You are on page 1of 17

Circulation Research

HYPERTENSION COMPENDIUM

Pathophysiology of Hypertension
The Mosaic Theory and Beyond
David G. Harrison , Thomas M. Coffman, Christopher S. Wilcox

ABSTRACT: Dr Irvine Page proposed the Mosaic Theory of Hypertension in the 1940s advocating that hypertension is the
result of many factors that interact to raise blood pressure and cause end-organ damage. Over the years, Dr Page modified
his paradigm, and new concepts regarding oxidative stress, inflammation, genetics, sodium homeostasis, and the microbiome
have arisen that allow further refinements of the Mosaic Theory. A constant feature of this approach to understanding
hypertension is that the various nodes are interdependent and that these almost certainly vary between experimental models
and between individuals with hypertension. This review discusses these new concepts and provides an introduction to other
reviews in this compendium of Circulation Research.

Key Words: blood pressure ◼ history ◼ hypertension ◼ inflammation ◼ reactive oxygen species

A PATHOPHYSIOLOGICAL/HISTORICAL
ccording to the 2017 guidelines of the American
Heart Association, almost one-half of the adult pop-
ulation in the United States has hypertension,1 and
CONCEPTS
the Global Burden of Disease study ranks elevated sys- The elusive nature of primary hypertension and the pre-
Downloaded from http://ahajournals.org by on April 23, 2024

tolic pressure as the leading cause of lost years of quality cise mechanisms underlying an elevation of blood pres-
life.2 Despite advances in medical therapy, hypertension sure (BP), even in cases of secondary hypertension and
remains a major risk factor for stroke, heart failure, renal in models of experimental hypertension, have remained
failure, atherosclerosis, and we now recognize dementia. a focus of debate and research for more than a cen-
In large part, this is because almost one-half of individu- tury. Beginning with seminal observations by Tigerstedt,4
als with hypertension are not aware that they have this Franz Volhard,5,6 and subsequently refined and elegantly
disease, and hypertension is not adequately treated in published by Goldblatt,7 it became apparent that the isch-
about one-half of those in whom the diagnosis is made.3 emic or underperfused kidney releases a substance or
Clinical hypertension can be grouped into 2 broad cat- substances that can raise the BP in recipient animals.
egories. Primary (or essential) hypertension represents These early findings set the groundwork for Irvine Page
between 85% and 95% of human cases and has an and Braun Menendez in 1939 to report their discovery
unidentified cause. In contrast, secondary hypertension is of a potent vasoconstrictor and pro-hypertensive agent
caused by identifiable underlying conditions, including renal isolated from renal extracts that they named hypertensin
artery stenosis, pheochromocytoma, adrenal adenoma, and angiotonin, respectively.8,9 They ultimately agreed to
or single-gene mutations. Historically, most patients were call it angiotensin.10
screened for secondary causes. However, it is now clear Irvine Page is among the most prominent pioneers of
that this is not cost-effective and that in the absence of hypertension research and is notable for his enduring
compelling clinical findings (abdominal bruit, hypokalemia, contributions. In his initial experimental observations with
clinical symptoms of a pheochromocytoma) or a pressing Oscar Helmer, he discovered that when preparations of
need to improve treatment (drug-resistant hypertension) renin were exposed to plasma containing what he termed
patients are generally treated without such an evaluation. “renin activator” a pressor substance was formed that

Correspondence to: David G. Harrison, MD, Betty and Jack Bailey Professor of Medicine, Pharmacology and Physiology, Director of Clinical Pharmacology, Room 536,
Robinson Research Bldg, 2220 Pierce Ave, 536 RRB, Vanderbilt University School of Medicine, Nashville, TN 37232-6602. Email david.g.harrison@vumc.org
For Sources of Funding and Disclosures, see page 858.
© 2021 American Heart Association, Inc.
Circulation Research is available at www.ahajournals.org/journal/res

Circulation Research. 2021;128:847–863. DOI: 10.1161/CIRCRESAHA.121.318082 April 2, 2021   847


Harrison et al Pathophysiology of Hypertension
Hypertension Compendium

discovery of this slow pressor response to angioten-


Nonstandard Abbreviations and Acronyms sin was, in itself, a major contribution to the field and
has given subsequent investigators a valuable model of
ACE angiotensin I–converting enzyme human hypertension. He reported further that these infu-
ADMA asymmetrical dimethylarginine sions of low dose Ang II caused the BP to become highly
Ang II angiotensin II labile, such that minimal environmental stimulation would
AP-1 activator protein 1 cause enormous increases in pressure, accompanied
AT1R angiotensin type 1 receptor by increases in heart rate. These increases in BP were
BP blood pressure
exacerbated by tyramine (that releases norepinephrine
from nerve terminals) and were blocked by guanethidine
EP3 E prostanoid receptor type 3
(that prevents norepinephrine release). Page concluded
GPCR G-protein coupled receptor
from these highly insightful studies that “angiotensin
IL interleukin causes hypertension by an indirect action mediated by
MCP1 monocyte chemoattractant protein 1 the central nervous system, an action independent of its
MEJ myoendothelial junction vasoconstrictor action.”13 He observed that renin was not
MR mineralocorticoid receptor elevated in the chronic phase of many cases of human
NHE3 sodium hydrogen exchanger 3 hypertension and surmised that human hypertension has
NOS NO synthase complex origins, and therefore, presumably was multifac-
Nox NADPH oxidase catalytic subunit torial. In his 1949 article, he chose to describe this mul-
PRR prorenin receptor tifactorial nature by the term “Mosaic,” pointing out that
PRS polygenic risk score “…many mechanisms are more or less involved. Elevated
RANTES regulated upon activation, normal T-cell
blood pressure is the resultant of multiple forces acting
expressed and presumably secreted on the variety of tissues which compose the circulatory
RAS renin-angiotensin system
apparatus.” In a subsequent review, Dr Page presented a
now-iconic octagonal diagram (Figure 1A) that has been
RGS regulator of G protein signaling
modified and often reproduced to illustrate his concept
RhoA rat sarcoma virus homologous protein A
that hypertension is caused by multiple factors, including
ROS reactive oxygen species neural and chemical perturbations, alterations of vascu-
SGK1 serum glucocorticoid kinase 1 lar caliber and elastance, cardiovascular reactivity, blood
Downloaded from http://ahajournals.org by on April 23, 2024

SGLT2 sodium-glucose–linked transporter type 2 volume, and viscosity.14 Dr Page subsequently revised
Sirt3 sirtuin3 the Mosaic Theory to include more general terms (Fig-
SOD superoxide dismutase ure 1B)15; however, the principle of his Mosaic Theory
Sox6 SRY-related HMG-box gene 6 has prevailed to the present day.16,17 It is fascinating to
TNF tumor necrosis factor ponder that the factors composing his Mosaic Theory
TonEBP tonicity-responsive enhancer-binding reflect an ongoing central discussion about the impor-
protein tance of the kidney (volume and cardiac output), the vas-
VEGF vascular endothelial growth factor culature (elastance, vascular caliber, and reactivity), and
the central nervous system (reactivity and neural). In the
past 2 decades, there have been numerous refinements
could raise BP up to 300 mm Hg. He named this sub- of our understanding of the nodes in Figure 1A and 1B
stance “angiotonin.”11 It would have been easy to assume and entirely new concepts. In this review, we will begin by
that this elevation of BP was entirely due to intense considering time-honored topics: the roles of the kidney,
vasoconstriction. However, his further observations sug- the vasculature, and the central nervous system and a
gested this was an inadequate explanation. In a classic prevailing effect of aldosterone in hypertension. We will
article in 1949, he summarized evidence that hyperten- then discuss new concepts that have emerged in the
sion could be mediated by the central nervous system, past 2 decades that Dr Page did not consider in his writ-
cardiovascular factors, endocrine factors, and perturba- ings but likely influence every node in the Mosaic dia-
tions of renal function.12 He provided evidence that the gram. Many of these topics are covered in greater depth
hypertension induced by acute injections of angiotonin by other articles in this compendium.
could be reversed by ganglionic blockade with tetraeth-
ylammonium. In a subsequent study, Dr Page and col-
leagues reported that low rates of Ang II (angiotensin
II) infusion had minimal or no effect on BP in the first
THE ROLE OF THE KIDNEY AND BODY
hours after starting the infusion, but increased BP by FLUID VOLUMES IN HYPERTENSION
30 mm Hg 24 hours later,13 suggesting the importance The kidney is an important node in Dr Page’s Mosaic
of mechanisms beyond immediate vasoconstriction. His Theory. Figure 1A refers to volume and cardiac output.

848   April 2, 2021 Circulation Research. 2021;128:847–863. DOI: 10.1161/CIRCRESAHA.121.318082


Harrison et al Pathophysiology of Hypertension

Hypertension Compendium
Figure 1. The original (A) and revised (B) Mosaic Theories proposed by Page.

Cardiac output is the product of heart rate and cardiac is acted on by the ACE to generate Ang II. Renin is the
stroke volume. Stroke volume is dependent on venous rate-limiting step for activation of the circulating renin-
return and is governed in large part by the kidneys and angiotensin system (RAS) and its synthesis and secre-
their modulation of volume homeostasis. Guyton et al18 tion by the kidney is tightly regulated.23 Renin is initially
built on these early concepts of Irvine Page to elabo- synthesized as the inactive precursor prorenin, which is
rate a complex mathematical model of the dynamic converted to active renin upon binding to the PRR (pro-
interactions between many of these Mosaic compo- renin receptor). Under normal conditions, renin is pro-
nents. The model predicted that the renal pressure duced almost exclusively by specialized juxtaglomerular
natriuresis mechanism regulated the body fluids and cells of the afferent arteriole. It is released in response to
thereby exerted a primary role in setting the level of BP, a reductions in perfusion pressure or delivery of sodium
Downloaded from http://ahajournals.org by on April 23, 2024

whereas the central nervous system and vascular func- chloride to macula densa cells, or by an increase in
tion, among others, provided important modulation. The sympathetic stimulation.24 Prolonged stimulation of the
critical role of the kidney has been illustrated repeatedly renal sympathetic nerves transforms a subset of vascu-
by studies in which the kidneys of hypertensive animals lar smooth muscle cells into renin-producing cells in the
raise BP, or permit hypertension, when transplanted afferent arteriole. This transformation is governed by the
into normal animals, dating to seminal work by Fasciolo transcription factor SRY-related HMG-box gene (Sox)-
et al.19 Dahl et al20 performed cross transplantation of 6.25 Principal cells of the collecting duct also can produce
kidneys between salt-sensitive and salt-resistant rats renin. This is mediated in part by a loss of NO production
and showed that hypertension followed the kidney. in various pathophysiological conditions.26 Although renin
Moreover, the transplant of kidneys from normotensive is normally released almost exclusively from the juxta-
donors into patients with renal failure from hypertensive glomerular apparatus of the kidney, the proximal tubules
nephrosclerosis has led in some cases to a slow rever- can become an important site of renin production during
sal of their hypertension and some of the accompanying oxidative stress or in response to increased glomerular
organ damage.21 Crowley et al22 subsequently reported protein filtration.27 Drs Gomez and Sequeira-Lopez will
that transplantation of kidneys from mice lacking the discuss these and other features of renin biology in their
angiotensin type 1 receptor protected wild-type recipi- review of this compendium.
ent mice from Ang II–induced hypertension, whereas Mice lacking the PRR in the collecting duct have a
transplantation of wild-type kidneys into AT1R (angio- reduced hypertensive response to Ang II infusion, in part,
tensin type 1 receptor) knockout mice restored their due to reduced-sodium resorption in this segment.28
ability to develop hypertension upon Ang II infusion. Direct actions of renin per se in clinical settings remain
This refinement relates a critical component of hyper- controversial. Treatment with the direct renin antagonist
tension from Ang II to activation of its principal recep- Aliskiren paradoxically worsened outcomes in humans
tor within the kidneys and provides further mechanisms with heart failure,29 and a recent meta-analysis reported
extending the theories of Page and Guyton. no improvement in outcomes in hypertension and a lower
The kidney has at least 4 major roles in hypertension. certainty of BP reduction for Aliskiren than for ACE
One is the production of renin, an aspartic protease that (angiotensin I–converting enzyme) inhibition.30 However,
cleaves angiotensinogen to angiotensin I, which in turn these disappointing reports may reflect pharmacokinetic

Circulation Research. 2021;128:847–863. DOI: 10.1161/CIRCRESAHA.121.318082 April 2, 2021   849


