Download as pdf or txt
Download as pdf or txt
You are on page 1of 24

Journal of Neurochemistry, 2006, 97, 310–333 doi:10.1111/j.1471-4159.2006.03717.

REVIEW The norepinephrine transporter and its regulation

Prashant Mandela* and Gregory A. Ordway 


*Departments of Pharmacology & Toxicology and Psychiatry and Human Behavior, University of
Mississippi Medical Center, Jackson, Mississippi, USA
 Department of Pharmacology, Quillen College of Medicine, East Tennessee State University, Johnson
City, Tennessee, USA

Abstract and thereby its function. The NET is a target of action of a


For many years, the norepinephrine transporter (NET) was number of drugs that are used long-term therapeutically or
considered a ‘static’ protein that contributed to the termination abused chronically. This has driven numerous investigations
of the action of norepinephrine in the synapse of noradren- of how the NET and its function are regulated by long-term
ergic neurons. The concept that the NET is dynamically exposure to drugs. While repeated exposure to many drugs
regulated, adjusting noradrenergic transmission by changing has been shown to affect NET function and expression, the
its function and/or expression, was considered initially in the intracellular mechanisms for these effects remains elusive.
mid 1980s. Since that time, a plethora of studies demonstrate Keywords: amphetamine, antidepressants, cocaine, endo-
that the NET is regulated by several intracellular and extra- cytosis, Na+/ Cl– -dependent transporters, regulation.
cellular signaling molecules, and that phosphorylation of the J. Neurochem. (2006) 97, 310–333.
NET is a major pathway regulating its cell surface expression

Norepinephrine (NE) is the principle chemical messenger


NET Structure and Function
employed in central noradrenergic and peripheral sympa-
thetic synapses. The norepinephrine transporter (NET), The NET belongs to a family of Na+/Cl– dependent transport-
present on the plasma membrane of noradrenergic neurons, ers that include transporters for serotonin, dopamine, glycine
limits the action of NE through reuptake into the cytoplasm. and GABA (Amara 1992; Masson et al. 1999; Robinson
The NET not only regulates the longevity of NE in the 2002). These transporters share similar amino acid sequences
synapse but also plays a very important role in presynaptic and presumed protein structures, and ‘pump’ mechanisms.
and postsynaptic homeostasis (Axelrod and Kopin 1969; Electrochemical energy derived from the inward gradient of
Iversen 1971; Xu et al. 2000; Zahniser and Doolen 2001). Na+ drives the intracellular accumulation of neurotransmitter
Many drugs that bind to the NET have been utilized via the NET. The function of the NET is selectively blocked by
therapeutically for the treatment of a number of disorders. the Na+/Cl– dependent binding of drugs such as desipramine
These NET-binding drugs directly alter its function, either by
acting as inhibitors or by competing as a substrate for uptake.
In 1983, the first report appeared that suggested that the NET Received September 9, 2005; revised manuscript received December 7,
could be regulated by drugs (Lee et al. 1983). These authors 2005; accepted December 12, 2005.
demonstrated that repeated exposure to drugs that modulate Address correspondence and reprint requests to Dr Gregory A.
noradrenergic transmission altered the expression of the Ordway, Department of Pharmacology, Quillen College of Medicine,
East Tennessee State University, PO Box 70577, Johnson City, TN
NET. These initial findings demonstrated that the neurons use 37614. E-mail: ordway@etsu.edu
changes in NET expression to adapt to changes in transmis- Abbreviations used: ANP, atrial natriuretic peptide; bNET, bovine
sion, and opened the door for studies of the cellular NET; BNP, brain natriuretic peptide; Br-, bromine; CaM kinase II,
mechanisms by which the NET is regulated. This paper calcium calmodulin kinase II; CNP, C-type natriuretic factor; DMI,
reviews research that has contributed to the current under- desipramine; hNET, human NET; MLC kinase II, myosin light chain
kinase II; NE, Norepinephrine; NET, norepinephrine transporter; NO,
standing of the regulation of the NET by numerous factors, nitric oxide; PI3 kinase, phosphotidyl-3-OH kinase; PKA, protein kinase
and the cellular and molecular mechanisms by which the A; PKC, protein kinase C; PMA, phorbol 12-myristate-13-acetate; rNET,
NET is regulated. rat NET; SERT, serotonin transporter.

 2006 The Authors


310 Journal Compilation  2006 International Society for Neurochemistry, J. Neurochem. (2006) 97, 310–333
Regulation of the norepinephrine transporter 311

(DMI) and nisoxetine. In fact, the binding properties of these NET in each species exhibits the same topology, including 12
drugs have been used to identify and characterize the NET, as transmembrane domains and sites for glycosylation. The
well as localize its distribution in the body (Iversen 1971; NET of each species is similar with respect to binding
Amara and Kuhar 1993; Bönisch and Brüss 1994; Ordway affinities for drugs, with some exception (Bönisch and Brüss
et al. 1997; Bauman et al. 2000; Blakely and Bauman 2000; 1994; Paczkowski et al. 1999). The structural composition of
Robinson 2002). Recent evidence indicates that the NET the hNET, rNET and bNET are discussed here, emphasizing
occurs as a homo-oligomer (Kocabas et al. 2003). similarities and differences between the species. It should be
Na+ and Cl– are the two critical ions involved in normal kept in mind that some of these differences could contribute
NET function. Br– (Bromine) is the only other ion that can to species-specific regulation of the NET.
partially mimic the function of Cl– (Friedrich and Bönisch The cloned hNET cDNA is an 1857 base pair open reading
1986; Bönisch and Brüss 1994). Kinetic studies of the frame, predicting a protein of 617 amino acids with a mass of
transporter suggest that binding of Na+ and then Cl– is a approximately 69 kDa (Pacholczyk et al. 1991). When this
prerequisite for the substrate to bind to the transporter cDNA is transfected into cultured cells, the protein that is
(Bönisch and Brüss 1994; Trendelenburg 1991). NE transport expressed demonstrates NE uptake properties and responds
by NET is electrogenic with a net gain of positive charge to the drugs known to affect NET function. Similar to other
(Trendelenburg 1991). Following the binding of substrate, species, the hNET protein consists of 12–13 hydrophobic
both Na+ and Cl– are cotransported along with the substrate segments each of 18–25 amino acids in length forming the
into the cytoplasm. Na+/K+-ATPase, a key ion pump, transmembrane domains. Both amino and carboxy termini of
provides the driving force for this secondarily active NET are located in the cytoplasmic side. The extracellular
cotransport by maintaining a Na+ concentration gradient loop between TM3 and TM4 has three potential N-glycosy-
across the plasma membrane. Na+/Cl– dependent transport of lation sites. These N-linked carbohydrate additions contrib-
NE into the cytoplasm also requires a critical balance of ute to one of the key mechanisms that regulate transporter
K+ ions across the cell membrane (Harder and Bönisch expression on the plasma membrane surface, which neces-
1985; Bönisch and Brüss 1994). A negative membrane sarily impacts the function of the NET. For example, addition
potential created by a high extracellular concentration of K+ of a carbohydrate moiety has been shown to dramatically
contributes to the driving force of amine transport (Harder and increase uptake (Melikian et al. 1994; Melikian et al. 1996;
Bönisch 1985). Removal of Na+ from the uptake buffer Nguyen and Amara 1996). There is a greater likelihood of
impairs uptake by the NET. In addition to contributing to NET glycosylated NET protein being expressed on the cell surface
function, Na+ can modulate both the affinity and velocity of than the unglycosylated forms or partially glycosylated
NE uptake. Changes in Na+ concentrations in the uptake forms. Poorly glycosylated forms or unglycosylated forms
buffer results in changes in the transporter Km and Vmax (Role are retained in the cytoplasm and are not directed to the
and Perlman 1983). Further, Na+ demand for NET function surface (Melikian et al. 1996; Nguyen and Amara 1996).
varies depending upon the extracellular concentration of NE The difference in cellular location of glycoyslated and
(Role and Perlman 1983). Alterations in membrane Na+/K+- unglycosylated forms, i.e. expressed on the surface and
ATPase activity can directly impact the ability of the NET to functional as opposed to intracellular and non-functional,
move NE across the membrane. Manipulations leading to a makes glycosylation sites potential domains for regulation of
decrease in Na+/K+-ATPase function are followed by a NET function. In addition to glycosylation sites, there are
decrease in uptake of NE and reversal of transporter function. sites for potential phosphorylation by protein kinase C and
Reversal of transporter function can only be detected if casein kinase II within the intracellular part of the transporter.
enough substrate is available in the cytoplasm. Drugs that Comparison of amino-acid sequences and the potential
block ATP synthesis (cyanide) or Na+/K+-ATPase pump regulatory sites reveal a close similarity with other trans-
(oubain) simultaneously decrease NET function (Harder and porters including GABA, glycine, SERT, and dopamine
Bönisch 1985; Bönisch and Brüss 1994). transporters (DAT; Pacholczyk et al. 1991; Amara 1992;
NET structure and systems regulating its function are Masson et al. 1999; Robinson 2002).
currently under intense investigation because, like the The rat NET and the bNET contain 617 and 615 amino
serotonin transporter (SERT), the NET is a target for acids, respectively. Like the hNET, both rNET and bNET
antidepressant and psychostimulant drugs. Analysis of NET share structural similarities with other Na+/Cl– neurotrans-
structure reveals critical information regarding the functional mitter transporters, such as 12 transmembrane domains with
residues and domains responsible for NET function, substrate both N- and C- termini on the cytoplasmic face and a large
binding, drug binding, and regulation. The human NET extracellular loop between TM3 and TM4 (Lingen et al.
(hNET), bovine NET (bNET), and rat NET (rNET) have 1994; Brüss et al. 1997). rNET has two and bNET (like
been cloned (Pacholczyk et al. 1991; Lingen et al. 1994; hNET) has three potential sites for N-glycosylation that are
Brüss et al. 1997). The hNET gene is localized at chromo- located in the extracellular loop (Lingen et al. 1994; Brüss
some 16q12.2 (Gelernter et al. 1993; Brüss et al. 1993). The et al. 1997). Analysis of the amino acid sequences reveals

 2006 The Authors


Journal Compilation  2006 International Society for Neurochemistry, J. Neurochem. (2006) 97, 310–333
312 P. Mandela and G.A. Ordway

two potential sites for phosphorylation by protein kinase C in the typical hNET (with exon 14), while expression of the short
rNET and three in bNET. A potential phosphorylation site at form of the exon 15 splice variant had no uptake activity and
amino acid 259 is conserved among the 3 species (rat, bovine was not expressed on the cell surface (Kitayama et al. 2001).
and human). Another site for phosphorylation is at amino The mechanism underlying the reduced surface expression of
acid 579 in the intracellular C-terminus (in rNET and bNET, the NET may be a result of the inability of the splice variant
but not in hNET) a third phosphorylation site is also in bNET hNET to bind to the PDZ-domain containing PICK1 protein
at the C-terminal (Lingen et al. 1994; Brüss et al. 1997). A (Distelmaier et al. 2004), a step crucial to cell surface targeting
potential casein kinase II phosphorylation site at amino acid (see below). Similar findings and the importance of the C-
583 near the C-terminus is also conserved among the 3 terminus in targeting of the NET to the plasma membrane has
species (rat, bovine, human). This conserved site for been demonstrated for the bNET expressed in HEK-293 cells
phosphorylation by casein kinase II among the species (Burton et al. 1998).
strongly suggests a role of casein kinase in regulating the
transporter function (Brüss et al. 1997) though no such
Regulation by Intracellular Signaling Molecules
involvement has yet been reported. Like hNET, bNET and
rNET show characteristic uptake and pharmacological prop- The NET is regulated by a number of intracellular signaling
erties when expressed transiently or stably in other cells molecules. One common pathway employed by these
systems. At the nucleotide level, rNET and hNET share 87% molecules is phosphorylation. Several signaling cascades
similarity in the coding region, while rNET and bNET share are considered below and these are summarized in Table 1.
84% similarity, and hNET and bNET share 90% similarity.
Even the amino acid sequences have very close similarities. Cyclic AMP and protein kinase A
Similarities among species reflect conserved structural fea- Considerable research demonstrates a role for cyclic AMP
tures that are more conserved than those of two different (cAMP) in regulating the NET and its function. Cyclic AMP
catecholamine transporters (e.g. NET and DAT) within the is a key component in the second messenger system linking
same species (Brüss et al. 1997). upstream G protein-coupled receptors to downstream sign-
The cellular trafficking of Na+/Cl– dependent transporters aling cascades and is produced by activation of adenylyl
was once thought to be limited to regulatory events (discussed cyclase. A membrane permeable, metabolically stable cAMP
below). However, recent studies on these transporters demon- analogue, 8 Br-cAMP, is commonly employed to study
strate that a dynamic basal transporter recycling occurs cAMP-mediated events. 8 Br-cAMP induces a concentration-
between the plasma membrane and the endosomal compart- dependent decrease in uptake of [3H]NE in bovine adrenal
ments that are capable of achieving a rate of 3–5% per minute medullary chromaffin cells (Bunn et al. 1992). In contrast,
(Deken et al. 2003; Loder and Melikian 2003). Recent studies forskolin, a drug that stimulates production of intracellular
by Holton et al. have identified a critical region in the C- cAMP by directly activating adenylyl cyclase, has a biphasic
terminus of these transporters that plays a vital role in their effect on NE uptake in chromaffin cells. At low concentra-
constitutive and regulation-dependent endocytosis (Holton tions, forskolin (1.0–3.0 lmol/L) stimulates an increase in
et al. 2005). Residues 584–593 in the C-terminus of the [3H]NE uptake, while at higher concentrations (10–
NET corresponding to amino acid sequence LWERLAYGIT 100 lmol/L) forskolin inhibits uptake and returns [3H]NE
are a vital component in the NET endocytotic process. This accumulation to basal levels (Bunn et al. 1992). In the same
region of the NET is conserved amongst Na+/Cl– dependent cell line pertussis toxin, which facilitates an increase in
transporters. Mutations of this sequence block the constitutive cAMP by inhibiting Gi, decreases uptake of [3H]NE. The
endocytosis of the transporter, and are likely to block its effect of pertussis toxin on uptake appears to be only partially
regulated endocytosis based on findings using a mutated DAT accounted for by cAMP actions (Bunn et al. 1992).
(Holton et al. 2005). Short-term (15 min) exposure of rat PC12 cells to forskolin
Alternative splicing has been shown to play a role in or a stable cAMP analogue (but not cGMP) has been shown to
hNET regulation (Pörzgen et al. 1995). The hNET gene decrease NET activity (Bryan-Lluka et al. 2001). This inhib-
consists of 14 exons, interrupted by 13 introns (Pörzgen et al. itory effect is rapidly reversed when the drugs are removed
1995). An additional exon (exon 15) has been identified and from the uptake assay. The effect of forskolin on NET function
can be spliced to exon 13, skipping exon 14. There are two is inhibited by staurosporine, a mixed protein kinase inhibitor
alternative 5¢-acceptor sites at exon 15 that can lead to two that has no effect on NET function by itself in PC12 cells.
variants of the alternatively spliced NET containing exon 15. Short-term exposure of PC12 cells to forskolin reduces the
The shorter and longer forms result in variants with 3 or 18 Vmax of the NET. Longer exposure (24 h) of PC12 cells to
amino acids, respectively, at the C-terminal end of the NET forskolin is accompanied by a reduction in NET gene
(Pörzgen et al. 1998). Transfection of cultured cells with the expression (Bryan-Lluka et al. 2001). While acute studies
long form of the exon 15 splice variant yielded NE uptake (30 min exposures) demonstrate that protein kinase C (PKC)
activity and NET cell surface expression that was lower than activation can decrease Vmax and move transporters away from

