2009.

27. Casey DP, Walker BG, Curry TB, Joyner MJ. Ageing reduces thecompensatory vasodilatation during hypoxic exercise: the role of nitricoxide. J Physiol 589: 1477–1488, 2011.

28. Collins DM, McCullough WT, Ellsworth ML. Conducted vascularresponses: communication across the capillary bed. Microvasc Res 56:43–53, 1998.

29. Crecelius AR, Kirby BS, Voyles WF, Dinenno FA. Augmented skel-etal muscle hyperaemia during hypoxic exercise in humans is blunted bycombined inhibition of nitric oxide and vasodilating prostaglandins. JPhysiol 589: 3671–3683, 2011.

Review

1535OXYGEN AVAILABILITY AND COMPENSATORY VASODILATION

J Appl Physiol • VOL 111 • DECEMBER 2011 • www.jap.org

by guest on April 7, 2013

http://jap.physiology.org/D

ownloaded from

Page 11: Local control of skeletal muscle blood flow during exercise: … · 2013-04-14 · exercise with normoxia, anemia, hypoxia, and anemia hyp-oxia were dependent on Ca O 2 not Pa O

30. Daley JC 3rd, Khan MH, Hogeman CS, Sinoway LI. Autonomic andvascular responses to reduced limb perfusion. J Appl Physiol 95: 1493–1498, 2003.

31. Dawes M, Chowienczyk PJ, Ritter JM. Effects of inhibition of theL-arginine/nitric oxide pathway on vasodilation caused by beta-adrener-gic agonists in human forearm. Circulation 95: 2293–2297, 1997.

32. DeLorey DS, Shaw CN, Shoemaker JK, Kowalchuk JM, PatersonDH. The effect of hypoxia on pulmonary O2 uptake, leg blood flow andmuscle deoxygenation during single-leg knee-extension exercise. ExpPhysiol 89: 293–302, 2004.

33. Delp MD, Laughlin MH. Regulation of skeletal muscle perfusion duringexercise. Acta Physiol Scand 162: 411–419, 1998.

34. Deussen A, Moser G, Schrader J. Contribution of coronary endothelialcells to cardiac adenosine production. Pflügers Arch 406: 608–614, 1986.

35. Dinenno FA, Joyner MJ. Alpha-adrenergic control of skeletal musclecirculation at rest and during exercise in aging humans. Microcirculation13: 329–341, 2006.

36. Dinenno FA, Joyner MJ. Blunted sympathetic vasoconstriction incontracting skeletal muscle of healthy humans: is nitric oxide obligatory?J Physiol 553: 281–292, 2003.

37. Dinenno FA, Joyner MJ, Halliwill JR. Failure of systemic hypoxia toblunt alpha-adrenergic vasoconstriction in the human forearm. J Physiol549: 985–994, 2003.

38. Duffy SJ, Castle SF, Harper RW, Meredith IT. Contribution ofvasodilator prostanoids and nitric oxide to resting flow, metabolic vaso-dilation, and flow-mediated dilation in human coronary circulation.Circulation 100: 1951–1957, 1999.

39. Dufour SP, Patel RP, Brandon A, Teng X, Pearson J, Barker H, AliL, Yuen AH, Smolenski RT, Gonzalez-Alonso J. Erythrocyte-depen-dent regulation of human skeletal muscle blood flow: role of variedoxyhemoglobin and exercise on nitrite, S-nitrosohemoglobin, and ATP.Am J Physiol Heart Circ Physiol 299: H1936–H1946, 2010.

40. Duncker DJ, Bache RJ. Inhibition of nitric oxide production aggravatesmyocardial hypoperfusion during exercise in the presence of a coronaryartery stenosis. Circ Res 74: 629–640, 1994.

41. Duncker DJ, Bache RJ. Regulation of coronary blood flow duringexercise. Physiol Rev 88: 1009–1086, 2008.

42. Ellsworth ML, Ellis CG, Goldman D, Stephenson AH, Dietrich HH,Sprague RS. Erythrocytes: oxygen sensors and modulators of vasculartone. Physiology 24: 107–116, 2009.

43. Engelke KA, Halliwill JR, Proctor DN, Dietz NM, Joyner MJ.Contribution of nitric oxide and prostaglandins to reactive hyperemia inhuman forearm. J Appl Physiol 81: 1807–1814, 1996.

