Download as pdf or txt
Download as pdf or txt
You are on page 1of 34

AHA Scientific Statement

Animal Models of Hypertension


A Scientific Statement From the American Heart Association
Lilach O. Lerman, MD, PhD, FAHA, Chair; Theodore W. Kurtz, MD, FAHA;
Rhian M. Touyz, BSc, MBBCh, MSc, PhD, FAHA; David H. Ellison, MD, FAHA;
Alejandro R. Chade, MD, FAHA; Steven D. Crowley, MD, FAHA;
David L. Mattson, PhD, FAHA; John J. Mullins, PhD, FRSE; Jeffrey Osborn, PhD, FAHA;
Alfonso Eirin, MD; Jane F. Reckelhoff, PhD, FAHA; Costantino Iadecola, MD;
Thomas M. Coffman, MD, FAHA, Co-Chair; on behalf of the American Heart Association Council on
Hypertension and Council on Clinical Cardiology

Abstract—Hypertension is the most common chronic disease in the world, yet the precise cause of elevated blood pressure
often cannot be determined. Animal models have been useful for unraveling the pathogenesis of hypertension and for
testing novel therapeutic strategies. The utility of animal models for improving the understanding of the pathogenesis,
prevention, and treatment of hypertension and its comorbidities depends on their validity for representing human forms
of hypertension, including responses to therapy, and on the quality of studies in those models (such as reproducibility
and experimental design). Important unmet needs in this field include the development of models that mimic the
discrete hypertensive syndromes that now populate the clinic, resolution of ongoing controversies in the pathogenesis
of hypertension, and the development of new avenues for preventing and treating hypertension and its complications.
Animal models may indeed be useful for addressing these unmet needs. (Hypertension. 2019;73:e•••–e•••. doi: 10.1161/
HYP.0000000000000090.)
Key Words: AHA Scientific Statements ■ blood pressure ■ hypertension ■ models, animal
Downloaded from http://ahajournals.org by on March 14, 2019

H ypertension is the most common chronic disease in the


world and produces substantial morbidity and mortal-
ity. However, in the majority of individuals, the precise cause
of hypertension, they can nonetheless be relatively common,
given the broad prevalence of hypertension itself. Malignant
hypertension is related to, but pathophysiologically distinct
of elevated blood pressure (BP) cannot be determined. Risk from, primary hypertension, as is the syndrome of preeclamp-
factors for primary (formerly called essential) hypertension sia. Secondary causes may involve the renal vasculature, endo-
include advancing age, obesity, high dietary NaCl consumption, crine organs, and kidney and may be involved in up to 20% of
and low dietary potassium intake, although these appear to con- cases of resistant hypertension. Finally, an increasing number
tribute to, but not cause, hypertension. Renin-sodium profiling of drugs used to treat cancer and other conditions are now
has been used to classify primary hypertension, suggesting that recognized as causing hypertension, which is often severe.
the phenotype is highly variable, but treatment remains largely Genetic forms of hypertension with mendelian inheritance are
empirical and influenced by race and comorbid disease. rare but have helped to identify important BP-regulating path-
A number of hypertensive subtypes also exist, and ways. Over the past 20 years, some the most important scien-
although they may make up only a small fraction of the cases tific breakthroughs have emanated from the discovery of the

The American Heart Association makes every effort to avoid any actual or potential conflicts of interest that may arise as a result of an outside relationship
or a personal, professional, or business interest of a member of the writing panel. Specifically, all members of the writing group are required to complete
and submit a Disclosure Questionnaire showing all such relationships that might be perceived as real or potential conflicts of interest.
This statement was approved by the American Heart Association Science Advisory and Coordinating Committee on November 15, 2018, and the
American Heart Association Executive Committee on November 27, 2018. A copy of the document is available at https://professional.heart.org/statements
by using either “Search for Guidelines & Statements” or the “Browse by Topic” area. To purchase additional reprints, call 843-216-2533 or e-mail kelle.
ramsay@wolterskluwer.com.
The American Heart Association requests that this document be cited as follows: Lerman LO, Kurtz TW, Touyz RM, Ellison DH, Chade AR, Crowley
SD, Mattson DL, Mullins JJ, Osborn J, Eirin A, Reckelhoff JF, Iadecola C, Coffman TM; on behalf of the American Heart Association Council on
Hypertension and Council on Clinical Cardiology. Animal models of hypertension: a scientific statement from the American Heart Association.
Hypertension. 2019;73:e•••–e•••. doi: 10.1161/HYP.0000000000000090.
The expert peer review of AHA-commissioned documents (eg, scientific statements, clinical practice guidelines, systematic reviews) is conducted by the
AHA Office of Science Operations. For more on AHA statements and guidelines development, visit https://professional.heart.org/statements. Select the
“Guidelines & Statements” drop-down menu, then click “Publication Development.”
Permissions: Multiple copies, modification, alteration, enhancement, and/or distribution of this document are not permitted without the express permission
of the American Heart Association. Instructions for obtaining permission are located at https://www.heart.org/permissions. A link to the “Copyright
Permissions Request Form” appears in the second paragraph (https://www.heart.org/en/about-us/statements-and-policies/copyright-request-form).
© 2019 American Heart Association, Inc.
Hypertension is available at https://www.ahajournals.org/journal/hyp DOI: 10.1161/HYP.0000000000000090

e1
e2  Hypertension   TBD 2019

basis of rare subtypes of human hypertension. Among these studies in translational medical research has come under
are the solution of nearly all the monogenic causes of hyper- increasing scrutiny because of low study reproducibility and
tension; identification of discrete somatic mutations that cause problems such as bias, poor experimental design and execu-
primary aldosteronism; the discovery that polymorphisms in tion, analytical and logical errors, and incomplete reporting.4–8
the APOL1 gene underlie some racial disparities in hyperten- Published recommendations on ways to mitigate these issues
sive kidney disease; the discovery that placental insufficiency should be considered for any studies using animal models. It
generates placental growth factor and sFlt-1 (soluble fms-like should be noted that >1000 scientific journals have endorsed
tyrosine kinase-1), factors that mark and contribute to pre- guidelines designed to improve the reporting of animal experi-
eclampsia; and finally, the recognition that certain anticancer ments. Nonetheless, these guidelines should be applied cau-
drugs commonly cause hypertension by impairing the func- tiously because excessive regulation may also hinder studies
tion of the vascular endothelium and the glomerulus. in animals.
The initial animal models of hypertension to be developed Various criteria have been used to assess the utility of
involved constriction of renal arteries (Goldblatt kidney) or animal models in translational medical research, including
parenchyma (Page kidney); the pathophysiology closely mim- face validity, construct validity, and predictive validity.9 By
icked their human analogs. However, renovascular hyperten- conventional definition, each animal model of hypertension
sion and Page kidney represent only a small fraction of human has at least some rudimentary degree of face validity in that
hypertension. Most experimental studies of hypertension each demonstrates the primary diagnostic feature, an increase
using animals have therefore focused on understanding the in BP compared with a level deemed to be normal. However,
mechanisms of primary hypertension. some models may have greater face validity than others with
Although excellent animal models with good human fidel- respect to other phenotypic aspects of hypertension such as
ity have been developed for many of these rare causes of hyper- age at onset, temporal course, severity, variability, and associ-
tension,1,2 models of primary hypertension have been more ated comorbidities. Given the clinical importance of hyperten-
difficult to develop, largely because the causes of the human sion-related target organ damage, it is noteworthy that models
disorder are unclear. Of National Institutes of Health–sponsored are also available exhibiting face validity with respect to risk
hypertension research, studies using Ang II (angiotensin II) for hypertension-related disturbances such as left ventricular
infusion make up a disproportionate share (nearly 50%).3 Only hypertrophy (LVH), metabolic abnormalities, heart failure,
4% of studies focus on aging and 4% focus on DOCA (deoxy- renal damage, and stroke (eg, spontaneously hypertensive
corticosterone acetate)–salt hypertension (which itself does not rats [SHRs], Dahl salt-sensitive [DSS] rats).10–16 However,
model primary aldosteronism). Thus, an important unmet need other hypertension-associated conditions such as spontaneous
Downloaded from http://ahajournals.org by on March 14, 2019

is to develop better animal models that more closely mimic the development of atherosclerosis or acute myocardial infarction
discrete hypertensive syndromes that now populate the clinic are not typically observed in current models.
such as primary aldosteronism. A corollary would be that the Although all typical animal models uniformly exhibit
portfolio of hypertension research might more closely mimic increased BP, the models vary considerably with respect to
the spectrum of human hypertension. construct validity, defined by how faithfully they recapitulate
A second important unmet need is to resolve ongoing key features of human hypertension such as genetic and envi-
controversies concerning pathogenesis. Proponents for indi- ronmental triggers or key pathophysiological mechanisms. As
vidual pathways, including the primacy of the nervous system, in other fields, there is no ideal animal model of human hyper-
kidney, and vasculature in the development of hypertension, tension; all have inherent limitations in construct validity. For
typically focused on their own views and interests, often example, there are striking differences between humans and
independently of considerations of heritability, environmen- animals with respect to factors that influence BP, including
tal exposure, and developmental programming. Despite >50 genetics, physiology, anatomy, behavior, environmental con-
years of work, there is no consensus integrating this range of ditions, and triggers. The nature of these differences, particu-
contributing causative factors. This persistent lack of conver- larly between humans and rodents, has motivated efforts to
gence slows bona fide progress and can limit the impact of the study hypertension and related disorders in larger animals
field. Addressing this unmet need will require that we bring and nonhuman primates. In addition, it should be emphasized
together diverse teams with competing views who are com- that the validity or utility of BP studies in animal models may
mitted to this common goal. be compromised by the use of BP measurement techniques
involving anesthesia or other forms of stress or those that do
Utility and Validity of Animal Models of not allow adequate assessment of BP over 24 hours and of key
Hypertension features of the BP waveform, including both systolic and dia-
Across a range of human diseases, including hypertension, stolic BPs, because they may influence risk for cardiovascular
animal models have been useful for unraveling disease patho- events.17–19
genesis by providing incisive experimental strategies not pos-
sible in human studies. In hypertension, the utility of animal Predictive Validity and the Primary Reason for
models for improving the understanding of the pathogenesis, Using Animal Models in Hypertension Research
prevention, and treatment of hypertension and its comorbidi- The main goal of studying animal models of hypertension is
ties depends on their validity for representing human forms to help develop improved approaches to preventing and treat-
of hypertension, including responses to therapy, and the qual- ing high BP and its complications. Therefore, from a practi-
ity of studies in those models. Recently, the utility of animal cal perspective, the most important aspect of such a model
Lerman et al   Animal Models of Hypertension   e3

is predictive validity, defined by its value for guiding devel- variables may also vary among models, the magnitude of the
opment of effective preventive or therapeutic interventions in BP response to a given antihypertensive treatment can vary
humans. This raises several corollaries. What are the major greatly among animal models, just as among different patient
obstacles and unmet needs for effectively preventing and treat- subgroups. The availability of a wide variety of animal models
ing hypertension and its complications? How useful are stud- is advantageous for generating hypotheses about the patho-
ies in a particular animal model of hypertension for addressing genesis, prevention, and treatment of different forms of high
these unmet needs? Some of the obstacles to achieving effec- BP in humans. Nonetheless, some approaches to lowering BP
tive BP control and reduction of associated cardiovascular risk were apparently first tested in humans (eg, ablation of sympa-
are related to behavioral issues leading to poor adherence to thetic nerves, diet therapies, weight reduction, supplemental
therapies or preventive measures, in which case application of potassium, and various drugs), bringing into question the need
animal models is unlikely to be productive. for research on animal models of human disorders.23 Finally,
On the other hand, the problem of resistant hypertension is because the main goal of treating hypertension is reducing the
an area in which animal models could have significant utility. risk for devastating cardiovascular complications, one could
Even in patients thought to be taking the requisite antihyper- argue that the most valuable animal models should provide
tensive drugs as prescribed, the prevalence of treatment-resis- insights into prevention and treatment of these complications.
tant hypertension is estimated to be in the range of 10% to Because the pathogenesis of human hypertension remains
15%.20 Although this may well be an overestimate, many indi- obscure, attempts to generalize study observations from a
viduals could still benefit from the availability of new thera- single animal model to the human circumstance should be
pies, particularly because this group of patients is at high risk viewed with considerable skepticism. Ideally, studies in mul-
for complications of hypertension. Development of successful tiple models may be most helpful in providing a more com-
vaccines or device-based therapies could be particularly help- plete view of the potential clinical relevance of mechanisms
ful for improving BP control in patients whose BPs cannot be and therapeutic responses observed in experimental studies of
controlled with conventional therapies, in those who are not hypertension and in stimulating hypotheses about responses
adherent to antihypertensive treatments, or in those prescribed in subsets of humans with hypertension. Nevertheless, stud-
suboptimal therapies. It is also conceivable that new antihy- ies in animal models of hypertension have successfully tested
pertensive therapies might reduce the sizeable cardiovascular important hypotheses relevant to human hypertension and
risk that persists in treated patients with hypertension with have motivated clinical research studies, leading to significant
seemingly good BP control and other determinants of cardio- improvements in clinical management and outcomes such as
vascular disease. Although traditional antihypertensive agents key applications of angiotensin-converting enzyme inhibi-
Downloaded from http://ahajournals.org by on March 14, 2019

do not necessarily completely abolish the cardiovascular risk tors,24,25 dihydropyridine calcium channel blockers, angio-
of the treated patient with hypertension,21 further research is tensin receptor blockers, and renin inhibitors to hypertension
required to determine the extent to which such residual risk treatment. The history of the development of renin inhibitors
is related to unrecognized inadequate BP control or to some illustrates both the value and potential pitfalls of using ani-
underlying mechanism of hypertension that is conferring mal models of hypertension to predict BP-lowering effects of
increased cardiovascular risk beyond just effects of elevated new molecules in humans with hypertension. Because of spe-
BP. Although better approaches addressing the issue of sub- cies differences in drug pharmacokinetics and in the amino
optimal BP control are required, important unmet needs exist acid sequence of renin, especially in rodents, results in both
for developing new avenues for treating hypertension and its nonprimate and primate models were critical for defining
complications.19–22 Animal models may indeed be useful for applications of these agents in humans.26–28 Overall, transla-
addressing these unmet needs. tional research in animal models of hypertension has largely
been a success story in modern medicine. We suggest that
Predictive Value of Animal Models for Improving judicious use of such models will continue to guide successful
Management of Hypertension identification and advancement of interventions.
The utility of animal models for developing better approaches
to the prevention and treatment of human disease has been Large Versus Small Animal Models of
controversial. A major concern is the poor success rate for Hypertension
new drugs advanced to clinical trials on the basis of preclinical In the selection of the most appropriate model of hyperten-
studies in animal models.9 On the other hand, animal models sion, one of the first decisions facing researchers is the choice
have a verified track record in some human disorders, includ- between small and large animal models. Several factors must
ing hypertension, in which all clinically effective antihyper- be considered, including the research scope and objectives,
tensive drugs lower BP.9 In this regard, all major classes of institutional resources, experimental cost, animal welfare, and
antihypertensive drugs in use today have been demonstrated practical suitability. The pros and cons of these models need
to substantially reduce BP in ≥1 of the most commonly used to be thoroughly evaluated to select the best model to meet a
animal models of hypertension (SHR, DSS rat, renal artery particular research purpose.
stenosis, mineralocorticoid-salt model).10,22 For example, Small animal models are most commonly used to study
the SHR responds to the antihypertensive effects of almost hypertension, providing useful insights. For example, these
all classes of drugs approved for treatment of hypertension. models may target specific factors implicated in the pathogen-
Because hypertension is a multifactorial heterogeneous esis of human hypertension, including salt sensitivity, activa-
disorder and because pharmacokinetic/pharmacodynamic tion of the renin-angiotensin-aldosterone system (RAAS),
e4  Hypertension   TBD 2019

and genetic factors. Rats and mice offer several advantages Ang II, glucocorticoids, or DOCA (with and without high-salt
over larger animal models, such as cost-effectiveness, a short diet) in pigs or dogs30–33 are less frequently used than in smaller
gestation period, and tractability for genetic manipulation. animals, partly because of the high cost of body size–titrated
However, reliable measurement of BP is challenging in small doses of drugs required over prolonged periods of time. On
animals, surgical procedures are technically difficult, and the the other hand, surgical induction of hypertension is relatively
amount of sample available, particularly plasma and urine, simple, widely used, and well tolerated and carries a low risk
may be limiting. Nevertheless, recent advances in imaging for surgery-related mortality. These interventions include con-
and surgical interventions have addressed some of these striction of the aorta by extravascular banding,34 implantable
issues and have greatly streamlined the assessment of target- adjustable occluders in the suprarenal aorta35,36 or renal arter-
organ damage.29 ies,37,38 or intravascular devices in the renal arteries.39 These
One of the most significant advantages of rodent mod- methods provide reliable models of chronic hypertension pri-
els is the ready availability of techniques for precise genetic marily of renovascular origin. The use of adjustable occluders
alterations through whole-body or cell-specific gene deletions to restrict blood flow allows the degree of insult leading to
(knockout) or gene editing, allowing mechanistic studies to hypertension to be controlled, which provides opportunities
elucidate molecular mechanisms and to identify novel tar- for determining the extent of BP elevation required to trigger
gets for therapy, which are further enhanced by the relatively target-organ injury and understanding how the process of end-
larger availability of specific antibodies for molecular studies organ damage unfolds. Intra-arterial devices such as coils39,40
compared with large animals. One great advantage of the rat that induce a progressive narrowing of the renal arterial lumen
is the existence of numerous genetic strains exhibiting robust may mirror the obstructive role of plaques in human renal
spontaneous hypertensive phenotypes at baseline or through artery stenosis and thus more closely recapitulate the patho-
induction by environmental conditions. In addition, the rat is physiology of this well-documented clinical condition. Unlike
easy and cost-effective to maintain, house, and breed yet large adjustable occluders, the resulting degree of obstruction and
enough for most analytical studies, including long-term stud- target-organ injury achieved by intravascular devices is often
ies, dynamic cardiovascular monitoring, and blood and tissue variable, again mimicking the clinical course of disease devel-
sampling. Because these rat models exhibit many phenotypic opment. Finally, recent data show the potential of the African
characteristics observed in human hypertension, they have green monkey as a model of spontaneous hypertension.41
been widely used to examine both the genetic and mechanistic Hypertension in this model seems to develop without the need
bases of hypertension. In recent years, many of these physi- of external interventions, exacerbates with aging, and is asso-
ological monitoring techniques have been adapted to mice, ciated with target-organ injury, which may offer a new avenue
Downloaded from http://ahajournals.org by on March 14, 2019

which bear lower experimental and maintenance costs com- for translational hypertension research.
pared with rats. In addition, a very wide range of specific anti-
bodies are available commercially for mice that can be used Platforms of Experimental Hypertension:
for monitoring and in vivo treatment studies. Genetic
Major advantages of large animal models such as pig and
primate are their anatomic, physiological, and hemodynamic Genetic Rat Models of Hypertension
similarities to humans, combined with developmental patho- The complex nature of hypertensive phenotypes in combina-
physiology in general and hypertension specifically, which tion with the polygenic mode of inheritance of hypertension
may also more closely resemble humans compared with small requires appropriate models amenable to study. Great insight
animal models. They are also particularly suitable for linear has been gained from genetic studies of human hyperten-
studies of hemodynamic consequences of long-term elevation sion and from mechanistic studies in experimental animal
of BP, with the added advantage of opportunities for repeated models of hypertension, summarized in other sections in this
sampling of plasma and abundant tissues in which to quan- statement.
tify and often follow functional and structural injury in target A number of rat genetic models of hypertension have been
organs in vivo and ex vivo. Hence, integrated longitudinal data used in genetic, (patho)physiological, and pharmacological
may be obtained in the same animal. A major disadvantage, studies.42–46 Rat strains exhibiting genetic hypertension include
however, is limited availability of genetically modified large the SHR, DSS rat, the fawn-hooded hypertensive (FHH) rat,
animal models of hypertension compared with the breadth the Milan hypertensive strain, the Lyon hypertensive rat, the
of genetically modified rodents. This is related largely to the Sabra hypertensive rat, the genetically hypertensive rat, and
higher costs of maintenance, longer reproductive cycles, and the inherited stress-induced arterial hypertension rat model.
labor-intensive experiments in large animals. Along with the Of these, the most commonly studied is the SHR. In the past
lack of other reagents such as specific antibodies, this restricts 10 years, >4500 articles were indexed in PubMed (https://
the mechanistic depth of some studies using large animals. www.ncbi.nlm.nih.gov/pubmed/) under the term spontane-
Finally, ethical issues have been raised for studies using non- ously hypertensive rats. In contrast, the next most commonly
human primates. cited model, the DSS rat, was indexed 585 times over the same
The most frequently used large animals for hyperten- time span, and the other genetic rat strains were indexed less
sion studies are swine, nonhuman primates, sheep, and, to frequently. In addition to the above-described inbred strains,
a declining extent, dogs. The means to induce hypertension a number of congenic and transgenic animals exhibit hyper-
generally require pharmacological or surgical approaches. tensive phenotypes. Although too numerous to conveniently
Pharmacological interventions using long-term infusions of list in this space, these other strains are commonly based on
Lerman et al   Animal Models of Hypertension   e5

the genetic background of the major strains listed above. The multiple generations to establish a strain that spontaneously
following sections provide a brief overview of the origin of develops hypertension soon after weaning and plateaus at 7
a number of these strains, their general experimental applica- to 8 weeks of age. Milan hypertensive strain rats have been
tions, considerations for choosing a rat genetic model of hyper- used to study primary hypertension. The Milan normotensive
tension, and the advantages and limitations of rat models. strain has also recently proved useful for investigating genetic
mechanisms mediating impaired myogenic responses and sus-
Commonly Used Strains
ceptibility to development of proteinuria and renal injury.56
The majority of the genetically hypertensive rats have been
derived from outbred Wistar or Sprague-Dawley (SD) breed- Lyon Hypertensive Rats
ing stock with selection for hypertension-related traits. These In the late 1960s, a group in France selected outbred SD rats
models provide reliable and reproducible phenotypes that are for elevated, normal, or decreased BP that were subsequently
often representative of clinical observations. The severity of inbred for multiple generations into strains with low, normal,
hypertension and of related phenotypes is different among or high BP.57,58 A separation of BP between the strains is evi-
strains and can be a consideration when an animal is chosen dent at a relatively early age (5 weeks) and is subsequently
for study; a number of reviews and resources provide compre- sustained. These strains have been used for renal, metabolic,
hensive information.42–46 Moreover, direct comparisons of the autonomic, cardiac, and genetic studies.59
hypertensive phenotypes and general body characteristics of
several of the genetic strains provide insight into the different Sabra Hypertensive Rats
degrees of disease attained under similar conditions. Sabra hypertensive rats were originally derived from Sabra
outbred rats by brother-sister mating and selection for high
SHR Strain BP after unilateral nephrectomy and treatment with DOCA
The SHR strain originated in Kyoto, Japan, from the cross of and dietary sodium chloride.60 A secondary round of inbreed-
an outbred Wistar male rat, which exhibited spontaneously ele- ing more recently rederived the Sabra hypertension-prone
vated BP, and a female with slightly elevated BP.47 Subsequent and -resistant rat models, which are also sensitive or resistant
brother-sister mating was continued with selection for animals to DOCA-salt treatment.62 The Sabra hypertensive rats have
with systolic BP >150 mm Hg. The inbred strain was sub- therefore been useful for genetic studies examining environ-
sequently established in the United States in the late 1960s mental interactions on BP.
after 20 generations of inbreeding at the National Institutes
Experimental Use And Considerations
of Health48 and spontaneously develops hypertension as adult
The most distinct advantage of the rat genetic models of hyper-
animals. The SHR is widely used in different studies as a rat
Downloaded from http://ahajournals.org by on March 14, 2019

