Download Causes and Consequences of Increased Sympathetic Activity in

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Neuropsychopharmacology wikipedia , lookup

Discovery and development of beta-blockers wikipedia , lookup

Neuropharmacology wikipedia , lookup

Bilastine wikipedia , lookup

Ofloxacin wikipedia , lookup

Discovery and development of angiotensin receptor blockers wikipedia , lookup

Transcript
Causes and Consequences of Increased Sympathetic Activity
in Renal Disease
Jaap A. Joles, Hein A. Koomans
Downloaded from http://hyper.ahajournals.org/ by guest on April 30, 2017
Abstract—Much evidence indicates increased sympathetic nervous activity (SNA) in renal disease. Renal ischemia is
probably a primary event leading to increased SNA. Increased SNA often occurs in association with hypertension.
However, the deleterious effect of increased SNA on the diseased kidney is not only caused by hypertension. Another
characteristic of renal disease is unbalanced nitric oxide (NO) and angiotensin (Ang) activity. Increased SNA in renal
disease may be sustained because a state of NO–Ang II unbalance is also present in the hypothalamus. Very few studies
have directly compared the efficacy of adrenergic blockade with other renoprotective measures. Third-generation
␤-blockers seem to have more protective effects than traditional ␤-blockers, possibly via stimulation of NO release.
Although it has been extensively documented that muscle SNA is increased in chronic renal failure, data on renal SNA
and cardiac SNA are not available for these patients before end-stage renal disease. It is also unknown whether
additional treatment with third-generation ␤-blockers can delay the progression of renal injury and prevent cardiac
injury in chronic renal failure more efficiently than conventional treatment with angiotensin-converting enzyme
inhibitors only. (Hypertension. 2004;43:699-706.)
Key Words: renal disease 䡲 antihypertensive agents 䡲 hypertension 䡲 diabetic nephropathy
䡲 sympathetic nervous system
I
In the clinical setting, however, the intracellular domain is
not yet accessible and gene therapy lies beyond the horizon.
Hence, the focus of much research, both clinical and experimental renal disease, is directed toward manipulation of
extracellular pathways. In this review, we focus on the role of
the sympathetic nervous system (SNS), which is activated in
subjects with renal disease.6 First, we briefly review potential
mechanisms that could be responsible for SNS stimulation in
renal disease. Then, we summarize the evidence that SNS
stimulation contributes to progression of renal disease. Mortality caused by cardiovascular disease is more than 3-times
higher in subjects with ESRD than in subjects with normal
renal function.7 Hence, we also review recent studies that
support a role for the SNS in the myocardial changes that are
inherent to uremia.
t is well known that irrespective of the primary cause of
renal injury, chronic renal disease often leads to renal
fibrosis and end-stage renal disease (ESRD). Delaying the
onset of renal replacement therapy is of utmost importance,
both medically and economically.1 Many quite different
interventions (dietary protein or phosphate restriction, inhibition of the renin-angiotensin system [RAS], endothelin
receptor antagonists, aldosterone antagonists, antioxidants,
renal denervation, HMG-CoA reductase inhibitors, etc) have
been shown to considerably slow this process in experimental
models. In fact, if these were all discrete pathways, then
combining such interventions would provide additive protective effects that would exceed the loss of function. Clearly,
this is not the case, suggesting that parallel extracellular
pathways converge on common intracellular signaling molecules, which in turn activate a limited number of transcription
factors that turn on excessive matrix synthesis. Recent studies
using modulation of intracellular signaling pathways, eg, by
gene transfer of Smad 7,2 or inhibition of transcription factors
with antisense technology3 show marked reduction of fibrosis
in robust models such as unilateral ureteral obstruction.
Another pathway that is gaining increasing attention is the
independent role of proteinuria in causing interstitial inflammation and ultimately fibrosis.4 Such inflammation causes
cytokine release and consequent monocyte influx. Preventing
this monocyte influx by anti-monocyte chemoattractant protein (MCP)-1 gene therapy5 also reduces injury.
What Stimulates the SNS in Renal Disease?
In the later stages of chronic renal disease, when renal
clearance is substantially decreased, renal catabolism of
peptides and low-molecular-weight proteins will decrease.
One such peptide is leptin, a peptide produced mainly in
adipose tissue, that decreases food intake and increases
energy expenditure. The latter is mediated by an increase in
SNS activity.8 Hyperleptinemia is common in ESRD.9 It has
been postulated that increased SNS activity associated with
hyperleptinemia may contribute to hypertension in ESRD10 as
Received October 10, 2003; first decision November 5, 2003; revision accepted February 3, 2004.
From the Department of Nephrology and Hypertension, University Medical Center, Utrecht, The Netherlands.
Correspondence to Dr Jaap A. Joles, Department of Nephrology and Hypertension (Room F03.226), University Medical Center, Heidelberglaan 100,
P.O. Box 85500, 3508 GA Utrecht, The Netherlands. E-mail [email protected]
© 2004 American Heart Association, Inc.
Hypertension is available at http://www.hypertensionaha.org
DOI: 10.1161/01.HYP.0000121881.77212.b1
699
700
Hypertension
April 2004
Downloaded from http://hyper.ahajournals.org/ by guest on April 30, 2017
it does in obesity.11 Accumulation of endogenous nitric oxide
synthase (NOS) inhibitors in chronic renal failure (CRF) such
as asymmetric dimethyl arginine could potentially directly
increase SNS activity by inhibiting cerebral NO production.
However, this is not likely because the levels are very low.12
Moreover, direct effects of uremia cannot be the whole story,
because SNS activity is already increased at earlier stages when
renal function is not or only slightly impaired13 and sympathetic
hyperactivity remains after correction of uremia by renal transplantation.14 Increased SNS activity in the presence of normal
renal function is also sometimes found in the nephrotic syndrome, in which central cardiovascular stimulation is thought to
play an important role in stimulating efferent renal sympathetic
nervous activity (RSNA).15 However, judging by noradrenaline
levels, this association only appears to be present in conjunction
with hypovolemic symptoms.16
Renal ischemia is probably an important primary event
leading to increased SNS activity. This was observed experimentally with acute renal artery stenosis.17 Restoration of
renal perfusion in humans with renovascular hypertension
reduces muscle sympathetic nerve activity (MSNA) to control levels along with normalization of blood pressure.18
Increased SNS activity has also been observed in hypertensive subjects with polycystic kidney disease and normal renal
function.13 It is conceivable that renal cysts cause localized
intrarenal ischemia, which in turn stimulates ␣2 adrenoceptors. Campese’s group has shown in rats that unilateral
intrarenal injection of a small quantity of phenol induces
hypertension that is associated with increased RSNA.19 Increased norepinephrine content of the posterior hypothalamic
nuclei is present in the 5/6-nephrectomy model.20 Importantly, in the 5/6-nephrectomy model, it was also shown that
central sympathetic activation could be dampened by central
production of NO from neuronal NOS.20 Intraventricular
administration of angiotensin (Ang) II also directly increases
central sympathetic output.21
Unbalanced NO Versus Angiotensin and
Sympathetic Activity in Renal Disease
Within the kidney, NOS activity progressively decreases in
CRF induced by ablation, as does urinary nitrite plus nitrate
excretion.22 Similar findings have been discovered in
immune-mediated glomerulonephritis.23 Moreover, proteinuria and renal injury induced by hypercholesterolemia are
also accompanied by a reduction in renal NOS activity.24 In
both ablation and hypercholesterolemia, proteinuria and injury are partially or wholly prevented by molsidomine, a NO
donor.24,25 Angiotensin-converting enzyme (ACE) inhibition
or AT-1 antagonism effectively reduces renal injury in the
ablation and NOS inhibition models,26 and this is also the
case for renal injury associated with hypercholesterolemia.27
Thus, a characteristic of renal disease is unbalanced NO and
Ang activity.28 However, this does not necessarily imply that
tissue Ang II is actually increased, but rather that it is high in
relation to the protection offered by NO activity. Indeed, Ang
II levels were neither increased in the intact portion of the
remnant kidney (away from the infarct scar)29 nor in the
whole kidney during NOS inhibition with a high dose of
NG-nitro-L-arginine (L-NNA), at least not before the develop-
ment of extensive injury and scarring.30 Increased SNS
activity in renal disease may be sustained because a state of
NO-Ang II imbalance is also present in the hypothalamus.20,21
Enhanced SNS activity appears to be a direct effect of
primary NO deficiency, even before renal insufficiency.