44. Friedman PL, Brown EJ Jr, Gunther S, Alexander RW, Barry WH,Mudge GH Jr, Grossman W. Coronary vasoconstrictor effect of indo-methacin in patients with coronary-artery disease. N Engl J Med 305:1171–1175, 1981.

45. Garovic VD, Joyner MJ, Dietz NM, Boerwinkle E, Turner ST.Beta(2)-adrenergic receptor polymorphism and nitric oxide-dependentforearm blood flow responses to isoproterenol in humans. J Physiol 546:583–589, 2003.

46. Gladwin MT. Evidence mounts that nitrite contributes to hypoxicvasodilation in the human circulation. Circulation 117: 594–597, 2008.

47. Gonzalez-Alonso J, Mortensen SP, Dawson EA, Secher NH, Dams-gaard R. Erythrocytes and the regulation of human skeletal muscle bloodflow and oxygen delivery: role of erythrocyte count and oxygenationstate of haemoglobin. J Physiol 572: 295–305, 2006.

48. Gonzalez-Alonso J, Olsen DB, Saltin B. Erythrocyte and the regulationof human skeletal muscle blood flow and oxygen delivery: role ofcirculating ATP. Circ Res 91: 1046–1055, 2002.

49. Gonzalez-Alonso J, Richardson RS, Saltin B. Exercising skeletalmuscle blood flow in humans responds to reduction in arterial oxyhae-moglobin, but not to altered free oxygen. J Physiol 530: 331–341, 2001.

50. Granger HJ, Goodman AH, Granger DN. Role of resistance andexchange vessels in local microvascular control of skeletal muscleoxygenation in the dog. Circ Res 38: 379–385, 1976.

51. Halliwill JR. Hypoxic regulation of blood flow in humans. Skeletalmuscle circulation and the role of epinephrine. Adv Exp Med Biol 543:223–236, 2003.

52. Halliwill JR, Minson CT. Effect of hypoxia on arterial baroreflexcontrol of heart rate and muscle sympathetic nerve activity in humans. JAppl Physiol 93: 857–864, 2002.

53. Hammond RL, Augustyniak RA, Rossi NF, Churchill PC, Lapa-nowski K, O’Leary DS. Heart failure alters the strength and mechanisms

of the muscle metaboreflex. Am J Physiol Heart Circ Physiol 278:H818–H828, 2000.

54. Hanada A, Sander M, Gonzalez-Alonso J. Human skeletal musclesympathetic nerve activity, heart rate and limb haemodynamics withreduced blood oxygenation and exercise. J Physiol 551: 635–647, 2003.

55. Hansen J, Sander M, Hald CF, Victor RG, Thomas GD. Metabolicmodulation of sympathetic vasoconstriction in human skeletal muscle:role of tissue hypoxia. J Physiol 527: 387–396, 2000.

56. Hansen J, Sayad D, Thomas GD, Clarke GD, Peshock RM, VictorRG. Exercise-induced attenuation of alpha-adrenoceptor mediated vaso-constriction in humans: evidence from phase-contrast MRI. CardiovascRes 41: 220–228, 1999.

57. Hansen J, Thomas GD, Harris SA, Parsons WJ, Victor RG. Differ-ential sympathetic neural control of oxygenation in resting and exercisinghuman skeletal muscle. J Clin Invest 98: 584–596, 1996.

58. Hansen M, Madsen J. Estimation of relative changes in resting muscleblood flow by 133Xe washout: the effect of oxygen. Scand J Clin LabInvest 31: 133–139, 1973.

59. Heinonen IH, Kemppainen J, Kaskinoro K, Peltonen JE, Borra R,Lindroos M, Oikonen V, Nuutila P, Knuuti J, Boushel R, KalliokoskiKK. Regulation of human skeletal muscle perfusion and its heterogeneityduring exercise in moderate hypoxia. Am J Physiol Regul Integr CompPhysiol 299: R72–R79, 2010.

60. Heistad DD, Abboud FM, Mark AL, Schmid PG. Effect of hypoxemiaon responses to norepinephrine and angiotensin in coronary and muscularvessels. J Pharmacol Exp Ther 193: 941–950, 1975.