 2006 The Authors


Journal Compilation  2006 International Society for Neurochemistry, J. Neurochem. (2006) 97, 310–333
Table 1 Intracellular signal regulation of the NET

Messenger Exposure NE Cell Surface NET


System Agent Cell type Time Uptake NET Kinetics Expression mRNA References

PKC bPMA SK-N-SH 30 min fl « Km fl Vmax fl – Apparsundaram et al. (1998a)

 2006 The Authors


bPMA COS-7 30 min fl – – – Bönisch et al. (1998)
(trasfected with hNET,
bNET, hNETs259A)
bPMA COS-7 24 h fl « Km fl Vmax – – Bönisch et al. (1998)
(transfected with hNET,
bNET, hNETs259A)
bPMA SK-N-SH-SY5Y 24 h fl « Km fl Vmax fl – Bönisch et al. (1998)
bPMA Placental 30 min fl « Km fl Vmax fl – Jayanthi et al. (2004)
trophoblasts (rat)
bPMA HEK293,LLC-PK1, 30 min fl « Km fl Vmax fl – Apparsundaram et al. (1998b)
(stably transfected)
cAMP/PKA cAMP analogues SK-N-SH-SY5Y 15 min, 24 h « – – – Bönisch et al. (1998)
(8Br-cAMP,db-cAMP)
cAMP anaologues SK-N-SH cells 40 min « – – – Apparsundaram et al. (1998a)
(8-pCT-cAMP,
Rp-8-pCT-cAMp)
8-Br-cAMP Chromaffin cells 25 min fl – – – Bunn et al. (1992)
dbcAMP PC12 cells 15 min, 24 h fl – – fl (24 h) Bryan-Lluka et al. (2001)
dbcAMP COS-7, transfected 15 min › – – – Bryan-Lluka et al. (2001)
with rat NET
dbcAMP COS-7, transfected 24 h › – – – Bryan-Lluka et al. (2001)
with hNET
Forskolin SK-N-SH-SY5Y 15 min, 24 h « – – – Bönisch et al. (1998)
Forskolin Chromaffin cells 25 min Biphasic – – – Bunn et al. (1992)
effect ›fl

Journal Compilation  2006 International Society for Neurochemistry, J. Neurochem. (2006) 97, 310–333
Forskolin PC12 cells 15 min fl « Km fl Vmax « – Bryan-Lluka et al. (2001)
Forskolin PC12 cells 24 h fl ? – fl Bryan-Lluka et al. (2001)
Forskolin COS-7 ratNET 15 min « – – – Bryan-Lluka et al. (2001)
Forskolin (100 lM) COS7 ratNET 24 h › – – – Bryan-Lluka et al. (2001)
Forskolin COS7 hNET 24 h › – – – Bryan-Lluka et al. (2001)
Cholera toxin SK-N-SH-SY5Y 15 min, 24 h « – – – Bönisch et al. (1998)
Cholera toxin PC12 24 h fl – – – Bönisch et al. (1998);
Bryan-Lluka et al. (2001)
Pertussis toxin Chromafin cells 90min fl « Km fl Vmax – – Bunn et al. (1992)
cGMP/PKG 8-br-cGMP PC12, SK-N-SH-SY5Y 15 min « – – – Bryan-Lluka et al. (2001)
Regulation of the norepinephrine transporter

CaM Kinase KN93 (inhibitor) SK-N-SH 40 min fl – – – Apparsundaram et al. (1998a)


313
314 P. Mandela and G.A. Ordway

Bauman et al. (2000)


the plasma membrane in SK-N-SH cells (Apparsundaram

Uchida et al. (1998)


Sung et al. (2003)

Sung et al. (2003)


Kaye et al. (1997)
et al. 1998a) (see below), such redistribution is not seen with
cAMP-induced regulation of NE uptake where the Bmax of

et al. (2004)
References
[3H]nisoxetine binding in intact cells does not change (Bryan-

Jayanthi
Lluka et al. 2001). It is noteworthy here that [3H]nisoxetine
binding assays in this latter study were performed in a manner
that limits [3H]nisoxetine binding to surface transporters
(Apparsundaram et al. 1998a; Distelmaier et al. 2004).
mRNA

cAMP-mediated regulation of NET in PC12 cells therefore


NET

may result from a change in the conformation of the transporter



?
that hinders reorientation of the transporter during the transport
cycle, and thereby decreases the Vmax without effecting the
Cell Surface
Expression

affinity (Km) of the transporter for the substrate and without


changing the level of expression of NET protein on the cell
surface. It is highly unlikely that cAMP has direct effects on the


NET, since the effects of cAMP are cell-line dependent (see


below).
NET Kinetics

Numerous studies demonstrate that cAMP-induced effects


fl Km › Vmax

on NET function are not consistent across cell lines or


species. In contrast to effects in PC12 cells, cAMP analogues
had no effects on NE uptake in human SK-N-SH-SY5Y cells

(naturally expressing the NET) or COS-7 cells transiently


transfected with rat or human NET cDNA (Bryan-Lluka
Uptake

et al. 2001). It is notable in this regard that PC12 cells and


NE


SK-N-SHSY5Y cells have different origins (rat, human).


Treatment of another human neuroblastoma cell line that
naturally expresses the hNET, SK-N-SH cells, with cAMP
Exposure

10 min
30 min
30 min

analogues also does not affect NE uptake (Apparsundaram


2 days
Time

1h

1h

et al. 1998a). Together, these studies suggest that cAMP-


induced regulation is not a general phenomenon in NET
regulation but is cell line- and species-dependent.
Vas deferens, C. cortex (rat)

Cyclic GMP
Vas deferens (inhibitor)

CAD-hNET, SK-N-SH,
Placental trophoblast

sympathetic neurons

The cGMP analogue, 8-bromo-c-GMP or dbcGMP has no


effect on NET activity in both PC12 and SK-N-SHSY5Y
cells, suggesting that cGMP and cGMP-dependent protein
PC12 and rat

CAD-hNET

PC12 cells

kinases do not play a role in regulating rat and human NET


Cell type

(Bryan-Lluka et al. 2001).

Protein kinase C (PKC)


The involvement of PKC in the regulation of transporter
SNAP (NOx donor)

function and membrane expression is proving to be universal


Extracellular Ca2+
oligonucleotides

among monoamine transporters. There are a number of


Okadaic acid
Okadaic acid

Syntaxin 1A

potential sites for phosphorylation by PKC of the NET in


BONT/C1
antisense
(inhibitor)

several species (see above). Information from these early


Agent

studies has been confirmed by multiple studies demonstrating


that the PKC pathway is the most common intracellular
mechanism through which transporters can be regulated.
Nitric oxide pathway

Studies involving both transfected cell lines and cell lines


Table 1 Continued

Dephosphorylation

Vesicular shuttling

naturally expressing the NET have shown that PKC can


regulate the expression of transporter on the cell surface, and
Messenger

thereby affect NET function.


System

To investigate the role of PKC phosphorylation sites on


Other

NET expression and function, Bönisch et al. (1998) utilized

 2006 The Authors


Journal Compilation  2006 International Society for Neurochemistry, J. Neurochem. (2006) 97, 310–333
Regulation of the norepinephrine transporter 315

COS-7 cells transfected with hNET (stated to have one site affinity (Km) for the substrate is unaffected by PKC
for PKC-induced phosphorylation, see below), bNET (has activation. Muscarinic receptor activation increases intracel-
three sites for PKC-induced phosphorylation) and a mutant lular concentrations of Ca2+ (through inositol triphosphate
hNET, hNETS259A. COS-7 cells transfected with generation) and diacylglycerol, and both Ca2+ and diacyl-
hNETS259 express a NET that has serine 259 replaced with glycerol activate PKC. PKC activation leads to phosphory-
alanine, thereby removing the putative single site for lation of the NET and decreased surface expression of the
phosphorylation by PKC. Acute treatment (10 min) of these NET. Inhibition of PKC blocks PMA-mediated effects on
cell lines with phorbol 12-myristate-13-acetate (PMA; PKC NET, but does not completely block muscarinic receptor-
agonist), okadaic acid (protein phosphotase inhibitor) and induced effects. Co-treatment of cells with methacholine
staurosporine (PKC antagonist) did not affect NE uptake. plus BAPTA/AM, a Ca2+ chelator, eliminates the effect of
However, longer exposure to PMA resulted in decreased methacholine on NE uptake. These observations suggest a
[3H]NE uptake via NET in all 3 cell lines. The effects of possible second mechanism, other than Ca2+-dependent PKC
PMA were inhibited by staurosporine, which had no effect on activation, through which intracellular Ca2+ can regulate NE
uptake on its own. The change in NE uptake following a 24 h uptake at the plasma membrane (Apparsundaram et al.
exposure to PMA was due to a reduction in transport velocity 1998a).
(Vmax) without a change in the affinity (Km) of the Recent studies on NET in placental trophoblasts have
transporter for the substrate. Reduced uptake induced by provided a wealth of information regarding PKC-induced
PMA exposure in COS-7 cells with all 3 varieties of phosphorylation and internalization of NET. A robust
transfected NET, as well as SK-N-SH-SY5Y cells (expres- expression of functional endogenous NET has been shown
sing the wild-type hNET), was accompanied by a reduction in rat placental trophoblasts (Jayanthi et al. 2002). As
of the binding of [3H]nisoxetine to NET on these cells demonstrated in other cell lines, PMA stimulation of PKC
(Bönisch et al. 1998). Okadaic acid, a protein phosphatase decreases [3H]NE uptake in a time- dependent and concen-
inhibitor, did not have any effect on PMA-induced decreases tration-dependent fashion, producing a robust reduction in
of uptake. Together, these findings demonstrate that NET Vmax with no effect on Km. [3H]Nisoxetine binding in whole
function is sensitive to PKC, although direct phosphorylation cell trophoblasts, as wells as cell surface biotinylation assays,
of the NET at serine 259 by PKC is not necessary to down- demonstrate reductions in surface expression of the NET
regulate its function. Furthermore, in contrast to cAMP- (Jayanthi et al. 2004). This effect of PMA is mediated by
dependent regulation, PKC-dependent regulation of the NET PKC in a Ca2+-independent manner. Removal of both
is irrespective of species and cell line. intracellular and extracellular Ca2+ did not prevent the effect
Further studies on stable and cell lines, HEK-293, LLC- of PMA on NET regulation. This observation suggests a role
PK1, and COS-7 cells, transiently transfected with NET have for Ca2+-independent PKC isoforms in NET regulation
confirmed NET internalization after PKC stimulation. PMA (Jayanthi et al. 2004).
decreases uptake in a time-dependent and dose-dependent Jayanthi et al. (2004) have further demonstrated that the
manner. PKC-induced regulation of surface transporter NET is directly phosphorylated by PKC activation. Exposure
function appears to be a specialized effect on NET function of rat placental trophoblasts to PMA or okadaic acid (a PP1/
in these cell lines because a similar effect is not seen with 2 A inhibitor) results in phosphorylation of the NET.
other transporter systems (e.g. Na+-dependent leucine trans- Inhibition of clathirin-mediated endocytosis by concanavalin
porter) present in the same cell lines. Activation of PKC A or monodansyl cadaverine does not block PMA-induced
results in phosphorylation of NET. Although the NET does down-regulation of NET, suggesting that NET regulation by
not appear to be regulated by PKC through phosphorylation PKC occurs via a clathrin-independent pathway. Placental
at serine 259 (see above; Bönisch and coworkers, 1998), it is trophoblasts expressing dominant negative mutants of dyn-
phosphorylated at other sites (Apparsundaram et al. 1998b). amin 1 or dynamin 2 were also susceptible to PMA-induced
Phosphorylation of the NET is followed by the internalizat- regulation of NET, again indicating that this system does not
ion of the transporter into the cytoplasm from the plasma play a role in NET endocytosis induced by PKC activation.
membrane. Reduced surface expression of NET is the In addition, placental trophoblast cells expressing dominant
primary mechanism contributing to PKC- induced decrease negative mutants of caveolin 1 or caveolin 3 did not disrupt
in NET activity (Apparsundaram et al. 1998b). PMA-induced internalization of NET. In contrast, the
Muscarinic receptors can signal through Ca2+- and PKC- cholesterol-disrupting agent, filipin, completely blocked
dependent mechanisms to modulate NET function (Appars- PMA-induced effects on the NET. The inhibitory effect of
undaram et al. 1998a). Stimulation of muscarinic receptors filipin appears to be limited to PKC-mediated NET inter-
on SK-N-SH cells with methacholine reduces [3H]NE nalization since other PMA-mediated inhibitions, e.g. taurine
uptake. Like direct PKC activation, muscarinic activation uptake, were unaffected by filipin. These latter observations
decreases maximum transport velocity (Vmax) secondary to a provide evidence for a role of lipid rafts in PMA-mediated
relocation of surface transporters to the cytoplasm. NET NET internalization in rat placental trophoblasts. The authors

 2006 The Authors


Journal Compilation  2006 International Society for Neurochemistry, J. Neurochem. (2006) 97, 310–333
316 P. Mandela and G.A. Ordway