Circulating catecholamines, particularly epinephrine, levels
were increased several-fold after chronic NG-nitro-L-arginine
methylester (L-NAME) treatment.31 Intracisternal NG-monomethyl-L-arginine (L-NMMA) administration resulted in a
mild pressor response but in marked stimulation of RSNA.32
Recently, it was shown that the acute central antihypertensive
action of clonidine, an ␣2-adrenergic agonist, is caused by the
release of NO in the nucleus tractus solitarii (NTS).33 Thus,
the marked antihypertensive effect of chronic clonidine administration observed in the intrarenal phenol model by Ye et
al was possibly not only caused by maintenance of intracerebral nNOS mRNA19 but also caused by direct stimulation of
NO release in the brain.
SNS activity plays an important role in the genesis of
hypertension in NO deficiency. Renal denervation can prevent L-NAME hypertension.34 Similarly, adrenergic blockade
can prevent hypertension when NO deficiency is induced in
pregnant rats35 and in diabetic rats.36
Catecholamines are potent stimulators of platelet aggregation.37 Conversely, chronic NO deficiency induced intraglomerular platelet aggregation and glomerular injury, which
was ameliorated by renal denervation.38 Moreover, increased
SNS activity may in fact aggravate the NO deficiency
induced by L-NAME, because the ␣1-adrenoceptor blocker,
doxazosin, increased the nitrite production of kidney tissue of
L-NAME–treated rats.39 However, this may be caused by the
partial correction of hypertension, because a calcium channel
antagonist and an ACE-inhibitor shared this effect on nitrite
production. Interestingly, renal nitrite production was increased above control levels when L-NAME–induced hypertension was treated with either the ␣1-adrenoceptor blocker
or the ACE inhibitor,39 suggesting that even under control
conditions both RSNA and intrarenal Ang II suppress renal
NO production. Thus once renal insufficiency starts to
develop, the resulting depression of renal NO synthesis and
enhancement of RSNA and the intrarenal RAS start to
reinforce each other in an ominous dance. Neutralization of
any one of these factors reduces progression of renal injury.
For example, in renal ablation renal function and structure
can be preserved by a NO donor,25 ACE inhibition, or AT-1
antagonism,26 or by ␣ and ␤ blockade.40 Moreover, even
though this model is classically regarded as being hemodynamically driven, protection by the lymphocyte inhibitor,
mycophenolate mofetil, indicates that inflammatory factors
play a key role in scarring.41 Combining hemodynamic and
non-hemodynamic interventions may provide somewhat better protection.41 The challenge is to identify and target
different multiple upstream pathways to halt activation of the
common downstream pathway.
How Does SNS Stimulation Contribute to
Progression of Renal Disease?
Is Hypertension Always Involved?
The deleterious effect of increased SNS activity on the
diseased kidney is not only caused by hypertension. In fact,
Joles and Koomans
Downloaded from http://hyper.ahajournals.org/ by guest on April 30, 2017
infusion of noradrenaline for 1 week did not induce proteinuria or decrease glomerular filtration rate (GFR), in contrast
to Ang II, despite similar development of severe hypertension.42 Note that longer periods of noradrenaline infusion,
even at a lower doses and, hence, blood pressure, do cause
vascular43 and renal injury44 vide infra. Moxonidine, an
I1-imidazoline agonist that inhibits the release of noradrenaline both in the kidney and in the brain,45 reduced glomerulosclerosis and albuminuria without changing blood pressure
in the subtotal nephrectomy model.46 Furthermore, in subtotally nephrectomized rats, subantihypertensive doses of ␣ and
␤ blockade, and particularly their combination, also reduced
progression of renal disease.40 Similarly, in extremely hypertensive stroke-prone spontaneously hypertensive rats (SHR)
on a high-fat, high-sodium diet, ␤ blockade increased survival and reduced glomerular, tubulointerstitial, and intrarenal vascular injury without any effect on systolic blood
pressure.47,48 This contrasts with the renoprotective effects of
ACE inhibition and aldosterone blockade in salt-loaded
stroke-prone SHR and of ACE-inhibition and AT-1–receptor
blockade in renal ablation that were consistently bloodpressure dependent.49,50 Note in the studies by Amann’s
group and in those of Bidani’s group that blood pressure was
measured continuously in conscious unrestrained animals
using telemetry.40,46,49,50 Thus renal protection by blockade of
the RAS or by reduction of SNS activity may not follow
identical pathways. This supports the notion that complementary combinations may have beneficial effects. Surprisingly,
little information is available on the effects of adrenergic
blockade in models of renal injury not associated with
hypertension, although it has been documented that in experimental nephrotic syndrome RSNA is increased.15
Sympathetic Hyperactivity: Mainly Vascular and
Glomerular Effects
Prolonged SNS hyperactivity can induce changes in intrarenal blood vessels. Catecholamines induce proliferation of
smooth muscle cells and adventitial fibroblasts in the vascular
wall.43,51 ␣1-Adrenoceptors mediate these effects.51 In vivo,
however, such effects are difficult to prove because the
simultaneous hemodynamic effects also have trophic effects.
However, suffusion of rat carotid arteries for 2 weeks with
noradrenaline at ⬇1000-times below the threshold for altering arterial pressure clearly caused hypertrophy.43 Interestingly, a new hypothesis relating peritubular capillary rarefaction to salt-sensitive hypertension52 was initially supported by
observations in rats infused with phenylephrine for 8 weeks.44
However, subsequently this has been observed in many other
primary models of hypertensive renal injury,52 so this cannot
be viewed as a specific adrenergic effect.