61. Heistad DD, Wheeler RC. Effect of acute hypoxia on vascular respon-siveness in man. I. Responsiveness to lower body negative pressure andice on the forehead. II. Responses to norepinephrine and angiotensin 3.Effect of hypoxia and hypocapnia. J Clin Invest 49: 1252–1265, 1970.

62. Houssiere A, Najem B, Cuylits N, Cuypers S, Naeije R, van de BorneP. Hyperoxia enhances metaboreflex sensitivity during static exercise inhumans. Am J Physiol Heart Circ Physiol 291: H210–H215, 2006.

63. Ichinose M, Delliaux S, Watanabe K, Fujii N, Nishiyasu T. Evaluationof muscle metaboreflex function through graded reduction in forearmblood flow during rhythmic handgrip exercise in humans. Am J PhysiolHeart Circ Physiol 301: H609–H616, 2011.

64. Jagger JE, Bateman RM, Ellsworth ML, Ellis CG. Role of erythrocytein regulating local O2 delivery mediated by hemoglobin oxygenation. AmJ Physiol Heart Circ Physiol 280: H2833–H2839, 2001.

65. Jones RD, Berne RM. Intrinsic regulation of skeletal muscle blood flow.Circ Res 14: 126–138, 1964.

66. Joyner MJ. Does the pressor response to ischemic exercise improveblood flow to contracting muscles in humans? J Appl Physiol 71:1496–1501, 1991.

67. Joyner MJ. Muscle chemoreflexes and exercise in humans. Clin AutonRes 2: 201–208, 1992.

68. Joyner MJ, Wilkins BW. Exercise hyperaemia: is anything obligatorybut the hyperaemia? J Physiol 583: 855–860, 2007.

69. Katayama K, Ishida K, Iwamoto E, Iemitsu M, Koike T, Saito M.Hypoxia augments muscle sympathetic neural response to leg cycling.Am J Physiol Regul Integr Comp Physiol 301: R456–R464, 2011.

70. Kaufman MP, Hayes SG. The exercise pressor reflex. Clin Auton Res12: 429–439, 2002.

71. Kilbom A, Wennmalm A. Endogenous prostaglandins as local regula-tors of blood flow in man: effect of indomethacin on reactive andfunctional hyperaemia. J Physiol 257: 109–121, 1976.

72. Kim JK, Sala-Mercado JA, Rodriguez J, Scislo TJ, O’Leary DS.Arterial baroreflex alters strength and mechanisms of muscle metabore-flex during dynamic exercise. Am J Physiol Heart Circ Physiol 288:H1374–H1380, 2005.

73. Klabunde RE. Conditions for dipyridamole potentiation of skeletalmuscle active hyperemia. Am J Physiol Heart Circ Physiol 250: H62–H67, 1986.

74. Knight DR, Schaffartzik W, Poole DC, Hogan MC, Bebout DE,Wagner PD. Effects of hyperoxia on maximal leg O2 supply andutilization in men. J Appl Physiol 75: 2586–2594, 1993.

75. Koch DW, Newcomer SC, Proctor DN. Blood flow to exercising limbsvaries with age, gender, and training status. Can J Appl Physiol 30:554–575, 2005.

76. Koch LG, Strick DM, Britton SL, Metting PJ. Reflex versus autoreg-ulatory control of hindlimb blood flow during treadmill exercise in dogs.Am J Physiol Heart Circ Physiol 260: H436–H444, 1991.

Review

1536 OXYGEN AVAILABILITY AND COMPENSATORY VASODILATION

J Appl Physiol • VOL 111 • DECEMBER 2011 • www.jap.org

by guest on April 7, 2013

http://jap.physiology.org/D

ownloaded from

Page 12: Local control of skeletal muscle blood flow during exercise: … · 2013-04-14 · exercise with normoxia, anemia, hypoxia, and anemia hyp-oxia were dependent on Ca O 2 not Pa O

77. Koskolou MD, Calbet JA, Radegran G, Roach RC. Hypoxia and thecardiovascular response to dynamic knee-extensor exercise. Am J PhysiolHeart Circ Physiol 272: H2655–H2663, 1997.

78. Koskolou MD, Roach RC, Calbet JA, Radegran G, Saltin B. Cardio-vascular responses to dynamic exercise with acute anemia in humans. AmJ Physiol Heart Circ Physiol 273: H1787–H1793, 1997.

