Download as pdf or txt
Download as pdf or txt
You are on page 1of 7

T R E N D S I N CA R D I OVA S C U L A R ME D I C I N E 23 (2013) 59–65

Available online at www.sciencedirect.com

www.elsevier.com/locate/tcm

Review article

Acid–base transporters modulate cell migration, growth


and proliferation: Implications for structure development
and remodeling of resistance arteries?

Ebbe Boedtkjern, and Christian Aalkjaer


Department of Biomedicine, Aarhus University, Denmark

artic le info abstract

Article history: Disturbed acid–base transport across the plasma membrane affects intracellular pH
Received 31 July 2012 control and has been shown—primarily based on studies with non-vascular cells—to
Received in revised form interfere with a number of fundamental cell functions including cell migration, growth
29 August 2012 and proliferation. Here, we evaluate the effects of acid–base transport and intracellular pH
Accepted 30 August 2012 on the morphology of the resistance artery wall, which is altered in a number of
Available online 21 December 2012 physiological and pathological conditions and is an independent predictor of cardiovas-
cular risk. The current evidence supports that disturbed function and/or expression of
acid–base transporters can alter resistance artery morphology—and potentially
atherosclerosis-prone conduit arteries—and hence should be considered as possible
mechanistic components and targets for treatment in cardiovascular disease. More
experimental evidence is required, however, to evaluate the cell biological effects of
acid–base transport in vascular cells, the roles of specific acid–base transporters in artery
remodeling, the relative mechanistic importance of acid–base transporters in the vascular
wall compared to other organs, and the therapeutic potential of modifying acid–base
transport activity pharmacologically or genetically.
& 2012 Elsevier Inc. All rights reserved.

Introduction 2002); and correspondingly, pulmonary artery remodeling is


associated with pulmonary hypertension (Hales et al., 1983;
The structure of resistance arteries (i.e. the media thickness Mandegar et al., 2004). Even though the association between
to lumen diameter ratio) contributes to determining vascular altered intravascular pressure and changes in artery struc-
resistance and is an independent predictor of cardiovascular ture has been described for decades, the temporal and
risk (Rizzoni et al., 2003; Mathiassen et al., 2007). In the mechanistic relationships remain to be defined in detail;
systemic circulation, morphological changes in the resis- and at present, it is unclear to what extent increased media
tance vasculature take place under a number of pathological thickness is a cause or consequence of hypertension.
conditions including diabetes (Sachidanandam et al., 2007), Vascular smooth muscle cell (VSMC) proliferation, hypertro-
hypotension (Li et al., 2002) and hypertension (Mulvany, phy and migration are obvious factors to consider when

n
Correspondence to: Department of Biomedicine, Aarhus University, Ole Worms Allé 6, Building 1180, DK-8000 Aarhus C, Denmark.
Tel.: þ45 87167716.
E-mail address: eb@fi.au.dk (E. Boedtkjer).

1050-1738/$ - see front matter & 2012 Elsevier Inc. All rights reserved.
http://dx.doi.org/10.1016/j.tcm.2012.09.001
60 TR E N D S I N CA R D I O VA S C U L A R M E D I C I N E 23 (2013) 59–65