also suggest that lipid rafts may function as transition phase buting to the increase in uptake, but the key in either case is
docking sites before NET internalization (Jayanthi et al. phosphorylation. Cells pretreated with 1 mM Ca2+ show a
2004). significant increase in the Bmax of [3H]DMI binding meas-
Taken together, these findings demonstrate that PKC can ured in crude membrane homogenates, raising the possibility
regulate NET function by altering the equilibrium between that Ca2+ induces relocation of transporter from cytoplasm to
the plasma membrane and the cytoplasmic fractions of the plasma membrane (Uchida et al. 1998). Interestingly,
transporters. PKC-induced reductions in NET function can pretreatment of SK-N-SH cells with KN-93, a CAM kinase II
function independently of the endocytosis pathway mediated inhibitor, reduces uptake independent of external Ca2+
by dynamin and clathrin, at least in rat placental trophoblasts (Apparsundaram et al. 1998a), and preliminary studies in
(Jayanthi et al. 2004). Recently, Holton et al. (2005) have our laboratory also are consistent with the idea that basal
demonstrated that an endocytic signal near the C terminus CaM kinase II activity is required for normal NET function.
of DAT, one that is shared by other Na+/Cl– dependent Apparently Ca2+, when applied externally, can influence
transporters including the NET, is vital to PKC-driven transporter function by increasing uptake through CaM
regulation as well as constitutive endocytotic cycling of the kinase II and MLC kinase rather than decreasing the uptake
DAT. Specific mutations within this sequence block consti- by taking the PKC route. Overall, current evidence suggests
tutive recycling without affecting PKC-induced regulation, that the pathway of activation by Ca2+ when applied
while other mutations block both constitutive endocytotic externally is different to the pathway activated when internal
cycling and PKC-induced regulation. Finally, while PKC- stores of Ca2+ are released.
induced reduction in NET function appears to be independ-
ent of species and cell line differences, the mechanisms of Protein phosphatases
NET sequestration following phosphorylation may exhibit Protein kinases and phosphatases control protein targeting
cell line-dependence. For example, while dynamin mutants and trafficking through phosphorylation and dephosphoryla-
did not effect PMA-induced effects on the NET in rat tion processes, respectively. The fine balance of the state of
trophoblasts, PMA had no effect on NET sequestration in phosphorylation is orchestrated by these two critical classes
HEK-293 cells coexpressing both human NET and a K44A of enzymes and represents a vital regulatory mechanism
mutant of dynamin 1 (Jayanthi et al. 2004). of protein function. Okadaic acid (OA) and calyculin A,
inhibitors of protein phosphatase 1/2A trigger SERT phos-
phorylation and reduce transporter activity (Ramamoorthy
Ca2+ and CaM kinase II
et al. 1998). Similar mechanisms have been demonstrated for
Extracellular Ca2+ plays an influential role in NET function. the regulation of the NET. Treatment of vas deferens with
PC12 cells preincubated with 0.3–10 mM Ca2+ for 10 min OA decreases NE uptake. Immunoprecipitation of the
show a marked increase in NE uptake (Uchida et al. 1998), extracts from the same tissue reveal the presence of NET/
characterized by an increase in maximum transport velocity PP2Ac complex. Further treatment of vas deferens with OA
(Vmax) and a decrease in substrate affinity (Km). Despite decreases PP2Ac levels in NET immunoprecipitants and no
removal of Ca2+ (1 mM) from the suspension medium after such effect is noticed in total extracts (Bauman et al. 2000).
the initial exposure, the increase in NE uptake persists, Okadaic acid treatment blocks protein phosphatase and thus
suggesting a role for an unknown indirect mechanism inhibits its interaction with NET. The phosphatase–NET
through which Ca2+ produces an effect (Uchida et al. interaction is essential for stabilizing the NET and for surface
1998). Ca2+-induced alterations of uptake kinetics are also expression of the NET.
observed for other monoamine transporters, including the Recent studies from rat placental trophoblasts also dem-
DAT and SERT, although there are qualitative differences onstrate a similar mechanism. Okadaic acid reduces NE
(Uchikawa et al. 1995; Yura et al. 1996). Regulation of uptake via the NET and also increases the phosphorylated
transporter function by changes in [Ca2+] could result from NET fraction (Jayanthi et al. 2004). These findings collec-
changes in the activity of Ca2+-dependent kinases such as tively demonstrate a role for phosphatases in phosphoryla-
PKC, Ca2+/calmodulin-dependent kinase II (CaM kinase II), tion-dependent regulation of NET. Blockade of phosphatase
and/or myosin light chain kinase (MLC kinase). However, function can severely restrict transporter recycling to the
PKC inhibitors have no affect on Ca2+-induced increases in plasma membrane by increasing the phosphorylated fraction
uptake. In contrast, CaM kinase II and MLC kinase inhibitors of the NET and leading to a decrease in NE uptake via NET
robustly inhibit Ca2+-dependent increases in NE uptake. (Jayanthi et al. 2004).
CaM kinase II could directly phosphorylate the transporter
and regulate its function, or it could work through activating ATP
MLC kinase and subsequent phosphorylation and movement ATP is an important molecule in biological systems that
of the transporter from the cytoplasm to the plasma caters to energy demands at the cellular level. Besides its role
membrane. MLC kinase could also be independently contri- as an energy ferrying molecule, ATP mediates a wide variety

 2006 The Authors


Journal Compilation  2006 International Society for Neurochemistry, J. Neurochem. (2006) 97, 310–333
Regulation of the norepinephrine transporter 317

of cellular functions by acting through signaling systems. ecto-kinase pathway could play a role at submicromolar
ATP present in the extracellular compartment is incapable of concentrations but at higher concentrations ATP could
penetrating cell/neuronal membranes, it thus exerts its directly interact with the transporter and alter its function
influence on cellular systems through a wide range of in an allosteric manner or act via the purinergic receptors and
mediators present externally (Caldwell et al. 1960; Burn- influence transport function via a second messenger system
stock et al. 1997). These mediators include ATP receptors (Eshleman et al. 1995). The differences between the two
(purinergic receptors) present on the plasma membrane studies mentioned above are likely due to various experi-
(Burnstock et al. 1997; Burnstock 1981) as well as externally mental conditions such as cell culture conditions and buffer
facing enzymes such as ecto-ATPases (Nagy et al. 1986) and media (Eshleman et al. 1995). These studies provide some
ecto-protein kinases (Ehrlich et al. 1986). important regulatory mechanisms through which ATP can
ATP is also present in vesicles loaded with neurotrans- work and exert its influence on NE uptake. More work is
mitters. The presence of ATP and neurotransmitters in the needed to understand the role and dynamics of ATP actions
same vesicular packet raises the possibility that ATP may at NE terminals. ATP effects on the NET seem to be of
function as a cotransmitter. ATP is stored in the synaptic particular physiological relevance, since ATP is released with
vesicles containing NE and is concomitantly released with NE from neuronal terminals.
NE during a depolarization event (Geffen and Livett 1971). It
is present in the extracellular medium while NE reuptake
is in progress (Cao et al. 1990). Furthermore, ATP can Proteins contributing to cellular NET trafficking
stimulate NE uptake as shown in rat iris preparations
(Angelakos 1964). ATP can also alter uptake of dopamine Synaptic proteins
and NE in rat occipital cortex synaptosomes (Cao et al. Recent evidence strongly implicates a role of synaptic
1990). These initial findings implicated a role for ATP in NE proteins in regulating the expression of cell surface proteins
uptake. such as transporters (summarized in Table 1). Synaptic
Further variations in the ionic environment (Ca2+, Mg2+) proteins presently known to regulate NET function are the
have been shown to play a critical role in ATP-induced SNARE (Sung et al. 2003) (soluble N-ethylamide-sensitive
regulation of NET (Hardwick et al. 1989). ATP alters NE factor attachment protein receptor) proteins and PICK1
uptake in a concentration- and cation-dependent fashion. (Torres et al. 2001). These studies are of considerable
ATP increases NE uptake at lower concentrations (0.1 lM) importance as they focus on the likelihood of NET surface
and decreases NE uptake at higher concentrations (1 lM). expression and localization being coupled to NE release.
The effects of ATP on altering NE uptake levels are greater in Syntaxin 1A, a SNARE protein, plays an important role in
buffers containing Ca2+ rather than Mg2+. Low concentra- the release of neurotransmitters. Recent evidence suggests
tions of ATP (0.1 lM) stimulate NE uptake in a Ca2+- that SNARE proteins interact with neurotransmitter trans-
dependent fashion in PC12 cells (Hardwick et al. 1989). porters (SERT, glycine, GABA) on the plasma membrane
These finding suggest that a Ca2+ rich environment is a and play an important regulatory role in determining their
requirement for ATP to exert its influence on NET. A non- function (Quick et al. 1997; Beckman et al. 1998; Geerlings
hydrolyzable analog of ATP has no effect on uptake, et al. 2000; Haase et al. 2001). Cells naturally expressing
suggesting that the actions of ATP and ATPcS are mediated NET (SK-N-SH) or cells transfected with NET (CAD hNET)
through an ecto-protein kinase pathway (Hardwick et al. show a decrease in NE uptake following treatment with
1989). antisense oligonucleotides to syntaxin. In addition, treatment
Some of the effects of ATP seen, such as the biphasic of cells with botulinum toxin C1 (BoNT/C1), which blocks
effect, were not noticed in another study involving the same syntaxin 1A association to the NET, decreases NE uptake by
cell line (PC12 cells; Eshleman et al. 1995). In this study, the NET. Using immunofluorescence, SDS-PAGE, and
ATP caused a concentration-dependent increase in uptake of immunoblotting methods, colocalization of syntaxin 1A with
NE rather than a biphasic effect. The ATP concentrations the NET has been demonstrated in noradrenergic varicosities.
ranged from 0.1 to 3 mM. In addition, GTP and UTP were In addition, SDS-PAGE and immunoblotting assays demon-
also effective in stimulating NE uptake. Further, the ATP- strate that syntaxin 1A is physically associated with the NET
induced increase in NE uptake was sensitive to temperature (Sung et al. 2003). Since high concentrations of syntaxin 1A
and dependent on Na+. The buffer media also played a are observed in noradrenergic varicosities, it is logical to
pivotal role in dictating ATP’s actions. ATP’s actions were assume that membrane insertion of NET via syntaxin 1A via
more profound in bicarbonate-buffered media than in HEPES a membrane fusion event could localize NET to the site of
or phosphate buffered media. Similar findings were seen in NE release, improving the efficiency of NE reuptake. SNAP-
ATP-induced reduction in the binding of DMI and cocaine to 25, another protein involved with neurotransmitter release, is
the transporter. This study suggests an altogether different not associated with NET/syntaxin complexes. Interestingly,
route for ATP-induced alterations in transporter function. The the interaction of NET with syntaxin 1A appears to be

 2006 The Authors


Journal Compilation  2006 International Society for Neurochemistry, J. Neurochem. (2006) 97, 310–333
318 P. Mandela and G.A. Ordway

required for protein kinase-induced NET regulation. When HEK-293 cells transfected with cDNAs encoding NET and
the NET/syntaxin 1A complex is disrupted with botulinum PICK1 reveal important information regarding the intimate
toxin, PKC-induced NET regulation is abolished (Sung et al. association between these two proteins (Torres et al. 2001;
2003). Sensitivity of both cytoplasmic and plasma membrane Distelmaier et al. 2004). PICK1 coimmunoprecipitates with
NET/syntaxin 1A complexes to phosphorylation by protein NET regardless of the NET antibody used. When expressed
kinases show that the surface pool of these complexes can be individually, PICK1 and hNET show different localization
destabilized, but not the cytoplamsic pool. These studies sites: NET is expressed predominantly on the plasma
reveal mechanisms by which neurotransmitter and transpor- membrane and PICK1 shows a diffuse intracellular distribu-
ter conservation is fine-tuned. Syntaxin, a protein mediating tion. Co-expression of both the proteins show a strong
release-induced insertion of the NET into the plasma colocalization and formation of clusters similar to that of
membrane links NET function to the release process. DAT association with PICK1. Primary cultures of rat locus
Dissociation of the syntaxin/NET complex would be expec- coeruleus neurons also demonstrate an association between
ted to result in a delayed clearance of the transmitter (Sung PICK1 and NET (Torres et al. 2001). Although association
et al. 2003). The NET has been shown to be colocalized with and colocalization of NET with PICK1 have been observed,
SNAP-25 and synaptophysin at axonal varicosities, also there is no clear-cut evidence showing that the association is
consistent with the link of NET function to release (Schroeter related to regulation of transporter function or to pharmaco-
et al. 2000). However, NET-immunoreactivity is also found logical properties of the NET. Since PICK1 associates with
in intervaricose segments of axons (Schroeter et al. 2000). DAT, PICK1 could regulate NET in a manner similar to DAT.
Interestingly, DAT immunolabeling in rat striatum has been PICK1 interacts with DAT in a positive manner by enhancing
shown in plasma membranes of axons and their terminals but DAT uptake activity in both mammalian cells and neurons
not at the active sites of release, indicating that DAT is not (Torres et al. 2001). Given the unknown mechanisms
localized to the immediate area of dopamine release (Hersch involved in inhibitor-induced regulation of the NET (see
et al. 1997). However, this same study also reported that in below), it is tempting to speculate that these drugs may work
rat substantia nigra, DAT was present close to the active zone by disrupting intimate relationships between the NET and
and most likely facilitates rapid uptake of dopamine. From proteins associated with intracellular trafficking.
these findings it has been speculated that transporter
localization to active release zones may be cell type- or
Regulation of the NET by neurotransmitters and
region-dependent.
hormones
PDZ domain-containing PICK1 protein NE and other neurotransmitters also play an important role in
PDZ domain-containing proteins play a role in cell signaling regulating the membrane expression of the NET and its
and membrane trafficking of proteins (see Table 1). PDZ function. Animal and cell culture studies have provided the
domains are interacting modules of globular proteins that evidence for these regulatory pathways. A few neuro-
play a crucial role in directing proteins to the cell membrane transmitter regulatory systems are discussed here and are
and organizing proteins to form supramolecular signaling summarized in Table 1. The regulation of the NET produced
molecules (Fan and Zhang 2002). The synaptic proteins, by NE itself is considered in the section below discussing
PICK1, are PDZ domain-containing proteins known to the effects of substrates.
interact with monoamine transporters. The PICK1 family is
demonstrating increasing importance in the targeting and Acetylcholine
clustering of receptors and ion channels. The DAT is known Acetylcholine (ACh) robustly decreases (by 90%) NE
to bind to PICK1, forming clusters and colocalizng with uptake in isolated guinea pig chromaffin cells. ACh alters
PICK1 in native dopamine-expressing neurons, as well as in Km, without altering the Vmax of the transporter (Role and
NET-transfected cell lines. An important prerequisite for Perlman 1983). Recent studies using SK-N-SH cells have
PDZ domain–mediated interactions is a consensus sequence shown that muscarinic receptor agonists can regulate trans-
located in the carboxy terminus of the interacting proteins. porter function via a PKC dependent pathway (Apparsunda-
NET has sequence similarities to the DAT and the NET ram et al. 1998a). ACh may work through a similar
carboxy terminus has a consensus sequence for a class II mechanism in isolated guinea pig chromaffin cells, although
PDZ domain binding, making the NET a likely candidate for further studies are required to prove this in these cells.
regulation by PICK1. In fact, the carboxy terminus of the Nicotine, working through cholinergic nicotinic receptors,
NET has been shown to interact with PICK1 in a yeast two- affects NET function. High concentrations of nicotine
hybrid system. PICK 1 interaction with NET is totally (5 mM) reduce uptake in synaptosomes prepared from rat
eliminated when the last three residues of the carboxy hippocampus and hypothalamus. In contrast, treatment with
terminus are deleted (Torres et al. 2001; Distelmaier et al. lower concentrations of nicotine produces a small increase in
2004). uptake (Balfour 1973).