79. Kravec TF, Eggers GW Jr, Kettel LJ. Influence of patient age onforearm and systemic vascular response to hypoxaemia. Clin Sci 42:555–565, 1972.

80. Laughlin MH. Skeletal muscle blood flow capacity: role of musclepump in exercise hyperemia. Am J Physiol Heart Circ Physiol 253:H993–H1004, 1987.

81. Laughlin MH, Armstrong RB. Muscle blood flow during locomotoryexercise. Exerc Sport Sci Rev 13: 95–136, 1985.

82. Laxson DD, Homans DC, Bache RJ. Inhibition of adenosine-mediatedcoronary vasodilation exacerbates myocardial ischemia during exercise.Am J Physiol Heart Circ Physiol 265: H1471–H1477, 1993.

83. Leuenberger U, Gleeson K, Wroblewski K, Prophet S, Zelis R,Zwillich C, Sinoway L. Norepinephrine clearance is increased duringacute hypoxemia in humans. Am J Physiol Heart Circ Physiol 261:H1659–H1664, 1991.

84. Leuenberger UA, Gray K, Herr MD. Adenosine contributes to hypox-ia-induced forearm vasodilation in humans. J Appl Physiol 87: 2218–2224, 1999.

85. Leuenberger UA, Johnson D, Loomis J, Gray KS, MacLean DA.Venous but not skeletal muscle interstitial nitric oxide is increased duringhypobaric hypoxia. Eur J Appl Physiol 102: 457–461, 2008.

86. Limberg JK, Evans TD, Blain GM, Pegelow DF, Danielson JR,Eldridge MW, Proctor LT, Sebranek JJ, Schrage WG. Effect ofobesity and metabolic syndrome on hypoxic vasodilation. Eur J ApplPhysiol (June 9, 2011) Epub ahead of print.

87. Lorentsen E. Systemic arterial blood pressure during exercise in patientswith atherosclerosis obliterans of the lower limbs. Circulation 46: 257–263, 1972.

88. Lundby C, Boushel R, Robach P, Moller K, Saltin B, Calbet JA.During hypoxic exercise some vasoconstriction is needed to match O2

delivery with O2 demand at the microcirculatory level. J Physiol 586:123–130, 2008.

89. Mark AL, Victor RG, Nerhed C, Wallin BG. Microneurographicstudies of the mechanisms of sympathetic nerve responses to staticexercise in humans. Circ Res 57: 461–469, 1985.

90. Markwald RR, Kirby BS, Crecelius AR, Carlson RE, Voyles WF,Dinenno FA. Combined inhibition of nitric oxide and vasodilatingprostaglandins abolishes forearm vasodilatation to systemic hypoxia inhealthy humans. J Physiol 589: 1979–1990, 2011.

91. Martin EA, Nicholson WT, Eisenach JH, Charkoudian N, JoynerMJ. Influences of adenosine receptor antagonism on vasodilator re-sponses to adenosine and exercise in adenosine responders and nonre-sponders. J Appl Physiol 101: 1678–1684, 2006.

92. Mazzeo RS, Bender PR, Brooks GA, Butterfield GE, Groves BM,Sutton JR, Wolfel EE, Reeves JT. Arterial catecholamine responsesduring exercise with acute and chronic high-altitude exposure. Am JPhysiol Endocrinol Metab 261: E419–E424, 1991.

93. McClain J, Hardy C, Enders B, Smith M, Sinoway L. Limb conges-tion and sympathoexcitation during exercise. Implications for congestiveheart failure. J Clin Invest 92: 2353–2359, 1993.

94. McCullough WT, Collins DM, Ellsworth ML. Arteriolar responses toextracellular ATP in striated muscle. Am J Physiol Heart Circ Physiol272: H1886–H1891, 1997.

95. Metting PJ, Weldy DL, Ronau TF, Britton SL. Effect of aminophyl-line on hindlimb blood flow autoregulation during increased metabolismin dogs. J Appl Physiol 60: 1857–1864, 1986.

96. Mortensen SP, Gonzalez-Alonso J, Bune LT, Saltin B, Pilegaard H,Hellsten Y. ATP-induced vasodilation and purinergic receptors in thehuman leg: roles of nitric oxide, prostaglandins, and adenosine. Am JPhysiol Regul Integr Comp Physiol 296: R1140–R1148, 2009.

