Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Brief Review

Myocardial Remodeling in Hypertension


Toward a New View of Hypertensive Heart Disease
Arantxa González, Susana Ravassa, Begoña López, María U. Moreno, Javier Beaumont,
Gorka San José, Ramón Querejeta, Antoni Bayés-Genís, Javier Díez

A rterial hypertension has been related to multiple out-


comes, including cardiac, cerebral, and renal.1 The burden
of arterial hypertension remains high, despite the availability of
Mechanisms and Consequences of MR in HHD
MR is a complex process driven by the responses of cardiomy-
ocytes, other resident cells of the myocardium (ie, fibroblasts,
preventive interventions and low-cost, effective antihyperten- endothelial cells, pericytes, and immune cells), and cells
sive medications.2 Therefore, to hypertension burden, beyond recruited from the circulation (eg, immune and inflammatory
new effective health strategies, novel pathophysiological and cells and progenitor cells) to a variety of dynamic stimuli,
clinical approaches are also required. including mechanical and nonmechanical stimuli, present in
Hypertension can cause or is related to various cardiac conditions of cardiac injury (Figure 1).9 As a consequence,
manifestations, including hypertensive heart disease (HHD) the volume, composition, and biophysiology of cardiomyo-
and obstructive coronary heart disease. HHD is defined by cytes, the interstitial space, and the coronary microvascula-
the presence of left ventricular (LV) hypertrophy (LVH) or ture develop a variety of interrelated alterations, which have a
LV systolic and diastolic dysfunction and their clinical mani- detrimental impact on both cardiac function and clinical out-
festations, such as arrhythmias and symptomatic heart failure comes of patients with HHD (Figure 1).9 Therefore, HHD is
(HF), appearing in patients with hypertension.3 The classical not just a matter of LVH but the result of complex myocardial,
view of HHD sustains that in conditions of pressure overload
cellular, and tissue arrangements leading to changes in shape
because of systemic hypertension and no other cardiac condi-
or size and function of the LV and other cardiac chambers.10
tions, the LV undergoes extensive growth, leading to LVH, in
an attempt to maintain cardiac output, despite the increased Alterations of Cardiomyocytes
afterload imposed by systemic hypertension.4 However, many
patients with high blood pressures do not present clinically Hypertrophy
Downloaded from http://ahajournals.org by on September 8, 2021

detectable LVH. Therefore, a new view of HHD is emerging, The hypertrophic growth of cardiomyocytes is the primary
sustaining that long-term exposure to the hemodynamic stress response by which the heart reduces the stress on the LV wall
imposed by hypertension, in combination with the influence imposed by pressure overload. It entails stimulation of intra-
of other factors, including comorbidities (eg, obesity, diabetes cellular signaling cascades that activate gene expression and
mellitus, and chronic kidney disease), sex, age, environmental promotes protein synthesis, protein stability, or both, with
exposures, and genetic factors, eventually leads to LV dys- consequent increases in protein content and in the size and
function and HF, as well as disturbances of the cardiac rhythm organization of force-generating units (sarcomeres), which, in
and the myocardial perfusion.5,6 This maladaptive change is turn, lead to increased size of individual cardiomyocytes. In
likely because of alterations in the structure and the function many cases, but not necessarily in all, the magnitude of the
of the myocardium that result in its remodeling (Figure 1).7,8 cardiomyocyte growth will determine that LV mass increases
This article reviews the major pathological components and, thus, LVH develops.11 Moreover, there are changes in sar-
of myocardial remodeling (MR) in HHD, highlighting their comeric proteins that regulate passive cardiomyocyte stiffness
main mechanisms and their impact on cardiac function and the (eg, titin).12
patient’s clinical outcome. In addition, we discuss the poten- The mechanisms whereby the mechanical stretch of
tial of circulating and imaging biomarkers to recognize diverse cardiomyocytes is transduced across the cell membrane are
phenotypes of MR and track their evolution and review cur- unclear. They probably involve stretch-sensitive ion chan-
rent and future therapies that may allow personalized HHD nels, a Na+/H+ exchanger, integrins and integrin-interacting
management targeting MR. molecules, and other internal and membrane-bound stretch

From the Program of Cardiovascular Diseases, Center for Applied Medical Research, University of Navarra, Pamplona, Spain (A.G., S.R., B.L., M.U.M.,
J.B., G.S.J., J.D.); Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona, Spain (A.G., S.R., B.L., M.U.M., J.B., G.S.J., J.D.); CIBERCV,
Carlos III Institute of Health, Madrid, Spain (A.G., S.R., B.L., M.U.M., J.B., G.S.J., A.B.-G., J.D.); Division of Cardiology, Donostia University Hospital,
University of the Basque Country, San Sebastián, Spain (R.Q.); Heart Failure Unit and Cardiology Service, Hospital Universitari Germans Trias i Pujol,
Badalona, Spain (A.B.-G.); Department of Medicine, Universitat Autònoma de Barcelona, Spain (A.B.-G.); and Department of Cardiology and Cardiac
Surgery (J.D.) and Department of Nephrology (J.D.), University of Navarra Clinic, University of Navarra, Pamplona, Spain.
The online-only Data Supplement is available with this article at https://www.ahajournals.org/doi/suppl/10.1161/HYPERTENSIONAHA.118.11125.
Correspondence to Javier Díez, Program of Cardiovascular Diseases, Center for Applied Medical Research, University of Navarra, Ave Pío XII, 55,
31008 Pamplona, Spain. Email jadimar@unav.es
(Hypertension. 2018;72:549-558. DOI: 10.1161/HYPERTENSIONAHA.118.11125.)
© 2018 American Heart Association, Inc.
Hypertension is available at https://www.ahajournals.org/journal/hyp DOI: 10.1161/HYPERTENSIONAHA.118.11125

549
550  Hypertension  September 2018

Figure 1.  Pathophysiological continuum of hypertensive heart disease, which describes the progressive process at cardiac and organ tissue levels that
manifest as clinical disease in hypertensive patients. BP indicates blood pressure; CFR, coronary flow reserve; CV, cardiovascular; and LVH, left ventricular
hypertrophy.

sensors in a complex network that links the extracellular Death


matrix (ECM), the cytoskeleton, the sarcomere, Ca2+-handling The hypertrophic response of the cardiomyocyte may be linked
proteins, and the nucleus.13 Local humoral mechanisms acti- to its death. The dysregulation of protein synthesis/processing
Downloaded from http://ahajournals.org by on September 8, 2021

vated in conditions of pressure overload, including neuro- during the hypertrophic process occurs within the endoplasmic
hormones as catecholamines14 and angiotensin II,15 as well reticulum and, when persistent, causes the accumulation of
as growth factors and cytokines released by noncardiomyo- unfolded proteins, thereby leading to endoplasmic reticulum
cytes, such as fibroblasts, vascular cells, and blood cells,16 stress and activation of the unfolded protein response, which,
can also play a role in activating gene expression and pro- in turn, may induce cardiomyocyte apoptosis.22 For instance,
moting hypertrophy of the cardiomyocyte through specific in cardiomyocytes isolated from spontaneously hypertensive
membrane receptors, although their quantitative importance rats, which present LVH and dysfunction, an association was
has not been determined. Because of the inter-relationships found between the activation of unfolded protein response and
of the cardiomyocyte responses to hypertrophic stimuli, they stimulation of apoptosis.23 Additionally, clinical evidence sug-
are likely to implicate common mediators. For instance, it has gests that in the hypertensive myocardium, there is a deficient
been recently reported that MRTF (myocardin-related tran- activity of the factors that prevent cardiomyocyte apoptosis
scription factor)-A mediates both mechanical stretch- and (eg, gp130/leukemia inhibitory factor receptor survival path-
neurohormonal stimulation-induced gene and hypertrophic way).24 Therefore, cardiomyocyte apoptosis is abnormally
responses in cardiomyocytes.17 stimulated in patients with HHD, namely in those with HF
Experimental evidence supports the notion that the genetic with reduced ejection fraction (Figure 2).25
reprogramming associated with cardiomyocyte hypertrophy The apoptosis of cardiomyocytes may contribute to the
may no longer be considered as an adaptive process.18 In fact, development of LV dysfunction or failure of the hypertensive
the genetic changes that accompany cardiomyocyte hyper- myocardium through 3 different pathways. First, it has been
trophy translate into derangements in energy metabolism, reported that the loss of cardiomyocytes caused by apoptosis
contractile cycle and excitation-contraction coupling, cyto- increases in parallel with the deterioration of systolic function
skeleton and membrane properties. These changes determine in spontaneously hypertensive rats,26 suggesting that apoptosis
mechanical dysfunction, which, in turn, provides the basis for may serve as one mechanism involved in the loss of contrac-
the cardiomyocyte malfunction, which is associated with LVH tile mass and function in patients with HHD. Second, some
and predisposes the LV to diastolic or systolic dysfunction. mechanisms that are activated during the apoptotic process
For instance, the analysis of experimental findings suggests may also interfere with the function of viable cardiomyo-
that the hypertrophic growth of the cardiomyocyte is associ- cytes before death.27 Caspase-3 cleaves cardiac myofibrillar
ated with metabolic changes that result in reduced fatty acid proteins, resulting in an impaired force/Ca2+ relationship and
oxidation and increased glucose use, the dysfunction of the myofibrillar ATPase activity. In addition, the release of cyto-
mitochondrial electron transport chain, and the subsequent chrome C from mitochondria during apoptosis may impair
diminution in ATP production.19–21 oxidative phosphorylation and ATP production, thus leading
González et al   Myocardial Remodeling in Hypertension   551