 2006 The Authors


Journal Compilation  2006 International Society for Neurochemistry, J. Neurochem. (2006) 97, 310–333
Regulation of the norepinephrine transporter 319

Angiotensin GABA
Angiotensin II affects blood pressure through actions on the GABA is an inhibitory neurotransmitter present in the
sympathetic nervous system, which uses NE as a neurotrans- nervous system. GABA’s role in altering NET uptake activity
mitter. Angiotensin II activates sympathetic pathways by has been studied in rat pineal gland. GABA decreases NE
regulating several of its components, such as the NET and uptake in a concentration-dependent manner with an IC50 of
transcriptional control of the synthesis and release of NE (Lu about 100 lM (Rosenstein et al. 1990). The physiological
et al. 1996; Yang et al. 1996; Yang and Raizada 1999). relevance of this effect is unknown, since extracellular
Exposure of neuronal cultures (hypothalamus and brain stem) GABA has been estimated to occur at low micromolar
from Wistar Kyoto (WKY) rats to angiotensin II at concen- concentrations (Timmerman and Westerink 1997; Shin et al.
trations as low as 100 nM results in time- and concentration- 2002) and to activate chloride conductance at these low
dependent increases in NE uptake. Angiotensin II stimulates concentrations (Mtchedlishvili and Kapur 2005). GABA’s
increases in NE uptake that reach maximum, i.e. 5-fold above effect on NE uptake is mimicked by muscimol, a GABA type
basal levels, after 30 min of exposure. In contrast to uptake, agonist. GABA’s action on NE uptake is inhibited by
angiotensin II induces a gradual increase in NET mRNA levels bicuculline a GABA antagonist. Bicuculline by itself does
starting at 2 h of incubation with angiotensin II, and reaches a not affect NE uptake. The effects of GABA on NE uptake are
peak (5 · basal levels) after 8 h of incubation (Lu et al. 1996). dependent on the concentration of NE used in the uptake
The AT1 receptor, but not the AT2 receptor, mediates the assay. While GABA decreases uptake at low NE concentra-
effects of angiotensin II on NET function. The acute (minutes tions (0.5 lM), GABA increases NE uptake at higher NE
of exposure) effects of angitensin II are mediated post- concentrations 5 lM (Rosenstein et al. 1990).
transcriptionally and are not PKC-dependent. In contrast, the
effects of longer exposure (hours) to angiotensin II involve Insulin
PKC, cFos and NET gene transcription (Lu et al. 1996). Insulin is a key endocrine hormone regulating glucose
Brains of spontaneously hypertensive (SH) rats express homeostasis in the body. Insulin also stimulates catecholam-
higher levels of angiotensin II, NET mRNA, and NE ine release from catecholaminergic neurons and can affect
compared with WKY rats. Neuronal cultures (hypothalamus the uptake of NE. The effect of insulin on NET function has
and brainstem) show a similar response to angiotensin II been studied in PC12 and SK-N-SH cells. Acute treatment of
treatment, but the relative increase in NE uptake and NET cells with insulin (20 min exposure) decreases NE uptake in
mRNA expression is much larger than that observed in WKY PC12 cells (Figlewicz et al. 1993a). This effect is resistant to
rats. High AT1 receptor gene expression, and parallel reserpine, a drug that depletes both NE and DA, demonstra-
increases in AT1 receptors and AT1 receptor-stimulated ting that the insulin-induced reduction in NE uptake is not
signalling in SH rats (Raizada et al. 1993) provide a possible related to competition of endogenous NE (released by
explanation for the differences seen between the two strains insulin) with radiolabelled NE in the uptake assay. PC12
of rats following angiotensin II treatment. cells treated with IGF-1 (insulin-like growth factor) show an
As noted above, angiotensin II employs more than one increase in uptake rather than a decrease, distinguishing
mechanism to regulate NET function. Furthermore, angio- between the 2 major routes through which insulin exerts its
tensin II can regulate NET function by affecting NET gene cellular effects (insulin receptors, insulin-like growth recep-
expression, and in SH rats can do this via multiple pathways. tors). The insulin-induced decrease in uptake is characterized
cFos antisense oligonucleotide treatment considerably limits by a decrease in Vmax, as a result of a decrease in transporter
the ability of angiotensin II to stimulate NET mRNA number on the cell surface (Figlewicz et al. 1993a). Insulin-
production (Lu et al. 1996). Antisense oligoneucleotide for dependent decreases in NE uptake may be due to direct
MAP kinase attenuates increases in NET gene expression action of insulin at the transporter or through influencing
following chronic treatment with angiotensin II in WKY rat transporter gene expression. Insulin is known to decrease
neuronal cultures. This reduction in NET mRNA expression NET mRNA expression in locus coeruleus neurons of the rat
is also accompanied by reduction in cfos and TH mRNA (Figlewicz et al. 1993b).
(Yang et al. 1996). The phosphatidylinositol-3-OH kinase Insulin’s action on the NET is dependent on the cell line in
(PI3 kinase) and PKB/Akt pathway is another route through which it is studied. Exposure of SK-N-SH cells to insulin
which NET mRNA production is altered by angiotensin II, increases the uptake of NE. Insulin in this cell line works
but in neuronal cultures from only SH rats (Yang and through multiple pathways to increase NET function. The
Raizada 1999). These complex findings suggest that angio- insulin receptor is a membrane bound tyrosine kinase that
tensin II affects NET function through multiple pathways. when activated phosphorylates tyrosine and other substrates
Dysregulation of the NET by angiotensin II likely contributes associated with it. Down stream signaling following insulin
to the pathogenesis of hypertension in SH rats, and receptor stimulation also includes activation of a lipid kinase,
implicates a possible role of angiotensin II-mediated regu- PI3 kinase. SK-N-SH cells treated with tyrosine kinase and
lation of the NET in human hypertension. PI3 kinase inhibitors show a drastic decrease in NE uptake.

 2006 The Authors


Journal Compilation  2006 International Society for Neurochemistry, J. Neurochem. (2006) 97, 310–333
320 P. Mandela and G.A. Ordway

This reveals that constitutive tyrosine kinase and PI3 kinase uptake regulation can be independent of changes in plasma
activity is necessary for basal NE transport activity (Appars- membrane NET expression (Apparsundaram et al. 2001).
undaram et al. 2001), at least in this cell line. Tyrosine kinase
and PI3 kinase inhibitors are not known to have any affinity Natriuretic peptides (NP)
for NET directly, suggesting mediation through intracellular Polypeptide hormones such as atrial natriuretic factor (ANF)
sigalling pathways. Furthermore, whole cell binding experi- (de Bold et al. 2001), brain natriuretic peptide (BNP) (Sudoh
ments reveal that these inhibitors decrease surface expression et al. 1989) and C-type natriuretic factor (CNP) belong to the
of the NET. The findings suggest that basal activities of PI3 family of natriuretic peptides (NP). These peptides constitute
kinase and tryosine kinase regulate cell surface expression of a major class of cardiac hormones that have strong influences
the NET (Apparsundaram et al. 2001). In contrast, insulin- on endocrine regulation of blood volume and vascular tone.
stimulated increases in uptake are independent of changes in ANF and BNP are also expressed outside the cardiac system
cell surface expression of the transporter (Apparsundaram in a wide variety of sites and are believed to act in an
et al. 2001), and therefore may involve an insulin-induced autocrine or paracrine fashion (Casco et al. 2002). CNP is
conformational change in the NET that changes its uptake also abundantly present in the nervous system and is also
activity. produced by vascular endothelium (Furuya et al. 1990;
Possible roles of ERK (extracellular signal receptor- Sudoh et al. 1990; Stingo et al. 1992). ANF exerts hypo-
activated kinase) and p38 MAPK (p38 mitogen-activated tensive effects on the cardiovascular system via its interac-
protein kinase) on insulin-induced regulation of NET tions with catecholamine metabolism and by modulating
function have also been investigated in SK-N-SH cells sympathetic activity (Vatta et al. 1993). ANF produces a
(Apparsundaram et al. 2001). The activity of GLUT 4, a time-dependent increase, reaching a plateau at 30 min, in NE
glucose transporter, is increased via p38 MAPK mechan- uptake via the NET in adrenal medullary slices from male
ism, suggesting a possible connection of p38 MAP kinase Wistar rats. ANF induced increases in NE uptake via NET
with insulin’s action on the NET. Inhibitors of p38 MAP may result from increasing NET availability at the plasma
kinase and ERK enzymes do not affect the basal NE membrane or by increasing NET synthesis (Vatta et al.
uptake, but a p38 MAP kinase inhibitor blocks insulin- 1993). Both BNP and CNP stimulate NE uptake via NET in
dependent increases in uptake. In contrast, ERK inhibition hypothalamic slices from male Wistar rats in both concen-
does not affect insulin’s action on the NET (Apparsundaram tration- and time-dependent fashions. The concentration
et al. 2001). required to produce an increase of NE uptake is 100-fold
The effect of insulin on the NET in SK-N-SH cells is also higher for BNP (100 nM) as compared to CNP (1 nM). (Vatta
mediated, at least in part, through a Ca2+-dependent pathway et al. 1996). Non-neuronal uptake of NE is unaffected by NF
(Apparsundaram et al. 2001). Uptake assays performed in peptides, although a specific effect on NET vs. other Na+-
the absence of extracellular Ca2+, or in the presence of dependent transporters has not been ruled out. Clearly
BAPTA/AM (an intracellular Ca2+ chelator) in a Ca2+- natriuretic peptides influence NE uptake, although the
containing buffer, robustly decreases insulin’s influence on cellular mechanisms mediating these effects are unknown.
uptake. Thapsigargin, an inhibitor of the sarcolemmal Ca2+ Further study of these mechanisms could help in under-
pump that depletes intracellular stores of Ca2+, decreases standing the intricate relationship between natriuretic pep-
insulin’s ability to stimulate NE uptake in SK-N-SH cells. tides and the cardiovascular system.
These findings suggest that the insulin-dependent increase in
uptake is secondary to an insulin-triggered influx of Ca2+ Nerve growth factor (NGF)
across the plasma membrane. Insulin does not release Ca2+ Exposure of PC12 cells to NGF for 24 h decreases NE
from intracellular stores, but the Ca2+ requirement for uptake. In contrast, if PC12 cells are grown in serum-free
insulin’s action is met through Ca2+ entry via voltage- media and exposed to NGF, NE uptake is slightly increased
dependent Ca2+ channels. The study by Apparsundaram and after a 24 h exposure to NGF, and then there is robustly
coworkers emphasizes the role of protein phosphatase2A decreased uptake after 96 h of exposure (Ikeda et al. 2001).
(PP2A) in NET regulation. Protein phosphatases were shown In serum-containing medium, NGF exposure decreases
to associate with NET (see above) and dictate NET’s uptake. Kinetic studies of uptake performed after 48 h of
phosphorylation state. This association between NET and NGF treatment show a decrease in Vmax without altering Km.
phosphatases is critical for insulin’s actions on NET since In addition to these effects, NGF also decreases NET mRNA
this NET–phosphatase association is vital for normal func- expression in PC12 cells. The decrease in mRNA levels
tion of NET. Further this study also demonstrates that NE could contribute to a decrease in availability of new NET
uptake function can be increased without altering NET protein thus resulting in the decrease of NE uptake (Ikeda
plasma membrane expression (Apparsundaram et al. 2001). et al. 2001). In bovine adrenal chromaffin cells, NGF
Together, these findings demonstrate that regulation of exposure increases NE uptake and increases NET mRNA
NET function by insulin involves multiple pathways and NE expression (Wakade et al. 1996). Other growth factors like

 2006 The Authors


Journal Compilation  2006 International Society for Neurochemistry, J. Neurochem. (2006) 97, 310–333
Regulation of the norepinephrine transporter 321

EGF have also been shown to affect (decrease) NE uptake in However, these compounds change the saturation kinetics of
PC12 cells without altering NET mRNA expression (Ikeda NE at the NET, although there are differences in the specific
et al. 2001). It is interesting to note that NGF’s effect on NE kinetic effects of bisphenol A and 17b-estradiol (Toyohira
uptake and NET mRNA levels varies with presence and et al. 2003). Hence, the effects of these compounds may be
absence of serum (Ikeda et al. 2001). The mechanism by mediated by direct modification of NET activity via modu-
which NGF affects the NET is poorly understood. latory binding sites on the transporter, rather than through
regulation of transporter expression or activity via intracell-
Nitric oxide ularly mediated pathways.
Nitric oxide (NO) and its congeners are key signaling
molecules in the cardiovascular system and have a putative
Regulation by Substrates and Inhibitors Acting at the
role as neurotransmitters in the nervous system. Regulation
NET
of noradrenergic signaling by NO and its congeners, nitrogen
oxides (NOx), have been studied in PC12 cells and rat The study of the regulation of the NET by substrates and
sympathetic neurons in primary culture. PC12 cells treated inhibitors of the NET is a growing area of research because
with NOx inhibit the uptake of NE. NOx signaling includes the NET is a site of action of drugs that are typically
both cGMP-dependent and independent pathways, but administered chronically. Many drugs can affect the NET
studies of the molecular mechanism of NOx-induced acutely, usually as a result of binding to the NET or
decreases in NE uptake indicate that the cGMP-independent disrupting ionic gradients. The studies reviewed below and
pathway is the most likely mediator. The cGMP analogues, summarized in Table 3 consider the effect of continuous or
8-BrcGMP and LY-83583, as well as inhibitors of soluble repeated exposure of the NET to NET substrates and NET
guanylyl cyclase, do not affect NOx-induced decreases in inhibitors on NET function and expression. Since these drugs
uptake (Kaye et al. 1997). It should be noted that besides bind the NET, a criterion for discussing the regulatory effects
cGMP signaling, NO inhibits Na+/K+-ATPase, a key pump in of these drugs is that the substrate or inhbitor is absent
maintaining the uptake process. Also, NO may exert its (except for NE) at the time that function and/or expression is
influence through a direct action on the transporter. This view assessed. The effect of drugs, treatments, or stress, which are
is supported by findings wherein treatment with 1 mM known to modulate NE, on NET expression are also
cysteine significantly decreases the effect of S-nitroso- reviewed below and summarized in Table 4.
acetylpencillamine (SNAP), a NOx donor, on uptake. This
observation suggests nonenzymatic reactions of NOx with Inhibitors
sulfhydryl groups. The NET has many cysteine residues that One of the earliest antidepressant drugs was the tricyclic
are candidates for trans-nitrosation (Kaye et al. 1997). In antidepressant, imipramine. Many early antidepressants,
SK-N-SH cells, pretreatment with the nitric oxide synthase some of which are still in use today, are variations on this
(NOS) inhibitor, l-NMMA, fails to alter methacholine- original structure, including DMI, amitriptyline, and nor-
induced reductions in uptake. SNAP treatments in SK-N- triptyline. These drugs bind to the NET and block the
SH cells show an increase rather than a decrease in uptake reuptake of NE. The pharmacological action of these drugs at
(Apparsundaram et al. 1998a). Overall, these findings dem- the synapse is rapid, i.e. uptake is inhibited upon binding to
onstrate that more research is needed to understand molecular the transporter. However, the therapeutic effect of these drugs
mechanisms of NO-induced regulation of the NET, and to in the treatment of depression requires several weeks of daily
elucidate its physiological significance. treatment. Thus, while uptake inhibition has been a valuable
index for identifying potential antidepressants, studies
Ovarian hormones designed to understand the relationship between biology
Estrogen plays a vital role in maintaining homeostatic and therapeutic action have concentrated on the effects of
conditions in the female reproductive system (Couse and long-term treatment with antidepressant drugs. Both acute
Korach 1999). Estrogens exert their effect on cell systems in and chronic exposure to tricyclic antidepressants decrease
both a genomic and a non-genomic way (McEwen 1991; uptake of NE, at least in part, by blockade of NET function.
Couse and Korach 1999). Environmental estrogens are In addition to inhibition of uptake as a result of binding to the
another class of substances that do not share the same NET, chronic treatment (e.g. 21 days) of rats with DMI
chemical structure or steroidal properties of estrogen, but results in a decrease in NET binding sites in specific brain
have similar actions. One such environmental estrogenic regions (Bauer and Tejani-Butt 1992; Hebert et al. 2001).
compound is bisphenol A (Krishnan et al. 1993). Bisphenol The effect of DMI treatment in vivo on NET expression is not
A, 17b-estradiol, and p-nonyl-phenol decrease NE uptake in seen following a single dose of DMI (Bauer and Tejani-Butt
bovine adrenal medullary cells following brief exposures 1992). The down-regulation of the NET produced by
(30 min) (Toyohira et al. 2003). The mechanism by which repeated DMI administration is accompanied by a reduction
these compounds influence NE uptake is not understood. in uptake (Benmansour et al. 2004). In addition, the