97. Mortensen SP, Gonzalez-Alonso J, Damsgaard R, Saltin B, HellstenY. Inhibition of nitric oxide and prostaglandins, but not endothelial-derived hyperpolarizing factors, reduces blood flow and aerobic energyturnover in the exercising human leg. J Physiol 581: 853–861, 2007.

98. Mortensen SP, Nyberg M, Thaning P, Saltin B, Hellsten Y. Adenosinecontributes to blood flow regulation in the exercising human leg byincreasing prostaglandin and nitric oxide formation. Hypertension 53:993–999, 2009.

99. Mortensen SP, Thaning P, Nyberg M, Saltin B, Hellsten Y. Localrelease of ATP into the arterial inflow and venous drainage of humanskeletal muscle: insight from ATP determination with the intravascularmicrodialysis technique. J Physiol 589: 1847–1857, 2011.

100. Neylon M, Marshall JM. The role of adenosine in the respiratory andcardiovascular response to systemic hypoxia in the rat. J Physiol 440:529–545, 1991.

101. Nicholson WT, Vaa B, Hesse C, Eisenach JH, Joyner MJ. Aging isassociated with reduced prostacyclin-mediated dilation in the humanforearm. Hypertension 53: 973–978, 2009.

102. Nowak J, Wennmalm A. A study on the role of endogenous prosta-glandins in the development of exercise-induced and post-occlusivehyperemia in human limbs. Acta Physiol Scand 106: 365–369, 1979.

103. Nyberg M, Mortensen SP, Thaning P, Saltin B, Hellsten Y. Interstitialand plasma adenosine stimulate nitric oxide and prostacyclin formationin human skeletal muscle. Hypertension 56: 1102–1108, 2010.

104. O’Leary DS, Augustyniak RA. Muscle metaboreflex increases ventric-ular performance in conscious dogs. Am J Physiol Heart Circ Physiol275: H220–H224, 1998.

105. O’Leary DS, Sheriff DD. Is the muscle metaboreflex important incontrol of blood flow to ischemic active skeletal muscle in dogs? Am JPhysiol Heart Circ Physiol 268: H980–H986, 1995.

106. Pasgaard T, Stankevicius E, Jorgensen MM, Ostergaard L, Simon-sen U, Frobert O. Hyperoxia reduces basal release of nitric oxide andcontracts porcine coronary arteries. Acta Physiol Scand 191: 285–296,2007.

107. Persson MG, Ohlen A, Lindbom L, Hedqvist P, Gustafsson LE. Roleof adenosine in functional hyperemia in skeletal muscle as indicated bypharmacological tools. Naunyn Schmiedebergs Arch Pharmacol 343:52–57, 1991.

108. Ping P, Johnson PC. Role of myogenic response in enhancing autoreg-ulation of flow during sympathetic nerve stimulation. Am J Physiol HeartCirc Physiol 263: H1177–H1184, 1992.

109. Pohl U, Busse R. Hypoxia stimulates release of endothelium-derivedrelaxant factor. Am J Physiol Heart Circ Physiol 256: H1595–H1600,1989.

110. Poucher SM. The role of the A(2A) adenosine receptor subtype infunctional hyperaemia in the hindlimb of anaesthetized cats. J Physiol492: 495–503, 1996.

111. Poucher SM, Nowell CG, Collis MG. The role of adenosine in exercisehyperaemia of the gracilis muscle in anaesthetized cats. J Physiol 427:19–29, 1990.

112. Radegran G, Calbet JA. Role of adenosine in exercise-induced humanskeletal muscle vasodilatation. Acta Physiol Scand 171: 177–185, 2001.

113. Ralevic V, Burnstock G. Receptors for purines and pyrimidines. Phar-macol Rev 50: 413–492, 1998.

114. Ray CJ, Abbas MR, Coney AM, Marshall JM. Interactions of aden-osine, prostaglandins and nitric oxide in hypoxia-induced vasodilatation:in vivo and in vitro studies. J Physiol 544: 195–209, 2002.

115. Ray CJ, Marshall JM. Measurement of nitric oxide release evoked bysystemic hypoxia and adenosine from rat skeletal muscle in vivo. JPhysiol 568: 967–978, 2005.