Figure 2.  Distribution of the cardiomyocyte


apoptotic index in normotensive subjects and
in hypertensive patients classified according
to stages of heart failure (HF). Box plots show
the 5th and 95th (vertical lines), 25th and 75th
(boxes), and 50th (horizontal line) percentile
values. Adapted from Ravassa et al25 with
permission. Copyright © 2007, The Author.

to energetic compromise and functional impairment. Third, release damage-associated molecular pattern molecules, DNA
in addition to contributing to histological remodeling of the fragments, heat shock proteins, and matricellular proteins,
myocardium, cardiomyocyte apoptosis may also contribute instructing surrounding healthy cardiomyocytes to produce
to geometric remodeling of the LV chamber. In fact, severe inflammatory mediators (eg, IL [interleukin]-1β, IL-6, macro-
cardiomyocyte apoptosis may lead to side-to-side slippage phage chemoattractant protein-1, and TNF-α [tumor necrosis
of cells, mural thinning, and chamber dilatation. Thus, wall factor α]), which in turn activate versatile signaling networks
restructuring secondary to severe cardiomyocyte apoptosis within surviving cardiomyocytes and trigger leukocyte activa-
Downloaded from http://ahajournals.org by on September 8, 2021

may create an irreversible state of the myocardium, condition- tion and recruitment.
ing progressive dilatation and the continuous deterioration of Recently published studies suggested that the myocardial
LV hemodynamics and performance with time.28 proinflammatory cytokine milieu, as well as changes in the
Although apoptosis is characteristic of HHD, it is impor- composition of the ECM, are crucial in the differentiation of
tant to point out that in the response to any given cardiac injury, resident cardiac fibroblasts to activated myofibroblasts, which
various modalities of cell death, including apoptosis, necrosis, initiate the process leading to myocardial fibrosis and its
and autophagy, are stimulated because they are interconnected detrimental consequences.40 Accordingly, experimental find-
by common cellular pathways at multiple points.29 In fact, ings support a substantial role of inflammation in myocardial
autophagy is activated during hypertensive LVH, serving to fibrosis and diastolic dysfunction in hypertensive hearts.41,42
maintain cellular homeostasis.30 Excessive autophagy, how- Additionally, a positive correlation between myocardial fibro-
ever, eliminates essential cellular elements and possibly pro- sis, as well as the amount of myocardial inflammatory cells,
vokes cardiomyocyte death, which further contributes to MR. and passive myocardial stiffness and diastolic dysfunction
was reported in patients (mostly hypertensive) with HF with
Alterations of the Interstitial Space preserved ejection fraction (HFpEF).43
Inflammation The role of systemic inflammation in MR in hypertension
Recent advances clearly show that inflammation and activa- also deserves to be considered. Evidence from experimental
tion of immunity are central drivers in the pathogenesis of models of hypertension and hypertensive patients suggests an
hypertension-induced target organ damage.31–33 For instance, imbalance of T effector and regulatory subsets of lymphocytes
experimental evidence supports a substantial involvement of in hypertension, causing low-grade inflammation and contrib-
the inflammatory component in hypertensive animal model- uting to blood pressure elevation and progression of end-organ
dependent cardiac damage, particularly in response to admin- damage.44 It has been demonstrated in numerous clinical trials
istration of exogenous angiotensin II,34 aldosterone35 and other that hypertensive patients commonly have increased plasma
mineralocorticoids,36 stimulation of the sympathetic nervous concentrations of C-reactive protein45 and proinflammatory
system,37 and induction of pressure overload.38 These models cytokines, such as IL-6, IL-1β, and TNF-α.46,47 In addition,
are associated in a variable degree with increased permeability circulating levels of CXCR3 (C-X-C chemokine receptor
of the capillary wall, induction of cytokines and chemokines, type 3) chemokines, which induce T-cell tissue homing, have
and recruitment of inflammatory cells that will infiltrate the been reported to be elevated in hypertensive patients.48 On the
myocardium. More recently, cardiomyocytes have emerged other hand, inverse correlations have been reported between
as additional key players in orchestrating the inflammatory regulatory T-cell and C-reactive protein levels in hypertensive
response in experimental HHD.39 Injured cardiomyocytes patients.49
552  Hypertension  September 2018

Recent findings in patients and rats with HFpEF support products and reactive oxygen species) stimuli acting on any
that systemic inflammation induces oxidative stress in the of these steps.
coronary microvascular endothelium that results in reduced Fibrosis may contribute to the pathophysiological changes
myocardial NO availability, leading to reduced protein kinase of HHD through diverse pathways. First, a linkage between
G in cardiomyocytes, which, therefore, become stiff and fibrosis and LV dysfunction may be established.54 Initially, the
hypertrophied.50 A high cardiomyocyte stiffness has been accumulation of collagen fibers compromises the rate of relax-
related to a diminished distensibility of the giant cytoskeletal ation, diastolic suction, and passive stiffness, thereby contrib-
protein titin, whose elastic properties are dynamically modu- uting to impaired diastolic function. Continued accumulation
lated by isoform shifts, phosphorylation, and oxidation.51 Of of collagen fibers, accompanied by changes in their spatial
interest, it has been reported that hypertensive patients with orientation, further impairs diastolic filling. In particular, an
HFpEF have markedly increased passive myocardial stiffness association of myocardial fibrosis with LV stiffness has been
because of increases in the contribution of both titin and col- described in patients with HHD without HF55 and in patients
lagen (Figure 3).12 with HFpEF (mostly hypertensives)56 and also that collagen
contributes to increased myocardial passive stiffness in these
Fibrosis patients.12 Additionally, these changes compromise cardio-
Myocardial fibrosis, secondary to the diffuse accumulation myocyte contraction and myocardial force development,
of collagen type I and type III fibers within the interstitium thus impairing systolic performance. Of interest, it has been
and surrounding intramural coronary arteries and arterioles, reported that acute chamber stiffening is the main mechanism
is one of the key features of hypertensive MR.52 The excess responsible for rising late-diastolic pressures when patients
of collagen relative to the mass of cardiomyocytes within the with HFpEF undergo hypertension transients.57 This stiffening
myocardium in HHD is suggested to be the result of a process is related to impaired dynamic systolic-diastolic interactions
with several consecutive steps:53 (1) the differentiation of resi- and correlates with the quantity and degree of cross-linking
dent fibroblasts and other cell types into myofibroblasts; (2) of collagen deposits. Second, perivascular fibrosis may con-
an increased synthesis and secretion of procollagen, procol- tribute to impaired coronary flow reserve (CFR) through
lagen processing enzymes, and profibrotic growth factors and the external compression of intramural coronary arteries.58
cytokines by myofibroblasts; (3) an increased extracellular Because the amount of perivascular collagen has been cor-
conversion of procollagen into microfibril-forming collagen related inversely with CFR in hypertensive animals59 and
by the action of specific proteinases; (4) an increased spon- patients60 with LVH. Third, interstitial fibrosis may also con-
taneous microfibril assembly to form fibrils; (5) an enhanced tribute to ventricular arrhythmias in hypertension.61 Patients
Downloaded from http://ahajournals.org by on September 8, 2021

cross-linking of fibrils to form fibers through chemical reac- with HHD and arrhythmias exhibit higher values of myocar-
tions catalyzed by lysyl oxidases and other enzymes; and (6) dial collagen than patients without arrhythmias, despite the
an unchanged or decreased fiber degradation by matrix metal- finding that ejection fraction and the frequency of coronary
loproteinases and other enzymes. This process can be trig- vessels with significant stenosis may be similar in the 2 groups
gered either to replace small foci of dead cardiomyocytes or as of patients. Fibrosis induces conduction abnormalities thereby
a reaction to a diversity of mechanical (eg, stretch), humoral promoting local reentry arrhythmias. Additionally, the myofi-
(eg, aldosterone), and chemical (eg, advanced glycation end broblasts may alter the electric activity of the cardiomyocyte
through either direct intercellular interactions or secretion of
paracrine factors.62
Of interest, myocardial fibrosis may also adversely influ-
ence the clinical outcome in patients with HHD, namely in
those with HF. For instance, it has been shown that whereas
increased collagen type I cross-linking is associated per se
with HF hospitalization in patients with HHD and HF,63 the
coincidence of increased collagen type I cross-linking with
extensive collagen type I deposition is associated with HF
hospitalization and cardiovascular and all-cause death.64

