ArticlePDF AvailableLiterature Review

Sodium Intake as a Cardiovascular Risk Factor: A Narrative Review

MDPI
Nutrients
Authors:

Abstract and Figures

While sodium is essential for human homeostasis, current salt consumption far exceeds physiological needs. Strong evidence suggests a direct causal relationship between sodium intake and blood pressure (BP) and a modest reduction in salt consumption is associated with a meaningful reduction in BP in hypertensive as well as normotensive individuals. Moreover, while long-term randomized controlled trials are still lacking, it is reasonable to assume a direct relationship between sodium intake and cardiovascular outcomes. However, a consensus has yet to be reached on the effectiveness, safety and feasibility of sodium intake reduction on an individual level. Beyond indirect BP-mediated effects, detrimental consequences of high sodium intake are manifold and pathways involving vascular damage, oxidative stress, hormonal alterations, the immune system and the gut microbiome have been described. Globally, while individual response to salt intake is variable, sodium should be perceived as a cardiovascular risk factor when consumed in excess. Reduction of sodium intake on a population level thus presents a potential strategy to reduce the burden of cardiovascular disease worldwide. In this review, we provide an update on the consequences of salt intake on human health, focusing on BP and cardiovascular outcomes as well as underlying pathophysiological hypotheses.
This content is subject to copyright.
nutrients
Review
Sodium Intake as a Cardiovascular Risk Factor:
A Narrative Review
David A. Jaques 1, * , Gregoire Wuerzner 2and Belen Ponte 1


Citation: Jaques, D.A.; Wuerzner, G.;
Ponte, B. Sodium Intake as a
Cardiovascular Risk Factor: A
Narrative Review. Nutrients 2021,13,
3177. https://doi.org/10.3390/
nu13093177
Academic Editor: Hirofumi Tanaka
Received: 8 August 2021
Accepted: 10 September 2021
Published: 12 September 2021
Publisher’s Note: MDPI stays neutral
with regard to jurisdictional claims in
published maps and institutional affil-
iations.
Copyright: © 2021 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
1Division of Nephrology and Hypertension, Department of Medicine, Geneva University Hospitals,
1205 Geneva, Switzerland; belen.ponte@hcuge.ch
2Division of Nephrology and Hypertension, Department of Medicine, Lausanne University Hospitals,
1011 Lausanne, Switzerland; gregoire.wuerzner@chuv.ch
*Correspondence: David.Jaques@hcuge.ch
Abstract:
While sodium is essential for human homeostasis, current salt consumption far exceeds
physiological needs. Strong evidence suggests a direct causal relationship between sodium intake
and blood pressure (BP) and a modest reduction in salt consumption is associated with a meaningful
reduction in BP in hypertensive as well as normotensive individuals. Moreover, while long-term
randomized controlled trials are still lacking, it is reasonable to assume a direct relationship between
sodium intake and cardiovascular outcomes. However, a consensus has yet to be reached on the
effectiveness, safety and feasibility of sodium intake reduction on an individual level. Beyond indirect
BP-mediated effects, detrimental consequences of high sodium intake are manifold and pathways
involving vascular damage, oxidative stress, hormonal alterations, the immune system and the
gut microbiome have been described. Globally, while individual response to salt intake is variable,
sodium should be perceived as a cardiovascular risk factor when consumed in excess. Reduction
of sodium intake on a population level thus presents a potential strategy to reduce the burden of
cardiovascular disease worldwide. In this review, we provide an update on the consequences of
salt intake on human health, focusing on BP and cardiovascular outcomes as well as underlying
pathophysiological hypotheses.
Keywords: salt; sodium; blood pressure; hypertension; cardiovascular
1. Introduction
Sodium (Na+), contained in dietary salt, is essential for human homeostasis. For
millions of years, our ancestors ate less than 0.25 g of salt per day, while the current
average daily consumption approaches 10 g in most countries [
1
,
2
]. Such an increase
over a comparatively modest time span imposes a significant physiological challenge in
evolutionary terms. Excessive sodium intake is thought to adversely affect our health
through effects on blood pressure (BP) and cardiovascular damages. Consequently, three
million deaths were attributed to high salt intake in 2017 [
3
]. Given that the majority
of cardiovascular burden affects individuals with high-normal BP or mild hypertension,
dietary and lifestyle programs including salt reduction constitute attractive and simple
public health measures [
4
]. Despite general agreement that excessive sodium consumption
is globally harmful, controversies still exist on the net benefit of sodium intake reduction
on a population level and the levels that should be targeted [
5
]. Moreover, far from a
simplistic cause–effect relationship, pathophysiological mechanisms linking sodium intake
and cardiovascular outcomes are diverse and intricate.
In this paper, we review the available evidence on the association between sodium
intake, BP and cardiovascular diseases. We more specifically discuss major pathophysio-
logical hypotheses underlying this relationship.
Nutrients 2021,13, 3177. https://doi.org/10.3390/nu13093177 https://www.mdpi.com/journal/nutrients
Nutrients 2021,13, 3177 2 of 15
2. Sodium Intake, Blood Pressure and Cardiovascular Outcomes
2.1. Blood Pressure
Convincing evidence suggest a direct and positive association between sodium intake
and BP regulation. At the population level, the INTERSALT (International Study of Sodium,
Potassium and Blood Pressure) study was the first international study to look at this
association [
6
]. This cross-sectional analysis described the relationship between sodium
intake based on 24-h urine collection and BP in over 10,000 participants aged 20 to 50 from
39 countries. The authors reported a significant association between sodium excretion and
BP at the individual level. Furthermore, sodium intake was also associated with age-related
hypertension, suggesting that sodium could also have a long-term impact in addition
to its immediate effect on BP regulation. More recently, the PURE (Prospective Urban
Rural Epidemiology) study, a large international report including more than 100,000 adult
participants from 18 countries, was published [
7
]. A positive curvilinear relationship
between sodium intake and BP was described. In line with INTERSALT, this relationship
was stronger in older individuals and those consuming low potassium diets. The UK
Biobank study is the largest report to date with urinary electrolyte data and BP measurements
in more than 450,000 adult subjects [
8
]. Authors found a positive linear association between
urinary sodium excretion and BP. On top of those cross-sectional data, longitudinal studies
are also available. In the EPOGH (European Project on Genes in Hypertension) study,
investigators followed a group of 1499 participants without cardiovascular disease over
6.1 years [
9
]. Compared to baseline values, increased sodium intake was associated with
an increase in BP after adjusting for potential confounders. In such observational studies, a
systolic blood pressure increase of 2 to 3 mmHg for each 1 g/day increment in estimated
sodium excretion was generally reported [7,8].
In addition to epidemiological studies, numerous randomized controlled trials have
confirmed the impact of dietary sodium on BP values and control of hypertension (
Table 1
).
The DASH-sodium (Dietary Approaches to Stop Hypertension) study included around
400 pre-hypertensive individuals [
10
]. Authors evaluated the impact on BP control of three
different diets with various sodium intake (1.5 g/day, 2.4 g/day and 3.3 g/day) consumed
for 30 days. On average, systolic BP values decreased by 2.1 mmHg when comparing
3.3 g/day to 2.4 g/day diets. An additional 4.6 mmHg decrease was reported when com-
paring 2.4 g/day to 1.5 g/day diets, highlighting a dose-dependent relationship between
sodium consumption and BP regulation. In the TOHP-II (Trial of Hypertension Prevention)
study, investigators evaluated the impact of long-term sodium intake reduction and weight
loss on blood pressure in 2382 individuals not taking antihypertensive medications using
a factorial design [
11
]. Despite a sodium intake target below 1.8 g/day, mean sodium
intake was 3.1 g/day and 3.2 g/day at 18 and 36 months, respectively. In comparison,
mean sodium intake was 4.0 g/day at 36 months in the control group. Consequently, the
improvement in BP values and hypertension prevalence achieved in the intervention group
decreased over time. As such, the primary efficacy endpoint, defined as the mean decrease
in diastolic BP, was not significant at 36 months. The TONE (Trial of Nonpharmacological
Intervention in the Elderly) trial also tested the impact of sodium reduction and weight loss
on BP control and cardiovascular events in 975 treated hypertensive adults aged between
60 and 80 [
12
]. As in the TOHP-II study, the sodium intake target in the intervention
group was 1.8 g/day. At 3-months follow-up, a 3.4 mmHg reduction in systolic BP was
achieved in the intervention group. Thirty months later, a higher proportion of patients
were withdrawn from antihypertensive medication in the intervention group compared to
the control group.
Nutrients 2021,13, 3177 3 of 15
Table 1. Selected RCTs on sodium intake, blood pressure and cardiovascular outcomes.
Study Population Intervention Outcome
Sodium intake and blood pressure
DASH-sodium
n= 412
Pre-HT; Age 47;
Male 41%; White 40%
Sodium intakes of 3.3, 2.4 and
1.5 g/day
SBP reduction of 2.1 mmHg (3.3 vs 2.4 g/day)
and 4.6 mmHg (2.4 vs 1.5 g/day) (p< 0.001)
TOHP-II
n= 2382
Not HT; Age 43.9;
Male 65.7%; White 79.3%
Sodium intake reduction to
80 mmol/day DBP reduction of 0.7 mmHg (p= 0.10)
TONE
n= 975
Treated HT; Age 65.8;
Male 53%; White 76%
Sodium intake reduction to
80 mmol/day SBP reduction of 3.4 mmHg (p< 0.001)
Sodium intake and cardiovascular outcomes a
Morgan et al.
n= 77
Untreated HT; Age 57.1;
Male 100%; White NA
Sodium intake reduction to
70–100 mmol/day Relative risk of CV event: 1.16 (0.39–3.45)
TOHP-I
n= 744
Not HT; Age 43.4;
Male 71.4%; White 77.2%
Sodium intake reduction to
80 mmol/day Relative risk of CV event: 0.51 (0.29–0.91)
TOHP-II
n= 2382
Not HT; Age 43.9;
Male 65.7%; White 79.3%
Sodium intake reduction to
80 mmol/day Relative risk of CV event: 0.88 (0.65–1.20)
TONE
n= 975
Treated HT; Age 65.8;
Male 53%; White 76%
Sodium intake reduction to
80 mmol/day Relative risk of CV event: 0.80 (0.53–1.21)
Abbreviations: RCT, randomized controlled trial; HT, hypertension; SBP systolic blood pressure: DBP, diastolic blood pressure; CV,
cardiovascular. Age is expressed as mean.
a
: Represented by follow-up studies including individuals who previously participated in RCT
of sodium intake reduction. Data from Taylor et al. meta-analysis.
Finally, meta-analyses of interventional trials globally confirmed an antihypertensive
effect of sodium reduction. A first meta-analysis, which included 36 randomized controlled
trials and 6736 adult individuals showed that reduced sodium intake was associated with a
decrease in BP of 3.39/1.54 mmHg without an adverse effect on renal function, or metabolic
or endocrine profile [
13
]. Of note, in sensitivity analyses, taking into account the study’s
duration, the authors reported a decrease in this beneficial effect with longer follow-up.
A second meta-analysis included randomized controlled trials investigating a modest
reduction in sodium intake with a minimum follow-up of four weeks [
14
]. A total of
34 trials and 3230 participants were identified. Investigators showed that a sodium intake
reduction of 4.4 g/day decreased BP of 5.4/2.8 mmHg in hypertensive individuals and
2.4/1.0 mmHg in normotensive individuals.
Although the beneficial effect on BP is clear, it has been previously suggested that
sodium intake reduction could potentially lead to harmful consequences. A meta-analysis
of 167 trials randomizing patients to low versus high sodium diets concluded that sodium
reduction resulted in an increase in renin, aldosterone, catecholamine and cholesterol
levels [
15
]. However, this report included studies involving large reductions in sodium
intake over a very short period of time. Such an abrupt decrease in sodium consumption
is expected to enhance several compensatory mechanisms, in contrast to what has been
described with a modest reduction over longer periods [
16
]. Second, a dose-response
relationship between sodium intake and BP has consistently been shown across obser-
vational as well as interventional trials, suggesting that benefits of sodium reduction
could extend to very low values [
6
,
10
,
17
,
18
]. Third, studies have generally shown that
sodium reduction allowed a significantly greater BP fall in older, hypertensive and Afro-
American individuals [
6
,
12
]. Such findings can be linked to varying sensitivities of the
renin–angiotensin–aldosterone system (RAAS) regulation in different populations [
19
,
20
].
Finally, sodium reduction has synergistic effects with pharmacological and conservative
measures on BP control. The DASH-sodium trial showed that a combination of low sodium
and a healthy diet has a greater effect on BP reduction than individual measures [
10
]. The
TONE and TOHP-II trials reported similar additive effects of sodium reduction with weight
reduction [
11
,
12
]. Taking into account such compensatory mechanism, a randomized
Nutrients 2021,13, 3177 4 of 15
controlled trial showed that sodium reduction allowed for a further reduction of BP in
hypertensive individuals treated with captopril as compared to normal sodium intake [
21
].
Sodium intake has varying effects on different subjects. In its simplest definition
“salt sensitivity” is a physiological trait by which BP exhibits changes parallel to sodium
intake [
22
]. Conversely, in “salt resistant” individuals, BP does not vary according to salt
loading. In humans, salt sensitivity is a continuous, normally distributed, quantitative
characteristic and any distinction between salt sensitive and salt resistant individuals is,
thus, somewhat arbitrary [
23
]. Although the definition and identification of salt sensitivity
lacks uniformity, it is suggested that up to 50% of hypertensive and 25% of normotensive in-
dividuals are salt sensitive [
24
]. Overall, the elderly, women and those with Afro-American
ethnicity, as well as patients suffering from chronic kidney disease, diabetes and primary
aldosteronism, are more prone to salt sensitivity [
25
27
]. Given the pathophysiological
complexity of hypertension, identification of genetic factors associated with salt sensi-
tivity is an intensive field of research [
28
]. Gene–environment interactions have been
characterized where polymorphisms in RAAS genes modified the effect of sodium intake
on BP in Japanese workers [
29
]. Such considerations could partially explain heteroge-
neous findings in clinical studies as well as facilitate personalized approaches to sodium
intake recommendations.
Globally, the causal relationship between sodium intake and BP control is, thus, well
established, and modest reduction in salt consumption is associated with a meaningful
reduction in BP on a population level. However, as the full effect of sodium intake reduction
on BP control is not reached until several weeks, results of interventional trials could have
underestimated the magnitude of this effect as most studies lasted a few weeks only [
14
,
30
].