Harrison et al Pathophysiology of Hypertension
Hypertension Compendium

defects of Aliskiren.31 Moreover, renin is a dual-purpose A third, recently recognized role of the kidney in
enzyme that both metabolizes angiotensinogen and acts hypertension is to modulate systemic sympathetic tone
as a complement C3 convertase.32,33 It has long been by generating reflex signals via renal afferent nerves.
known that circulating levels of C3 predict the subse- Approximately 90% of renal nerves are efferent nerves
quent development of hypertension,34 likely by promot- sending sympathetic signals to the kidney, thereby
ing innate and adaptive immune responses.35 Therefore, enhancing tubular sodium resorption, renin release, and
a role may yet emerge for improved direct renin inhibi- vasomotor tone depending on the intensity of nerve traf-
tors in the treatment of hypertension and for reducing fic.46 However, a smaller proportion of renal nerves are
complement C3 activation. afferents that generate signals from within the kidney
A second major role of the kidney in hypertension is and transmit them to the brain stem where they can initi-
to reset or alter the pressure diuresis and natriuresis. As ate reflexes that promote increases in efferent sympa-
originally described by Guyton et al,18 a rise in BP causes thetic nervous system tone and induce hypertension.47
brisk diuresis and natriuresis, thereby returning BP towards More than 20 years ago, Campese and Kogosov48
normal values. Guyton36 defined the relationship between reported that the hypertension in rats with 5/6 nephrec-
BP with sodium and water excretion as the pressure natri- tomy was reduced markedly after severing renal afferent
uresis curve and reasoned that there must be a rightward nerve traffic by bilateral spinal dorsal rhizotomy. This was
resetting of this curve for hypertension to be sustained. accompanied by a marked reduction in norepinephrine
Although the mechanisms for pressure natriuresis and its turnover in the posterior and lateral hypothalamus. In
resetting are multifactorial, substantial evidence implicates a subsequent study, this group reported that injection
increases in renal interstitial pressure and alterations of the of a small bolus of phenol into one pole of the kidney
sodium transporters, such as the NHE3 (sodium hydrogen also increased renal afferent nerve activity and norepi-
exchanger 3) and the sodium-phosphate cotransporter iso- nephrine production by the posterior hypothalamus led
form 2 in the proximal tubule.37 Acute hypertension retracts to hypertension. These events were abrogated by renal
these from the luminal cell membrane to the base of the denervation, suggesting that excitation of renal affer-
apical microvilli in the proximal tubule where they cannot ent nerves played a critical role. These findings are in
participate in sodium reabsorption. Likewise, an increase keeping with other observations that the hypertension
in BP can relocate the thiazide-sensitive sodium chloride associated with cyclosporine administration in renal
cotransporter in the distal tubule. However, during chronic allograft recipients is in part mediated by afferent nerve
hypertension, these transporters are translocated to the activation.49 More recently, it has been recognized that
Downloaded from http://ahajournals.org by on April 23, 2024

apical microvilli, where they enhance sodium reabsorption the renal afferent nerves have a special role in salt-
and likely sustain hypertension.38 The SGLT2 (sodium-glu- dependent hypertension. Banek et al50 reported that
cose–linked transporter type 2) in the proximal tubule is renal deafferentation reduced hypertension in rats with
also upregulated by Ang II acting on AT1 receptors during deoxycorticosterone acetate–salt hypertension. The
renovascular hypertension39 where it is functionally linked ability of renal afferent nerves to initiate an increase in
to NHE3 via the microtubule-associated protein 340 to global sympathetic outflow might explain the pleiotropic
enhance proximal Na+ reabsorption during reactive oxygen effects of renal denervation.51 In Ang II–induced hyper-
species (ROS) stimulation.41 Perfusion of an SGLT2 inhibi- tension, renal efferent, rather than afferent nerves, seem
tor into the proximal tubule inhibits Na+:H+ exchange.42 to have a predominant role.52,53
Because NHE3 transports more Na+ than SGLT2, this Although the precise sites in the kidney where
functional interaction between the 2 is likely to contribute afferent nerves are activated remain undefined, recent
to the surprisingly robust 27% inhibition of proximal Na+ studies have thrown light on some of the underlying
and fluid reabsorption by the SGLT2 inhibitor dapagliflozin mechanisms. A high-salt diet in the context of both
in diabetic rats.43 This may underlie the reported effects of acute54 or chronic47 causes the expected reduction
SGLT2 inhibitors to reduce the plasma volume in heart fail- in the circulating RAS but paradoxically activates the
ure and the BP in hypertension.44,45 Multiple other sodium intrarenal RAS via increased ROS and activates renal
transport mechanisms are similarly affected as has been afferent nerve traffic to reflexly increase BP.
expertly reviewed elsewhere.38 A fourth important role of the kidney in hyperten-
Shifts in transporter location and function can be sion is to serve as a site of immune activation. As dis-
mediated by Ang II, inflammatory cytokines, loss of NO, cussed below, Ang II–induced hypertension activates
and adrenergic stimulation. It is important to note that antigen-presenting dendritic cells in the kidney that
shifts in pressure natriuresis are often not reflected by migrate to secondary lymphoid organs to activate T
overt changes in renal function as measured by usual cells which in turn return back to the kidney.53 This pro-
clinical parameters, such as blood urea nitrogen, creati- cess is blocked by renal denervation, suggesting that
nine, or creatinine clearance. Moreover, renal Na+ reten- efferent renal nerves promote neoantigen formation in
tion in hypertension is not usually accompanied by overt the kidney in hypertension. Single-cell sequencing has
signs of fluid overload. shown that the kidney, as opposed to blood vessels

850   April 2, 2021 Circulation Research. 2021;128:847–863. DOI: 10.1161/CIRCRESAHA.121.318082


Harrison et al Pathophysiology of Hypertension

Hypertension Compendium
or the spleen, accumulates T cells with an oligoclonal critical for promoting hypertension.59 More recently, Patel
T-cell receptor population, supporting the role of the et al60 defined an important role of RGS1 in vascular
kidney in antigen formation.55 control in atherosclerotic ApoE-deficient mice where
the knockout of RGS1 evoked a marked increase in BP
and vasoconstriction to phenylephrine, thromboxane, and
Ang II. Bone marrow transplant studies demonstrated
THE ROLE OF THE VASCULATURE IN
that this phenotype was not mediated by hematopoietic
HYPERTENSION cells. Rather, these investigators showed that mRNA
Several of the nodes in Dr Page’s Mosaic diagram refer to expression of RGS1 is attenuated by chronic Ang II infu-
potential vascular mechanisms, including vascular caliber, sion and suggested that hypertension leads to a loss of
reactivity, elasticity, and, indirectly, cardiac output. Sys- this important modulatory pathway. In contrast, RGS5
temic vascular resistance is almost uniformly enhanced seems to have modest to no effects on BP by itself but
in adults with hypertension, and many common agents activates RhoA (rat sarcoma virus homologous protein
used for the treatment of hypertension are vasodilators. A) stress fiber formation and a contractile phenotype of
Historically, Coleman et al56 proposed that in response vascular smooth muscle.61 RhoA is a critical activator of
to an initial expansion of blood volume and elevation of Rho kinase that targets contractile proteins including
cardiac output, “systemic autoregulation” caused vaso- the myosin light chain in vascular smooth muscle to pro-
constriction and a further increase in BP. This leads to mote vasoconstriction.62 Endothelial Rho kinase phos-
additional pressure natriuresis and returned the blood phorylates NO synthase on threonine 495 to inhibit NO
volume to normal but at the expense of sustained hyper- production, which also increases vasoconstriction.63 The
tension. Somewhat in keeping with this scenario, Fagard Rho kinase inhibitor Fasudil is approved for use in Japan
and Staessen57 reported an increased cardiac output in and China for the treatment of cerebral vasospasm and
hypertensive individuals <25 years of age but a normal pulmonary hypertension and has BP-lowering effects
cardiac output in older subjects. In addition to systemic in genetic- and salt-induced forms of experimental
autoregulation, which is a physiological response, it is hypertension.64
now clear that there are long-term pathophysiological Hypertension is also associated with impaired vaso-
manifestations of hypertension that increase vascular dilatation. For many years, it has been recognized that
tone and resistance. In the following paragraphs, we will endothelium-dependent vasodilatation and NO signal-
review some new observations about how vascular func- ing are reduced in hypertension, as recently reviewed.65
Downloaded from http://ahajournals.org by on April 23, 2024

tion is altered and contributes to hypertension. We will There are fundamental differences in the mecha-
attempt to relate these to older concepts embodied in nisms underlying endothelium-dependent vasodilata-
Page’s Mosaic theory. tion between larger conduit arteries, branch arteries,
There are at least 4 vascular perturbations that occur and resistance arterioles. Larger vessels predominantly
in and contribute to hypertension. The first is an enhanced employ NO, whereas smaller arteries and arterioles are
milieu of vasoconstrictor hormones, including Ang II, cat- also modulated by endothelium-dependent hyperpolar-
echolamines, and vasopressin, coupled with alterations ization and vasodilator prostaglandins. In larger vessels,
in vascular function that promote vasoconstriction and defects in NO synthesis and bioavailability include loss
diminish vasodilatation. Studies of isolated vessels have of critical cofactors for NO synthase, including L-arginine
demonstrated that both conduit vessels and arterioles and tetrahydrobiopterin,66,67 inhibition of NOS (NO syn-
from hypertensive animals display enhanced vasocon- thase) activity by asymmetrical dimethylarginine (ADMA),
striction to a variety of agents. Of particular interest are and oxidative inactivation of NO.68 It should be stressed
genes that modulate GPCR (G protein–coupled recep- that alterations of NO production extend beyond the
tor) signaling, including the RGS (regulators of G protein vasculature and have implications for renal and central
signaling) proteins, that induce GTP hydrolysis to termi- nervous system dysfunction in hypertension. In smaller
nate G protein signaling. The RGS proteins 1 and 2 are arteries and arterioles endothelium-dependent vasodila-
linked to GPCRs that enhance responses to vasocon- tation is mediated by hyperpolarization, but the factors
strictors including thromboxane, Ang II, and norepineph- responsible for this have largely defied identification. In
rine. Almost 20 years ago, Heximer et al58 showed that the past decade, there has been increasing interest in
mice lacking RGS2, expressed predominantly in vascular the role of direct communications or gap junctions that
smooth muscle, develop baseline hypertension that is permit transmission of signals between the endothelium
dependent on the angiotensin type 1 receptor. Additional and vascular smooth muscle cells. Early studies using
studies showed that vascular smooth muscle cells from intracellular labeling showed polarized unidirectional
these mice displayed augmented calcium transients, with movement of fluorescent dyes from the endothelium to
delayed declines to baseline following agonist stimula- adjacent smooth muscle cells.69 These dyes were shown
tion. A key role for RGS2 was highlighted in subsequent to traverse MEJs (myoendothelial junctions) that were
work showing that RGS2 in the renal vasculature was subsequently identified as holes in the internal elastic

Circulation Research. 2021;128:847–863. DOI: 10.1161/CIRCRESAHA.121.318082 April 2, 2021   851


Harrison et al Pathophysiology of Hypertension
Hypertension Compendium

membrane lying between the endothelium and vascular inflammatory cytokines released by immune cells. Many
smooth muscle. MEJs are particularly common in smaller of these enhance the expression or activity of matrix
arterioles70 and are sites of signaling events and signal- metalloproteinases that promote cell migration, hyper-
ing molecules, including calcium pulsars, the inositol 3 trophy, and reorganization.
phosphate receptor, and calcium-sensitive potassium A related alteration of vascular function that likely
channels.71 Endothelium-dependent hyperpolarization is contributes to hypertension is stiffening of large conduit
transmitted via MEJs directly to vascular smooth muscle arteries, and, in particular, the proximal aorta, via mecha-
cells. MEJs also facilitate the transfer of NO generated nisms that continue to be investigated. A healthy aorta
by NOS that is expressed at these junctions. The NO distends during systole and recoils in diastole. This leads
travels from the endothelium to the vascular smooth to a reduction in the systolic pressure while maintain-
muscle where it regulates connexin43 to modulate the ing diastolic pressure and perfusion. The aorta stiffens
permeability of the MEJs.72 MEJs also contain hemoglo- in common conditions, including aging, diabetes, obesity,
bin alpha, which controls the passage of NO from the and tobacco use, thereby reducing this critical Windkes-
endothelium to the vascular smooth muscle cells in a sel function.84–87 Increased pulse wave velocity, reflecting
redox-dependent fashion.73 Intriguingly, a peptide mimic aortic stiffening, has been reported in population stud-
that disrupts the binding of NO to hemoglobin alpha ies to precede hypertension by several years and is a
lowers BP abruptly in wild-type animals and reverses harbinger of stroke, myocardial infarction, and renal fail-
experimental hypertension caused by Ang II infusion.74 ure.88 Aortic stiffening can be both a cause and a conse-
Importantly, the density of MEJs is reduced and their quence of hypertension. It develops well before the onset
local inositol trisphosphate–stimulated calcium signals of hypertension in spontaneously hypertensive rats,89 in
are blunted in mesenteric arterioles of spontaneously diet-induced obesity,90 and in 2 models of chronic vascu-
hypertensive rats, indicating that these signaling loci are lar oxidative stress.91 In contrast, Wolinsky92 reported that
deficient in hypertensive resistance vessels.75 In keeping aortic collagen increases following the onset of Goldblatt
with this, mice lacking the MEJ component connexin40 hypertension, and we found that experimental hyperten-
exhibit hypertension and reduced endothelium-depen- sion in mice causes aortic stiffening. Mechanical stretch
dent vasodilatation in response to the potassium channel of cultured aortic murine fibroblasts increases their
activator SKA-31. Likewise, endothelial-specific expres- expression of collagens I, III, and V. Wu et al91 found that
sion of a mutant connexin40, which causes electrical aortic stiffening occurs before BP elevation in 2 models
impairment of gap junctions, leads to exercise-induced of chronic vascular oxidative stress. It is thus likely that
Downloaded from http://ahajournals.org by on April 23, 2024

hypertension.76 Recently, it has been shown that chronic aortic stiffening and hypertension act in concert, with one
sympathetic stimulation in the setting of hypertension leading to the other in a feed-forward fashion.93
due to experimental chronic kidney disease decreases Although the mechanisms by which aortic stiffening
the expression of connexin43, leading to disruption of causes end-organ damage are not completely under-
MEJ function and enhanced vasoconstriction.77 These stood, a consequence is enhanced propagation of pres-
various lines of evidence strongly support the role of sure into the microcirculation, leading to barotrauma
MEJs in modulation of microvascular tone and abnor- in target organs, in part via increasing cyclical stretch
malities thereof in hypertension. of microvascular endothelial cells. Indeed, in an ele-
Perturbations of both microvascular and large ves- gant analysis of the components of the pulse wave, an
sel structure represent a second vascular contribution increased forward pressure wave was associated with an
to hypertension. More than 60 years ago, Folkow et increased risk for incident cardiovascular disease.94 Drs
al78 proposed that vascular smooth muscle hypertro- Boutouyrie, Chowienczyk, and Mitchell discuss vascular
phy and concomitant narrowing of the arteriolar lumen stiffening in greater detail in their review of this issue.
increases systemic vascular resistance. This phenom- A fourth role of the vasculature in hypertension is to
enon has been detected in the retinal arterioles by fun- serve as a source and target of immune activation. This
doscopic examination in clinical hypertension79 where is in part mediated by a cross-talk between the endo-
a reduced arteriole to venule ratio increases the odds thelium and immune cells that is covered in depth in the
of developing hypertension. This indicates that arterio- section on immune mechanisms below.
lar remodeling can precede, and possibly predispose, to Finally, there is an impaired vessel wall defense
hypertension.80 The application of artificial intelligence against thrombosis in hypertension that has been related
and machine learning methods to quantitate parameters to increased endothelial expression of tissue factor and
of retinal arterial morphology have further demonstrated the vascular cell adhesion molecule 1. This enhances
that retinal artery morphology correlates with cardiovas- platelet-dependent leukocyte adhesion and is driven
cular outcomes.81 There are multifactorial mechanisms by thrombin factor XI and the platelet factor XI recep-
of arteriolar remodeling in hypertension.82 These include tor.95 Inhibition of factor XI in angiotensin-infused rats
the direct hypertrophic/growth–promoting actions of and mice prevents platelet thrombin formation, vascu-
Ang II and catecholamines, oxidative signaling,83 and lar leukocyte infiltration, endothelial dysfunction, and