evaluating changes in artery structure although their relative VSMCs and the effects of acid–base transporters and disturbed
importance depends on the cardiovascular disturbance in pHi on structure development and remodeling of resistance
question. There is general agreement that essential hyperten- arteries.
sion is associated with eutrophic inward remodeling of resis-
tance arteries (i.e. rearrangement of the same amount of
media material around a smaller lumen diameter (Mulvany, Regulation of intracellular pH in the vascular wall
2002)), while resistance arteries from patients with reno-
vascular hypertension (Rizzoni et al., 2000) and rats chronically Similar to other cell types, pHi of VSMCs and endothelial cells
infused with adrenergic receptor agonists (Dao et al., 2001) (ECs) is primarily influenced by metabolic acid production,
are characterized by hypertrophic inward remodeling physicochemical buffering and transport of acid–base
(i.e. increased amounts of media material around a smaller equivalents across the cell membrane, see also (Boedtkjer
lumen diameter) mainly due to VSMC hypertrophy and hyper- and Aalkjaer, 2012). Typically, since cell metabolism has the
plasia, respectively. Migration and proliferation of VSMCs are net effect of adding acid equivalents to the cytosol and the
also key pathogenic factors in atherosclerosis. negative membrane potential favors net influx of Hþ-equiva-
Considering the independent predictive value of resistance lents, most cells rely on efficient mechanisms of acid extru-
artery morphology in cardiovascular risk assessment (Rizzoni sion to avoid development of intracellular acidification. Net
et al., 2003; Mathiassen et al., 2007), treatment strategies that acid extrusion from VSMCs in mesenteric small arteries is
modify artery structure are desired. The inward remodeling mediated by the Naþ,HCO 3 -cotransporter NBCn1 (slc4a7)
of arteries from hypertensive patients can be partially (Boedtkjer et al., 2006, 2011) and the Naþ/Hþ-exchanger
reversed by antihypertensive drugs (Heagerty et al., 1988); NHE1 (slc9a1) (Boedtkjer et al., 2012b). Although, the net
however, in order to develop pharmacological therapies that effect of both of these secondary active transport mechan-
target artery structure directly, an in-depth molecular under- isms is extrusion of one Hþ-equivalent in exchange for one
standing of the pathways controlling arterial structure devel- Naþ (Fig. 1), they appear to have different roles in pHi control:
opment and remodeling is required. NHE1 is predominantly active at low pHi values and plays a
Acid–base disturbances are commonly seen in renal, gastro- major role for acid extrusion during conditions of severe
intestinal and pulmonary disease and can be associated with intracellular acidification (Boedtkjer et al., 2012b) whereas
conditions of altered metabolism, such as in diabetes, cancer NBCn1 is active at both low and near-physiological pHi values
and ischemia. Local changes in pH can also be observed and therefore contributes to acid extrusion during intracel-
physiologically during strenuous exercise or high neuronal lular acidification as well as control of steady-state pHi
activity. Changes in pH affect numerous cellular responses; (Boedtkjer et al., 2006, 2011). While these disparate roles of
and as outlined below (section Importance of acid–base trans- NBCn1 and NHE1 for pHi regulation have been most clearly
porters for cell migration and Importance of acid–base trans- defined for mouse mesenteric arteries (Boedtkjer et al., 2006,
porters for cell proliferation, survival and hypertrophy), 2011, 2012b), they also seem to apply to coronary and cerebral
intracellular pH (pHi) is of particular importance for cellular arteries (Boedtkjer et al., 2006) and thus probably are valid for
migration, hypertrophy and proliferation. Although these phe- resistance arteries in general.
nomena have so far primarily been investigated in cultured Obviously, the requirement for cellular acid extrusion
cells of non-vascular origin—and further studies are required differs under various physiological and pathological condi-
to determine the consequences of disturbed pHi regulation in tions. As illustrated in Fig. 2, it has been suggested that
vascular cells—acid–base transporters should be regarded as mechanical stimuli (e.g. shear stress (Ziegelstein et al., 1992)),
potential new therapeutic targets for modifying the structure neurohumoral factors (e.g. insulin (Boedtkjer and Aalkjaer,
of both conduit and resistance arteries. Here, we evaluate the 2009) and norepinephrine (Aalkjaer and Cragoe, 1988)) and
current evidence concerning mechanisms of pHi control in metabolic disturbances (e.g. during hypoxia (Foy et al., 1997))
Cl-

AE2

HCO3-
CAII
CO2 + H2O HCO3- + H+

HCO3- H+

NBCn1 NHE1

H2O + CO2 HCO3- Na+ Na+


H+

Fig. 1 – Schematic showing the major net acid and base extruders in VSMCs. In addition to the transport of acid–base
equivalents, the transporters also permit uptake of Naþ and Cl and hence contribute to volume regulation. Note that
NBCn1 and NHE1 perform similar net transport functions when the CO2/HCO 3 buffer system is in equilibrium. CO2 may
pass through the lipid bilayer directly or through gas channels (e.g. aquaporins).
TR E N D S I N CA R D I OVA S C U L A R M E D I C I N E 23 (2013) 59–65 61

Fig. 2 – Diagram of the proposed role of acid–base transporters in modulating vascular structure and function. Note that the
specified enzymes are only examples of mechanistic pathways involved in modifying artery structure; see the main text for
additional details.

affect acid–base transport activity in the vascular wall or in


isolated vascular cells but the signaling pathways have yet to
be defined in detail, see also (Boedtkjer and Aalkjaer, 2012;
Boedtkjer et al., 2012a).