On the other hand, the few existing long-term interventional trials showed that maintaining
a lower salt intake on a prolonged time period is challenging from an individual perspective
given the societal food environment [11,31].
2.2. Cardiovascular Outcomes
As sodium intake reduction lowers BP in normotensive and hypertensive individuals,
it could also be expected to improve cardiovascular outcomes. However, evidence from
large, long-term, randomized controlled trials on an individual level is currently lacking.
Over the years, numerous cohort studies have been published exploring the rela-
tionship between sodium intake and cardiovascular outcomes. Several meta-analyses
have pooled those studies and come to the conclusion that salt consumption directly and
negatively impacts cardiovascular prognosis [
13
,
32
,
33
]. Interestingly, several independent
cohort studies have reported a U-shaped association where both low and high sodium
consumption increased the risk of cardiovascular events and mortality when compared
with moderate intake [
34
37
]. Despite this, major potential confounding factors impede
definite conclusions and a causative association between low sodium intake and increased
cardiovascular risk for several reasons. First, reverse causality likely introduces a signifi-
cant bias as those studies included subjects at high cardiovascular risk, probably already
advised to lower their salt consumption. The increased morbidity associated with de-
creased sodium intake might therefore represent the confounding effect of underlying
cardiovascular diseases. Second, most of those studies relied on a single morning fasting
urine sample to estimate 24 h sodium consumption. Kawasaki’s formula is usually used in
such circumstances, including parameters such as age, gender, height and weight [
38
]. As
these variables are themselves potentially associated with the adverse prognosis, this could
further confound the association between sodium intake and the considered outcomes. As
such, 24 h urinary collection should be considered the gold standard to reliably estimate
sodium intake in such a setting. Globally, cohort studies that relied on multiple 24 h urinary
collections universally reported a direct positive linear relationship between sodium intake
and cardiovascular events [
39
42
]. Moreover, post hoc analysis of the data from the TOHP
trial has shown that the association between mortality and sodium intake was J or U-shaped
only when using formula-derived spot estimates but linear when measured on 24 h urine
Nutrients 2021,13, 3177 5 of 15
collections [
43
]. In agreement with these reports, an observational study recently showed
that 24 h estimates of sodium consumption derived from formulae based on spot urine
measurement overestimated sodium consumption at lower levels and underestimated
sodium consumption at higher levels when compared to directly measured 24 h sodium
urinary excretion [
44
]. Consequently, while a linear association existed between systolic
BP and measured 24 h sodium excretion, formula-derived estimates resulted in a J-shaped
association, thus altering the true relationship between dietary sodium and BP.
As patients with chronic kidney disease are at increased risk of cardiovascular events
compared with the general population, the effect of sodium consumption in this specific
sub-group is of pivotal importance. To answer this question, a prospective cohort study
included more than 3700 participants with chronic kidney disease and evaluated the as-
sociation between urinary sodium excretion and clinical cardiovascular events with a
median follow-up of 6.8 years [
41
]. Higher sodium consumption was associated with
increased risk of cardiovascular events in this population. Patients suffering from heart
disease also constitute a sub-group of special relevance. Left ventricular hypertrophy
is considered a major predictor of cardiovascular prognosis and, since left ventricular
hypertrophy is directly caused by hypertension, it could be expected that salt consumption
would indirectly induce ventricular remodelling through raised BP [
45
]. However, there
exists increasing evidence suggesting that sodium intake may directly induce left ven-
tricular hypertrophy, independent of BP [
46
,
47
]. A review of nine cross-sectional studies
described a strong correlation between sodium intake as assessed by 24 h urinary collection
and left ventricular mass, independent of relevant confounders as well as BP [
48
]. In a
prospective cohort study including more than 10,000 participants, higher salt consumption
was associated with increased risk of developing heart failure over a follow-up of almost
20 years [
49
]. This association was, however, significant in overweight individuals only.
Conversely, a low salt diet is a mainstay of congestive heart failure management. As such,
in a cohort of 443 patients with preserved ejection fraction, those who were recommended
a sodium-restricted diet had lower risk of death and hospital readmission at 30 days [50].
Estimation of sodium consumption on a population level is more reliable than at an
individual level [51]. Overall, evidence derived from public health interventions describe
a robust relationship between sodium intake and cardiovascular outcomes. For example,
Finland, and more recently the UK, have led salt reduction campaigns in an effort to
improve national health and both were successful at significantly reducing BP and car-
diovascular burden over the course of several years [
52
,
53
]. In Finland, salt consumption
decreased from 14 g/day in 1972 to 9 g/day in 2002 [
53
]. During this time period, mean
systolic and diastolic BP decreased by 10 mmHg and cardiovascular mortality by 75%.
While other factors could have played a role, obesity and alcohol consumption have both
increased during this time period suggesting a pivotal impact of sodium restriction. In
the UK, salt consumption decreased from 9.5 g/day in 2003 to 8.1 g/day in 2011 [
52
] and
a mean decrease in systolic BP of 2.7 mmHg was reported after adjustment for potential
confounders, while stroke and ischemic heart disease mortality decreased by 36%.
Finally, in a cluster randomized controlled trial including five veteran’s retirement
homes in Taiwan [
54
], sodium intake was reduced from 5.2 to 3.8 g/day in the intervention
group in combination with increased potassium, resulting in a reduction in cardiovascular
mortality. Follow-up studies have also been conducted including individuals who previ-
ously participated in randomized controlled trials of sodium intake reduction (
Table 1
),
later aggregated in meta-analyses. Despite including seven studies pooling 6489 partici-
pants with or without hypertension, the first study found no significant association between
reduced sodium intake and cardiovascular events [
55
]. Authors concluded that there is
insufficient power to exclude a clinically important effect of reduced sodium consumption
on cardiovascular morbidity and called for future, large, long-term, randomized controlled
trials focusing on clinical outcomes. According to their own calculation, 2500 cardiovascu-
lar events would be needed to achieve sufficient power to detect a clinically meaningful
effect. Such a trial would require thousands of participants adhering to specific sodium
Nutrients 2021,13, 3177 6 of 15
diets over several years. Imposing a high salt diet on a large scale for an extended pe-
riod would raise serious ethical and methodological concerns, rendering such a study
unlikely to ever be conducted. A second meta-analysis conducted by another group later
reanalysed the same dataset in with a slightly different methodology [
56
]. Indeed, one
study was excluded from the pooled analysis as it involved heart failure patients who
were likely to be sodium depleted before randomization as they had already been treated
with furosemide, spironolactone, captopril and fluid restriction [
57
]. Then, instead of
considering two distinct groups of patients, normotensive and hypertensive individuals
were analysed collectively, thereby increasing statistical power. By implementing those
methodological modifications, authors showed that reduced sodium intake was associated
with a significant decrease in cardiovascular events and a non-significant trend toward
lower mortality.
Considering the available evidence, it is reasonable to assume a direct and continuous
relationship between sodium intake, BP control and cardiovascular outcomes from a popu-
lation standpoint. However, as definite evidence from long-term randomized controlled
trials is still lacking, a consensus has yet to be reached on the effectiveness, safety and
feasibility of sodium intake reduction on an individual level. Until then, it seems judicious
to routinely evaluate sodium intake in patients at high cardio-renal risk and recommend
adherence to low sodium diet as part of a multifaceted treatment approach in an effort to
reduce morbidity and mortality in those patients [58,59].
3. Pathophysiological Considerations
3.1. Basic Principles
Multiple mechanisms are responsible for the association between sodium intake and
BP. The key role of the kidney in this relationship has been clearly demonstrated in several
transplant experiments [
60
,
61
]. Guyton formalized the basic principle of kidney BP reg-
ulation as the pressure natriuresis response [
62
]. In its simplest form, this concept states
that when BP increases, the excess pressure causes the kidney to excrete more sodium and
water. This, subsequently, decreases extracellular blood volume and, thus, preload as well
as cardiac output, thereby restoring BP to lower levels. On the other hand, variations in
sodium intake induce parallel changes in plasma sodium content, both in hypertensive and
normotensive individuals [
63
,
64
]. A rise in plasma sodium increases osmolarity, thus induc-
ing a fluid shift from the intracellular to the extracellular compartment. A small increase
in plasma osmolarity also stimulates vasopressin secretion, thus resulting in water reten-
tion. Both these mechanisms restore plasma sodium to its original level but also increase
extracellular fluid volume thereby increasing BP [
65
]. Importantly, evidence exists that a
variation in plasma sodium can influence BP regulation independently of blood volume
variation [
66
]. Such additive effects could be mediated by the direct influence of plasma
sodium on the hypothalamus; the vasculature as well as the immune system [6769].
3.2. Organ Damage and Cardiovascular Impact
Sodium intake indirectly affects target organs via its effect on BP. Hypertension is
associated with endothelial dysfunction and platelet activation eventually resulting in
microvascular as well as macrovascular disease and target organ damage [
70
]. However,
evidence also points towards direct adverse consequences of sodium load on cardiovascular
prognosis independent of BP (Figure 1). In this section, we review the main mechanisms
involved in this pathophysiological process.
Nutrients 2021,13, 3177 7 of 15
Nutrients 2021, 13, x FOR PEER REVIEW 7 of 16
Figure 1. Pathophysiological pathways linking sodium intake to target organ damage independently of BP-mediated ef-
fects.
Excessive sodium has been shown to be involved in different pathways such as ox-
idative stress, inflammation and fibrosis, which are determinant in target organs being
damaged. In patients with non-diabetic chronic kidney disease, sodium restriction in-
creases the concentration of anti-inflammatory and anti-fibrotic peptides on top of RAAS
blockade, providing pathophysiological insights into the synergistic benefit of sodium
reduction and RAAS blockade [71]. Animal studies confirmed a direct pro-fibrotic effect
of sodium on glomeruli mediated via an increased local expression of transforming
growth factor (TGF) β [72,73]. Additionally, sodium intake directly influences nitric oxide
(NO) generation and local oxidative stress in rats kidneys [74,75]. Thus, in middle-aged
hypertensive adults, dietary sodium restriction largely reversed macro- and microvas-
cular endothelial dysfunction by enhancing NO bioavailability and decreasing oxidative
stress, thus supporting a direct vascular protective effect of sodium restriction beyond
any influence on BP regulation [76].
Some evidence suggests that sodium is involved in cellular senescence as a high
NaCl environment inhibits the activity of key components of the classical DNA damage
response, such as Mre11 exonuclease in cell culture [77,78]. Exposure to NaCl was also
associated, both in cell culture as well as in vivo, with increased senescence-associated
β-galactosidase activity and p16
INK4
expression as well as reduced levels of Hsp70, all of
which are indicative of cellular senescence [79]. High salt intake is also associated with
cell death in animal experiments as cytosolic accumulation of caspase-independent
apoptotic factors, such as apoptosis-induced factor (AIF) and HtrA2/Omi, was described
in response to sodium loading [80]. Finally, apoptosis signal-regulating kinase 1 (ASK1)
has been involved in sodium-induced organ damage in animal models. ASK1 deficiency
abolished sodium-induced cardiac and vascular pathological alterations in normotensive
mice [81]. In a second experimental study, ASK1 deficiency also improved aldoste-
rone/salt-induced cardiac inflammation and fibrosis. Furthermore, the enhancement of
NADPH oxidase-induced cardiac oxidative stress caused by aldosterone and sodium
was notably decreased by ASK1 deficiency [82]. A schematic representation of potential
pathways involved in sodium-induced cellular injury is given in Figure 2.
Figure 1.
Pathophysiological pathways linking sodium intake to target organ damage independently
of BP-mediated effects.
Excessive sodium has been shown to be involved in different pathways such as ox-
idative stress, inflammation and fibrosis, which are determinant in target organs being
damaged. In patients with non-diabetic chronic kidney disease, sodium restriction in-
creases the concentration of anti-inflammatory and anti-fibrotic peptides on top of RAAS
blockade, providing pathophysiological insights into the synergistic benefit of sodium
reduction and RAAS blockade [
71
]. Animal studies confirmed a direct pro-fibrotic ef-
fect of sodium on glomeruli mediated via an increased local expression of transforming
growth factor (TGF)
β
[
72
,
73
]. Additionally, sodium intake directly influences nitric oxide
(NO) generation and local oxidative stress in rats’ kidneys [
74
,
75
]. Thus, in middle-aged
hypertensive adults, dietary sodium restriction largely reversed macro- and microvascu-
lar endothelial dysfunction by enhancing NO bioavailability and decreasing oxidative
stress, thus supporting a direct vascular protective effect of sodium restriction beyond any
influence on BP regulation [76].
Some evidence suggests that sodium is involved in cellular senescence as a high
NaCl environment inhibits the activity of key components of the classical DNA damage
response, such as Mre11 exonuclease in cell culture [
77
,
78
]. Exposure to NaCl was also
associated, both in cell culture as well as
in vivo
, with increased senescence-associated
β
-galactosidase activity and p16
INK4
expression as well as reduced levels of Hsp70, all of
which are indicative of cellular senescence [
79
]. High salt intake is also associated with cell
death in animal experiments as cytosolic accumulation of caspase-independent apoptotic
factors, such as apoptosis-induced factor (AIF) and HtrA2/Omi, was described in response
to sodium loading [
80
]. Finally, apoptosis signal-regulating kinase 1 (ASK1) has been
involved in sodium-induced organ damage in animal models. ASK1 deficiency abolished
sodium-induced cardiac and vascular pathological alterations in normotensive mice [
81
].
In a second experimental study, ASK1 deficiency also improved aldosterone/salt-induced
cardiac inflammation and fibrosis. Furthermore, the enhancement of NADPH oxidase-
induced cardiac oxidative stress caused by aldosterone and sodium was notably decreased
by ASK1 deficiency [
82
]. A schematic representation of potential pathways involved in
sodium-induced cellular injury is given in Figure 2.
Nutrients 2021,13, 3177 8 of 15
Nutrients 2021, 13, x FOR PEER REVIEW 8 of 16
Figure 2. Schematic representation of potential pathways involved in sodium-induced cellular in-
jury. SA-beta-gal, senescence-associated β-galactosidase; AIF, apoptosis-induced factor; Nox,
NADPH oxidase; ASK1, apoptosis signal-regulating kinase 1.