852   April 2, 2021 Circulation Research. 2021;128:847–863. DOI: 10.1161/CIRCRESAHA.121.318082


Harrison et al Pathophysiology of Hypertension

Hypertension Compendium
hypertension. Further studies of the mechanisms that nerve stimulation is the heart. Infusion of saline into
activate thrombosis in hypertension may provide insight cardiac transplant recipients with no evidence of car-
into stroke, atherosclerosis, and end-organ damage. Drs diac dysfunction failed to reduce their sympathetic
Webb, Eguchi, and Tostes cover the role of the vascula- nervous system activity, arginine vasopressin levels or
ture in greater depth in their review. their renin, angiotensin, and aldosterone levels and led
to hypertension.103,104 This hypertensive response was
linked to a failure to suppress Ang II during blood vol-
THE CENTRAL NERVOUS SYSTEM IN ume expansion and was prevented by angiotensin-con-
verting enzyme inhibition.105
HYPERTENSION A summary of some of the roles and perturbations of
Dr Page’s revised diagram (Figure 1B) included the term renal, vascular, endocrine, and neural control in hyperten-
“neural.” Indeed Page demonstrated that Ang II caused sion are depicted in Figure 2. These are organ-specific
hypertension predominantly by sympathetic neural acti- features of the Mosaic Theory and hypertension patho-
vation.13 Measures of muscle sympathetic nerve activity, physiology that remain the focus of ongoing research. In
norepinephrine spillover, and heart rate variability have the past 2 decades, new factors and phenomena have
suggested that humans with hypertension commonly emerged that likely affect the mosaic factor in a global
have increased sympathetic outflow and enhanced cat- fashion. These include oxidative signaling, inflammation,
echolamine mediated vasoconstriction.96 Ang II and ROS and the microbiome. In addition, a new role of sodium
in the brain stem nuclei enhance vascular resistance by has been identified. These are briefly considered in the
in part by inhibition of the microvascular endothelium- following paragraphs.
dependent hyperpolarizing factor that normally reduces
vascular tone.77 In keeping with these central vascu-
lar effects, ganglionic blockade acutely lowers BP in ALDOSTERONE, THE
humans with hypertension to a variable extent,97 particu- MINERALOCORTICOID RECEPTOR
larly in those who are obese.98
AND THEIR UBIQUITOUS ROLE IN THE
The many mechanisms whereby sympathetic tone
modulates hypertension include enhanced vasocon- MOSAIC THEORY
striction and vascular remodeling, renal renin production A major development since the initial Mosaic theory is the
via beta 1 adrenergic receptors in the juxtaglomerular evolving understanding of aldosterone in hypertension.
Downloaded from http://ahajournals.org by on April 23, 2024

apparatus, and enhanced renal sodium resorption and Aldosterone is produced by the zona granulosa cells of
inflammation. the adrenal gland in response to Ang II and elevations of
The central pathways involved in hypertension have extracellular potassium. Until recently, the sole effect of
been reviewed elsewhere.99 An important initiating site aldosterone was thought to enhance sodium reabsorp-
is the laminal terminalis, located anterior to the third ven- tion in the collecting duct, and aldosterone blockade was
tricle, which contains the subfornical organ, the median felt to predominantly promote diuresis. It has become
preoptic eminence, and the organum vasculosum of the obvious that the MR (mineralocorticoid receptor) is
lamina terminalis. The subfornical organ and organum expressed in numerous organs and cells, including the
vasculosum of the lamina terminalis are circumventricu- heart, blood vessels, immune cells, and in the brain. Stud-
lar organs that have a poorly formed blood-brain barrier ies using cre-lox technology have shown important roles
that permits blood-born Ang II and sodium to activate of many of these extra-renal sites in hypertension. As
neuronal firing. Indeed, the organum vasculosum of the an example, mice with vascular smooth muscle deletion
lamina terminalis is responsive to even modest increases of MR have decreased BP with aging, and their vessels
in sodium to initiate salt-sensitive hypertension.100 Xiao exhibit diminished vascular myogenic tone, agonist-
et al101 recently identified a role of the prostaglandin EP3 dependent contraction, vascular superoxide production
(E prostanoid receptor type 3) receptor in this response, and expression, and L-type calcium channel activity.106
leading to a cascade of ROS production in renal arteri- Likewise, Jia et al107 showed that endothelial cell MR
oles, isolevuglandin accumulation in dendritic cells, and deficiency prevents aortic stiffening caused by feeding
immune activation, thereby providing a contemporary a Western diet and established a role for the endothelial
example of the mosaic interaction between neural regu- MR receptor in modulating endothelial sodium channel
lation, vasculature, and inflammation. activation, oxidative stress, and endothelial NO synthase
In addition to efferent signals promoting hyperten- activation. Sun et al108 used Cre-Lox technology to elimi-
sion, there is ample evidence that afferent pathways are nate the MR in all T cells and showed that this abrogated
likewise involved. In addition to renal afferent activation, T-cell IFN (interferon) γ production and the hyperten-
discussed above, afferent nerves from adipose tissue sion caused by chronic infusion of Ang II. These authors
are triggered by a high-fat diet to reflexively increase showed that the MR interacted with other transcription
BP and insulin resistance.102 Another source of afferent factors to mediate IFNγ production and that its deletion

Circulation Research. 2021;128:847–863. DOI: 10.1161/CIRCRESAHA.121.318082 April 2, 2021   853


Harrison et al Pathophysiology of Hypertension
Hypertension Compendium

Figure 2. Perturbations of the kidney (A), vasculature (B), and central nervous system (C) contributing to hypertension.
ADMA indicates asymmetrical dimethylarginine; GPCR, G protein–coupled receptor; JG, juxtaglomerular apparatus; ROS, reactive oxygen species;
and VSM, vascular smooth muscle.

from T cells attenuated fibrosis in the aorta and kidneys, caliber and reactivity. ROS are important for vascular
reduced albuminuria, and preserved endothelium-depen- stiffening and for remodeling of microarterioles.83,91 In
Downloaded from http://ahajournals.org by on April 23, 2024

dent vasodilatation. A role for the MR in macrophage the kidney, ROS enhance renal sodium reabsorption117
activation has been reviewed elsewhere.109 and thus enhance volume and cardiac output. They have
These myriad effects of the MR receptor are impor- also been implicated in activation of matrix metallopro-
tant because at least 5% of all cases of hypertension are teinases and thus promote tissue remodeling.118 ROS
due to primary hyperaldosteronism and this is likely much likewise increase neuronal firing in critical brain centers,
higher among those with resistant hypertension. Brown et including the subfornical organ,119,120 and brain stem cen-
al recently reported that urinary aldosterone levels (in the ters,121 thus altering neural control.
sodium loaded state) parallel the severity of hypertension Rajagopalan et al114 and Griendling et al122 showed in
across all subjects,110 suggesting that hyperaldosteronism the 1990s that Ang II could stimulate superoxide pro-
is much more prevalent than previously considered, even duction in cultured vascular smooth muscle cells and in
among those without severe hypertension. vivo by activating an enzyme similar to the phagocytic
NADPH oxidase. The catalytic subunit of the NADPH
oxidase is Nox (NADPH oxidase catalytic subunit) 2, and
THE ROLE OF ROS IN HYPERTENSION homologs of this are activated in vascular smooth mus-
It was first observed in the 1990s that scavenging super- cle cells, the endothelium, renal tubular cells, and in the
oxide could lower BP in rats with hypertension due to central nervous system in hypertensive animal models.
chronic Ang II infusion and in spontaneously hyperten- There is clearly an interaction of ROS derived from the
sive rats.111–114 It has subsequently been recognized that NADPH oxidase and with other enzymatic sources, such
several enzymatic sources of ROS are activated in tis- that ROS from one can trigger ROS production by others
sues of hypertensive animals and humans, including the in a feed-forward fashion.
nicotinamide adenine dinucleotide phosphate (NADPH) Although early studies emphasized the role of oxida-
oxidases, the mitochondria, and uncoupled NO synthase. tive stress in vascular cells, the kidney and its vasculature
ROS likely contribute to almost each node of Dr Page’s develop oxidative stress with increased NADPH oxidase and
diagram. Superoxide and other related ROS can oxi- reduced SOD (superoxide dismutase) activity both during
datively inactivate NO and oxidize tetrahydrobiopterin, Ang II infusion and during high salt intake. High salt intake
a critical cofactor for NO synthase.115,116 ROS can also enhances renal expression of NADPH oxidase and NOX2
enhance vasoconstriction and thereby alter vascular and reduces SOD1 and SOD2,123 whereas Ang II infusion

854   April 2, 2021 Circulation Research. 2021;128:847–863. DOI: 10.1161/CIRCRESAHA.121.318082


Harrison et al Pathophysiology of Hypertension

Hypertension Compendium
increases renal NADPH/p22phox and reduces SOD 3 expres- the interstitium of the kidney and blood vessels where they
sion.124 Renal ROS can activate renal afferent nerves,125 and release potent cytokines, ROS, and metalloproteinases that
increase renal and circulating levels of ADMA,126 by reduc- modulate renal and vascular function and structure. It is
ing its metabolism by dimethylarginine dimethylaminohy- likely that these mediators affect virtually every node of Dr
drolase and increasing its generation by protein arginine Page’s Mosaic diagram. A notable example is the cytokine
methyltransferase.66 Thus, an increase in circulating ADMA IL-17A (interleukin 17A) produced by a subset of T cells,
is one means whereby ROS in the kidney may enhance innate lymphocytes, histiocytes, and renal tubular cells. IL-
peripheral vasoconstriction. ADMA inhibits endothelial func- 17A affects renal tubular handling of sodium and seems
tion and flow-mediated vasodilation and activation of the to modulate pressure natriuresis.129 This cytokine stimu-
sympathetic nervous system during high salt intake could lates vascular superoxide production,130 causes inhibitory
contribute to the failure of salt-sensitive subjects to reduce phosphorylation of the endothelial NOS,131 and therefore,
their peripheral vascular resistance during increases in car- reduces the caliber of blood vessels. Over the long term, IL-
diac output evoked by a high salt intake. In the kidney, the 17A promotes vascular fibrosis and impairs vascular elas-
proximal tubular cells take up oxidatively modified albumin ticity.132 In contrast, regulatory T cells and anti-inflammatory
that induces a robust local activation of the RAS via NADPH cytokines, such as IL-10, have antihypertensive effects
oxidase, protein kinase C, NFκB (nuclear factor-κB), and mediated, in part, by counteracting IL-17A.133,134
AP-1 (activator protein 1).27 These together increase proxi- The manner by which immune cells are activated in
mal tubular Ang II that initiates renal inflammation and fibro- hypertension is a matter of ongoing investigation. Accu-
sis. This proximal tubule ROS pathway provides a potential mulating data suggest that ROS in antigen-presenting
link between hypertension, systemic oxidative stress, and cells promote oxidation of fatty acids to form highly reac-
proteinuria and the activation of the intrarenal RAS. These tive isolevuglandin products that covalently modify pro-
events damage the kidney and can thereby perpetuate salt- tein lysines.135 These altered proteins are antigenic and
sensitivity and worsening hypertension. activate both CD8+ and CD4+ T cells.
Of interest is an emerging role of the mitochondria as There is substantial evidence that sympathetic outflow
a source of ROS in hypertension. Recent studies suggest promotes immune activation in hypertension. Lesions of the
that aging and hypertension reduce the function of Sirt3 forebrain that prevent sympathetic outflow reduce T-cell
(sirtuin3), a histone deacetylase that modulates the acety- activation,119,136 whereas manipulations that increase sym-
lation status of SOD2. Mice lacking Sirt3 are protected pathetic outflow promote T-cell activation.137 Recent data
against hypertension and its associated vascular dysfunc- suggest that renal sympathetic tone modulates trafficking
Downloaded from http://ahajournals.org by on April 23, 2024

tion in response to Ang II infusion, and this is prevented by of immune cells to and from the kidney and the homing
mitochondrial-targeted antioxidants, including mitoTempol of memory T cells to the bone marrow, where they can
and mitoEbselen.127 These alterations of the mitochon- be reactivated upon future hypertensive challenges.53,138 In
dria in hypertension likely have myriad effects on cellular the vasculature, T cells accumulate in the perivascular fat
metabolism and function that contribute to the pathophysi- and adventitia of larger vessels in hypertension, likely via
ology of hypertension through yet undefined mechanisms. expression of chemokines, such as RANTES (regulated
Therapeutically, scavenging ROS is not clinically upon activation, normal T-cell expressed and presumably
employed, and there is evidence that high-dose vitamin secreted) and MCP (monocyte chemoattractant pro-
antioxidants can be harmful, possibly because ROS can tein)-1.139,140 This adventitial inflammatory response likely
have important signaling function.128 Targeted scaveng- triggers collagen deposition, as described above. There is
ing of ROS using agents like mitoTempol or treatment also an immune reflex, whereby local tissue signals trig-
with agents that inhibit untoward activation of ROS form- ger a vagally mediated anti-inflammatory response.141 This
ing enzymes are viable future options. Indeed, one of the reflex seems to be miswired to promote inflammation in
major benefits of Ang II blockade might be due to inhibi- prehypertensive animals.142
tion of ROS formation. This topic is reviewed in depth in Hypertension is associated with endothelial activation.
this compendium by Drs Griendling and Touyz. It has long been recognized that the activated endothe-
lium promotes rolling, adhesion, and transmigration of
leukocytes. As monocytes transmigrate the endothelium,
INFLAMMATORY AND IMMUNE they are transformed into inflammatory macrophages and
dendritic cells that emerge as activated monocytes with
MECHANISMS IN HYPERTENSION enhanced potential to produce cytokines and activate T
It has been recognized for >50 years that inflammatory cells.143,144 Endothelial cell stretch, oxidative stress, and
cells accumulate in the kidney and blood vessels of hyper- loss of NO in hypertension can all enhance endothelial/
tensive humans and animals with experimental hyper- leukocyte interaction and increase the endothelial activa-
tension. It is now recognized that virtually every type of tion of monocytes. Human monocytes activated in this way
immune cell, including those of innate and adaptive immu- by the endothelium produce copious quantities of IL-6,
nity, contributes to hypertension. These transmigrate into IL-23, IL-1β, and stimulate autologous T cells potently.145