tension is the similarities of the BP/hypertension phenotypes


model of primary or essential hypertension. This strain or sub-
to those observed in patients and the genetic basis of disease
strains such as the stroke-prone SHR49,50 have proved useful
development that occurs in these animals. In addition, because
in studies of stroke, vascular function, autonomic regulation,
of the relatively low costs associated with maintenance of rat
renal function, therapeutic interventions, and the genetics of
colonies, the ease of experimental studies, and their relatively
primary hypertension.
rapid growth and reproductive rate, the genetic rat models
DSS Rats have been popular for experimental studies of hypertension.
DSS rats were developed by Lewis Dahl, who observed
Genetics of Hypertension
the beneficial effects of low sodium–containing diets in the
The inbred genetic rats have served as useful model sys-
1950s51 and examined the influence of different salt diets on
tems to identify BP quantitative trait loci (QTL), regions on
BP in outbred SD rats. Selective breeding of those rats fed
the genome that contribute to the elevation of BP. With the
high salt that developed hypertension led to the inbred DSS
development and application of polymorphic microsatellite
rats, which are often used in experiments examining the kid-
markers to identify BP QTL, linkage-mapping approaches
ney, vasculature, and genetics in hypertension.51 Selective
were extensively used to study inheritance of hypertension in
breeding of rats resistant to salt-sensitive hypertension led to
rat models. Subsequent studies to validate these QTL were
the inbred Dahl salt-resistant rats.
performed through the generation of congenic or consomic
FHH Rat strains.46,63 The subsequent identification of genes within these
The FHH model was derived by inbreeding the outbred fawn- QTL has depended on complementary approaches, including
hooded (or fawn-headed) rat originally characterized for its transcriptomic analyses and gene-sequencing and gene-edit-
bleeding disorder.52 The outbred rats were demonstrated ing approaches. Perhaps the ultimate question related to the
to have an elevated mean arterial pressure compared with use of rat genetic models is the applicability of findings to
Wistar rats and were subsequently inbred by Provoost and human hypertension. Encouragingly, comparative mapping
De Keijzer53 to produce 2 strains designated the hypertensive strategies have identified overlap of QTLs detected in rat and
FHH and the normotensive fawn-hooded (also known as FHL) human linkage studies,64 indicating that findings in rat models
rat. The FHH rat has been useful in addressing the genetics of of hypertension may be translatable to human disease.
hypertension and chronic kidney disease.
Phenotypes and Mechanism of Hypertension
Milan Hypertensive Strain In the past 10 years, a major technological breakthrough
The Milan hypertensive strain rats were derived from Wistar occurred with the use of gene-editing technology to manip-
rats observed to have elevated BP54,55; the rats were inbred for ulate the rat genome. The zinc finger nuclease (ZFN)
e6  Hypertension   TBD 2019

methodology for gene manipulation, pioneered by Geurts et (and differences) between these species. The Millennium
al,65 has permitted assessment of function of genes identified Genome Project for Hypertension73 stated as its aim the iden-
in human association studies in animals with a hypertensive tification of hypertension-susceptibility genes and pathways
genetic background.45,66 This approach has permitted elucida- by a systemic multiple candidate gene approach. Candidate
tion of the function of a number of genes that are associated genes identified as possible contributors by GWAS screening
with hypertension in human genome-wide association studies can be interrogated in animal models with transgenesis, that
(GWASs). Of note, the functional importance of various gene is, the stable introduction of modifications into the genome.
products with previously unrecognized function in the regula- Gene addition, resulting in overexpression of a given
tion of BP (eg, an adherens junction protein, immune signal- gene of interest, was classically achieved by microinjection
ing proteins, a secreted metalloproteinase) has been revealed of DNA into the single-cell embryo and monitoring of its
through the use of this technology in genetically hypertensive subsequent expression and phenotypic effect in the mouse74,75
rats. Similar studies are also underway using the more recently and subsequently in rats and other species. Alternatively, the
developed methods of gene manipulation involving clustered use of bacterial artificial chromosomes, which incorporate
regularly interspaced short palindromic repeat (CRISPR)/Cas hundreds of kilobases of DNA, human chromagranin vari-
system–based technologies. ants have been analyzed in mouse models, for their ability
to reduce the risk of hypertension.76,77 Gene targeting, result-
Considerations ing in loss of gene function, was also achieved initially in the
Important considerations to take into account with the use mouse after the development of embryonic stem cells (ESCs).
of genetic rat models include the choice of the appropriate The gene-targeting construct is introduced by transfection
model for a particular research question, the need to ensure or electroporation into ESCs, and those cells that have been
an appropriate genetic background of the experimental and correctly targeted by homologous recombination are selected
control rats, and the requirement for a controlled environ- and injected into blastocysts. The resultant chimeras are then
ment. It should be noted that the basic genetic architecture of bred, and their progenies are screened for incorporation of the
the SHR, and of other inbred strains in which the animals are genetic modification.
homozygous at virtually all loci, is quite different from the ESC technology led to the supremacy of the mouse for
genetic architecture of humans. The various genetic rat strains generating new animal modeling of human disease over the
can be used as models of slowly developing primary hyper- following 25 years. Many developments, improvements,
tension,48,67,68 juvenile hypertension,69,70 salt-induced hyperten- and refinements in gene targeting during this time allowed
sion,51 and hypertension associated with end-organ damage.56 the researcher to introduce precise and ever more ingenious
Downloaded from http://ahajournals.org by on March 14, 2019

However, understanding the genetic composition of the hyper- genetic changes (Table 1).
tensive strain and choosing an appropriate control strain are Gene expression cassettes may be targeted to safe havens
important.66 A direct comparison of 2 strains of commonly such as the ROSA26 locus.110,111 Fluorescent reporters,
used DSS rats demonstrated >1.3 million different base pairs expressed in place of the gene of interest but under its endog-
(bps) between the strains and a significant difference in BP enous promoter, highlight the tissue- or development-specific
when the rats were fed a high-salt diet.45 Genetic differences pattern of expression. In some cases, knockout of a gene may
between control strains must also be considered. Kurtz and prove to be embryonic lethal because it contributes to a criti-
Morris48 demonstrated that the Wistar Kyoto rat, a common cal stage in development. By flanking key exons of the gene
genetic control for the SHR, had profound differences in phe- with loxP sites and crossing the transgenic animal with ani-
notypes (growth rate and BP) when obtained from different mals that express the enzyme Cre recombinase in a tissue- or
commercial suppliers. Moreover, all control strains are neces- development-specific manner, investigators can knock out the
sarily limited in the absence of a complete understanding of gene later in development. Alternatively, transgene expression
the genetic differences between the control and hypertensive may be driven by an inducible promoter, so its expression is
strains. These examples emphasize the importance of careful under the control of the researcher.
identification of and consistent use of strains when performing Before 2008, gene knockout in the rat or rabbit relied on
experiments. Equally important are environmental influences random mutagenesis with N-ethyl-N-nitrosourea, transposon-
on phenotypes. In inbred DSS rats, a simple substitution of based systems such as Sleeping Beauty, or spermatagonial
the sodium-independent components of the diet could pro- stem cell targeting. After identification of the signaling path-
foundly alter the salt-sensitive hypertension and renal damage ways that control self-renewal and differentiation of ESCs, it
phenotype.71 was discovered that incorporation of certain inhibitors in rat
In summary, genetic rat models have demonstrated great ESC growth medium was sufficient to maintain them in the
utility and provided exceptional insight into the genetics and self-renewal, pluripotent state,112,113 and a variety of species
pathophysiology of hypertension. A researcher interested in are now targetable through the ESC route. Hence, all the tech-
using such models should carefully consider the disease traits niques developed in mice are potentially applicable to other
of the different strains and appropriate genetic controls and species such as APOE knockout in rabbits.103
pay careful attention to controlling the environment. More recently, generation of transgenic animals has been
transformed by the introduction of gene-editing technologies
Transgenic Models of Hypertension using sequence-specific nucleases. The first of these, ZFN
The construction of high-density comparative genetic maps targeting, requires 2 ZFNs to bind upstream and downstream
between mouse, rat, and human72 reinforces the similarities of the target site, eventuating in a double-strand break at the
Lerman et al   Animal Models of Hypertension   e7

Table 1.  Examples of Transgenic Techniques Related to Hypertensive Models phosphodiesterase 1A–null mouse lines, which revealed a role
Transgenic for phosphodiesterase 1 in BP regulation in addition to renal
Technique Species Examples pathogenesis.105
The third and most readily available nuclease class is the
Gene overexpression M Mendelian models of hypertension
CRISPR/Cas9 system derived from the bacterial immune sys-
M/R Salt-sensitive hypertension78,79 tem of Streptococcus pyogenes. Although the CRISPR/Cas9
Inducible expression R Inducible hypertension and end- system is susceptible to off-target events, it has proved its
organ damage80 worth in generating knockout models in a wide range of spe-
BAC incorporation M Reduced risk of hypertension76,77 cies. Of note is the recent CRISPR/Cas9–targeted knockout
and subsequent knock-in of a 19-bp indel polymorphism in
Global gene knockout M Mendelian models of
hypertension81–85 a rat long noncoding RNA (Rffl-inc1).106 Recent advances in
CRISPR technology include the generation of Cas9 deriva-
M/R Salt-sensitive hypertension78,86–90
tives with increased specificity117,118 and altered protospacer
M Pulmonary hypertension91 adjacent motif recognition sequences119 and the development
Renin hypotension92–94 of other CRISPRs from alternative bacterial sources.120 The
Gene knock-in M/R Renin95; RAS humanization96,97 versatility of nucleases extends to the generation of large-
Targeted gene M Kidney-specific Hsd11b2 knockout88 scale deletions98 and insertions,99 which allows the humaniza-
knockout tion of animal models.
Gene reduction or knockout has also been achieved with
Safe-haven targeting R ROSA2698,99
siRNAs (small interfering RNAs). To prevent rapid degrada-
Conditional knockout M Pulmonary hypertension100,101 tion in vivo, miRNA (microRNA) mimics or anti-miR oligo-
Rat/rabbit ESCs R TALEN targeting102 nucleotides require chemical modifications such as locked
Rb ApoE knockout103 nucleic acid modifications. Locked nucleic acid–modified
anti-miRs proved to be effective in targeting miR-29b, which
ZFNs R Salt-sensitive hypertension104
affects collagen gene expression in the renal medulla in DSS
TALENs M/R Pde1a105 rats.109 Genetic modification can be achieved with recombi-
CRISPR/Cas9 R Humanization and ROSA2698,99,106 nant lentiviral or adeno-associated virus–mediated delivery.
Adverse reactions to the delivery vehicle need to be addressed,
Gene knockdown/ M/R AT1A receptor107; reduced
anti-miRs/siRNA hypertension in aldosterone/ in addition to target specificity and cellular uptake.121
Downloaded from http://ahajournals.org by on March 14, 2019

salt–treated mice108; reduced Cell ablation is a transgenic technique with numerous


hypertension in SHRs109 potential applications. Classically, diphtheria toxin A was
ApoE indicates apolipoprotein E; AT1A, angiotensin receptor 1A; BAC, bacterial used to achieve cell ablation.122 More recently, the nitro-
artificial chromosomes; CRISPR, clustered regularly interspaced short palindromic reductase gene, together with pharmacological treatment with
repeat; ESC, embryonic stem cell; Hsd11b2, hydroxysteroid dehydrogenase 11b2; a prodrug, Metronidazole, or KillerRed, a far-red fluorescent
M, mouse; miR, microRNA; Pde1a, phosphodiesterase 1a; R, rat; RAS, renin- protein, which is phototoxic to the cell on exposure to appro-
angiotensin system; Rb, rabbit; SHR, spontaneously hypertensive rat; siRNA, priate laser light, has been used.123 Optogenetic cell ablation
small interfering RNA; TALEN, transcription activator–like effector nuclease; and
ZFN, zinc finger nuclease.
can be tightly controlled both spatially and temporally, allow-
ing single cells to be ablated.124

target site. The cell repairs the break, but it often results in the Application of Transgenesis to Hypertension Research
introduction of small deletions or insertions. If the target site The utilities of a transgenic model for investing the role of
is within an exon, then such changes may lead to missense a specific gene in hypertension might include (1) elucidat-
or nonsense mutations, effectively knocking out the target ing its basic function, (2) mechanistically understanding its
gene.114 Alternatively, a single-strand oligo or plasmid can involvement in a particular pathway, (3) determining its con-
be added to direct a desired sequence alteration or insertion tribution to primary hypertension, and (4) faithfully modeling
at the target site. Geurts et al,65 Geurts and Moreno,115 and the pathogenesis of human hypertension, with a view toward
Jacob et al116 used ZFNs (and subsequently CRISPR/Cas9) identifying new therapeutic targets and drug treatments.
to systematically knock out a number of GWAS candidate Many candidate genes for human hypertension identified
cardiovascular disease–related genes104 through the PhysGen by GWASs have been individually found to make only small
Knockout program. contributions to BP (<1 mm Hg), suggesting that there are no
Transcription activator–like effector nucleases are a sec- common gene variants with major effects to promote primary
ond class of nucleases that also bind upstream and down- hypertension.125–127 Although considerable basic knowledge
stream of the target site and require the close proximity of about the complexities of homeostatic BP control has been
FokI monomers to introduce a double-strand break. Unlike gleaned from these studies, the lack of clinically relevant
ZFNs, transcription activator–like effector nucleases targeting therapeutic targets emerging from this work is disappointing.
a specific sequence can be readily constructed from libraries of Recently, a single nucleotide polymorphism in the third intron
domains, each of which recognizes 1 nucleotide, making them of the PHACTR1 gene, identified through GWASs, has been
widely accessible and relatively cheap. Recently, transcrip- shown to enhance ET-1 (endothelin 1) expression in the vas-
tion activator–like effector nucleases were used to generate 2 culature.128 This is an example of a common noncoding variant
e8  Hypertension   TBD 2019

contributing to vascular disease and hypertension. Similarly, allowed identification of angiotensin-converting enzyme
Ji et al129 found that rare gene variants in 3 genes, Na+-K+- as a modifier of end-organ damage.131 The model has been
2Cl− cotransporter (NKCC)-2, ROMK and NCC, causing rare improved further by placing expression of the mouse renin
mendelian syndromes with recessive inheritance, are surpris- gene under an inducible (Cyp1a1) promoter.80 The timing and
ingly common, with 1 in 64 subjects in the Framingham Heart severity of hypertension in the Cyp1a1(Ren2) transgenic rat
Study cohort carrying such a mutation.129 Such rare variants are thus under the control of the researcher.
cannot be detected by GWASs, which suggests that whole- The action of Ang II in the proximal tubule was deter-
genome sequencing may be a more fruitful source of rare gene mined in angiotensin receptor (AT1A)–deficient mice. Kidney
variants or epigenetic modifications that potentially contrib- expression of AT1A is necessary for Ang II–dependent hyper-
ute to individual propensity for hypertension. Relevant trans- tension, and specific removal79,95 or overexpression79 in proxi-
genic models might then be designed to confirm the effect of mal tubules causes hypotension or hypertension, respectively,
such variants on BP and to provide a valuable test bed for on normal-salt diet. Meanwhile, animals lacking all 3 Ang
therapeutic development. Although the clinical relevance of a II receptors reveal that Ang II controls BP by acting solely
particular model is an important consideration, advancing our through these receptors.132 Humanization of both rats and
understanding of basic mechanisms of hypertension is also of mice with human renin and angiotensin genes96,97 has been
value and not incompatible with long-term translational goals. useful for establishing species specificity of the RAAS, but
With the abundance of gene-targeting tools, particu- more important, such transgenics are useful for investigat-
larly for the mouse, substantial basic understanding of rare ing the function of different haplotypes and species-specific
mendelian disorders associated with altered BP and renal RAAS inhibition.
sodium handling has been gained. A mouse model of Liddle Conditional knockouts have been used to investigate the
syndrome2 containing a gain-of-function stop codon in roles of ET-1 and its receptor, ETA (endothelin A). Specific
β-epithelial sodium channel (reflecting the human mutation) knockout of ET-1 in the collecting duct causes hypertension
exhibits normal BP unless placed on a high-salt diet, which and sodium retention,100 whereas knockout of the ETA receptor
causes mice to develop hypertension and hypokalemia. The in collecting duct prevents receptor antagonist–related fluid
hypervolemia suggested by increased sodium reabsorption retention.101
and low aldosterone on normal-salt diet, together with salt Gene knockdown is a potential therapeutic strategy. When
sensitivity, replicates the human syndrome. The basic mech- adenovirus was used to direct anti-miR against the AT1A recep-
anisms of positive modulators of epithelial sodium channel tor to the paraventricular nucleus of SHR, hypertension was
trafficking or expression such as Sgk1 and Af17 and negative attenuated.107 Likewise, siRNA against NADPH (nicotinamide
Downloaded from http://ahajournals.org by on March 14, 2019

modifiers such as Nedd4-2 have all been revealed with respec- adenine dinucleotide phosphate) oxidase (Nox) 2 or Nox4
tive animal knockout models. has been used to attenuate BP in the aldosterone-salt mouse
Mice lacking NKCC2, which models Bartter syndrome, model.108 Transgenic technology has been used to ascertain the
die of dehydration before weaning because of uncompensated causative genes in multiple models of primary and salt-sensi-
polyuria.85 Bartter syndrome can also be modeled by knock- tive hypertension. For example, knocking out either Nox4 or
out of ROMK,84 with animals exhibiting hypotension. NCC the subunit p67phox on the DSS background significantly ame-
knockout, despite recapitulating features of Gitelman syn- liorates both salt sensitivity and albuminuria.89,90 An extensive
drome such as hypocalciuria, causes no reduction in BP unless list of transgenic models that exhibit salt-sensitive hyperten-
the animals are put on a low-salt diet.83 Renal knockout of sion is given in a recent American Heart Association review.78
Nedd4-282 leads to salt-sensitive hypertension with increased The size of the mouse can sometimes preclude its use in
NCC phosphorylation. NCC is also activated in Gordon syn- studies in which substantial or multiple sampling is required.
drome through mutations in WNK4,81 leading to hypertension Although surgical instrumentation is routine (eg, telemetric
and hyperkalemia. devices), there may be time constraints in which, for example,
Rare conditions such as the syndrome of apparent min- battery life is limiting. Historically, the rat has been the model
eralocorticoid excess, in which Hsd (hydroxysteroid dehy- of choice in the pharmaceutical industry, so there is a wealth
drogenase) 11b2 deficiency allows the mineralocorticoid of physiological knowledge associated with this species.133
receptor to be activated illicitly by glucocorticoids, have been With the development of protocols for ESC isolation and gene
modeled in both mouse86–88 and rat114 models. The global editing, rats, rabbits, and larger animals are now more likely to
mouse knockout87 replicates the human syndrome with be considered useful genetic model systems. With the rapidly
hypertension and hypokalemia, and heterozygous animals expanding range of tissue and spatiotemporal controls that can
exhibit salt sensitivity.86 Recently, kidney-specific knockout be placed on the genes of interest and with the possibility for
of mouse Hsd11b2 was found to be sufficient to cause salt- multiple targeting or humanization, animal models can now
sensitive hypertension attributed to epithelial sodium channel be designed to answer ever more complex questions relating
and NCC activation.88 to human disease.
Renin knockout in both mouse92,94 and rat93 results in sig- There are caveats to the use of transgenic modeling, and
nificant hypotension. Knockout of the duplicated renin gene, these caveats must be considered on a case-by-case basis.
Ren2, has no effect on BP in the mouse,94 but its overexpres- Vector sequences may cause anomalous expression of a
sion in the SD rat leads to extreme hypertension.130 Subsequent reporter, as reported in the vasculature of fli1:EGFP (enhanced
crossing of the transgene onto inbred strains of rat that are sus- green fluorescent protein) zebrafish.134 The pharmacologi-
ceptible (F344) or resistant (Lewis) to malignant hypertension cal profiles of the mouse and rat may differ from that of the
Lerman et al   Animal Models of Hypertension   e9

human, exemplified by the TRPA1 channel.135 Homologs to BP.144 The swine model also demonstrates spontaneous hyper-
human genes may be absent in animal models or vice versa. tension, resulting from a neurogenic mechanism potentially
Finally, there may also be species-specific differences in related to increased renal sympathetic outflow. However,
genetic mechanisms of disease progression or end-organ unlike nonhuman primate models, minipigs do not typically
damage.136 have elevated heart rates.
The optically clear zebrafish has been very successful in A nonhuman primate model of systemic hypertension
informing basic understanding of organ development or func- was first reported in a small number of individual African
tion.137,138 The advantage of ex vivo development, together green monkeys or vervets.145 These findings have recently
with optical clarity, facilitates the use of unparalleled optical been extensively expanded through phenotyping of nearly
and imaging techniques such as selective plane illumination 400 animals by forearm plethysmography.41 In this model,
microscopy, optogenetics, and optical ablation techniques. the mechanism is likely to be neurological in origin on the
Although zebrafish is a relatively new species in cardiovascu- basis of the direct correlation between BP and elevated heart
lar science, its use in studies of heart development and func- rate. Although RAAS components were unaltered, these
tion and the cardiorenal axis, combined with its facile genetic monkeys developed significant renal pathology in glomeruli
tractability and utility for high-throughput screening of drugs and small vessels, resembling humans with primary hyper-
and small molecules, positions it as a likely important species tension. The vervet genome has now been fully sequenced
in future translational studies. and a basic annotation has been published,146 so defining the
In summary, transgenic technology affords a great deal of genomic mechanisms underlying the development of spon-
scope for the generation of informative animal models, from taneous hypertension may be possible. Accordingly, this
identification of gene function to humanization. Single nucle- nonhuman primate model may have substantial utility for
otide polymorphisms, suspected of being disease related, can translational research on the genetic basis of human primary
be investigated in vivo, and potential therapeutic strategies can hypertension.
be tested. In addition, with the latest developments in stem cell
technology and genome editing, the choice of species is no Platforms of Experimental Hypertension:
longer limited to small rodents. Induced
Large Animal Genetic Models Renovascular
Large animal models of spontaneous hypertension have been The pioneering work by Goldblatt et al37 and later Page147 in
recognized and developed over the past 50 years. Spontaneous the 1930s planted the seed for the development of surgically
Downloaded from http://ahajournals.org by on March 14, 2019