Alterations of the Coronary Microvasculature


Together with alterations in the coronary macrocirculation
(eg, obstructive disease of the epicardial coronary arteries),
several structural alterations develop in the coronary micro-
vasculature (ie, prearterioles 100–500 μm in diameter and
arterioles <100 μm in diameter that make up the coronary
Figure 3.  Collagen-dependent and titin-dependent myocardial stress at a
sarcomere length (SL) of 2.6 μm for referent control patients (green bars),
microcirculation, capillaries, and venules) as part of MR. In
patients with hypertension but without heart failure (HF; orange bars), and combination with reduced diastolic myocardial perfusion
patients with hypertension and HF with preserved ejection fraction (red pressure because of increased arterial stiffness, they render
bars). Total stress is the numeric sum of collagen- and titin-specific data.
the hypertensive heart an ischemic organ.65.66 On one hand,
LV indicates left ventricular. *P<0.01 vs referent control patients, †P<0.01
vs patients with hypertension but without HF. Adapted from Zile et al12 with hyperplasia or hypertrophy and altered vascular smooth
permission. Copyright © 2015, American Heart Association, Inc. muscle cellular alignment may promote encroachment of the
González et al   Myocardial Remodeling in Hypertension   553

tunica media into the lumen, thereby causing both increased Phenotyping MR
medial thickness/lumen ratio or reduced maximal cross-sec- Whereas cardiovascular magnetic resonance (CMR) is the
tional area of prearterioles and arterioles. On the other hand, gold standard for the diagnosis of LVH, echocardiography is
vascular density in LVH becomes relatively decreased. This the most frequently used in clinical practice because of its low
seems to result from capillary rarefaction or inadequate vas- cost and availability. However, a recent analysis of 39 studies
cular growth in response to the increasing muscle mass. These focusing on LVH in hypertension reveals that LVH was vari-
alterations may depend, in part, on the abnormal transmission ably defined according to as many as 19 different echocardio-
of highly pulsatile blood pressure into microvascular net- graphic criteria,82 thus making the assessment of this entity
works, especially in highly perfused organs with low vascular highly imprecise and, more importantly, limiting the possibil-
resistance, such as the heart. Additionally, alterations in peri- ity to discriminate phenotypes truly representative of the diver-
cytes (ie, smooth muscle-like cells of mesenchymal origin that sity of mechanistic pathways and alterations underlying HHD.
surround capillary endothelial cells) may be involved in the As regards the phenotyping of MR, although the endo-
paucity of microvessels in HHD as it has been demonstrated myocardial biopsy is relatively safe and might serve to this
in other cardiac diseases.67 Finally, deposition of fibrotic tissue purpose taking advantage of the use of both histopathologic
around prearterioles and arterioles (ie, perivascular fibrosis) and molecular methodologies,83 alternative noninvasive meth-
increases oxygen diffusion distance leading to impairment of ods are needed for routine practice. In this regard, it is desir-
oxygen supply to cardiomyocytes.68 able that circulating or imaging biomarkers of MR are useful
These structural alterations, namely those of the coro- for classifying and staging HHD (Table 1).
nary microcirculation, together with endothelial dysfunc-
tion,69 contribute to reduction of CFR in patients with HHD.70 Circulating Biomarkers
Interestingly, associations between decreased CFR and LV The investigation of circulating biomarkers for MR detectable
systolic and diastolic dysfunction71,72 and mortality73 have been in blood by immunochemical methods has been accelerating
found in patients with HHD (Figure 4). In addition, it has been at a remarkable pace. These investigations have deluged the
proposed that the combination of microvascular ischemia and clinical and research communities with numerous candidates,
myocardial fibrosis may be involved in the facilitation of ven- few of which are, however, likely to survive as useful clinical
tricular tachyarrythmias and sudden cardiac death in patients tools in terms of diagnosis, prognosis, and therapy monitor-
with HHD.74,75 Of interest, it has been recently reported that ing. One possible explanation for this failure is that most of
the proportion of sudden cardiac deaths attributable to HHD the proposed biomarkers lack proof that they actually reflect
in the absence of coronary artery disease has increased during the structural, functional, or molecular alterations associated
with MR in patients with HHD. As shown in Table 1, several
Downloaded from http://ahajournals.org by on September 8, 2021

the last years.76


circulating parameters have been proposed as biomarkers of
Diagnosis and Treatment of MR in HHD MR in HHD. However, a strong evidence of association with
As MR negatively influences the clinical evolution of MR alterations has not been demonstrated for all of them.
patients with HHD, integrating its assessment into the eval- The clinical usefulness of circulating biomarkers of HHD
uation and management of these patients may be warranted. can be inferred from considering some examples. Recent find-
Therefore, it has been proposed that beyond the current ings demonstrate that the presence of echocardiographic LVH
clinical protocols based on detecting LVH and LV dysfunc- in conjunction with elevated circulating biomarkers for cardio-
tion and LVH regression,77 the time has come for additional myocyte injury/stress (high‐sensitivity cardiac troponin T and
strategies, aimed at both the noninvasive biochemical or amino-terminal pro‐B‐type natriuretic peptide) can identify a
imaging diagnosis78,79 and therapeutic repair80,81 of hyper- malignant phenotype of LVH more likely to progress to LV
tensive MR, to be considered. dysfunction, HF with reduced ejection fraction, or cardiovas-
cular death.84–86 In another study, it was found that the combi-
nation of 2 circulating biomarkers associated with a phenotype
of complex myocardial fibrosis characterized by both high
collagen type I deposition (ie, high serum PICP [C-terminal
propeptide of procollagen type I] levels)87,88 and high collagen
type I cross-linking (ie, low serum CITP [C-terminal telopep-
tide of collagen type I]:MMP-1 [matrix metalloproteinase-1
ratio])63 identifies a subgroup of ≈30% of patients with HHD
and HF with higher independent risk of HF hospitalization
and cardiovascular mortality than the remaining patients.64
Therefore, the combination of circulating biomarkers–based
phenotyping of HHD defines subpopulations of patients who
may benefit from enhanced surveillance and intervention to
prevent progression of the disease.
Imaging Biomarkers
Figure 4.  Survival curves of hypertensive patients with heart failure
CMR is currently the imaging method of choice to assess myo-
classified according to coronary flow reserve (CFR) threshold values
recorded in healthy controls. Adapted from Pereira et al73 with permission. cardial fibrosis (Table 1). In fact, CMR-assessed myocardial
Copyright © 2010, American Society of Hypertension. extracellular volume fraction (ECV) quantifies the interstitial
554  Hypertension  September 2018