Circulation Research. 2021;128:847–863. DOI: 10.1161/CIRCRESAHA.121.318082 April 2, 2021   855


Harrison et al Pathophysiology of Hypertension
Hypertension Compendium

In addition, endothelial transmigration likely promotes populations, sodium-sensitivity, defined as an exagger-


myeloid cell accumulation in the corticomedullary junction ated change in BP in response to extremes of dietary
of the kidney, where supraphysiological levels of sodium salt intake, is a relatively common phenotype associated
may provide a stimulus to activate monocytes.146 with increased risk for hypertension and cardiovascular
This vascular/immune cross-talk again reflects the events.153 The BP of about one-quarter of normal sub-
mosaic nature of hypertension, whereby a primary per- jects and one-half of hypertensive increases with salt
turbation, in this case of endothelial (vascular) function, intake.154 Although salt-sensitive hypertension entails a
in turn, generates mediators that promote inflammation renal defect in salt excretion, the increase in BP with salt
that likely affects multiple other organs and cells. In this intake is caused by increased peripheral resistance that
compendium, Drs Madhur, Kirabo, and colleagues pro- requires a communication between the kidneys and the
vide an in-depth review that discusses more fully the role peripheral vasculature. Indeed, enhanced vasoconstric-
of inflammation in hypertension. tion and endothelial dysfunction,155 rather than a primary
increase in renal Na+ retention, can initiate salt-sensitive
hypertension.156 Salt-sensitive subjects are unable to
GENETIC INFLUENCES increase peripheral vascular resistance appropriately dur-
ing a low salt intake157 or to reduce their peripheral vascu-
Genetic alterations are atop Dr Page’s revised Mosaic
lar resistance during a high salt intake,158 thereby causing
diagram (Figure 1B). Several single-gene mutations,
their BP to be unusually dependent on salt intake. There is
including those responsible for Liddle syndrome, pseu-
an accompanying failure of salt-sensitive subjects to sup-
dohyperaldosteronism, aldosterone-producing adenomas,
press the sympathetic nervous system during salt load-
glucocorticoid remedial hypertension, and missense muta-
ing,159 accompanied by an increase in circulating ADMA
tions of the mineralocorticoid receptor have been identi-
during salt loading only in salt-sensitive subjects.158
fied to be responsible for highly heritable Mendelian forms
Classic Guytonian models suggest that a defect in
of hypertension.147 Although these single-gene mutations
sodium excretion by the kidney is the primary basis for
have provided insight into the pathogenesis of hyperten-
hypertension, with impaired elimination of sodium during
sion, they are rare and do not explain the high prevalence
high salt feeding leading directly to expanded extracel-
of familial hypertension commonly observed in the clinic. In
lular fluid volume promoting increased BP.18 Studies in
contrast, single nucleotide polymorphisms that often occur
recent years indicate that the impact of dietary sodium
in nonprotein coding regions of the genome and usually
intake upon BP is complex, beyond the simple construct
Downloaded from http://ahajournals.org by on April 23, 2024

do not alter protein function are common. Genome-wide


of intravascular volume expansion balanced by excretory
association studies in the past decade have identified
functions of the kidney. For example, it is now apparent
>1000 single nucleotide polymorphisms that associate
that during high salt feeding, sodium accumulates in the
with hypertension.148 Although individual single nucleotide
interstitium of the skin and other tissues where it may
polymorphisms have small effects on BP, when multiple
be stored at hypertonic concentrations in complexes
single nucleotide polymorphisms are analyzed as a poly-
with proteoglycans, acting as a reservoir and buffer-
genic risk score (PRS), they can account for as much as
ing the impact of sodium accumulation on intravascular
13 mm Hg of BP variability. Vauraa et al149 recently showed
volume and BP.160 This interstitial sodium storage can
such a PRS is predictive of early-onset hypertension in a
be visualized and quantified in humans using magnetic
progressive fashion, such that those with the highest 2.5%
resonance imaging of the 23sodium isotope.161 Accu-
of PRS have an almost 3-fold risk of developing hyper-
mulation of sodium in tissues increases with high salt
tension, whereas a low PRS is protective. Refinements of
intake or aging and has been associated with hyperten-
the PRS promise to contribute to our future understanding
sion and increased cardiovascular risk.162 In the sub-
and treatment of hypertension. The genetics in hyperten-
dermal space, sodium can also stimulate macrophages,
sion will be reviewed in detail by Dr Caufield in a subse-
triggering expression of TonEBP (tonicity-responsive
quent section of this compendium.
enhancer-binding protein), a transcription factor regulat-
ing the expression of osmo-protective genes including
VEGF (vascular endothelial growth factor)-C, a potent
NOVEL CONCEPTS RELATED TO SALT inducer of lymphangiogenesis.160 It has been suggested
AND SODIUM INTAKE that these immunomodulatory actions of salt deposited
Dr Page’s revised Mosaic Diagram (Figure 1B) included in skin may have evolved to subserve a barrier function to
the environment, which includes diet, and in particular prevent invasion by micro-organisms, but in the modern
salt intake. There are numerous large epidemiological world where dietary salt is ubiquitous, this may now be
studies linking high levels of dietary sodium intake with a maladaptive response.163 Other recent studies indicate
risk for hypertension.150 Conversely, reducing dietary salt that very high levels of sodium intake can trigger arche-
or administration of diuretic agents, such as thiazides, typal metabolic changes aimed at conserving water, with
are effective treatments for elevated BP.151,152 In human a side effect of raising BP.164

856   April 2, 2021 Circulation Research. 2021;128:847–863. DOI: 10.1161/CIRCRESAHA.121.318082


Harrison et al Pathophysiology of Hypertension

Hypertension Compendium
Elevated concentrations of tissue sodium can also contributions of the microbiome in the pathogenesis of
activate immune cells that contribute to hypertension. several diseases, including hypertension.174,175 This work
Modest elevations of sodium can stimulate T cells to has also identified conditioning effects of diet and drugs
produce the cytokine IL-17A,165,166 which as mentioned on the composition of the gut microbiota and high-
above, promotes vascular remodeling, endothelial dys- lighted the potential of these disturbances to affect host
function, and renal sodium retention. Sodium entry via responses including BP regulation.
an amiloride-sensitive sodium channel can also drive In healthy individuals, the gut microbiome is typically
dendritic cell activation167 and promotes transformation in a stable state of eubiosis and in relative equilibrium
of human monocytes to a dendritic cell–like phenotype with the surrounding community.176 By contrast, signifi-
that produces cytokines like IL-6, TNF (tumor necrosis cant alteration in the microbiome (dysbiosis) has been
factor) α, and IL-1β.146 Upon entry of sodium into myeloid observed in range of disorders including hypertension.
cells, calcium activation of the NADPH oxidase ensues For example, in humans with hypertension and prehy-
leading to ROS formation and the generation of isolev- pertension, characteristic alterations in the microbiome
uglandin adducts, which as discussed above act as neo- have been observed.177,178 Dysbiosis has also been
antigens in these cells. seen in experimental models of hypertension, such as
In both T cells and myeloid cells, the effect of sodium Dahl salt–sensitive and spontaneously hypertensive
is mediated by the SGK1 (serum glucocorticoid kinase rats,179,180 and in mice chronically administered Ang II
1). Wu et al165 showed that high salt feeding stimulated or deoxycorticosterone acetate+high-salt diet.181 Fecal
Th17 (T-helper cell 17) cells in the central nervous transfer from hypertensive mice to germ-free mice sen-
system and mesenteric lymph nodes and exacerbated sitizes the latter to hypertension, providing strong evi-
experimental allergic encephalitis, a model of multiple dence for a role of the microbiome.178 In hypertension,
sclerosis mediated by IL-17A. Interestingly, mice with alterations in the gut microbiome have been associated
T-cell SGK1 deficiency exhibited reduced experimen- with inflammation, structural changes in the intestinal
tal allergic encephalitis incidence and severity that was wall, and enhanced gut permeability.182 Altered intesti-
not exacerbated by high salt feeding. Norlander et al168 nal permeability may facilitate egress of mediators, such
showed that deletion of SGK1 in T cells blunts the as microbial products, hormones, and immune cells into
hypertensive response to Ang II and deoxycorticosterone the circulation where they can impact peripheral and
acetate–salt challenge in mice and abrogates renal and central BP control mechanisms.
vascular inflammation in these models. In this study, a Along with associations between hypertension and gut
Downloaded from http://ahajournals.org by on April 23, 2024

T-cell sodium-potassium-2 chloride cotransporter 1 was dysbiosis, there is a direct relationship between diet and
found to mediate the salt-induced increase in SGK1 and the composition of gut microbiota with potential to affect
the IL-23R (IL-23 receptor), which in turn promoted IL- propensity for hypertension. For example, increased
17A production. Likewise, deletion of this kinase specifi- dietary salt intake causes significant alterations in the
cally in antigen-presenting cells markedly reduces renal composition of microbiota and has been associated with
inflammation and hypertension in a salt-sensitive model accumulation of proinflammatory TH17 cells discussed
of hypertension.169 In this myeloid cell subset, SGK1 pro- above.170 Sodium is not the only dietary constituent with
motes assembly of the alpha and gamma subunits of the potential to impact BP through effects on the microbi-
epithelial sodium channel, which further promotes sodium ome. Multiple studies have shown that diets enriched
entry. These studies suggest that diuretic agents previ- in fruit, vegetables, and fiber can lower BP and reduce
ously thought to act only on the kidney might prove use- risk for hypertension.183 Moreover, dietary fiber increases
ful in reducing salt-driven inflammation in hypertension. bacterial populations in the gut responsible for gener-
ating short-chain fatty acids through fermentation.184 In
the human circulation, short-chain fatty acids are almost
exclusively of microbial origin,185 and have been impli-
MICROBIOME AS AN EMERGING
cated as a key pathway whereby microbiota influence
MODULATOR OF BP host physiology,186 perhaps through a family of nutrient-
Along with effects on fluid homeostasis and immune sensing GCPRs expressed in vascular smooth muscle
activation, sodium as a dietary constituent may promote cells, the autonomic nervous system and renal juxtaglo-
hypertension by modifying gut microbiota.170 Indeed, merular cells influencing renin release.171,187 Thus, the gut
many recent studies have highlighted the potential microbiome can influence BP control through pathways
of the microbiome to influence BP.171–173 Advances in affecting many of Page’s Mosaic components, raising
unbiased sequencing techniques have allowed precise the possibility that prebiotics and probiotics, antibiotics,
identification and assessment of commensal bacterial and specific dietary formulations could be used to modify
species in the gut that cannot be cultured, transforming gut microbiome and its metabolites to prevent and treat
understanding of the role of the microbiome in health hypertension. The potential impact of the microbiome in
and disease. Such studies have provided evidence for hypertension will be reviewed by Drs Muller and Luft.

Circulation Research. 2021;128:847–863. DOI: 10.1161/CIRCRESAHA.121.318082 April 2, 2021   857


Harrison et al Pathophysiology of Hypertension
Hypertension Compendium

Disclosures
None.

REFERENCES
1. Whelton PK, Carey RM, Aronow WS, Casey DE Jr, Collins KJ,
Dennison Himmelfarb C, DePalma SM, Gidding S, Jamerson KA, Jones
DW, et al. 2017 ACC/AHA/AAPA/ABC/ACPM/AGS/APhA/ASH/
ASPC/NMA/PCNA guideline for the prevention, detection, evaluation,
and management of high blood pressure in adults: a report of the Ameri-
can College of Cardiology/American Heart Association Task Force on
Clinical Practice Guidelines. J Am Coll Cardiol. 2018;71:e127–e248. doi:
10.1016/j.jacc.2017.11.006
2. GBD 2019 Risk Factors Collaborators. Global burden of 87 risk factors
in 204 countries and territories, 1990-2019: a systematic analysis for the
Global Burden of Disease Study 2019. Lancet. 2020;396:1223–1249. doi:
10.1016/S0140-6736(20)30752-2
3. Mills KT, Bundy JD, Kelly TN, Reed JE, Kearney PM, Reynolds K, Chen
Figure 3. Mosaic Theories Proposed by Page. J, He J. Global disparities of hypertension prevalence and control: a sys-
A revised Mosaic Theory incorporating new understanding of cellular, tematic analysis of population-based studies from 90 countries. Circulation.
environmental, and genetic mechanisms. 2016;134:441–450. doi: 10.1161/CIRCULATIONAHA.115.018912
4. Tigerstedt R, Bergman P. Niere und Kreislauf. Scand Arch Physiol.
1898;8:223–271.
SUMMARY 5. Sarre H. Franz Vollhard: Roter und blasser Hochdruck aus heutiger sicht.
Med Klin. 1973;68:1309–1312.
These considerations of the Mosaic Theory and beyond 6. Luft FC, Dietz R. Franz Volhard in historical perspective. Hypertension.
reflect the prescient nature of Dr Page’s original con- 1993;22:253–256. doi: 10.1161/01.hyp.22.2.253
7. Goldblatt H, Lynch J, Hanzal RF, Summerville WW. Studies on experimen-
cept, depicted in Figures 1A and 1B, which is notable tal hypertension: I. The production of persistent elevation of systolic blood
for the interdependence of the various components, pressure by means of renal ischemia. J Exp Med. 1934;59:347–379. doi:
such that a change in one can almost certainly perturb 10.1084/jem.59.3.347
8. Braun-Menendez E, Fasciolo JC, Leloir LF, Muñoz JM. The substance caus-
the others. As research has evolved, a newer Mosaic ing renal hypertension. J Physiol. 1940;98:283–298. doi: 10.1113/jphysiol.
diagram could be constructed as shown in Figure 3, 1940.sp003850
emphasizing the interplay of factors like inflamma- 9. Page IH. On the nature of the pressor action of renin. J Exp Med.
1939;70:521–542. doi: 10.1084/jem.70.5.521
tion, oxidative stress, the microbiome with alterations
Downloaded from http://ahajournals.org by on April 23, 2024