Implications of low intracellular pH for vasomotor


function

Artery remodeling has been proposed to be wall-stress-sensitive


and depend on VSMC activation (Jacobsen et al., 2008). Hence, it
is relevant to consider the effects of disturbed acid–base trans-
port and pHi on the vasomotor function of resistance arteries;
see also (Boedtkjer and Aalkjaer, 2012) for further details. As
illustrated in Fig. 3, acute intracellular acidification of VSMCs Fig. 3 – Diagram of the predominant effects of acute and
(induced within tens of seconds) causes a marked increase in long-term changes in pHi on tone development of
intracellular [Ca2þ] and an associated vasoconstriction (see resistance arteries. Intracellular pH affects a number of
(Boedtkjer and Aalkjaer, 2012)) whereas prolonged intracellular targets including Ca2þ buffers, ion channels, intracellular
acidification of VSMCs (lasting several minutes to hours) attenu- enzymes and possibly cellular receptors or sensors. During
ates rho-kinase-dependent Ca2þ-sensitivity (Boedtkjer et al., acute intracellular acidification (induced within tens of
2011, 2012b). The inhibition of rho-kinase-dependent signaling seconds), an increase in [Ca2þ] is evident causing
seems—at least in part—attributable to a moderate intrinsic pH- vasoconstriction. During sustained intracellular
sensitivity of the rho-kinase, which is inhibited at pHi levels acidification (lasting several minutes to hours), modulation
both below and above normal physiological values (Boedtkjer of enzyme activity is prominent and both vasocontractile
et al., 2011). The reduced Ca2þ-sensitivity of VSMCs with low pHi (e.g. due to reduced rho-kinase signaling) and vasodilatory
tends to attenuate agonist-induced vasocontractile responses (e.g. due to reduced NO-synthase activity) responses are
(Boedtkjer et al., 2006, 2011, 2012b); however, in endothelium- inhibited. See section Implications of low intracellular pH
intact arteries from global knockout mice, where EC pHi is also for vasomotor function for further details.
low, the expected vasocontractile dysfunction is masked by a
reduced vasodilatory function caused by a pHi-mediated inhibi- pressure regulation in NBCn1 knockout mice (Boedtkjer et al.,
tion of NO-synthase activity in the ECs (Boedtkjer et al., 2011, 2011). These mice are mildly hypertensive at rest and show
2012b), see Fig. 3. It is relevant that both the rho-kinase and the attenuated blood pressure responses to NO-synthase inhibi-
NO-synthase pathway have complex effects on VSMC prolifera- tion (by L-NAME ingestion) and to stimulation (by angioten-
tion and migration which are independent of contractility sin II infusion) or inhibition (by Y-27632 injection) of
(Chang et al., 2002; Wirth, 2010). rho-kinase-dependent signaling (Boedtkjer et al., 2011).
Disturbed cellular acid–base regulation also has cardiovas- These rather complex perturbations of blood pressure prob-
cular implications in vivo as illustrated by the disturbed blood ably reflect that the net impact of NBCn1 knockout is a
62 TR E N D S I N CA R D I O VA S C U L A R M E D I C I N E 23 (2013) 59–65