Perivascular nerves in the adventitia release ATP with
noradrenaline and neuropeptide Y from sympathetic nerves to
act at smooth muscle P2X purinoceptors resulting in vasoconstriction.53 In preglomerular vessels, ATP and ADP stimulate P2 receptors and cause vasoconstriction by opening
calcium channels, whereas AMP and adenosine stimulate P1
receptors (adenosine A1 receptors) and cause relaxation by
increasing cAMP formation.54 Although adrenergic fibers do
not penetrate the glomerulus,55 podocytes56 have adrenocep-
Sympathetic Activity in Renal Disease
701
tors, and all glomerular cell types have purinoceptors.57 In
contrast to the preglomerular vessels in which ATP causes
constriction,54 ATP efflux from endothelial cells in isolated
glomeruli stimulates P2Y purinoceptor-mediated NO release
and causes dilation of glomerular capillaries.58 This microvascular endothelial ATP release is potently reinforced by
third-generation ␤-blockers.58 These apparently conflicting
actions of extracellular ATP are possible because extracellular ATPase limits the half-life of extracellular ATP to 0.2
seconds.57
Dissecting the exact mechanism by which sympathetic
activity damages the glomerulus independently of Ang II is
not trivial. Podocyte injury is a pivotal step in the development of glomerulosclerosis, and an early event is probably
calcium influx, constriction, and hence decreased glomerular
permselectivity.56 Adrenergic and purinergic receptors appear
to be present on podocytes, because adrenergic and purinergic
agonists induced calcium influx.56 Podocytes also have Ang
II receptors, and Ang II can also induce calcium influx in
podocytes ex vivo.56 Thus direct effects of both the RAS and
the SNS may lead to podocyte constriction and proteinuria,
independently of their hemodynamic effects. Interestingly, in
a 2-week experiment with dietary hypercholesterolemia, ie,
before the development of proteinuria, we observed a decrease in renal NOS activity and protective effects of losartan
on podocyte activation without a significant decrease in blood
pressure.59 Similarly, Amann et al observed podocyte protection with adrenergic blockade without a change in blood
pressure in the subtotal nephrectomy model.40
Secondary Tubulointerstitial Changes
It is well known that the tubular epithelium is densely
innervated and well endowed with both adrenergic55 and
purinergic57 receptors. However, in contrast to Ang II that
directly stimulates proliferation, apoptosis and collagen synthesis via its receptors on the tubular epithelium,60 adrenergic
and purinergic receptors do not appear to be directly involved
in the characteristic tubulointerstitial changes of chronic renal
disease. Catecholamine infusion, in contrast to Ang II, did not
stimulate tubular epithelial or interstitial apoptosis or proliferation.61 The overriding tubulotoxic effects of proteinuria4
preclude dissection of a sympathetic contribution to interstitial fibrosis in most models.
Adrenergic Blockade in Different Models of
Renal Disease
␤-Adrenergic Receptor Blockade
There are several studies showing renal protection by
␤-adrenergic receptor blockade in 5/6 nephrectomy.40,62– 65
Renal protection by ␤-blockade has also been observed in
NOS inhibition66 – 68 and in stroke-prone SHR on high-salt
intake.47 Experimental studies of renal injury that evaluated
adrenergic blockade are listed in Table 1. Very few studies
have directly compared the efficacy of adrenergic blockade
with other renoprotective measures. In the study by Brooks et
al,65 very similar protection was found for carvedilol and
captopril. There may also be substantial differences between
different ␤-blocking drugs. Third-generation ␤-blockers,
702
Hypertension
April 2004
TABLE 1. Effects of Renal Denervation or Pharmacological Blockade of the Sympathetic Nervous System on Blood Pressure and
Renal Injury in Different Models of Renal Disease
Model
Treatment
Study
Length
Glomerular
Blood Filtration
Glomerular Tubulo-int. Vascular
Pressure
Rate
UalbV UprotV Injury
Injury
Injury Reference
Downloaded from http://hyper.ahajournals.org/ by guest on April 30, 2017
5/6 NX excision
Renal denervation
12 wk
7
7
2
2
2
2
75
5/6 NX excision
Moxonidine 1.5 mg/kg per day
12 wk
7
7
2
2
7
2
46
5/6 NX excision
Phenoxybenzamine 5 mg/kg per day
12 wk
7
7
2
2
2
2
40
5/6 NX excision
Metoprolol 150 mg/kg per day
12 wk
7
7
2
2
2
2
40
5/6 NX excision
Phenoxybenzamine⫹Metoprolol
12 wk
7
7
2
22
22
22
40
5/6 NX excision
Nipradilol 10 or 15 mg/kg per day
12 wk
2
1
2
2
N/A
N/A
64
5/6 NX excision
Propanolol 50 mg/kg per day
12 wk
2
1
2
7
N/A
N/A
64
5/6 NX excision⫹L-NAME
6 mg/kg per day
Nipradilol 10 mg/kg per day
12 wk
2
1/7
7
2/7
N/A
N/A
64
5/6 NX excision⫹L-NAME
6 mg/kg per day
Propanolol 50 mg/kg per day
12 wk
2
7
7
7
N/A
N/A
64
5/6 NX excision
Carvedilol 30 mg/kg per day
11 wk
2
1
2
2
N/A
N/A
63
5/6 NX excision
Propanolol 30 mg/kg per day
11 wk
2
1
2
7
N/A
N/A
63
98
5/6 NX infarction
Renal denervation
6 wk
2
1
N/A
2
N/A
N/A
5/6 NX infarction
Carvedilol 55 mg/kg per day
6 wk
2
1
N/A
2
2
N/A
65
5/6 NX infarction
Carvedilol 5 or 10 mg/kg per day
11 wk
7/2
1
2
2
2
N/A
62
5/6 NX infarction
Carvedilol 20 mg/kg per day
11 wk
2
1
22
22
22
N/A
62
Obese SHR
Moxonidine 8 mg/kg per day
13 wk
2
N/A
2
N/A
N/A
N/A
88
SHR/N-cp diabetes
Doxazosin 40 mg/kg per day*
16 wk
2
N/A
2
2
N/A
N/A
73
STZ diabetes
Doxazosin 30 mg/kg per day*
20 wk
2
N/A
22
7
N/A
N/A
82
Renal denervation
4 wk
2
N/A
2
N/A.
N/A
N/A
34
L-NAME
70 mg/kg per day
L-NAME
80 mg/kg per day
Renal denervation
4 wk
N/A
N/A
2
22
2
N/A
38
L-NAME
60 mg/kg per day
Doxazosin 10 mg/kg per day
10 wk, treated
last 4 wk
2
N/A
N/A
2
N/A
N/A
39
L-NAME
5 mg/kg per day*
Doxazosin 30 mg/kg per day
12 wk, treated
last 6 wk
2
7
2
N/A
N/A
N/A
66
L-NAME
5 mg/kg per day*
Metoprolol 350 mg/kg per day
12 wk, treated
last 6 wk
22
7
22
N/A
N/A
N/A
66
L-NAME
35 mg/kg per day
Prazosin 5 mg/kg per day*
6 wk
2
7
22
22
22
22
68
Prazosin 5 mg/kg per day*
6 wk
2
7
22
22
22
22
68
SHR⫹L-NAME
10 mg/kg per day*
Nipradilol 20 mg/kg per day
3 wk
2
1
N/A
2
N/A
N/A
67
Aging SHRSP
Moxonidine 8 mg/kg per day
3 month
2
N/A
N/A
2
2
2
99
L-NAME⫹DOCA
SHRSP⫹high salt
Carvedilol 200 mg/kg per day*
18 wk (survival)
7
7
N/A
2
2
2
47
SHRSP⫹high salt
Propanolol 200 mg/kg per day*
18 wk (survival)
7
7
N/A
2
2
2
47
SHRSP⫹high salt
Carvedilol 200 mg/kg per day*
7 wk
7
1
2
2
2
2
48
Pregnancy⫹L-NAME
10 mg/kg per day
Methyldopa 400 mg/kg per day
D11 to term
2
1
2
N/A
N/A
N/A
35
Carvedilol 2 mg/kg per day
intraperitoneal
8d
N/A
1
N/A
N/A
22
N/A
69
Gentamicin acute renal failure
N/A indicates not available; tubulo-int., tubulointerstitial; NX, nephrectomy; 1, increased; 2, decreased; 7, unchanged (all in comparison to the levels found in
the non-treated model).