In healthy adults, endothelial function, as measured by flow-mediated dilatation
and arterial stiffness, was negatively impacted by high sodium intake [83,84], and re-
ducing sodium intake had the opposite effect [85]. Importantly, the adverse influence of
sodium on vascular function was unequivocally found to be independent of BP [86–88].
Sodium intake was also positively and independently associated with renal resistive in-
dex (RRI) in an adult general population [89]. As RRI could represent systemic vascular
damage and correlates with adverse cardiovascular outcomes, this suggests that sodium
consumption impacts renal hemodynamic as a reflection of a broader systemic alteration
[90,91]. An effect of sodium intake on systemic vascular properties independent of BP has
been confirmed in other studies. High sodium consumption was associated with in-
creased arterial stiffness as measured by pulse wave velocity after adjustment for BP in
Chinese communities [92]. The same group showed that pulse wave velocity was de-
creased in Australian normotensive subjects adhering to a low sodium diet compared
with age and BP matched controls [93]. More recently, a meta-analysis comprising 11
randomized controlled trials and 14 independent cohorts reported a positive association
between sodium intake and pulse wave velocity beyond BP control. Globally, those
findings are all the more important in that the severity of endothelial dysfunction relates
to the global cardiovascular risk [94]. On a cellular level, dietary salt has direct effects on
vascular endothelium NO production [95]. In bovine aortic endothelial cells, sodium
exposure caused a significant decrease in NO synthase (NOS) activity in a
dose-dependent manner, thereby explaining a salt-induced reduction in endothelial NO
generation [96]. High salt intake led to increased superoxide production and decreased
NO bioavailability in mice aortas [97]. Furthermore, a high salt diet impaired aortic ring
endothelium-dependent relaxation via reduced NO levels and increased superoxide
production in rat aorta [98]. In spontaneously hypertensive rats, excessive dietary salt
decreased cyclic GMP production in the aorta, leading to the impairment of
NO-mediated vascular relaxation despite increased NO production [99]. Such alterations
Figure 2.
Schematic representation of potential pathways involved in sodium-induced cellular injury.
SA-beta-gal, senescence-associated
β
-galactosidase; AIF, apoptosis-induced factor; Nox, NADPH
oxidase; ASK1, apoptosis signal-regulating kinase 1.
In healthy adults, endothelial function, as measured by flow-mediated dilatation and
arterial stiffness, was negatively impacted by high sodium intake [
83
,
84
], and reducing
sodium intake had the opposite effect [
85
]. Importantly, the adverse influence of sodium
on vascular function was unequivocally found to be independent of BP [
86
88
]. Sodium
intake was also positively and independently associated with renal resistive index (RRI)
in an adult general population [
89
]. As RRI could represent systemic vascular damage
and correlates with adverse cardiovascular outcomes, this suggests that sodium consump-
tion impacts renal hemodynamic as a reflection of a broader systemic alteration [
90
,
91
].
An effect of sodium intake on systemic vascular properties independent of BP has been
confirmed in other studies. High sodium consumption was associated with increased
arterial stiffness as measured by pulse wave velocity after adjustment for BP in Chinese
communities [
92
]. The same group showed that pulse wave velocity was decreased in
Australian normotensive subjects adhering to a low sodium diet compared with age and BP
matched controls [
93
]. More recently, a meta-analysis comprising 11 randomized controlled
trials and 14 independent cohorts reported a positive association between sodium intake
and pulse wave velocity beyond BP control. Globally, those findings are all the more
important in that the severity of endothelial dysfunction relates to the global cardiovascular
risk [
94
]. On a cellular level, dietary salt has direct effects on vascular endothelium NO
production [
95
]. In bovine aortic endothelial cells, sodium exposure caused a significant
decrease in NO synthase (NOS) activity in a dose-dependent manner, thereby explaining a
salt-induced reduction in endothelial NO generation [
96
]. High salt intake led to increased
superoxide production and decreased NO bioavailability in mice aortas [
97
]. Furthermore,
a high salt diet impaired aortic ring endothelium-dependent relaxation via reduced NO
levels and increased superoxide production in rat aorta [
98
]. In spontaneously hypertensive
rats, excessive dietary salt decreased cyclic GMP production in the aorta, leading to the im-
pairment of NO-mediated vascular relaxation despite increased NO production [
99
]. Such
alterations of the NO/cyclic GMP system were restored by dietary salt restriction but not by
Nutrients 2021,13, 3177 9 of 15
antihypertensive therapy in a later study [
100
]. Activation of vasopressor mechanisms were
also described, as a high salt diet increased the expression of cytochrome p450 4A enzymes
in rat mesenteric arteries, thus upregulating the production of 20-hydroxyeicosatetraenoic
acid (20-HETE) [
101
]. In turn, 20-HETE contributed to the vasoconstrictor response to
norepinephrine in those arteries [
101
]. A schematic representation of potential pathways
involved in sodium-induced vascular alterations is given in Figure 3.
Nutrients 2021, 13, x FOR PEER REVIEW 9 of 16
of the NO/cyclic GMP system were restored by dietary salt restriction but not by anti-
hypertensive therapy in a later study [100]. Activation of vasopressor mechanisms were
also described, as a high salt diet increased the expression of cytochrome p450 4A en-
zymes in rat mesenteric arteries, thus upregulating the production of
20-hydroxyeicosatetraenoic acid (20-HETE) [101]. In turn, 20-HETE contributed to the
vasoconstrictor response to norepinephrine in those arteries [101]. A schematic repre-
sentation of potential pathways involved in sodium-induced vascular alterations is given
in Figure 3.
Figure 3. Schematic representation of potential pathways involved in sodium-induced vascular
alterations. VSM, vascular smooth muscle; NOS, NO synthase; NO, nitric oxide; cGMP, cyclic
GMP; 20-HETE, 20-hydroxyeicosatetraenoic acid.
The adverse effect of sodium on target organs could be partially mediated by hor-
monal interactions. In a case-control study including 21 patients with primary aldoste-
ronism and 21 hypertensive control patients, 24 h sodium excretion was associated with
left ventricular mass and thickness only in patients with primary aldosteronism [102].
Aldosterone excess may thus play a permissive role in sodium induced target organ
damage. Another study prospectively investigated the influence of sodium intake on
cardiac outcomes of patients both before and after treatment of primary aldosteronism
[103]. Interestingly, sodium intake interacted with aldosterone in inducing cardiac
changes over time, while left ventricular mass was associated with both sodium intake
and aldosterone levels before treatment. The decrease in ventricular mass obtained after
treatment was greater in patients whose sodium intake also decreased. Change of ven-
tricular mass was also associated with sodium intake independently of BP and other po-
tential confounders. In another study including 90 adults with essential hypertension, left
ventricular mass was associated with plasma aldosterone level after, but not prior to, in-
travenous saline load, implying that a limited ability of sodium to supress aldosterone
production could contribute to organ damage [104]. Finally, in a longitudinal study, 182
adults with essential hypertension and left ventricular hypertrophy were treated with
RAAS blockade [105]. The observed decrease in left ventricular mass over time was cor-
Figure 3.
Schematic representation of potential pathways involved in sodium-induced vascular
alterations. VSM, vascular smooth muscle; NOS, NO synthase; NO, nitric oxide; cGMP, cyclic GMP;
20-HETE, 20-hydroxyeicosatetraenoic acid.
The adverse effect of sodium on target organs could be partially mediated by hormonal
interactions. In a case-control study including 21 patients with primary aldosteronism and
21 hypertensive control patients, 24 h sodium excretion was associated with left ventricular
mass and thickness only in patients with primary aldosteronism [
102
]. Aldosterone excess
may thus play a permissive role in sodium induced target organ damage. Another study
prospectively investigated the influence of sodium intake on cardiac outcomes of patients
both before and after treatment of primary aldosteronism [
103
]. Interestingly, sodium intake
interacted with aldosterone in inducing cardiac changes over time, while left ventricular
mass was associated with both sodium intake and aldosterone levels before treatment.
The decrease in ventricular mass obtained after treatment was greater in patients whose
sodium intake also decreased. Change of ventricular mass was also associated with
sodium intake independently of BP and other potential confounders. In another study
including 90 adults with essential hypertension, left ventricular mass was associated with
plasma aldosterone level after, but not prior to, intravenous saline load, implying that
a limited ability of sodium to supress aldosterone production could contribute to organ
damage [
104
]. Finally, in a longitudinal study, 182 adults with essential hypertension
and left ventricular hypertrophy were treated with RAAS blockade [
105
]. The observed
decrease in left ventricular mass over time was correlated with change in BP, 24-h sodium
urinary excretion and plasma aldosterone concentration. At the end of the follow-up,
the combination of high sodium intake and high aldosterone levels was associated with
Nutrients 2021,13, 3177 10 of 15
increased left ventricular mass. In contrast, in patients with low sodium intake, no influence
of aldosterone levels was detected. These results together suggest that persistence of organ
damage despite adequate BP control may result from the combined effect of excessive
sodium intake and breakthrough of aldosterone despite pharmacological blockage. Basic
research studies also support the interplay between sodium and aldosterone in organ
damage physiopathology as aldosterone-induced superoxide over-production and vascular
smooth muscle hypertrophy in cell culture, a phenomenon synergistically augmented with
sodium chloride [106].
Finally, sodium could impact cardiovascular prognosis via other less studied path-
ways. Evidence suggests that sodium has immunomodulatory properties as interleukin-17
producing T lymphocytes are highly pro-inflammatory cells whose differentiation was
enhanced
in vitro
and
in vivo
by a modest sodium load [
107
]. Extensive renal T cell infil-
tration has otherwise been described in numerous
in vivo
experiments under a high salt
diet [
108
,
109
]. In parallel, interleukin-17 was reported to decrease nitric oxide bioavail-
ability in smooth muscle cells and fibroblast, thereby promoting endothelial dysfunction
and arterial stiffness [
67
]. The impact of dietary salt intake on the immune response has
also been described in humans in a post-hoc analysis of a controlled simulated spaceflight
program termed Mras520 [
110
]. Authors described an increase in inflammatory cytokines
(IL-6 and IL-23) as well as a decrease in anti-inflammatory cytokines (IL-10) in the plasma
of subjects submitted to high salt intake as compared to low salt intake. Recently, the gut
microbiome has been proposed as a potential intermediary between sodium intake and
clinical outcomes. The impact of a high sodium diet on gut microbial composition has been
illustrated in various animal models [
111
,
112
]. In mice, chronic high sodium load induced
qualitative and quantitative alterations in intestinal flora [
113
]. This study also revealed
that sodium altered intestinal immunological gene expression and enhanced gut perme-
ability as well as enteric bacterial translocation to the kidney. Excessive dietary sodium led
to expansion of interleukin-17 producing T cells in the small intestine of mice, resulting
in increased levels of circulating interleukin-17 [
114
]. High sodium intake affected mice
gut microbiome by depleting Lactobacillus murinus in another study [
115
]. Consequently,
treatment of those mice with L. murinus prevented salt-induced hypertension by modulat-
ing interleukin-17 producing T cells. Differences in gut microbial composition have been
shown between salt-sensitive and salt-resistant Dahl rats and faecal transplantation from
one strain to the other was able to change the BP pattern [116].
Altogether, the available evidence suggests that sodium not only affects target organs
via its indirect effect on BP but, clearly, also through complex interconnected pathways
involving oxidative, inflammatory, endocrine, immune and microbiological mechanisms.
4. Conclusions
The available evidence points toward a causal role of sodium intake on BP and
cardiovascular prognosis. While the pathophysiological link between hypertension and
cardiovascular events is relatively straightforward, a large body of data now suggests
that sodium directly damages target organs independently of BP control via multiple
intricate pathways. Although gaps in knowledge still exist, reduction in sodium intake
on a population level represents a feasible strategy to reduce the burden of cardiovascular
morbidity and mortality worldwide.
Author Contributions:
D.A.J. wrote the manuscript. G.W. and B.P. revised the manuscript and
gave final approval for publication. All authors have read and agreed to the published version of
the manuscript.
Funding: This research received no external funding.
Institutional Review Board Statement: Not applicable.
Informed Consent Statement: Not applicable.
Data Availability Statement: Not applicable.
Nutrients 2021,13, 3177 11 of 15
Acknowledgments:
We thank Eric Feraille (Department of Cellular Physiology and Metabolism,
University of Geneva) for the insights in the molecular pathways of sodium–cell interaction. We
also thank Jack Galliford (Department of Nephrology, North Bristol NHS Trust) for proofreading
the manuscript.
Conflicts of Interest: Authors declare no conflict of interest.
References
1. He, F.J.; MacGregor, G.A. Salt, blood pressure and cardiovascular disease. Curr. Opin. Cardiol. 2007,22, 298–305. [CrossRef]
2.
Thout, S.R.; Santos, J.A.; McKenzie, B.; Trieu, K.; Johnson, C.; McLean, R.; Arcand, J.A.; Campbell, N.R.C.; Webster, J. The Science
of Salt: Updating the evidence on global estimates of salt intake. J. Clin. Hypertens. 2019,21, 710–721. [CrossRef]
3.
Afshin, A.; Sur, P.J.; Fay, K.A.; Cornaby, L.; Ferrara, G.; Salama, J.S.; Mullany, E.C.; Abate, K.H.; Abbafati, C.; Abebe, Z.; et al.
Health effects of dietary risks in 195 countries, 1990–2017: A systematic analysis for the Global Burden of Disease Study 2017.
Lancet 2019,393, 1958–1972. [CrossRef]
4.
Lewington, S.; Clarke, R.; Qizilbash, N.; Peto, R.; Collins, R. Age-specific relevance of usual blood pressure to vascular mortality:
A meta-analysis of individual data for one million adults in 61 prospective studies. Lancet 2002,360, 1903–1913. [CrossRef]
5.
O’Donnell, M.; Mente, A.; Alderman, M.H.; Brady, A.J.B.; Diaz, R.; Gupta, R.; López-Jaramillo, P.; Luft, F.C.; Lüscher, T.F.; Mancia,
G.; et al. Salt and cardiovascular disease: Insufficient evidence to recommend low sodium intake. Eur. Heart J.
2020
,41, 3363–3373.
[CrossRef] [PubMed]
6.
Rose, G.; Stamler, J.; Stamler, R.; Elliott, P.; Marmot, M.; Pyorala, K.; Kesteloot, H.; Joossens, J.; Hansson, L.; Mancia, G.; et al.