Table 1.  Proposed Biomarkers for the Phenotyping of Myocardial Remodeling

Cardiomyocyte Alterations Interstitial Alterations Microvascular Alterations


Injury Endothelial Activation Coronary Flow
Type of Biomarker Hypertrophy Death Inflammation Fibrosis Dysfunction Dysregulation
Circulating CT-1 hs-TnT Gal-3 PICP VCAM-1
Annexin A5 sST-2 PIIINP P-selectin
CRP CITP:MMP-1 E-selectin
IL-6 Gal-3
GDF-15 sST-2
Imaging LV mass: 99 m Tc-annexin A5 VEGF receptors ECV (assessed by αvβ3 integrins Coronary flow reserve
(1.05×[1−ECV]) (assessed by SPECT) (assessed by PET) CMR) (assessed by PET and
(assessed by CMR) SPECT)
LV mass: (1.05×ECV)
(assessed by CMR)
Late gadolinium
enhancement
(assessed by CMR)
Backscatter amplitude
(assessed by
ultrasound)
Perfusable tissue index
(assessed by PET)
See references for each proposed biomarker in Table S1 in the online-only Data Supplement. CITP indicates carboxy-terminal telopeptide of collagen type I; CMR,
cardiac magnetic resonance; CRP, C-reactive protein; CT-1, cardiotrophin-1; ECV, extracellular volume; Gal-3, galectin-3; GDF-15, growth/differentiation factor 15;
hs-TnT, high-sensitivity troponin T; IL, interleukin; LV, left ventricular; MMP-1, matrix metalloproteinase-1; PET, positron emission tomography; PICP, carboxy-terminal
propeptide of procollagen type I; PIIINP, amino-terminal propeptide of procollagen type III; SPECT, single photon emission computed tomography; sST-2, soluble
suppressor of tumorigenicity-2; VCAM-1, vascular cell adhesion molecule-1; and VEGF, vascular endothelial growth factor.
Downloaded from http://ahajournals.org by on September 8, 2021

space, and if there is no myocardial edema or amyloidosis, to baseline coronary blood flow or CFR in response to vari-
then ECV is an adequate measurement of diffuse myocardial ous vasoactive stimuli).69,92 CFR is an integrated measure of
fibrosis.89 CMR-quantified myocardial ECV has been shown flow through both the large epicardial arteries and the coro-
to be increased in hypertension and associated with LVH and nary microcirculation. In the absence of obstructive steno-
dysfunction.90 But beyond the assessment of myocardial fibro- sis of the epicardial arteries, reduced CFR is a biomarker of
sis, recent advances suggest that CMR may have a role in a coronary microvascular dysfunction, but because obstructive
more global assessment of MR. As proposed by Treibel et al,91 disease of the epicardial arteries and coronary microvascu-
using T1 mapping CMR can split LV mass into cellular and lar dysfunction often coexist in the hypertensive patient,65,66
ECM components by measuring ECV and then calculating the discrimination of these 2 conditions on CFR is challenging.
cell volume as LV mass 1.05×(1−ECV) and the ECM volume
as LV mass 1.05×ECV (1.05 being the specific gravity of the Targeting MR
myocardium). With this approach, the authors demonstrated Recent findings show that regression of electrocardiographic93
that cell and ECM volumes increase proportionally in physi- and echocardiographic94 LVH is associated with lower inci-
ological and pathological LVH, but the ratio of proportional dence of cardiovascular morbidity and mortality in patients
increase differs depending on the pathogenesis of the LVH. with HHD independently of blood pressure reduction.
Because no patients with hypertensive LVH were included in However, the residual risk is still unacceptably high. In addi-
this study, further studies are required to histopathologically tion, the reduction in LV mass is not always achieved, and even
validate the method and ascertain its usefulness in phenotyp- de novo LVH may develop in treated patients, despite a good
ing MR in HHD. Furthermore, it can be anticipated that the control of blood pressure.95 Additionally, although in small
combination of these measurements with circulating biomark- and short-duration clinical studies, some conventional thera-
ers of cardiomyocyte hypertrophy and myocardial fibrosis pies, such as renin-angiotensin-aldosterone system inhibitors,
would enhance the sensitivity for phenotyping the diversity of reduce MR in patients with HHD,96,97 the alterations persist,
MR patterns in HHD. indicating a need to develop novel and effective anti-MR thera-
Currently, no technique allows for the direct visualization peutic strategies. In this conceptual framework, 2 approaches
of the coronary microvasculature in vivo in humans. Coronary are proposed: one relies on the biomarker-oriented use of drugs
microcirculation function can be indirectly assessed using currently used to treat hypertension that have proven ability to
several techniques that enable the measurement of param- repair MR, and another strategy is based on the development
eters that are strongly dependent on the functional integrity of new pharmacological agents capable of targeting the mecha-
of the coronary microcirculation (eg, the ratio of hyperemic nisms underlying the alterations constitutive of MR.
González et al   Myocardial Remodeling in Hypertension   555

Biomarker-Oriented Therapy Table 2.  Proposed Novel Strategies With Antiremodeling Therapeutic Potential
The use of panels combining circulating and imaging bio- Proposed Aims Molecular Targets Candidate Agents
markers of MR in translational research studies may integrate
At the cardiomyocyte level
diverse levels of information, overcome methodological limi-
tations, and contribute to a precise phenotyping of patients,   Prevent injury and death Drp1 Mdivi-1
with a view to personalizing HHD therapy. From this perspec- Cardiolipin MTP, SS-31
tive, and as an example, it can be hypothesized that patients
  Prevent hypertrophy Socs1 Anti-miR 155
presenting with the biomarker-based phenotype of complex
myocardial fibrosis, characterized by the combination of low CXCL1 Antibody
serum CITP:MMP-1 ratio (an index of increased myocardial CXCR2 SB265610
collagen type I cross-linking) and high serum PICP (an index   Preserve function Cardiolipin MTP
of increased myocardial collagen type I deposition), should be
At the interstitial level
treated with therapies with proven capacity to correct or atten-
uate both alterations of extracellular collagen processing and   Inhibit inflammation Rho-kinase Fasudil
consequently to reduce LV stiffness and improve LV function. P38 MAPK FR167653
In this context, recent data show that patients with HFpEF,
Socs1 Anti-miR 155
because of HHD among other etiologies, with a high degree of
collagen cross-linking (ie, low CITP:MMP-1) escape the ben- α7nAChR PNU282987
eficial effects of spironolactone, which is not able to reduce TLR2 Antibody
collagen type I deposition (ie, PICP levels) and to improve CXCL1 Antibody
diastolic function in this subgroup of patients.98 Interestingly,
CXCR2 SB265610
in a small pilot study, it has been shown that chronic treatment
with torasemide is able to reduce collagen cross-linking and   Reverse fibrosis Rho-kinase Fasudil
myocardial fibrosis in patients with HF with HHD.99 P38 MAPK FR167653

New Antiremodeling Strategies CXCL1 Antibody


During the last years, several new molecular targets and drugs CXCR2 SB265610
have been developed with therapeutic potential on MR act-
TLR2 Antibody
ing at 3 levels80,81: preventing cardiomyocyte hypertrophy and
TGF-β1 Receptor
Downloaded from http://ahajournals.org by on September 8, 2021

death to preserve its function, reversing interstitial alterations


antagonists
to reduce stiffness, and boosting angiogenesis to increase
oxygen and nutrition supply (Table 2). The selection of the PCP/PCPE Torasemide
candidates should be oriented to target common mechanisms LOX Torasemide,
affecting simultaneously multiple pathways involved in MR. EXP3179
As an example, novel, growing evidence identifies new poten- ADAM-17 A9B8 antibody
tial targets within the immune system for therapeutic interven-
miR Let-7i miR mimic
tions (eg, promoting immunosuppressive T-regulatory activity
and reducing proinflammatory T effector lymphocytes or miR-133a miR mimic
the products or targets thereof) that could contribute to limit LOXL2 Antibody
hypertensive MR in terms of avoiding cardiomyocyte hyper- Lnc Wisper ASO
trophy,100 suppressing myocardial inflammation101 and fibro-
sis,102 and protecting coronary microcirculation.103 At the microvascular level
Although many emerging therapies targeting MR are   Stimulate angiogenesis AMPKα2/VEGF Anti-miR-195a-3p
promising in preclinical models, many clinical trials of novel CD151 Anti-miR-124
antiremodeling drugs have failed. Therefore, it is important
  Restore endothelial function Cardiolipin MTP
to rationalize drug discovery by using meaningful in vivo and
in vitro models to discard irrelevant molecules in terms of PDE5 Sildenafil
efficacy and pharmacokinetic and toxicological profiles at an   Reverse vascular remodeling ADAM-17 A9B8 antibody
early stage. In this regard, an algorithm for selection of new See references for each molecular target and candidate agent in Table S2.
antifibrotic agents to be further tested as potential therapeutic α7nAChR indicates α7 nicotinic acetylcholine receptor; ADAM, disintegrin
targets has recently been proposed.104 and metalloproteinase; AMPK, AMP-activated protein kinase; ASO, antisense
oligonucleotides; CD151, Cluster of Differentiation 151; CXCL1, chemokine
Conclusions and Perspectives (C-X-C motif) ligand 1; CXCR2, chemokine (C-X-C motif) receptor 2; Drp1,
dynamin-related protein-1; Lnc, long noncoding; LOX, lysyl oxidase; LOXL2, lysyl
The alterations that develop in the myocardium of the oxidase-like 2; MAPK, p38 mitogen-activated protein kinase; Mdivi-1, chemical
hypertensive LV leading to its remodeling provide struc- compound mitochondrial division inhibitor-1; MTP, mitochondrial-targeted
tural and functional support for the alterations of cardiac peptide; PCP, procollagen type I carboxy-terminal proteinase; PCPE, procollagen
function and electric activity that adversely influence the type I carboxy-terminal proteinase enhancer; PDE5, phosphodiesterase type 5;
clinical evolution of patients with HHD. Therefore, a major Socs1, suppressor of cytokine signaling 1; TGF-β, transforming growth factor-β;
unmet need in HHD management is the ability to identify TLR2, toll-like receptor 2; and VEGF, vascular endothelial growth factor.
556  Hypertension  September 2018