10. Braun-Menendez E, Page IH. Suggested revision of nomenclature–angio-


of renal, neural and vascular function. It should be tensin. Science. 1958;127:242. doi: 10.1126/science.127.3292.242-a
stressed that the role of these and other factors almost 11. Page IH, Helmer OM. A crystalline pressor substance (Angiotonin) result-
ing from the reaction between renin and renin-activator. J Exp Med.
certainly vary from one hypertensive human to the next 1940;71:29–42. doi: 10.1084/jem.71.1.29
or upon the experimental model studied. Importantly, 12. Page IH. Pathogenesis of arterial hypertension. J Am Med Assoc.
another factor that should be considered is that BP 1949;140:451–458. doi: 10.1001/jama.1949.02900400005002
13. McCubbin JW, DeMoura RS, Page IH, Olmsted F. Arterial hypertension
elevation can influence hypertension by initially caus- elicited by subpressor amounts of angiotensin. Science. 1965;149:1394–
ing baroreflex activation and pressure natriuresis (to 1395. doi: 10.1126/science.149.3690.1394
lower BP) and ultimately by producing pathophysi- 14. Page IH. The nature of arterial hypertension. Arch Intern Med.
1963;111:103–115. doi: 10.1001/archinte.1963.03620250107016
ological effects that progressively raise BP in a feed- 15. Page IH. The mosaic theory 32 years later. Hypertension. 1982;4:177. doi:
forward fashion. These features of the Mosaic Theory 10.1161/01.hyp.4.2.177
will undoubtedly guide hypertension research in the 16. Lip S, Padmanabhan S. Genomics of blood pressure and hyperten-
sion: extending the mosaic theory toward stratification. Can J Cardiol.
foreseeable future. 2020;36:694–705. doi: 10.1016/j.cjca.2020.03.001
17. Harrison DG. The mosaic theory revisited: common molecular mechanisms
coordinating diverse organ and cellular events in hypertension. J Am Soc
ARTICLE INFORMATION Hypertens. 2013;7:68–74. doi: 10.1016/j.jash.2012.11.007
18. Guyton AC, Coleman TG, Cowley AW Jr, Liard JF, Norman RA Jr, Manning
Affiliations RD Jr. Systems analysis of arterial pressure regulation and hypertension.
Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Ann Biomed Eng. 1972;1:254–281. doi: 10.1007/BF02584211
Medical Center, Nashville, TN (D.G.H.). Cardiovascular and Metabolic Disorders 19. Fasciolo JC, Houssay BA, Taquini AC. The blood-pressure raising secre-
Research Program, Duke-National University of Singapore Medical School, Sin- tion of the ischaemic kidney. J Physiol. 1938;94:281–293. doi: 10.1113/
gapore (T.M.C.). Division of Nephrology and Hypertension Center, Georgetown jphysiol.1938.sp003680
University, District of Columbia (C.S.W.). 20. Dahl LK, Heine M, Thompson K. Genetic influence of renal homografts on
the blood pressure of rats from different strains. Proc Soc Exp Biol Med.
Sources of Funding 1972;140:852–856. doi: 10.3181/00379727-140-36566
Supported by National Institutes of Health (NIH) HL5R35HL140016, 21. Curtis JJ, Luke RG, Dustan HP, Kashgarian M, Whelchel JD, Jones P, Diethelm
P01HL129941, UL1TR001881, and T32HL144446 to D.G. Harrison, by the AG. Remission of essential hypertension after renal transplantation. N Engl
Singapore National Medical Research Council grant OFLCG/001/201 to T.M. J Med. 1983;309:1009–1015. doi: 10.1056/NEJM198310273091702
Coffman and by NIH grants RO-1 HL-134511 and R21-HD101340 and by 22. Crowley SD, Gurley SB, Herrera MJ, Ruiz P, Griffiths R, Kumar AP, Kim HS,
funds from the Walter Family Chair of Cardiovascular Research, the Smith-Ko- Smithies O, Le TH, Coffman TM. Angiotensin II causes hypertension and
god Family Foundation, the Gildenhorn-Speisman Family Foundation, and the cardiac hypertrophy through its receptors in the kidney. Proc Natl Acad Sci
Georgetown University Hypertension Research Center to C.S. Wilcox. USA. 2006;103:17985–17990. doi: 10.1073/pnas.0605545103

858   April 2, 2021 Circulation Research. 2021;128:847–863. DOI: 10.1161/CIRCRESAHA.121.318082


Harrison et al Pathophysiology of Hypertension

Hypertension Compendium
23. Guessoum O, de Goes Martini A, Sequeira-Lopez MLS, Gomez RA. Deci- diuretic and cardiorenal effects. Circulation. 2020;142:1028–1039. doi:
phering the identity of renin cells in health and disease. Trends Mol Med. 10.1161/CIRCULATIONAHA.120.045691
2021;27:280–292. doi: 10.1016/j.molmed.2020.10.003 45. Wilcox CS. Antihypertensive and renal mechanisms of SGLT2 (Sodium-
24. Castrop H, Höcherl K, Kurtz A, Schweda F, Todorov V, Wagner C. Physiol- Glucose Linked Transporter 2) inhibitors. Hypertension. 2020;75:894–901.
ogy of kidney renin. Physiol Rev. 2010;90:607–673. doi: 10.1152/physrev. doi: 10.1161/HYPERTENSIONAHA.119.11684
00011.2009 46. DiBona GF. Neural control of the kidney: functionally specific renal
25. Saleem M, Hodgkinson CP, Xiao L, Gimenez-Bastida JA, Rasmussen ML, sympathetic nerve fibers. Am J Physiol Regul Integr Comp Physiol.
Foss J, Payne AJ, Mirotsou M, Gama V, Dzau VJ, et al. Sox6 as a new 2000;279:R1517–R1524. doi: 10.1152/ajpregu.2000.279.5.R1517
modulator of renin expression in the kidney. Am J Physiol Renal Physiol. 47. Cao W, Li A, Wang L, Zhou Z, Su Z, Bin W, Wilcox CS, Hou FF. A Salt-
2020;318:F285–F297. doi: 10.1152/ajprenal.00095.2019 Induced reno-cerebral reflex activates renin-angiotensin systems and pro-
26. Curnow AC, Gonsalez SR, Gogulamudi VR, Visniauskas B, Simon EE, motes CKD progression. J Am Soc Nephrol. 2015;26:1619–1633. doi:
Gonzalez AA, Majid DSA, Lara LS, Prieto MC. Low nitric oxide bioavail- 10.1681/ASN.2014050518
ability increases renin production in the collecting duct. Front Physiol. 48. Campese VM, Kogosov E. Renal afferent denervation prevents hyperten-
2020;11:559341. doi: 10.3389/fphys.2020.559341 sion in rats with chronic renal failure. Hypertension. 1995;25:878–882. doi:
27. Cao W, Zhou QG, Nie J, Wang GB, Liu Y, Zhou ZM, Hou FF. Albumin over- 10.1161/01.hyp.25.4.878
load activates intrarenal renin-angiotensin system through protein kinase 49. Moss NG, Powell SL, Falk RJ. Intravenous cyclosporine activates afferent and
C and NADPH oxidase-dependent pathway. J Hypertens. 2011;29:1411– efferent renal nerves and causes sodium retention in innervated kidneys in rats.
1421. doi: 10.1097/HJH.0b013e32834786f0 Proc Natl Acad Sci USA. 1985;82:8222–8226. doi: 10.1073/pnas.82.23.8222
28. Prieto MC, Reverte V, Mamenko M, Kuczeriszka M, Veiras LC, Rosales CB, 50. Banek CT, Knuepfer MM, Foss JD, Fiege JK, Asirvatham-Jeyaraj N,
McLellan M, Gentile O, Jensen VB, Ichihara A, et al. Collecting duct pro- Van Helden D, Shimizu Y, Osborn JW. Resting afferent renal nerve discharge
renin receptor knockout reduces renal function, increases sodium excre- and renal inflammation: elucidating the role of afferent and efferent renal
tion, and mitigates renal responses in ANG II-induced hypertensive mice. nerves in deoxycorticosterone acetate salt hypertension. Hypertension.
Am J Physiol Renal Physiol. 2017;313:F1243–F1253. doi: 10.1152/ 2016;68:1415–1423. doi: 10.1161/HYPERTENSIONAHA.116.07850
ajprenal.00152.2017 51. Donazzan L, Mahfoud F, Linz D, Ewen S, Ukena C, Böhm M. Novel and
29. McMurray JJ, Krum H, Abraham WT, Dickstein K, Køber LV, Desai AS, nonpharmacologic approaches to cardio-protection in hypertension. Curr
Solomon SD, Greenlaw N, Ali MA, Chiang Y, et al; ATMOSPHERE Commit- Hypertens Rep. 2014;16:430. doi: 10.1007/s11906-014-0430-3
tees Investigators. Aliskiren, enalapril, or aliskiren and enalapril in heart fail- 52. Foss JD, Fiege J, Shimizu Y, Collister JP, Mayerhofer T, Wood L, Osborn JW.
ure. N Engl J Med. 2016;374:1521–1532. doi: 10.1056/NEJMoa1514859 Role of afferent and efferent renal nerves in the development of AngII-salt
30. Wang GM, Li LJ, Tang WL, Wright JM. Renin inhibitors versus angiotensin hypertension in rats. Physiol Rep. 2018;6:e13602.
converting enzyme (ACE) inhibitors for primary hypertension. Cochrane 53. Xiao L, Kirabo A, Wu J, Saleh MA, Zhu L, Wang F, Takahashi T, Loperena R,
Database Syst Rev. 2020;10:CD012569. doi: 10.1002/14651858. Foss JD, Mernaugh RL, et al. Renal denervation prevents immune cell acti-
CD012569.pub2 vation and renal inflammation in angiotensin II-induced hypertension. Circ
31. Wal P, Wal A, Rai AK, Dixit A. Aliskiren: an orally active renin inhibitor. J Res. 2015;117:547–557. doi: 10.1161/CIRCRESAHA.115.306010
Pharm Bioallied Sci. 2011;3:189–193. doi: 10.4103/0975-7406.80764 54. Cao W, Li A, Li J, Wu C, Cui S, Zhou Z, Liu Y, Wilcox CS, Hou FF. Reno-
32. Békássy ZD, Kristoffersson AC, Rebetz J, Tati R, Olin AI, Karpman D. Cerebral reflex activates the renin-angiotensin system, promoting oxidative
Aliskiren inhibits renin-mediated complement activation. Kidney Int. stress and renal damage after ischemia-reperfusion injury. Antioxid Redox
2018;94:689–700. doi: 10.1016/j.kint.2018.04.004 Signal. 2017;27:415–432. doi: 10.1089/ars.2016.6827
33. Perez-Gomez MV, Ortiz A. Aliskiren and the dual complement inhibition con- 55. Trott DW, Thabet SR, Kirabo A, Saleh MA, Itani H, Norlander AE, Wu J,
Downloaded from http://ahajournals.org by on April 23, 2024

cept. Clin Kidney J. 2020;13:35–38. doi: 10.1093/ckj/sfz142 Goldstein A, Arendshorst WJ, Madhur MS, et al. Oligoclonal CD8+ T cells
34. Engström G, Hedblad B, Berglund G, Janzon L, Lindgärde F. Plasma lev- play a critical role in the development of hypertension. Hypertension.
els of complement C3 is associated with development of hypertension: 2014;64:1108–1115. doi: 10.1161/HYPERTENSIONAHA.114.04147
a longitudinal cohort study. J Hum Hypertens. 2007;21:276–282. doi: 56. Coleman TG, Granger HJ, Guyton AC. Whole-body circulatory autoregula-
10.1038/sj.jhh.1002129 tion and hypertension. Circ Res. 1971;28(suppl 2):76–87.
35. Wenzel UO, Bode M, Kurts C, Ehmke H. Salt, inflammation, IL-17 and hyper- 57. Fagard R, Staessen J. Relation of cardiac output at rest and during exer-
tension. Br J Pharmacol. 2019;176:1853–1863. doi: 10.1111/bph.14359 cise to age in essential hypertension. Am J Cardiol. 1991;67:585–589. doi:
36. Guyton AC. Renal function curve–a key to understanding the pathogenesis 10.1016/0002-9149(91)90896-s
of hypertension. Hypertension. 1987;10:1–6. doi: 10.1161/01.hyp.10.1.1 58. Heximer SP, Knutsen RH, Sun X, Kaltenbronn KM, Rhee MH, Peng N,
37. Wilcox CS, Sterzel RB, Dunckel PT, Mohrmann M, Perfetto M. Renal interstitial Oliveira-dos-Santos A, Penninger JM, Muslin AJ, Steinberg TH, et al. Hyper-
pressure and sodium excretion during hilar lymphatic ligation. Am J Physiol. tension and prolonged vasoconstrictor signaling in RGS2-deficient mice. J
1984;247:F344–F351. doi: 10.1152/ajprenal.1984.247.2.F344 Clin Invest. 2003;111:445–452. doi: 10.1172/JCI15598
38. McDonough AA, Nguyen MT. Maintaining balance under pressure: inte- 59. Gurley SB, Griffiths RC, Mendelsohn ME, Karas RH, Coffman TM. Renal
grated regulation of renal transporters during hypertension. Hypertension. actions of RGS2 control blood pressure. J Am Soc Nephrol. 2010;21:1847–
2015;66:450–455. doi: 10.1161/HYPERTENSIONAHA.115.04593 1851. doi: 10.1681/ASN.2009121306
39. Bautista R, Manning R, Martinez F, Avila-Casado Mdel C, Soto V, Medina 60. Patel J, Chuaiphichai S, Douglas G, Gorvin CM, Channon KM. Vascular wall
A, Escalante B. Angiotensin II-dependent increased expression of Na+- regulator of G-protein signalling-1 (RGS-1) is required for angiotensin II-
glucose cotransporter in hypertension. Am J Physiol Renal Physiol. mediated blood pressure control. Vascul Pharmacol. 2018;108:15–22. doi:
2004;286:F127–F133. doi: 10.1152/ajprenal.00113.2003 10.1016/j.vph.2018.04.002
40. Huber G, Matus A. Microtubule-associated protein 3 (MAP3) expression in 61. Arnold C, Demirel E, Feldner A, Genové G, Zhang H, Sticht C, Wieland T,
non-neuronal tissues. J Cell Sci. 1990;95(pt 2):237–246. Hecker M, Heximer S, Korff T. Hypertension-evoked RhoA activity in vas-
41. Panico C, Luo Z, Damiano S, Artigiano F, Gill P, Welch WJ. Renal proxi- cular smooth muscle cells requires RGS5. FASEB J. 2018;32:2021–2035.
mal tubular reabsorption is reduced in adult spontaneously hyperten- doi: 10.1096/fj.201700384RR
sive rats: roles of superoxide and Na+/H+ exchanger 3. Hypertension. 62. Shimokawa H, Sunamura S, Satoh K. RhoA/Rho-kinase in the cardiovas-
2009;54:1291–1297. doi: 10.1161/HYPERTENSIONAHA.109.134783 cular system. Circ Res. 2016;118:352–366. doi: 10.1161/CIRCRESAHA.
42. Pessoa TD, Campos LC, Carraro-Lacroix L, Girardi AC, Malnic G. Functional 115.306532
role of glucose metabolism, osmotic stress, and sodium-glucose cotrans- 63. Sugimoto M, Nakayama M, Goto TM, Amano M, Komori K, Kaibuchi K. Rho-
porter isoform-mediated transport on Na+/H+ exchanger isoform 3 activity kinase phosphorylates eNOS at threonine 495 in endothelial cells. Biochem
in the renal proximal tubule. J Am Soc Nephrol. 2014;25:2028–2039. doi: Biophys Res Commun. 2007;361:462–467. doi: 10.1016/j.bbrc.2007.07.030
10.1681/ASN.2013060588 64. Behuliak M, Bencze M, Vaněčková I, Kuneš J, Zicha J. Basal and activated
43. Thomson SC, Rieg T, Miracle C, Mansoury H, Whaley J, Vallon V, Singh P. calcium sensitization mediated by RhoA/Rho kinase pathway in rats with
Acute and chronic effects of SGLT2 blockade on glomerular and tubular genetic and salt hypertension. Biomed Res Int. 2017;2017:8029728. doi:
function in the early diabetic rat. Am J Physiol Regul Integr Comp Physiol. 10.1155/2017/8029728
2012;302:R75–R83. doi: 10.1152/ajpregu.00357.2011 65. Li Q, Youn JY, Cai H. Mechanisms and consequences of endothelial nitric
44. Griffin M, Rao VS, Ivey-Miranda J, Fleming J, Mahoney D, Maulion C, Suda oxide synthase dysfunction in hypertension. J Hypertens. 2015;33:1128–
N, Siwakoti K, Ahmad T, Jacoby D, et al. Empagliflozin in heart failure: 1136. doi: 10.1097/HJH.0000000000000587