composite of effects on multiple signaling cascades and sufficient detail to evaluate whether also in these respects
determined among others by the relative activities in the Naþ,HCO 3 -cotransporters may perform functions similar to
rho-kinase- and NO-dependent signaling pathways. those of NHE1. While the current experimental evidence sug-
gests that another Naþ,HCO 3 -cotransporter NBCe1 (slc4a4)
affects cell migration, the effect is rather modest: in transformed
Importance of acid–base transporters for cell renal epithelial cells, inhibition of NBCe1 activity reduced
migration migration velocity after an intracellular acid load and on a short
time scale of 30 min, while—in contrast to NHE1—no effect on
In both eutrophic and hypertrophic resistance artery remo- migration was seen after longer observation times (Schwab
deling and in atherosclerosis, cell migration is required for et al., 2005). In a recent study using the human breast cancer
the reorganization of the media material. Studies have shown cell line MCF-7 stimulated by overexpression of a constitutively
that the overlap between adjacent VSMCs increases within a active, amino-truncated ErbB2 receptor, no role for NBCn1 in cell
few hours of stimulation with vascular agonists (Martinez- migration was found (Lauritzen et al., 2012). Studies investigat-
Lemus et al., 2004) suggesting that VSMCs are indeed highly ing the importance of Naþ,HCO 3 -cotransport for cellular migra-
mobile cells allowing for great structural plasticity. This likely tion have so far been restricted by the lack of specific
plays an important physiological role by minimizing energy pharmacological inhibitors (Boedtkjer et al., 2012a); and clearly,
expenditure during prolonged vasoconstriction (Martinez- more information is needed to fully determine its potential role.
Lemus et al., 2004) and contributes to the pathological artery
remodeling seen among others during hypertension. There is
consistent evidence (Schwab et al., 2005; Stock and Schwab, Importance of acid–base transporters for cell
2006) that NHE1 is important for migration of cultured cells proliferation, survival and hypertrophy
and pharmacological inhibition of NHE1 has been reported to
inhibit migration velocity as much as 70%. Also for pulmonary In hypertrophic artery remodeling, the amount of media material
artery VSMCs, down-regulation of NHE1 has been shown to increases and hence cell proliferation or hypertrophy is required.
attenuate migration (Yu and Hales, 2011). It is generally accepted that an increase in pHi enhances cell
Multiple mechanisms likely contribute to the effects of NHE1 growth and proliferation, which partially appears to be due to
on cell migration and these can be roughly categorized into alkaline pHi promoting cell-cycle progression and accelerated
some that depend on ion translocation and others that depend synthesis of protein, RNA and DNA (Pedersen, 2006), see Fig. 2.
on NHE1 as a scaffold interacting physically with intra- and NHE1 seems particularly important and, in addition to effects on
extracellular components. The transport-dependent mechan- pHi, likely promotes an increase in cell volume and uptake of
isms support directed locomotion by inducing local volume inorganic ions important for cell growth and division (Pedersen,
changes (by coupled Naþ/Hþ-exchange, Cl/HCO 3 -exchange 2006). In addition, NHE1 may also independently of pHi activate
and water transport) and modifying local pHi-mediated reg- relevant intracellular signaling cascades (e.g. ERK-activation
ulation of actin polymerization (Stock and Schwab, 2006), see and MAPK-signaling) (Pedersen, 2006). Consistent with a role
Fig. 2. Important in these respects is the uneven distribution of for Naþ/Hþ-exchange and pHi in cell growth and proliferation,
NHE1 on the plasma membrane with particularly high expres- growth factors have been shown to activate Naþ/Hþ-exchange
sion levels at the leading edge of lamellipodia (Grinstein et al., activity in fibroblasts and cause intracellular alkalinization
1993). The uneven distribution of NHE1 is also crucial for the (Moolenaar et al., 1983). Using isolated VSMCs from pulmonary
generation and maintenance of a longitudinal pH gradient, arteries, previous studies have shown that knockdown of NHE1
which accompanies cell polarity (Stock et al., 2007). The or pharmacological inhibition result in a hypotrophic VSMC
extracellular acidification associated with increased net acid phenotype (LaPointe and Batlle, 1994; Yu and Hales, 2011).
extrusion also likely contributes to activation of extracellular In addition to effects on proliferation and growth, NHE1
enzymatic activity involved in matrix degradation and hence has been shown to inhibit apoptosis (Pedersen, 2006). NHE1
allows for migration of cells into tissues (Stock and Schwab, likely inhibits apoptosis by defending cells against shrinkage
2006). The transport-independent mechanisms include plas- and maintaining pHi more alkaline than the optimum for
malemmal anchoring of actin filaments (e.g. via endonucleases, caspases and cathepsins critical for induc-
ezrin–radixin–moesin proteins), interactions with extracellular tion of programmed cell death but may also act in part
matrix components (e.g. via integrins) and coupling to intra- through a transport-independent scaffolding role (Pedersen,
cellular signaling cascades altering intracellular kinase activity 2006). It should be noted that enhanced NHE1 activity in the
or gene expression (Stock and Schwab, 2006). Most of these vascular wall may under other circumstances—particularly
mechanisms, however, require verification in VSMCs. during hypoxia/reoxygenation events—contribute to cell
The contribution of Naþ,HCO 3 -cotransporters to cell migra- death, in part due to intracellular Naþ- and consequent
tion has so far only been scarcely investigated. Focusing on the Ca2þ-overload (Boedtkjer and Aalkjaer, 2012).
mechanistic pathways described above, the very similar trans-
port functions of Naþ/Hþ-exchangers and Naþ,HCO 3 -cotran-
sporters (Fig. 1) suggest that Naþ,HCO 3 -cotransporters may Effects of acid–base transporters on the structure
substitute for NHE1 at least in aspects requiring ion transloca- of systemic small arteries
tion. The interaction of NBCn1 and other Naþ,HCO 3 -cotranspor-
ters with the cytoskeleton, extracellular matrix components and Although there is strong evidence that disturbed acid–base
intracellular signaling cascades have not yet been determined in transporter function and/or expression affect cell migration,
TR E N D S I N CA R D I OVA S C U L A R M E D I C I N E 23 (2013) 59–65 63