(or primary) elevation of BP has been reported in a variety of induced models of hypertension and opened a new chapter for
large animal species, including chickens, turkeys,139 rabbits,140 hypertension research. Their work positioned the kidney and
swine,141 dogs,142 and nonhuman primates.41 In the avian spe- renal arteries on the complex map of the pathophysiology of
cies, elevated BP likely does not represent true hypertension hypertension and disclosed the unique relationship between
because BP differences are characteristic of comparisons with the kidney and BP control that also propelled the develop-
other species rather than intraspecies variation. Other large ment of various therapeutic interventions. Surgical induc-
animal models show intraspecies variation in BP by individu- tion of hypertension by reducing blood flow in the suprarenal
als, indicative of hypertension and similar to that observed aorta or main renal arteries with and without removal of renal
in humans. Those species all exhibit clear genetic correlates mass, inducing compression of the renal parenchyma, sub-
among individuals with strong degrees of heritability. Similar total nephrectomy, or sinoaortic baroreceptor denervation is
to the SHR, selective breeding of New Zealand and Dutch applicable to both small and large animal models with similar
white rabbits generated one of the earliest large animal mod- outcomes, underscoring the pathophysiological mechanisms
els of hypertension.140 Hypertensive animals tended to exhibit of hypertension that likely are conserved across species.
varying degrees of renal pathologies, possibly reflecting the A common pathophysiological feature shared by models
cause or consequence of the hypertension. of renovascular hypertension is the driver of hypertension and
Large animal models of spontaneous hypertension in dogs target-organ damage: a reduction in blood flow to the kidneys,
and swine exhibit significant elevation in BP that is spon- resulting in decreased perfusion pressure and activating the
taneous and may be expanded by selective breeding. Both RAAS, leading to vasoconstriction and salt and water reten-
dog142,143 and pig141 models of spontaneous hypertension also tion, with systemic hypertension developing in a matter of
have LVH, one of the most common cardiovascular compli- days. Systemic hypertension induces progressive endothelial
cations of human hypertension portending significant risk for dysfunction, stretch, and organ damage, whereas long-term
morbidity and mortality. The canine model was derived from reduction of blood flow to the kidneys leads to tissue isch-
diagnosed primary hypertension in a female Siberian husky emia and subsequent release of hypoxia-activated factors and
selectively bred over a 5-year period to normotensive males. oxidative stress that activate inflammation. These processes
The offspring produced a colony of hypertensive animals in turn induce microvascular remodeling, fibrosis, and loss of
with dominant heritability. Similar to the rabbit model, sub- renal function, which likely play a dual role by both maintain-
sets of Guizhou minipigs and Sichuan domestic pigs exhibit ing hypertension and promoting target-organ injury.
renal disease with mild renal fibrosis, which may be partly Alternative approaches to surgically induced hyper-
responsible for hypertension.141 In this spontaneous model of tension, although less widely used, include direct dam-
systemic hypertension, bilateral renal denervation normalized age of the renal parenchyma through compression,148 renal
e10  Hypertension   TBD 2019

microembolization,149 and ureteral obstruction,150,151 which is long-term subcutaneous infusion of Ang II. The utility
often also activate the RAAS and elicit inflammation, renal of this model accrues in part from resembling some forms
fibrosis, and subsequent loss of renal function. A potential of human hypertension. The RAAS is broadly activated in
limitation of these approaches is a difficulty in controlling the human primary hypertension, and the level of BP elevation
degree of renal damage; thus, the development and severity of achieved with commonly used doses of Ang II in mice is on
hypertension are less predictable. Another potential limitation par with that seen in uncontrolled stage 2 hypertension. After
is clinical relevance; hypertension caused by parenchymal 4 weeks of long-term Ang II infusion in susceptible rodent
compression is infrequent in humans. Nevertheless, hyperten- strains, target-organ damage is quite similar to that seen in
sion can be observed in humans with renal masses such as human patients with sustained elevations in BP, including car-
subcapsular hematomas and tumors. Parenchymal compres- diac hypertrophy, vascular remodeling, and chronic kidney
sion has also been implicated in development of hypertension disease.161–164 Nevertheless, because the renal vasoconstric-
associated with obesity,152 thus potentially conferring signifi- tion attributable to Ang II can induce ischemia, particularly at
cance for these surgical models of hypertension. higher doses, long-term Ang II infusion more closely models
the renal injury that accrues from chronic renal ischemia in
Summary: Recommendations for Using Animal human hypertension than that from barotrauma.165
Models of Renovascular Hypertension The Ang II infusion model was used in early dog stud-
Although reduction of blood flow to the kidneys and induc- ies that characterized the functions of the RAAS166,167 but
tion of parenchymal injury represent <10% of the cases of was adapted for long-term subcutaneous infusion in rodents.
clinical hypertension, target-organ injury develops in a pat- Among rodents, rats develop target-organ injury in response
tern similar to primary hypertension. However, models of to Ang II more readily than mice and can more easily be surgi-
secondary hypertension might be more suitable for assess- cally implemented with BP monitoring devices. On the other
ing target-organ injury and developing therapeutic inter- hand, mice breed more quickly, have a rapid onset of hyperten-
ventions than for understanding the pathogenesis of human sion, and are amenable to gene targeting even within selected
primary hypertension. Furthermore, the prominent involve- cell lineages. Among mouse strains, C57LB/6 mice are more
ment of the RAAS in many of these models may distinguish resistant to renal injury and are not salt sensitive, making this
secondary hypertension from some forms of low-renin strain ideal for investigating RAAS-dependent hypertension
hypertension. in the absence of these 2 conditions.168,169 In contrast, 129SVE
Large animal models of renovascular hypertension may mice are salt sensitive, are more susceptible to kidney injury,
have several advantages beyond their anatomic, physiologi- and manifest greater levels of BP elevation at similar Ang II
cal, and pathophysiological similarities to humans. For
Downloaded from http://ahajournals.org by on March 14, 2019

doses relative to C57BL/6 mice.95 Mice from the FVB strain


example, they may be more amenable than rodents for stud- develop marked injury in several compartments within the
ies in chronic stable hypertension, allowing longitudinal kidney during Ang II infusion, particularly when combined
quantification of relevant hemodynamic and biochemical with unilateral nephrectomy.170 Thus, the choices of species
parameters that characterize the progression of hyperten- and strain are key considerations in the design of experiments
sion, target-organ injury, and response to therapeutic inter- using long-term Ang II infusion.
ventions in a translational fashion.153 On the other hand, In mouse studies, several doses of Ang II have been used
targeted interventions to resolve the vascular occlusion154–158 to induce and analyze hypertension of different severities. The
are now feasible in both small and large animal models. slow-pressor dose of 400 ng·kg−1·min−1 Ang II may mimic the
Thus, both types of models might be used for teasing out the gradual onset of hypertension in humans with primary hyper-
driving force initiating hypertension, studying the poten- tension.171 The intermediate dose of 490 to 500 ng·kg−1·min−1
tial for reversibility of target-organ injury, and identifying has been the most widely used in recent years.162 A higher dose
mechanisms of organ damage that may be independent from of 1000 ng·kg−1·min−1 Ang II was used to dissect the functions
hypertension. of AT1 receptors in distinct tissue pools during hypertension
The ability to induce renovascular hypertension in geneti- and, in conjunction with other modifications, can provoke
cally modified rodents offers unique opportunities for in- measurable renal damage in susceptible strains.163 Even higher
depth elucidation of pathophysiological mechanisms, opening doses are used, albeit less commonly, to provoke cardiac
the possibility for a more comprehensive understanding of the fibrosis.172 Notably, these doses far exceed the Ang II levels
consequences of hypertension. Finally, both large and small observed naturally in humans with hypertension.
animal models of renovascular hypertension offer the pos- The duration of infusion can be adjusted on the basis
sibility of imposing other comorbidities such as metabolic of the cardiovascular control center studied. Immediate BP
derangements and determining their contributions to target- effects of Ang II can be appreciated within seconds to min-
organ damage,40,159,160 thereby realistically mimicking the clin- utes,173 whereas with long-term subcutaneous infusions of ≥
ical population of elderly patients with hypertension in whom 500 ng·kg−1·min−1 Ang II, increases in BP emerge within the
multiple cardiovascular risk factors often coexist. first 24 hours.161 Vascular remodeling resulting from Ang II
is evident within 2 weeks,162 and cardiac hypertrophy can be
Ang II–Dependent Hypertension seen at 2 to 4 weeks.161,174 Renal injury is reproducible at 4
The RAAS plays a fundamental role in normal sodium and weeks in susceptible strains163,175 but is far more robust after
water homeostasis. Accordingly, one of the most widely used 8 weeks.170 Thus, investigators should titrate the Ang II dose
preclinical models of hypertension, particularly in rodents, and study duration on the basis of the experimental question.
Lerman et al   Animal Models of Hypertension   e11

The current state-of-the-art methodology for measuring initial studies emphasized the need for extremely high DOCA
BPs during long-term Ang II infusion is radiotelemetry.176 doses, lower doses reproducibly induce hypertension; doses
With the older method of tail-cuff plethysmography, the level typically currently range from 20 to 50 mg/kg (in rats).180
of vasoconstriction with higher-dose Ang II is so profound165,169 Notably, using saline drinking solution without access to free
that extrapolating BP from blood flow velocity through the tail water imposes a nonphysiological stress on the animals that
circulation may be misleading. Radiotelemetry also permits may affect the results; furthermore, the models often become
accurate measurement of heart rate and arrhythmias. hypokalemic, and some investigators have therefore supple-
A few modifications to murine Ang II infusion can mented potassium. Although rats are most commonly used for
enhance its applicability to studies of injury and salt sensitiv- this model, it has been used successfully with many differ-
ity. Unilateral nephrectomy, high-salt diet, and an extended ent species, including mice, dogs, sheep, and pigs. The model
infusion period (8 weeks) can yield more robust renal dam- manifests low plasma renin activity (and low circulating Ang
age in mice.163,170 Because Ang II promotes sodium retention, II), so it bears some similarity to a common form of primary
a low-salt diet can be added to long-term Ang II to under- human hypertension, low-renin hypertension.
stand the extent to which the BP elevation seen during Ang II The DOCA-salt model appears to have volume-dependent,
accrues from its capacity to promote sodium retention.164 vascular, and neurogenic components. Mineralocorticoids,
dietary salt loading, and uninephrectomy would each be
Summary and Future Considerations
expected to promote increases in extracellular fluid volume,
Although an aggressive pharmacological challenge to induce
and it is thus not surprising that salt and water balances can be
hypertension has obvious limitations, in several circum-
positive, especially early in the course of mineralocorticoid-
stances, long-term Ang II infusion offers distinct advantages. salt administration. Both plasma volume and extracellular
First, it is the most direct approach for investigating the in fluid volume are expanded,181 and these effects may appear
vivo actions of angiotensin receptors and their downstream to be sustained.182 However, the extent to which a high-salt
signaling cascades. Second, pairing long-term subcutaneous diet causes greater sodium retention and volume expansion in
Ang II infusions with short-term intravenous infusions allows animals treated with deoxycorticosterone or aldosterone than
a comprehensive profile of the vascular and renal functions in nontreated salt-loaded controls remains controversial.183
of a protein during RAAS activation. Third, the model is Although the classic model of mineralocorticoid-salt hyper-
reproducible across species. Finally, long-term Ang II infu- tension typically involves expansion of blood volume and
sion engages all the cardiovascular control centers in patho- extracellular fluid volume, hypertension may occur even when
genesis, allowing in vivo assessment of interactions between prominent volume expansion does not occur, as in the setting
sympathetic and immune activation, systemic and renal vaso-
Downloaded from http://ahajournals.org by on March 14, 2019

of a normal-salt diet.182 It should be noted that administration


constriction, and renal sodium transport. Thus, long-term of mineralocorticoids to animals given a low-salt diet usually
Ang II infusion remains a useful tool to dissect coordinated has little or no effect on BP.
contributions of multiple cardiovascular control centers in Shortly after DOCA-salt hypertension was described, it
hypertension. was recognized to have a substantial neurogenic component.
DOCA-salt animals typically manifest sympathetic hyperac-
Mineralocorticoid-Salt Hypertension tivity, perhaps resulting in part from increased plasma osmo-
Administration of mineralocorticoids together with a high-salt lality, which regulates lumbar sympathetic nerve activity.
diet can induce hypertension in both large and small animals. DOCA has been postulated to sensitize the brain to salt and
In animals with a high salt intake, administration of deoxycor- osmolality.184 Sympathetic hyperactivity increases vasocon-
ticosterone, usually in the form of DOCA, has been the most striction in both arterioles and the venous circulation. The
widely used approach for inducing mineralocorticoid-salt importance of these effects has been supported by reports that
hypertension. Deoxycorticosterone appears to have both glu- lesions in the area postrema, anterolateral third ventricle, and
cocorticoid and mineralocorticoid properties, but its tendency paraventricular nucleus of the hypothalamus attenuate the
to cause sodium retention appears central to the DOCA-salt hypertension.185
model. Its mineralocorticoid potency, however, is less than Although DOCA-salt hypertension is characterized by
that of aldosterone itself. Although elevated levels of deoxy- suppressed plasma renin activity, Ang II concentrations in the
corticosterone can contribute to some rare human forms of brain may actually increase, likely contributing to sympathetic
hypertension, the most common human form of mineralocor- activation and salt and water retention through effects on brain
ticoid-dependent hypertension involves hyperaldosteronism. AT1 receptors.186 There is also evidence for a role of the pro-
Thus, the development of animal models of hyperaldosteron- renin receptor in these processes because its blockade reduces
ism is a subject of considerable scientific interest.177 Recently, BP in DOCA-salt animals.187 In contrast, intrarenal RAAS
for example, a mouse model of aldosterone-salt hypertension does not appear to play a major role. The early studies of Selye
was developed by transgenically expressing the human gene and colleagues188 indicated that DOCA-salt animals exhibited
for aldosterone synthase under control of the human promoter substantial systemic inflammation. More recently, interest in
for the gene encoding 11β hydroxylase.178 the immune contribution to hypertension generally has re-
Early studies showed that the effects of mineralocorti- emerged. Guzik and colleagues162 have shown that T cells are
coids like DOCA were greatly enhanced in animals sensitized important for the full effects of DOCA-salt to increase BP and
to its actions by a high salt intake (typically 0.6%–1% NaCl to enhance superoxide production, clearly implicating a role
in drinking water) and often by uninephrectomy.179 Although for T cells in the model.
e12  Hypertension   TBD 2019

In summary, the combination of mineralocorticoid treat- Summary and Future Considerations


ment and high-salt intake, sometimes with uninephrectomy, In the future, subtotal nephrectomy will likely remain a
provides a reliable animal model that can develop severe useful model of hypertension relevant to patients with
hypertension with some features of human low-renin hyper- advanced and progressive CKD. The salt sensitivity in
tension. Studies of such models might provide insight into the remnant kidney model seen with rats or 129SVE mice
the salutary effects of mineralocorticoid receptor blockade in enhances its relevance to human subpopulations such as
the setting of resistant hypertension, even in patients without blacks, who are prone to salt sensitivity. Abnormalities in
frank hyperaldosteronism. Thus, the mineralocorticoid-salt cardiac morphology and function such as LVH and altered
models may have broad applicability to those patients with diastolic relaxation seen with subtotal nephrectomy will
hypertension in whom controlling BP presents a challenge. provide a timely model of cardiorenal syndrome.190,192 The
Such models could also have value for understanding why glomerulosclerosis seen particularly in the rat model mim-
cardiovascular risk may be higher in humans with primary ics the secondary focal segmental glomerulosclerosis that
aldosteronism than in individuals with similar BP levels and develops after several years of uncontrolled hypertension.
hypertension of unknown origin. The gradual increase in BP in this model emulates the
BP trajectory seen in human primary hypertension more
Renoprival Hypertension smoothly than abrupt induction of hypertension via phar-
The prevalence of hypertension among patients with kid- macological perturbations. Nevertheless, hybrid models
ney disease highlights the relevance of renoprival models to of renal mass reduction are now being used to enhance
human hypertension; >90% of patients with end-stage kidney injury or to shorten the time required for readouts. For
disease are afflicted with hypertension. Preclinical renoprival example, when vigorous RAAS activation is needed, uni-
models correspond precisely to the phenotype in the nephrol- lateral nephrectomy combined with Ang II infusion yields
ogy clinic that garners concern: a patient with hypertension robust glomerular and tubular damage after 8 weeks or
who has developed chronic kidney disease. Another salient after 4 weeks when a high-salt diet is administered.163,170
feature of renoprival models is the impairment in salt excre- Alternatively, Ang II infusion combined with true subtotal
tion that accrues from reduced nephron mass, allowing one to nephrectomy in salt-resistant mouse strains like C57BL/6
study the salt sensitivity that afflicts roughly half of human can overcome resistance to hypertension in these strains.194
patients with hypertension. Thus, at institutions with skilled surgical personnel, the
Remnant kidney models were developed in rats to remnant kidney model alone or in combination with other
explore the adaptive effects of renal mass reduction on approaches will continue to yield data relevant to human
Downloaded from http://ahajournals.org by on March 14, 2019

the remaining nephrons.189 Accordingly, use of this model patients with hypertension and CKD.
revealed the detrimental consequences of glomerular
hyperfiltration and the benefits of lowering glomerular Nitric Oxide System
pressure via inhibition of the RAAS. Extrapolation from Nitric oxide (NO) is catalyzed by eNOS (endothelial NO
these rat models provided some of the rationale for the synthase), and its local release occurs on a continual basis,
use of RAAS blockers in patients with kidney disease and thereby modulating effects of local and systemic vasocon-
hypertension. Infarction of two-thirds of 1 rat kidney leads strictors and fine-tuning of BP and organ blood flow. On the
to BP elevation, whereas two-thirds nephrectomy together basis of the premise that NO inhibition would lead to pre-
with total contralateral nephrectomy, the modern subtotal dominance of vasoconstrictors and consequent increase in
nephrectomy model, yields both hypertension and glomer- BP, researchers began to explore the possibility that NO syn-
ulosclerosis. In rats, two-thirds nephrectomy is typically thase inhibition would cause hypertension in animal models.
performed on the left kidney and achieved via ligation of In 1990, Gardiner et al195 showed that short-term treatment of
the posterior branch and the inferior segment of the renal Brattelboro rats with L-NMMA (NG-monomethyl-l-arginine),
artery.190 Adaptation to mice, typically as a three-fourths a methyl derivative of arginine and NO synthase inhibitor,
nephrectomy (1 nephrectomy plus resection of half the caused an increase in BP. Ribeiro et al196 followed up on
contralateral kidney), is technically challenging but has these studies and showed that long-term treatment of Wistar
been executed with ligation of renal artery segments as in rats with the NO inhibitor L-NAME (Nω-nitro-l-arginine), a
rats191 or with direct excision of half a kidney with electro- nitro derivative of l-arginine, increased systolic BP by >60
cautery or glue used to achieve hemostasis.192 Hypertension mm Hg. L-NAME–treated rats also show renal vasoconstric-
develops universally in remnant kidney rats. In contrast, tion and hypoperfusion.195–197 As the disease progresses, it is
subtotal nephrectomy in mice induces hypertension in the characterized by renal dysfunction; renal hypertensive micro-
129SVE salt-sensitive strain but not in the C57BL/6 strain, angiopathy; cardiac, vascular, and renal fibrosis; and features
which is relatively salt resistant.191,193,194 Hypertension and of malignant hypertension. These studies defined a new model
kidney disease progress more slowly and less severely in of hypertension induced by long-term NO inhibition and pro-
this model than in more aggressive pharmacological hyper- vided the hypertension community with a robust experimen-
tension models, with studies reporting several months of tal model of severe/malignant hypertension with evidence of
data in rats and mice.193,194 In rats, tail-cuff plethysmogra- target-organ damage.
phy may be sufficient to detect a BP rise, but given the The mechanisms underlying L-NAME–induced hyper-
variability of responses in mice, radiotelemetry is recom- tension seem to involve processes beyond inhibition of
mended for BP monitoring in mice. endothelium-derived NO because infusion of l-arginine,
Lerman et al   Animal Models of Hypertension   e13

which activates eNOS to produce NO, does not completely heavy metals, stress (eg, air jet, psychosocial, cold induced),
reverse hypertension. Long-term inhibition of NO likely has medications and herbal supplements (eg, acetaminophen, non-
an impact on BP regulatory systems beyond direct effects on steroidal anti-inflammatory drugs, licorice), hypothyroidism,
vasodilation and vascular tone. In particular, persistent inhi- hyperparathyroidism, and others, which have been described
bition of NO biosynthesis with L-NAME is associated with elsewhere.
profound vasoconstriction, activation of the sympathetic ner-
vous system and RAAS, oxidative stress, kidney damage, and Obesity
structural alterations of the vascular wall. Because RAAS Overweight and obesity contribute up to 75% of the risk for
inhibitors fail to completely ameliorate L-NAME–induced primary hypertension and to most cases of treatment-resis-
hypertension, other humoral factors such as ET-1 have also tant hypertension. Although the physiological and molecular
been implicated.197 mechanisms of obesity-related hypertension remain unclear,
As with all experimental models of human disease, there sodium retention, RAAS activation, increased sympathetic
are strengths and limitations that need to be considered for the activity, leptin resistance, and endothelial dysfunction have
long-term NO inhibition model of hypertension. The strengths been implicated. Elucidating the mechanisms responsible for
of this pharmacological model of hypertension include the obesity-related hypertension may allow the development of
relatively simple technical approach, reproducibility of the novel strategies for treating these patients.
model, development of systemic hypertension, robust nature Overall, animal models that exhibit concomitant obesity
of severe hypertension, evidence of target-organ damage, and and hypertension can be divided into 2 distinct categories:
reversibility of hypertension with l-arginine and various com- models of obesity that spontaneously develop hyperten-
monly used antihypertensive drugs.198 However, the mecha- sion and models of hypertension with superimposed obesity.
nisms underlying L-NAME likely involve processes beyond Despite their different pathogeneses, both types of models
NO synthase inhibition, and more important, the pathophysi- allow exploration of the role of obesity in the development
ological role of decreased NO biosynthesis in human hyper- and progression of hypertension. Although models of primary
tension remains unclear. Moreover, the BP increase in NO obesity permit evaluating the mechanisms by which obesity
deficiency hypertension occurs rapidly, usually within hours predisposes to hypertension, models of hypertension enable
of L-NAME or L-NMMA infusion, whereas the development studying how obesity alters its course.
of hypertension in humans occurs slowly and becomes estab- Several models of experimental obesity spontaneously
lished after many years. Hence, although the L-NAME/L- develop hypertension as they gain body weight. Genetically
NMMA rat is an excellent model of hypertension-induced induced obesity models that develop hypertension include the
Downloaded from http://ahajournals.org by on March 14, 2019