Circulation Research. 2021;128:847–863. DOI: 10.1161/CIRCRESAHA.121.318082 April 2, 2021   859


Harrison et al Pathophysiology of Hypertension
Hypertension Compendium

66. Wilcox CS. Asymmetric dimethylarginine and reactive oxygen species: 85. Agnoletti D, Lieber A, Zhang Y, Protogerou AD, Borghi C, Blacher J, Safar ME.
unwelcome twin visitors to the cardiovascular and kidney disease tables. Central hemodynamic modifications in diabetes mellitus. Atherosclerosis.
Hypertension. 2012;59:375–381. doi: 10.1161/HYPERTENSIONAHA. 2013;230:315–321. doi: 10.1016/j.atherosclerosis.2013.07.054
111.187310 86. Safar ME, Czernichow S, Blacher J. Obesity, arterial stiffness, and cardio-
67. Harrison DG, Chen W, Dikalov S, Li L. Regulation of endothelial cell tetrahy- vascular risk. J Am Soc Nephrol. 2006;17:S109–S111. doi: 10.1681/ASN.
drobiopterin pathophysiological and therapeutic implications. Adv Pharmacol. 2005121321
2010;60:107–132. doi: 10.1016/B978-0-12-385061-4.00005-2 87. Yun M, Li S, Sun D, Ge S, Lai CC, Fernandez C, Chen W, Srinivasan
68. Touyz RM. Oxidative stress and vascular damage in hypertension. Curr SR, Berenson GS. Tobacco smoking strengthens the association of el-
Hypertens Rep. 2000;2:98–105. doi: 10.1007/s11906-000-0066-3 evated blood pressure with arterial stiffness: the Bogalusa Heart Study. J
69. Little TL, Xia J, Duling BR. Dye tracers define differential endothelial Hypertens. 2015;33:266–274. doi: 10.1097/HJH.0000000000000410
and smooth muscle coupling patterns within the arteriolar wall. Circ Res. 88. Vasan RS, Short MI, Niiranen TJ, Xanthakis V, DeCarli C, Cheng S,
1995;76:498–504. doi: 10.1161/01.res.76.3.498 Seshadri S, Mitchell GF. Interrelations between arterial stiffness, target
70. Sandow SL, Hill CE. Incidence of myoendothelial gap junctions in the organ damage, and cardiovascular disease outcomes. J Am Heart Assoc.
proximal and distal mesenteric arteries of the rat is suggestive of a role in 2019;8:e012141. doi: 10.1161/JAHA.119.012141
endothelium-derived hyperpolarizing factor-mediated responses. Circ Res. 89. van Gorp AW, Schenau DS, Hoeks AP, Boudier HA, de Mey JG, Reneman
2000;86:341–346. doi: 10.1161/01.res.86.3.341 RS. In spontaneously hypertensive rats alterations in aortic wall proper-
71. Ledoux J, Taylor MS, Bonev AD, Hannah RM, Solodushko V, Shui B, ties precede development of hypertension. Am J Physiol Heart Circ Physiol.
Tallini Y, Kotlikoff MI, Nelson MT. Functional architecture of inosi- 2000;278:H1241–H1247. doi: 10.1152/ajpheart.2000.278.4.H1241
tol 1,4,5-trisphosphate signaling in restricted spaces of myoendothe- 90. Weisbrod RM, Shiang T, Al Sayah L, Fry JL, Bajpai S, Reinhart-King
lial projections. Proc Natl Acad Sci USA. 2008;105:9627–9632. doi: CA, Lob HE, Santhanam L, Mitchell G, Cohen RA, et al. Arterial stiffen-
10.1073/pnas.0801963105 ing precedes systolic hypertension in diet-induced obesity. Hypertension.
72. Straub AC, Billaud M, Johnstone SR, Best AK, Yemen S, Dwyer ST, 2013;62:1105–1110. doi: 10.1161/HYPERTENSIONAHA.113.01744
Looft-Wilson R, Lysiak JJ, Gaston B, Palmer L, et al. Compartmentalized 91. Wu J, Saleh MA, Kirabo A, Itani HA, Montaniel KR, Xiao L, Chen W,
connexin 43 s-nitrosylation/denitrosylation regulates heterocellular com- Mernaugh RL, Cai H, Bernstein KE, et al. Immune activation caused by
munication in the vessel wall. Arterioscler Thromb Vasc Biol. 2011;31:399– vascular oxidation promotes fibrosis and hypertension. J Clin Invest.
407. doi: 10.1161/ATVBAHA.110.215939 2016;126:50–67. doi: 10.1172/JCI80761
73. Straub AC, Lohman AW, Billaud M, Johnstone SR, Dwyer ST, Lee MY, Bortz 92. Wolinsky H. Response of the rat aortic media to hypertension.
PS, Best AK, Columbus L, Gaston B, et al. Endothelial cell expression of Morphological and chemical studies. Circ Res. 1970;26:507–522. doi:
haemoglobin α regulates nitric oxide signalling. Nature. 2012;491:473–477. 10.1161/01.res.26.4.507
doi: 10.1038/nature11626 93. Humphrey JD, Harrison DG, Figueroa CA, Lacolley P, Laurent S. Central
74. Straub AC, Butcher JT, Billaud M, Mutchler SM, Artamonov MV, Nguyen AT, artery stiffness in hypertension and aging: a problem with cause and con-
Johnson T, Best AK, Miller MP, Palmer LA, et al. Hemoglobin α/eNOS cou- sequence. Circ Res. 2016;118:379–381. doi: 10.1161/CIRCRESAHA.
pling at myoendothelial junctions is required for nitric oxide scavenging dur- 115.307722
ing vasoconstriction. Arterioscler Thromb Vasc Biol. 2014;34:2594–2600. 94. Cooper LL, Rong J, Benjamin EJ, Larson MG, Levy D, Vita JA, Hamburg
doi: 10.1161/ATVBAHA.114.303974 NM, Vasan RS, Mitchell GF. Components of hemodynamic load and cardio-
75. Wilson C, Zhang X, Buckley C, Heathcote HR, Lee MD, McCarron JG. vascular events: the Framingham Heart Study. Circulation. 2015;131:354–
Increased vascular contractility in hypertension results from impaired 361. doi: 10.1161/CIRCULATIONAHA.114.011357
endothelial calcium signaling. Hypertension. 2019;74:1200–1214. doi: 95. Kossmann S, Lagrange J, Jackel S, Jurk K, Ehlken M, Schonfelder T,
Downloaded from http://ahajournals.org by on April 23, 2024

10.1161/HYPERTENSIONAHA.119.13791 Weihert Y, Knorr M, Brandt M, Xia N, et al. Platelet-localized FXI promotes


76. Morton SK, Chaston DJ, Howitt L, Heisler J, Nicholson BJ, Fairweather S, a vascular coagulation-inflammatory circuit in arterial hypertension. Sci
Bröer S, Ashton AW, Matthaei KI, Hill CE. Loss of functional endothelial Transl Med. 2017;9:eaah4923.
connexin40 results in exercise-induced hypertension in mice. Hypertension. 96. Head GA. The sympathetic nervous system in hypertension: assess-
2015;65:662–669. doi: 10.1161/HYPERTENSIONAHA.114.04578 ment by blood pressure variability and ganglionic blockade. J Hypertens.
77. Cao W, Wu L, Zhang X, Zhou J, Wang J, Yang Z, Su H, Liu Y, Wilcox 2003;21:1619–1621. doi: 10.1097/00004872-200309000-00006
CS, Hou FF. Sympathetic overactivity in CKD disrupts buffering of neu- 97. Diedrich A, Jordan J, Tank J, Shannon JR, Robertson R, Luft FC, Robertson
rotransmission by endothelium-derived hyperpolarizing factor and D, Biaggioni I. The sympathetic nervous system in hypertension: assess-
enhances vasoconstriction. J Am Soc Nephrol. 2020;31:2312–2325. doi: ment by blood pressure variability and ganglionic blockade. J Hypertens.
10.1681/ASN.2020030234 2003;21:1677–1686. doi: 10.1097/00004872-200309000-00017
78. Folkow B, Grimby G, Thulesius O. Adaptive structural changes of the vas- 98. Gamboa A, Figueroa R, Paranjape SY, Farley G, Diedrich A, Biaggioni I.
cular walls in hypertension and their relation to the control of the peripheral Autonomic blockade reverses endothelial dysfunction in obesity-associ-
resistance. Acta Physiol Scand. 1958;44:255–272. doi: 10.1111/j.1748- ated hypertension. Hypertension. 2016;68:1004–1010. doi: 10.1161/
1716.1958.tb01626.x HYPERTENSIONAHA.116.07681
79. Lehmann MV, Schmieder RE. Remodeling of retinal small arteries in hyper- 99. DeLalio LJ, Sved AF, Stocker SD. Sympathetic nervous system contribu-
tension. Am J Hypertens. 2011;24:1267–1273. doi: 10.1038/ajh.2011.166 tions to hypertension: updates and therapeutic relevance. Can J Cardiol.
80. Wong TY, Klein R, Sharrett AR, Duncan BB, Couper DJ, Klein BE, Hubbard 2020;36:712–720. doi: 10.1016/j.cjca.2020.03.003
LD, Nieto FJ; Atherosclerosis Risk in Communities Study. Retinal arteriolar 100. Kinsman BJ, Simmonds SS, Browning KN, Stocker SD. Organum vas-
diameter and risk for hypertension. Ann Intern Med. 2004;140:248–255. culosum of the lamina terminalis detects NaCl to elevate sympathetic
doi: 10.7326/0003-4819-140-4-200402170-00006 nerve activity and blood pressure. Hypertension. 2017;69:163–170. doi:
81. Poplin R, Varadarajan AV, Blumer K, Liu Y, McConnell MV, Corrado GS, Peng 10.1161/HYPERTENSIONAHA.116.08372
L, Webster DR. Prediction of cardiovascular risk factors from retinal fun- 101. Xiao L, Itani HA, do Carmo LS, Carver LS, Breyer RM, Harrison DG.
dus photographs via deep learning. Nat Biomed Eng. 2018;2:158–164. doi: Central EP3 (E Prostanoid 3) receptors mediate salt-sensitive hyper-
10.1038/s41551-018-0195-0 tension and immune activation. Hypertension. 2019;74:1507–1515. doi:
82. Heagerty AM, Heerkens EH, Izzard AS. Small artery structure and 10.1161/HYPERTENSIONAHA.119.13850
function in hypertension. J Cell Mol Med. 2010;14:1037–1043. doi: 102. Cao W, Shi M, Wu L, Li J, Yang Z, Liu Y, Wilcox CS, Hou FF. Adipocytes
10.1111/j.1582-4934.2010.01080.x initiate an adipose-cerebral-peripheral sympathetic reflex to induce insulin
83. Li L, Feng D, Luo Z, Welch WJ, Wilcox CS, Lai EY. Remodeling of affer- resistance during high-fat feeding. Clin Sci (Lond). 2019;133:1883–1899.
ent arterioles from mice with oxidative stress does not account for doi: 10.1042/CS20190412
increased contractility but does limit excessive wall stress. Hypertension. 103. Braith RW, Mills RM Jr, Wilcox CS, Convertino VA, Davis GL, Limacher MC,
2015;66:550–556. doi: 10.1161/HYPERTENSIONAHA.115.05631 Wood CE. Fluid homeostasis after heart transplantation: the role of cardiac
84. Vaitkevicius PV, Fleg JL, Engel JH, O’Connor FC, Wright JG, Lakatta denervation. J Heart Lung Transplant. 1996;15:872–880.
LE, Yin FC, Lakatta EG. Effects of age and aerobic capacity on arte- 104. Braith RW, Mills RM Jr, Wilcox CS, Davis GL, Wood CE. Breakdown of blood
rial stiffness in healthy adults. Circulation. 1993;88:1456–1462. doi: pressure and body fluid homeostasis in heart transplant recipients. J Am
10.1161/01.cir.88.4.1456 Coll Cardiol. 1996;27:375–383. doi: 10.1016/0735-1097(95)00467-x