homogeneous subsets of patients whose underlying myo- 14. Scheuer J. Catecholamines in cardiac hypertrophy. Am J Cardiol.
1999;83:70H–74H.
cardial lesions are driven by specific mechanisms that can
15. Lijnen P, Petrov V. Renin-angiotensin system, hypertrophy and gene
be solely targeted using personalized treatment. From this expression in cardiac myocytes. J Mol Cell Cardiol. 1999;31:949–970.
perspective, the noninvasive detection and effective repair doi: 10.1006/jmcc.1999.0934.
of MR must be considered as part of the overall clinical 16. Kamo T, Akazawa H, Komuro I. Cardiac nonmyocytes in the
hub of cardiac hypertrophy. Circ Res. 2015;117:89–98. doi:
care in patients with HHD. In doing so, adverse outcomes 10.1161/CIRCRESAHA.117.305349.
associated with this cardiac disease could be prevented 17. Kuwahara K, Kinoshita H, Kuwabara Y, Nakagawa Y, Usami S, Minami T,
more effectively. Clearly, further research is needed both in Yamada Y, Fujiwara M, Nakao K. Myocardin-related transcription factor
the experimental and clinical settings to reach these aims. A is a common mediator of mechanical stress- and neurohumoral stimula-
tion-induced cardiac hypertrophic signaling leading to activation of brain
natriuretic peptide gene expression. Mol Cell Biol. 2010;30:4134–4148.
Sources of Funding doi: 10.1128/MCB.00154-10.
This study was supported by the Ministry of Economy and 18. Kong SW, Bodyak N, Yue P, Liu Z, Brown J, Izumo S, Kang PM. Genetic
Competitiveness, Spain (Instituto de Salud Carlos III grants expression profiles during physiological and pathological cardiac hyper-
CB16/11/00483 and PI15/01909 cofinanced by FEDER [Fondo trophy and heart failure in rats. Physiol Genomics. 2005;21:34–42. doi:
Europeo de DEsarrollo Regional] funds), the European Commission 10.1152/physiolgenomics.00226.2004.
FP7 Programme (HOMAGE [Heart Omics in Ageing] project 2012– 19. Facundo HDTF, Brainard RE, Caldas FRL, Lucas AMB. Mitochondria
305507), the European Research Area Network on Cardiovascular and cardiac hypertrophy. Adv Exp Med Biol. 2017;982:203–226. doi:
Diseases Joint Transnational Call 2016 LYMIT-DIS (Targeted 10.1007/978-3-319-55330-6_11.
Lymphatic and Microvessel Treatment in Metabolic Disease HFpEF; 20. Tuomainen T, Tavi P. The role of cardiac energy metabolism in car-
AC16/00020), and LIPCAR-HF (Long Intergenei Noncoding RNA diac hypertrophy and failure. Exp Cell Res. 2017;360:12–18. doi:
Predicting Cardiac Remodeling; AC16/00016). 10.1016/j.yexcr.2017.03.052.
21. Neubauer S. The failing heart–an engine out of fuel. N Engl J Med.
2007;356:1140–1151. doi: 10.1056/NEJMra063052.
Disclosures 22. Dickhout JG, Carlisle RE, Austin RC. Interrelationship between cardiac
None. hypertrophy, heart failure, and chronic kidney disease: endoplasmic retic-
ulum stress as a mediator of pathogenesis. Circ Res. 2011;108:629–642.
References doi: 10.1161/CIRCRESAHA.110.226803.
23. Sun Y, Liu G, Song T, Liu F, Kang W, Zhang Y, Ge Z. Upregulation
1. Lawes CM, Vander Hoorn S, Rodgers A; International Society of
of GRP78 and caspase-12 in diastolic failing heart. Acta Biochim Pol.
Hypertension. Global burden of blood-pressure-related disease, 2001.
Lancet. 2008;371:1513–1518. doi: 10.1016/S0140-6736(08)60655-8. 2008;55:511–516.
2. Forouzanfar MH, Liu P, Roth GA, et al. Global burden of hypertension 24. González A, Ravassa S, Loperena I, López B, Beaumont J, Querejeta R,
and systolic blood pressure of at least 110 to 115 mm Hg, 1990-2015. Larman M, Díez J. Association of depressed cardiac gp130-mediated anti-
JAMA. 2017;317:165–182. doi: 10.1001/jama.2016.19043. apoptotic pathways with stimulated cardiomyocyte apoptosis in hyperten-
sive patients with heart failure. J Hypertens. 2007;25:2148–2157. doi:
Downloaded from http://ahajournals.org by on September 8, 2021