target-organ damage mimicking many of the complications obese Zucker rat, ZSF1 rat, Wistar fatty rat, and ob/ob mouse,
observed in human hypertension, it is probably not an appro- which result from mutations that interfere with leptin signal-
priate model for primary hypertension, which has a slow ing and transduction. These models are the most commonly
onset that develops over the lifetime, and caution needs to be used and offer the advantage of minimizing the impact of
exercised when findings from this model are extrapolated to confounding factors, allowing mechanistic studies to investi-
the clinic.199 On the other hand, superimposing the L-NAME gate the role of specific genes in obesity-induced hyperten-
model onto conventional diabetic mouse strains accelerates sion. Genetically induced obesity-hypertension can also be
kidney injury, recapitulating many features of human diabetic achieved in large animal models. For example, DNA transpo-
nephropathy.200,201 sition of D374Y gain-of-function cDNA of chimp proprotein
convertase subtilisin/kexin type-9 is associated with elevated
Summary and Future Directions BP.202 However, coexisting obesity and hypertension result
What we have learned from the long-term NO synthase from the combination of genetic and environmental epigen-
inhibition model is that NO is a critical factor involved in etic factors. Thus, genetically induced obesity models may not
the physiological regulation of cardiovascular function and fully address this need.
homeostasis and is a key regulatory molecule involved in Diets rich in saturated fats and refined carbohydrates
multiple functions, including vascular tone, renal function, induce weight gain and changes in body composition and
salt-volume homeostasis, and renin secretion. Therefore, adipose tissue cellularity in both rodents203,204 and several
it is a potentially attractive therapeutic target. Although large animals, including dogs,205 rabbits,206 and pigs.207
organic nitrates have been used to treat angina since the The rat might be the optimal small animal model for diet-
1800s, these compounds have a short half-life with little induced hypertension because of its size and propensity to
benefit in chronic hypertension. Hence, there is now grow- develop hypertension faster than mice. High-carbohydrate
ing interest in the antihypertensive effects of inorganic diets, alone or in combination with high-fat diets, have been
nitrates in the diet and in the development of modulators shown to induce obesity, hypertension, hyperlipidemia, and
of NO that are stable and that will deliver NO in a tissue- glucose intolerance in several rat strains, including SD and
and site-specific manner. Animal models may be useful in Wistar rats, within just 3 weeks.208,209 However, C57BL/6
understanding the therapeutic potential of NO modulation mice fed a high-fat/high-carbohydrate diet supplemented
in human hypertension. with NaCl may need as much as 3 months to develop hyper-
Several additional forms of hypertension that may develop tension.203 Unlike rodents, development of hypertension is
in animal models are less common and are not described here. relatively similar among large animal models of obesity,
These include models of hypertension induced by infection, ranging from 5 weeks in the dog210 to 12 to 16 weeks in
e14  Hypertension   TBD 2019

the pig.207 Although a mixture of chow and added fat may Sex Differences and Aging in Hypertensive
be sufficient to develop obesity-hypertension, purified- Small Animal Models
ingredient diets are the preferred choice because of their Profound sex differences are an important feature of hyper-
low batch-to-batch variability and lack of plant-derived tension and cardiovascular diseases in humans. Similarly, sex
phytochemicals, which may alter disease progression.211 differences in BP have been noted in most animals, including
Purified diets contain high levels of carbohydrates, which rodents, dogs, chickens, and rabbits,220,221 but the mechanisms
are indispensable to achieve several features of metabolic responsible for the sex differences in hypertension or BP con-
syndrome, particularly hypertension. Pigs fed a high-fat diet trol have been studied mainly in rodents (Table 2). Almost
alone are characterized by dyslipidemia and vascular dys- without exception, young adult male rats have higher BP than
function but do not have increased BP levels.212 Contrarily, females regardless of whether they are normotensive (SD) or
feeding domestic pigs a diet containing high levels of carbo- hypertensive (SHR, DSS), just as in humans. In normoten-
hydrates, fat, and cholesterol over 16 weeks induces spon- sive SD rats, mean BP is lower in females than in males,222
taneous hypertension.207 Although high-fructose diets seem possibly because of lower proximal sodium reabsorption and
to be more effective in inducing metabolic syndrome com- greater ability to excrete sodium.223 There are also greater
pared with high-sucrose diets, the diet-induced increases in pressor responses in males with DOCA-salt hypertension224
BP levels are comparable.213 and DSS rats.225 In both mice and rats, normotensive males
Lastly, chemical agents (eg, monosodium glutamate) also develop higher BP in response to a slow-pressor dose of
used primarily to induce obesity may result in spontaneous Ang II.226,227 The mechanisms responsible for the elevated BP
hypertension.214 However, the association between obesity and or response to Ang II vary with the strain and genetics of the
hypertension in these models is often weak. rodents. The kidney and perhaps the sex steroid milieu might
In addition to models of primary obesity, models of be responsible for the sex differences in the Ang II–mediated
concurrent, independently achieved hypertension are hypertension. However, in general, radiotelemetry monitoring
critical to answer the fundamental question of whether shows that the estrus cycle of the female does not affect daily
obesity exacerbates existing hypertension. For example, BP markedly.
fat loading in DSS rats aggravated BP and salt-induced Aging in humans is associated with an increase in BP. In
renal damage, preceded by body weight gain, visceral fat hypertensive rat models, BP increases in females as they stop
accumulation, and insulin resistance.215 Similarly, studies estrous cycling. In SHRs, BP is lower in females before they
in Ossabaw pigs with coexisting obesity and renal artery stop estrous cycling and then increases progressively such that
stenosis demonstrated that obesity amplifies both renal by 16 months of age, BP in females is similar to or higher than
Downloaded from http://ahajournals.org by on March 14, 2019

and cardiac injury, yet BP levels remained unaltered.216,217 that in males.228 In DSS rats on a low-salt diet, aging in both
These observations suggest that the primary mechanism males and females is also associated with increases in BP.229
implicated in the development of hypertension is an In many hypertensive rat models, hypertension is mediated by
important determinant of the effect of obesity. Although different mechanisms for males and females. Contributions of
obesity exacerbates the salt sensitivity of BP, the effect of hormones, sympathetic and renal nervous systems, immune
hypertension may be dissociated from obesity in surgically system, metabolic syndrome effects, and oxidative stress,
induced hypertensive models. which are different between males and females, must be taken
Alternatively, the impact of obesity on hypertension into account. Thus, sex differences in BP control in animals
could be studied by genetic or dietary manipulations. In the are as complicated as in humans.
spontaneously hypertensive obese (Koletsky) rat, hyper-
tension and obesity are induced by independent genetic Role of Sex Steroids
mutations.218 BP levels are comparable to those of rats The presence or absence of sex steroids is an important medi-
with salt-sensitive hypertension, suggesting that obesity ator of hypertension in many of the spontaneously hyper-
induced by this mutation does not exacerbate genetically tensive rodent models. Castration of male SHR, DSS, and
induced hypertension. Contrarily, DOCA-salt treatment mREN2 (transgenic rat line bearing the murine Ren [renin]
of obese Zucker rats increases BP in as few as 4 days.219 2 gene) models causes a reduction in BP and ovariectomy
Therefore, these models are suitable to explore whether increases BP in DS rats on a high- or low-salt diet229 and in
obesity increases the sensitivity of animals to experimen- mREN2 females.224 In contrast, ovariectomy of SHRs has no
tally induced hypertension. Presumably, the involvement of effect on their BP.225 Exogenous androgens given long term
the RAAS, oxidative stress, or other mechanisms may dic- increase BP in female SD rats, a model of polycystic ovary
tate the sensitivity of the models to coexisting obesity and syndrome.230 The mechanisms by which sex steroids can affect
hypertension. BP include stimulation of renal angiotensinogen synthesis by
The choice between small or large animal models of androgens and estrogen-mediated stimulation of the vasodi-
obesity-hypertension should consider several elements. lator arm of the RAAS or decreased AT1 receptor synthesis.
Specifically, large animal models of diet-induced obesity may Estradiol also increases the expression of eNOS.
require a long period of time to develop spontaneous hyperten-
sion, increasing experimental costs. In addition, body weight, Sex and Aging-Related Differences in Mechanisms
length, and fat percentage may reach significant proportions, Responsible for Hypertension in Young SHRs
limiting animal mobility and increasing the risk for infections In young male and female SHRs, the mechanisms responsible
and surgical complications. for hypertension are different. For example, in young adult
Lerman et al   Animal Models of Hypertension   e15

Table 2.  Sex Differences in BP and Response to Treatment in Animal Models

Males Females
Aging (After
Strain/Model Young Adult Aging Young Adult Cycling)
SHR
  No treatment M>F M≤F F<M F≥M
 ETAR antagonism No effect No effect No effect Modest fall
 Enalapril/losartan Normalize Normalize Normalize Decrease
 20-HETE ... Yes No effect Decrease F>M
 Gonadectomy Castrated<M Castrated<M No effect, OVX=F No effect, OVX=F
  Adrenergic block/renal denervation Decrease M>F Decrease Decrease Decrease
  MC4R antagonist Decrease Decrease No effect No effect
 NO NOx/NOS1/NOS3 E2 increase eNOS synthesis ... ...
activities M<F
 Antioxidants Decrease Decrease If start before puberty No effect
  T cells ... ... Tregs F>M ...
 Pregnancy ... ... Falls last trimester ...
DSS
  Low NaCl M>F Rise with aging F<M Rise with aging
  Plus high-NaCl diet M>F ... ... ...
 Gonadectomy Castrated<M Castrated<M OVX>F OVX=F rise with age
 Pregnancy ... ... No fall last trimester ...
mREN2
Downloaded from http://ahajournals.org by on March 14, 2019

  No treatment M>F ... F<M ...


 Gonadectomy Castrated<M ... OVX>F OVX=F
Normotensive strains
  No treatment (rats) M>F M<F F<M F≥M by 18 mo
  Plus DOCA-salt M>F ... ... ...
  Plus Ang II ... ... ... ...
  Plus ACE inhibitor M>F (mice) ... F>M (rats) ...
  Minus ACE inhibitor M>F (mice/rats) ... ... ...
  Plus ACE inhibitor+high NaCl M=salt sensitive ... F=not salt sensitive ...
  T cells M>F ... F<M ...
Pregnancy Decrease late
  Plus RUPP ... ... Increase late ...
  Plus L-NAME ... ... Increase ...
Offspring effect RUPP dams M>F Normalizes F<M Increase F>M
Obesity (mice) M>F ... No effect ...
Rabbits M>F ... F<M ...
Dogs + Ang II Increase M>F ... Increase F<M ...
Chickens M>F ... F<M ...
20-HETE indicates 20-hydroxyeicosatetraenoic acid; ACE, angiotensin-converting enzyme; Ang II, angiotensin II; BP, blood pressure; DOCA, deoxycorticosterone
acetate; DSS, Dahl salt-sensitive; E2, estrogen; ellipses (...), data unavailable; eNOS, endothelial nitric oxide synthase; ETAR, endothelin-A receptor; F, Female;
L-NAME, Nω-nitro-l-arginine; M, male; MC4R, melanocortin 4 receptor; mREN2, transgenic rat line bearing the murine Ren (renin) 2 gene; NO, nitric oxide; NOS, nitric
oxide synthase; NOx, NADPH (nicotinamide adenine dinucleotide phosphate) oxidase; OVX, ovariectomized female; RUPP, reduced uterine perfusion pressure; SHR,
spontaneously hypertensive rats; and Treg, regulatory T cell.
e16  Hypertension   TBD 2019

male SHRs, hypertension is mediated by androgens because suggesting that other components contribute to the hyperten-
castration reduces BP to levels similar to those in females, and sion in aging SHRs. Similarly, blockade of the RAAS reduces
androgen supplementation restores hypertension,231 whereas BP but becomes less efficacious with aging in females235 but
ovariectomy has no effect in females, suggesting that estro- normalizes BP in aging males as in young males. Whereas
gens have no effect on BP in females. Removal of the renal the endothelin system plays no role in mediating hyperten-
nerves or adrenergic blockade normalizes BP in both male sion in young male or female SHRs, with aging, ETA receptor
and female SHRs.232 Central MC4R (melanocortin 4 recep- antagonism modestly reduces BP in females but not males.230
tor) antagonism also reduces BP in young male SHRs233 but Blockade of 20-HETE synthesis fails to reduce BP in young
not young females,234 suggesting that sympathetic activation female SHRs, whereas 20-HETE synthesis inhibition reduces
in males may be the result of MC4R activation. Blockade of BP in old females,242 suggesting that, unlike in young females,
the RAAS with angiotensin-converting enzyme inhibitor231,235 20-HETE contributes to regulation of BP in old female SHRs.
normalizes BP in both young male and female SHRs. In DSS rats, BP in females increases with ovariectomy and
The role of oxidative stress in mediating hypertension in decreases with estradiol supplements.229 By 12 months of age,
SHRs is also sex dependent, although females have similar estradiol-supplemented ovariectomized DSS rats, ovariecto-
or higher levels of oxidative stress markers. Overall, anti- mized DSS rats, and intact DSS rats have similar BPs,229 sug-
oxidants prevent the development of hypertension in both gesting that with aging changes occur in the estrogen receptors
sexes of SHRs,236,237 but once hypertension is established, its or intracellular signaling pathways that make the estradiol no
maintenance is independent of oxidative stress in females longer able to attenuate the BP.
but not males. The NO system may also be more activated Other comparisons for BP regulation in male and female
in young female than male SHRs.238 Tetrahydrobiopterin sup- DSS rats are not as comprehensive as in SHRs and thus are not
plementation reduces BP in male SHRs, but this results from discussed further.
a concomitant reduction in androgen levels rather than pro-
tecting against eNOS uncoupling.239 Inhibition of 20-HETE Other Mechanisms Underlying Sex Differences in
(20-hydroxyeicosatetraenoic acid), synthesis, by either a Hypertension
nonspecific arachidonic acid metabolism inhibitor or a spe- Sex differences in BP have been reported in most models
cific ω-hydroxylase inhibitor, reduces BP in male SHRs240 but of diet-induced obesity. Male C57Bl/6 mice on a 16-week
not young females.230 Blockade of the RAAS reduces BP to high-fat diet gained less weight than females but developed
similar levels in both male and female SHRs when they are hypertension.243 Male obese Zucker rats have higher BP than
young231 but becomes less efficacious with aging in females.235 females.244 The mechanisms responsible for the sex differ-
Downloaded from http://ahajournals.org by on March 14, 2019

In addition, the endothelin and the 20-HETE eicosanoid sys- ences in BP in these rodent models remain unclear.
tems may contribute to hypertension in aging females. In large animal models, many studies have been performed
Sex differences in inflammation and the immune system in dogs, although the sex has not often been considered. Ang
also contribute to hypertension (Table 2). Both male and II given intracerebroventricularly to dogs caused a pressor and
female SHRs have a depressor response to the lymphocyte dipsogenic effect in male dogs but not females.245 Male rabbits
inhibitor mycophenolate mofetil, but the response was greater have higher BPs than females, but neither is salt sensitive.221
in females.241 Females have more circulating CD3+, CD4+, In summary, many rodent models show sex differences in
and proinflammatory CD3+CD4+RORγ+ Th17 cells, whereas the BP levels, and the hypertensive mechanisms responsible
males have more immune-suppressive CD3+CD4+Foxp3+ for them are different. As Table 2 shows, in general, in female
regulatory T cells. In the kidney, females had greater num- animal models, the mechanisms responsible for hyperten-
bers of CD8+ and regulatory T-cell infiltration, whereas sion are more multifaceted than in males, and these mecha-
males had greater CD4+ and Th17 cell infiltration. The nisms tend to increase in number and significance with aging.
role of T cells in mediating hypertension is emerging. For Therefore, investigators should be cognizant of and look for
example, females show greater pressor response to Ang II sex differences in BP, recognize that there may be sex dif-
after male-to-female T-cell transfer, and males become less ferences in the mechanisms responsible for BP control, and
responsive after female-to-male T-cell transfer. Future stud- recognize that there also may be age-related differences in the
ies are necessary to determine the role that T cells play in mechanisms responsible for BP control.
mediating primary hypertension in humans in order to refine
studies in animal models. End-Organ Damage
The impact of hypertension on human health occurs through
Aging and Sex Differences in Mechanisms damage to critical target organ systems, including the brain,
Responsible for Hypertension heart, kidneys, and vasculature. Investigations of target organ
As noted, with aging and cessation of estrous cycling, BP damage in many established rodent models of hypertension
increases in female SHRs to levels similar to or higher than have revealed insights into the mechanisms of BP-induced tis-
those in males,228 despite the lack of effect of ovariectomy sue injury. However, with the growing importance of comor-
on BP in young females.231 The mechanisms responsible bidities and aging in hypertension and its complications,
for hypertension are also different by sex in aging SHRs. studying aged animals and animals with induced comor-
For example, although BP falls with renal denervation or bidities such as hyperglycemia, hyperlipidemia, and obesity
adrenergic blockade, both aging males and females remain would further increase the translational relevance of animal
hypertensive,234 which does not occur in young animals, models of human hypertension.
Lerman et al   Animal Models of Hypertension   e17

Vasculature: Vascular Dysfunction and Remodeling and passive properties of the vessel.283 Pressure myography
In hypertension, endothelial dysfunction is associated with is used to assess small vessel function and structure under
target-organ damage and has been demonstrated in peripheral, nearly physiological conditions of pressure and flow by digi-
coronary, cerebral, renal, and conduit vessels in experimen- tally tracking diameter and flow in real time.284 Parameters
tal and clinical hypertension. Molecular processes causing studied by pressure myography include media structure, wall
endothelial dysfunction include decreased endothelial NO stress, strain, and myogenic tone, which allow evaluation of
production, increased bioavailability of reactive oxygen spe- responses to increases in pressure, flow, and pharmacologi-
cies, increased ET-1 and Ang II production, and immune cal stimuli. The system can be linked to imaging equipment,
mechanisms. Hypercontractility also contributes to high vas- for example, to assess calcium transients. These approaches
cular tone in hypertension. Processes underlying this involve have been used extensively in various rodent models of hyper-
increased vascular smooth muscle cell contraction and tension, including L-NAME rats, SHRs, stroke-prone SHRs,
reduced relaxation, mediated in large part through changes in salt-sensitive rats, renovascular models, Ang II–induced
intracellular calcium concentration and activation of RhoA- hypertension, and transgenic mice, and they have contributed
Rho kinase pathways. enormously to elucidating the vascular phenotype as a cause
In addition to functional alterations, small and large arter- and target of hypertension.
ies exhibit structural changes in hypertension, characterized In addition to impaired endothelial function, hyperreactiv-
by remodeling, fibrosis, and inflammation, processes that are ity, and structural remodeling, hypertension-induced vascular
amplified with aging in SHRs.246 Resistance artery narrowing damage may involve rarefaction, which is characterized by a
and large artery stiffening not only are target-organ effects of decrease in microvascular density. Rarefaction may be func-
high BP but also contribute to the development of hyperten- tional (vasoconstriction) or structural and may contribute to
sion by increasing peripheral resistance and compromising ≈25% of peripheral resistance in experimental hypertension.249
arterial compliance.
Structural remodeling of the vascular wall leads to Heart: Cardiac Fibrosis, Hypertrophy, and
reduced lumen diameter and thickening of the vascular media. Hypertensive Heart Disease
Remodeling of small arteries may be the first manifestation Cardiac injury is a major consequence of persistent, uncon-
of target-organ damage in hypertension. In clinical studies, trolled hypertension. Elevated BP culminates in myocar-
100% of patients with stage 1 hypertension have small vessel dial strain, resulting in LVH, an independent risk factor for
remodeling, whereas only 60% have endothelial dysfunction cardiovascular mortality. Disruption in cardiac architecture
and 45% have LVH.247 The concept of vascular remodeling with LVH is associated with aberrant electrical conduction,
Downloaded from http://ahajournals.org by on March 14, 2019

was first suggested in 1989 when pial arterioles from stroke- leading to atrial or ventricular arrhythmias and sudden death.
prone SHRs were found to have significant structural altera- When the heart can no longer sustain normal function in the
tions.248 Arterial remodeling is characteristically associated face of elevated afterload, persistent hypertension leads to
with increased wall thickness, of which 2 types are described: diastolic and ultimately systolic heart failure. Accordingly,
inward or outward, depending on whether the lumen diam- hypertension is a leading cause of congestive heart failure
eter is reduced or increased, respectively. Remodeling is fur- in humans.
ther classified into eutrophic, hypotrophic, or hypertrophic, In many rodent models of hypertension, approximating
depending on whether there is no change in or reduced or human stage 2 hypertension leads to LVH within 2 to 4 weeks,
increased vascular material (vascular smooth muscle cells, measured by augmented ratios of heart to body weight or heart
extracellular matrix), respectively.249,250 Eutrophic inward to tibia length. The level of BP measured by radiotelemetry
remodeling is usually found in SHRs and Ang II–induced correlates with the extent of cardiac hypertrophy,161 making
hypertension, whereas hypertrophic remodeling is found in heart weight a possible surrogate for hypertension when direct
renovascular hypertension, salt-sensitive hypertension, aldo- measurements are unavailable. Rodent echocardiography
sterone-induced hypertension, and other forms of secondary allows direct assessment of changes in cardiac filling patterns
hypertension (Table 3). and left ventricular wall thickness. These detailed measure-
Different types of remodeling may occur in differ- ments allow discrimination between signaling pathways that
ent vascular beds. For example, in SHRs, small resistance favor physiological versus pathogenic cardiac hypertrophy.
arteries exhibit eutrophic inward remodeling, whereas con- After 1 month, hypertensive cardiac injury in rodents is
duit arteries undergo hypertrophic remodeling. Mechanical marked at the histological level by myocyte damage, mild
changes that promote arterial stiffness, decreased elastic- perivascular fibrosis, and sparse mononuclear cell infil-
ity, and reduced distensibility are associated with structural trates,161 which nonetheless modulate cardiac injury during
alterations, which further contribute to vascular target-organ hypertension. At the molecular level, cardiac hypertrophy
damage. is characterized by recapitulation of fetal gene expression
The most commonly used approach to directly assess in experimental hypertension.285 Severe hypertension pro-
vascular function (endothelial function, vasoconstrictor and vides a model of cardiac fibrosis, which can be quantified
vasodilator properties), along with structure and mechanical by immunohistochemistry and molecular signatures of car-
properties, is myography (wire and pressure). Wire myog- diac scar formation. Scarring disrupts electric conduction in
raphy allows ex vivo measurement of transverse isometric the heart with consequent discrete dysrhythmias that can be
tension in an arterial segment in response to different fac- captured and quantified with current radiotelemetry moni-
tors and assessment of biochemical and molecular pathways toring systems.286
e18  Hypertension   TBD 2019

Table 3.  Vascular Target-Organ Damage in Different Experimental Models of Hypertension

Hypertension Model Vascular Target-Organ Damage Reference


Genetic rat models
 SHR Inward eutrophic remodeling (resistance artery) 251–256
Hypertrophic remodeling (conduit artery)
Rarefaction
Endothelium-dependent dysfunction
Impaired endothelium-independent vasorelaxation (conduit
arteries)
 SHRSP Hypertrophic remodeling 257, 258
Fibrosis
Vascular hypercontractility
Endothelial dysfunction
Increased myogenic tone
 GHR Hypotrophic outward remodeling (basilar artery) 259
Salt-sensitive hypertension
 DSS Hypertrophic remodeling 51, 260–262
Endothelial dysfunction
Impaired myogenic response (cerebral artery)
 DOCA-salt Hypertrophic remodeling 254, 263
Endothelial dysfunction
Vascular inflammation
Renovascular
  1-Clip Goldblatt Inward eutrophic remodeling (resistance artery) 264
Downloaded from http://ahajournals.org by on March 14, 2019

Hypertrophic remodeling (conductance artery)


 2K-1C Hypertrophic remodeling (aorta but not mesenteric artery) 265
NO-dependent models
 L-NAME Outward hypotrophic remodeling 266–269
Aortic stiffness
Increased pulse-wave velocity
Impaired endothelium-independent vasorelaxation
Cerebral artery remodeling
 SHR/L-NAME Aortic stiffness 270
Reduced distensibility
Excessive fibrosis
Ang II–dependent models
  REN2 transgenic rats Aortic endothelial dysfunction 271
Decreased aortic contraction
Fibrosis
Endothelial dysfunction
 dTGR Medial hypertrophy 272
Intimal thickening
Fibrinoid necrosis
 Ang II infused (400 ng·kg ·min ;
−1 −1
Inward eutrophic remodeling 161–164,
slow pressor) 273–275
Endothelial dysfunction
Vascular hypercontractility
Low-grade vascular inflammation

(Continued )
Lerman et al   Animal Models of Hypertension   e19

Table 3.  Continued

Hypertension Model Vascular Target-Organ Damage Reference


 Ang II infused (≥1000  Aortic medial thickening 276–279
ng·kg−1·min−1)
Outward aortic remodeling
Vascular inflammation
Vascular hyperreactivity
Increased vascular tone
Increased aortic stiffness
Endothelial dysfunction
Large mammals
 AGM Renal vascular remodeling 41, 280
Vascular hypertrophy
  Hypertensive obese pig Cardiac microvascular remodeling 281
  Fat-fed minipig Endothelial dysfunction 282
Vascular inflammation
Hypertrophic remodeling
AGM indicates African green monkey; Ang II, angiotensin II; DOCA, deoxycorticosterone acetate; DSS, Dahl salt-sensitive; dTGR, human
renin-angiotensinogen double-transgenic rat; GHR, genetically hypertensive rat; L-NAME, Nω-nitro-l-arginine; NO, nitric oxide; REN2,
Renin-2; SHR, spontaneously hypertensive rat; SHRSP, stroke-prone spontaneously hypertensive rat; and 2K-1C, 2-kidney, 1-clip.