116. Reich T, Tuckman J, Naftchi NE, Jacobson JH. Effect of normo- andhyperbaric oxygenation on resting and postexercise calf blood flow. JAppl Physiol 28: 275–278, 1970.

117. Remensnyder JP, Mitchell JH, Sarnoff SJ. Functional sympatholysisduring muscular activity. Observations on influence of carotid sinus onoxygen uptake. Circ Res 11: 370–380, 1962.

118. Richardson DW, Kontos HA, Raper AJ, Patterson JL Jr. Modifica-tion by beta-adrenergic blockade of the circulatory response to acutehypoxia in man. J Clin Invest 46: 77–85, 1967.

119. Richardson RS, Grassi B, Gavin TP, Haseler LJ, Tagore K, Roca J,Wagner PD. Evidence of O2 supply-dependent VO2 max in the exercise-trained human quadriceps. J Appl Physiol 86: 1048–1053, 1999.

120. Richardson RS, Knight DR, Poole DC, Kurdak SS, Hogan MC,Grassi B, Wagner PD. Determinants of maximal exercise VO2 duringsingle leg knee-extensor exercise in humans. Am J Physiol Heart CircPhysiol 268: H1453–H1461, 1995.

121. Richardson RS, Noyszewski EA, Saltin B, Gonzalez-Alonso J. Effectof mild carboxy-hemoglobin on exercising skeletal muscle: intravascularand intracellular evidence. Am J Physiol Regul Integr Comp Physiol 283:R1131–R1139, 2002.

Review

1537OXYGEN AVAILABILITY AND COMPENSATORY VASODILATION

J Appl Physiol • VOL 111 • DECEMBER 2011 • www.jap.org

by guest on April 7, 2013

http://jap.physiology.org/D

ownloaded from

Page 13: Local control of skeletal muscle blood flow during exercise: … · 2013-04-14 · exercise with normoxia, anemia, hypoxia, and anemia hyp-oxia were dependent on Ca O 2 not Pa O

122. Roach RC, Koskolou MD, Calbet JA, Saltin B. Arterial O2 content andtension in regulation of cardiac output and leg blood flow during exercisein humans. Am J Physiol Heart Circ Physiol 276: H438–H445, 1999.

123. Rowell LB. How are cardiovascular and metabolic functions matchedduring exercise: what is the exercise stimulus? In: Human Circulation:Regulation During Physical Stress. New York: Oxford Univ. Press,1986, p. 287–327.

124. Rowell LB, Blackmon JR. Human cardiovascular adjustments to acutehypoxaemia. Clin Physiol 7: 349–376, 1987.

125. Rowell LB, O’Leary DS. Reflex control of the circulation duringexercise: chemoreflexes and mechanoreflexes. J Appl Physiol 69: 407–418, 1990.

126. Rowell LB, Saltin B, Kiens B, Christensen NJ. Is peak quadricepsblood flow in humans even higher during exercise with hypoxemia? AmJ Physiol Heart Circ Physiol 251: H1038–H1044, 1986.

127. Rowell LB, Savage MV, Chambers J, Blackmon JR. Cardiovascularresponses to graded reductions in leg perfusion in exercising humans. AmJ Physiol Heart Circ Physiol 261: H1545–H1553, 1991.

128. Rubanyi GM, Romero JC, Vanhoutte PM. Flow-induced release ofendothelium-derived relaxing factor. Am J Physiol Heart Circ Physiol250: H1145–H1149, 1986.

129. Ruocco NA, Most AS, Sasken H, Steiner M, Gewirtz H. Role ofendogenous prostacyclin in myocardial blood flow regulation distal to asevere coronary stenosis. Cardiovasc Res 22: 511–519, 1988.

130. Saito M, Mano T, Iwase S, Koga K, Abe H, Yamazaki Y. Responsesin muscle sympathetic activity to acute hypoxia in humans. J ApplPhysiol 65: 1548–1552, 1988.

131. Saltin B. Exercise hyperaemia: magnitude and aspects on regulation inhumans. J Physiol 583: 819–823, 2007.

132. Saltin B, Radegran G, Koskolou MD, Roach RC. Skeletal muscleblood flow in humans and its regulation during exercise. Acta PhysiolScand 162: 421–436, 1998.