Abbreviations are defined in the text.
*Calculated from intake and body weight.
such as carvedilol and nebivolol, seem to have more protective effects than traditional ␤-blockers,47,63 possibly via stimulation of NO release.58 In fact, carvedilol is even protective
in acute renal failure after gentamicin.69 Third-generation
␤-blockers are now rapidly becoming the drug of choice in
heart failure in the general population.70 However, their use in
patients with advanced renal failure who experience myocardial infarction is grossly neglected.71 Hopefully, the marked
improvement of heart function and survival of dialysis
patients with heart failure achieved by carvedilol72 will
improve this treatment deficit. The important question is
whether additional treatment with these drugs can delay the
Joles and Koomans
progression of renal injury and prevent cardiac injury in
chronic renal failure more efficiently than conventional treatment with ACE inhibitors only. To our knowledge, not a
single experimental study is available that addresses this
question.
␣-Adrenergic Receptor Blockade
Downloaded from http://hyper.ahajournals.org/ by guest on April 30, 2017
Renal protection by ␣-adrenergic receptor blockade has also
been observed in the 5/6-nephrectomy model,40 in NOS
inhibition,39,66,68 and in type 2 diabetes in corpulent SHR.73
Combining ␣-adrenergic blockade (doxazosin) with ACE
inhibition only provided slightly better protection (ie, less
mesangial expansion) than single-drug treatment in spontaneous type 2 diabetes.73 However, the use of ␣-adrenergic
blockade has been superseded by the findings of the Antihypertensive and Lipid-Lowering Treatment to Prevent Heart
Attack Trial (ALLHAT) study in which the most recent
analysis conclusively shows a higher risk of stroke and
combined cardiovascular disease in patients treated with
doxazosin than in those using the diuretic chlorthalidone.74
Inclusion criteria for the ALLHAT study were age (older than
55 years), hypertension, and at least one additional risk factor.
Note that renal disease was not a recognized risk factor, and
⬇36% of the patients had type 2 diabetes;74 thus, the results
of this study cannot be directly extrapolated to chronic renal
disease.
␣ Plus ␤-Adrenergic Receptor Blockade
A combination of ␣ blockade and ␤ blockade (phenoxybenzamine plus metoprolol) aimed at preventing both ␣-adrenergic–mediated ATP release and ␤-adrenergic–mediated renin release was more effective in reducing renal damage
(glomerular, tubulointerstitial, and vascular) in the subtotal
nephrectomy model than ␣ blockade only.40 Similar beneficial effects were observed in this model with the central
sympatholytic agent moxonidine46 and renal denervation.75 A
recent comparative study in patients with advanced CRF
showed that moxonidine superimposed on some form of RAS
inhibition delayed loss of renal function over a 24-week
period, whereas superimposing nitrendipine did not.76 However, as pointed out by Laverman and Remuzzi, this observation needs a follow-up because at the start of the trial, the
albumin excretion was substantially lower (1.3 versus 1.9
g/d) in the group subsequently treated with moxonidine.77
Diabetes and Obesity
Diabetes triggers mechanisms that in parallel enhance and
suppress NO bioavailability in the kidney. In the early phases
of diabetic nephropathy, the balance between these opposing
forces appears to be shifted toward increased NO, whereas in
a later phase, factors that suppress NO bioavailability prevail.78 A similar situation may apply to SNS activity. MSNA
is typically reduced in insulin-dependent diabetes mellitus
patients without diabetic complications,79 and RSNA is decreased after 2 weeks in normotensive streptozotocin (STZ)induced diabetes in rats.80 Indeed, Matsuoka found that renal
denervation before induction of diabetes with STZ enhanced
albuminuria.81 At a later stage, however, sympathetic nerve
activity is clearly deleterious, because when ␣-adrenergic
Sympathetic Activity in Renal Disease
703
blockade was started 1 week after STZ, albuminuria decreased.82 Normotensive type 1 diabetic patients who had had
diabetes for ⬎15 years showed a reduction in microalbuminuria after treatment with a dose of moxonidine that did not
affect blood pressure.83 However, when diabetic nephropathy
is allowed to progress, renal denervation is no longer able to
decrease albumin and transforming growth factor (TGF)-␤
excretion.84 In meta-analyses of studies exclusively directed
at diabetes, ACE-inhibition was approximately twice as
effective as diuretics and/or ␤-blockers in reducing proteinuria85 and cardiovascular events.86 The only exception was
the United Kingdom Prospective Diabetes Study (UKPDS),
which did not show any difference and involved 758 of 2180
patients in the meta-analysis by Pahor et al.86 Thus, in the
race to save diabetic patients from renal and cardiovascular
disease, the conventional ␤-blockers seem to have been
beaten by the ACE inhibitors. However, in the next run-off,
third-generation ␤-blockers have good chances.
A common disorder that is closely linked to diabetes is
obesity. Obesity is also associated with microalbuminuria.87
As mentioned, doxazosin reduced albuminuria in corpulent
SHR,73 and proteinuria was also lower in obese SHR treated
with moxonidine.88 Interestingly, in the latter study the
antihypertensive and renoprotective actions of moxonidine
were accompanied by improvement of glucose tolerance and
insulin sensitivity.88 Although renal SNS activity is clearly
increased in normotensive and in hypertensive obesity,89 an
effect of adrenergic blockade on microalbuminuria has yet to
be shown in obesity.
SNS Stimulation and Cardiac Injury in
Chronic Renal Disease
Experimental Studies
Experimental studies that evaluated effects of adrenergic
blockade on cardiac injury in chronic renal disease are listed
in Table 2. Although most of the experimental studies in
which blood pressure is decreased by sympathetic inhibition
also found a decrease in left ventricular hypertrophy,68 this
cannot be viewed as a specific effect. In renal ablation
studies, both moxonidine90 and carvedilol62 reduced myocardial interstitial volume and restored the abnormal myocardial
capillary density. Similar findings were documented in aging
stroke-prone SHR.91 Because blood pressure decreased in
these studies, it remains difficult to distinguish a specific
effect of decreased adrenergic action on the heart. However,
in stroke-prone SHR on a high-salt diet, good cardiac92 and
renal47 protection was observed with carvedilol without any
change in blood pressure. In this model, we have previously
found partial reduction of proteinuria and total remission of
cerebral edema for many months after late initiation of ACE
inhibition or AT-1 receptor blockade once injury was present,
despite persistent severe hypertension.93 Thus, part of the
protective action of carvedilol in this model may be caused by
a decrease in tissue Ang II activity in target organs.
Clinical Studies
Cardiac sympathetic nervous activity (CSNA), MSNA, and
RSNA are coordinately increased in essential hypertension
704
Hypertension
April 2004
TABLE 2. Effects of Pharmacological Blockade of the Sympathetic Nervous System on Blood Pressure and Cardiac Injury in Different
Models of Renal Disease
Treatment
Study
Length
Blood
Pressure
Left
Ventricular
Hypertrophy
Arteriolar
Thickening
Capillary
Density
Interstitial
Volume
Inflammation,
Microthrombosis,
and Infarction
Reference
70 mg/kg
Propanolol 100 mg/kg per day*
8 wk
2
2
N/A.
N/A.
N/A.