proliferation, growth and survival, it is difficult to extrapolate 2012b) (see section Effects of acid-base transporters on the
from results based on isolated (and typically non-vascular) cells structure of systemic small arteries)—NHE1 knockout mice
to effects on artery morphology seen under in vivo conditions. (Yu et al., 2008) and rats treated with Naþ/Hþ-exchange
Work on spontaneously hypertensive rats supports, however, inhibitors (Quinn et al., 1998) have been shown to be
that Naþ/Hþ-exchange activity is enhanced and media thickness unsusceptible to hypoxia-induced pulmonary artery remo-
increased in this model of hypertension compared to control deling and development of pulmonary hypertension. Later,
Wistar-Kyoto rats (Izzard and Heagerty, 1989), see also (Boedtkjer the same authors showed that reduced NHE1 expression
and Aalkjaer, 2012) for details. Although this does not necessarily inhibits proliferation, hypertrophy and migration of pulmon-
imply a causative association, the findings are consistent with a ary artery VSMCs through a mechanism that depends on the
role for acid extrusion in regulation of blood pressure and transcription factor E2F1 (Yu and Hales, 2011). Furthermore,
resistance artery structure. Further supporting this notion, we rho-kinase (ROCK1 and ROCK2) expression was strongly
have shown that NHE1 knockout mice are hypotensive; and in reduced in pulmonary arteries from NHE1 deficient mice
mesenteric arteries from NHE1 knockout mice, the media thick- while a paradoxical increase in phosphorylation of its
ness and the media thickness to lumen diameter ratio are immediate downstream target MYPT1 was observed (Yu
reduced compared to arteries from wild type mice (Boedtkjer et al., 2008). This suggests a complex disturbance of rho-
et al., 2012b). Interestingly, this morphological change was due to kinase signaling, which may be important for the reduced
smaller VSMCs while the number of VSMCs per unit artery tendency of NHE1 knockout mice to develop pulmonary
length was unchanged (Boedtkjer et al., 2012b). The smaller hypertension. The involvement of the rho-kinase pathway
volume of the VSMCs was explained by a smaller cross- in development of pulmonary hypertension is supported by
sectional area, while cell length was unaffected (Boedtkjer other studies (Xu et al., 2010; Peng et al., 2012); and recently, it
et al., 2012b). Importantly, in the presence of CO2/HCO 3 when was shown that sustained hypercapnia inhibits rho-kinase
pHi is similar between VSMCs from NHE1 knockout and wild activity and ameliorates pulmonary hypertension in rats
type mice, VSMC function was otherwise unaffected by the (Peng et al., 2012). The mechanistic background needs to be
absence of NHE1 (Boedtkjer et al., 2012b). The normal (global) further explored, but since hypercapnia would be expected to
pHi of VSMCs in arteries from NHE1 knockout mice in the cause intracellular acidification of VSMCs—and no significant
presence of CO2/HCO 3 may suggest that transport-independent change in rho-kinase expression was seen in pulmonary
effects of NHE1 (see section Importance of acid–base transporters arteries from the hypercapnic rats—the reported pH sensi-
for cell migration) could play a major role, although local effects tivity of the rho-kinase pathway (Boedtkjer et al., 2011) (see
on pH or volume in intra- or extracellular restricted compart- section Implications of low intracellular pH for vasomotor
ments could also play a role. Nonetheless, the role of pHi for function) could play a prominent role.
arterial structure is still controversial; and it seems relevant to
address whether NBCn1 and Naþ,HCO 3 -cotransport, which plays
no obvious role for artery structure under physiological condi- Conclusions and future directions
tions (Boedtkjer et al., 2011), may play a role under stimulated or
pathological conditions. The current evidence supports that acid–base transporters
It remains a particular challenge to determine whether are important for structure development and remodeling of
in vivo inhibition of acid–base transport modifies the mor- resistance arteries in both the systemic and the pulmonary
phology of the vascular wall due to direct effects on the circulation. Hence, acid–base transporters may play a con-
VSMCs or secondary to altered hemodynamic control or siderable role in development of hypertension and athero-
neurohumoral signaling. Tissue-specific knockout mice for sclerosis and provide a promising target for new treatment
NBCn1 and NHE1 are not available at this time but could strategies to improve cardiovascular survival.
provide important clues to this question. Techniques to Nevertheless, several important questions should be
measure pHi in the vascular wall in vivo under various addressed to substantiate the role of acid–base transporters
physiological and pathological conditions and under influ- for control of resistance artery structure: (1) The link between
ence of e.g. the flowing blood, circulating humoral factors acid–base transport and changes in cell migration, prolifera-
and transmitters released from perivascular nerves would tion, growth and survival is well established for cultured non-
also represent an important step forward. vascular cells and supported by initial studies on VSMCs.
More work is required, however, to conclusively determine
the relevance of acid–base transport for cell migration,
Effects of acid–base transporters on the structure proliferation, growth and survival in VSMCs both in culture
of pulmonary small arteries and in vivo. (2) Acid–base transporters clearly play a promi-
nent role by controlling pHi but also have pHi-independent
An association between pulmonary hypertension and NHE1 effects due to their function as molecular scaffolds and their
expression and function has been suggested based on involvement in cellular signaling cascades. It should be
increased NHE1 mRNA and protein expression as well as determined to what extent the effects of disturbed
enhanced Naþ/Hþ-exchange activity in pulmonary artery acid–base transporter expression on artery structure are
VSMCs from mice with hypoxia-induced pulmonary hyper- secondary to an altered pHi regulatory function. (3) In addi-
tension (Rios et al., 2005; Shimoda et al., 2006). In line with tion to direct effects on VSMCs, disturbed acid–base transport
this—and consistent with the hypotrophic VSMCs in sys- function could alter neurohumoral signaling or water and
temic arteries from NHE1 deficient mice (Boedtkjer et al., electrolyte balance—due to or independent of changes in
64 TR E N D S I N CA R D I O VA S C U L A R M E D I C I N E 23 (2013) 59–65