 2006 The Authors


Journal Compilation  2006 International Society for Neurochemistry, J. Neurochem. (2006) 97, 310–333
322 P. Mandela and G.A. Ordway

structurally dissimilar NET inhibitor, reboxetine, also down- While DMI does not produce effects on NET binding
regulates NET (as measured by radioligand binding) follow- following a single dose in rats, in vitro studies show an acute
ing its chronic administration to rats (Gould et al. 2003), regulation of the transporter by antidepressants. Ordway and
while paroxetine, a SERT inhibitor with low affinity for the coworkers (Zhu and Ordway 1997; Zhu et al. 1998)
NET, does not affect NET binding or function (Benmansour originally reported that short exposures (4 h) to DMI could
et al. 2004). The ability of DMI and reboxetine to down- down-regulate the NET. However, more recently these
regulate the NET, an effect that is functionally similar to authors (Ordway et al. 2005) have demonstrated that in vitro
acute blockade of the NET, has raised speculation that this studies of DMI’s effects on NET function and binding can be
regulatory effect may contribute to the utility of transporter complicated by the tenacity at which DMI is retained in cell
inhibitors in the treatment of depression. membranes (Zhu et al. 2004). This property of DMI, and
In vivo experiments designed to address mechanisms of presumably other lipophilic psychotherapeutic compounds,
DMI-induced regulation of the NET reveal an ambiguous creates an obstacle to the study of the acute effects on
role of changes in NET gene expression on DMI-induced regulation of transporter function. In fact, it is speculated that
changes in the NET. Repeated treatment of rats with retention of transporter inhibitors in the brain following
antidepressants for 14 days, but not 3 days, down-regulates cessation of treatments to rats, could potentially complicate
NET-immunoreactivity in the locus coeruleus, as measured binding assays (see Zhu et al. 2004 for discussion). This
by Western blotting (Zhu et al. 2002). However, NET complication arises because of the membrane retention of
mRNA levels have been reported to increase (Szot et al. DMI (and nisoxetine) and because these drugs have a very
1993) or decrease (Zhu et al. 2002) after short-term high affinity for the very protein being studied, i.e. the NET.
treatment of DMI to rats, and increase after 14 days Uptake and binding assays are performed in very small
treatment (Zhu et al. 2002). Hence, changes in NET mRNA volumes and redistribution of DMI from membranes to the
levels in the locus coeruleus following DMI treatment do not small volume of the reaction mixture can result in pharma-
parallel changes in NET binding. cologically relevant concentrations of NET inhibitor in the
In an effort to elucidate the molecular mechanisms assay (Zhu et al. 2004; Ordway et al. 2005). The retention
involved in inhibitor-induced regulation of the NET, the and redistribution of DMI hampers evaluation of NET by
effect of exposure to NET inhibitors on NET expression and certain radioligand binding assays and uptake assays, but is
function has been studied recently in in vitro cell culture not expected to interfere with immunologically detected NET
systems. Since the inhibitors themselves affect NET function using methods such as Western blotting. It is noted, however,
acutely, it is important to note that the functional (uptake) that a lack of an effect of DMI on antibody binding to the
effects of the drugs are measured under conditions that NET has not been specifically demonstrated. Ordway et al.
assume that the drug has been washed from the cells (2005) recently developed an assay to evaluate binding and
following the exposure (see below for pitfalls in this uptake at time points as short as 2 h following DMI
assumption). Exposure (3 days) of both PC12 cells (produce exposure, without the interference of residual drug. Using
NE and NET) and 293-hNET cells (HEK cells transfected these methods, exposure to DMI for 16 h is necessary before
with hNET, but do not produce NE) to DMI produces reductions of NET function (uptake) in the absence of DMI
reductions in uptake capacity, radioligand binding to the can be measured.
NET, and NET-immunoreactivity (Zhu and Ordway 1997; The molecular mechanisms responsible for NET inhibitor-
Zhu et al. 1998). These effects of DMI appear to be specific induced regulation of the NET expression and function are
to NET binding antidepressants, since similar exposure of only beginning to be elucidated. One possible mediator of
cells to the selective serotonin uptake inhibitor, citalopram, this event could be NE itself. Inhibition of NE uptake by
does not affect NET binding. Furthermore, exposure to NET inhibitors could result in extracellular actions of NE, i.e.
nisoxetine, a NET inhibitor structurally dissimilar from DMI, through receptors for NE that lead to intracellular events
produces effects on the NET very similar to DMI. NET mediating NET down-regulation. However, treatment of
binding and NET immunoreactivity slowly recovers (over the PC12 cells with concentrations of NE as high as 1 mM did
course of days) to normal levels following the removal of not affect radioligand binding to the NET (Zhu and Ordway
DMI. Hence, NET regulation in response to NET inhibitor 1997). Furthermore, exposure of 293-hNET cells, which
exposure in cell culture systems appears to be similar to express the NET but do not synthesize NE, to DMI or
NET regulation as induced by prolonged exposure to NET nisoxetine down-regulates the NET just as it does in PC12
inhibitors in vivo. It is worth noting however, that if NET cells that synthesize NE. Together, these findings strongly
binding by the inhibitor were the only factor contributing to suggest that NE does not play a role in the down-regulation
regulation in vivo during NET inhibitor treatment, then it of the NET produced by NET inhibitor exposure.
becomes difficult to explain why the NET is not regulated Several lines of evidence suggest that down-regulation of
similarly (Bauer and Tejani-Butt 1992) in all brain regions the NET by DMI occurs as a consequence of binding to the
after chronic NET inhibitor treatment. transporter, rather than as a result of secondary effects.

 2006 The Authors


Journal Compilation  2006 International Society for Neurochemistry, J. Neurochem. (2006) 97, 310–333
Regulation of the norepinephrine transporter 323

Similar time-dependent and concentration-dependent down- decrease radioligand binding to the NET in postnatal rat
regulations of the NET are produced by DMI exposure in rat cardiac tissue (Zhao and Sun 2004), and to have no effect on
PC12 cells (Zhu and Ordway 1997), human SK-N-SHSY5Y NET protein in postnatal rat brain (Shearman and Meyer
cells (Zavosh et al. 1999), and in non-catecholaminergic 1999). Benmansour et al. (1992) and Belej et al. (1996)
293-hNET cells (Zhu et al. 1998). Also, the structurally reported that repeated cocaine administration to rats does
dissimilar NET inhibitors, nisoxetine and reboxetine, down- produce major effects on radioligand binding to the brain
regulate the NET (Zhu and Ordway 1997; Gould et al. NET, although both increases and decreases have has been
2003). Although these data suggest that regulation of the demonstrated in very discrete brain regions (Belej et al.
NET by inhibitors is a basic property of inhibitors, it is 1996). Chronic self-administration of cocaine in non-human
emphasized that cocaine, an inhibitor of the NET, does not primates results in an increase in radioligand binding to the
down-regulate the NET in cultured cells (Zhu et al. 2000). In NET in several brain regions (Macey et al. 2003; Beveridge
addition, exposure of NET-expressing cells to amphetamine, et al. 2005). In the human brain, as assessed postmortem in
a substrate for the NET, also results in down-regulation of the chronic cocaine users, cocaine has been reported to increase
NET (Zhu et al. 2000). Hence, it can be speculated that NET protein (Mash et al. 2005). Hence, while studies of the
uptake inhibition by itself is not sufficient to initiate the effects of antidepressant inhibitors of the NET consistently
process of down-regulation. Rather, it seems to be some demonstrate down-regulation of NET protein and/or func-
specific domain(s) of the NET protein engaging binding to tion, studies examining chronic cocaine exposure have
the ligand that is required for down-regulation. Oligomeri- yielded incongruent findings. Exposure in vivo to cocaine
zation is another important process that plays a vital role in appears to increase NET protein. Since this effect does not
NET function (Kocabas et al. 2003) and drug-induced occur in cultured cells expressing the NET and lacking
disruption of oligmerization could conceivably contribute synaptic contacts, the effect of cocaine on the NET in vivo
to disruption of transporter function and to its ultimate down- may be mediated through actions of cocaine other than direct
regulation. binding to the NET.
The possibility that DMI-induced regulation of the NET is Overall, numerous investigators have demonstrated that
mediated at the level of changes in NET mRNA has been specific NET ligands (e.g. DMI, nisoxetine, amphetamine),
investigated using cultured cells. In 293 h-hNET cells, DMI but not all NET inhibitors (e.g. cocaine), down-regulate the
exposure, at times (e.g. 3 days) and concentrations (e.g. NET. The specific molecular mechanisms responsible for
500 nM) that down-regulate NET protein, does not affect ligand-induced regulation have not been fully elucidated.
NET mRNA levels (Zhu et al. 1998). However, it is worth In vivo and in vitro studies demonstrate that the mechanisms
noting that this transfected cell line has a very active involved in DMI-induced regulation of NET protein are
promoter driving NET gene transcription. In the SK-N- complex, may be brain region-specific, and are not tightly
BE(2)M17 cell line, which expresses native hNET, exposure associated with NET gene transcription. Preliminary studies
to DMI for 3 days reduces levels of NET mRNA, whereas in this laboratory demonstrate that initially, DMI induces a
exposure for 14 days increases NET mRNA. Despite the redistribution of NET away from the plasma membrane
fluctuation in mRNA levels following DMI exposure, NET surface (Pan et al. 2002), and that DMI’s effects on the
protein levels are down-regulated following 3 and 14 days of transporter are unaffected by inhibitors of several protein
DMI exposure. Similar experiments have been performed in kinases and protein phosphatases (Pan et al. 2003). Antide-
the NET expressing cell line SK-N-SHSY5Y. Cells treated pressant-block of the NET antagonizes NET function (reup-
with DMI for 1 and 3 days demonstrate a decrease in take) and may give the NET a non-functional tag. Unknown
radioligand binding to the NET. After 1 day of DMI cellular proteins may recognize this inactivity and work
exposure, there is no effect on NET mRNA, but a 3 day together to internalize the NET protein in order to recycle
exposure decreases NET mRNA. Since a decrease in mRNA new functional NET proteins to the surface. This process
content followed a decrease in protein content, the change in would be expected to take some time and could explain the
mRNA content is at best a contributor to reduce NET protein, lack of immediate recovery following antidepressant expo-
but not the initial cause of loss of NET protein. sure. Unfortunately, the intracellular mediators for this
Cocaine is also a NET inhibitor and a number of studies process remain elusive.
using different treatment paradigms have investigated the
effects of cocaine on the NET. Continuous exposure of Substrates
293hNET cells to cocaine causes a small reduction in uptake Studies using 293hNET cells have shown that NET function
and no effects on NET protein (Zhu et al. 2000). Studies (uptake) is reduced as a result of exposure to NE followed by
examining the effect of repeated treatment of rats with cell washing. This effect of NE is concentration-dependent
cocaine have revealed conflicting findings. Prenatal exposure (Zhu and Ordway 1997). NE’s effect on the NET is opposite
to cocaine has been reported to increase radioligand binding to that observed for serotonin and its cognate monoamine
to the NET in the placenta (Shearman and Meyer 1999), to transporter, the SERT. Blakely and coworkers have demon-

 2006 The Authors


Journal Compilation  2006 International Society for Neurochemistry, J. Neurochem. (2006) 97, 310–333
324 P. Mandela and G.A. Ordway

strated that SERT regulation is activity dependent, such that et al. 1997). Likewise, repeated exposure of rats to cold
the presence of serotonin increases transporter activity stress reduces NE uptake capacity and NET binding sites in
(Ramamoorthy and Blakely 1999). Recently, Mao et al. brown adipose tissue (King et al. 1999). An elevation of
(2004) have demonstrated that the reduction in NET activity NET gene expression has been reported following exposure
(reduced Vmax) produced by exposure (24 h) of PC12 cells to of rats to repeated restraint stress, but this effect is not
NE is likely mediated by oxidative stress pathways (Mao observed in the locus coeruleus (Rusnak et al. 2001), the
et al. 2004). NE produces cellular oxidative stress that is principal source of brain NE. One human postmortem study
enhanced by copper, and exposure to NE plus copper measuring binding to the NET reports reduced NET in the
increases the magnitude of NE-induced reductions in NET locus coeruleus in subjects with major depression, a stress-
activity and NET protein. NE appears to reduce NET activity, related disorder, as compared to psychiatrically normal
at least in part, by decreasing N-glycosylation of the NET, an control subjects (Klimek et al. 1997). Together, these studies
effect induced by endoplasmic reticulum stress pathways suggest that repeated stress decreases NET protein and
induced by NE-derived oxidative metabolites (Mao et al. function, and these effects are consistent with the effects of
2005). pharmacological depletion of NE on the NET as discussed
Amphetamine is a psychostimulant that induces neuro- above. The mechanisms by which stress regulate the NET
transmitter release from neuron terminals. It also is a may be related to reduced activity of the transporter in
substrate for the NET. Amphetamine works through intra- response to stress-induced reduction in substrate. Alternat-
cellular Ca2+ and protein kinase-dependent mechanisms to ively, stress hormones such as CRF and glucocorticoids may
reverse transporter function. Amphetamine-mediated neuro- mediate changes in the expression of the NET through as yet
transmitter release through the NET is attenuated by protein unidentified signaling cascades. Clearly, a considerably
kinase inhibitors and BAPTA/AM an intracellular calcium amount of research effort has to be focused on this issue to
chelator (Kantor and Gnegy 1998; Kantor et al. 2001). As understand how stress regulates the NET and whether this
mentioned above, exposures of cultured NET-expressing effect is mediated by change in synaptic concentrations of
cells to amphetamine results in a down-regulation of NET NE or through other mechanisms.
and its function (Zhu et al. 2000). The molecular mecha-
nisms responsible for this effect have not been revealed.
Regulation of the NET by other drugs
Treatments that reduce or elevate endogenous NE regulate
the expression of the NET. These treatments might produce Certain drugs affect NET function through mechanisms that
their effects through actions of the substrate (NE) or through are largely unknown. The regulatory effect of these drugs on
other unknown direct or indirect effects on the NET. The NET function, following continuous exposure of the drugs to
earliest study demonstrated that manipulation of NE levels the NET, have been studied in some cases. These effects are
by administration of reserpine (depletes NE and other discussed here and summarized in Table 2.
monoamines) or monoamine oxidase inhibitors (elevate
NE) to rats results in reciprocal effects on radioligand Anesthetics
binding to the NET in the cerebral cortex (Lee et al. 1983). Anesthetics both general and local exert their effect by
Reserpine treatment reduces radioligand binding to the NET, altering neurotransmission. Ketamine has anesthetic and
whereas monoamine oxidase inhibitor treatment increases analgesic properties, and also has a wide variety of
radioligand binding to the NET. Subsequently, others have psychotropic and sympathomimetic effects. Ketamine acts
shown that depletion of NE in rats by either reserpine on numerous targets including glutamatergic, opioidergic,
(Cubells et al. 1995) or a-methyl-p-tyrosine treatment (Xiao and monoaminergic systems. The sympathomimetic effects
et al. 1995) reduces NET mRNA levels in the locus of ketamine result from its inhibitory actions on NE reup-
coeruleus. take (Taube et al. 1975; Graf et al. 1995). However, the
Stress induces the release of NE in the brain and chronic actions of ketamine on the NET are complex and vary with
stress can deplete NE. Hence, it is interesting to consider the the length of time of exposure to ketamine. Acute exposure
effects of stress on NET expression in the context of potential of bovine adrenal medullary cells to ketamine (30 min)
substrate-mediated effects. Exposure of rats to acute stress decreases NE uptake, characterized by decreased Vmax and
does not affect radioligand binding to the NET (Zafar et al. no apparent change in Km (Hara et al. 1998). In stark
1997) or NET gene expression (Sands et al. 2000). In these contrast, exposure of bovine adrenal medullary cells to
studies, NET binding or gene expression was measured ketamine for longer than 3 h increases NE uptake (Hara et al.
shortly after the stressor. When the NET is measured several 2002; Table 2). The effects of the longer exposure are
days after acute exposure of mice to a stressor, NET binding accompanied by a change in the Vmax without changes in
has been found to be increased (Hwang et al. 1999). In Km. The effects of ketamine are modestly attenuated in the
contrast to acute stress, repeated restraint stress decreases presence of the RNA polymerase inhibitor, actinomycin D,
NET binding sites in the brain (Tejani-Butt et al. 1994; Zafar and blocked in the presence of the ribosomal protein