3. Frohlich ED, Apstein C, Chobanian AV, Devereux RB, Dustan HP, Dzau
V, Fauad-Tarazi F, Horan MJ, Marcus M, Massie B, Pfeffer MA, Re RN, 10.1097/HJH.0b013e32828626e2.
Roccella EJ, Savage D, Shub C. The heart in hypertension. N Engl J Med. 25. Ravassa S, González A, López B, Beaumont J, Querejeta R, Larman M,
1992;327:998–1008. Díez J. Upregulation of myocardial Annexin A5 in hypertensive heart dis-
4. Izzo JL Jr, Gradman AH. Mechanisms and management of hypertensive ease: association with systolic dysfunction. Eur Heart J. 2007;28:2785–
heart disease: from left ventricular hypertrophy to heart failure. Med Clin 2791. doi: 10.1093/eurheartj/ehm370.
North Am. 2004;88:1257–1271. doi: 10.1016/j.mcna.2004.06.002. 26. Li Z, Bing OH, Long X, Robinson KG, Lakatta EG. Increased cardio-
5. Lazzeroni D, Rimoldi O, Camici PG. From left ventricular hyper- myocyte apoptosis during the transition to heart failure in the spontane-
trophy to dysfunction and failure. Circ J. 2016;80:555–564. doi: ously hypertensive rat. Am J Physiol. 1997;272(5 pt 2):H2313–H2319.
10.1253/circj.CJ-16-0062. doi: 10.1152/ajpheart.1997.272.5.H2313.
6. Drazner MH. The progression of hypertensive heart disease. Circulation. 27. Narula J, Arbustini E, Chandrashekhar Y, Schwaiger M. Apoptosis and the
2011;123:327–334. doi: 10.1161/CIRCULATIONAHA.108.845792. systolic dysfunction in congestive heart failure. Story of apoptosis inter-
7. Díez J, González A, López B, Querejeta R. Mechanisms of disease: patho- ruptus and zombie myocytes. Cardiol Clin. 2001;19:113–126.
logic structural remodeling is more than adaptive hypertrophy in hyper- 28. Chandrashekhar Y. Role of apoptosis in ventricular remodeling. Curr
tensive heart disease. Nat Clin Pract Cardiovasc Med. 2005;2:209–216. Heart Fail Rep. 2005;2:18–22.
doi: 10.1038/ncpcardio0158. 29. Whelan RS, Kaplinskiy V, Kitsis RN. Cell death in the pathogen-
8. Shimizu I, Minamino T. Physiological and pathological car- esis of heart disease: mechanisms and significance. Annu Rev Physiol.
diac hypertrophy. J Mol Cell Cardiol. 2016;97:245–262. doi: 2010;72:19–44. doi: 10.1146/annurev.physiol.010908.163111.
10.1016/j.yjmcc.2016.06.001. 30. Wang ZV, Rothermel BA, Hill JA. Autophagy in hypertensive heart dis-
9. Swynghedauw B. Molecular mechanisms of myocardial remodeling. ease. J Biol Chem. 2010;285:8509–8514. doi: 10.1074/jbc.R109.025023.
Physiol Rev. 1999;79:215–262. doi: 10.1152/physrev.1999.79.1.215. 31. Kvakan H, Luft FC, Muller DN. Role of the immune system in hyperten-
10. Knöll R, Iaccarino G, Tarone G, Hilfiker-Kleiner D, Bauersachs J, Leite- sive target organ damage. Trends Cardiovasc Med. 2009;19:242–246. doi:
Moreira AF, Sugden PH, Balligand JL; European Society of Cardiology. 10.1016/j.tcm.2010.02.004.
Towards a re-definition of ‘cardiac hypertrophy’ through a rational charac- 32. McMaster WG, Kirabo A, Madhur MS, Harrison DG. Inflammation,
terization of left ventricular phenotypes: a position paper of the Working immunity, and hypertensive end-organ damage. Circ Res. 2015;116:1022–
Group ‘Myocardial Function’ of the ESC. Eur J Heart Fail. 2011;13:811– 1033. doi: 10.1161/CIRCRESAHA.116.303697.
819. doi: 10.1093/eurjhf/hfr071. 33. Altara R, Mallat Z, Booz GW, Zouein FA. The CXCL10/CXCR3 axis
11. Sugden PH, Clerk A. Cellular mechanisms of cardiac hypertrophy. J Mol and cardiac inflammation: implications for immunotherapy to treat
Med (Berl). 1998;76:725–746. infectious and noninfectious diseases of the heart. J Immunol Res.
12. Zile MR, Baicu CF, Ikonomidis JS, Stroud RE, Nietert PJ, Bradshaw 2016;2016:4396368. doi: 10.1155/2016/4396368.
AD, Slater R, Palmer BM, Van Buren P, Meyer M, Redfield MM, 34. Behr TM, Willette RN, Coatney RW, Berova M, Angermann CE, Anderson
Bull DA, Granzier HL, LeWinter MM. Myocardial stiffness in K, Sackner-Bernstein JD, Barone FC. Eprosartan improves cardiac per-
patients with heart failure and a preserved ejection fraction: contribu- formance, reduces cardiac hypertrophy and mortality and downregulates
tions of collagen and titin. Circulation. 2015;131:1247–1259. doi: myocardial monocyte chemoattractant protein-1 and inflammation in
10.1161/CIRCULATIONAHA.114.013215. hypertensive heart disease. J Hypertens. 2004;22:583–592.
13. Clerk A, Cullingford TE, Fuller SJ, Giraldo A, Markou T, Pikkarainen S, 35. Yoshida K, Kim-Mitsuyama S, Wake R, Izumiya Y, Izumi Y, Yukimura
Sugden PH. Signaling pathways mediating cardiac myocyte gene expres- T, Ueda M, Yoshiyama M, Iwao H. Excess aldosterone under normal
sion in physiological and stress responses. J Cell Physiol. 2007;212:311– salt diet induces cardiac hypertrophy and infiltration via oxidative stress.
322. doi: 10.1002/jcp.21094. Hypertens Res. 2005;28:447–455. doi: 10.1291/hypres.28.447.
González et al   Myocardial Remodeling in Hypertension   557

36. Ogata T, Miyauchi T, Sakai S, Takanashi M, Irukayama-Tomobe Y, 55. Díez J, Querejeta R, López B, González A, Larman M, Martínez Ubago
Yamaguchi I. Myocardial fibrosis and diastolic dysfunction in deoxycor- JL. Losartan-dependent regression of myocardial fibrosis is associ-
ticosterone acetate-salt hypertensive rats is ameliorated by the peroxi- ated with reduction of left ventricular chamber stiffness in hypertensive
some proliferator-activated receptor-alpha activator fenofibrate, partly patients. Circulation. 2002;105:2512–2517.
by suppressing inflammatory responses associated with the nuclear 56. Kasner M, Westermann D, Lopez B, Gaub R, Escher F, Kühl U,
factor-kappa-B pathway. J Am Coll Cardiol. 2004;43:1481–1488. doi: Schultheiss HP, Tschöpe C. Diastolic tissue Doppler indexes correlate
10.1016/j.jacc.2003.11.043. with the degree of collagen expression and cross-linking in heart failure
37. Levick SP, Murray DB, Janicki JS, Brower GL. Sympathetic ner- and normal ejection fraction. J Am Coll Cardiol. 2011;57:977–985. doi:
vous system modulation of inflammation and remodeling in 10.1016/j.jacc.2010.10.024.
the hypertensive heart. Hypertension. 2010;55:270–276. doi: 57. Pérez Del Villar C, Savvatis K, López B, Kasner M, Martinez-Legazpi P,
10.1161/HYPERTENSIONAHA.109.142042. Yotti R, González A, Díez J, Fernández-Avilés F, Tschöpe C, Bermejo J.
38. Deckx S, Heggermont W, Carai P, Rienks M, Dresselaers T, Himmelreich Impact of acute hypertension transients on diastolic function in patients
U, van Leeuwen R, Lommen W, van der Velden J, Gonzalez A, Diez J, with heart failure with preserved ejection fraction. Cardiovasc Res.
Papageorgiou AP, Heymans S. Osteoglycin prevents the development of 2017;113:906–914. doi: 10.1093/cvr/cvx047.
age-related diastolic dysfunction during pressure overload by reducing 58. Schwartzkopff B, Motz W, Frenzel H, Vogt M, Knauer S, Strauer BE.
cardiac fibrosis and inflammation. Matrix Biol. 2018;66:110–124. doi: Structural and functional alterations of the intramyocardial coro-
10.1016/j.matbio.2017.09.002. nary arterioles in patients with arterial hypertension. Circulation.
39. Ghigo A, Franco I, Morello F, Hirsch E. Myocyte signalling in leuco- 1993;88:993–1003.
cyte recruitment to the heart. Cardiovasc Res. 2014;102:270–280. doi: 59. Brilla CG, Janicki JS, Weber KT. Impaired diastolic function and coro-
10.1093/cvr/cvu030. nary reserve in genetic hypertension. Role of interstitial fibrosis and
40. Shinde AV, Frangogiannis NG. Mechanisms of fibroblast activation in medial thickening of intramyocardial coronary arteries. Circ Res.
the remodeling myocardium. Curr Pathobiol Rep. 2017;5:145–152. doi: 1991;69:107–115.
10.1007/s40139-017-0132-z. 60. Dai Z, Aoki T, Fukumoto Y, Shimokawa H. Coronary perivascular fibro-
41. Kuwahara F, Kai H, Tokuda K, Takeya M, Takeshita A, Egashira K, sis is associated with impairment of coronary blood flow in patients
Imaizumi T. Hypertensive myocardial fibrosis and diastolic dysfunction: with non-ischemic heart failure. J Cardiol. 2012;60:416–421. doi:
another model of inflammation? Hypertension. 2004;43:739–745. doi: 10.1016/j.jjcc.2012.06.009.
10.1161/01.HYP.0000118584.33350.7d. 61. McLenachan JM, Dargie HJ. Ventricular arrhythmias in hypertensive
42. Kai H, Kuwahara F, Tokuda K, Imaizumi T. Diastolic dysfunction in hyper- left ventricular hypertrophy. Relationship to coronary artery disease,
tensive hearts: roles of perivascular inflammation and reactive myocardial left ventricular dysfunction, and myocardial fibrosis. Am J Hypertens.
fibrosis. Hypertens Res. 2005;28:483–490. doi: 10.1291/hypres.28.483. 1990;3:735–740.
43. Westermann D, Lindner D, Kasner M, Zietsch C, Savvatis K, Escher 62. Rohr S. Arrhythmogenic implications of fibroblast-myocyte inter-
F, von Schlippenbach J, Skurk C, Steendijk P, Riad A, Poller W, actions. Circ Arrhythm Electrophysiol. 2012;5:442–452. doi:
Schultheiss HP, Tschöpe C. Cardiac inflammation contributes to 10.1161/CIRCEP.110.957647.
changes in the extracellular matrix in patients with heart failure 63. López B, Ravassa S, González A, Zubillaga E, Bonavila C, Bergés
and normal ejection fraction. Circ Heart Fail. 2011;4:44–52. doi: M, Echegaray K, Beaumont J, Moreno MU, San José G, Larman M,
10.1161/CIRCHEARTFAILURE.109.931451. Querejeta R, Díez J. Myocardial collagen cross-linking is associated with
44. Idris-Khodja N, Mian MO, Paradis P, Schiffrin EL. Dual opposing roles heart failure hospitalization in patients with hypertensive heart failure. J
Downloaded from http://ahajournals.org by on September 8, 2021