pH—and thus cause a secondary change in artery morphology. Boedtkjer E, Damkier HH, Aalkjaer C. NHE1 knockout reduces
The chain of events linking acid–base transport function to blood pressure and arterial media/lumen ratio with no effect
artery morphology and the potential role of disturbed blood on resting pHi in the vascular wall. Journal of Physiology
2012;590:1895–906.
pressure regulation in this response should be further investi-
Boedtkjer E, Praetorius J, Aalkjaer C. NBCn1 (slc4a7) mediates the
gated. (4) While the discussion above has focused mainly on
Naþ-dependent bicarbonate transport important for regula-
the benefits of inhibiting artery remodeling during pathological tion of intracellular pH in mouse vascular smooth muscle
states, it should be kept in mind that artery remodeling is also cells. Circulation Research 2006;98:515–23.
an important physiological response to sustained vasoconstric- Boedtkjer E, Praetorius J, Matchkov VV, Stankevicius E, Mogensen S,
tion (Martinez-Lemus et al., 2004). The potential negative Füchtbauer AC, et al. Disruption of Naþ,HCO 3 -cotransporter

effects of inhibiting this physiological process require further NBCn1 (slc4a7) inhibits NO-mediated vasorelaxation, smooth
muscle Ca2þ-sensitivity and hypertension development in mice.
investigations.
Circulation 2011;124:1819–29.
Finally, in order to advance studies on the role of acid–base
Ch’en FF, Villafuerte FC, Swietach P, Cobden PM, Vaughan-Jones RD.
transporters in the vascular wall both in vitro and in vivo and S0859, an N-cyanosulphonamide inhibitor of sodium-bicarbonate
exploit potential therapeutic effects of acid–base transport cotransport in the heart. British Journal of Pharmacology 2008;
inhibition, it is imperative that selective pharmacological 153:972–82.
inhibitors of NBCn1 and NHE1 with appropriate pharmaco- Chang Y, Ceacareanu B, Dixit M, Sreejayan N, Hassid A. Nitric
kinetic profiles be developed. A number of Naþ/Hþ-exchange oxide-induced motility in aortic smooth muscle cells: role of
protein tyrosine phosphatase SHP-2 and GTP-binding protein
inhibitors with selectivity towards NHE1 are available and
Rho. Circulation Research 2002;91:390–7.
have been administered clinically although neurological side-
Dao HH, Lemay J, de CJ, deBlois D, Moreau P. Norepinephrine-
effects have precluded wider clinical use, see also (Boedtkjer induced aortic hyperplasia and extracellular matrix deposi-
et al., 2012a). Specific pharmacological inhibition of Naþ, tion are endothelin-dependent. Journal of Hypertension 2001;
HCO 3 -cotransport is still a major challenge (for an in-depth 19:1965–73.
discussion, see (Boedtkjer et al., 2012a)) and although the Foy RA, Shimizu S, Paul RJ. The effects of hypoxia on pHi in
compound S0859 is a promising non-selective inhibitor of porcine coronary artery endothelium and smooth muscle.
Naþ,HCO A novel method for measurements in endothelial cells
3 -cotransport (Ch’en et al., 2008; Larsen et al., 2012),
in situ. Circulation Research 1997;80:21–7.
a selective NBCn1 inhibitor is desired to reduce side-effects
Grinstein S, Woodside M, Waddell TK, Downey GP, Orlowski J,
due to inhibition of other Naþ,HCO 3 -cotransporters. S0859 Pouyssegur J, et al. Focal localization of the NHE-1 isoform of