2
100
70 mg/kg
Atenolol 100 mg/kg
per day*
8 wk
2
2
N/A.
N/A.
N/A.
2
100
35 mg/kg
per day⫹DOCA
Prazosin 5 mg/kg
per day*
6 wk
2
2
N/A.
N/A.
N/A.
N/A.
68
Model
L-NAME
per day*
L-NAME
per day*
L-NAME
Downloaded from http://hyper.ahajournals.org/ by guest on April 30, 2017
5/6 NX infarction
Carvedilol 5 or 10 mg/kg per day
11 wk
7/2
2
N/A.
2
2
2
62
5/6 NX infarction
Carvedilol 20 mg/kg
per day
11 wk
2
2
N/A.
22
22
2
62
SHRSP⫹high salt
Carvedilol 200 mg/kg per day*
18 wk
(survival)
7
2
2
N/A.
2
22
92
5/6 NX excision
Moxonidine 10 mg/kg per day
8 wk
2
2
7/2
2
2
N/A.
90
Moxonidine 8 mg/kg
per day
3 month
2
2
2
2
2
22
91, 99
Aging SHRSP
Abbreviations are defined in the text.
N/A indicates not available; 1, increased; 2, decreased; 7, unchanged (all in comparison to the levels found in the non-treated model).
*Calculated from intake and body weight.
associated with left ventricular (LV) hypertrophy.94 Similarly, CSNA95 and RSNA96 are increased in congestive heart
failure, which provides the theoretical foundation for the
successful use of ␤-blockers in heart failure. Unfortunately,
although it has been extensively documented that MSNA is
increased in CRF,6,13 data on RSNA and CSNA are not
available for these patients before ESRD. The recent report
that carvedilol markedly improves survival in dialysis patients with congestive heart failure72 suggests that at this stage
CSNA is increased, just as it is in the general population.95
Whether sympathetic hyperactivity in CRF specifically contributes to the cardiac damage beyond its effect on blood
pressure has received relatively scant attention considering
the magnitude of the problem in the clinic. However, considering the excellent reduction in LV hypertrophy and hypertension achieved recently in the general population with an
ACE inhibitor plus a selective aldosterone blocker,97 it would
appear logical in predialysis patients to add a third-generation
␤-blocker to such a combination.
Conclusion
Although many experimental and clinical studies in diverse
models of renal disease document renoprotective effects of
␣-blockers and ␤-blockers, it remains unclear how adrenergic
blockade can best be used in chronic renal disease. There are
very few comparative trials. For instance, to date no comparison has been made between the third-generation ␤-blockers
and an AT-1 receptor blocker, whereas there is ample reason
to believe that both agents will also enhance NO activity.
Unbalanced NO versus Ang II and sympathetic activity is
characteristic of renal disease. In this setting, both Ang II and
sympathetic activity become damaging factors for kidney and
heart. Given the overwhelming evidence of renoprotection
and cardioprotection provided by interference with the reninangiotensin-aldosterone system, now is the time to investigate
whether complementary combinations are even more effective in the clinical setting.
Acknowledgment
Sandy van Laar is acknowledged for her expert secretarial assistance.
References
1. Owen WF, Jr. Patterns of care for patients with chronic kidney disease
in the United States: dying for improvement. J Am Soc Nephrol. 2003;
14:S76 –S80.
2. Lan HY, Mu W, Tomita N, Huang XR, Li JH, Zhu HJ, Morishita R,
Johnson RJ. Inhibition of renal fibrosis by gene transfer of inducible Smad7
using ultrasound-microbubble system in rat UUO model. J Am Soc Nephrol.
2003;14:1535–1548.
3. Isaka Y, Tsujie M, Ando Y, Nakamura H, Kaneda Y, Imai E, Hori M.
Transforming growth factor-beta 1 antisense oligodeoxynucleotides block
interstitial fibrosis in unilateral ureteral obstruction. Kidney Int. 2000;58:
1885–1892.
4. Ruggenenti P, Schieppati A, Remuzzi G. Progression, remission, regression
of chronic renal diseases. Lancet. 2001;357:1601–1608.
5. Shimizu H, Maruyama S, Yuzawa Y, Kato T, Miki Y, Suzuki S, Sato W,
Morita Y, Maruyama H, Egashira K, Matsuo S. Anti-monocyte chemoattractant protein-1 gene therapy attenuates renal injury induced by proteinoverload proteinuria. J Am Soc Nephrol. 2003;14:1496–1505.
6. Ligtenberg G, Blankestijn PJ, Oey PL, Klein IH, Dijkhorst-Oei LT,
Boomsma F, Wieneke GH, van Huffelen AC, Koomans HA. Reduction of
sympathetic hyperactivity by enalapril in patients with chronic renal failure.
N Engl J Med. 1999;340:1321–1328.
7. Manjunath G, Tighiouart H, Ibrahim H, MacLeod B, Salem DN, Griffith JL,
Coresh J, Levey AS, Sarnak MJ. Level of kidney function as a risk factor for
atherosclerotic cardiovascular outcomes in the community. J Am Coll
Cardiol. 2003;41:47–55.
8. Matsumura K, Tsuchihashi T, Fujii K, Iida M. Neural regulation of blood
pressure by leptin and the related peptides. Regul Pept. 2003;114:79–86.
9. Sharma K, Considine RV, Michael B, Dunn SR, Weisberg LS, Kurnik BR,
Kurnik PB, O’Connor J, Sinha M, Caro JF. Plasma leptin is partly cleared
by the kidney and is elevated in hemodialysis patients. Kidney Int. 1997;
51:1980–1985.
10. Wolf G, Chen S, Han DC, Ziyadeh FN. Leptin and renal disease. Am J
Kidney Dis. 2002;39:1–11.
11. Hall JE, Hildebrandt DA, Kuo J. Obesity hypertension: role of leptin and
sympathetic nervous system. Am J Hypertens. 2001;14:103S–115S
Joles and Koomans
Downloaded from http://hyper.ahajournals.org/ by guest on April 30, 2017
12. Anderstam B, Katzarski K, Bergstrom J. Serum levels of NG, NG-dimethylL-arginine, a potential endogenous nitric oxide inhibitor in dialysis patients.
J Am Soc Nephrol. 1997;8:1437–1442.
13. Klein IH, Ligtenberg G, Oey PL, Koomans HA, Blankestijn PJ. Sympathetic activity is increased in polycystic kidney disease and is associated
with hypertension. J Am Soc Nephrol. 2001;12:2427–2433.
14. Hausberg M, Kosch M, Harmelink P, Barenbrock M, Hohage H, Kisters K,
Dietl KH, Rahn KH. Sympathetic nerve activity in end-stage renal disease.
Circulation. 2002;106:1974 –1979.
15. Dibona GF, Jones SY, Sawin LL. Reflex influences on renal nerve activity
characteristics in nephrosis and heart failure. J Am Soc Nephrol. 1997;8:
1232–1239.
16. Vande Walle JG, Donckerwolcke RA, van Isselt JW, Derkx FH, Joles JA,
Koomans HA. Volume regulation in children with early relapse of minimalchange nephrosis with or without hypovolaemic symptoms. Lancet. 1995;
346:148–152.
17. Faber JE, Brody MJ. Afferent renal nerve-dependent hypertension following acute renal artery stenosis in the conscious rat. Circ Res. 1985;57:
676–688.