133. Saunders NR, Dinenno FA, Pyke KE, Rogers AM, Tschakovsky ME.Impact of combined NO and PG blockade on rapid vasodilation in aforearm mild-to-moderate exercise transition in humans. Am J PhysiolHeart Circ Physiol 288: H214–H220, 2005.

134. Schrage WG, Joyner MJ, Dinenno FA. Local inhibition of nitric oxideand prostaglandins independently reduces forearm exercise hyperaemiain humans. J Physiol 557: 599–611, 2004.

135. Seals DR, Johnson DG, Fregosi RF. Hyperoxia lowers sympatheticactivity at rest but not during exercise in humans. Am J Physiol RegulIntegr Comp Physiol 260: R873–R878, 1991.

136. Seals DR, Johnson DG, Fregosi RF. Hypoxia potentiates exercise-induced sympathetic neural activation in humans. J Appl Physiol 71:1032–1040, 1991.

137. Shepherd JT. Circulation to skeletal muscle. In: Handbook of Physiol-ogy. The Cardiovascular System. Peripheral Circulation and OrganBlood Flow. Bethesda, MD: Am Physiol Soc, 1983, sect 2, vol. 3, p.319–370.

138. Sheriff DD. Latency of muscle chemoreflex to vascular occlusion ofactive muscle during dynamic exercise. Am J Physiol Heart Circ Physiol272: H1981–H1985, 1997.

139. Sheriff DD, Wyss CR, Rowell LB, Scher AM. Does inadequate oxygendelivery trigger pressor response to muscle hypoperfusion during exer-cise? Am J Physiol Heart Circ Physiol 253: H1199–H1207, 1987.

140. Singel DJ, Stamler JS. Chemical physiology of blood flow regulation byred blood cells: the role of nitric oxide and S-nitrosohemoglobin. AnnuRev Physiol 67: 99–145, 2005.

141. Skinner MR, Marshall JM. Studies on the roles of ATP, adenosine andnitric oxide in mediating muscle vasodilatation induced in the rat byacute systemic hypoxia. J Physiol 495: 553–560, 1996.

142. Smith TP Jr, Canty JM Jr. Modulation of coronary autoregulatoryresponses by nitric oxide Evidence for flow-dependent resistance adjust-ments in conscious dogs. Circ Res 73: 232–240, 1993.

143. Somers VK, Mark AL, Zavala DC, Abboud FM. Influence of venti-lation and hypocapnia on sympathetic nerve responses to hypoxia innormal humans. J Appl Physiol 67: 2095–2100, 1989.

144. Sprague RS, Ellsworth ML, Stephenson AH, Kleinhenz ME, LonigroAJ. Deformation-induced ATP release from red blood cells requiresCFTR activity. Am J Physiol Heart Circ Physiol 275: H1726–H1732,1998.

145. Sprague RS, Ellsworth ML, Stephenson AH, Lonigro AJ. Participa-tion of cAMP in a signal-transduction pathway relating erythrocyte

deformation to ATP release. Am J Physiol Cell Physiol 281: C1158–C1164, 2001.

146. Stainsby WN. Autoregulation of blood flow in skeletal muscle duringincreased metabolic activity. Am J Physiol 202: 273–276, 1962.

147. Stamler JS, Jia L, Eu JP, McMahon TJ, Demchenko IT, Bonaven-tura J, Gernert K, Piantadosi CA. Blood flow regulation by S-nitrosohemoglobin in the physiological oxygen gradient. Science 276:2034–2037, 1997.

148. Stickland MK, Smith CA, Soriano BJ, Dempsey JA. Sympatheticrestraint of muscle blood flow during hypoxic exercise. Am J PhysiolRegul Integr Comp Physiol 296: R1538–R1546, 2009.

149. Sundberg CJ, Kaijser L. Effects of graded restriction of perfusion oncirculation and metabolism in the working leg; quantification of a humanischaemia-model. Acta Physiol Scand 146: 1–9, 1992.

150. Thomas GD, Hansen J, Victor RG. Inhibition of alpha 2-adrenergicvasoconstriction during contraction of glycolytic, not oxidative, rathindlimb muscle. Am J Physiol Heart Circ Physiol 266: H920–H929,1994.