of adaptive immunity in hypertension. Eur Heart J. 2014;35:1238–1244. Am Coll Cardiol. 2016;67:251–260. doi: 10.1016/j.jacc.2015.10.063.
doi: 10.1093/eurheartj/ehu119. 64. Ravassa S, López B, Querejeta R, Echegaray K, San José G, Moreno MU,
45. Blake GJ, Rifai N, Buring JE, Ridker PM. Blood pressure, C-reactive pro- Beaumont FJ, González A, Díez J. Phenotyping of myocardial fibrosis in
tein, and risk of future cardiovascular events. Circulation. 2003;108:2993– hypertensive patients with heart failure. Influence on clinical outcome. J
2999. doi: 10.1161/01.CIR.0000104566.10178.AF. Hypertens. 2017;35:853–861. doi: 10.1097/HJH.0000000000001258.
46. Bautista LE, Vera LM, Arenas IA, Gamarra G. Independent association 65. Feihl F, Liaudet L, Levy BI, Waeber B. Hypertension and micro-
between inflammatory markers (C-reactive protein, interleukin-6, and vascular remodelling. Cardiovasc Res. 2008;78:274–285. doi:
TNF-alpha) and essential hypertension. J Hum Hypertens. 2005;19:149– 10.1093/cvr/cvn022.
154. doi: 10.1038/sj.jhh.1001785. 66. Feihl F, Liaudet L, Waeber B. The macrocirculation and microcirculation
47. Dalekos GN, Elisaf M, Bairaktari E, Tsolas O, Siamopoulos KC. of hypertension. Curr Hypertens Rep. 2009;11:182–189.
Increased serum levels of interleukin-1beta in the systemic circulation of 67. Avolio E, Madeddu P. Discovering cardiac pericyte biology: from
patients with essential hypertension: additional risk factor for atherogen- physiopathological mechanisms to potential therapeutic applications
esis in hypertensive patients? J Lab Clin Med. 1997;129:300–308. in ischemic heart disease. Vascul Pharmacol. 2016;86:53–63. doi:
48. Youn JC, Yu HT, Lim BJ, Koh MJ, Lee J, Chang DY, Choi YS, Lee 10.1016/j.vph.2016.05.009.
SH, Kang SM, Jang Y, Yoo OJ, Shin EC, Park S. Immunosenescent 68. Frohlich ED. Fibrosis and ischemia: the real risks in hypertensive heart
CD8+ T cells and C-X-C chemokine receptor type 3 chemokines are disease. Am J Hypertens. 2001;14:194S–199S.
increased in human hypertension. Hypertension. 2013;62:126–133. doi: 69. Treasure CB, Klein JL, Vita JA, Manoukian SV, Renwick GH, Selwyn
10.1161/HYPERTENSIONAHA.113.00689. AP, Ganz P, Alexander RW. Hypertension and left ventricular hypertrophy
49. De Ciuceis C, Agabiti-Rosei C, Rossini C, et al. Relationship between are associated with impaired endothelium-mediated relaxation in human
different subpopulations of circulating CD4+ T lymphocytes and coronary resistance vessels. Circulation. 1993;87:86–93.
microvascular or systemic oxidative stress in humans. Blood Press. 70. Kelm M, Strauer BE. Coronary flow reserve measurements in hyperten-
2017;26:237–245. doi: 10.1080/08037051.2017.1292395. sion. Med Clin North Am. 2004;88:99–113.
50. Franssen C, Chen S, Unger A, Korkmaz HI, De Keulenaer GW, Tschöpe 71. Galderisi M, Cicala S, Caso P, De Simone L, D’Errico A, Petrocelli A, de
C, Leite-Moreira AF, Musters R, Niessen HW, Linke WA, Paulus WJ, Divitiis O. Coronary flow reserve and myocardial diastolic dysfunction in
Hamdani N. Myocardial microvascular inflammatory endothelial activa- arterial hypertension. Am J Cardiol. 2002;90:860–864.
tion in heart failure with preserved ejection fraction. JACC Heart Fail. 72. Ikonomidis I, Tzortzis S, Paraskevaidis I, Triantafyllidi H, Papadopoulos
2016;4:312–324. doi: 10.1016/j.jchf.2015.10.007. C, Papadakis I, Trivilou P, Parissis J, Anastasiou-Nana M, Lekakis
51. Linke WA, Hamdani N. Gigantic business: titin properties and func- J. Association of abnormal coronary microcirculatory function with
tion through thick and thin. Circ Res. 2014;114:1052–1068. doi: impaired response of longitudinal left ventricular function during adenos-
10.1161/CIRCRESAHA.114.301286. ine stress echocardiography in untreated hypertensive patients. Eur Heart
52. Berk BC, Fujiwara K, Lehoux S. ECM remodeling in hypertensive heart J Cardiovasc Imaging. 2012;13:1030–1040. doi: 10.1093/ehjci/jes071.
disease. J Clin Invest. 2007;117:568–575. doi: 10.1172/JCI31044. 73. Pereira VF, de Carvalho Frimm C, Rodrigues AC, Cúri M. Coronary
53. González A, Schelbert EB, Díez J, Butler J. Myocardial interstitial fibro- reserve impairment prevents the improvement of left ventricular dysfunc-
sis in heart failure: biological and translational perspectives. J Am Coll tion and adversely affects the long-term outcome of patients with hyper-
Cardiol. 2018;71:1696–1706. doi: 10.1016/j.jacc.2018.02.021. tensive dilated cardiomyopathy. J Am Soc Hypertens. 2010;4:14–21. doi:
54. Brower GL, Gardner JD, Forman MF, Murray DB, Voloshenyuk T, 10.1016/j.jash.2009.12.002.
Levick SP, Janicki JS. The relationship between myocardial extracellu- 74. Zehender M, Faber T, Koscheck U, Meinertz T, Just H. Ventricular
lar matrix remodeling and ventricular function. Eur J Cardiothorac Surg. tachyarrhythmias, myocardial ischemia, and sudden cardiac death in
2006;30:604–610. doi: 10.1016/j.ejcts.2006.07.006. patients with hypertensive heart disease. Clin Cardiol. 1995;18:377–383.
558  Hypertension  September 2018