18. Miyajima E, Yamada Y, Yoshida Y, Matsukawa T, Shionoiri H, Tochikubo
O, Ishii M. Muscle sympathetic nerve activity in renovascular hypertension
and primary aldosteronism. Hypertension. 1991;17:1057–1062.
19. Ye S, Zhong H, Yanamadala V, Campese VM. Renal injury caused by
intrarenal injection of phenol increases afferent and efferent renal sympathetic nerve activity. Am J Hypertens. 2002;15:717–724.
20. Ye S, Nosrati S, Campese VM. Nitric oxide (NO) modulates the neurogenic
control of blood pressure in rats with chronic renal failure (CRF). J Clin
Invest. 1997;99:540–548.
21. Reid IA. Interactions between ANG II, sympathetic nervous system, and
baroreceptor reflexes in regulation of blood pressure. Am J Physiol. 1992;
262:E763–E778.
22. Aiello S, Noris M, Todeschini M, Zappella S, Foglieni C, Benigni A, Corna
D, Zoja C, Cavallotti D, Remuzzi G. Renal and systemic nitric oxide
synthesis in rats with renal mass reduction. Kidney Int. 1997;52:171–181.
23. Wagner L, Riggleman A, Erdely A, Couser W, Baylis C. Reduced nitric
oxide synthase activity in rats with chronic renal disease due to glomerulonephritis. Kidney Int. 2002;62:532–536.
24. Attia DM, Ni ZN, Boer P, Attia MA, Goldschmeding R, Koomans HA,
Vaziri ND, Joles JA. Proteinuria is preceded by decreased nitric oxide
synthesis and prevented by a NO donor in cholesterol-fed rats. Kidney Int.
2002;61:1776 –1787.
25. Benigni A, Zoja C, Noris M, Corna D, Benedetti G, Bruzzi I, Todeschini M,
Remuzzi G. Renoprotection by nitric oxide donor and lisinopril in the
remnant kidney model. Am J Kidney Dis. 1999;33:746–753.
26. Taal MW, Brenner BM. ACE-I vs angiotensin II receptor antagonists:
prevention of renal injury in chronic rat models. J Hum Hypertens. 1999;
13:S51–S56; discussion S61.
27. Atarashi K, Takagi M, Minami M, Ishiyama A. Effects of alacepril and
amlodipine on the renal injury induced by a high-cholesterol diet in rats.
J Hypertens. 1999;17:1983–1986.
28. Bataineh A, Raij L. Angiotensin II, nitric oxide, and end-organ damage in
hypertension. Kidney Int Suppl. 1998;68:S14–S19.
29. Mackie FE, Campbell DJ, Meyer TW. Intrarenal angiotensin and bradykinin
peptide levels in the remnant kidney model of renal insufficiency. Kidney
Int. 2001;59:1458 –1465.
30. Verhagen AM, Braam B, Boer P, Grone HJ, Koomans HA, Joles JA.
Losartan-sensitive renal damage caused by chronic NOS inhibition does not
involve increased renal angiotensin II concentrations. Kidney Int. 1999;56:
222–231.
31. Zanchi A, Schaad NC, Osterheld MC, Grouzmann E, Nussberger J, Brunner
HR, Waeber B. Effects of chronic NO synthase inhibition in rats on reninangiotensin system and sympathetic nervous system. Am J Physiol. 1995;
268:H2267–H2273.
32. Togashi H, Sakuma I, Yoshioka M, Kobayashi T, Yasuda H, Kitabatake A,
Saito H, Gross SS, Levi R. A central nervous system action of nitric oxide
in blood pressure regulation. J Pharmacol Exp Ther. 1992;262:343–347.
33. Dobrucki LW, Cabrera CL, Bohr DF, Malinski T. Central hypotensive
action of clonidine requires nitric oxide. Circulation. 2001;104:1884–1886.
34. Matsuoka H, Nishida H, Nomura G, Van Vliet BN, Toshima H. Hypertension induced by nitric oxide synthesis inhibition is renal nerve dependent.
Hypertension. 1994;23:971–975.
35. Podjarny E, Benchetrit S, Katz B, Green J, Bernheim J. Effect of methyldopa on renal function in rats with L-NAME-induced hypertension in
pregnancy. Nephron. 2001;88:354–359.
Sympathetic Activity in Renal Disease
705
36. Fitzgerald SM, Brands MW. Hypertension in L-NAME-treated diabetic rats
depends on an intact sympathetic nervous system. Am J Physiol Regul
Integr Comp Physiol. 2002;282:R1070–R1076.
37. Ashby B, Daniel JL, Smith JB. Mechanisms of platelet activation and
inhibition. Hematol Oncol Clin North Am 1990;4:1–26.
38. Nakashima A, Matsuoka H, Yasukawa H, Kohno K, Nishida H, Nomura G,
Imaizumi T, Morimatsu M. Renal denervation prevents intraglomerular
platelet aggregation and glomerular injury induced by chronic inhibition of
nitric oxide synthesis. Nephron. 1996;73:34–40.
39. Tojo A, Kobayashi N, Kimura K, Hirata Y, Matsuoka H, Yagi S, Omata M.
Effects of antihypertensive drugs on nitric oxide synthase activity in rat
kidney. Kidney Int Suppl. 1996;55:S138–S140.
40. Amann K, Koch A, Hofstetter J, Gross ML, Haas C, Orth SR, Ehmke H,
Rump LC, Ritz E. Glomerulosclerosis and progression: effect of subantihypertensive doses of alpha and beta blockers. Kidney Int. 2001;60:
1309–1323.
41. Fujihara CK, De Lourdes Noronha I, Malheiros, Antunes GR, de Oliveira
IB, Zatz R. Combined mycophenolate mofetil and losartan therapy arrests
established injury in the remnant kidney. J Am Soc Nephrol. 2000;11:
283–290.
42. Aizawa T, Ishizaka N, Taguchi J, Nagai R, Mori I, Tang SS, Ingelfinger JR,
Ohno M. Heme oxygenase-1 is upregulated in the kidney of angiotensin
II-induced hypertensive rats : possible role in renoprotection. Hypertension.
2000;35:800–806.
43. Erami C, Zhang H, Ho JG, French DM, Faber JE. Alpha(1)-adrenoceptor
stimulation directly induces growth of vascular wall in vivo. Am J Physiol
Heart Circ Physiol. 2002;283:H1577–H1587.
44. Johnson RJ, Gordon KL, Suga S, Duijvestijn AM, Griffin K, Bidani A.
Renal injury and salt-sensitive hypertension after exposure to catecholamines. Hypertension. 1999;34:151–159.
45. Rump LC, Amann K, Orth S, Ritz E. Sympathetic overactivity in renal
disease: a window to understand progression and cardiovascular complications of uraemia? Nephrol Dial Transplant. 2000;15:1735–1748.
46. Amann K, Rump LC, Simonaviciene A, Oberhauser V, Wessels S, Orth SR,
Gross ML, Koch A, Bielenberg GW, Van Kats JP, Ehmke H, Mall G, Ritz
E. Effects of low dose sympathetic inhibition on glomerulosclerosis and
albuminuria in subtotally nephrectomized rats. J Am Soc Nephrol. 2000;11:
1469–1478.
47. Barone FC, Nelson AH, Ohlstein EH, Willette RN, Sealey JE, Laragh JH,
Campbell WG, Jr., Feuerstein GZ. Chronic carvedilol reduces mortality and
renal damage in hypertensive stroke-prone rats. J Pharmacol Erxp The.