Intersalt: An international study of electrolyte excretion and blood pressure. Results for 24 hour urinary sodium and potassium
excretion. Br. Med. J. 1988,297, 319–328. [CrossRef]
7.
Mente, A.; O’Donnell, M.J.; Rangarajan, S.; McQueen, M.J.; Poirier, P.; Wielgosz, A.; Morrison, H.; Li, W.; Wang, X.; Di, C.; et al.
Association of Urinary Sodium and Potassium Excretion with Blood Pressure. N. Engl. J. Med. 2014,371, 601–611. [CrossRef]
8.
Welsh, C.E.; Welsh, P.; Jhund, P.; Delles, C.; Celis-Morales, C.; Lewsey, J.D.; Gray, S.; Lyall, D.; Iliodromiti, S.; Gill, J.M.R.; et al.
Urinary Sodium Excretion, Blood Pressure, and Risk of Future Cardiovascular Disease and Mortality in Subjects without Prior
Cardiovascular Disease. Hypertension 2019,73, 1202–1209. [CrossRef] [PubMed]
9.
Stolarz-Skrzypek, K.; Kuznetsova, T.; Thijs, L.; Tikhonoff, V.; Seidlerová, J.; Richart, T.; Jin, Y.; Olszanecka, A.; Malyutina, S.;
Casiglia, E.; et al. Fatal and nonfatal outcomes, incidence of hypertension, and blood pressure changes in relation to urinary
sodium excretion. J. Am. Med. Assoc. 2011,305, 1777–1785. [CrossRef] [PubMed]
10.
Sacks, F.M.; Svetkey, L.P.; Vollmer, W.M.; Appel, L.J.; Bray, G.A.; Harsha, D.; Obarzanek, E.; Conlin, P.R.; Miller, E.R.; Simons-
Morton, D.G.; et al. Effects on Blood Pressure of Reduced Dietary Sodium and the Dietary Approaches to Stop Hypertension
(DASH) Diet. N. Engl. J. Med. 2001,344, 3–10. [CrossRef]
11.
Cutler, J.A. Effects of weight loss and sodium reduction intervention on blood pressure and hypertension incidence in overweight
people with high-normal blood pressure: The trials of hypertension prevention, phase II. Arch. Intern. Med.
1997
,157, 657–667.
[CrossRef]
12.
Whelton, P.K.; Appel, L.J.; Espeland, M.A.; Applegate, W.B.; Ettinger, W.H.; Kostis, J.B.; Kumanyika, S.; Lacy, C.R.; Johnson, K.C.;
Folmar, S.; et al. Sodium reduction and weight loss in the treatment of hypertension in older persons: A randomized controlled
trial of nonpharmacologic interventions in the elderly (TONE). J. Am. Med. Assoc. 1998,279, 839–846. [CrossRef]
13.
Aburto, N.J.; Ziolkovska, A.; Hooper, L.; Elliott, P.; Cappuccio, F.P.; Meerpohl, J.J. Effect of lower sodium intake on health:
Systematic review and meta-analyses. BMJ 2013,346, f1326. [CrossRef]
14.
He, F.J.; Li, J.; MacGregor, G.A. Effect of longer term modest salt reduction on blood pressure: Cochrane systematic review and
meta-analysis of randomised trials. BMJ 2013,346, f1325. [CrossRef]
15.
Graudal, N.A.; Hubeck-Graudal, T.; Jurgens, G. Effects of low sodium diet versus high sodium diet on blood pressure, renin,
aldosterone, catecholamines, cholesterol, and triglyceride. In Cochrane Database of Systematic Reviews; John Wiley & Sons, Ltd.:
Hoboken, NJ, USA, 2011.
16.
Rhee, O.J.; Rhee, M.Y.; Oh, S.W.; Shin, S.J.; Gu, N.; Nah, D.Y.; Kim, S.W.; Lee, J.H. Effect of sodium intake on renin level: Analysis
of general population and meta-analysis of randomized controlled trials. Int. J. Cardiol. 2016,215, 120–126. [CrossRef]
17. He, F.J.; MacGregor, G.A. How Far Should Salt Intake Be Reduced? Hypertension 2003,42, 1093–1099. [CrossRef]
18.
Macgregor, G.A.; Sagnella, G.A.; Markandu, N.D.; Singer, D.R.J.; Cappuccio, F.P. Double-blind study of three sodium intakes and
long-term effects of sodium restriction in essential hypertension. Lancet 1989,334, 1244–1247. [CrossRef]
19.
He, F.J.; Markandu, N.D.; MacGregor, G.A. Importance of the renin system for determining blood pressure fall with acute salt
restriction in hypertensive and normotensive whites. Hypertension 2001,38, 321–325. [CrossRef] [PubMed]
20.
He, F.J.; Markandu, N.D.; Sagnella, G.A.; MacGregor, G.A. Importance of the renin system in determining blood pressure fall
with salt restriction in black and white hypertensives. Hypertension 1998,32, 820–824. [CrossRef] [PubMed]
21.
MacGregor, G.A.; Markandu, N.D.; Jsinger, D.R.; Cappuccio, F.P.; Shore, A.C.; Sagnella, G.A. Moderate sodium restriction with
angiotensin converting enzyme inhibitor in essential hypertension: A double blind study. Br. Med. J. (Clin. Res. Ed.)
1987
,294,
531–534. [CrossRef] [PubMed]
Nutrients 2021,13, 3177 12 of 15
22.
Elijovich, F.; Weinberger, M.H.; Anderson, C.A.M.; Appel, L.J.; Bursztyn, M.; Cook, N.R.; Dart, R.A.; Newton-Cheh, C.H.; Sacks,
F.M.; Laffer, C.L. Salt sensitivity of blood pressure: A scientific statement from the American Heart Association. Hypertension
2016,68, e7–e46. [CrossRef]
23.
Weinberger, M.H.; Miller, J.Z.; Luft, F.C.; Grim, C.E.; Fineberg, N.S. Definitions and characteristics of sodium sensitivity and blood
pressure resistance. Hypertension 1986,8, II-127–II-134. [CrossRef]
24. Balafa, O.; Kalaitzidis, R.G. Salt sensitivity and hypertension. J. Hum. Hypertens. 2021,35, 184–192. [CrossRef]
25.
Rocchini, A.P. Obesity hypertension, salt sensitivity and insulin resistance. Nutr. Metab. Cardiovasc. Dis.
2000
,10, 287–294.
[PubMed]
26.
Weir, M.R.; Chrysant, S.G.; McCarron, D.A.; Canossa-Terris, M.; Cohen, J.D.; Gunter, P.A.; Lewin, A.J.; Mennella, R.F.; Kirkegaard,
L.W.; Hamilton, J.H.; et al. Influence of race and dietary salt on the antihypertensive efficacy of an angiotensin-converting enzyme
inhibitor or a calcium channel antagonist in salt-sensitive hypertensives. Hypertension
1998
,31, 1088–1096. [CrossRef] [PubMed]
27.
He, J.; Gu, D.; Chen, J.; Jaquish, C.E.; Rao, D.C.; Hixson, J.E.; Chen, J.C.; Duan, X.; Huang, J.F.; Chen, C.S.; et al. Gender difference
in blood pressure responses to dietary sodium intervention in the GenSalt study. J. Hypertens.
2009
,27, 48–54. [CrossRef]
[PubMed]
28.
Feng, W.; Dell’Italia, L.J.; Sanders, P.W. Novel paradigms of salt and hypertension. J. Am. Soc. Nephrol.
2017
,28, 1362–1369.
[CrossRef] [PubMed]
29. Zhang, L.; Miyaki, K.; Araki, J.; Song, Y.; Kimura, T.; Omae, K.; Muramatsu, M. Interaction of angiotensin I-Converting enzyme
insertion-deletion polymorphism and daily salt intake influences hypertension in Japanese men. Hypertens. Res.
2006
,29, 751–758.
[CrossRef] [PubMed]
30.
Juraschek, S.P.; Woodward, M.; Sacks, F.M.; Carey, V.J.; Miller, E.R.; Appel, L.J. Time Course of Change in Blood Pressure from
Sodium Reduction and the DASH Diet. Hypertension 2017,70, 923–929. [CrossRef]
31.
Whelton, P.K.; Appel, L.; Charleston, J. The Effects of Nonpharmacologic Interventions on Blood Pressure of Persons With High
Normal Levels: Results of the Trials of Hypertension Prevention, Phase I. J. Am. Med. Assoc. 1992,267, 1213–1220. [CrossRef]
32.
Strazzullo, P.; D’Elia, L.; Kandala, N.B.; Cappuccio, F.P. Salt intake, stroke, and cardiovascular disease: Meta-analysis of
prospective studies. BMJ 2009,339, 1296. [CrossRef]
33.
Poggio, R.; Gutierrez, L.; Matta, M.G.; Elorriaga, N.; Irazola, V.; Rubinstein, A. Daily sodium consumption and CVD mortality
in the general population: Systematic review and meta-analysis of prospective studies. Public Health Nutr.
2015
,18, 695–704.
[CrossRef]
34.
Graudal, N.; Jürgens, G.; Baslund, B.; Alderman, M.H. Compared with usual sodium intake, low- and excessive-sodium diets are
associated with increased mortality: A meta-analysis. Am. J. Hypertens. 2014,27, 1129–1137. [CrossRef] [PubMed]
35.
Mente, A.; O’Donnell, M.; Rangarajan, S.; Dagenais, G.; Lear, S.; McQueen, M.; Diaz, R.; Avezum, A.; Lopez-Jaramillo, P.; Lanas,
F.; et al. Associations of urinary sodium excretion with cardiovascular events in individuals with and without hypertension: A
pooled analysis of data from four studies. Lancet 2016,388, 465–475. [CrossRef]
36.
O’Donnell, M.J.; Yusuf, S.; Mente, A.; Gao, P.; Mann, J.F.; Teo, K.; McQueen, M.; Sleight, P.; Sharma, A.M.; Dans, A.; et al. Urinary
sodium and potassium excretion and risk of cardiovascular events. J. Am. Med. Assoc. 2011,306, 2229–2238. [CrossRef]
37.
O’Donnell, M.; Mente, A.; Rangarajan, S.; McQueen, M.J.; Wang, X.; Liu, L.; Yan, H.; Lee, S.F.; Mony, P.; Devanath, A.; et al.
Urinary Sodium and Potassium Excretion, Mortality, and Cardiovascular Events. N. Engl. J. Med.
2014
,371, 612–623. [CrossRef]
[PubMed]
38.
Kawasaki, T.; Itoh, K.; Uezono, K.; Sasaki, H. A simple method for estimating 24 h urinary sodium and potassium excretion from
second morning voiding urine specimen in adults. Clin. Exp. Pharmacol. Physiol. 1993,20, 7–14. [CrossRef]
39.
Cook, N.R.; Appel, L.J.; Whelton, P.K. Sodium Intake and All-Cause Mortality Over 20 Years in the Trials of Hypertension
Prevention. J. Am. Coll. Cardiol. 2016,68, 1609–1617. [CrossRef] [PubMed]
40.
Olde Engberink, R.H.G.; Van Den Hoek, T.C.; Van Noordenne, N.D.; Van Den Born, B.J.H.; Peters-Sengers, H.; Vogt, L. Use of a
single baseline versus multiyear 24-hour urine collection for estimation of long-term sodium intake and associated cardiovascular
and renal risk. Circulation 2017,136, 917–926. [CrossRef]
41.
Mills, K.T.; Chen, J.; Yang, W.; Appel, L.J.; Kusek, J.W.; Alper, A.; Delafontaine, P.; Keane, M.G.; Mohler, E.; Ojo, A.; et al. Sodium
excretion and the risk of cardiovascular disease in patients with chronic kidney disease. J. Am. Med. Assoc.
2016
,315, 2200–2210.
[CrossRef]
42.
Cook, N.R.; Appel, L.J.; Whelton, P.K. Lower levels of sodium intake and reduced cardiovascular risk. Circulation
2014
,129,
981–989. [CrossRef]
43.
He, F.J.; Ma, Y.; Campbell, N.R.C.; Macgregor, G.A.; Cogswell, M.E.; Cook, N.R. Formulas to estimate dietary sodium intake from
spot urine alter sodium-mortality relationship. Hypertension 2019,74, 572–580. [CrossRef] [PubMed]
44.
Naser, A.M.; He, F.J.; Rahman, M.; Campbell, N.R.C. Spot Urine Formulas to Estimate 24-Hour Urinary Sodium Excretion Alter
the Dietary Sodium and Blood Pressure Relationship. Hypertension 2021,77, 2127–2137. [CrossRef] [PubMed]
45.
Levy, D.; Garrison, R.J.; Savage, D.D.; Kannel, W.B.; Castelli, W.P. Prognostic Implications of Echocardiographically Determined
Left Ventricular Mass in the Framingham Heart Study. N. Engl. J. Med. 1990,322, 1561–1566. [CrossRef] [PubMed]
46.
Jin, Y.; Kuznetsova, T.; Maillard, M.; Richart, T.; Thijs, L.; Bochud, M.; Herregods, M.C.; Burnier, M.; Fagard, R.; Staessen, J.A.
Independent relations of left ventricular structure with the 24-hour urinary excretion of sodium and aldosterone. Hypertension
2009,54, 489–495. [CrossRef] [PubMed]
Nutrients 2021,13, 3177 13 of 15
47.
Burnier, M.; Phan, O.; Wang, Q. High salt intake: A cause of blood pressure-independent left ventricular hypertrophy? Nephrol.
Dial. Transplant. 2007,22, 2426–2429. [CrossRef]
48.
Messerli, F.H.; Schmieder, R.E.; Weir, M.R. Salt: A perpetrator of hypertensive target organ disease? Arch. Intern. Med.
1997
,157,
2449–2452. [CrossRef] [PubMed]
49.
He, J.; Ogden, L.G.; Bazzano, L.A.; Vupputuri, S.; Loria, C.; Whelton, P.K. Dietary sodium intake and incidence of congestive
heart failure in overweight US men and women: First national health and nutrition examination survey epidemiologic follow-up
study. Arch. Intern. Med. 2002,162, 1619–1624. [CrossRef] [PubMed]
50.