Kidney: Renal Fibrosis and Hypertensive characteristic features of human malignant hypertension can
Kidney Failure also be observed in Ren2 transgenic rats.
Hypertension-induced renal damage comprises at least 3 pat- In the setting of underlying renal disease, the relation-
terns: benign nephrosclerosis, malignant nephrosclerosis, and ship between arterial pressure and kidney damage shifts, and
Downloaded from http://ahajournals.org by on March 14, 2019

hypertension-accelerated kidney disease.287 Benign neph- BPs that do not normally lead to progressive damage do so.
rosclerosis is characterized by arteriosclerosis, interstitial Mechanisms involved are controversial and depend on the
fibrosis, and global glomerulosclerosis. The individual risk models used. Many studies use five-sixths nephrectomy (see
of end-stage kidney disease from benign nephrosclerosis is the Renoprival Hypertension section). Kidney damage has
surprisingly small, but the net effect of benign hypertension been suggested to result from resultant dilation of the affer-
is significant because hypertension itself is so common. In ent arteriole with efferent vasoconstriction, which together
contrast, malignant hypertension, which itself is uncommon, increase glomerular capillary pressure independently of
typically leads to kidney damage, often associated with fibri- changes in arterial pressure. In contrast, a surgical approach
noid necrosis and thrombosis of small vessels and glomeruli. to reduce renal mass without generating hypertension showed
Hypertension-induced renal damage most commonly occurs that systemic hypertension is required for renal damage.290
in the setting of underlying kidney disease, in which hyper- Mouse strains vary in their susceptibility to kidney damage.
tension accelerates the progression, for instance, of diabetic Rodent models of diabetic kidney disease, for example, that
kidney disease. are induced by streptozotocin on a 129SvE background, have
The 3 subtypes of hypertension-induced renal damage been used to demonstrate the impact of superimposed hyper-
have been replicated in animal models. Rodent models like tension on baseline kidney damage.291
the SHR develop kidney damage very slowly. This appears
to reflect preserved renal vascular autoregulation, with nor- Brain: Hypertensive Cerebral Damage
mal pressure-induced afferent vasoconstriction, preventing Hypertension is a major risk factor for cerebrovascular dis-
high arterial pressure from being transmitted to the glomeru- eases such as stroke (ischemic and hemorrhagic) and vascular
lar capillaries.288 This model resembles benign hypertension dementia but also for neurodegenerative diseases, including
in humans, in which the risk for hypertensive nephrosclero- Alzheimer disease.292 Hypertension has damaging effects
sis is low, and damage is restricted primarily to precapillary on cerebral blood vessels, which have been implicated in its
blood vessels and interstitium. In contrast, when arterial pres- harmful effects on the brain. Lacking energy reserves, the
sure rises above a critical threshold, for example, in stroke- brain is highly susceptible to alterations in blood supply, and
prone SHR rats exposed to high salt intake, renal damage hypertension can promote both acute and chronic ischemic
develops rapidly, with lesions characteristic of malignant brain injury.293 As in systemic arteries, hypertension acceler-
hypertension.289 In this case, the pressure is above the auto- ates atherosclerosis and induces stiffening, remodeling, and
regulatory range and is therefore transmitted directly to the hypertrophy in cerebral arteries. Distinctive alterations, similar
glomerulus. This causes proteinuria and rapidly progressive to those observed in the kidney (lipohyalinosis), are observed
renal dysfunction resulting from glomerular damage. These in penetrating arterioles of the brainstem and basal ganglia.
e20  Hypertension   TBD 2019

Functionally, hypertension alters myogenic tone and cere- Of particular interest are models in larger animals such
brovascular autoregulation, induces endothelial dysfunction, as pigs and monkeys, in which brain size, gray-white matter
impairs the ability of neural activity to increase cerebral blood ratio, vascular topology, cognitive testing, and cardiovascu-
flow (neurovascular coupling), and damages the blood-brain lar function have greater translational relevance.344 Monkeys
barrier. These structural and functional alterations promote made hypertensive by aortic coarctation exhibit white matter
vascular occlusions, leading to acute ischemic brain injury and lesions and microinfarcts and, like the pig model, lend them-
chronic vascular insufficiency, causing white matter damage. A selves to more detailed assessment of cognitive end points
major consequence of the hypertensive white matter damage is (Table 4). However, these models are expensive, not well
cognitive impairment. Indeed, hypertension is the major cause suited to high-throughput investigations, and less amenable to
of cognitive impairment on a vascular basis, the most com- genetic manipulations.
mon cause of dementia after Alzheimer disease.292 Executive In summary, although investigations into target-organ
dysfunction and psychomotor slowing are the typical cogni- damage in animal models of human hypertension have
tive deficits, but memory impairment, more characteristic of focused mainly on vessels, the heart, and the kidney, there is
Alzheimer disease, can also occur in more advanced cases.292 a paucity of information on the brain effects in these models.
In addition, hypertension induces rupture of cerebral microves- This is particularly evident in renovascular hypertension and
sels, causing intracerebral hemorrhage, typically in the basal low-renin hypertension. Considering the devastating impact
ganglia, or bursting of aneurysmal dilatation in arteries at the of hypertension on the brain and its vessels and its pathogenic
base of the brain, resulting in subarachnoid hemorrhage. role in a wide variety of brain diseases, there is a strong ratio-
Several animal models of hypertension have been used to nale for expanding application of state-of-the-art cerebrovas-
investigate the effects of hypertension on the brain (Table 4). cular and neurovascular investigative tools to delve deeper
Although these models do not fully recapitulate the harmful into the mechanisms through which hypertension promotes
effects of hypertension, they have provided valuable knowl- neurovascular and neurodegenerative diseases.
edge of the potential mechanisms underlying the susceptibil-
ity of the brain to hypertension. Most of the models used for Applications of Genetics and Systems
cerebrovascular research have been in rodents, although there Biology to Animal Models
have been studies in larger animals, mainly pigs and monkeys.
Great insight has been gained from the genetic study of
As noted, models based on administration of pharmacologi-
human hypertension. Studies of monogenic forms of hyper-
cal agents have the advantage that the cause of hypertension
tension have revealed the molecular basis of several related
is known and hypertension can be induced in a defined time
syndromes. More recently, GWAS analyses uncovered com-
Downloaded from http://ahajournals.org by on March 14, 2019

frame and in transgenic animals, allowing the study of early


mon variants of modest effect and low-frequency variants that
mechanisms of disease at the molecular level, as well as cog-
contribute to BP variation in patients. These studies provide
nitive dysfunction. A disadvantage is that the hypertension is
important insight into human disease, which can be comple-
limited in time (usually weeks) and does not mimic the long-
mented by animal studies, often in models exhibiting pheno-
lasting impact on the brain of the human disease. Nevertheless,
these models have been some of the most commonly used. typic characteristics observed in human hypertension, which
Genetic models based on intercrossing and selecting for can provide mechanistic biological insight into gene function
the hypertensive phenotype, for example, stroke-prone SHRs and underlying cardiovascular risk.
and blood pressure high-2 (BPH2) mice, exhibit lifelong
Identification of QTL Influencing BP Traits
hypertension and provide insight into the effects of hyperten-
sion on the brain, including cognitive dysfunction, over the life Of the different inbred species used for genetic studies of
course. However, the precise cause of the hypertension remains hypertension, the rat has been widely used for the identifica-
unknown, raising the possibility that the cerebrovascular altera- tion of QTL with linkage analysis approaches. This has been
tions are not attributable to hypertension but to unrelated genetic driven by the large number of rat genetic models of hyperten-
factors. For example, the increased susceptibility to ischemic sion, the relatively low cost of rat experimentation, and the
brain injury in SHRs and stroke-prone SHRs could be related, ease and accessibility of techniques for assessing cardiovas-
in part, to an inherited vulnerability of neurons to excitotoxic- cular phenotypes in rats. The functional validation of QTL
ity.342,343 Some transgenic models have lifelong hypertension has been enabled by the generation of congenic or consomic
with a known cause, for example, mice overexpressing human strains, in which defined segments of DNA from 1 strain are
angiotensinogen and renin (R+/A+) or lacking eNOS, and have introgressed onto the genetic background of a second strain
been very useful for investigating the role of specific pathways with a genetic marker–assisted breeding strategy. With this
and mediators in the effects of lifelong hypertension on the brain. approach, phenotypic differences detected between the paren-
Some hypertension models that produce brain lesions tal and congenic strains can indicate that a gene or genes
(infarcts, hemorrhages, or white matter lesions) usually within a particular substituted region of genomic DNA have
require the combination of pharmacological, dietary, genetic, an influence on the functional trait of interest. The subsequent
and surgical manipulations to enhance the effects of hyperten- identification of genes within these QTL has been difficult
sion on the brain (Table 4). Although this mimics the neuro- and depends on complementary approaches, including tran-
pathological impact of hypertension, the time when lesions scriptomic analyses, gene sequencing, and gene editing.45,63
develop cannot be predicted, and the location of the lesions is Development of modern sequencing techniques and sequenc-
highly variable. ing of the full rat genome provided further opportunities to
Lerman et al   Animal Models of Hypertension   e21

Table 4.  Cerebrovascular Pathologies in Selected Animal Models of Hypertension

Hypertension Model Species Neurovascular Pathology References


Ang II infusion Mouse Hypertrophy and remodeling 294–298
(2–4 wk) Rat Neurovascular dysfunction
Increased BBB permeability
Inflammation
Oxidative stress
Cognitive deficits
Brain amyloid-β accumulation
Long-term Ang II+short-term Ang Mouse Microhemorrhages 299, 300
II+L-NAME
Increased BBB permeability
Inflammation
Cognitive deficits
Ren/Agt overexpression Mouse Hypertrophy and remodeling (R+/A+ 301–305
Rat mouse)
Increased stiffness (renin rat)
Endothelial dysfunction
Cognitive deficits (R+/A+ mouse)
Larger infarcts (after MCAO)
Ren/Agt overexpression+L- Mouse Microhemorrhages 306
NAME+high salt
Inflammation
Oxidative stress
ET-1 overexpression in endothelial Mouse Larger infarcts 307, 308
Downloaded from http://ahajournals.org by on March 14, 2019

cells+MCAO
Increased BBB permeability
Cognitive deficits
eNOS deficiency Mouse Hypertrophy 309–312
L-NAME hypertension Rat Endothelial dysfunction
Larger infarcts (after MCAO)
Chronic intermittent hypoxia/ Mouse Neurovascular dysfunction 313–317
obstructive sleep apnea Rat Oxidative stress
Larger infarcts (after MCAO) only with
more severe cyclic hypoxia (6% O2)
BPH2 Mouse Hypertrophy 295, 302, 318
Neurovascular dysfunction
Increased BBB permeability
Cognitive deficits
SHRSP+Western diet Rat Infarcts and hemorrhages 319–321
Neurovascular dysfunction
Increased BBB permeability
Retinopathy
SHR+MCAO Rat Larger infarcts 322, 323
SHRSP+carotid occlusion Rat White matter lesions 324
Increased BBB permeability
Inflammation
Cognitive deficits

(Continued )
e22  Hypertension   TBD 2019

Table 4.  Continued

Hypertension Model Species Neurovascular Pathology References


DSS rat+high salt Rat Oxidative stress 15, 325, 326
Reduced BBB marker proteins
Increased BBB permeability
Loss of myogenic tone
Infarcts and hemorrhages
DOCA-salt Mouse Hypertrophy 298,327–330
Rat Neurovascular dysfunction
Inflammation
Oxidative stress
Cognitive deficits
No change in infarct size or larger
infarcts (after MCAO)
DOCA-salt+elastase Mouse Cerebral aneurysms formation and 328, 331, 332
Ang II (2 wk)+ elastase subarachnoid hemorrhage
Carotid and renal artery
ligation+Ang II (2 wk) +elastase
Renovascular hypertension Rat Infarcts and hemorrhages 333, 334
Larger lesions (after MCAO)
Aortic coarctation Mouse Hypertrophy 335, 336
Endothelial dysfunction
Oxidative stress
Brain amyloid-β accumulation
Downloaded from http://ahajournals.org by on March 14, 2019

Cognitive impairment
Yucatan pig Vascular stiffening 337, 338
Cognitive deficits
Cynomolgus or Rhesus White and gray matter microinfarcts 339
monkey (<1 mm)
Cognitive deficits
Aortic coarctation+high-fat Cynomolgus or Rhesus Worse cognitive deficits 340, 341
diet±aging monkey
A indicates angiotensinogen; Ang II, angiotensin II; BBB, blood-brain barrier; BPH2, blood pressure high-2; DOCA,
deoxycorticosterone acetate; DSS, Dahl salt-sensitive; eNOS, endothelial nitric oxide synthase; ET-1, endothelin 1; L-NAME,
Nω-nitro-l-arginine; MCAO, middle cerebral artery occlusion; R, renin; Ren/Agt, renin/angiotensinogen; SHR, spontaneously
hypertensive rat; and SHRSP, stroke-prone spontaneously hypertensive rat.

fully define and design experiments to elucidate key sequence DNA methylation, posttranslational histone modifications,
differences of candidate genes within a QTL. and noncoding RNAs. Of these factors, DNA methylation
has been most studied. For example, elevated methylation
Epigenetics of the promoter region of HSd11b2 has been correlated with
Rodent models have served as excellent platforms to validate reduced activity of the enzyme and hypertension in patients.345
the impact of deletion or overexpression of individual genes Similarly, the promoter of NKCC-1 is hypomethylated in the
associated with hypertensive traits in GWASs or other link- aorta and heart of SHRs compared with the Wistar Kyoto rat,
age studies. In addition to their value for genetic and genomic correlating with increased NKCC-1 activity in those tissues
studies, these models have contributed to recognition of the and more severe hypertension.346 The use of animal models to
influence of environmental factors on disease phenotypes, assess epigenetic regulation of gene expression in hyperten-
including hypertension, fueling the study of epigenetics. sion promises to be a productive area of focus in the future.
Epigenetics refers to the effects of environmental factors that
induce changes in an organism resulting from modifications Noncoding RNAs
in gene expression rather than a direct alteration of DNA RNAs that do not code for proteins can influence disease patho-
sequence. These modifications commonly occur through genesis by regulating the effectiveness of gene expression
Lerman et al   Animal Models of Hypertension   e23

through modulation of mRNA levels and repression of mRNA genomics, proteomics, and analysis of the microbiome show
translation. Various forms of noncoding RNAs such as miR- promise for advancing basic understanding of human hyper-
NAs, long noncoding RNAs, and circular RNAs have been tension. These approaches should also allow better validation
proposed to play a role in the regulation of BP, the risk for of animal models based on identification and recapitulation of
hypertension, and the risk for target-organ damage associated specific “omic” signatures derived from human hypertension.
with hypertension.106,347,348 Although controversy surrounds
the appropriate criteria for identifying noncoding RNAs of Summary: Relevance of Models to Human
functional significance, there is growing interest in studying Hypertension: Concordances and Gaps
them in animal models of hypertension and hypertension- Animal models of human disorders have proved to be
related cardiovascular disorders from both a mechanistic and immensely useful in translational research in a number of
a therapeutic perspective.106,347–349 Furthermore, noncoding fields, including hypertension. These models allow incisive
RNAs may be involved in the mechanisms of kidney injury in approaches not possible in clinical studies for understand-
some forms of hypertension.350 ing pathophysiology, genetic mechanisms, identification of
new disease markers, and potential therapeutic targets. As we
Microbiome have highlighted here, insights derived from animal models of
Recent studies in humans and animal models have highlighted hypertension have contributed significantly to our understand-
the powerful impact of the microbiome on a range of disor- ing of this highly prevalent human disorder. Here, we sum-
ders. Likewise, gut microbiota can have a profound influence marize a few key points that are especially relevant for those
on BP regulation. In animal models of hypertension, recent working in the field.
observations suggest that there are differences in the micro- Recent high-profile publications have decried the poor
biota between DSS and Dahl R rats351 and between SHRs and reproducibility of published studies using animal models for
Wistar Kyoto rats.352 The mechanisms of action whereby the preclinical assessment of therapeutic agents in human disor-
gut microbiota influence BP are not fully elucidated, but the ders from cancer358 to neurological disease.359 Various factors
release or stimulation of trimethylamine N-oxide, short-chain have been implicated to explain these inconsistencies, includ-
fatty acids, or other factors can influence cardiovascular phe- ing deploying insufficient numbers of experimental animals,
notypes.353,354 In addition, recent studies have identified spe- inadequate power calculations and statistical analyses, assess-
cific changes in the gut microbiome that influence BP effects ment of outcomes by individuals who are not blinded to exper-
of a high-salt diet.355 As this field matures, it is likely that the imental groups, and failure to pursue independent replication
influence of the microbiome on complex phenotypes such as of critical experiments. Accordingly, these remediable meth-
Downloaded from http://ahajournals.org by on March 14, 2019

hypertension will receive wider recognition. Animal modeling odological problems must be taken into account in the design
will likely be very useful in unraveling these actions. of any study using animal models of hypertension.4–8
Another key issue is consideration of how closely the
Systems Biology experimental model truly captures what is observed in humans
The integrated scientific approach to interrogate and under- (see the discussion of construct validity in Utility and Validity
stand the contributions of individual biological components, of Animal Models of Hypertension section). In this regard, 1
including genes, transcripts, proteins, metabolites, epigenetic limitation to developing animal models of human hyperten-
modifications, the microbiome, and environmental modifiers, sion is that the pathogenesis of the human disease is not well
along with the integrated function of these components to a understood, and the primary cause of elevated BP is not appar-
cell, tissue, organ, organ system, or organism, is called sys- ent in the vast majority of affected individuals. On the other
tems biology. Systems biology approaches use high-through- hand, the cardinal feature of human hypertension, elevated BP,
put analytical and bioinformatic tools to understand the entire can be modeled relatively easily in animals by activating sys-
system rather than any single individual aspect. There is no tems known to be involved in human hypertension, including
individual systems biology approach, nor is there agreement the renin-angiotensin system and sympathetic nervous system,
on the precise definition of this approach,356,357 but systems or inhibition of protective factors such as NO. However, these
biology approaches, integrating quantitative measurement of models all suffer from their limited duration of hypertension
biological variables obtained in animal models, mathematical compared with humans. The obscure pathogenesis of human
modeling, reconstruction, and theory,357 show great promise hypertension also complicates interpretation and relevance
for understanding complex multifactorial human diseases of existing animal models with spontaneous hypertension.
such as hypertension. Nonetheless, most of these models respond to antihyperten-
In summary, studies at the level of genetics, epigenetics, sive therapies used in humans and develop similar long-term
the microbiome, and systems biology show great promise for complications, suggesting overlaps in pathogenesis. Recent
hypertension research. At present, however, as a result of sub- progress in understanding genetic mechanisms of primary
stantial gaps in understanding the pathogenesis and genetic hypertension in humans should provide opportunities for gen-
determinants of human hypertension, opportunities to incor- erating more reliable models and for analyzing the veracity of
porate relevant causal pathways or genetic variants into ani- existing models with systems biology approaches.
mal modeling efforts have been limited. For the same reasons, Although elevated BP is the key diagnostic feature of
it has not been possible to use molecular profiling to verify the human hypertension, its morbidity and mortality result from
authenticity of existing animal models. However, data emerg- complications in the brain, heart, kidney, and vasculature.
ing from agnostic systems biology studies using genetics, The contribution of elevated BP to these complications has
e24  Hypertension   TBD 2019

been well established in clinical trials showing that BP low- expertise. No individual model will recapitulate all features
ering reduces complications. However, understanding the of human hypertension, and all have advantages and short-
molecular mechanisms of these complications is an unmet comings, which we have highlighted in this document and the
need, and animal models should continue to add value by accompanying tables. These factors obviously must be taken
identifying pathways and markers associated with increased into account in the design and interpretation of experiments,
risk for complications, understanding mechanisms for known and the most powerful insights will often be derived from
risk factors such as APOL1, and potentially identifying thera- studies carried out in multiple, complementary models.
pies that can protect against complications, above and beyond
BP control. Conclusions
Another key feature of human hypertension is its frequent Hypertension is the most common chronic disease in the
association with chronic comorbidities such as obesity, dia- world, and increased understanding of the pathogenesis, pre-
betes mellitus, and heart and kidney disease, which can influ- vention, and treatment of hypertension and its comorbidities
ence the disease characteristics and outcomes. Likewise, sex is imperative. Animal models of hypertension have been, and
and ethnicity also have major impacts on human hypertension. will likely remain, very useful in providing insights into the
These factors are not typically incorporated into most animal pathogenesis and novel treatment options of hypertension.
models of hypertension but should be to improve concordance Clearly, investigators need to make informed choices as to
with the human condition. the appropriate animal model for specific application, and
The choice of a suitable model such as small versus large the experiments need to be carefully designed, executed, and
animal and spontaneous versus induced hypertension will interpreted. In this scientific statement, we summarize a few
depend on a number of factors, including the specific experi- key points that are especially relevant for those working in the
mental question and the investigator’s available resources and field and may aid in propelling it forward.