75. Shenasa M, Shenasa H. Hypertension, left ventricular hypertro- 90. Wang S, Hu H, Lu M, et al. Myocardial extracellular volume fraction
phy, and sudden cardiac death. Int J Cardiol. 2017;237:60–63. doi: quantified by cardiovascular magnetic resonance is increased in hyper-
10.1016/j.ijcard.2017.03.002. tension and associated with left ventricular remodeling. Eur Radiol.
76. Junttila MJ, Hookana E, Kaikkonen KS, Kortelainen ML, Myerburg RJ, 2017;27:4620–4630. doi: 10.1007/s00330-017-4841-9.
Huikuri HV. Temporal trends in the clinical and pathological characteris- 91. Treibel TA, Kozor R, Menacho K, Castelletti S, Bulluck H, Rosmini
tics of victims of sudden cardiac death in the absence of previously identi- S, Nordin S, Maestrini V, Fontana M, Moon JC. Left ventricu-
fied heart disease. Circ Arrhythm Electrophysiol. 2016;9:e003723. lar hypertrophy revisited: cell and matrix expansion have disease-
77. Schiattarella GG, Hill JA. Inhibition of hypertrophy is a good therapeutic specific relationships. Circulation. 2017;136:2519–2521. doi:
strategy in ventricular pressure overload. Circulation. 2015;131:1435– 10.1161/CIRCULATIONAHA.117.029895.
1447. doi: 10.1161/CIRCULATIONAHA.115.013894. 92. Camici PG, d’Amati G, Rimoldi O. Coronary microvascular dysfunction:
78. González A, López B, Ravassa S, Beaumont J, Arias T, Hermida N, mechanisms and functional assessment. Nat Rev Cardiol. 2015;12:48–
Zudaire A, Díez J. Biochemical markers of myocardial remodelling 62. doi: 10.1038/nrcardio.2014.160.
in hypertensive heart disease. Cardiovasc Res. 2009;81:509–518. doi: 93. Bang CN, Devereux RB, Okin PM. Regression of electrocardiographic
10.1093/cvr/cvn235. left ventricular hypertrophy or strain is associated with lower incidence
79. Hoey ET, Pakala V, Teoh JK, Simpson H. The role of imaging of cardiovascular morbidity and mortality in hypertensive patients inde-
in hypertensive heart disease. Int J Angiol. 2014;23:85–92. doi: pendent of blood pressure reduction - A LIFE review. J Electrocardiol.
10.1055/s-0034-1370885. 2014;47:630–635. doi: 10.1016/j.jelectrocard.2014.07.003.
80. Tarone G, Balligand JL, Bauersachs J, Clerk A, De Windt L, Heymans 94. de Simone G, Devereux RB, Izzo R, Girfoglio D, Lee ET, Howard BV,
S, Hilfiker-Kleiner D, Hirsch E, Iaccarino G, Knöll R, Leite-Moreira Roman MJ. Lack of reduction of left ventricular mass in treated hyper-
AF, Lourenço AP, Mayr M, Thum T, Tocchetti CG. Targeting myocar- tension: the strong heart study. J Am Heart Assoc. 2013;2:e000144. doi:
dial remodelling to develop novel therapies for heart failure: a posi- 10.1161/JAHA.113.000144.
tion paper from the Working Group on Myocardial Function of the 95. Pierdomenico SD, Cuccurullo F. Risk reduction after regression of echo-
European Society of Cardiology. Eur J Heart Fail. 2014;16:494–508. cardiographic left ventricular hypertrophy in hypertension: a meta-anal-
doi: 10.1002/ejhf.62. ysis. Am J Hypertens. 2010;23:876–881. doi: 10.1038/ajh.2010.80.
81. González A, Ravassa S, Beaumont J, López B, Díez J. New targets to 96. López B, Querejeta R, Varo N, González A, Larman M, Martínez Ubago
treat the structural remodeling of the myocardium. J Am Coll Cardiol. JL, Díez J. Usefulness of serum carboxy-terminal propeptide of procol-
2011;58:1833–1843. doi: 10.1016/j.jacc.2011.06.058. lagen type I in assessment of the cardioreparative ability of antihyperten-
82. Cuspidi C, Esposito A, Negri F, Sala C, Masaidi M, Giudici V, Zanchetti sive treatment in hypertensive patients. Circulation. 2001;104:286–291.
A, Mancia G. Studies on left ventricular hypertrophy regression in arte- 97. Brilla CG, Funck RC, Rupp H. Lisinopril-mediated regression of myo-
rial hypertension: a clear message for the clinician? Am J Hypertens. cardial fibrosis in patients with hypertensive heart disease. Circulation.
2008;21:458–463. doi: 10.1038/ajh.2007.85. 2000;102:1388–1393.
83. Chimenti C, Frustaci A. Contribution and risks of left ventricular endo- 98. Ravassa S, Trippel T, Bach D, Bachran D, González A, López B,
myocardial biopsy in patients with cardiomyopathies: a retrospective Wachter R, Hasenfuss G, Delles C, Dominiczak AF, Pieske B, Díez
study over a 28-year period. Circulation. 2013;128:1531–1541. doi: J, Edelmann F. Biomarker-based phenotyping of myocardial fi-
10.1161/CIRCULATIONAHA.13.001414. brosis identifies patients with heart failure with preserved ejec-
84. Neeland IJ, Drazner MH, Berry JD, Ayers CR, deFilippi C, Seliger SL, tion fraction resistant to the beneficial effects of spironolactone:
Nambi V, McGuire DK, Omland T, de Lemos JA. Biomarkers of chronic results from the Aldo-DHF trial [published online April 30, 2018].
Downloaded from http://ahajournals.org by on September 8, 2021

cardiac injury and hemodynamic stress identify a malignant phenotype of Eur J Heart Fail. doi: 10.1002/ejhf.1194. https://onlinelibrary.
left ventricular hypertrophy in the general population. J Am Coll Cardiol. wiley.com/doi/abs/10.1002/ejhf.1194.
2013;61:187–195. doi: 10.1016/j.jacc.2012.10.012. 99. López B, Querejeta R, González A, Beaumont J, Larman M, Díez J.
85. Seliger SL, de Lemos J, Neeland IJ, Christenson R, Gottdiener J, Impact of treatment on myocardial lysyl oxidase expression and collagen
Drazner MH, Berry J, Sorkin J, deFilippi C. Older adults, “Malignant” cross-linking in patients with heart failure. Hypertension. 2009;53:236–
left ventricular hypertrophy, and associated cardiac-specific bio- 242. doi: 10.1161/HYPERTENSIONAHA.108.125278.
marker phenotypes to identify the differential risk of new-onset 100. Fabbiano S, Menacho-Márquez M, Robles-Valero J, Pericacho M,

reduced versus preserved ejection fraction heart failure: CHS Matesanz-Marín A, García-Macías C, Sevilla MA, Montero MJ, Alarcón
(Cardiovascular Health Study). JACC Heart Fail. 2015;3:445–455. doi: B, López-Novoa JM, Martín P, Bustelo XR. Immunosuppression-
10.1016/j.jchf.2014.12.018. independent role of regulatory T cells against hypertension-driv-
86. Peters MN, Seliger SL, Christenson RH, Hong-Zohlman SN, Daniels en renal dysfunctions. Mol Cell Biol. 2015;35:3528–3546. doi:
LB, Lima JAC, de Lemos JA, Neeland IJ, deFilippi CR. “Malignant” left 10.1128/MCB.00518-15.
ventricular hypertrophy identifies subjects at high risk for progression 101. Kvakan H, Kleinewietfeld M, Qadri F, Park JK, Fischer R, Schwarz
to asymptomatic left ventricular dysfunction, heart failure, and death: I, Rahn HP, Plehm R, Wellner M, Elitok S, Gratze P, Dechend R,
MESA (Multi-Ethnic Study of Atherosclerosis). J Am Heart Assoc. Luft FC, Muller DN. Regulatory T cells ameliorate angiotensin II-
2018;7:e006619. induced cardiac damage. Circulation. 2009;119:2904–2912. doi:
87. Querejeta R, López B, González A, Sánchez E, Larman M, 10.1161/CIRCULATIONAHA.108.832782.
Martínez Ubago JL, Díez J. Increased collagen type I synthe- 102. Kanellakis P, Dinh TN, Agrotis A, Bobik A. CD4⁺CD25⁺Foxp3⁺ regu-
sis in patients with heart failure of hypertensive origin: relation latory T cells suppress cardiac fibrosis in the hypertensive heart. J
to myocardial fibrosis. Circulation. 2004;110:1263–1268. doi: Hypertens. 2011;29:1820–1828. doi: 10.1097/HJH.0b013e328349c62d.
10.1161/01.CIR.0000140973.60992.9A. 103. Matrougui K, Abd Elmageed Z, Zakaria AE, Kassan M, Choi S, Nair
88. López B, Querejeta R, González A, Sánchez E, Larman M, Díez J. Effects D, Gonzalez-Villalobos RA, Chentoufi AA, Kadowitz P, Belmadani S,
of loop diuretics on myocardial fibrosis and collagen type I turnover Partyka M. Natural regulatory T cells control coronary arteriolar endo-
in chronic heart failure. J Am Coll Cardiol. 2004;43:2028–2035. doi: thelial dysfunction in hypertensive mice. Am J Pathol. 2011;178:434–
10.1016/j.jacc.2003.12.052. 441. doi: 10.1016/j.ajpath.2010.11.034.
89. Diao KY, Yang ZG, Xu HY, Liu X, Zhang Q, Shi K, Jiang L, Xie LJ, Wen 104. Gyöngyösi M, Winkler J, Ramos I, Do QT, Firat H, McDonald K,
LY, Guo YK. Histologic validation of myocardial fibrosis measured by González A, Thum T, Díez J, Jaisser F, Pizard A, Zannad F. Myocardial
T1 mapping: a systematic review and meta-analysis. J Cardiovasc Magn fibrosis: biomedical research from bench to bedside. Eur J Heart Fail.
Reson. 2016;18:92. doi: 10.1186/s12968-016-0313-7. 2017;19:177–191. doi: 10.1002/ejhf.696.

You might also like