Hummel, S.L.; DeFranco, A.C.; Skorcz, S.; Montoye, C.K.; Koelling, T.M. Recommendation of Low-Salt Diet and Short-term
Outcomes in Heart Failure with Preserved Systolic Function. Am. J. Med. 2009,122, 1029–1036. [CrossRef]
51.
Cogswell, M.E.; Mugavero, K.; Bowman, B.A.; Frieden, T.R. Dietary Sodium and Cardiovascular Disease Risk—Measurement
Matters. N. Engl. J. Med. 2016,375, 580–586. [CrossRef]
52.
He, F.J.; Pombo-Rodrigues, S.; MacGregor, G.A. Salt reduction in England from 2003 to 2011: Its relationship to blood pressure,
stroke and ischaemic heart disease mortality. BMJ Open 2014,4, 4549. [CrossRef]
53.
Laatikainen, T.; Critchley, J.; Vartiainen, E.; Salomaa, V.; Ketonen, M.; Capewell, S. Explaining the decline in coronary heart
disease mortality in Finland between 1982 and 1997. Am. J. Epidemiol. 2005,162, 764–773. [CrossRef]
54.
Chang, H.Y.; Hu, Y.W.; Yue, C.S.J.; Wen, Y.W.; Yeh, W.T.; Hsu, L.S.; Tsai, S.Y.; Pan, W.H. Effect of potassium-enriched salt on
cardiovascular mortality and medical expenses of elderly men. Am. J. Clin. Nutr. 2006,83, 1289–1296. [CrossRef]
55.
Taylor, R.S.; Ashton, K.E.; Moxham, T.; Hooper, L.; Ebrahim, S. Reduced dietary salt for the prevention of cardiovascular disease.
Cochrane Database Syst. Rev. 2011,2011, CD009217.
56.
He, F.J.; MacGregor, G.A. Salt reduction lowers cardiovascular risk: Meta-analysis of outcome trials. Lancet
2011
,378, 380–382.
[CrossRef]
57.
Paterna, S.; Gaspare, P.; Fasullo, S.; Sarullo, F.M.; Di Pasquale, P. Normal-sodium diet compared with low-sodium diet in
compensated congestive heart failure: Is sodium an old enemy or a new friend? Clin. Sci. 2008,114, 221–230. [CrossRef]
58.
Sasso, F.C.; De Nicola, L.; Carbonara, O.; Nasti, R.; Minutolo, R.; Salvatore, T.; Conte, G.; Torella, R. Cardiovascular risk factors
and disease management in type 2 diabetic patients with diabetic nephropathy. Diabetes Care
2006
,29, 498–503. [CrossRef]
[PubMed]
59.
Sasso, F.C.; Pafundi, P.C.; Simeon, V.; De Nicola, L.; Chiodini, P.; Galiero, R.; Rinaldi, L.; Nevola, R.; Salvatore, T.; Sardu, C.; et al.
Efficacy and durability of multifactorial intervention on mortality and MACEs: A randomized clinical trial in type-2 diabetic
kidney disease. Cardiovasc. Diabetol. 2021,20, 145. [CrossRef]
60.
Dahl, L.; Heine, M.; Thompson, K. Genetic influence of the kidneys on blood pressure. Evidence from chronic renal homografts
in rats with opposite predispositions to hypertension. Circ. Res. 1977,40, 94–101. [CrossRef]
61.
Dahl, L.K.; Heine, M.; Thompson, K. Genetic Influence of Renal Homografts on the Blood Pressure of Rats from Different Strains.
Proc. Soc. Exp. Biol. Med. 1972,140, 852–856. [CrossRef] [PubMed]
62. Guyton, A.C. Blood pressure control-Special role of the kidneys and body fluids. Science 1991,252, 1813–1816. [CrossRef]
63. de Wardener, H.E.; He, F.J.; MacGregor, G.A. Plasma sodium and hypertension. Kidney Int. 2004,66, 2454–2466. [PubMed]
64.
He, F.J.; Markandu, N.D.; Sagnella, G.A.; De Wardener, H.E.; MacGregor, G.A. Plasma sodium: Ignored and underestimated.
Hypertension 2005,45, 98–102. [CrossRef] [PubMed]
65.
He, F.J.; Markandu, N.D.; Sagnella, G.A.; MacGregor, G.A. Effect of salt intake on renal excretion of water in humans. Hypertension
2001,38, 317–320. [CrossRef] [PubMed]
66.
Friedman, S.M.; McIndoe, R.A.; Tanaka, M. The relation of blood sodium concentration to blood pressure in the rat. J. Hypertens.
1990,8, 61–66. [CrossRef] [PubMed]
67.
McMaster, W.G.; Kirabo, A.; Madhur, M.S.; Harrison, D.G. Inflammation, Immunity, and Hypertensive End-Organ Damage. Circ.
Res. 2015,116, 1022–1033. [CrossRef]
68.
Edwards, D.G.; Farquhar, W.B. Vascular effects of dietary salt. Curr. Opin. Nephrol. Hypertens.
2015
,24, 8–13. [CrossRef] [PubMed]
69. De Wardener, H.E. The hypothalamus and hypertension. Physiol. Rev. 2001,81, 1599–1658. [CrossRef]
70.
Nadar, S.; Tayebjee, M.; Messerli, F.; Lip, G. Target Organ Damage in Hypertension: Pathophysiology and Implications for Drug
Therapy. Curr. Pharm. Des. 2006,12, 1581–1592. [CrossRef] [PubMed]
71.
Kwakernaak, A.J.; Waanders, F.; Slagman, M.C.J.; Dokter, M.M.; Laverman, G.D.; De Boer, R.A.; Navis, G. Sodium restriction
on top of renin-angiotensin-aldosterone system blockade increases circulating levels of N-acetyl-seryl-aspartyl-lysyl-proline in
chronic kidney disease patients. J. Hypertens. 2013,31, 2425–2432. [CrossRef]
72.
Yu, H.C.M.; Burrell, L.M.; Black, M.J.; Wu, L.L.; Dilley, R.J.; Cooper, M.E.; Johnston, C.I. Salt induces myocardial and renal fibrosis
in normotensive and hypertensive rats. Circulation 1998,98, 2621–2628. [CrossRef]
73.
Ying, W.Z.; Sanders, P.W. Dietary salt modulates renal production of transforming growth factor-
β
in rats. Am. J. Physiol-Ren.
Physiol. 1998,274, F635–F641. [CrossRef] [PubMed]
74.
Kitiyakara, C.; Chabrashvili, T.; Chen, Y.; Blau, J.; Karber, A.; Aslam, S.; Welch, W.J.; Wilcox, C.S. Salt Intake, Oxidative Stress, and
Renal Expression of NADPH Oxidase and Superoxide Dismutase. J. Am. Soc. Nephrol.
2003
,14, 2775–2782. [CrossRef] [PubMed]
75.
Tojo, A.; Kimoto, M.; Wilcox, C.S. Renal expression of constitutive NOS and DDAH: Separate effects of salt intake and angiotensin.
Kidney Int. 2000,58, 2075–2083. [CrossRef]
Nutrients 2021,13, 3177 14 of 15
76.
Jablonski, K.L.; Racine, M.L.; Geolfos, C.J.; Gates, P.E.; Chonchol, M.; McQueen, M.B.; Seals, D.R. Dietary sodium restriction
reverses vascular endothelial dysfunction in middle-aged/older adults with moderately elevated systolic blood pressure. J. Am.
Coll. Cardiol. 2013,61, 335–343. [CrossRef]
77.
Dmitrieva, N.I.; Burg, M.B. Living with DNA breaks is an everyday reality for cells adapted to high NaCl. Cell Cycle
2004
,3,
559–561. [CrossRef]
78.
Dmitrieva, N.I.; Bulavin, D.V.; Burg, M.B. High NaCl causes Mre11 to leave the nucleus, disrupting DNA damage signaling and
repair. Am. J. Physiol-Ren. Physiol. 2003,285, F266–F274. [CrossRef]
79. Dmitrieva, N.I.; Burg, M.B. High NaCl promotes cellular senescence. Cell Cycle 2007,6, 3108–3113. [CrossRef]
80.
Siu, P.M.; Bae, S.; Bodyak, N.; Rigor, D.L.; Kang, P.M. Response of caspase-independent apoptotic factors to high salt diet-induced
heart failure. J. Mol. Cell. Cardiol. 2007,42, 678–686. [CrossRef]
81.
Kataoka, K.; Tokutomi, Y.; Yamamoto, E.; Nakamura, T.; Fukuda, M.; Dong, Y.F.; Ichijo, H.; Ogawa, H.; Kim-Mitsuyama, S.
Apoptosis signal-regulating kinase 1 deficiency eliminates cardiovascular injuries induced by high-salt diet. J. Hypertens.
2011
,29,
76–84. [CrossRef] [PubMed]
82.
Nakamura, T.; Kataoka, K.; Fukuda, M.; Nako, H.; Tokutomi, Y.; Dong, Y.F.; Ichijo, H.; Ogawa, H.; Kim-Mitsuyama, S. Critical role
of apoptosis signal-regulating kinase 1 in aldosterone/salt-induced cardiac inflammation and fibrosis. Hypertension
2009
,54,
544–551. [CrossRef]
83.
Dickinson, K.M.; Clifton, P.M.; Burrell, L.M.; Barrett, P.H.R.; Keogh, J.B. Postprandial effects of a high salt meal on serum sodium,
arterial stiffness, markers of nitric oxide production and markers of endothelial function. Atherosclerosis
2014
,232, 211–216.
[CrossRef]
84.
Dickinson, K.M.; Clifton, P.M.; Keogh, J.B. Endothelial function is impaired after a high-salt meal in healthy subjects. Am. J. Clin.
Nutr. 2011,93, 500–505. [CrossRef]
85. Dickinson, K.M.; Clifton, P.M.; Keogh, J.B. A reduction of 3 g/day from a usual 9 g/day salt diet improves endothelial function
and decreases endothelin-1 in a randomised cross_over study in normotensive overweight and obese subjects. Atherosclerosis
2014,233, 32–38. [CrossRef]
86.
Matthews, E.L.; Brian, M.S.; Ramick, M.G.; Lennon-Edwards, S.; Edwards, D.G.; Farquhar, W.B. High dietary sodium reduces
brachial artery flow-mediated dilation in humans with salt-sensitive and salt-resistant blood pressure. J. Appl. Physiol.
2015
,118,
1510–1515. [CrossRef]
87.
DuPont, J.J.; Greaney, J.L.; Wenner, M.M.; Lennon-Edwards, S.L.; Sanders, P.W.; Farquhar, W.B.; Edwards, D.G. High dietary
sodium intake impairs endothelium-dependent dilation in healthy salt-resistant humans. J. Hypertens.
2013
,31, 530–536.
[CrossRef] [PubMed]
88.
Greaney, J.L.; Dupont, J.J.; Lennon-Edwards, S.L.; Sanders, P.W.; Edwards, D.G.; Farquhar, W.B. Dietary sodium loading impairs
microvascular function independent of blood pressure in humans: Role of oxidative stress. J. Physiol.
2012
,590, 5519–5528.
[CrossRef]
89.
Jaques, D.A.; Pruijm, M.; Ackermann, D.; Vogt, B.; Guessous, I.; Burnier, M.; Pechere-Bertschi, A.; Bochud, M.; Ponte, B. Sodium
Intake Is Associated with Renal Resistive Index in an Adult Population-Based Study. Hypertension
2020
,76, 1898–1905. [CrossRef]
90.
Doi, Y.; Iwashima, Y.; Yoshihara, F.; Kamide, K.; Hayashi, S.I.; Kubota, Y.; Nakamura, S.; Horio, T.; Kawano, Y. Renal resistive
index and cardiovascular and renal outcomes in essential hypertension. Hypertension 2012,60, 770–777. [CrossRef] [PubMed]
91.
Hamano, K.; Nitta, A.; Ohtake, T.; Kobayashi, S. Associations of renal vascular resistance with albuminuria and other microan-
giopathy in type 2 diabetic patients. Diabetes Care 2008,31, 1853–1857. [CrossRef] [PubMed]
92.
Avolio, A.P.; Fa-Quan, D.; Wei-Qiang, L. Effects of aging on arterial distensibility in populations with high and low prevalence of
hypertension: Comparison between urban and rural communities in China. Circulation 1985,71, 202–210. [CrossRef] [PubMed]
93.
Avolio, A.P.; Clyde, K.M.; Beard, T.C.; Cooke, H.M.; Ho, K.K.; O’Rourke, M.F. Improved arterial distensibility in normotensive
subjects on a low salt diet. Arteriosclerosis 1986,6, 166–169. [CrossRef]
94.
Widlansky, M.E.; Gokce, N.; Keaney, J.F.; Vita, J.A. The clinical implications of endothelial dysfunction. J. Am. Coll. Cardiol.
2003
,
42, 1149–1160. [CrossRef]
95.
Ying, W.Z.; Sanders, P.W. Increased dietary salt activates rat aortic endothelium. Hypertension
2002
,39, 239–244. [CrossRef]
[PubMed]
96.
Li, J.; White, J.; Guo, L.; Zhao, X.; Wang, J.; Smart, E.J.; Li, X.A. Salt inactivates endothelial nitric oxide synthase in endothelial
cells. J. Nutr. 2009,139, 447–451. [CrossRef]
97.
Ma, S.; Wang, Q.; Zhang, Y.; Yang, D.; Li, D.; Tang, B.; Yang, Y. Transgenic overexpression of uncoupling protein 2 attenuates
salt-induced vascular dysfunction by inhibition of oxidative stress. Am. J. Hypertens. 2014,27, 345–354. [CrossRef] [PubMed]
98.
Zhu, J.; Mori, T.; Huang, T.; Lombard, J.H. Effect of high-salt diet on NO release and superoxide production in rat aorta. Am. J.
Physiol. Heart Circ. Physiol. 2004,286, H575–H583. [CrossRef] [PubMed]
99.
Kagota, S.; Tamashiro, A.; Yamaguchi, Y.; Sugiura, R.; Kuno, T.; Nakamura, K.; Kunitomo, M. Downregulation of vascular soluble
guanylate cyclase induced by high salt intake in spontaneously hypertensive rats. Br. J. Pharmacol.
2001
,134, 737–744. [CrossRef]
100.
Kagota, S.; Tamashiro, A.; Yamaguchi, Y.; Nakamura, K.; Kunitomo, M. High salt intake impairs vascular nitric oxide/cyclic
guanosine monophosphate system in spontaneously hypertensive rats. J. Pharmacol. Exp. Ther. 2002,302, 344–351. [CrossRef]
101.