1996;279:948–955.
48. Wong VY, Laping NJ, Nelson AH, Contino LC, Olson BA, Gygielko E,
Campbell WG, Jr., Barone F, Brooks DP. Renoprotective effects of
carvedilol in hypertensive-stroke prone rats may involve inhibition of TGF
beta expression. Br J Pharmacol. 2001;134:977–984.
49. Griffin KA, Abu-Amarah I, Picken M, Bidani AK. Renoprotection by ACE
inhibition or aldosterone blockade is blood pressure-dependent. Hypertension. 2003;41:201–206.
50. Bidani AK, Griffin KA, Bakris G, Picken MM. Lack of evidence of blood
pressure-independent protection by renin-angiotensin system blockade after
renal ablation. Kidney Int. 2000;57:1651–1661.
51. Zhang H, Faber JE. Trophic effect of norepinephrine on arterial
intima-media and adventitia is augmented by injury and mediated by different alpha1-adrenoceptor subtypes. Circ Res. 2001;89:815–822.
52. Kang DH, Kanellis J, Hugo C, Truong L, Anderson S, Kerjaschki D,
Schreiner GF, Johnson RJ. Role of the microvascular endothelium in progressive renal disease. J Am Soc Nephrol. 2002;13:806–816.
53. Burnstock G. Purinergic signaling and vascular cell proliferation and death.
Arterioscler Thromb Vasc Biol. 2002;22:364–373.
54. Inscho EW. P2 receptors in regulation of renal microvascular function. Am J
Physiol Renal Physiol. 2001;280:F927–F944.
55. DiBona GF, Kopp UC. Neural control of renal function. Physiol Rev
1997;77:75–197.
56. Pavenstadt H, Kriz W, Kretzler M. Cell biology of the glomerular podocyte.
Physiol Rev. 2003;83:253–307.
57. Schwiebert EM, Kishore BK. Extracellular nucleotide signaling along the
renal epithelium. Am J Physiol Renal Physiol. 2001;280:F945–F963.
58. Kalinowski L, Dobrucki LW, Szczepanska-Konkel M, Jankowski M,
Martyniec L, Angielski S, Malinski T. Third-generation beta-blockers
stimulate nitric oxide release from endothelial cells through ATP efflux: a
novel mechanism for antihypertensive action. Circulation. 2003;107:
2747–2752.
59. Attia DM, Verhagen AM, Stroes ES, van Faassen EE, Grone HJ, De Kimpe
SJ, Koomans HA, Braam B, Joles JA. Vitamin E alleviates renal injury, but
706
60.
61.
62.
63.
64.
65.
Downloaded from http://hyper.ahajournals.org/ by guest on April 30, 2017
66.
67.
68.
69.
70.
71.
72.
73.
74.
75.
76.
77.
78.
79.
Hypertension
April 2004
not hypertension, during chronic nitric oxide synthase inhibition in rats.
J Am Soc Nephrol. 2001;12:2585–2593.
Wolf G, Butzmann U, Wenzel UO. The renin-angiotensin system and
progression of renal disease: from hemodynamics to cell biology. Nephron
Physiol 2003;93:P3–P13.
Aizawa T, Ishizaka N, Kurokawa K, Nagai R, Nakajima H, Taguchi J, Ohno
M. Different effects of angiotensin II and catecholamine on renal cell
apoptosis and proliferation in rats. Kidney Int 2001;59:645–653.
Rodriguez-Perez JC, Losada A, Anabitarte A, Cabrera J, Llobet J, Palop L,
Plaza C. Effects of the novel multiple-action agent carvedilol on severe
nephrosclerosis in renal ablated rats. J Pharmacol Exp Ther. 1997;283:
336–344.
Van den Branden C, Gabriels M, Vamecq J, Vanden Houte K, Verbeelen D.
Carvedilol protects against glomerulosclerosis in rat remnant kidney without
general changes in antioxidant enzyme status. A comparative study of two
beta-blocking drugs, carvedilol and propanolol. Nephron. 1997;77:
319–324.
Takamitsu Y, Nakanishi T, Nishihara F, Hasuike Y, Izumi M, Inoue T,
Hiraoka K, Itahana R, Miyagawa K. A nitric oxide-generating beta-blocking
agent prevents renal injury in the rat remnant kidney model. Comparative
study of two beta-blocking drugs, nipradilol and propranolol. Nephron
Physiol. 2003;93:42-P50.
Brooks DP, Short BG, Cyronak MJ, Contino LC, DiCristo M, Wang YX,
Ruffolo RR, Jr. Comparison between carvedilol and captopril in rats with
partial ablation-induced chronic renal failure. Br J Pharmacol. 1993;109:
581–586.
Erley CM, Rebmann S, Strobel U, Schmidt T, Wehrmann M, Osswald H,
Risler T. Effects of antihypertensive therapy on blood pressure and renal
function in rats with hypertension due to chronic blockade of nitric oxide
synthesis. Exp Nephrol. 1995;3:293–299.
Inada H, Ono H, Minami J, Ishimitsu T, Matsuoka H. Nipradilol prevents
L-NAME-exacerbated nephrosclerosis with decreasing of caspase-3
expression in SHR. Hypertens Res. 2002;25:433–440.
Wangensteen R, O’Valle F, Del Moral R, Vargas F, Osuna A. Chronic
alpha1-adrenergic blockade improves hypertension and renal injury in
L-NAME and low-renin L-NAME-DOCA hypertensive rats. Med Sci
Monit. 2002;8:BR378–BR384.
Kumar KV, Shifow AA, Naidu MU, Ratnakar KS. Carvedilol: a beta
blocker with antioxidant property protects against gentamicin-induced nephrotoxicity in rats. Life Sci. 2000;66:2603–2611.
Packer M, Fowler MB, Roecker EB, Coats AJ, Katus HA, Krum H,
Mohacsi P, Rouleau JL, Tendera M, Staiger C, Holcslaw TL, Amann-Zalan
I, DeMets DL. Effect of carvedilol on the morbidity of patients with severe
chronic heart failure: results of the carvedilol prospective randomized cumulative survival (COPERNICUS) study. Circulation. 2002;106:2194–2199.
Shlipak MG, Heidenreich PA, Noguchi H, Chertow GM, Browner WS,
McClellan MB. Association of renal insufficiency with treatment and
outcomes after myocardial infarction in elderly patients. Ann Intern Med.
2002;137:555–562.
Cice G, Ferrara L, D’Andrea A, D’Isa S, Di Benedetto A, Cittadini A, Russo
PE, Golino P, Calabro R. Carvedilol increases two-year survivalin dialysis
patients with dilated cardiomyopathy: a prospective, placebo-controlled
trial. J Am Coll Cardiol. 2003;41:1438 –1444.
Reddi AS, Nimmagadda VR, Lefkowitz A, Kuo HR, Bollineni JS. Effect of
antihypertensive therapy on renal injury in type 2 diabetic rats with hypertension. Hypertension. 2000;36:233–238.
Diuretic versus alpha-blocker as first-step antihypertensive therapy: final
results from the Antihypertensive and Lipid-Lowering Treatment to Prevent
Heart Attack Trial (ALLHAT). Hypertension. 2003;42:239–246.
Odoni G, Ogata H, Viedt C, Amann K, Ritz E, Orth SR. Cigarette smoke
condensate aggravates renal injury in the renal ablation model. Kidney Int.
2002;61:2090 –2098.