 2006 The Authors


Journal Compilation  2006 International Society for Neurochemistry, J. Neurochem. (2006) 97, 310–333
 2006 The Authors
Table 2 Regulation of the NET by neurotransmitters, hormones, and drugs

Messenger Exposure Agent Cell Surface


System Cell type Time NE Uptake NET Kinetics Expression NET mRNA References

2+
Methacholine SK-N-SH cells 30min Ca , PKC fl « Km fl Vmax fl – Apparsundaram et al.
(1998a)
Insulin SK-N-SH cells 60 min PI3 Kinase › « Km › Vmax « – Apparsundaram et al.
(2001)
PC 12 cells (reserpine treated) 20 min – fl ? fl – Figlewicz et al. (1993a)
Rat locus coeruleus – – – – – fl Figlewicz et al. (1993b)
Angiotensin II Primary culture of 30 min, MAP kinase, › – – › only with Lu et al. (1996);
WKY rat neurons 4h cfos 4 h treatment Yang and Raizada (1998)
Primary culture of 30 min, MAP kinase, › – – › only with Lu et al. (1996);
SH rat neurons 4h cfos 4 h treatment
ATP PC12 cells 5 min ? › « Km › Vmax – – Eshleman et al. (1995)
PC12 cells 3 min – › (0.1 lM) – – – Hardwick et al. (1989)
only in Ca
KRH buffer,
fl (1 lM)
ATPcS PC12 cells 30 min – › « Km › Vmax – – Hardwick et al. (1989)
pre-treatment
ANF Adrenal medullary slices (Wistar rats) 60 min – › – – – Vatta et al. (1993)
BNP, CNP Hypothalamus slices (Wistar rats) 60 min – › – – – Vatta et al. (1996)

Journal Compilation  2006 International Society for Neurochemistry, J. Neurochem. (2006) 97, 310–333
Ketamine Bovine adrenal medullary cells 12 h – › « Km › Vmax › NP › Hara et al. (2002)
Propofol Bovine adrenal medullary cells 6–24 h – › – › NP › Hara et al. (2000)
Clozapine Bovine adrenal medullary cells 2–168 h – fl (2–48 h) fl Vmax (6 h) fl NP (6 h) › (12–96 h) Yoshimura et al. (2001)
› (72–168 h) › Vmax (96 h) › (96 h)
Regulation of the norepinephrine transporter
325
Table 3 Regulation of the NET by substrates and inhibitors of the NET

Messenger NET protein (NP)*


Cell type or Time period of system or Cell Surface
Agent Species exposure involved NE uptake NET Kinetics Expression (CSE) NET mRNA References

Amphetamine 293hNET cells 3 days – fl « Km fl Vmax fl NP « Zhu et al. (2000)


326 P. Mandela and G.A. Ordway

Citalopram PC12 cells, 3 days – – – « NP – Zhu and Ordway (1997)


293-hNET Zhu et al. (1998)
Cocaine Rat, pregnant 3 days – – – › NP (placenta) « – Shearman and Meyer (1999)
dams NP (fetal brain)
Rat, pregnant Perinatal – fl (myocardial) – « NP (myocardial) – Zhao and Sun (2004)
dams
Rat 10 days – – – « NP (brain) Benmansour et al. (1992)
Rat 4 week – – – ›, fl, « NPdepending – Belej et al. (1996)

 2006 The Authors


on brain region
Non-human 100 days – – – › NP (brain) – Macey et al. (2003)
primate Beveridge et al. (2005)
Human, Chronic – – – › NP (brain) – Mash et al. (2005)
postmortem
293hNET cells 3 days – fl (small change) « Km « Vmax « NP « Zhu et al. (2000)
Desipramine Rat 1 day, – – – fl (brain; 21 days) – Bauer and Tejani-Butt (1992)
21 days i.p. Hebert et al. (2001)
Rat 3 week, 6 week – fl – fl (brain) « Benmansour et al. (2004)
PC12 and 3 days – fl – fl NP – Zhu and Ordway (1997)
293hNET cells 1–21 days – fl (3 days) – fl (‡ 2 days) NP « Zhu et al. (1998)
PC12 cells 4 h)3 days – fl (‡ 16 h) – fl (‡ 3 days) NP – Ordway et al. (2005)
293-hNET fl ((‡ 16 h) CSE
Nisoxetine PC12 3 days – – – fl NP – Zhu and Ordway (1997)
Norepinephrine PC12 3 days – – – « – Zhu and Ordway (1997)
PC12 24 h Free radicals fl « Km fl Vmax fl NP fl CSE « Mao et al. (2004) Mao
et al. (2005)
Paroxetine Rat 3 week, 6 week – « – « – Gould et al. (2003)
Benmansour et al. (2004)
Reboxetine Rat 3 week, 6 week – – – fl – Gould et al. (2003)

*NET protein measured by binding of [3H]nisoxetine or with NET antibody using crude membrane homogenates.

Journal Compilation  2006 International Society for Neurochemistry, J. Neurochem. (2006) 97, 310–333
 2006 The Authors
Table 4 Regulation of the NET by other drug treatments, stress, or stress-related disease

NET protein (NP)* or


Cell type or Time period of NET Cell Surface
Agent/Illness Action Species exposure NE uptake Kinetics Expression (CSE) NET mRNA References

Iproniazid MAO inhibitor Rat 18 days fl** › NP (brain – Lee et al. 1983;
Pargyline MAO inhibitor Rat 10, 18 days fl** – › NP (brain) – Lee et al. (1983)
Reserpine Monoamine Rat 1–10 days fl – fl (brain) – Lee et al. (1983)
depletion Rat 1 day – – – fl (brain and Cubells et al. (1995)
adrenal)
a-methyl-p-tyrosine Catecholamine Rat 1, 3, 7 days – – – fl 3, 7 days Xiao et al. (1995)
depeletion (brain)
Restraint Stress Rat 30 min – – – « Sands et al. (2000)
Rat 2 h/day for – – fl 7 days (brain) – Zafar et al. (1997)
1 or 7 days
Mice 2h › NP (brain) – Hwang et al. (1999)
Rat 21 days, including – – fl (brain) – Tejani-Butt et al. (1994)
other stressors
Rat 2 h/day for 41 days – – – › (isolated Rusnak et al. (2001)
changes)
Cold Stress Rat 2–7 days fl 7 days – fl NP 3–7 days – King et al. (1999)
(adipose)
Major depression Stress-related Human – – – fl (brain) – Klimek et al. (1997)
Citalopram Serotonin PC12 and, 3 days – – « NP – Zhu and Ordway (1997)

Journal Compilation  2006 International Society for Neurochemistry, J. Neurochem. (2006) 97, 310–333
uptake inhibitor 293-hNET cells Zhu et al. (1998)
Paroxetine Serotonin Rat 3 week, 6 week « – « NP – Gould et al. (2003)
uptake inhibitor Benmansour et al. (2004)

*NET protein measured by binding of [3H]nisoxetine or with NET antibody using crude membrane homogenates.
**These drugs have modest acute inhibitory effects on uptake, and were considered by the authors to be present in the uptake assay (Lee et al. 1983).
Regulation of the norepinephrine transporter
327
328 P. Mandela and G.A. Ordway

synthesis inhibitor cycloheximide. Following a 24 h incuba- ter and either inhibit its function, essentially as an antagonist,
tion with ketamine, NET mRNA expression increases (Hara or alter its function by acting as a competitive substrate.
et al. 2002). The effect of ketamine is not stereo-selective, Besides these functional changes in the NET as a result of
that is both (+) ketamine and (–) ketamine have similar binding to the NET, drugs, hormones, and other transmitters
effects on NE uptake (Hara et al. 2002). Although protein can regulate the function of the NET through interactions
synthesis clearly plays a role, the mechanism by which with other receptors or binding sites, either by activation of
ketamine affects transporter function is largely unknown. It is known second messenger systems, or by activating or
intriguing to consider the possibility that ketamine may be inhibiting other signaling pathways that are currently
affecting the function of the norepinephrine transporter unknown. In addition, repeated or continuous exposure of
through its actions at the NMDA receptor. the NET to substrates or inhibitors regulates the physical
expression of the NET on the cell surface, through as yet
Other anesthetics unknown cellular mechanisms.
Other anesthetics such as propofol, thiamylal and diazepam Although drugs that directly manipulate the function of the
inhibit NE uptake in a concentration-dependent manner in NET have been used for over a half century, it is only within
bovine adrenal medullary cells. Propofol demonstrates the the past 10–15 years that considerable advances have been
highest potency followed by diazepam and thiamylal. In made in understanding how the NET functions at the
addition, propofol exhibits a biphasic effect on NE uptake molecular level and how its function is regulated by repeated
depending on the times of exposure (Hara et al. 2000; or continuous exposure to drugs or hormones. The key
Table 2). These anesthetics exhibit similar properties to those development in studying this transporter was its cloning by
of ketamine in altering NET expression and uptake kinetics. Pacholczyk et al. in 1991. Since then, cellular transfection
Further these findings carry some pharmacological import- studies of the NET across species have paved way for many
ance as it has been demonstrated that propofol and thiamylal important studies yielding information about multiple cellu-
can further increase epinephrine-induced arrhythmias, an lar systems that participate in regulating the transporters.
effect likely to be mediated via the inhibition of NET NET function is regulated by a wide variety of drugs,
function. Furthermore, some of the antinociceptive actions of neurotransmitters, peptides, psychostimulants, temperature,
these anesthetics may be mediated by NE secondary to NET ionic environment, nucleotides and various pathological
inhibition (Hara et al. 2000). conditions. Across the species, phosphorylation is the most
significant intracellular event that dictates NET expression
Clozapine and function, although the number of potential sites for
Clozapine is an atypical antipsychotic that has mixed actions phosphorylation varies from one species to another. Second
on NE uptake. Clozapine has relatively low affinity for the messenger systems and subsequent intracellular signaling
NET, i.e. in the micromolar range (Fernandez et al. 2005). cascades, particularly but not limited to PKC, are intracel-
Exposure of bovine adrenal medullary cells to clozapine has lular triggers that regulate NET surface expression and
biphasic actions on the NET, decreasing NET function function. Besides working through second messenger sys-
initially followed by increasing NET function. Both phases tems, it is interesting to consider the potential for allosteric
of clozapine’s action on the NET require continuous inhibition of the NET. For example, insulin increases NE
exposure and appear to be regulatory in nature, rather than uptake in SK-N-SH cells without altering the membrane
as a result of simply acute binding of clozapine to the expression of NET (Apparsundaram et al. 2001). Though
transporter. Both phases of action of clozapine are accom- allosteric mechanisms have been proposed by various
panied by correlative changes in NET protein, although only investigators, it is not known how allosterism operates at
the longer clozapine-induced increase in NET function is the molecular level, i.e. how agents bring about conforma-
accompanied by both increases in NET protein and NET tional or topographic changes to increase or decrease NET
mRNA (Yoshimura et al. 2001). The unusual actions of function. Another secondary mechanism of regulating NET
clozapine on NET function require further study, particularly function is through altering Na+/K+-ATPase activity or by
with regard to whether the actions of clozapine on the NET otherwise disturbing the ionic gradient required for NET
contribute to its unique actions in the treatment of schizo- function. In addition, altering the oligomerization of the NET
phrenia and whether NET effects extend to other atypical represents another potential regulatory pathway.
antipsychotic drugs. Clearly, most studies of the regulation of NET have
utilized in vitro systems to identify cellular mechanisms. In
order for the field to move forward, more research is needed
Summary
to try to understand how regulation of the transporter affects
The NET plays a major role in modulating transmission at noradrenergic signaling in vivo, and to delineate the physio-
noradrenergic synapses. The function of the NET can be logical consequences of these effects. Given the cell-line
altered by numerous drugs that bind directly to the transpor- dependence of many cellular mechanisms involved in

 2006 The Authors


Journal Compilation  2006 International Society for Neurochemistry, J. Neurochem. (2006) 97, 310–333
Regulation of the norepinephrine transporter 329