Wang, J.; Roman, R.J.; Falck, J.R.; De La Cruz, L.; Lombard, J.H. Effects of high-salt diet on CYP450-4A
ω
-hydroxylase expression
and active tone in mesenteric resistance arteries. Am. J. Physiol. Heart Circ. Physiol. 2005,288, H1557–H1565. [CrossRef]
Nutrients 2021,13, 3177 15 of 15
102.
Pimenta, E.; Gordon, R.D.; Ahmed, A.H.; Cowley, D.; Leano, R.; Marwick, T.H.; Stowasser, M. Cardiac dimensions are largely
determined by dietary salt in patients with primary aldosteronism: Results of a case-control study. J. Clin. Endocrinol. Metab.
2011,96, 2813–2820. [CrossRef]
103.
Catena, C.; Colussi, G.L.; Novello, M.; Verheyen, N.D.; Bertin, N.; Pilz, S.; Tomaschitz, A.; Sechi, L.A. Dietary Salt Intake Is a
Determinant of Cardiac Changes After Treatment of Primary Aldosteronism: A Prospective Study. Hypertension
2016
,68, 204–212.
[CrossRef]
104.
Catena, C.; Verheyen, N.D.; Url-Michitsch, M.; Kraigher-Krainer, E.; Colussi, G.; Pilz, S.; Tomaschitz, A.; Pieske, B.; Sechi, L.A.
Association of Post-Saline Load Plasma Aldosterone Levels with Left Ventricular Hypertrophy in Primary Hypertension. Am. J.
Hypertens. 2016,29, 303–310. [CrossRef]
105.
Du Cailar, G.; Fesler, P.; Ribstein, J.; Mimran, A. Dietary sodium, aldosterone, and left ventricular mass changes during long-term
inhibition of the renin-angiotensin system. Hypertension 2010,56, 865–870. [CrossRef] [PubMed]
106.
Fan, C.Y.; Kawai, Y.; Inaba, S.; Arakawa, K.; Katsuyama, M.; Kajinami, K.; Yasuda, T.; Yabe-Nishimura, C.; Konoshita, T.;
Miyamori, I. Synergy of aldosterone and high salt induces vascular smooth muscle hypertrophy through up-regulation of NOX1.
J. Steroid Biochem. Mol. Biol. 2008,111, 29–36. [CrossRef]
107.
Wu, C.; Yosef, N.; Thalhamer, T.; Zhu, C.; Xiao, S.; Kishi, Y.; Regev, A.; Kuchroo, V.K. Induction of pathogenic TH 17 cells by
inducible salt-sensing kinase SGK1. Nature 2013,496, 513–517. [CrossRef] [PubMed]
108.
Mattson, D.L. Infiltrating immune cells in the kidney in salt-sensitive hypertension and renal injury. Am. J. Physiol. Ren. Physiol.
2014,307, F499–F508. [CrossRef] [PubMed]
109.
Lu, X.; Crowley, S.D. Inflammation in Salt-Sensitive Hypertension and Renal Damage. Curr. Hypertens. Rep.
2018
,20, 103.
[CrossRef] [PubMed]
110.
Yi, B.; Titze, J.; Rykova, M.; Feuerecker, M.; Vassilieva, G.; Nichiporuk, I.; Schelling, G.; Morukov, B.; Choukèr, A. Effects of dietary
salt levels on monocytic cells and immune responses in healthy human subjects: A longitudinal study. Transl. Res.
2015
,166,
103–110. [CrossRef] [PubMed]
111.
Miranda, P.M.; De Palma, G.; Serkis, V.; Lu, J.; Louis-Auguste, M.P.; McCarville, J.L.; Verdu, E.F.; Collins, S.M.; Bercik, P. High salt
diet exacerbates colitis in mice by decreasing Lactobacillus levels and butyrate production. Microbiome 2018,6, 57. [CrossRef]
112.
Wang, C.; Huang, Z.; Yu, K.; Ding, R.; Ye, K.; Dai, C.; Xu, X.; Zhou, G.; Li, C. High-salt diet has a certain impact on protein
digestion and gut microbiota: A sequencing and proteome combined study. Front. Microbiol. 2017,8. [CrossRef] [PubMed]
113.
Hu, J.; Luo, H.; Wang, J.; Tang, W.; Lu, J.; Wu, S.; Xiong, Z.; Yang, G.; Chen, Z.; Lan, T.; et al. Enteric dysbiosis-linked gut barrier
disruption triggers early renal injury induced by chronic high salt feeding in mice. Exp. Mol. Med. 2017,49. [CrossRef]
114.
Faraco, G.; Brea, D.; Garcia-Bonilla, L.; Wang, G.; Racchumi, G.; Chang, H.; Buendia, I.; Santisteban, M.M.; Segarra, S.G.; Koizumi,
K.; et al. Dietary salt promotes neurovascular and cognitive dysfunction through a gut-initiated TH17 response. Nat. Neurosci.
2018,21, 240–249. [CrossRef] [PubMed]
115.
Wilck, N.; Matus, M.G.; Kearney, S.M.; Olesen, S.W.; Forslund, K.; Bartolomaeus, H.; Haase, S.; Mahler, A.; Balogh, A.; Marko, L.;
et al. Salt-responsive gut commensal modulates TH17 axis and disease. Nature 2017,551, 585–589. [CrossRef]
116.
Mell, B.; Jala, V.R.; Mathew, A.V.; Byun, J.; Waghulde, H.; Zhang, Y.; Haribabu, B.; Vijay-Kumar, M.; Pennathur, S.; Joe, B. Evidence
for a link between gut microbiota and hypertension in the Dahl rat. Physiol. Genom. 2015,47, 187–197. [CrossRef] [PubMed]
... HS diets are associated with several cardiovascular and inflammatory diseases resulting in ischemic injury and end-organ stress [35][36][37]. An average Western diet is considered to have a 2-3 times higher salt content than the American Heart Association daily recommendation [38]. ...
Article
Full-text available
Several chronic inflammatory diseases have been linked to high-salt (HS) diets. Chronic inflammation is an established causative hallmark of cancer. However, a direct role of HS diets in tumorigenesis is yet to be defined. Previous orthotopic murine breast tumor studies have shown that short-term HS diets caused inhibition of tumor growth through the activation of cytotoxic adaptive immune responses. However, there have been experimental challenges in developing a viable chronic HS-diet-based murine tumor model. To address this, we have developed a novel chronic HS diet tumor model through the sequential passaging of tumor cells in mice under HS dietary conditions. Two orthotopic murine triple-negative breast cancer models, 4T1 tumor cells injected into BALB/c mice and Py230 tumor cells injected into C57Bl/6 mice, were utilized in our study. For the HS diet cohort, prior to orthotopic injection with tumor cells, the mice were kept on a 4% NaCl diet for 2 weeks. For the regular salt (RS) diet cohort, the mice were kept on a 1% NaCl diet. Following syngeneic cancer cell injection, tumors were allowed to grow for 28 days, following which they were collected to isolate immune cell-depleted cancer cells (passage 1, P1). The tumor cells from P1 were reinjected into the next set of non-tumor-bearing mice. This procedure was repeated for three cycles (P2–P4). In P1, compared to the RS diet cohort, we observed reduced tumor kinetics in both murine tumor models on the HS diet. In contrast, by P4, there was significantly higher tumor progression in the HS diet cohort over the RS diet cohort. Flow cytometry analysis demonstrated an 8-fold increase in tumor-initiating stem cells (TISCs) from P1 to P4 of the HS diet cohort, while there were no significant change in TISC frequency with sequential passaging in the RS diet cohort. Molecular studies showed enhanced expression of TGFβR2 and CD80 on TISCs isolated from the P4 HS diet cohort. In vitro studies demonstrated that TGFβ stimulation of these TISCs increased the cellular expression of CD80 molecules. Further, the chronic HS diet selectively induced the glycolytic metabolic phenotype over the mitochondrial oxidative phosphorylation phenotype in TISCs, which is needed for the production of metabolites during tumor cell differentiation and proliferation. The infiltrating CD8 and CD4 T-lymphocytes in P4 tumors demonstrated increased expression of the immune checkpoint inhibitor (ICI) CTLA4, a known binding partner of CD80, to cause immune exhaustion and pro-tumorigenic effects. Interestingly, anti-TGFβ monoclonal antibodies (mAbs) played a synergistic role in further enhancing the anti-tumor effect of anti-CTLA4 mAb. In summary, our findings demonstrated that chronic HS diet increased the frequency of TISCs in tumors leading to blunting of cytotoxic adaptive immune responses causing tumor proliferation. Furthermore, a combination of anti-TGFβ with current ICI-based immunotherapies could exert more favorable anti-cancer clinical outcomes.
... The high salt meal is hypothesized to increase BP by adversely increasing oxidative stress (Dickinson et al., 2011), alterations in the renin-angiotensin system (RAS) (Drenjancevic-Peric et al., 2011), and inactivation of endothelial nitric oxide synthase (eNOS) activity. Consequently, the decreased eNOS activity lowers nitric oxide (NO) production, resulting in endothelial dysfunction (Dickinson et al., 2011;Grillo et al., 2019;Jaques et al., 2021). Longitudinal studies have also provided evidence that salt reduction is linearly associated with BP reduction (Filippini et al., 2021). ...
Article
Full-text available
Cocoa powder is an important dietary source of flavanols that modulate elevated blood pressure (BP). This study aimed to investigate the acute effects of cocoa beverage co-consumption with a high-fat-high-salt meal (HFHSM) on postprandial systolic blood pressure (SBP), diastolic blood pressure (DBP), and heart rate in older adults living with uncontrolled hypertension. The study was a randomized crossover trial and involved older adults (aged 52.50 ± 9.36 years) (n = 28) living with hypertension. Participants, following an overnight fast, consumed either an HFHSM and cocoa beverage comprising 15 g cocoa powder in 250 mL water (HFHSM + CB; intervention) or HFHSM and 250 mL water (HFHSM + W; control). Resting SBP, DBP, and the heart rate of participants were measured at baseline (−5 min and 0 min) and 15, 30, 45, 60, 90, and 120 min following the consumption of either HFHSM + CB or HFHSM + W using an automated BP monitor. Repeated measures linear mixed model was used to compare the effect of cocoa beverage and water on postprandial outcomes over a 2-h period. A significant reduction (−3.8 ± 0.6 mmHg, p < .05) in resting SBP was observed postprandially following the intake of the HFHSM + CB over the 120-min period compared to the HFHSM + W. The effect was more prominent during the 90-and 120-min time points of the trial duration. No significant change in the DBP and heart rate following the consumption of HFHSM + CB compared to the HFHSM + W was observed. Cocoa beverage co-consumption with HFHSM improved postprandial SBP in older adults living with hypertension. K E Y W O R D S cocoa beverage, diastolic blood pressure, heart rate, hypertension, systolic blood pressure This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.
... Berbagai mekanisme bertanggung jawab atas hubungan antara asupan natrium dan tekanan darah. Peran penting ginjal dalam hubungan ini telah ditunjukkan dengan jelas dalam beberapa percobaan transplantasi (12).Guyton mengembangkan model regulasi tekanan darah yang kompleks, di mana ginjal adalah pengatur utama yang menjaga keseimbangan antara asupan natrium, volume ekstraseluler, dan tekanan darah. Dia memperkenalkan konsep penting tekanan natriuresis sebagai mekanismenya melalui ginjal memiliki kemampuan untuk mempertahankan tekanan darah normal melalui fungsinya mengatur homeostatis volume dan reabsorpsi natrium (13). ...
Article
Full-text available
Hipertensi merupakan masalah kesehatan masyarakat yang utama di Indonesia karena prevalensinya yang tinggi dan merupakan faktor risiko penyakit jantung, gagal ginjal, dan stroke.Hipertensi diderita oleh satu miliar orang di seluruh dunia dan diperkirakan pada tahun 2025 akan meningkat menjadi 1,5 miliar orang. Berdasarkan data Riskesdas tahun 2018, prevalensi hipertensi mengalami peningkatan dari 25,8% menjadi 34,1%. Salah satu faktor resiko hipertensi yang dapat dimodifikasi adalah diet tinggi garam. Konsumsi natrium yang berlebihan merupakan penyebab utama tingginya penyakit hipertensi. Oleh karena itu, mengurangi jumlah asupan natrium akan membantu menurunkan tingkat tekanan darah. Tujuan dari telaah singkat ini untuk menyajikan semua bukti mengenai hubungan diet rendah garam terhadap penurunan tekanan darah pada penderita hipertensi. Pencarian literatur dilakukan pada database PubMed, Science Direct, Google Scholar dan Springer Link pada topik diet rendah garam terhadap hipertensi. Hasil telaah menujukkan penelitian tentang hubungan diet rendah garam pada hipertensi telah dilaporkan. Berdasarkan bukti yang ada, diet tinggi sodium atau garam dapat meningkatkan tekanan darah. Kesimpulannya bahwa diet rendah garam dapat berpengaruh terhadap tekanan darah pasien hipertensi.
... The role of excessive salt intake in hypertension and the health benefits of salt reduction are very well documented [1][2][3]. Although sodium is essential for almost all physiological functions, from nutrient absorption to nervous impulse transmission and muscle contraction [4][5][6], in excess it adversely impacts the metabolism [7], immunity [8], fibrosis [9], and cardiopulmonary work [10][11][12] among many other effects. In a rat model, salt-elevated food with NaCl concentration exceeding 4% (like in the human-used processed meats and soups) was shown to exacerbate the development of various types of cardiomyopathy [13] leading to heart failure. ...
Article
Full-text available
Low-salt diet (LSD) is a constant recommendation to hypertensive patients, but the genomic mechanisms through which it improves cardiac pathophysiology are still not fully understood. Our publicly accessible transcriptomic dataset of the left ventricle myocardium of adult male mice subjected to prolonged LSD or normal diet was analyzed from the perspective of the Genomic Fabric Paradigm. We found that LSD shifted the metabolic priorities by increasing the transcription control for fatty acids biosynthesis while decreasing it for steroid hormone biosynthesis. Moreover, LSD remodeled pathways responsible for cardiac muscle contraction (CMC), chronic Chagas (CHA), diabetic (DIA), dilated (DIL), and hypertrophic (HCM) cardiomyopathies, and their interplays with the glycolysis/glucogenesis (GLY), oxidative phosphorylation (OXP), and adrenergic signaling in cardiomyocytes (ASC). For instance, the statistically (p < 0.05) significant coupling between GLY and ASC was reduced by LSD from 13.82% to 2.91% (i.e., −4.75×), and that of ASC with HCM from 10.50% to 2.83% (−3.71×). The substantial up-regulation of the CMC, ASC, and OXP genes, and the significant weakening of the synchronization of the expression of the HCM, CHA, DIA, and DIL genes within their respective fabrics justify the benefits of the LSD recommendation.