furthermore shows strong binding to plasma proteins the Naþ/Hþ antiport: assessment of effects on intracellular
restricting its use to isolated cells in saline solutions pH. EMBO Journal 1993;12:5209–18.
(Larsen et al., 2012) and underscoring the need for further Hales CA, Kradin RL, Brandstetter RD, Zhu YJ. Impairment of
advancements both in terms of pharmacokinetic and phar- hypoxic pulmonary artery remodeling by heparin in mice.
macodynamic properties. American Review of Respiratory Disease 1983;128:747–51.
Heagerty AM, Bund SJ, Aalkjaer C. Effects of drug treatment on
human resistance arteriole morphology in essential hyper-
tension: direct evidence for structural remodelling of resis-
tance vessels. Lancet 1988;2:1209–12.
Acknowledgments Izzard AS, Heagerty AM. The measurement of internal pH in
resistance arterioles: evidence that intracellular pH is more
Related work in the authors laboratories is supported by the alkaline in SHR than WKY animals. Journal of Hypertension
Danish Council for Independent Research (10-094816 to E.B. 1989;7:173–80.
and 271-06-0472 to C.A.), the Danish Heart Foundation (08-10- Jacobsen JC, Mulvany MJ, Holstein-Rathlou NH. A mechanism for
R68-A2179-B719-22494 to E.B.) and the Lundbeck Foundation arteriolar remodeling based on maintenance of smooth mus-
cle cell activation. American Journal of Physiology Regulatory-
(R93-A8859 to E.B.).
Integrative and Comparative Physiology 2008;294:R1379–89.
LaPointe MS, Batlle DC. Naþ/Hþ exchange and vascular smooth
ref eren ces muscle proliferation. American Journal of Medical Science
1994;307(Suppl 1):S9–16.
Larsen AM, Krogsgaard-Larsen, Lauritzen G, Olesen CW, Honoré
Aalkjaer C, Cragoe Jr EJ. Intracellular pH regulation in resting and Hansen S, Boedtkjer E, et al. Gram-scale solution-phase
contracting segments of rat mesenteric resistance vessels. synthesis of selective sodium bicarbonate co-transport inhi-
Journal of Physiology 1988;402:391–410. bitor S0859: in vitro efficacy studies in breast cancer cells.
Boedtkjer E, Aalkjaer C. Insulin inhibits Naþ/Hþ exchange in ChemMedChem 2012;7:1808–14.
vascular smooth muscle and endothelial cells in situ: involve- Lauritzen G, Stock CM, Lemaire J, Lund SF, Jensen MF, Damsgaard
ment of H2O2 and tyrosine phosphatase SHP-2. American B, et al. The Naþ/Hþ exchanger NHE1, but not the Naþ,HCO 3
Journal of Physiology Heart and Circulatory Physiology 2009; cotransporter NBCn1, regulates motility of MCF7 breast can-
296:H247–55. cer cells expressing constitutively active ErbB2. Cancer Letters
Boedtkjer E, Aalkjaer C. Intracelullar pH in the resistance vascu- 2012;317:172–83.
lature: regulation and functional implications. Journal of Li ZJ, Huang W, Fung YC. Changes of zero-bending-moment
Vascular Research 2012;49:479–96. states and structures of rat arteries in response to a step
Boedtkjer E, Bunch L, Pedersen SF. Physiology, pharmacology and lowering of the blood pressure. Annals of Biomedical Engi-
pathophysiology of the pH regulatory transport proteins NHE1 neering 2002;30:379–91.
and NBCn1: similarities, differences and implications for Mandegar M, Thistlethwaite PA, Yuan JX. Molecular biology of
cancer therapy. Current Pharmaceutical Design 2012a;18: primary pulmonary hypertension. Cardiology Clinics 2004;22:
1345–71. 417–29 vi.
TR E N D S I N CA R D I OVA S C U L A R M E D I C I N E 23 (2013) 59–65 65