regulation, it is important to identify which mechanisms are Beckman M. L., Bernstein E. M. and Quick M. W. (1998) Protein kinase
relevant to the in vivo situation, and to also consider the C regulates the interaction between a GABA transporter and syn-
taxin 1A. J. Neurosci. 18, 6103–6112.
possibility of species-specificity. Recent studies demonstra-
Belej T., Manji D., Sioutis S., Barros H. M. and Nobrega J. N. (1996)
ting that the NET is predominately located in the noradren- Changes in serotonin and norepinephrine uptake sites after chronic
ergic terminal cytoplasm, rather in the plasma membrane, in cocaine: pre- vs. post-withdrawal effects. Brain Res. 736, 287–296.
certain brain regions (Miner et al. 2003) emphasizes the Benmansour S., Altamirano A. V., Jones D. J., SancheZ. T. A., Gould G.
dearth of understanding of the physiological significance of G., Pardon M. C., Morilak D. A. and Frazer A. (2004) Regulation
of the norepinephrine transporter by chronic administration of
NET regulation in vivo. Elucidation of cellular pathways for
antidepressants. Biol. Psychiatry 55, 313–316.
NET regulation in vivo will move the field one step closer to Benmansour S., Tejani-Butt S. M., Hauptmann M. and Brunswick D. J.
developing novel drugs capable of modulating NET activity. (1992) Lack of effect of high-dose cocaine on monoamine uptake
These types of drugs could have unique activities for the sites in rat brain measured by quantitative autoradiography. Psy-
treatment of CNS and PNS disorders, such as depression, chopharmacology (Berl). 106, 459–462.
Beveridge T. J., Smith H. R., Nader M. A. and Porrino L. J. (2005)
attention deficit, cardiovascular, or other disorders where
Effects of chronic cocaine self-administration on norepinephrine
modulation of noradrenergic transmission can yield thera- transporters in the nonhuman primate brain. Psychopharmacology
peutic effects. (Berl) 2005 March 1; [Epub ahead of print].
Blakely R. D. and Bauman A. L. (2000) Biogenic amine transporters:
regulation in flux. Curr. Opin. Neurobiol. 10, 328–336.
Acknowledgements de Bold A. J., Borenstein H. B., Veress A. T. and Sonnenberg H. (2001)
A rapid and potent natriuretic response to intravenous injection of
This work was supported in part by the National Institute of Mental atrial myocardial extract in rats. Reprinted from Life Sci. 28: 89–
Health (MH58211 and MH02031). 94, 1981. J. Am. Soc. Nephrol. 12, 403–409.
Bönisch H. and Brüss M. (1994) The noradrenaline transporter of
the neuronal plasma membrane. Ann. N. Y. Acad. Sci. 733, 193–202.
References Bönisch H., Hammermann R. and Brüss M. (1998) Role of protein
Amara S. G. (1992) Neurotransmitter transporters. A tale of two families. kinase C and second messengers in regulation of the norepineph-
Nature 360, 420–421. rine transporter. Adv. Pharmacol. 42, 183–186.
Amara S. G. and Kuhar M. J. (1993) Neurotransmitter transporters: Brüss M., Kunz J., Lingen B. and Bonisch H. (1993) Chromosomal
recent progress. Annu. Rev. Neurosci. 16, 73–93. mapping of the human gene for the tricyclic antidepressant-sensi-
Angelakos E. T. (1964) Histochemical demonstration of exogenous tive noradrenalin transporter. Hum. Genet. 91, 278–280.
norepinephrine by adrenergic fibers in vitro. Science 145, 503– Brüss M., Pörzgen P., Bryan-Lluka L. J. and Bönisch H. (1997) The rat
504. norepinephrine transporter: molecular cloning from PC12 cells and
Apparsundaram S., Galli A., DeFelice L. J., Hartzell H. C. and Blakely functional expression. Brain Res. Mol. Brain Res. 52, 257–262.
R. D. (1998a) Acute regulation of norepinephrine transport: I. Bryan-Lluka L. J., Paczkowski F. A. and Bönisch H. (2001) Effects of
protein kinase C-linked muscarinic receptors influence transport short- and long-term exposure to c-AMP and c-GMP on the nor-
capacity and transporter density in SK-N-SH cells. J. Pharmacol. adrenaline transporter. Neuropharmacology 40, 607–617.
Exp. Ther. 287, 733–743. Bunn S. J., O’Brien K. J., Boyd T. L. and Powis D. A. (1992) Pertussis
Apparsundaram S., Schroeter S., Giovanetti E. and Blakely R. D. toxin inhibits noradrenaline accumulation by bovine adrenal
(1998b) Acute regulation of norepinephrine transport. II. PKC- medullary chromaffin cells. Naunyn Schmiedebergs Arch. Phar-
modulated surface expression of human norepinephrine transporter macol. 346, 649–656.
proteins. J. Pharmacol. Exp. Ther. 287, 744–751. Burnstock G. (1981) Review lecture. Neurotransmitters and trophic
Apparsundaram S., Sung U., Price R. D. and Blakely R. D. (2001) factors in the autonomic nervous system. J. Physiol. 313, 1–35.
Trafficking-dependent and – independent pathways of neurotrans- Burnstock G., Campbell G., Satchell D. and Smythe A. (1997) Evidence
mitter transporter regulation differentially involving p38 mitogen- that adenosine triphosphate or a related nucleotide is the transmitter
activated protein kinase revealed in studies of insulin modulation substance released by non-adrenergic inhibitory nerves in the gut
of norepinephrine transport in SK-N-SH cells. J. Pharmacol. Exp. 1970. Br. J. Pharmacol. 120, 337–357.
Ther. 299, 666–677. Burton L. D., Kippenberger A. G., Lingen B., Brüss M., Bönisch H. and
Axelrod J. and Kopin I. J. (1969) The uptake, storage, release and Christie D. L. (1998) A variant of the bovine noradrenaline
metabolism of noradrenaline in sympathetic nerves. Prog. Brain transporter reveals the importance of the C-terminal region for
Res. 31, 21–32. correct targeting to the membrane and functional expression. Bio-
Balfour D. J. (1973) Effects of nicotine on the uptake and retention of chem. J. 330, 909–914.
14C-noradrenaline and 14C-5-hydroxytryptamine by rat brain Caldwell P. C., Hodgkin A. L., Keynes R. D. and Shaw T. L. (1960) The
homogenates. Eur. J. Pharmacol. 23, 19–26. effects of injecting ‘energy-rich’ phosphate compounds on the
Bauer M. E. and Tejani-Butt S. M. (1992) Effects of repeated adminis- active transport of ions in the giant axons of Loligo. J. Physiol.
tration of desipramine or electroconvulsive shock on norepineph- 152, 561–590.
rine uptake sites measured by [3H]nisoxetine autoradiography. Cao C. J., Eldefrawi A. T. and Eldefrawi M. E. (1990) ATP-regulated
Brain Res. 582, 208–214. neuronal catecholamine uptake: a new mechanism. Life Sci. 47,
Bauman A. L., Apparsundaram S., Ramamoorthy S., Wadzinski B. E., 655–667.
Vaughan R. A. and Blakely R. D. (2000) Cocaine and antide- Casco V. H., Veinot J. P., Kuroski de Bold M. L., Masters R. G.,
pressant-sensitive biogenic amine transporters exist in regulated Stevenson M. M. and de Bold A. J. (2002) Natriuretic peptide
complexes with protein phosphatase 2A. J. Neurosci. 20, 7571– system gene expression in human coronary arteries. J. Histochem.
7578. Cytochem. 50, 799–809.

 2006 The Authors


Journal Compilation  2006 International Society for Neurochemistry, J. Neurochem. (2006) 97, 310–333
330 P. Mandela and G.A. Ordway

Couse J. F. and Korach K. S. (1999) Estrogen receptor null mice: what transporter in response to prolonged exposure to ketamine. Naunyn
have we learned and where will they lead us? Endocr. Rev. 20, Schmiedebergs Arch. Pharmacol. 365, 406–412.
358–417. Hara K., Yanagihara N., Minami K., Hirano H., Sata T., Shigematsu A.
Cubells J. F., Kim K. S., Baker H., Volpe B. T., Chung Y., Houpt T. A., and Izumi F. (2000) Dual effects of intravenous anesthetics on the
Wessel T. C. and Joh T. H. (1995) Differential in vivo regulation of function of norepinephrine transporters. Anesthesiology 93, 1329–
mRNA encoding the norepinephrine transporter and tyrosine hy- 1335.
droxylase in rat adrenal medulla and locus ceruleus. J. Neurochem. Hara K., Yanagihara N., Minami K., Ueno S., Toyohira Y., Sata T.,
65, 502–509. Kawamura M., Brüss M., Bönisch H., Shigematsu A. and Izumi F.
Deken S. L., Wang D. and Quick M. W. (2003) Plasma membrane (1998) Ketamine interacts with the noradrenaline transporter at a
GABA transporters reside on distinct vesicles and undergo rapid site partly overlapping the desipramine binding site. Naunyn Sch-
regulated recycling. J. Neurosci. 23, 1563–1568. miedebergs Arch. Pharmacol. 358, 328–333.
Distelmaier F., Wiedemann P., Brüss M. and Bönisch H. (2004) Func- Harder R. and Bönisch H. (1985) Effects of monovalent ions on the
tional importance of the C-terminus of the human norepinephrine transport of noradrenaline across the plasma membrane of neuronal
transporter. J. Neurochem. 91, 537–546. cells (PC-12 cells). J. Neurochem. 45, 1154–1162.
Ehrlich Y. H., Davis T. B., Bock E., Kornecki E. and Lenox R. H. (1986) Hardwick J. C., Ehrlich Y. H. and Hendley E. D. (1989) Extracellular
Ecto-protein kinase activity on the external surface of neural cells. ATP stimulates norepinephrine uptake in PC12 cells. J. Neuro-
Nature 320, 67–70. chem. 53, 1512–1518.
Eshleman A., Dunigan C., Shamoo A. and Eldefrawi M. (1995) ATP Hebert C., Habimana A., Elie R. and Reader T. A. (2001) Effects of
enhances catecholamine uptake into PC12 cells. Life Sci. 56, 1613– chronic antidepressant treatments on 5-HT and NA transporters
1621. in rat brain: an autoradiographic study. Neurochem. Int. 38,
Fan J. S. and Zhang M. (2002) Signaling complex organization by PDZ. 63–74.
domain proteins. Neurosignals. 11, 315–321. Hersch S. M., Yi H., Heilman C. J., Edwards R. H. and Levey A. I.
Fernandez J., Alonso J. M., Andres J. I., Cid J. M., DiaZ. A., Iturrino (1997) Subcellular localization and molecular topology of the
L., Gil P., Megens A., Sipido V. K. and Trabanco A. A. (2005) dopamine transporter in the striatum and substantia nigra. J. Comp
Discovery of new tetracyclic tetrahydrofuran derivatives as Neurol. 388, 211–227.
potential broad-spectrum psychotropic agents. J. Med. Chem. 48, Holton K. L., Loder M. K. and Melikian H. E. (2005) Nonclassical,
1709–1712. distinct endocytic signals dictate constitutive and PKC-regulated
Figlewicz D. P., Bentson K. and Ocrant I. (1993a) The effect of insulin neurotransmitter transporter internalization. Nat. Neurosci. 8, 881–
on norepinephrine uptake by PC12 cells. Brain Res. Bull. 32, 425– 888.
431. Hwang B. H., Kunkler P. E., Tarricone B. J., Hingtgen J. N. and
Figlewicz D. P., Szot P., Israel P. A., Payne C. and Dorsa D. M. (1993b) Nurnberger J. I. Jr (1999) Stress-induced changes of norepineph-
Insulin reduces norepinephrine transporter mRNA in vivo in rat rine uptake sites in the locus coeruleus of C57BL/6J and DBA/2J
locus coeruleus. Brain Res. 602, 161–164. mice: a quantitative autoradiographic study using [3H]-tomoxetine.
Friedrich U. and Bönisch H. (1986) The neuronal noradrenaline trans- Neurosci. Lett. 265, 151–154.
port system of PC-12 cells: kinetic analysis of the interaction Ikeda T., Kitayama S., Morita K. and Dohi T. (2001) Nerve growth
between noradrenaline, Na+ and Cl) in transport. Naunyn factor down-regulates the expression of norepinephrine transporter
Schmiedebergs Arch. Pharmacol. 333, 246–252. in rat pheochromocytoma (PC12) cells. Brain Res. Mol. Brain Res.
Furuya M., Takehisa M., Minamitake Y., Kitajima Y., Hayashi Y., 86, 90–100.
Ohnuma N., Ishihara T., Minamino N., Kangawa K. and Matsuo H. Iversen L. L. (1971) Role of transmitter uptake mechanisms in synaptic
(1990) Novel natriuretic peptide, CNP, potently stimulates cyclic neurotransmission. Br. J. Pharmacol. 41, 571–591.
GMP production in rat cultured vascular smooth muscle cells. Jayanthi L. D., Samuvel D. J. and Ramamoorthy S. (2004) Regulated
Biochem. Biophys. Res. Commun. 170, 201–208. internalization and phosphorylation of the native norepinephrine
Geerlings A., Lopez-Corcuera B. and Aragon C. (2000) Characteri- transporter in response to phorbol esters. Evidence for localization
zation of the interactions between the glycine transporters in lipid rafts and lipid raft-mediated internalization. J. Biol. Chem.
GLYT1 and GLYT2 and the SNARE protein syntaxin 1A. FEBS 279, 19 315–19 326.
Lett. 470, 51–54. Jayanthi L. D., Vargas G. and DeFelice L. J. (2002) Characterization
Geffen L. B. and Livett B. G. (1971) Synaptic vesicles in sympathetic of cocaine and antidepressant-sensitive norepinephrine trans-
neurons. Physiol. Rev. 51, 98–157. porters in rat placental trophoblasts. Br. J. Pharmacol. 135,
Gelernter J., Kruger S., Pakstis A. J., Pacholczyk T., Sparkes R. S., Kidd 1927–1934.
K. K. and Amara S. (1993) Assignment of the norepinephrine Kantor L. and Gnegy M. E. (1998) Protein kinase C inhibitors block
transporter protein (NET1) locus to chromosome 16. Genomics 18, amphetamine-mediated dopamine release in rat striatal slices.
690–692. J. Pharmacol. Exp. Ther. 284, 592–598.
Gould G. G., Pardon M. C., Morilak D. A. and Frazer A. (2003) Reg- Kantor L., Hewlett G. H., Park Y. H., Richardson-Burns S. M.,
ulatory effects of reboxetine treatment alone, or following par- Mellon M. J. and Gnegy M. E. (2001) Protein kinase C and
oxetine treatment, on brain noradrenergic and serotonergic intracellular calcium are required for amphetamine-mediated
systems. Neuropsychopharmacology 28, 1633–1641. dopamine release via the norepinephrine transporter in undif-
Graf B. M., Vicenzi M. N., Martin E., Bosnjak Z. J. and Stowe D. F. ferentiated PC12 cells. J. Pharmacol. Exp. Ther. 297,
(1995) Ketamine has stereospecific effects in the isolated perfused 1016–1024.
guinea pig heart. Anesthesiology 82, 1426–1437. Kaye D. M., Wiviott S. D., Kobzik L., Kelly R. A. and Smith T. W.
Haase J., Killian A. M., Magnani F. and Williams C. (2001) Regulation (1997) S-nitrosothiols inhibit neuronal norepinephrine transport.
of the serotonin transporter by interacting proteins. Biochem. Soc. Am. J. Physiol. 272, H875–H883.
Trans. 29, 722–728. King V. L., Dwoskin L. P. and Cassis L. A. (1999) Cold exposure
Hara K., Minami K., Ueno S., Toyohira Y., Tsutsui M., Shigematsu A. regulates the norepinephrine uptake transporter in rat brown adi-
and Yanagihara N. (2002) Up-regulation of noradrenaline pose tissue. Am. J. Physiol. 276, R143–R151.

 2006 The Authors


Journal Compilation  2006 International Society for Neurochemistry, J. Neurochem. (2006) 97, 310–333
Regulation of the norepinephrine transporter 331