... It is cholesterol-free and contains 2.7 g/100 g of protein, 62.3 g/100 g of carbohydrates and 177 mg/100 g of sodium (Na), which is approximately half the sodium content found in other creamer products on the market (Table 8). Excessive sodium intake is believed to have adverse effects on our health, including impacts on blood pressure and cardiovascular health (Afshin et al., 2019;Jaques et al., 2021). ...
Article
Full-text available
A study was conducted to develop non‐dairy creamer production with a modified fatty acid profile having high levels of omega‐3 and 6, by using raw materials collected from the Republic Foods industrial non‐dairy creamer plant. The non‐dairy creamer formulation aimed to maintain a balanced 1:1:1 ratio of polyunsaturated, monounsaturated and saturated fatty acids. The ingredients used included palm olein, stearin, RBD palm oil, rice bran oil, soybean oil, coconut oil and palm kernel oil. The mixture proportions tested were determined using Design Expert software, for in total 20 different formulations. From these, six formulations were selected based on their fatty acid profiles, which closely approximated the desired 1:1:1 ratio of polyunsaturated, monounsaturated and saturated fatty acids. To complement the physicochemical results, a quality control and research officer from Republic Foods industrial conducted a comprehensive evaluation of various properties of the non‐dairy cream samples intended for use in coffee. It is noteworthy that formulation number 20 received the highest scores across all tested properties. This formulation had 50.53% palm olein, 1.00% stearin, 1.00% RBD palm oil, 1.00% rice bran oil, 44.47% soybean oil, 1.00% coconut oil and 1.00% palm kernel oil. This combination results in a fatty acid ratio of 1:1:0.8 (PUFA:MUFA:SFA) at the least production cost. The developed non‐dairy creamer exhibited the highest total energy content, namely 519 kilocalories/100 g, when compared with commercial brands. However, after a storage period of 90 days, the peroxide value reached 6.18 ± 0.22 mEq kg⁻¹ of fat, which is below the FDA standard limit of 10.0 mEq kg⁻¹ fat. This trend in peroxide value increase over the 90‐day storage period aligns with the behaviour of TBA, which also showed an upward trend during storage.
... Salt intake is also related to BP-independent effects. The most well-known are endothelial dysfunction and remodelling [13,14], atherosclerosis [6], inflammation [6], water retention, increased left ventricular mass, increased risk of stroke and stomach cancer, deterioration of renal function, renal stones and osteoporosis, asthma and obesity [5]. Recently, alterations of gut microbiota composition and diversity following elevated salt intake have been reported [15,16], with reduction of probiotic Lactobacillusspp [17].. ...
Article
Full-text available
High salt intake and compliance to low-sodium (LS) diets are critical in hypertension. Salt reduction in processed foods can help to achieve the target sodium intake. To verify the hypothesis that an innovative LS formulation of a traditional bread could result in a reduction of sodium intake and blood pressure, we performed a 6-month randomized controlled pilot trial on hypertensive patients. We additionally explored the effects of sodium restriction on blood pressure and fecal cultivable bacteria. Fifty-seven patients were randomized in three groups. Group A (n = 19) followed a free diet using standard bread (750 mg Na/100 g), group B (n = 18) followed a LS diet (2300 mg Na/die) using standard bread, group C (n = 20) followed a LS diet (2300 mg Na/die) using LS bread (280 mg Na/100 g). We measured 24-h urinary sodium, blood pressure, routine parameters, fecal microbial counts (26 patients). After 6 months, as compared to group A, group C showed a reduction of 24-h urinary sodium excretion (-908 mg/24 h), diastolic pressure (-9 mmHg) and microbial counts of Bacteroides, Porphyromonas, Prevotella, Enterobacteriaceae, Staphylococcus, Micrococcus. These results suggest that LS bread could increase the adherence to a LS diet, reducing sodium excretion, diastolic pressure and abundance of some fecal cultivable bacteria. Trial registration Registration nr. NCT03127553, on 25/04/2017.
... (www.preprints.org) | NOT PEER-REVIEWED | Posted: 29 January 2024 doi:10.20944/preprints202401.1981.v111 ...
Preprint
Full-text available
Keywords: Adrenergic signaling in cardiomyocytes; cardiac muscle contraction; Chagas disease; diabetic cardiomyopathy; dilated cardiomyopathy; glycerolipid metabolism; glycolisis/glucogenesis; hypertrophic cardiomyopathy; purine metabolism; steroid hormone biosynthesis.
Article
Full-text available
Background Across the globe, a vast number of people, amounting to 1.28 billion adults aged 30–79 years, suffer from hypertension. Two-thirds of them reside in low- and middle-income countries, and a significant 46% of these adults with hypertension are unaware of their condition. Hence the study aims to determine the Antihypertensive use insights and experiences among hypertensive patients at Korle-Bu Teaching Hospital. Methods A qualitative exploratory design was employed to recruit thirty 30 hypertensive patients from both male and female medical wards as well as the hypertensive clinic at the OPD of Korle Bu Teaching Hospital. The patients were selected using a purposive sampling technique, following which they were involved in face-face in-depth interviews which were audiotaped. Recorded data was then transcribed and analyzed with content analysis. Findings The analysis of the data resulted in three (3) main themes and 10 sub-themes. The study uncovered a general lack of knowledge about antihypertensive medications. This limited understanding resulted in a negative attitude among most patients toward the use of prescribed antihypertensive drugs. When it came to their experiences with antihypertensive medications, patients shared a wide range of experiences. Conclusion It was concluded that there is a need to improve the knowledge and attitude of patients as these play pivotal roles in determining adherence levels. Thus, interventions such as the organization of educational programs and awareness creation is recommended to improve adherence level and in turn, decrease the prevalence of hypertensive complications associated with poor management.
Article
Excessive salt intake is accompanied by the development of cardiovascular disorders, not always associated with an increase in blood pressure (BP). Given the cardioprotective effect of soy proteins in chronic kidney disease, the question arose of the possibility of leveling dietary interventions, in particular soy proteins, the damaging effect of a high-salt diet on the cardiovascular system. The aim of this work is to study the effect of long-term use of a diet high in NaCl and soy protein on myocardial remodeling and skin histomorphology in monkeys. The study was performed on male Javan macaques (Macaca fascicularis). The control group received a standard diet (2 g NaCl/kg feed). The second was a high-salt diet (8 g NaCl/kg feed), the third was a high-salt diet and SUPRO760 soy protein (200 g/kg feed). Echocardiography, registration of BP and heart rate were performed at baseline, after 4 and 12 months. After 12 months, a histological examination of the musculocutaneous flap was performed. In all animals, BP and heart rate did not change significantly during 12 months. In macaques on a high-salt diet, by the end of the study, deterioration in systolic and diastolic functions of the left ventricle (LV) was noted. In animals receiving additional soy protein, these changes leveled out. After 12 months, macaques fed soy protein had LV myocardial mass smaller and higher LV contractility than animals fed excess salt without soy protein. In monkeys on a high-salt diet, accumulation of collagen fibers in the hypodermis, hyalinization of the cytoplasm of capillary smooth muscle cells, perivascular and perineural edema of the reticular dermis were revealed. In animals treated with soy protein, skin capillary remodeling was less pronounced. Thus, high salt intake leads to adverse structural and functional disorders of the heart and blood vessels in cynomolgus monkeys, not associated with an increase in blood pressure. The inclusion of soy isolate in the diet reduces the negative effects of a high-salt diet on the cardiovascular system.
Article
Full-text available
Background The impact of sodium intake on cardiovascular disease (CVD) health and mortality has been studied for decades, including the well-established association with blood pressure. However, non-linear patterns, dose–response associations, and sex differences in the relationship between sodium and potassium intakes and overall and cause-specific mortality remain to be elucidated and a comprehensive examination is lacking. Our study objective was to determine whether intake of sodium and potassium and the sodium–potassium ratio are associated with overall and cause-specific mortality in men and women. Methods We conducted a prospective analysis of 237,036 men and 179,068 women in the National Institutes of Health-AARP Diet and Health Study. Multivariable-adjusted Cox proportional hazard regression models were utilized to calculate hazard ratios. A systematic review and meta-analysis of cohort studies was also conducted. Results During 6,009,748 person-years of follow-up, there were 77,614 deaths, 49,297 among men and 28,317 among women. Adjusting for other risk factors, we found a significant positive association between higher sodium intake (≥ 2,000 mg/d) and increased overall and CVD mortality (overall mortality, fifth versus lowest quintile, men and women HRs = 1.06 and 1.10, Pnonlinearity < 0.0001; CVD mortality, fifth versus lowest quintile, HRs = 1.07 and 1.21, Pnonlinearity = 0.0002 and 0.01). Higher potassium intake and a lower sodium–potassium ratio were associated with a reduced mortality, with women showing stronger associations (overall mortality, fifth versus lowest quintile, HRs for potassium = 0.96 and 0.82, and HRs for the sodium–potassium ratio = 1.09 and 1.23, for men and women, respectively; Pnonlinearity < 0.05 and both P for interaction ≤ 0.0006). The overall mortality associations with intake of sodium, potassium and the sodium–potassium ratio were generally similar across population risk factor subgroups with the exception that the inverse potassium-mortality association was stronger in men with lower body mass index or fruit consumption (Pinteraction < 0.0004). The updated meta-analysis of cohort studies based on 42 risk estimates, 2,085,904 participants, and 80,085 CVD events yielded very similar results (highest versus lowest sodium categories, pooled relative risk for CVD events = 1.13, 95% CI: 1.06–1.20; Pnonlinearity < 0.001). Conclusions Our study demonstrates significant positive associations between daily sodium intake (within the range of sodium intake between 2,000 and 7,500 mg/d), the sodium–potassium ratio, and risk of CVD and overall mortality, with women having stronger sodium–potassium ratio-mortality associations than men, and with the meta-analysis providing compelling support for the CVD associations. These data may suggest decreasing sodium intake and increasing potassium intake as means to improve health and longevity, and our data pointing to a sex difference in the potassium-mortality and sodium–potassium ratio-mortality relationships provide additional evidence relevant to current dietary guidelines for the general adult population. Systematic review registration PROSPERO Identifier: CRD42022331618.
Article
Full-text available
Background: Multiple modifiable risk factors for late complications in patients with diabetic kidney disease (DKD), including hyperglycemia, hypertension and dyslipidemia, increase the risk of a poor outcome. DKD is associated with a very high cardiovascular risk, which requires simultaneous treatment of these risk factors by implementing an intensified multifactorial treatment approach. However, the efficacy of a multifactorial intervention on major fatal/non-fatal cardiovascular events (MACEs) in DKD patients has been poorly investigated. Methods: Nephropathy in Diabetes type 2 (NID-2) study is a multicentre, cluster-randomized, open-label clinical trial enrolling 395 DKD patients with albuminuria, diabetic retinopathy (DR) and negative history of CV events in 14 Italian diabetology clinics. Centres were randomly assigned to either Standard-of-Care (SoC) (n = 188) or multifactorial intensive therapy (MT, n = 207) of main cardiovascular risk factors (blood pressure < 130/80 mmHg, glycated haemoglobin < 7%, LDL, HDL and total cholesterol < 100 mg/dL, > 40/50 mg/dL for men/women and < 175 mg/dL, respectively). Primary endpoint was MACEs occurrence by end of follow-up phase. Secondary endpoints included single components of primary endpoint and all-cause death. Results: At the end of intervention period (median 3.84 and 3.40 years in MT and SoC group, respectively), targets achievement was significantly higher in MT. During 13.0 years (IQR 12.4-13.3) of follow-up, 262 MACEs were recorded (116 in MT vs. 146 in SoC). The adjusted Cox shared-frailty model demonstrated 53% lower risk of MACEs in MT arm (adjusted HR 0.47, 95%CI 0.30-0.74, P = 0.001). Similarly, all-cause death risk was 47% lower (adjusted HR 0.53, 95%CI 0.29-0.93, P = 0.027). Conclusion: MT induces a remarkable benefit on the risk of MACEs and mortality in high-risk DKD patients. Clinical Trial Registration ClinicalTrials.gov number, NCT00535925. https://clinicaltrials.gov/ct2/show/NCT00535925.
Article
Full-text available
We evaluated the relationship between estimated 24-hour urinary sodium excretion from the Kawasaki, Tanaka, and INTERSALT (International Study of Sodium, Potassium, and Blood Pressure) formulas and blood pressure (BP). We pooled 10 034 person-visit data from 3 cohort studies in Bangladesh that had measured 24-hour urine sodium (m-24hUNa), potassium, creatinine excretion, and BP. We used m-24hUNa, potassium, and creatinine where necessary, rather than spot urine values in the formulas. Bland-Altman plots were used to determine the bias associated with formula-estimated sodium relative to m-24hUNa. We compared the sodium excretion and BP relationships from m-24hUNa versus formula-estimated sodium excretions, using restricted cubic spline plots for adjusted multilevel linear models. All formulas overestimated 24-hour sodium at lower levels but underestimated 24-hour sodium at higher levels. There was a linear relationship between m-24hUNa excretion and systolic BP, while estimated sodium excretion from all 3 formulas had a J-shaped relationship with systolic BP. The relationships between urine sodium excretion and diastolic BP were more complex but were also altered by using formulas. All formulas had associations with BP when a sex-specific constant sodium concentration was inserted in place of measured sodium. Since we used the m-24hUNa, potassium, and creatinine concentrations in formulas, the J-shaped relationships are due to intrinsic problems in the formulas, not due to spot urine sampling. Formula-estimated 24-hour urine sodium excretion should not be used to examine the relationship between sodium excretion and BP since they alter the real associations.