Disclosures
Writing Group Disclosures

Other Speakers’
Writing Group Research Bureau/ Expert Ownership Consultant/
Member Employment Research Grant Support Honoraria Witness Interest Advisory Board Other
Downloaded from http://ahajournals.org by on March 14, 2019

Lilach O. Mayo Clinic NIH† None None None None None None
Lerman
Thomas M. Duke University Medical None None None None None None None
Coffman Center and Durham VA
Medical Center
Alejandro R. University of Mississippi NIH-HL095638 (RO1 None None None None None None
Chade Medical Center grant-PI)†; AHA-18490005
(Established Investigator
Award–PI)†; AHA-
IPA34170267 (Innovative
Project Award)†
Steven D. Duke University Medical None None None None None None None
Crowley Center
Alfonso Eirin Mayo Clinic None None None None None None None
David H. Ellison Oregon Health and NIDDK†; Department of None None None None None None
Science University Veterans Affairs†; Fondation
LeDucq†
Costantino Weill Cornell Medicine/ NIH† None None None None Broadview None
Iadecola Feil Family Brain and Ventures†
Mind Research Institute
Theodore W. University of California, None None None None None None None
Kurtz San Francisco
David L. Medical College of NIH†; AHA† None None None None None None
Mattson Wisconsin
John J. Mullins University of Edinburgh None None None None None None None
Medical School
United Kingdom
Jeffrey Osborn University of Kentucky None None None None None None None

(Continued )
Lerman et al   Animal Models of Hypertension   e25

Writing Group Disclosures Continued

Other Speakers’
Writing Group Research Bureau/ Expert Ownership Consultant/
Member Employment Research Grant Support Honoraria Witness Interest Advisory Board Other
Jane F. University of Mississippi NIH (PI, project on P01, PI, None None None None None None
Reckelhoff Medical Center P20, Co-PI, R01)†
Rhian M. Touyz University of British Heart Foundation None None Novartis† None Alnylam* None
Glasgow Institute of (operating grant)†; Medical
Cardiovascular and Research Council (operating
Medical Sciences grant)†
United Kingdom
This table represents the relationships of writing group members that may be perceived as actual or reasonably perceived conflicts of interest as reported on the
Disclosure Questionnaire, which all members of the writing group are required to complete and submit. A relationship is considered to be “significant” if (a) the person
receives $10 000 or more during any 12-month period, or 5% or more of the person’s gross income; or (b) the person owns 5% or more of the voting stock or share of
the entity or owns $10 000 or more of the fair market value of the entity. A relationship is considered to be “modest” if it is less than “significant” under the preceding
definition.
*Modest.
†Significant.

Reviewer Disclosures

Speakers’
Research Other Research Bureau/ Expert Ownership Consultant/
Reviewer Employment Grant Support Honoraria Witness Interest Advisory Board Other
Stephen R. Daniels University of Colorado None None None None None None None
Donald D. Heistad University of Iowa None None None None None None None
R. Clinton Webb Augusta University None None None None None None None
This table represents the relationships of reviewers that may be perceived as actual or reasonably perceived conflicts of interest as reported on the Disclosure
Questionnaire, which all reviewers are required to complete and submit. A relationship is considered to be “significant” if (a) the person receives $10 000 or more during
any 12-month period, or 5% or more of the person’s gross income; or (b) the person owns 5% or more of the voting stock or share of the entity, or owns $10 000 or
Downloaded from http://ahajournals.org by on March 14, 2019

more of the fair market value of the entity. A relationship is considered to be “modest” if it is less than “significant” under the preceding definition.

References 11. Pfeffer JM, Pfeffer MA, Fishbein MC, Frohlich ED. Cardiac function
and morphology with aging in the spontaneously hypertensive rat. Am J
1. Yang SS, Morimoto T, Rai T, Chiga M, Sohara E, Ohno M, Uchida K, Lin
Physiol 1979;237:H461–H468.
SH, Moriguchi T, Shibuya H, Kondo Y, Sasaki S, Uchida S. Molecular
12. Bing OH, Conrad CH, Boluyt MO, Robinson KG, Brooks WW. Studies of
pathogenesis of pseudohypoaldosteronism type II: generation and analy-
prevention, treatment and mechanisms of heart failure in the aging sponta-
sis of a Wnk4(D561A/+) knockin mouse model. Cell Metab. 2007;5:331–
neously hypertensive rat. Heart Fail Rev. 2002;7:71–88.
344. doi: 10.1016/j.cmet.2007.03.009
13. Hultström M. Development of structural kidney damage in sponta-
2. Pradervand S, Wang Q, Burnier M, Beermann F, Horisberger JD,
neously hypertensive rats. J Hypertens. 2012;30:1087–1091. doi:
Hummler E, Rossier BC. A mouse model for Liddle’s syndrome. J Am
10.1097/HJH.0b013e328352b89a
Soc Nephrol. 1999;10:2527–2533. 14. Bailey EL, Smith C, Sudlow CL, Wardlaw JM. Is the spontaneously
3. Galis ZS, Thrasher T, Reid DM, Stanley DV, Oh YS. Investing in high blood hypertensive stroke prone rat a pertinent model of sub cortical isch-
pressure research: a National Institutes of Health perspective. Hypertension. emic stroke? A systematic review. Int J Stroke. 2011;6:434–444. doi:
2013;61:757–761. doi: 10.1161/HYPERTENSIONAHA.111.00770 10.1111/j.1747-4949.2011.00659.x
4. Reproducibility Issues in Research with Animals and Animal Models: 15. Werber AH, Baumbach GL, Wagner DV, Mark AL, Heistad DD.
Workshop in Brief. Washington, DC: National Academies Press; 2015:8. Factors that influence stroke in Dahl salt-sensitive rats. Hypertension.
5. Sjoberg EA. Logical fallacies in animal model research. Behav Brain 1985;7:59–64.
Funct. 2017;13:3. doi: 10.1186/s12993-017-0121-8 16. Doi R, Masuyama T, Yamamoto K, Doi Y, Mano T, Sakata Y, Ono K,
6. Zeiss CJ, Johnson LK. Bridging the gap between reproducibility and Kuzuya T, Hirota S, Koyama T, Miwa T, Hori M. Development of dif-
translation: data resources and approaches. ILAR J. 2017;58:1–3. doi: ferent phenotypes of hypertensive heart failure: systolic versus diastolic
10.1093/ilar/ilx017 failure in Dahl salt-sensitive rats. J Hypertens. 2000;18:111–120.
7. Avey MT, Moher D, Sullivan KJ, Fergusson D, Griffin G, Grimshaw 17. Wilde E, Aubdool AA, Thakore P, Baldissera L Jr, Alawi KM, Keeble
JM, Hutton B, Lalu MM, Macleod M, Marshall J, Mei SH, Rudnicki J, Nandi M, Brain SD. Tail-cuff technique and its influence on central
M, Stewart DJ, Turgeon AF, McIntyre L; Canadian Critical Care blood pressure in the mouse. J Am Heart Assoc. 2017;6;e005204. doi:
Translational Biology Group. The devil is in the details: incomplete 10.1161/JAHA.116.005204
reporting in preclinical animal research. PLoS One. 2016;11:e0166733. 18. Li Y, Wei FF, Wang S, Cheng YB, Wang JG. Cardiovascular risks asso-
doi: 10.1371/journal.pone.0166733 ciated with diastolic blood pressure and isolated diastolic hypertension.
8. Reichlin TS, Vogt L, Würbel H. The researchers’ view of scientific rigor- Curr Hypertens Rep. 2014;16:489. doi: 10.1007/s11906-014-0489-x
survey on the conduct and reporting of in vivo research. PLoS One. 19. Kurtz TW, Griffin KA, Bidani AK, Davisson RL, Hall JE.
2016;11:e0165999. doi: 10.1371/journal.pone.0165999 Recommendations for blood pressure measurement in humans and exper-
9. McGonigle P, Ruggeri B. Animal models of human disease: challenges imental animals, part 2: blood pressure measurement in experimental
in enabling translation. Biochem Pharmacol. 2014;87:162–171. doi: animals: a statement for professionals from the subcommittee of profes-
10.1016/j.bcp.2013.08.006 sional and public education of the American Heart Association Council
10. Pinto YM, Paul M, Ganten D. Lessons from rat models of hypertension: on High Blood Pressure Research. Hypertension. 2005;45:299–310. doi:
from Goldblatt to genetic engineering. Cardiovasc Res. 1998;39:77–88. 10.1161/01.HYP.0000150857.39919.cb
e26  Hypertension   TBD 2019

20. Sim JJ, Bhandari SK, Shi J, Liu IL, Calhoun DA, McGlynn EA, Kalantar- 44. Doggrell SA, Brown L. Rat models of hypertension, cardiac hypertrophy
Zadeh K, Jacobsen SJ. Characteristics of resistant hypertension in a large, and failure. Cardiovasc Res. 1998;39:89–105.
ethnically diverse hypertension population of an integrated health system. 45. Padmanabhan S, Joe B. Towards precision medicine for hypertension: a
Mayo Clin Proc. 2013;88:1099–1107. doi: 10.1016/j.mayocp.2013.06.017 review of genomic, epigenomic, and microbiomic effects on blood pres-
21. Mancia G, Dell’Oro R, Trevano FQ, Grassi G. Novel agents in develop- sure in experimental rat models and humans. Physiol Rev. 2017;97:1469–
ment. In: McGinnis GT, ed. Clinical Pharmacology and Therapeutics of 1528. doi: 10.1152/physrev.00035.2016
Hypertension. Amsterdam: Elsevier; 2008:391–412. 46. Rapp JP. Genetic analysis of inherited hypertension in the rat. Physiol Rev.
22. Zandberg P. Animal models in experimental hypertension: relevance to 2000;80:135–172. doi: 10.1152/physrev.2000.80.1.135
drug testing and discovery. In: Van Zwieten PA, ed. Pharmacology of 47. Okamoto K, Aoki K. Development of a strain of spontaneously hyperten-
Antihypertensive Drugs. Amsterdam: Elsevier; 1984:6–45. sive rats. Jpn Circ J. 1963;27:282–293.
23. Greek CR, Greek JS. Sacred Cows and Golden Geese: The Human Cost 48. Kurtz TW, Morris RC Jr. Biological variability in Wistar-Kyoto rats:
of Experiments on Animals. New York: Continuum; 2000. implications for research with the spontaneously hypertensive rat.
24. Ondetti MA. From peptides to peptidases: a chronicle of drug Hypertension. 1987;10:127–131.
discovery. Annu Rev Pharmacol Toxicol. 1994;34:1–16. doi: 49. Okamoto K, Yamoci Y, Nagaoka A. Establishment of the stroke-
10.1146/annurev.pa.34.040194.000245 prone spontaneously hypertensive rat (SHR). Circ Res. 1974;34(suppl
25. Pfeffer MA. Janice M. Pfeffer Memorial Lecture. J Card Fail. I):I-143–I153.
2002;8(suppl):S248–S252. doi: 10.1054/jcaf.2002.129275 50. Nabika T, Ohara H, Kato N, Isomura M. The stroke-prone spontaneously
26. Wood JM, Maibaum J, Rahuel J, Grütter MG, Cohen NC, Rasetti V, Rüger hypertensive rat: still a useful model for post-GWAS genetic studies?
H, Göschke R, Stutz S, Fuhrer W, Schilling W, Rigollier P, Yamaguchi Y, Hypertens Res. 2012;35:477–484. doi: 10.1038/hr.2012.30
Cumin F, Baum HP, Schnell CR, Herold P, Mah R, Jensen C, O’Brien 51. Joe B. Dr Lewis Kitchener Dahl, the Dahl rats, and the “inconvenient
E, Stanton A, Bedigian MP. Structure-based design of aliskiren, a truth” about the genetics of hypertension. Hypertension. 2015;65:963–
novel orally effective renin inhibitor. Biochem Biophys Res Commun. 969. doi: 10.1161/HYPERTENSIONAHA.114.04368
2003;308:698–705. 52. Tschopp TB, Baumgartner HR. Defective platelet adhesion and aggregation
27. Wood JM, Schnell CR, Cumin F, Menard J, Webb RL. Aliskiren, a novel, on subendothelium exposed in vivo or in vitro to flowing blood of fawn-
orally effective renin inhibitor, lowers blood pressure in marmosets and hooded rats and storage pool disease. Thromb Haemost. 1977;38:620–629.
spontaneously hypertensive rats. J Hypertens. 2005;23:417–426. 53. Provoost A, De Keijzer M. The fawn-hooded rat: a model for chronic
28. Jensen C, Herold P, Brunner HR. Aliskiren: the first renin inhibi- renal failure. In: Gretz N, Strauch M, eds. Experimental and Genetic Rat
tor for clinical treatment. Nat Rev Drug Discov. 2008;7:399–410. doi: Models of Chronic Renal Failure. Basel: Karger; 1993:100–114.
10.1038/nrd2550 54. Bianchi G, Fox U, Imbasciati E. The development of a new strain of spon-
29. Tsui BM, Kraitchman DL. Recent advances in small-animal cardiovas- taneously hypertensive rats. Life Sci. 1974;14:339–347.
cular imaging. J Nucl Med. 2009;50:667–670. doi: 10.2967/jnumed. 55. Bianchi G, Fox U, Di Francesco GF, Giovanetti AM, Pagetti D. Blood
108.058479 pressure changes produced by kidney cross-transplantation between
30. Zambraski EJ, Ciccone CD, Izzo JL Jr. The role of the sympathetic ner- spontaneously hypertensive rats and normotensive rats. Clin Sci Mol Med.
vous system in 2-kidney DOCA-hypertensive Yucatan miniature swine. 1974;47:435–448.
Clin Exp Hypertens A. 1986;8:411–424. 56. Ge Y, Fan F, Didion SP, Roman RJ. Impaired myogenic response of
31. Ciccone CD, Zambraski EJ. Effects of acute renal denervation on the afferent arteriole contributes to the increased susceptibility to renal
kidney function in deoxycorticosterone acetate-hypertensive swine. disease in Milan normotensive rats. Physiol Rep. 2017;5:e13089. doi:
Downloaded from http://ahajournals.org by on March 14, 2019

Hypertension. 1986;8:925–931. 10.14814/phy2.13089


32. Olmsted F, Page IH. Hemodynamic aspects of prolonged infusion of 57. Dupont J, Dupont JC, Froment A, Milon H, Vincent M. Selection of
angiotensin into unanesthetized dogs. Circ Res. 1965;16:140–149. three strains of rats with spontaneously different levels of blood pressure.
33. McCubbin JW, DeMoura RS, Page IH, Olmsted F. Arterial hyper- Biomedicine. 1973;19:36–41.
tension elicited by subpressor amounts of angiotensin. Science. 58. Vincent M, Dupont J, Sassard J. Plasma renin activity as a function of age
1965;149:1394–1395. in two new strains of spontaneously hypertensive and normotensive rats.
34. Hofstaetter JG, Blouin S, Friehs I, Klaushofer K, Roschger P. No effect of Clin Sci Mol Med. 1976;50:103–107.
short-term hypertension on bone matrix mineralization in a surgical ani- 59. Ma MCJ, Pettus JM, Jakoubek JA, Traxler MG, Clark KC, Mennie
mal model in immature rabbits. Clin Exp Hypertens. 2012;34:107–112. AK, Kwitek AE. Contribution of independent and pleiotropic genetic
doi: 10.3109/10641963.2011.601382 effects in the metabolic syndrome in a hypertensive rat. PLoS One.
35. Fossum TW, Baltzer WI, Miller MW, Aguirre M, Whitlock D, Solter P, 2017;12:e0182650. doi: 10.1371/journal.pone.0182650
Makarski LA, McDonald MM, An MY, Humphrey JD. A novel aortic 60. Ben-Ishay D, Zamir N, Feurstein G, Kobrin I, Le Quan-Bui KH, Devynck
coarctation model for studying hypertension in the pig. J Invest Surg. MA. Distinguishing traits in the Sabra hypertension-prone (SBH) and
2003;16:35–44. hypertension-resistant (SBN) rats. Clin Exp Hypertens. 1981;3:737–747.
36. Cody RJ Jr, Rodger RF, Hartley LH, Burton J, Herd JA. Acute hyperten- 61. Deleted in proof.
sion in a nonhuman primate: humoral and hemodynamic mechanisms. 62. Yagil C, Katni G, Rubattu S, Stolpe C, Kreutz R, Lindpaintner K, Ganten
Hypertension. 1982;4:219–225. D, Ben-Ishay D, Yagil Y. Development, genotype and phenotype of a new
37. Goldblatt H, Lynch J, Hanzal RF, Summerville WW. Studies on experi- colony of the Sabra hypertension prone (SBH/y) and resistant (SBN/y) rat
mental hypertension, I: the production of persistent elevation of systolic model of slat sensitivity and resistance. J Hypertens. 1996;14:1175–1182.
blood pressure by means of renal ischemia. J Exp Med. 1934;59:347–379. 63. Cowley AW Jr. The genetic dissection of essential hypertension. Nat Rev
38. Panek RL, Ryan MJ, Weishaar RE, Taylor DG Jr. Development of a Genet. 2006;7:829–840. doi: 10.1038/nrg1967
high renin model of hypertension in the cynomolgus monkey. Clin Exp 64. Stoll M, Jacob HJ. Genetic rat models of hypertension: relationship to
Hypertens A. 1991;13:1395–1414. human hypertension. Curr Hypertens Rep. 2001;3:157–164.
39. Lerman LO, Schwartz RS, Grande JP, Sheedy PF, Romero JC. Noninvasive 65. Geurts AM, Cost GJ, Freyvert Y, Zeitler B, Miller JC, Choi VM, Jenkins
evaluation of a novel swine model of renal artery stenosis. J Am Soc SS, Wood A, Cui X, Meng X, Vincent A, Lam S, Michalkiewicz M,
Nephrol. 1999;10:1455–1465. Schilling R, Foeckler J, Kalloway S, Weiler H, Ménoret S, Anegon I,
40. Chade AR, Rodriguez-Porcel M, Grande JP, Krier JD, Lerman A, Romero Davis GD, Zhang L, Rebar EJ, Gregory PD, Urnov FD, Jacob HJ, Buelow
JC, Napoli C, Lerman LO. Distinct renal injury in early atherosclerosis R. Knockout rats via embryo microinjection of zinc-finger nucleases.
and renovascular disease. Circulation. 2002;106:1165–1171. Science. 2009;325:433. doi: 10.1126/science.1172447
41. Rhoads MK, Goleva SB, Beierwaltes WH, Osborn JL. Renal vascu- 66. Rudemiller NP, Mattson DL. Candidate genes for hypertension: insights
lar and glomerular pathologies associated with spontaneous hyper- from the Dahl S rat. Am J Physiol Renal Physiol. 2015;309:F993–F995.
tension in the nonhuman primate Chlorocebus aethiops sabaeus. Am doi: 10.1152/ajprenal.00092.2015
J Physiol Regul Integr Comp Physiol. 2017;313:R211–R218. doi: 67. Pravenec M, Křen V, Landa V, Mlejnek P, Musilová A, Šilhavý J,
10.1152/ajpregu.00026.2017 Šimáková M, Zídek V. Recent progress in the genetics of spontaneously
42. Rat genome database. http://rgd.mcw.edu/rgdweb/search/strains.html?ter hypertensive rats. Physiol Res. 2014;63(suppl 1):S1–S8.
m=hypertensive+rats&obj=strain). Accessed October 18, 2017. 68. Doris PA. Genetics of hypertension: an assessment of progress in the
43. Festing MFW. Index of major rat strains. http://www.informatics.jax.org/ spontaneously hypertensive rat. Physiol Genomics. 2017;49:601–617.
inbred_strains/rat/STRAINS.shtml. Accessed October 18, 2017. doi: 10.1152/physiolgenomics.00065.2017
Lerman et al   Animal Models of Hypertension   e27

69. Bianchi G, Ferrari P, Barber B. The Milan hypertensive strain. In: de 85. Takahashi N, Chernavvsky DR, Gomez RA, Igarashi P, Gitelman HJ,
Jong W, ed. Handbook of Hypertension. Amsterdam: Elsevier; 1984: Smithies O. Uncompensated polyuria in a mouse model of Bartter’s
328–349. syndrome. Proc Natl Acad Sci U  S A. 2000;97:5434–5439. doi:
70. Cusi D, Alberghini E, Pati P, Tripodi G, Barlassina C, Colombo R, Cova 10.1073/pnas.090091297
T, Niutta E, Vezzoli G, Bianchi G. Pathogenetic mechanisms in essential 86. Bailey MA, Craigie E, Livingstone DEW, Kotelevtsev YV, Al-Dujaili
hypertension: analogies between a rat model and the human disease. Int J EAS, Kenyon CJ, Mullins JJ. Hsd11b2 haploinsufficiency in mice causes
Cardiol. 1989;25(suppl 1):S29–S36. salt sensitivity of blood pressure. Hypertension. 2011;57:515–520. doi:
71. Mattson DL, Meister CJ, Marcelle ML. Dietary protein source determines 10.1161/HYPERTENSIONAHA.110.163782
the degree of hypertension and renal disease in the Dahl salt-sensitive 87. Kotelevtsev Y, Brown RW, Fleming S, Kenyon C, Edwards CR, Seckl JR,
rat. Hypertension. 2005;45:736–741. doi: 10.1161/01.HYP.0000153318. Mullins JJ. Hypertension in mice lacking 11beta-hydroxysteroid dehy-
74544.cc drogenase type 2. J Clin Invest. 1999;103:683–689. doi: 10.1172/JCI4445
72. Kwitek AE, Tonellato PJ, Chen D, Gullings-Handley J, Cheng YS, 88. Ueda K, Nishimoto M, Hirohama D, Ayuzawa N, Kawarazaki W,
Twigger S, Scheetz TE, Casavant TL, Stoll M, Nobrega MA, Shiozawa Watanabe A, Shimosawa T, Loffing J, Zhang MZ, Marumo T, Fujita T.
M, Soares MB, Sheffield VC, Jacob HJ. Automated construction of high- Renal dysfunction induced by kidney-specific gene deletion of Hsd11b2
density comparative maps between rat, human, and mouse. Genome Res. as a primary cause of salt-dependent hypertension. Hypertension.
2001;11:1935–1943. doi: 10.1101/gr.173701 2017;70:111–118. doi: 10.1161/HYPERTENSIONAHA.116.08966
73. Kohara K, Tabara Y, Nakura J, Imai Y, Ohkubo T, Hata A, Soma M, 89. Cowley AW Jr, Yang C, Zheleznova NN, Staruschenko A, Kurth T, Rein L,
Nakayama T, Umemura S, Hirawa N, Ueshima H, Kita Y, Ogihara T, Kumar V, Sadovnikov K, Dayton A, Hoffman M, Ryan RP, Skelton MM,
Katsuya T, Takahashi N, Tokunaga K, Miki T. Identification of hyper- Salehpour F, Ranji M, Geurts A. Evidence of the importance of Nox4
tension-susceptibility genes and pathways by a systemic multiple candi- in production of hypertension in Dahl salt-sensitive rats. Hypertension.
date gene approach: the Millennium Genome Project for Hypertension. 2016;67:440–450. doi: 10.1161/HYPERTENSIONAHA.115.06280
Hypertens Res. 2008;31:203–212. doi: 10.1291/hypres.31.203 90. Feng D, Yang C, Geurts AM, Kurth T, Liang M, Lazar J, Mattson DL,
74. Gordon JW, Scangos GA, Plotkin DJ, Barbosa JA, Ruddle FH. Genetic O’Connor PM, Cowley AW Jr. Increased expression of NAD(P)H oxi-
transformation of mouse embryos by microinjection of purified DNA. dase subunit p67(phox) in the renal medulla contributes to excess oxida-
Proc Natl Acad Sci U S A. 1980;77:7380–7384. tive stress and salt-sensitive hypertension. Cell Metab. 2012;15:201–208.
75. Palmiter RD, Brinster RL. Transgenic mice. Cell. 1985;41:343–345. doi: 10.1016/j.cmet.2012.01.003
76. Zhang K, Mir SA, Hightower CM, Miramontes-Gonzalez JP, Maihofer 91. Gomez-Arroyo J, Voelkel NF, Bogaard HJ, Taraseviciene-Stewart L.
AX, Chen Y, Mahata SK, Nievergelt CM, Schork NJ, Freedman BI, Usefulness of a mouse model of reversible pulmonary arterial hyper-
Vaingankar SM, O’Connor DT. Molecular mechanism for hypertensive tension: be cautious, choose carefully. Am J Respir Crit Care Med.
renal disease: differential regulation of chromogranin A expression at 2012;185:1326. doi: 10.1164/ajrccm.185.12.1326a
3’-untranslated region polymorphism C+87T by microRNA-107. J Am 92. Clark AF, Sharp MG, Morley SD, Fleming S, Peters J, Mullins JJ.
Soc Nephrol. 2015;26:1816–1825. doi: 10.1681/ASN.2014060537 Renin-1 is essential for normal renal juxtaglomerular cell granulation
77. Mir SA, Zhang K, Milic M, Gu Y, Rieg T, Ziegler M, Vaingankar SM. and macula densa morphology. J Biol Chem. 1997;272:18185–18190.
Analysis and validation of traits associated with a single nucleotide poly- 93. Moreno C, Hoffman M, Stodola TJ, Didier DN, Lazar J, Geurts
morphism Gly364Ser in catestatin using humanized chromogranin A mouse AM, North PE, Jacob HJ, Greene AS. Creation and characteriza-
models. J Hypertens. 2016;34:68–78. doi: 10.1097/HJH.0000000000000760 tion of a renin knockout rat. Hypertension. 2011;57:614–619. doi:
78. Elijovich F, Weinberger MH, Anderson CA, Appel LJ, Bursztyn M, 10.1161/HYPERTENSIONAHA.110.163840
Downloaded from http://ahajournals.org by on March 14, 2019