Kitayama S., Morita K. and Dohi T. (2001) Functional characterization granule cells. Mol Pharmacol. November 10 Issue, EPublications
of the splicing variants of human norepinephrine transporter. ahead of print.
Neurosci. Lett. 312, 108–112. Nagy A. K., Shuster T. A. and gado-Escueta A. V. (1986) Ecto-ATPase
Klimek V., Stockmeier C. A., Overholser J., Meltzer H. Y., Kalka S., of mammalian synaptosomes: identification and enzymic charac-
Dilley G. and Ordway G. A. (1997) Reduced levels of norepi- terization. J. Neurochem. 47, 976–986.
nephrine transporters in the locus coeruleus in major depression. Nguyen T. T. and Amara S. G. (1996) N-linked oligosaccharides are
J. Neurosci. 17, 8451–8458. required for cell surface expression of the norepinephrine trans-
Kocabas A. M., Rudnick G. and Kilic F. (2003) Functional consequences porter but do not influence substrate or inhibitor recognition.
of homo- but not hetero-oligomerization between transporters for J. Neurochem. 67, 645–655.
the biogenic amine neurotransmitters. J. Neurochem. 85, 1513– Ordway G. A., Jia W., Li J., Zhu M. Y., Mandela P. and Pan J. (2005)
1520. Norepinephrine transporter function and desipramine: residual
Krishnan A. V., Stathis P., Permuth S. F., Tokes L. and Feldman D. drug effects versus short-term regulation. J. Neurosci. Meth 143,
(1993) Bisphenol-A: an estrogenic substance is released from 217–225.
polycarbonate flasks during autoclaving. Endocrinology 132, Ordway G. A., Stockmeier C. A., Cason G. W. and Klimek V. (1997)
2279–2286. Pharmacology and distribution of norepinephrine transporters in
Lee C. M., Javitch J. A. and Snyder S. H. (1983) Recognition sites for the human locus coeruleus and raphe nuclei. J. Neurosci. 17, 1710–
norepinephrine uptake: regulation by neurotransmitter. Science 1719.
220, 626–629. Pacholczyk T., Blakely R. D. and Amara S. G. (1991) Expression clo-
Lingen B., Brüss M. and Bönisch H. (1994) Cloning and expression of ning of a cocaine- and antidepressant-sensitive human noradren-
the bovine sodium- and chloride-dependent noradrenaline trans- aline transporter. Nature 350, 350–354.
porter. FEBS Lett. 342, 235–238. Paczkowski F. A., Bryan-Lluka L. J., Pörzgen P., Brüss M. and Bönisch
Loder M. K. and Melikian H. E. (2003) The dopamine transporter H. (1999) Comparison of the pharmacological properties of cloned
constitutively internalizes and recycles in a protein kinase C- rat, human, and bovine norepinephrine transporters. J. Pharmacol.
regulated manner in stably transfected PC12 cell lines. J. Biol. Exp. Ther. 290, 761–767.
Chem. 278, 22 168–22 174. Pan J., Jia W., Hume S. A., Mandela P. and Ordway G. A. (2003) Elusive
Lu D. YuK., Paddy M. R., Rowland N. E. and Raizada M. K. (1996) mechanism underlying regulation of the norepinephrine transporter
Regulation of norepinephrine transport system by angiotensin II in by inhibitors. Abstract Viewer/Itinerary Planner, Washington, DC.
neuronal cultures of normotensive and spontaneously hypertensive Society for Neuroscience, Abstract no. 253.5.
rat brains. Endocrinology 137, 763–772. Pan J., Jia W. H. and Ordway G. A. (2002) Cell surface expression of
Macey D. J., Smith H. R., Nader M. A. and Porrino L. J. (2003) Chronic norepinephrine transporter (NET) is reduced followed following
cocaine self-administration upregulates the norepinephrine trans- exposure of 293hNET cells to desipramine. Abstract Viewer/Itin-
porter and alters functional activity in the bed nucleus of the stria erary Planner, Washington, DC. Society for Neuroscience, Abstract
terminalis of the rhesus monkey. J. Neurosci. 23, 12–16. no. 646.12.
Mao W., Iwai C., Qin F. and Liang C. S. (2005) Norepinephrine induces Pörzgen P., Bönisch H. and Brüss M. (1995) Molecular cloning and
endoplasmic reticulum stress and downregulation of norepineph- organization of the coding region of the human norepinephrine
rine transporter density in PC12 cells via oxidative stress. Am. J. transporter gene. Biochem. Biophys. Res. Commun. 215, 1145–1150.
Physiol. Heart Circ. Physiol. 288, H2381–H2389. Pörzgen P., Bönisch H., Hammermann R. and Brüss M. (1998) The
Mao W., Qin F., Iwai C., Vulapalli R., Keng P. C. and Liang C. S. (2004) human noradrenaline transporter gene contains multiple polyade-
Extracellular norepinephrine reduces neuronal uptake of norepi- nylation sites and two alternatively spliced C-terminal exons.
nephrine by oxidative stress in PC12 cells. Am. J. Physiol. Heart Biochim. Biophys. Acta 1398, 365–370.
Circ Physiol. 287, H29–H39. Quick M. W., Corey J. L., Davidson N. and Lester H. A. (1997) Second
Mash D. C., Ouyang Q., Qin Y. and Pablo J. (2005) Norepinephrine messengers, trafficking-related proteins, and amino acid residues
transporter immunoblotting and radioligand binding in cocaine that contribute to the functional regulation of the rat brain GABA
abusers. J. Neurosci. Meth 143, 79–85. transporter GAT1. J. Neurosci. 17, 2967–2979.
Masson J., Sagne C., Hamon M. and El M. S. (1999) Neurotransmitter Raizada M. K., Lu D., Tang W., Kurian P. and Sumners C. (1993)
transporters in the central nervous system. Pharmacol. Rev. 51, Increased angiotensin II type-1 receptor gene expression in neur-
439–464. onal cultures from spontaneously hypertensive rats. Endocrinology
McEwen B. S. (1991) Non-genomic and genomic effects of steroids on 132, 1715–1722.
neural activity. Trends Pharmacol. Sci. 12, 141–147. Ramamoorthy S. and Blakely R. D. (1999) Phosphorylation and
Melikian H. E., McDonald J. K., Gu H., Rudnick G., Moore K. R. and sequestration of serotonin transporters differentially modulated by
Blakely R. D. (1994) Human norepinephrine transporter. Biosyn- psychostimulants. Science 285, 763–766.
thetic studies using a site-directed polyclonal antibody. J. Biol. Ramamoorthy S., Giovanetti E., Qian Y. and Blakely R. D. (1998)
Chem. 269, 12290–12297. Phosphorylation and regulation of antidepressant-sensitive sero-
Melikian H. E., Ramamoorthy S., Tate C. G. and Blakely R. D. (1996) tonin transporters. J. Biol. Chem. 273, 2458–2466.
Inability to N-glycosylate the human norepinephrine transporter Robinson M. B. (2002) Regulated trafficking of neurotransmitter trans-
reduces protein stability, surface trafficking, and transport activity porters: common notes but different melodies. J. Neurochem. 80,
but not ligand recognition. Mol. Pharmacol. 50, 266–276. 1–11.
Miner L. H., Schroeter S., Blakely R. D. and Sesack S. R. (2003) Ul- Role L. W. and Perlman R. L. (1983) Catecholamine uptake into isolated
trastructural localization of the norepinephrine transporter in adrenal chromaffin cells: inhibition of uptake by acetylcholine.
superficial and deep layers of the rat prelimbic prefrontal cortex Neuroscience 10, 987–996.
and its spatial relationship to probable dopamine terminals. Rosenstein R. E., Chuluyan H. E. and Cardinali D. P. (1990) Presynaptic
J. Comp Neurol. 466, 478–494. effects of gamma-aminobutyric acid on norepinephrine release and
Mtchedlishvili Z. and Kapur J. (2005) High affinity, slowly desensitizing uptake in rat pineal gland. J. Neural Transm. General Sect. 82,
GABAA receptors mediate tonic inhibition in hippocampal dentate 131–140.

 2006 The Authors


Journal Compilation  2006 International Society for Neurochemistry, J. Neurochem. (2006) 97, 310–333
332 P. Mandela and G.A. Ordway

Rusnak M., Kvetnansky R., Jelokova J. and Palkovits M. (2001) Effect PC12 cells through calmodulin-dependent kinases. Brain Res.
of novel stressors on gene expression of tyrosine hydroxylase and 809, 155–164.
monoamine transporters in brainstem noradrenergic neurons of Uchikawa T., Kiuchi Y., Yura A., Nakachi N., Yamazaki Y., Yokomizo
long-term repeatedly immobilized rats. Brain Res. 899, 20–35. C. and Oguchi K. (1995) Ca (2+)-dependent enhancement of
Sands S. A., Strong R., Corbitt J. and Morilak D. A. (2000) Effects of [3H]dopamine uptake in rat striatum: possible involvement of
acute restraint stress on tyrosine hydroxylase mRNA expression in calmodulin-dependent kinases. J. Neurochem. 65, 2065–2071.
locus coeruleus of Wistar and Wistar-Kyoto rats. Brain Res. Mol Vatta M. S., Bianciotti L. G. and Fernandez. B. E. (1993) Influence of
Brain Res. 75, 1–7. atrial natriuretic factor on uptake, intracellular distribution, and
Schroeter S., Apparsundaram S., Wiley R. G., Miner L. H., Sesack S. R. release of norepinephrine in rat adrenal medulla. Can. J. Physiol.
and Blakely R. D. (2000) Immunolocalization of the cocaine- and Pharmacol. 71, 195–200.
antidepressant-sensitive 1-norepinephrine transporter. J. Comp Vatta M. S., Presas M., Bianciotti L. G., Zarrabeitia V. and Fernandez. B.
Neurol. 420, 211–232. E. (1996) B and C types natriuretic peptides modulate norepi-
Shearman L. P. and Meyer J. S. (1999) Cocaine up-regulates norepi- nephrine uptake and release in the rat hypothalamus. Regul. Pept.
nephrine transporter binding in the rat placenta. Eur J. Pharmacol. 65, 175–184.
386, 1–6. Wakade A. R., Wakade T. D., Poosch M. and Bannon M. J. (1996)
Shin R. S., Anisman H., Merali Z. and McIntyre D. C. (2002) Changes Noradrenaline transport and transporter mRNA of rat chromaffin
in extracellular levels of amygdala amino acids in genetically fast cells are controlled by dexamethasone and nerve growth factor.
and slow kindling rat strains. Brain Res. 946, 31–42. J. Physiol. 494, 67–75.
Stingo A. J., Clavell A. L., Aarhus L. L. and Burnett J. C. Jr (1992) Xiao Q., Yao Y. and Tejani-Butt S. M. (1995) Acute administration of
Cardiovascular and renal actions of C-type natriuretic peptide. Am. alpha-methyl-para-tyrosine alters levels of norepinephrine trans-
J. Physiol. 262, H308–H312. porter mRNA in the rat brainstem. Brain Res. Mol Brain Res. 30,
Sudoh T., Maekawa K., Kojima M., Minamino N., Kangawa K. and 389–392.
Matsuo H. (1989) Cloning and sequence analysis of cDNA enco- Xu F., Gainetdinov R. R., Wetsel W. C., Jones S. R., Bohn L. M., Miller
ding a precursor for human brain natriuretic peptide. Biochem. G. W., Wang Y. M. and Caron M. G. (2000) Mice lacking the
Biophys. Res. Commun. 159, 1427–1434. norepinephrine transporter are supersensitive to psychostimulants.
Sudoh T., Minamino N., Kangawa K. and Matsuo H. (1990) C-type Nat. Neurosci. 3, 465–471.
natriuretic peptide (CNP): a new member of natriuretic peptide Yang H., Lu D., YuK. and Raizada M. K. (1996) Regulation of neuro-
family identified in porcine brain. Biochem. Biophys. Res. Com- modulatory actions of angiotensin II in the brain neurons by the
mun. 168, 863–870. Ras-dependent mitogen-activated protein kinase pathway. J. Neu-
Sung U., Apparsundaram S., Galli A., Kahlig K. M., Savchenko V., rosci. 16, 4047–4058.
Schroeter S., Quick M. W. and Blakely R. D. (2003) A regulated Yang H. and Raizada M. K. (1998) MAP kinase-independent signaling
interaction of syntaxin 1A with the antidepressant-sensitive nor- in angiotensin II regulation of neuromodulation in SHR neurons.
epinephrine transporter establishes catecholamine clearance capa- Hypertension 32, 473–481.
city. J. Neurosci. 23, 1697–1709. Yang H. and Raizada M. K. (1999) Role of phosphatidylinositol 3-kinase
Szot P., Ashliegh E. A., Kohen R., Petrie E., Dorsa D. M. and Veith R. in angiotensin II regulation of norepinephrine neuromodulation in
(1993) Norepinephrine transporter mRNA is elevated in the locus brain neurons of the spontaneously hypertensive rat. J. Neurosci.
coeruleus following short- and long-term desipramine treatment. 19, 2413–2423.
Brain Res. 618, 308–312. Yoshimura R., Yanagihara N., Hara K., Nakamura J., Toyohira Y., Ueno
Taube H. D., Montel H., Hau G. and Starke K. (1975) Phencyclidine and S. and Izumi F. (2001) Dual phases of functional change in nor-
ketamine: comparison with the effect of cocaine on the noradren- epinephrine transporter in cultured bovine adrenal medullary cells
ergic neurones of the rat brain cortex. Naunyn Schmiedebergs Arch. by long-term treatment with clozapine. J. Neurochem. 77, 1018–
Pharmacol. 291, 47–54. 1026.
Tejani-Butt S. M., Pare W. P. and Yang J. (1994) Effect of repeated novel Yura A., Kiuchi Y., Uchikawa T., Uchida J., Yamazaki K. and Oguchi K.
stressors on depressive behavior and brain norepinephrine receptor (1996) Possible involvement of calmodulin-dependent kinases in
system in Sprague-Dawley and Wistar Kyoto (WKY) rats. Brain Ca (2+)-dependent enhancement of [3H]5-hydroxytryptamine up-
Res. 649, 27–35. take in rat cortex. Brain Res. 738, 96–102.
Timmerman W. and Westerink B. H. (1997) Brain microdialysis of Zafar H. M., Pare W. P. and Tejani-Butt S. M. (1997) Effect of acute or
GABA and glutamate: what does it signify? Synapse 27, 242– repeated stress on behavior and brain norepinephrine system in
261. Wistar-Kyoto (WKY) rats. Brain Res. Bull. 44, 289–295.
Torres G. E., Yao W. D., Mohn A. R., Quan H., Kim K. M., Levey A. Zahniser N. R. and Doolen S. (2001) Chronic and acute regulation of
I., Staudinger J. and Caron M. G. (2001) Functional interaction Na+/Cl – dependent neurotransmitter transporters: drugs, sub-
between monoamine plasma membrane transporters and the strates, presynaptic receptors, and signaling systems. Pharmacol.
synaptic PDZ. domain-containing protein PICK1. Neuron 30, Ther. 92, 21–55.
121–134. Zavosh A., Schaefer J., Ferrel A. and Figlewicz. D. P. (1999) Desipr-
Toyohira Y., Utsunomiya K., Ueno S., Minami K., Uezono Y., amine treatment decreases 3H-nisoxetine binding and norepi-
Yoshimura R., Tsutsui M., Izumi F. and Yanagihara N. (2003) nephrine transporter mRNA in SK-N-SHSY5Y cells. Brain Res.
Inhibition of the norepinephrine transporter function in cultured Bull. 49, 291–295.
bovine adrenal medullary cells by bisphenol A. Biochem. Zhao Y. and Sun L. (2004) Perinatal cocaine exposure reduces myo-
Pharmacol. 65, 2049–2054. cardial norepinephrine transporter function in the neonatal rat.
Trendelenburg U. (1991) The TiPS lecture: functional aspects of the Neurotoxicol. Teratol. 26, 443–450.
neuronal uptake of noradrenaline. Trends Pharmacol. Sci. 12, 334– Zhu M. Y., Blakely R. D., Apparsundaram S. and Ordway G. A. (1998)
337. Down-regulation of the human norepinephrine transporter in intact
Uchida J., Kiuchi Y., Ohno M., Yura A. and Oguchi K. (1998) Ca 293-hNET cells exposed to desipramine. J. Neurochem. 70, 1547–
(2+)-dependent enhancement of [3H]noradrenaline uptake in 1555.

 2006 The Authors


Journal Compilation  2006 International Society for Neurochemistry, J. Neurochem. (2006) 97, 310–333
Regulation of the norepinephrine transporter 333

Zhu M. Y., Kim C. H., Hwang D. Y., Baldessarini R. J. and Kim K. S. Zhu M. Y. and Ordway G. A. (1997) Down-regulation of norepinephrine
(2002) Effects of desipramine treatment on norepinephrine trans- transporters on PC12 cells by transporter inhibitors. J. Neurochem.
porter gene expression in the cultured SK-N-BE 2, M17 cells and 68, 134–141.
rat brain tissue. J. Neurochem. 82, 146–153. Zhu M. Y., Shamburger S., Li J. and Ordway G. A. (2000) Regulation of
Zhu M. Y., Kyle P. B., Hume A. S. and Ordway G. A. (2004) The the human norepinephrine transporter by cocaine and amphetam-
persistent membrane retention of desipramine causes lasting inhi- ine. J. Pharmacol. Exp. Ther. 295, 951–959.
bition of norepinephrine transporter function. Neurochem. Res. 29,
419–427.

 2006 The Authors


Journal Compilation  2006 International Society for Neurochemistry, J. Neurochem. (2006) 97, 310–333

You might also like