Martinez-Lemus LA, Hill MA, Bolz SS, Pohl U, Meininger GA. Sachidanandam K, Portik-Dobos V, Harris AK, Hutchinson JR,
Acute mechanoadaptation of vascular smooth muscle cells in Muller E, Johnson MH, et al. Evidence for vasculoprotective
response to continuous arteriolar vasoconstriction: implica- effects of ETB receptors in resistance artery remodeling in
tions for functional remodeling. FASEB Journal 2004;18:708–10. diabetes. Diabetes 2007;56:2753–8.
Mathiassen ON, Buus NH, Sihm I, Thybo NK, Mørn B, Schroeder Schwab A, Rossmann H, Klein M, Dieterich P, Gassner B, Neff C,
AP, et al. Small artery structure is an independent predictor of et al. Functional role of Naþ-HCO 3 cotransport in migration of
cardiovascular events in essential hypertension. Journal of transformed renal epithelial cells. The Journal of Physiology
Hypertension 2007;25:1021–6. 2005;568:445–58.
Moolenaar WH, Tsien RY, van der Saag PT, de Laat SW. Naþ/Hþ Shimoda LA, Fallon M, Pisarcik S, Wang J, Semenza GL. HIF-1
exchange and cytoplasmic pH in the action of growth factors regulates hypoxic induction of NHE1 expression and alkalini-
in human fibroblasts. Nature 1983;304:645–8. zation of intracellular pH in pulmonary arterial myocytes.
Mulvany MJ. Small artery remodeling and significance in the American Journal of Physiology-Lung, Cellular and Molecular
development of hypertension. News in Physiological Sciences
Physiology 2006;291:L941–9.
2002;17:105–9.
Stock C, Mueller M, Kraehling H, Mally S, Noël J, Eder C, et al. pH
Pedersen SF. The Naþ/Hþ exchanger NHE1 in stress-induced
nanoenvironment at the surface of single melanoma cells.
signal transduction: implications for cell proliferation and
Cell Physiology and Biochemistry 2007;20:679–86.
cell death. Pflugers Archiv 2006;452:249–59.
Stock C, Schwab A. Role of the Na/H exchanger NHE1 in cell
Peng G, Ivanovska J, Kantores C, Van Vliet T, Engelberts D,
migration. Acta Physiologica 2006;187:149–57 (Oxford).
Kavanagh BP, et al. Sustained therapeutic hypercapnia attenu-
Wirth A. Rho kinase and hypertension. Biochimica et Biophysica
ates pulmonary arterial rho-kinase activity and ameliorates
Acta 2010;1802:1276–84.
chronic hypoxic pulmonary hypertension in juvenile rats.
Xu EZ, Kantores C, Ivanovska J, Engelberts D, Kavanagh BP,
American Journal of Physiology-Heart and Circulatory Phy-
McNamara PJ, et al. Rescue treatment with a rho-kinase
siology 2012;302:H2599–611.
inhibitor normalizes right ventricular function and reverses
Quinn DA, Du HK, Thompson BT, Hales CA. Amiloride analogs
inhibit chronic hypoxic pulmonary hypertension. American Jour- remodeling in juvenile rats with chronic pulmonary hyper-
nal of Respiratory and Critical Care Medicine 1998;157:1263–8. tension. American Journal of Physiology-Heart and Circula-
Rios EJ, Fallon M, Wang J, Shimoda LA. Chronic hypoxia elevates tory Physiology 2010;299:H1854–64.
intracellular pH and activates Naþ/Hþ exchange in pulmonary Yu L, Hales CA. Silencing of sodium–hydrogen exchanger 1 attenu-
arterial smooth muscle cells. American Journal of Physiology- ates the proliferation, hypertrophy, and migration of pulmonary
Lung, Cellular and Molecular Physiology 2005;289:L867–74. artery smooth muscle cells via E2F1. American Journal of
Rizzoni D, Porteri E, Boari GE, De Ciuceis C, Sleiman I, Muiesan ML, Respiratory Cell and Molecular Biology 2011;45:923–30.
et al. Prognostic significance of small-artery structure in hyper- Yu L, Quinn DA, Garg HG, Hales CA. Deficiency of the NHE1 gene
tension. Circulation 2003;108:2230–5. prevents hypoxia-induced pulmonary hypertension and vas-
Rizzoni D, Porteri E, Guefi D, Piccoli A, Castellano M, Pasini G, cular remodeling. American Journal of Respiratory and Criti-
et al. Cellular hypertrophy in subcutaneous small arteries of cal Care Medicine 2008;177:1276–84.
patients with renovascular hypertension. Hypertension Ziegelstein RC, Cheng L, Capogrossi MC. Flow-dependent cytosolic
2000;35:931–5. acidification of vascular endothelial cells. Science 1992;258:656–9.

You might also like