Article
Full-text available
Renal resistive index (RRI) has been associated with adverse renal and cardiovascular outcomes. Although traditionally considered a marker of intrinsic renal damage, RRI could also re ect systemic vascular dysfunction. As sodium intake was linked to alterations in vascular properties, we wished to characterize the association of salt consumption with RRI in the general adult population. Participants were recruited in a population-based study in Switzerland. RRI was measured by ultrasound in 3 segmental arteries. Sodium intake (UNa; mmol/24 h) was estimated on 24-hour urine samples. Carotido-femoral pulse wave velocity was obtained by applanation tonometry. Mixed multivariate regression models were used with RRI or pulse wave velocity as independent variables and UNa as dependent variable, adjusting for possible confounders. We included 1002 patients in the analyses with 528 (52.7%) women and mean age of 47.2±17.4. Mean values of UNa and RRI were 141.8±61.1 mmol/24 h and 63.8±5.5%, respectively. In multivariate analysis, UNa was positively associated with RRI (P=0.002) but not with pulse wave velocity (P=0.344). Plasma renin activity and aldosterone did not modify the relationship between UNa and RRI (P=0.087 for interaction). UNa/urinary potassium ratio was positively associated with pulse wave velocity ≥12 m/s (P=0.033). Our results suggest that dietary salt consumption has a direct impact on renal hemodynamic in the adult general population. Alterations in vascular properties likely explain those ndings, but inadequate renal vaso-motor response is also possible. Sodium intake could thus potentially be linked to underlying structural systemic damages affecting this population.
Article
Full-text available
Salt sensitivity refers to the physiological trait present in mammals, including humans, by which the blood pressure (BP) of some members of the population exhibits changes parallel to changes in salt intake. It is commoner in elderly, females, Afro-Americans, patients with chronic kidney disease (CKD) and insulin resistance. Increased salt intake promotes an expansion of extracellular fluid volume and increases cardiac output. Salt-sensitive individuals present an abnormal kidney reaction to salt intake; the kidneys retain most of the salt due to an abnormal over-reactivity of sympathetic nervous system and a blunted suppression of renin-angiotensin axis. Moreover, instead of peripheral vascular resistance falling, salt-sensitive subjects present increased vascular resistance due mainly to impaired nitric oxide synthesis in endothelium. Recent studies have shown that part of the dietary salt loading accumulates in skin. Hypertensive and patients with CKD seem to have more sodium in skin comparing to healthy ones. However, we still have not fully explained the link between skin sodium, BP and salt sensitivity. Finally, although salt sensitivity plays a meaningful role in BP pathophysiology, it cannot be used by the physician in everyday patient's care, mainly due to lack of a simple and practical diagnostic test.
Article
Full-text available
The Global Burden of Disease (GBD) 2010 study estimated national salt intake for 187 countries based on data available up to 2010. The purpose of this review was to identify studies that have measured salt intake in a nationally representative population using the 24‐hour urine collection method since 2010, with a view to updating evidence on population salt intake globally. Studies published from January 2011 to September 2018 were searched for from MEDLINE, Scopus, and Embase databases using relevant terms. Studies that provided nationally representative estimates of salt intake among the healthy adult population based on the 24‐hour urine collection were included. Measured salt intake was extracted and compared with the GBD estimates. Of the 115 identified studies assessed for eligibility, 13 studies were included: Four studies were from Europe, and one each from the United States, Canada, Benin, India, Samoa, Fiji, Barbados, Australia, and New Zealand. Mean daily salt intake ranged from 6.75 g/d in Barbados to 10.66 g/d in Portugal. Measured mean population salt intake in Italy, England, Canada, and Barbados was lower, and in Fiji, Samoa, and Benin was higher, in recent surveys compared to the GBD 2010 estimates. Despite global targets to reduce population salt intake, only 13 countries have published nationally representative salt intake data since the GBD 2010 study. In all countries, salt intake levels remain higher than the World Health Organization's recommendation, highlighting the need for additional global efforts to lower salt intake and monitor salt reduction strategies.
Article
Full-text available
Background: Suboptimal diet is an important preventable risk factor for non-communicable diseases (NCDs); however, its impact on the burden of NCDs has not been systematically evaluated. This study aimed to evaluate the consumption of major foods and nutrients across 195 countries and to quantify the impact of their suboptimal intake on NCD mortality and morbidity. Methods: By use of a comparative risk assessment approach, we estimated the proportion of disease-specific burden attributable to each dietary risk factor (also referred to as population attributable fraction) among adults aged 25 years or older. The main inputs to this analysis included the intake of each dietary factor, the effect size of the dietary factor on disease endpoint, and the level of intake associated with the lowest risk of mortality. Then, by use of disease-specific population attributable fractions, mortality, and disability-adjusted life-years (DALYs), we calculated the number of deaths and DALYs attributable to diet for each disease outcome. Findings: In 2017, 11 million (95% uncertainty interval [UI] 10-12) deaths and 255 million (234-274) DALYs were attributable to dietary risk factors. High intake of sodium (3 million [1-5] deaths and 70 million [34-118] DALYs), low intake of whole grains (3 million [2-4] deaths and 82 million [59-109] DALYs), and low intake of fruits (2 million [1-4] deaths and 65 million [41-92] DALYs) were the leading dietary risk factors for deaths and DALYs globally and in many countries. Dietary data were from mixed sources and were not available for all countries, increasing the statistical uncertainty of our estimates. Interpretation: This study provides a comprehensive picture of the potential impact of suboptimal diet on NCD mortality and morbidity, highlighting the need for improving diet across nations. Our findings will inform implementation of evidence-based dietary interventions and provide a platform for evaluation of their impact on human health annually. Funding: Bill & Melinda Gates Foundation.
Article
Background: Recent cohort studies show that salt intake below 6 g is associated with increased mortality. These findings have not changed public recommendations to lower salt intake below 6 g, which are based on assumed blood pressure (BP) effects and no side-effects. Objectives: To assess the effects of sodium reduction on BP, and on potential side-effects (hormones and lipids) SEARCH METHODS: The Cochrane Hypertension Information Specialist searched the following databases for randomized controlled trials up to April 2018 and a top-up search in March 2020: the Cochrane Hypertension Specialised Register, the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE (from 1946), Embase (from 1974), the World Health Organization International Clinical Trials Registry Platform, and ClinicalTrials.gov. We also contacted authors of relevant papers regarding further published and unpublished work. The searches had no language restrictions. The top-up search articles are recorded under "awaiting assessment." Selection criteria: Studies randomizing persons to low-sodium and high-sodium diets were included if they evaluated at least one of the outcome parameters (BP, renin, aldosterone, noradrenalin, adrenalin, cholesterol, high-density lipoprotein, low-density lipoprotein and triglyceride,. Data collection and analysis: Two review authors independently collected data, which were analysed with Review Manager 5.3. Certainty of evidence was assessed using GRADE. Main results: Since the first review in 2003 the number of included references has increased from 96 to 195 (174 were in white participants). As a previous study found different BP outcomes in black and white study populations, we stratified the BP outcomes by race. The effect of sodium reduction (from 203 to 65 mmol/day) on BP in white participants was as follows: Normal blood pressure: SBP: mean difference (MD) -1.14 mmHg (95% confidence interval (CI): -1.65 to -0.63), 5982 participants, 95 trials; DBP: MD + 0.01 mmHg (95% CI: -0.37 to 0.39), 6276 participants, 96 trials. Hypertension: SBP: MD -5.71 mmHg (95% CI: -6.67 to -4.74), 3998 participants,88 trials; DBP: MD -2.87 mmHg (95% CI: -3.41 to -2.32), 4032 participants, 89 trials (all high-quality evidence). The largest bias contrast across studies was recorded for the detection bias element. A comparison of detection bias low-risk studies versus high/unclear risk studies showed no differences. The effect of sodium reduction (from 195 to 66 mmol/day) on BP in black participants was as follows: Normal blood pressure: SBP: mean difference (MD) -4.02 mmHg (95% CI:-7.37 to -0.68); DBP: MD -2.01 mmHg (95% CI:-4.37, 0.35), 253 participants, 7 trials. Hypertension: SBP: MD -6.64 mmHg (95% CI:-9.00, -4.27); DBP: MD -2.91 mmHg (95% CI:-4.52, -1.30), 398 participants, 8 trials (low-quality evidence). The effect of sodium reduction (from 217 to 103 mmol/day) on BP in Asian participants was as follows: Normal blood pressure: SBP: mean difference (MD) -1.50 mmHg (95% CI: -3.09, 0.10); DBP: MD -1.06 mmHg (95% CI:-2.53 to 0.41), 950 participants, 5 trials. Hypertension: SBP: MD -7.75 mmHg (95% CI:-11.44, -4.07); DBP: MD -2.68 mmHg (95% CI: -4.21 to -1.15), 254 participants, 8 trials (moderate-low-quality evidence). During sodium reduction renin increased 1.56 ng/mL/hour (95%CI:1.39, 1.73) in 2904 participants (82 trials); aldosterone increased 104 pg/mL (95%CI:88.4,119.7) in 2506 participants (66 trials); noradrenalin increased 62.3 pg/mL: (95%CI: 41.9, 82.8) in 878 participants (35 trials); adrenalin increased 7.55 pg/mL (95%CI: 0.85, 14.26) in 331 participants (15 trials); cholesterol increased 5.19 mg/dL (95%CI:2.1, 8.3) in 917 participants (27 trials); triglyceride increased 7.10 mg/dL (95%CI: 3.1,11.1) in 712 participants (20 trials); LDL tended to increase 2.46 mg/dl (95%CI: -1, 5.9) in 696 participants (18 trials); HDL was unchanged -0.3 mg/dl (95%CI: -1.66,1.05) in 738 participants (20 trials) (All high-quality evidence except the evidence for adrenalin). Authors' conclusions: In white participants, sodium reduction in accordance with the public recommendations resulted in mean arterial pressure (MAP) decrease of about 0.4 mmHg in participants with normal blood pressure and a MAP decrease of about 4 mmHg in participants with hypertension. Weak evidence indicated that these effects may be a little greater in black and Asian participants. The effects of sodium reduction on potential side effects (hormones and lipids) were more consistent than the effect on BP, especially in people with normal BP.
Article
Several blood pressure guidelines recommend low sodium intake (<2.3 g/day, 100 mmol, 5.8 g/day of salt) for the entire population, on the premise that reductions in sodium intake, irrespective of the levels, will lower blood pressure, and, in turn, reduce cardiovascular disease occurrence. These guidelines have been developed without effective interventions to achieve sustained low sodium intake in free-living individuals, without a feasible method to estimate sodium intake reliably in individuals, and without high-quality evidence that low sodium intake reduces cardiovascular events (compared with moderate intake). In this review, we examine whether the recommendation for low sodium intake, reached by current guideline panels, is supported by robust evidence. Our review provides a counterpoint to the current recommendation for low sodium intake and suggests that a specific low sodium intake target (e.g. <2.3 g/day) for individuals may be unfeasible, of uncertain effect on other dietary factors and of unproven effectiveness in reducing cardiovascular disease. We contend that current evidence, despite methodological limitations, suggests that most of the world's population consume a moderate range of dietary sodium (2.3-4.6g/day; 1-2 teaspoons of salt) that is not associated with increased cardiovascular risk, and that the risk of cardiovascular disease increases when sodium intakes exceed 5 g/day. While current evidence has limitations, and there are differences of opinion in interpretation of existing evidence, it is reasonable, based upon observational studies, to suggest a population-level mean target of <5 g/day in populations with mean sodium intake of >5 g/day, while awaiting the results of large randomized controlled trials of sodium reduction on incidence of cardiovascular events and mortality.
Article
To study the effect of formulas on the estimation of dietary sodium intake (sodium intake) and its association with mortality, we analyzed the TOHP (Trials of Hypertension Prevention) follow-up data. Sodium intake was assessed by measured 24-hour urinary sodium excretion and estimations from sodium concentration using the Kawasaki, Tanaka, and INTERSALT (International Cooperative Study on Salt, Other Factors, and Blood Pressure) formulas. We used both the average of 3 to 7 urinary measurements during the trial period and the first measurement at the beginning of each trial. Additionally, we kept sodium concentration constant to test whether the formulas were independently associated with mortality. We included 2974 individuals aged 30 to 54 years with prehypertension, not assigned to sodium intervention. During a median 24-year follow-up, 272 deaths occurred. The average measured sodium intake was 3766±1290 mg/d. All estimated values, including those with constant sodium concentration, were systematically biased with overestimation at lower levels and underestimation at higher levels. There was a significant linear association between the average measured sodium intake (ie, gold standard method) and mortality. This relationship was altered by using the estimated sodium intakes. There appeared to be a J- or U-shaped relationship for the average estimated sodium by all formulas. Despite variations in the sodium-mortality relationship among various formulas, a common pattern was that all estimated values including those with constant sodium appeared to be inversely related to mortality at lower levels of sodium intake. These results demonstrate that inaccurate estimates of sodium cannot be used in association studies, particularly as the formulas per se seem to be related to mortality independent of sodium.
Article
Hypertension is a risk factor for cardiovascular disease. Increased urinary sodium excretion, representing dietary sodium intake, is associated with hypertension. Low sodium intake has been associated with increased mortality in observational studies. Further studies should assess whether confounding relationships explain associations between sodium intake and outcomes. We studied UK Biobank participants (n=457 484; mean age, 56.3 years; 44.7% men) with urinary electrolytes and blood pressure data. Estimated daily urinary sodium excretion was calculated using Kawasaki formulae. We analyzed associations between sodium excretion and blood pressure in subjects without cardiovascular disease, treated hypertension, or diabetes mellitus at baseline (n=322 624). We tested relationships between sodium excretion, incidence of fatal and nonfatal cardiovascular disease, heart failure, and mortality. Subjects in higher quintiles of sodium excretion were younger, with more men and higher body mass index. There was a linear relationship between increasing urinary sodium excretion and blood pressure. During median follow-up of 6.99 years, there were 11 932 deaths (1125 cardiovascular deaths) with 10 717 nonfatal cardiovascular events. There was no relationship between quintile of sodium excretion and outcomes. These relationships were unchanged after adjustment for comorbidity or excluding subjects with events during the first 2 years follow-up. No differing risk of incident heart failure (1174 events) existed across sodium excretion quintiles. Urinary sodium excretion correlates with elevated blood pressure in subjects at low cardiovascular risk. No pattern of increased cardiovascular disease, heart failure, or mortality risk was demonstrated with either high or low sodium intake.