Cook NR, Dart RA, Newton-Cheh CH, Sacks FM, Laffer CL; on 94. Sharp MG, Fettes D, Brooker G, Clark AF, Peters J, Fleming S, Mullins
behalf of the American Heart Association Professional and Public JJ. Targeted inactivation of the Ren-2 gene in mice. Hypertension.
Education Committee of the Council on Hypertension; Council 1996;28:1126–1131.
on Functional Genomics and Translational Biology; and Stroke 95. Gurley SB, Riquier-Brison ADM, Schnermann J, Sparks MA, Allen
Council. Salt sensitivity of blood pressure: a scientific statement AM, Haase VH, Snouwaert JN, Le TH, McDonough AA, Koller BH,
from the American Heart Association [published correction appears Coffman TM. AT1A angiotensin receptors in the renal proximal
in Hypertension. 2016;88:e62]. Hypertension. 2016;68:e7–e46. doi: tubule regulate blood pressure. Cell Metab. 2011;13:469–475. doi:
10.1161/HYP.0000000000000047 10.1016/j.cmet.2011.03.001
79. Li H, Weatherford ET, Davis DR, Keen HL, Grobe JL, Daugherty A, 96. Fukamizu A, Sugimura K, Takimoto E, Sugiyama F, Seo MS, Takahashi
Cassis LA, Allen AM, Sigmund CD. Renal proximal tubule angiotensin S, Hatae T, Kajiwara N, Yagami K, Murakami K. Chimeric renin-
AT1A receptors regulate blood pressure. Am J Physiol Regul Integr Comp angiotensin system demonstrates sustained increase in blood pressure of
Physiol. 2011;301:R1067–R1077. doi: 10.1152/ajpregu.00124.2011 transgenic mice carrying both human renin and human angiotensinogen
80. Kantachuvesiri S, Fleming S, Peters J, Peters B, Brooker G, Lammie genes. J Biol Chem. 1993;268:11617–11621.
AG, McGrath I, Kotelevtsev Y, Mullins JJ. Controlled hypertension, 97. Ganten D, Wagner J, Zeh K, Bader M, Michel JB, Paul M, Zimmermann
a transgenic toggle switch reveals differential mechanisms under- F, Ruf P, Hilgenfeldt U, Ganten U. Species specificity of renin kinetics
lying vascular disease. J Biol Chem. 2001;276:36727–36733. doi: in transgenic rats harboring the human renin and angiotensinogen genes.
10.1074/jbc.M103296200 Proc Natl Acad Sci U S A. 1992;89:7806–7810.
81. Chowdhury JA, Liu CH, Zuber AM, O’Shaughnessy KM. An inducible 98. Yoshimi K, Kaneko T, Voigt B, Mashimo T. Allele-specific genome
transgenic mouse model for familial hypertension with hyperkalaemia editing and correction of disease-associated phenotypes in rats
(Gordon’s syndrome or pseudohypoaldosteronism type II). Clin Sci using the CRISPR-Cas platform. Nat Commun. 2014;5:4240. doi:
(Lond). 2013;124:701–708. doi: 10.1042/CS20120430 10.1038/ncomms5240
82. Ronzaud C, Loffing-Cueni D, Hausel P, Debonneville A, Malsure SR, 99. Yoshimi K, Kunihiro Y, Kaneko T, Nagahora H, Voigt B, Mashimo T.
Fowler-Jaeger N, Boase NA, Perrier R, Maillard M, Yang B, Stokes JB, ssODN-mediated knock-in with CRISPR-Cas for large genomic regions
Koesters R, Kumar S, Hummler E, Loffing J, Staub O. Renal tubular in zygotes. Nat Commun. 2016;7:10431. doi: 10.1038/ncomms10431
NEDD4-2 deficiency causes NCC-mediated salt-dependent hypertension. 100. Ahn D, Ge Y, Stricklett PK, Gill P, Taylor D, Hughes AK, Yanagisawa
J Clin Invest. 2013;123:657–665. doi: 10.1172/JCI61110 M, Miller L, Nelson RD, Kohan DE. Collecting duct-specific knockout

83. Schultheis PJ, Lorenz JN, Meneton P, Nieman ML, Riddle TM, of endothelin-1 causes hypertension and sodium retention. J Clin Invest.
Flagella M, Duffy JJ, Doetschman T, Miller ML, Shull GE. Phenotype 2004;114:504–511. doi: 10.1172/JCI21064
resembling Gitelman’s syndrome in mice lacking the apical 101. Stuart D, Chapman M, Rees S, Woodward S, Kohan DE. Myocardial,
Na+-Cl- cotransporter of the distal convoluted tubule. J Biol Chem. smooth muscle, nephron, and collecting duct gene targeting reveals
1998;273:29150–29155. the organ sites of endothelin A receptor antagonist fluid retention. J
84. Zhou X, Zhang Z, Shin MK, Horwitz SB, Levorse JM, Zhu L, Sharif- Pharmacol Exp Ther. 2013;346:182–189. doi: 10.1124/jpet.113.205286
Rodriguez W, Streltsov DY, Dajee M, Hernandez M, Pan Y, Urosevic- 102. Ponce de Leon V, Merillat AM, Tesson L, Anegon I, Hummler E. Generation
Price O, Wang L, Forrest G, Szeto D, Zhu Y, Cui Y, Michael B, Balogh of TALEN-mediated GRdim knock-in rats by homologous recombina-
LA, Welling PA, Wade JB, Roy S, Sullivan KA. Heterozygous disrup- tion. PLoS One. 2014;9:e88146. doi: 10.1371/journal.pone.0088146
tion of renal outer medullary potassium channel in rats is associated 103. Ji D, Zhao G, Songstad A, Cui X, Weinstein EJ. Efficient creation of
with reduced blood pressure. Hypertension. 2013;62:288–294. doi: an APOE knockout rabbit. Transgenic Res. 2015;24:227–235. doi:
10.1161/HYPERTENSIONAHA.111.01051 10.1007/s11248-014-9834-8
e28  Hypertension   TBD 2019

104. Flister MJ, Tsaih SW, O’Meara CC, Endres B, Hoffman MJ, Geurts AM, protein. Methods Mol Biol. 2014;1148:229–238. doi: 10.1007/
Dwinell MR, Lazar J, Jacob HJ, Moreno C. Identifying multiple caus- 978-1-4939-0470-9_15
ative genes at a single GWAS locus. Genome Res. 2013;23:1996–2002. 124. Buckley C, Carvalho MT, Young LK, Rider SA, McFadden C, Berlage
doi: 10.1101/gr.160283.113 C, Verdon RF, Taylor JM, Girkin JM, Mullins JJ. Precise spatio-tem-
105. Wang X, Yamada S, LaRiviere WB, Ye H, Bakeberg JL, Irazabal MV, poral control of rapid optogenetic cell ablation with mem-KillerRed in
Chebib FT, van Deursen J, Harris PC, Sussman CR, Behfar A, Ward CJ, Zebrafish. Sci Rep. 2017;7:5096. doi: 10.1038/s41598-017-05028-2
Torres VE. Generation and phenotypic characterization of Pde1a mutant 125. Wain LV, Verwoert GC, O’Reilly PF, Shi G, Johnson T, Johnson AD,
mice. PLoS One. 2017;12:e0181087. doi: 10.1371/journal.pone.0181087 Bochud M, Rice KM, Henneman P, Smith AV, Ehret GB, Amin N,
106. Cheng X, Waghulde H, Mell B, Morgan EE, Pruett-Miller SM, Joe Larson MG, Mooser V, Hadley D, Dörr M, Bis JC, Aspelund T, Esko T,
B. Positional cloning of quantitative trait nucleotides for blood pres- Janssens AC, Zhao JH, Heath S, Laan M, Fu J, Pistis G, Luan J, Arora P,
sure and cardiac QT-interval by targeted CRISPR/Cas9 editing of a Lucas G, Pirastu N, Pichler I, Jackson AU, Webster RJ, Zhang F, Peden
novel long non-coding RNA. PLoS Genet. 2017;13:e1006961. doi: JF, Schmidt H, Tanaka T, Campbell H, Igl W, Milaneschi Y, Hottenga
10.1371/journal.pgen.1006961 JJ, Vitart V, Chasman DI, Trompet S, Bragg-Gresham JL, Alizadeh BZ,
107. Fan ZD, Zhang L, Shi Z, Gan XB, Gao XY, Zhu GQ. Artificial
Chambers JC, Guo X, Lehtimäki T, Kühnel B, Lopez LM, Polašek O,
microRNA interference targeting AT(1a) receptors in paraventricular Boban M, Nelson CP, Morrison AC, Pihur V, Ganesh SK, Hofman A,
nucleus attenuates hypertension in rats. Gene Ther. 2012;19:810–817. Kundu S, Mattace-Raso FU, Rivadeneira F, Sijbrands EJ, Uitterlinden
doi: 10.1038/gt.2011.145 AG, Hwang SJ, Vasan RS, Wang TJ, Bergmann S, Vollenweider P,
108. Xue B, Beltz TG, Johnson RF, Guo F, Hay M, Johnson AK. PVN adeno- Waeber G, Laitinen J, Pouta A, Zitting P, McArdle WL, Kroemer HK,
virus-siRNA injections silencing either NOX2 or NOX4 attenuate aldo- Völker U, Völzke H, Glazer NL, Taylor KD, Harris TB, Alavere H, Haller
sterone/NaCl-induced hypertension in mice. Am J Physiol Heart Circ T, Keis A, Tammesoo ML, Aulchenko Y, Barroso I, Khaw KT, Galan P,
Physiol. 2012;302:H733–H741. doi: 10.1152/ajpheart.00873.2011 Hercberg S, Lathrop M, Eyheramendy S, Org E, Sõber S, Lu X, Nolte
109. Liu Y, Taylor NE, Lu L, Usa K, Cowley AW Jr, Ferreri NR, Yeo NC, IM, Penninx BW, Corre T, Masciullo C, Sala C, Groop L, Voight BF,
Liang M. Renal medullary microRNAs in Dahl salt-sensitive rats: Melander O, O’Donnell CJ, Salomaa V, d’Adamo AP, Fabretto A, Faletra
miR-29b regulates several collagens and related genes. Hypertension. F, Ulivi S, Del Greco F, Facheris M, Collins FS, Bergman RN, Beilby
2010;55:974–982. doi: 10.1161/HYPERTENSIONAHA.109.144428 JP, Hung J, Musk AW, Mangino M, Shin SY, Soranzo N, Watkins H,
110. Hasegawa Y, Daitoku Y, Sekiguchi K, Tanimoto Y, Mizuno-Iijima S, Goel A, Hamsten A, Gider P, Loitfelder M, Zeginigg M, Hernandez D,
Mizuno S, Kajiwara N, Ema M, Miwa Y, Mekada K, Yoshiki A, Takahashi Najjar SS, Navarro P, Wild SH, Corsi AM, Singleton A, de Geus EJ,
S, Sugiyama F, Yagami K. Novel ROSA26 Cre-reporter knock-in Willemsen G, Parker AN, Rose LM, Buckley B, Stott D, Orru M, Uda
C57BL/6N mice exhibiting green emission before and red emission after M, van der Klauw MM, Zhang W, Li X, Scott J, Chen YD, Burke GL,
Cre-mediated recombination. Exp Anim. 2013;62:295–304. Kähönen M, Viikari J, Döring A, Meitinger T, Davies G, Starr JM,
111. Yang D, Song J, Zhang J, Xu J, Zhu T, Wang Z, Lai L, Chen YE. Emilsson V, Plump A, Lindeman JH, Hoen PA, König IR, Felix JF,
Identification and characterization of rabbit ROSA26 for gene knock- Clarke R, Hopewell JC, Ongen H, Breteler M, Debette S, Destefano AL,
in and stable reporter gene expression. Sci Rep. 2016;6:25161. doi: Fornage M, Mitchell GF, Smith NL, Holm H, Stefansson K, Thorleifsson
10.1038/srep25161 G, Thorsteinsdottir U, Samani NJ, Preuss M, Rudan I, Hayward C,
112. Buehr M, Meek S, Blair K, Yang J, Ure J, Silva J, McLay R, Hall J, Ying Deary IJ, Wichmann HE, Raitakari OT, Palmas W, Kooner JS, Stolk RP,
QL, Smith A. Capture of authentic embryonic stem cells from rat blasto- Jukema JW, Wright AF, Boomsma DI, Bandinelli S, Gyllensten UB,
cysts. Cell. 2008;135:1287–1298. doi: 10.1016/j.cell.2008.12.007 Wilson JF, Ferrucci L, Schmidt R, Farrall M, Spector TD, Palmer LJ,
Downloaded from http://ahajournals.org by on March 14, 2019

113. Li P, Tong C, Mehrian-Shai R, Jia L, Wu N, Yan Y, Maxson RE, Schulze Tuomilehto J, Pfeufer A, Gasparini P, Siscovick D, Altshuler D, Loos RJ,
EN, Song H, Hsieh CL, Pera MF, Ying QL. Germline competent embry- Toniolo D, Snieder H, Gieger C, Meneton P, Wareham NJ, Oostra BA,
onic stem cells derived from rat blastocysts. Cell. 2008;135:1299–1310. Metspalu A, Launer L, Rettig R, Strachan DP, Beckmann JS, Witteman
doi: 10.1016/j.cell.2008.12.006 JC, Erdmann J, van Dijk KW, Boerwinkle E, Boehnke M, Ridker PM,
114. Mullins LJ, Kenyon CJ, Bailey MA, Conway BR, Diaz ME, Mullins Jarvelin MR, Chakravarti A, Abecasis GR, Gudnason V, Newton-Cheh
JJ. Mineralocorticoid excess or glucocorticoid insufficiency: renal and C, Levy D, Munroe PB, Psaty BM, Caulfield MJ, Rao DC, Tobin MD,
metabolic phenotypes in a rat Hsd11b2 knockout model. Hypertension. Elliott P, van Duijn CM; LifeLines Cohort Study; EchoGen Consortium;
2015;66:e20. doi: 10.1161/HYP.0000000000000035 AortaGen Consortium; CHARGE Consortium Heart Failure Working
115. Geurts AM, Moreno C. Zinc-finger nucleases: new strategies to
Group; KidneyGen Consortium; CKDGen Consortium; Cardiogenics
target the rat genome. Clin Sci (Lond). 2010;119:303–311. doi: Consortium; CardioGram. Genome-wide association study identifies
10.1042/CS20100201 six new loci influencing pulse pressure and mean arterial pressure. Nat
116. Jacob HJ, Lazar J, Dwinell MR, Moreno C, Geurts AM. Gene targeting Genet. 2011;43:1005–1011. doi: 10.1038/ng.922
in the rat: advances and opportunities. Trends Genet. 2010;26:510–518. 126. Newton-Cheh C, Johnson T, Gateva V, Tobin MD, Bochud M, Coin L,
doi: 10.1016/j.tig.2010.08.006 Najjar SS, Zhao JH, Heath SC, Eyheramendy S, Papadakis K, Voight
117. Kleinstiver BP, Pattanayak V, Prew MS, Tsai SQ, Nguyen NT, Zheng BF, Scott LJ, Zhang F, Farrall M, Tanaka T, Wallace C, Chambers JC,
Z, Joung JK. High-fidelity CRISPR-Cas9 nucleases with no detect- Khaw KT, Nilsson P, van der Harst P, Polidoro S, Grobbee DE, Onland-
able genome-wide off-target effects. Nature. 2016;529:490–495. doi: Moret NC, Bots ML, Wain LV, Elliott KS, Teumer A, Luan J, Lucas G,
10.1038/nature16526 Kuusisto J, Burton PR, Hadley D, McArdle WL, Brown M, Dominiczak
118. Slaymaker IM, Gao L, Zetsche B, Scott DA, Yan WX, Zhang F.
A, Newhouse SJ, Samani NJ, Webster J, Zeggini E, Beckmann JS,
Rationally engineered Cas9 nucleases with improved specificity. Science. Bergmann S, Lim N, Song K, Vollenweider P, Waeber G, Waterworth
2016;351:84–88. doi: 10.1126/science.aad5227 DM, Yuan X, Groop L, Orho-Melander M, Allione A, Di Gregorio A,
119. Kleinstiver BP, Prew MS, Tsai SQ, Nguyen NT, Topkar VV, Zheng Z, Guarrera S, Panico S, Ricceri F, Romanazzi V, Sacerdote C, Vineis P,
Joung JK. Broadening the targeting range of Staphylococcus aureus Barroso I, Sandhu MS, Luben RN, Crawford GJ, Jousilahti P, Perola
CRISPR-Cas9 by modifying PAM recognition. Nat Biotechnol. M, Boehnke M, Bonnycastle LL, Collins FS, Jackson AU, Mohlke
2015;33:1293–1298. doi: 10.1038/nbt.3404 KL, Stringham HM, Valle TT, Willer CJ, Bergman RN, Morken MA,
120. Zetsche B, Gootenberg JS, Abudayyeh OO, Slaymaker IM, Makarova KS, Döring A, Gieger C, Illig T, Meitinger T, Org E, Pfeufer A, Wichmann
Essletzbichler P, Volz SE, Joung J, van der Oost J, Regev A, Koonin EV, HE, Kathiresan S, Marrugat J, O’Donnell CJ, Schwartz SM, Siscovick
Zhang F. Cpf1 is a single RNA-guided endonuclease of a class 2 CRISPR- DS, Subirana I, Freimer NB, Hartikainen AL, McCarthy MI, O’Reilly
Cas system. Cell. 2015;163:759–771. doi: 10.1016/j.cell.2015.09.038 PF, Peltonen L, Pouta A, de Jong PE, Snieder H, van Gilst WH, Clarke
121. Kriegel AJ, Mladinov D, Liang M. Translational study of microRNAs R, Goel A, Hamsten A, Peden JF, Seedorf U, Syvänen AC, Tognoni G,
and its application in kidney disease and hypertension research. Clin Sci Lakatta EG, Sanna S, Scheet P, Schlessinger D, Scuteri A, Dörr M, Ernst
(Lond). 2012;122:439–447. doi: 10.1042/CS20110159 F, Felix SB, Homuth G, Lorbeer R, Reffelmann T, Rettig R, Völker
122. Ivanova A, Signore M, Caro N, Greene ND, Copp AJ, Martinez-
U, Galan P, Gut IG, Hercberg S, Lathrop GM, Zelenika D, Deloukas
Barbera JP. In vivo genetic ablation by Cre-mediated expression of P, Soranzo N, Williams FM, Zhai G, Salomaa V, Laakso M, Elosua R,
diphtheria toxin fragment A. Genesis. 2005;43:129–135. doi: 10.1002/ Forouhi NG, Völzke H, Uiterwaal CS, van der Schouw YT, Numans ME,
gene.20162 Matullo G, Navis G, Berglund G, Bingham SA, Kooner JS, Connell JM,
123. Teh C, Korzh V. In vivo optogenetics for light-induced oxidative
Bandinelli S, Ferrucci L, Watkins H, Spector TD, Tuomilehto J, Altshuler
stress in transgenic zebrafish expressing the KillerRed photosensitizer D, Strachan DP, Laan M, Meneton P, Wareham NJ, Uda M, Jarvelin MR,
Lerman et al   Animal Models of Hypertension   e29