151. Thomas GD, Victor RG. Nitric oxide mediates contraction-inducedattenuation of sympathetic vasoconstriction in rat skeletal muscle. JPhysiol 506: 817–826, 1998.

152. Tschakovsky ME, Joyner MJ. Nitric oxide and muscle blood flow inexercise. Appl Physiol Nutr Metab 33: 151–161, 2008.

153. Tschakovsky ME, Sujirattanawimol K, Ruble SB, Valic Z, JoynerMJ. Is sympathetic neural vasoconstriction blunted in the vascular bed ofexercising human muscle? J Physiol 541: 623–635, 2002.

154. Victor RG, Bertocci LA, Pryor SL, Nunnally RL. Sympathetic nervedischarge is coupled to muscle cell pH during exercise in humans. J ClinInvest 82: 1301–1305, 1988.

155. Victor RG, Seals DR. Reflex stimulation of sympathetic outflow duringrhythmic exercise in humans. Am J Physiol Heart Circ Physiol 257:H2017–H2024, 1989.

156. Wallin BG, Victor RG, Mark AL. Sympathetic outflow to restingmuscles during static handgrip and postcontraction muscle ischemia. AmJ Physiol Heart Circ Physiol 256: H105–H110, 1989.

157. Weisbrod CJ, Eastwood PR, O’Driscoll G, Walsh JH, Best M,Halliwill JR, Green DJ. Vasomotor responses to hypoxia in type 2diabetes. Diabetes 53: 2073–2078, 2004.

158. Weisbrod CJ, Minson CT, Joyner MJ, Halliwill JR. Effects ofregional phentolamine on hypoxic vasodilatation in healthy humans. JPhysiol 537: 613–621, 2001.

159. Welch HG, Bonde-Petersen F, Graham T, Klausen K, Secher N.Effects of hyperoxia on leg blood flow and metabolism during exercise.J Appl Physiol 42: 385–390, 1977.

160. Wilkins BW, Pike TL, Martin EA, Curry TB, Ceridon ML, JoynerMJ. Exercise intensity-dependent contribution of beta-adrenergic recep-tor-mediated vasodilatation in hypoxic humans. J Physiol 586: 1195–1205, 2008.

161. Wilkins BW, Schrage WG, Liu Z, Hancock KC, Joyner MJ. Systemichypoxia and vasoconstrictor responsiveness in exercising human muscle.J Appl Physiol 101: 1343–1350, 2006.

162. Win TS, Marshall JM. Contribution of prostaglandins to the dilationthat follows isometric forearm contraction in human subjects: effects ofaspirin and hyperoxia. J Appl Physiol 99: 45–52, 2005.

163. Wolfel EE, Selland MA, Cymerman A, Brooks GA, Butterfield GE,Mazzeo RS, Grover RF, Reeves JT. O2 extraction maintains O2 uptakeduring submaximal exercise with beta-adrenergic blockade at 4,300 m. JAppl Physiol 85: 1092–1102, 1998.

164. Wyss CR, Ardell JL, Scher AM, Rowell LB. Cardiovascular responsesto graded reductions in hindlimb perfusion in exercising dogs. Am JPhysiol Heart Circ Physiol 245: H481–H486, 1983.

165. Yamauchi K, Tsutsui Y, Endo Y, Sagawa S, Yamazaki F, Shiraki K.Sympathetic nervous and hemodynamic responses to lower body nega-tive pressure in hyperbaria in men. Am J Physiol Regul Integr CompPhysiol 282: R38–R45, 2002.

166. Zamir M, Goswami R, Salzer D, Shoemaker JK. Role of vascular bedcompliance in vasomotor control in human skeletal muscle. Exp Physiol92: 841–848, 2007.

167. Zhilyaev SY, Moskvin AN, Platonova TF, Gutsaeva DR, ChurilinaIV, Demchenko IT. Hyperoxic vasoconstriction in the brain is mediatedby inactivation of nitric oxide by superoxide anions. Neurosci BehavPhysiol 33: 783–787, 2003.

Review

1538 OXYGEN AVAILABILITY AND COMPENSATORY VASODILATION

J Appl Physiol • VOL 111 • DECEMBER 2011 • www.jap.org

by guest on April 7, 2013

http://jap.physiology.org/D

ownloaded from