860   April 2, 2021 Circulation Research. 2021;128:847–863. DOI: 10.1161/CIRCRESAHA.121.318082


Harrison et al Pathophysiology of Hypertension

Hypertension Compendium
105. Braith RW, Mills RM, Wilcox CS, Mitchell MJ, Hill JA, Wood CE. High dose 123. Kitiyakara C, Chabrashvili T, Chen Y, Blau J, Karber A, Aslam S, Welch WJ,
angiotensin-converting enzyme inhibition prevents fluid volume expansion Wilcox CS. Salt intake, oxidative stress, and renal expression of NADPH
in heart transplant recipients. J Am Coll Cardiol. 2000;36:487–492. doi: oxidase and superoxide dismutase. J Am Soc Nephrol. 2003;14:2775–
10.1016/s0735-1097(00)00753-1 2782. doi: 10.1097/01.asn.0000092145.90389.65
106. McCurley A, Pires PW, Bender SB, Aronovitz M, Zhao MJ, Metzger D, 124. Welch WJ, Chabrashvili T, Solis G, Chen Y, Gill PS, Aslam S, Wang X, Ji H,
Chambon P, Hill MA, Dorrance AM, Mendelsohn ME, et al. Direct regula- Sandberg K, Jose P, et al. Role of extracellular superoxide dismutase in the
tion of blood pressure by smooth muscle cell mineralocorticoid receptors. mouse angiotensin slow pressor response. Hypertension. 2006;48:934–
Nat Med. 2012;18:1429–1433. doi: 10.1038/nm.2891 941. doi: 10.1161/01.HYP.0000242928.57344.92
107. Jia G, Habibi J, Aroor AR, Martinez-Lemus LA, DeMarco VG, 125. Ye S, Zhong H, Yanamadala S, Campese VM. Oxidative stress medi-
Ramirez-Perez FI, Sun Z, Hayden MR, Meininger GA, Mueller KB, et ates the stimulation of sympathetic nerve activity in the phenol renal
al. Endothelial mineralocorticoid receptor mediates diet-induced aor- injury model of hypertension. Hypertension. 2006;48:309–315. doi:
tic stiffness in females. Circ Res. 2016;118:935–943. doi: 10.1161/ 10.1161/01.HYP.0000231307.69761.2e
CIRCRESAHA.115.308269 126. Jayachandran I, Sundararajan S, Venkatesan S, Paadukaana S,
108. Sun XN, Li C, Liu Y, Du LJ, Zeng MR, Zheng XJ, Zhang WC, Liu Y, Zhu Balasubramanyam M, Mohan V, Manickam N. Asymmetric dimethylarginine
M, Kong D, et al. T-Cell mineralocorticoid receptor controls blood pressure (ADMA) accelerates renal cell fibrosis under high glucose condition through
by regulating interferon-gamma. Circ Res. 2017;120:1584–1597. doi: NOX4/ROS/ERK signaling pathway. Sci Rep. 2020;10:16005. doi: 10.1038/
10.1161/CIRCRESAHA.116.310480 s41598-020-72943-2
109. Bene NC, Alcaide P, Wortis HH, Jaffe IZ. Mineralocorticoid recep- 127. Dikalova AE, Itani HA, Nazarewicz RR, McMaster WG, Flynn CR, Uzhachenko
tors in immune cells: emerging role in cardiovascular disease. Steroids. R, Fessel JP, Gamboa JL, Harrison DG, Dikalov SI. Sirt3 impairment and
2014;91:38–45. doi: 10.1016/j.steroids.2014.04.005 SOD2 hyperacetylation in vascular oxidative stress and hypertension. Circ
110. Brown JM, Siddiqui M, Calhoun DA, Carey RM, Hopkins PN, Williams Res. 2017;121:564–574. doi: 10.1161/CIRCRESAHA.117.310933
GH, Vaidya A. The unrecognized prevalence of primary aldosteron- 128. Irazabal MV, Torres VE. Reactive oxygen species and redox signaling in
ism: a cross-sectional study. Ann Intern Med. 2020;173:10–20. doi: chronic kidney disease. Cells. 2020;9:1342.
10.7326/M20-0065 129. Norlander AE, Saleh MA, Kamat NV, Ko B, Gnecco J, Zhu L, Dale BL,
111. Schnackenberg CG, Welch WJ, Wilcox CS. Normalization of blood pressure Iwakura Y, Hoover RS, McDonough AA, et al. Interleukin-17A regu-
and renal vascular resistance in SHR with a membrane-permeable super- lates renal sodium transporters and renal injury in angiotensin II-
oxide dismutase mimetic: role of nitric oxide. Hypertension. 1998;32:59– induced hypertension. Hypertension. 2016;68:167–174. doi: 10.1161/
64. doi: 10.1161/01.hyp.32.1.59 HYPERTENSIONAHA.116.07493
112. Schnackenberg CG, Wilcox CS. Two-week administration of tempol attenu- 130. Madhur MS, Lob HE, McCann LA, Iwakura Y, Blinder Y, Guzik TJ,
ates both hypertension and renal excretion of 8-Iso prostaglandin f2alpha. Harrison DG. Interleukin 17 promotes angiotensin II-induced hyperten-
Hypertension. 1999;33:424–428. doi: 10.1161/01.hyp.33.1.424 sion and vascular dysfunction. Hypertension. 2010;55:500–507. doi:
113. Laursen JB, Rajagopalan S, Galis Z, Tarpey M, Freeman BA, Harrison 10.1161/HYPERTENSIONAHA.109.145094
DG. Role of superoxide in angiotensin II-induced but not catecholamine- 131. Nguyen H, Chiasson VL, Chatterjee P, Kopriva SE, Young KJ, Mitchell
induced hypertension. Circulation. 1997;95:588–593. doi: 10.1161/01 BM. Interleukin-17 causes Rho-kinase-mediated endothelial dys-
.cir.95.3.588 function and hypertension. Cardiovasc Res. 2013;97:696–704. doi:
114. Rajagopalan S, Kurz S, Münzel T, Tarpey M, Freeman BA, Griendling KK, 10.1093/cvr/cvs422
Harrison DG. Angiotensin II-mediated hypertension in the rat increases 132. Wu J, Thabet SR, Kirabo A, Trott DW, Saleh MA, Xiao L, Madhur MS,
vascular superoxide production via membrane NADH/NADPH oxidase Chen W, Harrison DG. Inflammation and mechanical stretch pro-
Downloaded from http://ahajournals.org by on April 23, 2024

activation. Contribution to alterations of vasomotor tone. J Clin Invest. mote aortic stiffening in hypertension through activation of p38 mi-
1996;97:1916–1923. doi: 10.1172/JCI118623 togen-activated protein kinase. Circ Res. 2014;114:616–625. doi:
115. Laursen JB, Somers M, Kurz S, McCann L, Warnholtz A, Freeman BA, 10.1161/CIRCRESAHA.114.302157
Tarpey M, Fukai T, Harrison DG. Endothelial regulation of vasomotion in 133. Kassan M, Galan M, Partyka M, Trebak M, Matrougui K. Interleukin-10
apoE-deficient mice: implications for interactions between peroxyni- released by CD4(+)CD25(+) natural regulatory T cells improves micro-
trite and tetrahydrobiopterin. Circulation. 2001;103:1282–1288. doi: vascular endothelial function through inhibition of NADPH oxidase activity
10.1161/01.cir.103.9.1282 in hypertensive mice. Arterioscler Thromb Vasc Biol. 2011;31:2534–2542.
116. Somers MJ, Mavromatis K, Galis ZS, Harrison DG. Vascular superoxide doi: 10.1161/ATVBAHA.111.233262
production and vasomotor function in hypertension induced by deoxy- 134. Matrougui K, Abd Elmageed Z, Zakaria AE, Kassan M, Choi S, Nair D,
corticosterone acetate-salt. Circulation. 2000;101:1722–1728. doi: Gonzalez-Villalobos RA, Chentoufi AA, Kadowitz P, Belmadani S, et al.
10.1161/01.cir.101.14.1722 Natural regulatory T cells control coronary arteriolar endothelial dys-
117. Araujo M, Wilcox CS. Oxidative stress in hypertension: role of the kidney. function in hypertensive mice. Am J Pathol. 2011;178:434–441. doi:
Antioxid Redox Signal. 2014;20:74–101. doi: 10.1089/ars.2013.5259 10.1016/j.ajpath.2010.11.034
118. Rajagopalan S, Meng XP, Ramasamy S, Harrison DG, Galis ZS. Reactive 135. Kirabo A, Fontana V, de Faria AP, Loperena R, Galindo CL, Wu J, Bikineyeva
oxygen species produced by macrophage-derived foam cells regulate AT, Dikalov S, Xiao L, Chen W, et al. DC isoketal-modified proteins activate
the activity of vascular matrix metalloproteinases in vitro. Implications for T cells and promote hypertension. J Clin Invest. 2014;124:4642–4656.
atherosclerotic plaque stability. J Clin Invest. 1996;98:2572–2579. doi: doi: 10.1172/JCI74084
10.1172/JCI119076 136. Marvar PJ, Thabet SR, Guzik TJ, Lob HE, McCann LA, Weyand C, Gordon
119. Lob HE, Schultz D, Marvar PJ, Davisson RL, Harrison DG. Role of FJ, Harrison DG. Central and peripheral mechanisms of T-lymphocyte
the NADPH oxidases in the subfornical organ in angiotensin II- activation and vascular inflammation produced by angiotensin II-
induced hypertension. Hypertension. 2013;61:382–387. doi: 10.1161/ induced hypertension. Circ Res. 2010;107:263–270. doi: 10.1161/
HYPERTENSIONAHA.111.00546 CIRCRESAHA.110.217299
120. Wang HW, Huang BS, White RA, Chen A, Ahmad M, Leenen FH. 137. Lob HE, Marvar PJ, Guzik TJ, Sharma S, McCann LA, Weyand C, Gordon
Mineralocorticoid and angiotensin II type 1 receptors in the subfornical FJ, Harrison DG. Induction of hypertension and peripheral inflammation
organ mediate angiotensin II - induced hypothalamic reactive oxygen spe- by reduction of extracellular superoxide dismutase in the central ner-
cies and hypertension. Neuroscience. 2016;329:112–121. doi: 10.1016/j. vous system. Hypertension. 2010;55:277–83, 6p following 283. doi:
neuroscience.2016.04.050 10.1161/HYPERTENSIONAHA.109.142646
121. Carvalho-Galvão A, Guimarães DD, De Brito Alves JL, Braga VA. 138. Xiao L, do Carmo LS, Foss JD, Chen W, Harrison DG. Sympathetic en-
Central inhibition of tumor necrosis factor alpha reduces hyperten- hancement of memory T-cell homing and hypertension sensitization. Circ
sion by attenuating oxidative stress in the rostral ventrolateral medulla Res. 2020;126:708–721. doi: 10.1161/CIRCRESAHA.119.314758
in renovascular hypertensive rats. Front Physiol. 2019;10:491. doi: 139. Guzik TJ, Skiba DS, Touyz RM, Harrison DG. The role of infiltrating immune
10.3389/fphys.2019.00491 cells in dysfunctional adipose tissue. Cardiovasc Res. 2017;113:1009–
122. Griendling KK, Minieri CA, Ollerenshaw JD, Alexander RW. Angiotensin 1023. doi: 10.1093/cvr/cvx108
II stimulates NADH and NADPH oxidase activity in cultured vascular 140. Mikolajczyk TP, Nosalski R, Szczepaniak P, Budzyn K, Osmenda G, Skiba
smooth muscle cells. Circ Res. 1994;74:1141–1148. doi: 10.1161/01. D, Sagan A, Wu J, Vinh A, Marvar PJ, et al. Role of chemokine RANTES
res.74.6.1141 in the regulation of perivascular inflammation, T-cell accumulation, and

Circulation Research. 2021;128:847–863. DOI: 10.1161/CIRCRESAHA.121.318082 April 2, 2021   861