Vonend O, Marsalek P, Russ H, Wulkow R, Oberhauser V, Rump LC.
Moxonidine treatment of hypertensive patients with advanced renal failure.
J Hypertens. 2003;21:1709–1717.
Laverman GD, Remuzzi G. Antihypertensive therapy in chronic renal
disease: a place for sympathicolytic agents? J Hypertens. 2003;21:
1625–1626.
Komers R, Anderson S. Paradoxes of nitric oxide in the diabetic kidney.
Am J Physiol Renal Physiol. 2003;284:F1121–F1137.
Hoffman RP, Sinkey CA, Kienzle MG, Anderson EA. Muscle sympathetic
nerve activity is reduced in IDDM before overt autonomic neuropathy.
Diabetes. 1993;42:375–380.
80. Maliszewska-Scislo M, Scislo TJ, Rossi NF. Effect of blockade of endogenous angiotensin II on baroreflex function in conscious diabetic rats. Am J
Physiol Heart Circ Physiol. 2003;284:H1601–H1611.
81. Matsuoka H. Protective role of renal nerves in the development of diabetic
nephropathy. Diabetes Res. 1993;23:19–29.
82. Jyothirmayi GN, Alluru I, Reddi AS. Doxazosin prevents proteinuria and
glomerular loss of heparan sulfate in diabetic rats. Hypertension. 1996;27:
1108–1114.
83. Strojek K, Grzeszczak W, Gorska J, Leschinger MI, Ritz E. Lowering of
microalbuminuria in diabetic patients by a sympathicoplegic agent: novel
approach to prevent progression of diabetic nephropathy? J Am Soc
Nephrol. 2001;12:602–605.
84. D’Agord Schaan B, Lacchini S, Bertoluci MC, Irigoyen MC, Machado UF,
Schmid H. Impact of renal denervation on renal content of GLUT1, albuminuria and urinary TGF-beta1 in streptozotocin-induced diabetic rats.
Auton Neurosci. 2003;104:88–94.
85. Bohlen L, de Courten M, Weidmann P. Comparative study of the effect of
ACE-inhibitors and other antihypertensive agents on proteinuria in diabetic
patients. Am J Hypertens. 1994;7:84S–92S.
86. Pahor M, Psaty BM, Alderman MH, Applegate WB, Williamson JD,
Furberg CD. Therapeutic benefits of ACE inhibitors and other antihypertensive drugs in patients with type 2 diabetes. Diabetes Care. 2000;23:
888–892.
87. Kambham N, Markowitz GS, Valeri AM, Lin J, D’Agati VD. Obesityrelated glomerulopathy: an emerging epidemic. Kidney Int. 2001;59:
1498–1509.
88. Ernsberger P, Ishizuka T, Liu S, Farrell CJ, Bedol D, Koletsky RJ, Friedman
JE. Mechanisms of antihyperglycemic effects of moxonidine in the obese
spontaneously hypertensive Kopetzky rat (SHROB). J Pharmacol Exp
Ther. 1999;288:139–147.
89. Esler M, Rumantir M, Wiesner G, Kaye D, Hastings J, Lambert G. Sympathetic nervous system and insulin resistance: from obesity to diabetes.
Am J Hypertens. 2001;14:304S–309S.
90. Tornig J, Amann K, Ritz E, Nichols C, Zeier M, Mall G. Arteriolar wall
thickening, capillary rarefaction and interstitial fibrosis in the heart of rats
with renal failure:the effects of ramipril, nifedipine and moxonidine. J Am
Soc Nephrol. 1996;7:667–675.
91. Amann K, Greber D, Gharehbaghi H, Wiest G, Lange B, Ganten U,
Mattfeldt T, Mall G. Effects of nifedipine and moxonidine on cardiac
structure in spontaneously hypertensive rats. Stereological studies on
myocytes, capillaries, arteries, and cardiac interstitium. Am J Hypertens.
1992;5:76–83.
92. Barone FC, Campbell WG, Jr., Nelson AH, Feuerstein GZ. Carvedilol
prevents severe hypertensive cardiomyopathy and remodeling. J Hypertens.
1998;16:871–884.
93. Blezer EL, Nicolay K, Koomans HA, Joles JA. Losartan versus enalapril on
cerebral edema and proteinuria in stroke-prone hypertensive rats. Am J
Hypertens. 2001;14:54–61.
94. Schlaich MP, Kaye DM, Lambert E, Sommerville M, Socratous F, Esler
MD. Relation between cardiac sympathetic activity and hypertensive left
ventricular hypertrophy. Circulation. 2003;108:560–565.
95. Brunner-La Rocca HP, Esler MD, Jennings GL, Kaye DM. Effect of cardiac
sympathetic nervous activity on mode of death in congestive heart failure.
Eur Heart J. 2001;22:1136–1143.
96. Pliquett RU, Cornish KG, Peuler JD, Zucker IH. Simvastatin normalizes
autonomic neural control in experimental heart failure. Circulation. 2003;
107:2493–2498.
97. Pitt B, Reichek N, Willenbrock R, Zannad F, Phillips RA, Roniker B,
Kleiman J, Krause S, Burns D, Williams GH. Effects of eplerenone, enalapril, and eplerenone/enalapril in patients with essential hypertension and
left ventricular hypertrophy: the 4E-left ventricular hypertrophy study. Circulation. 2003;108:1831–1838.
98. Campese VM, Kogosov E, Koss M. Renal afferent denervation prevents the
progression of renal disease in the renal ablation model of chronic renal
failure in the rat. Am J Kidney Dis. 1995;26:861–865.
99. Irzyniec T, Mall G, Greber D, Ritz E. Beneficial effect of nifedipine and
moxonidine on glomerulosclerosis in spontaneously hypertensive rats. A
micromorphometric study. Am J Hypertens. 1992;5:437–443.
100. Pacca SR, de Azevedo AP, De Oliveira CF, De Luca IM, De Nucci G,
Antunes E. Attenuation of hypertension, cardiomyocyte hypertrophy, and
myocardial fibrosis by beta-adrenoceptor blockers in rats under long-term
blockade of nitric oxide synthesis. J Cardiovasc Pharmacol. 2002;39:
201–207.
Causes and Consequences of Increased Sympathetic Activity in Renal Disease
Jaap A. Joles and Hein A. Koomans
Downloaded from http://hyper.ahajournals.org/ by guest on April 30, 2017
Hypertension. 2004;43:699-706; originally published online February 23, 2004;
doi: 10.1161/01.HYP.0000121881.77212.b1
Hypertension is published by the American Heart Association, 7272 Greenville Avenue, Dallas, TX 75231
Copyright © 2004 American Heart Association, Inc. All rights reserved.
Print ISSN: 0194-911X. Online ISSN: 1524-4563
The online version of this article, along with updated information and services, is located on the
World Wide Web at:
http://hyper.ahajournals.org/content/43/4/699
Permissions: Requests for permissions to reproduce figures, tables, or portions of articles originally published
in Hypertension can be obtained via RightsLink, a service of the Copyright Clearance Center, not the Editorial
Office. Once the online version of the published article for which permission is being requested is located,
click Request Permissions in the middle column of the Web page under Services. Further information about
this process is available in the Permissions and Rights Question and Answer document.
Reprints: Information about reprints can be found online at:
http://www.lww.com/reprints
Subscriptions: Information about subscribing to Hypertension is online at:
http://hyper.ahajournals.org//subscriptions/