Harrison et al Pathophysiology of Hypertension
Hypertension Compendium

vascular dysfunction in hypertension. FASEB J. 2016;30:1987–1999. doi: inhibition of vasodilatation by asymmetrical dimethylarginine. Hypertension.
10.1096/fj.201500088R 2011;58:380–385. doi: 10.1161/HYPERTENSIONAHA.111.170175
141. Tracey KJ. The inflammatory reflex. Nature. 2002;420:853–859. doi: 159. Campese VM, Romoff MS, Levitan D, Saglikes Y, Friedler RM, Massry SG.
10.1038/nature01321 Abnormal relationship between sodium intake and sympathetic nervous
142. Harwani SC, Chapleau MW, Legge KL, Ballas ZK, Abboud FM. system activity in salt-sensitive patients with essential hypertension. Kidney
Neurohormonal modulation of the innate immune system is proinflam- Int. 1982;21:371–378. doi: 10.1038/ki.1982.32
matory in the prehypertensive spontaneously hypertensive rat, a genetic 160. Machnik A, Neuhofer W, Jantsch J, Dahlmann A, Tammela T, Machura K,
model of essential hypertension. Circ Res. 2012;111:1190–1197. doi: Park JK, Beck FX, Müller DN, Derer W, et al. Macrophages regulate salt-
10.1161/CIRCRESAHA.112.277475 dependent volume and blood pressure by a vascular endothelial growth
143. Jakubzick C, Gautier EL, Gibbings SL, Sojka DK, Schlitzer A, Johnson factor-C-dependent buffering mechanism. Nat Med. 2009;15:545–552.
TE, Ivanov S, Duan Q, Bala S, Condon T, et al. Minimal differentiation of doi: 10.1038/nm.1960
classical monocytes as they survey steady-state tissues and transport 161. Kopp C, Linz P, Wachsmuth L, Dahlmann A, Horbach T, Schöfl C, Renz
antigen to lymph nodes. Immunity. 2013;39:599–610. doi: 10.1016/j. W, Santoro D, Niendorf T, Müller DN, et al. (23)Na magnetic reso-
immuni.2013.08.007 nance imaging of tissue sodium. Hypertension. 2012;59:167–172. doi:
144. Randolph GJ, Beaulieu S, Lebecque S, Steinman RM, Muller WA. 10.1161/HYPERTENSIONAHA.111.183517
Differentiation of monocytes into dendritic cells in a model of transen- 162. Kopp C, Linz P, Dahlmann A, Hammon M, Jantsch J, Müller DN,
dothelial trafficking. Science. 1998;282:480–483. doi: 10.1126/science. Schmieder RE, Cavallaro A, Eckardt KU, Uder M, et al. 23Na magnetic
282.5388.480 resonance imaging-determined tissue sodium in healthy subjects and
145. Loperena R, Van Beusecum JP, Itani HA, Engel N, Laroumanie F, Xiao hypertensive patients. Hypertension. 2013;61:635–640. doi: 10.1161/
L, Elijovich F, Laffer CL, Gnecco JS, Noonan J, et al. Hypertension and HYPERTENSIONAHA.111.00566
increased endothelial mechanical stretch promote monocyte differentia- 163. Jantsch J, Schatz V, Friedrich D, Schröder A, Kopp C, Siegert I, Maronna
tion and activation: roles of STAT3, interleukin 6 and hydrogen peroxide. A, Wendelborn D, Linz P, Binger KJ, et al. Cutaneous Na+ storage
Cardiovasc Res. 2018;114:1547–1563. doi: 10.1093/cvr/cvy112 strengthens the antimicrobial barrier function of the skin and boosts
146. Barbaro NR, Van Beusecum J, Xiao L, do Carmo L, Pitzer A, Loperena macrophage-driven host defense. Cell Metab. 2015;21:493–501. doi:
R, Foss JD, Elijovich F, Laffer CL, Montaniel KR, et al. Sodium activates 10.1016/j.cmet.2015.02.003
human monocytes via the NADPH oxidase and isolevuglandin formation 164. Kitada K, Daub S, Zhang Y, Klein JD, Nakano D, Pedchenko T, Lantier L,
Accessed March 14, 2021. https://academic.oup.com/cardiovascres/ LaRocque LM, Marton A, Neubert P, et al. High salt intake reprioritizes
advance-article/doi/10.1093/cvr/cvaa207/5872525. osmolyte and energy metabolism for body fluid conservation. J Clin Invest.
147. Lifton RP, Gharavi AG, Geller DS. Molecular mechanisms of human hyper- 2017;127:1944–1959. doi: 10.1172/JCI88532
tension. Cell. 2001;104:545–556. doi: 10.1016/s0092-8674(01)00241-0 165. Wu C, Yosef N, Thalhamer T, Zhu C, Xiao S, Kishi Y, Regev A, Kuchroo
148. Cabrera CP, Ng FL, Nicholls HL, Gupta A, Barnes MR, Munroe PB, Caulfield VK. Induction of pathogenic TH17 cells by inducible salt-sensing kinase
MJ. Over 1000 genetic loci influencing blood pressure with multiple sys- SGK1. Nature. 2013;496:513–517. doi: 10.1038/nature11984
tems and tissues implicated. Hum Mol Genet. 2019;28:R151–R161. doi: 166. Kleinewietfeld M, Manzel A, Titze J, Kvakan H, Yosef N, Linker RA, Muller
10.1093/hmg/ddz197 DN, Hafler DA. Sodium chloride drives autoimmune disease by the in-
149. Vauraa F, Kaukoa a, Suvilaa K, Havulinnac A, Marsd N, Salomaac V, duction of pathogenic TH17 cells. Nature. 2013;496:518–522. doi:
Chenge S, Niiranena T. Polygenic risk scores predict hypertension onset 10.1038/nature11868
and cardiovascular risk. Hypertension. 2021;77:1119–1127. doi:10.1161/ 167. Barbaro NR, Foss JD, Kryshtal DO, Tsyba N, Kumaresan S, Xiao L,
HYPERTENSIONAHA.120.16471 Mernaugh RL, Itani HA, Loperena R, Chen W, et al. Dendritic cell amiloride-
Downloaded from http://ahajournals.org by on April 23, 2024

150. He FJ, MacGregor GA. A comprehensive review on salt and health and cur- sensitive channels mediate sodium-induced inflammation and hypertension.
rent experience of worldwide salt reduction programmes. J Hum Hypertens. Cell Rep. 2017;21:1009–1020. doi: 10.1016/j.celrep.2017.10.002
2009;23:363–384. doi: 10.1038/jhh.2008.144 168. Norlander AE, Saleh MA, Pandey AK, Itani HA, Wu J, Xiao L, Kang J, Dale
151. He FJ, Li J, Macgregor GA. Effect of longer term modest salt reduction BL, Goleva SB, Laroumanie F, et al. A salt-sensing kinase in T lympho-
on blood pressure: cochrane systematic review and meta-analysis of ran- cytes, SGK1, drives hypertension and hypertensive end-organ damage. JCI
domised trials. BMJ. 2013;346:f1325. doi: 10.1136/bmj.f1325 Insight. 2017;2:e92801.
152. ALLHAT Officers and Coordinators for the ALLHAT Collaborative Research 169. Van Beusecum JP, Barbaro NR, McDowell Z, Aden LA, Xiao L, Pandey AK,
Group. The Antihypertensive and Lipid-Lowering Treatment to Prevent Itani HA, Himmel LE, Harrison DG, Kirabo A. High salt activates CD11c+
Heart Attack Trial. Major outcomes in high-risk hypertensive patients ran- antigen-presenting cells via SGK (Serum Glucocorticoid Kinase) 1 to
domized to angiotensin-converting enzyme inhibitor or calcium channel promote renal inflammation and salt-sensitive hypertension. Hypertension.
blocker vs diuretic: the Antihypertensive and Lipid-Lowering Treatment to 2019;74:555–563. doi: 10.1161/HYPERTENSIONAHA.119.12761
Prevent Heart Attack Trial (ALLHAT). JAMA. 2002;288:2981–2997. doi: 170. Wilck N, Matus MG, Kearney SM, Olesen SW, Forslund K, Bartolomaeus
10.1001/jama.288.23.2981 H, Haase S, Mähler A, Balogh A, Markó L, et al. Salt-responsive gut com-
153. Elijovich F, Weinberger MH, Anderson CA, Appel LJ, Bursztyn M, Cook mensal modulates TH17 axis and disease. Nature. 2017;551:585–589.
NR, Dart RA, Newton-Cheh CH, Sacks FM, Laffer CL; American Heart doi: 10.1038/nature24628
Association Professional and Public Education Committee of the Council 171. Marques FZ, Mackay CR, Kaye DM. Beyond gut feelings: how the gut mi-
on Hypertension; Council on Functional Genomics and Translational crobiota regulates blood pressure. Nat Rev Cardiol. 2018;15:20–32. doi:
Biology; and Stroke Council. Salt sensitivity of blood pressure: a sci- 10.1038/nrcardio.2017.120
entific statement from the American Heart Association. Hypertension. 172. Thornton AM, Shevach EM. CD4+CD25+ immunoregulatory T cells sup-
2016;68:e7–e46. doi: 10.1161/HYP.0000000000000047 press polyclonal T cell activation in vitro by inhibiting interleukin 2 produc-
154. Weinberger MH. Salt sensitivity of blood pressure in humans. Hypertension. tion. J Exp Med. 1998;188:287–296. doi: 10.1084/jem.188.2.287
1996;27:481–490. doi: 10.1161/01.hyp.27.3.481 173. Raizada MK, Joe B, Bryan NS, Chang EB, Dewhirst FE, Borisy GG, Galis
155. Choi HY, Park HC, Ha SK. Salt sensitivity and hypertension: a para- ZS, Henderson W, Jose PA, Ketchum CJ, et al. Report of the National
digm shift from kidney malfunction to vascular endothelial dysfunction. Heart, Lung, and Blood Institute Working Group on the Role of microbiota
Electrolyte Blood Press. 2015;13:7–16. doi: 10.5049/EBP.2015.13.1.7 in blood pressure regulation: current status and future directions. https://
156. Morris RC Jr, Schmidlin O, Sebastian A, Tanaka M, Kurtz TW. www.ahajournals.org/doi/10.1161/HYPERTENSIONAHA.117.09699
Vasodysfunction that involves renal vasodysfunction, not abnormally 174. Tang WHW, Li DY, Hazen SL. Dietary metabolism, the gut microbiome,
increased renal retention of sodium, accounts for the initiation of salt- and heart failure. Nat Rev Cardiol. 2019;16:137–154. doi: 10.1038/
induced hypertension. Circulation. 2016;133:881–893. doi: 10.1161/ s41569-018-0108-7
CIRCULATIONAHA.115.017923 175. Forsburg SL, Guarente L. Communication between mitochondria and the
157. Laffer CL, Scott RC III, Titze JM, Luft FC, Elijovich F. Hemodynamics nucleus in regulation of cytochrome genes in the yeast Saccharomyces
and salt-and-water balance link sodium storage and vascular dysfunc- cerevisiae. Annu Rev Cell Biol. 1989;5:153–180. doi: 10.1146/
tion in salt-sensitive subjects. Hypertension. 2016;68:195–203. doi: annurev.cb.05.110189.001101
10.1161/HYPERTENSIONAHA.116.07289 176. Human Microbiome Project Consortium. Structure, function and diver-
158. Schmidlin O, Forman A, Leone A, Sebastian A, Morris RC Jr. Salt sen- sity of the healthy human microbiome. Nature. 2012;486:207–214. doi:
sitivity in blacks: evidence that the initial pressor effect of NaCl involves 10.1038/nature11234

862   April 2, 2021 Circulation Research. 2021;128:847–863. DOI: 10.1161/CIRCRESAHA.121.318082


Harrison et al Pathophysiology of Hypertension

Hypertension Compendium
177. Li J, Zhao F, Wang Y, Chen J, Tao J, Tian G, Wu S, Liu W, Cui Q, pathophysiological alterations in the gut. Circ Res. 2017;120:312–323.
Geng B, et al. Gut microbiota dysbiosis contributes to the development doi: 10.1161/CIRCRESAHA.116.309006
of hypertension. Microbiome. 2017;5:14. doi: 10.1186/s40168- 183. Sacks FM, Svetkey LP, Vollmer WM, Appel LJ, Bray GA, Harsha D,
016-0222-x Obarzanek E, Conlin PR, Miller ER III, Simons-Morton DG, et al; DASH-
178. Ferguson JF, Aden LA, Barbaro NR, Van Beusecum JP, Xiao L, Simmons Sodium Collaborative Research Group. Effects on blood pressure of re-
AJ, Warden C, Pasic L, Himmel LE, Washington MK, et al. High dietary salt- duced dietary sodium and the Dietary Approaches to Stop Hypertension
induced dendritic cell activation underlies microbial dysbiosis-associated (DASH) diet. DASH-Sodium Collaborative Research Group. N Engl J Med.
hypertension. JCI Insight. 2019;5:e126241. 2001;344:3–10. doi: 10.1056/NEJM200101043440101
179. Mell B, Jala VR, Mathew AV, Byun J, Waghulde H, Zhang Y, Haribabu B, 184. David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE,
Vijay-Kumar M, Pennathur S, Joe B. Evidence for a link between gut micro- Ling AV, Devlin AS, Varma Y, Fischbach MA, et al. Diet rapidly and repro-
biota and hypertension in the Dahl rat. Physiol Genomics. 2015;47:187– ducibly alters the human gut microbiome. Nature. 2014;505:559–563. doi:
197. doi: 10.1152/physiolgenomics.00136.2014 10.1038/nature12820
180. Yang T, Santisteban MM, Rodriguez V, Li E, Ahmari N, Carvajal JM, Zadeh 185. Maslowski KM, Vieira AT, Ng A, Kranich J, Sierro F, Yu D, Schilter HC,
M, Gong M, Qi Y, Zubcevic J, et al. Gut dysbiosis is linked to hypertension. Rolph MS, Mackay F, Artis D, et al. Regulation of inflammatory respons-
Hypertension. 2015;65:1331–1340. doi: 10.1161/HYPERTENSIONAHA. es by gut microbiota and chemoattractant receptor GPR43. Nature.
115.05315 2009;461:1282–1286. doi: 10.1038/nature08530
181. Marques FZ, Nelson E, Chu PY, Horlock D, Fiedler A, Ziemann 186. Koh A, De Vadder F, Kovatcheva-Datchary P, Bäckhed F. From dietary fiber
M, Tan JK, Kuruppu S, Rajapakse NW, El-Osta A, et al. High- to host physiology: short-chain fatty acids as key bacterial metabolites. Cell.
fiber diet and acetate supplementation change the gut microbiota 2016;165:1332–1345. doi: 10.1016/j.cell.2016.05.041
and prevent the development of hypertension and heart failure in 187. Pluznick JL, Protzko RJ, Gevorgyan H, Peterlin Z, Sipos A, Han J, Brunet
hypertensive mice. Circulation. 2017;135:964–977. doi: 10.1161/ I, Wan LX, Rey F, Wang T, et al. Olfactory receptor responding to gut mi-
CIRCULATIONAHA.116.024545 crobiota-derived signals plays a role in renin secretion and blood pres-
182. Santisteban MM, Qi Y, Zubcevic J, Kim S, Yang T, Shenoy V, Cole-Jeffrey sure regulation. Proc Natl Acad Sci USA. 2013;110:4410–4415. doi:
CT, Lobaton GO, Stewart DC, Rubiano A, et al. Hypertension-linked 10.1073/pnas.1215927110
Downloaded from http://ahajournals.org by on April 23, 2024

Circulation Research. 2021;128:847–863. DOI: 10.1161/CIRCRESAHA.121.318082 April 2, 2021   863

You might also like