Mooser V, Melander O, Loos RJ, Elliott P, Abecasis GR, Caulfield M, 144. Osborn JW, Foss JD. Renal nerves and long-term control of arterial pres-
Munroe PB; Wellcome Trust Case Control Consortium. Genome-wide sure. Compr Physiol. 2017;7:263–320. doi: 10.1002/cphy.c150047
association study identifies eight loci associated with blood pressure. Nat 145. Martin S, Palmour RM, Goldwater R, Gutkowsa J, Hughes C, Hamet
Genet. 2009;41:666–676. doi: 10.1038/ng.361 P. Ervin FR. The response of hypertensive and normotensive male ver-
127. Levy D, Ehret GB, Rice K, Verwoert GC, Launer LJ, Dehghan A, Glazer vets (Cercopithecus aethiops) to cold pressor stress, captopril admin-
NL, Morrison AC, Johnson AD, Aspelund T, Aulchenko Y, Lumley T, istration and acute bolus of atrial natriuretic factor. Am J Hypertens.
Köttgen A, Vasan RS, Rivadeneira F, Eiriksdottir G, Guo X, Arking DE, 1990;3:27–32.
Mitchell GF, Mattace-Raso FU, Smith AV, Taylor K, Scharpf RB, Hwang SJ, 146. Harding J. Genomic tools for the use of nonhuman primates in transla-
Sijbrands EJ, Bis J, Harris TB, Ganesh SK, O’Donnell CJ, Hofman A, Rotter tional research. ILAR J. 2017;58:59–68. doi: 10.1093/ilar/ilw042
JI, Coresh J, Benjamin EJ, Uitterlinden AG, Heiss G, Fox CS, Witteman 147. Page IH. A method for producing persistent hypertension by cellophane.
JC, Boerwinkle E, Wang TJ, Gudnason V, Larson MG, Chakravarti A, Psaty Science. 1939;89:273–274.
BM, van Duijn CM. Genome-wide association study of blood pressure and 148. DeForrest JM, Scalese RJ, Oehl RS, Waldron TL, Mitch S, Brittain RJ,
hypertension. Nat Genet. 2009;41:677–687. doi: 10.1038/ng.384 Free CA, Asaad M, Burkett D. Perinephritis hypertension in Macaca
128. Gupta RM, Hadaya J, Trehan A, Zekavat SM, Roselli C, Klarin D, Emdin fascicularis (cynomolgus monkey): studies of the renin-angioten-
CA, Hilvering CRE, Bianchi V, Mueller C, Khera AV, Ryan RJH, Engreitz sin-aldosterone axis and renal hemodynamic function. J Hypertens.
JM, Issner R, Shoresh N, Epstein CB, de Laat W, Brown JD, Schnabel 1989;7:763–767.
RB, Bernstein BE, Kathiresan S. A genetic variant associated with five 149. Misra S, Gordon JD, Fu AA, Glockner JF, Chade AR, Mandrekar J,
vascular diseases is a distal regulator of endothelin-1 gene expression. Lerman L, Mukhopadhyay D. The porcine remnant kidney model
Cell. 2017;170:522–533.e15. doi: 10.1016/j.cell.2017.06.049 of chronic renal insufficiency. J Surg Res. 2006;135:370–379. doi:
129. Ji W, Foo JN, O’Roak BJ, Zhao H, Larson MG, Simon DB, Newton- 10.1016/j.jss.2006.04.001
Cheh C, State MW, Levy D, Lifton RP. Rare independent mutations 150. Carlstrom M, Wahlin N, Skott O, Persson AE. Relief of chronic partial
in renal salt handling genes contribute to blood pressure variation. Nat ureteral obstruction attenuates salt-sensitive hypertension in rats. Acta
Genet. 2008;40:592–599. doi: 10.1038/ng.118 Physiol (Oxf). 2007:189:67–75.
130. Mullins JJ, Peters J, Ganten D. Fulminant hypertension in transgenic 151. Carlström M, Wåhlin N, Sällström J, Skøtt O, Brown R, Persson AE.
rats harbouring the mouse Ren-2 gene. Nature. 1990;344:541–544. doi: Hydronephrosis causes salt-sensitive hypertension in rats. J Hypertens.
10.1038/344541a0 2006;24:1437–1443. doi: 10.1097/01.hjh.0000234126.78766.00
131. Liu X, Bellamy CO, Bailey MA, Mullins LJ, Dunbar DR, Kenyon CJ, 152. Hall JE. Pathophysiology of obesity hypertension. Curr Hypertens Rep.
Brooker G, Kantachuvesiri S, Maratou K, Ashek A, Clark AF, Fleming 2000;2:139–147.
S, Mullins JJ. Angiotensin-converting enzyme is a modifier of hyper- 153. Lerman LO, Chade AR, Sica V, Napoli C. Animal models of hyper-
tensive end organ damage. J Biol Chem. 2009;284:15564–15572. doi: tension: an overview. J Lab Clin Med. 2005;146:160–173. doi:
10.1074/jbc.M806584200 10.1016/j.lab.2005.05.005
132. Gembardt F, Heringer-Walther S, van Esch JH, Sterner-Kock A, van 154. Chade AR, Tullos N, Stewart NJ, Surles B. Endothelin-a recep-

Veghel R, Le TH, Garrelds IM, Coffman TM, Danser AH, Schultheiss tor antagonism after renal angioplasty enhances renal recovery in
HP, Walther T. Cardiovascular phenotype of mice lacking all three sub- renovascular disease. J Am Soc Nephrol. 2015;26:1071–1080. doi:
types of angiotensin II receptors. FASEB J. 2008;22:3068–3077. doi: 10.1681/ASN.2014040323
10.1096/fj.08-108316 155. Urbieta-Caceres VH, Zhu XY, Gibson ME, Favreau FD, Jordan K,

133. Aitman TJ, Critser JK, Cuppen E, Dominiczak A, Fernandez-Suarez Lerman A, Lerman LO. Reversal of experimental renovascular hyper-
Downloaded from http://ahajournals.org by on March 14, 2019

XM, Flint J, Gauguier D, Geurts AM, Gould M, Harris PC, Holmdahl tension restores coronary microvascular function and architecture. Am J
R, Hubner N, Izsvák Z, Jacob HJ, Kuramoto T, Kwitek AE, Marrone Hypertens. 2011;24:458–465. doi: 10.1038/ajh.2010.259
A, Mashimo T, Moreno C, Mullins J, Mullins L, Olsson T, Pravenec M, 156. Chade AR, Kelsen S. Renal microvascular disease determines the

Riley L, Saar K, Serikawa T, Shull JD, Szpirer C, Twigger SN, Voigt B, responses to revascularization in experimental renovascular dis-
Worley K. Progress and prospects in rat genetics: a community view. Nat ease. Circ Cardiovasc Interv. 2010;3:376–383. doi: 10.1161/
Genet. 2008;40:516–522. doi: 10.1038/ng.147 CIRCINTERVENTIONS.110.951277
134. Liu Z, Liu F. Cautious use of fli1a:EGFP transgenic zebrafish in vas- 157. Chelko SP, Schmiedt CW, Lewis TH, Lewis SJ, Robertson TP. A novel
cular research. Biochem Biophys Res Commun. 2012;427:223–226. doi: vascular clip design for the reliable induction of 2-kidney, 1-clip hyper-
10.1016/j.bbrc.2012.09.064 tension in the rat. J Appl Physiol. 2012;112:362–366.
135. Bianchi BR, Zhang XF, Reilly RM, Kym PR, Yao BB, Chen J. Species 158. Huang WC, Tsai RY, Fang TC. Nitric oxide modulates the development
comparison and pharmacological characterization of human, monkey, and surgical reversal of renovascular hypertension in rats. J Hypertens.
rat, and mouse TRPA1 channels. J Pharmacol Exp Ther. 2012;341:360– 2000;18:601–613.
368. doi: 10.1124/jpet.111.189902 159. Hollander W, Prusty S, Kemper T, Rosene DL, Moss MB. The effects
136. Conway BR, Rennie J, Bailey MA, Dunbar DR, Manning JR, Bellamy of hypertension on cerebral atherosclerosis in the cynomolgus monkey.
CO, Hughes J, Mullins JJ. Hyperglycemia and renin-dependent hyper- Stroke. 1993;24:1218–1226.
tension synergize to model diabetic nephropathy. J Am Soc Nephrol. 160. Xu CP, Glagov S, Zatina MA, Zarins CK. Hypertension sustains plaque
2012;23:405–411. progression despite reduction of hypercholesterolemia. Hypertension.
137. Kotb AM, Müller T, Xie J, Anand-Apte B, Endlich K, Endlich N.
1991;18:123–129.
Simultaneous assessment of glomerular filtration and barrier function in 161. Crowley SD, Gurley SB, Herrera MJ, Ruiz P, Griffiths R, Kumar AP,
live zebrafish. Am J Physiol Renal Physiol. 2014;307:F1427–F1434. doi: Kim HS, Smithies O, Le TH, Coffman TM. Angiotensin II causes hyper-
10.1152/ajprenal.00029.2014 tension and cardiac hypertrophy through its receptors in the kidney.
138. Huang J, McKee M, Huang HD, Xiang A, Davidson AJ, Lu HA. A Proc Natl Acad Sci U S A. 2006;103:17985–17990. doi: 10.1073/pnas.
zebrafish model of conditional targeted podocyte ablation and regenera- 0605545103
tion. Kidney Int. 2013;83:1193–1200. doi: 10.1038/ki.2013.6 162. Guzik TJ, Hoch NE, Brown KA, McCann LA, Rahman A, Dikalov S,
139. Schlager G. Spontaneous hypertension in laboratory animals: a review of Goronzy J, Weyand C, Harrison DG. Role of the T cell in the genesis
the genetic implications. J Hered. 1972;63:35–38. of angiotensin II induced hypertension and vascular dysfunction. J Exp
140. Alexander N, Hinshaw LB, Drury DR. Further observations on develop- Med. 2007;204:2449–2460. doi: 10.1084/jem.20070657
ment of a colony of spontaneously hypertensive rabbits. Proc Soc Exp 163. Crowley SD, Frey CW, Gould SK, Griffiths R, Ruiz P, Burchette JL,
Biol Med. 1956;92:249–253. Howell DN, Makhanova N, Yan M, Kim HS, Tharaux PL, Coffman TM.
141. Li D, Wang Q, Zhang Y, Li D, Yang D, Wei S, Su L, Ye T, Zheng X, Peng Stimulation of lymphocyte responses by angiotensin II promotes kidney
K, Zhang L, Zhang Y, Yang Y, Ma S. A novel swine model of spontane- injury in hypertension. Am J Physiol Renal Physiol. 2008;295:F515–
ous hypertension with sympathetic hyperactivity responds well to renal F524. doi: 10.1152/ajprenal.00527.2007
denervation. Am J Hypertens. 2016;29:63–72. doi: 10.1093/ajh/hpv066 164. Crowley SD, Zhang J, Herrera M, Griffiths R, Ruiz P, Coffman TM.
142. Frederick E. Tippett GA, Eyster G, Blanchard G, Bell T. Primary hyper- Role of AT₁ receptor-mediated salt retention in angiotensin II-dependent
tension in a colony of dogs. Hypertension. 1987;9:49–58. hypertension. Am J Physiol Renal Physiol. 2011;301:F1124–F1130. doi:
143. Littman MP, Robertson JL, Bovée KC. Spontaneous systemic hyper- 10.1152/ajprenal.00305.2011
tension in dogs: five cases (1981–1983). J Am Vet Med Assoc. 165. Polichnowski AJ, Griffin KA, Picken MM, Licea-Vargas H, Long J,
1988;193:486–494. Williamson GA, Bidani AK. Hemodynamic basis for the limited renal
e30  Hypertension   TBD 2019

injury in rats with angiotensin II-induced hypertension. Am J Physiol effects of deoxycorticosterone (DOCA)-salt in C57 mice. Hypertension.
Renal Physiol. 2015;308:F252–F260. 2011;57:600–607. doi: 10.1161/HYPERTENSIONAHA.110.165829
166. Olsen ME, Hall JE, Montani JP, Guyton AC, Langford HG, Cornell JE. 187. Danser AH. The role of the (pro)renin receptor in hypertensive disease.
Mechanisms of angiotensin II natriuresis and antinatriuresis. Am J Physiol. Am J Hypertens. 2015;28:1187–1196. doi: 10.1093/ajh/hpv045
1985;249(pt 2):F299–F307. doi: 10.1152/ajprenal.1985.249.2.F299 188. Selye H, Hall CE, Rowley EM. Malignant hypertension produced by
167. Cowley AW, Miller JP, Guyton AC. Open-loop analysis of the renin- treatment with desoxycorticosterone acetate and sodium chloride. Can
angiotensin system in the dog. Circ Res. 1971;28:568–581. Med Assoc J. 1943;49:88–92.
168. Hartner A, Cordasic N, Klanke B, Veelken R, Hilgers KF. Strain differ- 189. Deen WM, Maddox DA, Robertson CR, Brenner BM. Dynamics of glo-
ences in the development of hypertension and glomerular lesions induced merular ultrafiltration in the rat, VII: response to reduced renal mass. Am
by deoxycorticosterone acetate salt in mice. Nephrol Dial Transplant. J Physiol. 1974;227:556–562. doi: 10.1152/ajplegacy.1974.227.3.556
2003;18:1999–2004. 190. Zhang Y, Kompa AR. A practical guide to subtotal nephrectomy in the rat
169. Sparks MA, Stegbauer J, Chen D, Gomez JA, Griffiths RC, Azad
with subsequent methodology for assessing renal and cardiac function.
HA, Herrera M, Gurley SB, Coffman TM. Vascular type 1A angio- Nephrology (Carlton). 2014;19:552–561.
tensin II receptors control BP by regulating renal blood flow and uri- 191. Salzler HR, Griffiths R, Ruiz P, Chi L, Frey C, Marchuk DA, Rockman
nary sodium excretion. J Am Soc Nephrol. 2015;26:2953–2962. doi: HA, Le TH. Hypertension and albuminuria in chronic kidney disease
10.1681/ASN.2014080816 mapped to a mouse chromosome 11 locus. Kidney Int. 2007;72:1226–
170. Lautrette A, Li S, Alili R, Sunnarborg SW, Burtin M, Lee DC, Friedlander 1232. doi: 10.1038/sj.ki.5002519
G, Terzi F. Angiotensin II and EGF receptor cross-talk in chronic kidney 192. Kennedy DJ, Elkareh J, Shidyak A, Shapiro AP, Smaili S, Mutgi K,
diseases: a new therapeutic approach. Nat Med. 2005;11:867–874. doi: Gupta S, Tian J, Morgan E, Khouri S, Cooper CJ, Periyasamy SM, Xie Z,
10.1038/nm1275 Malhotra D, Fedorova OV, Bagrov AY, Shapiro JI. Partial nephrectomy
171. Kawada N, Imai E, Karber A, Welch WJ, Wilcox CS. A mouse model of as a model for uremic cardiomyopathy in the mouse. Am J Physiol Renal
angiotensin II slow pressor response: role of oxidative stress. J Am Soc Physiol. 2008;294:F450–F454. doi: 10.1152/ajprenal.00472.2007
Nephrol. 2002;13:2860–2868. 193. Kren S, Hostetter TH. The course of the remnant kidney model in mice.
172. Sopel MJ, Rosin NL, Lee TD, Légaré JF. Myocardial fibrosis in response Kidney Int. 1999;56:333–337. doi: 10.1046/j.1523-1755.1999.00527.x
to angiotensin II is preceded by the recruitment of mesenchymal pro- 194. Leelahavanichkul A, Yan Q, Hu X, Eisner C, Huang Y, Chen R,

genitor cells. Lab Invest. 2011;91:565–578. doi: 10.1038/labinvest. Mizel D, Zhou H, Wright EC, Kopp JB, Schnermann J, Yuen PS, Star
2010.190 RA. Angiotensin II overcomes strain-dependent resistance of rapid
173. Oliverio MI, Best CF, Kim HS, Arendshorst WJ, Smithies O, Coffman CKD progression in a new remnant kidney mouse model. Kidney Int.
TM. Angiotensin II responses in AT1A receptor-deficient mice: a role for 2010;78:1136–1153. doi: 10.1038/ki.2010.287
AT1B receptors in blood pressure regulation. Am J Physiol. 1997;272(pt 195. Gardiner SM, Kemp PA, Bennett T, Palmer RM, Moncada S. Nitric
2):F515–F520. doi: 10.1152/ajprenal.1997.272.4.F515 oxide synthase inhibitors cause sustained, but reversible, hypertension
174. Zhang J, Rudemiller NP, Patel MB, Karlovich NS, Wu M, McDonough and hindquarters vasoconstriction in Brattleboro rats. Eur J Pharmacol.
AA, Griffiths R, Sparks MA, Jeffs AD, Crowley SD. Interleukin-1 recep- 1992;213:449–451.
tor activation potentiates salt reabsorption in angiotensin II-induced 196. Ribeiro MO, Antunes E, de Nucci G, Lovisolo SM, Zatz R. Chronic inhi-
hypertension via the NKCC2 co-transporter in the nephron. Cell Metab. bition of nitric oxide synthesis: a new model of arterial hypertension.
2016;23:360–368. doi: 10.1016/j.cmet.2015.11.013 Hypertension. 1992;20:298–303.
175. Zhang JD, Patel MB, Song YS, Griffiths R, Burchette J, Ruiz P, Sparks 197. Qiu C, Engels K, Baylis C. Angiotensin II and alpha 1-adrenergic tone
Downloaded from http://ahajournals.org by on March 14, 2019

MA, Yan M, Howell DN, Gomez JA, Spurney RF, Coffman TM, Crowley in chronic nitric oxide blockade-induced hypertension. Am J Physiol.
SD. A novel role for type 1 angiotensin receptors on T lymphocytes to 1994;266(pt 2):R1470–R1476. doi: 10.1152/ajpregu.1994.266.5.R1470
limit target organ damage in hypertension. Circ Res. 2012;110:1604– 198. Jain M, Bhosale V, Tripathi D, Singh H, Pal N, Hanif K, Jagavelu K.
1617. doi: 10.1161/CIRCRESAHA.111.261768 Antihypertensive drugs aliskiren, nebivolol, and olmesartan reduce
176. Butz GM, Davisson RL. Long-term telemetric measurement of cardiovas- hypertension by reducing endothelial microparticles and regulat-
cular parameters in awake mice: a physiological genomics tool. Physiol ing angiogenesis. J Cardiovasc Pharmacol. 2017;70:176–183. doi:
Genomics. 2001;5:89–97. doi: 10.1152/physiolgenomics.2001.5.2.89 10.1097/FJC.0000000000000503
177. Aragao-Santiago L, Gomez-Sanchez CE, Mulatero P, Spyroglou A,
199. Zatz R, Baylis C. Chronic nitric oxide inhibition model six years on.
Reincke M, Williams TA. Mouse models of primary aldosteronism: from Hypertension. 1998;32:958–964.
physiology to pathophysiology. Endocrinology. 2017;158:4129–4138. 200. Mohan S, Reddick RL, Musi N, Horn DA, Yan B, Prihoda TJ, Natarajan
doi: 10.1210/en.2017-00637 M, Abboud-Werner SL. Diabetic eNOS knockout mice develop distinct
178. Gu H, Ma Z, Wang J, Zhu T, Du N, Shatara A, Yi X, Kowala MC, Du Y. macro- and microvascular complications. Lab Invest. 2008;88:515–528.
Salt-dependent blood pressure in human aldosterone synthase-transgenic doi: 10.1038/labinvest.2008.23
mice. Sci Rep. 2017;7:492. doi: 10.1038/s41598-017-00461-9 201. Zhao HJ, Wang S, Cheng H, Zhang MZ, Takahashi T, Fogo AB, Breyer
179. Selye H. Production of hypertension and hyalinosis by desoxocortisone. MD, Harris RC. Endothelial nitric oxide synthase deficiency pro-
Br Med J. 1950;1:203–206. duces accelerated nephropathy in diabetic mice. J Am Soc Nephrol.
180. Basting T, Lazartigues E. DOCA-salt hypertension: an update. Curr 2006;17:2664–2669. doi: 10.1681/ASN.2006070798
Hypertens Rep. 2017;19:32. doi: 10.1007/s11906-017-0731-4 202. Hedayat AF, Park KH, Kwon TG, Woollard JR, Jiang K, Carlson DF,
181. Haack D, Möhring J, Möhring B, Petri M, Hackenthal E. Comparative study Lerman A, Lerman LO. Peripheral vascular atherosclerosis in a novel
on development of corticosterone and DOCA hypertension in rats. Am J PCSK9 gain-of-function mutant Ossabaw miniature pig model. Transl
Physiol. 1977;233:F403–F411. doi: 10.1152/ajprenal.1977.233.5.F403 Res. 2018;192:30–45. doi: 10.1016/j.trsl.2017.10.007
182. Tajima Y, Ichikawa S, Sakamaki T, Matsuo H, Aizawa F, Kogure M, 203. Yu Q, Larson DF, Slayback D, Lundeen TF, Baxter JH, Watson RR.
Yagi S, Murata K. Body fluid distribution in the maintenance of DOCA- Characterization of high-salt and high-fat diets on cardiac and vascular
salt hypertension in rats. Am J Physiol. 1983;244:H695–H700. doi: function in mice. Cardiovasc Toxicol. 2004;4:37–46.
10.1152/ajpheart.1983.244.5.H695 204. Dobrian AD, Davies MJ, Prewitt RL, Lauterio TJ. Development of

183. Kurtz TW, Dominiczak AF, DiCarlo SE, Pravenec M, Morris RC
hypertension in a rat model of diet-induced obesity. Hypertension.
Jr. Molecular-based mechanisms of Mendelian forms of salt-depen- 2000;35:1009–1015.
dent hypertension: questioning the prevailing theory. Hypertension. 205. Hariri N, Thibault L. High-fat diet-induced obesity in animal models.
2015;65:932–941. doi: 10.1161/HYPERTENSIONAHA.114.05092 Nutr Res Rev. 2010;23:270–299. doi: 10.1017/S0954422410000168
184. O’Donaughy TL, Qi Y, Brooks VL. Central action of increased
206. Carroll JF, Dwyer TM, Grady AW, Reinhart GA, Montani JP, Cockrell
osmolality to support blood pressure in deoxycorticosterone acetate- K, Meydrech EF, Mizelle HL. Hypertension, cardiac hypertrophy,
salt rats. Hypertension. 2006;48:658–663. doi: 10.1161/01.HYP. and neurohumoral activity in a new animal model of obesity. Am J
0000238140.06251.7a Physiol. 1996;271(pt 2):H373–H378. doi: 10.1152/ajpheart.1996.
185. Fink GD, Pawloski CM, Blair ML, Mangiapane ML. The area postrema 271.1.H373
in deoxycorticosterone-salt hypertension in rats. Hypertension. 1987;9(pt 207. Pawar AS, Zhu XY, Eirin A, Tang H, Jordan KL, Woollard JR, Lerman
2):III206–III209. A, Lerman LO. Adipose tissue remodeling in a novel domestic porcine
186. Grobe JL, Buehrer BA, Hilzendeger AM, Liu X, Davis DR, Xu D, model of diet-induced obesity. Obesity (Silver Spring). 2015;23:399–
Sigmund CD. Angiotensinergic signaling in the brain mediates metabolic 407. doi: 10.1002/oby.20971
Lerman et al   Animal Models of Hypertension   e31

208. Wong SK, Chin KY, Suhaimi FH, Fairus A, Ima-Nirwana S. Animal mod- 229. Hinojosa-Laborde C, Lange DL, Haywood JR. Role of female sex

els of metabolic syndrome: a review. Nutr Metab (Lond). 2016;13:65. hormones in the development and reversal of dahl hypertension.
doi: 10.1186/s12986-016-0123-9 Hypertension. 2000;35(pt 2):484–489.
209. Thirunavukkarasu V, Anitha Nandhini AT, Anuradha CV. Lipoic acid 230. Yanes LL, Romero DG, Moulana M, Lima R, Davis DD, Zhang H,
attenuates hypertension and improves insulin sensitivity, kallikrein Lockhart R, Racusen LC, Reckelhoff JF. Cardiovascular-renal and meta-
activity and nitrite levels in high fructose-fed rats. J Comp Physiol B. bolic characterization of a rat model of polycystic ovary syndrome. Gend
2004;174:587–592. doi: 10.1007/s00360-004-0447-z Med. 2011;8:103–115. doi: 10.1016/j.genm.2010.11.013
210. Rocchini AP, Moorehead C, Wentz E, Deremer S. Obesity-induced
231. Reckelhoff JF, Zhang H, Srivastava K. Gender differences in develop-
hypertension in the dog. Hypertension. 1987;9:III64–III68. ment of hypertension in spontaneously hypertensive rats: role of the
211. Thigpen JE, Setchell KD, Saunders HE, Haseman JK, Grant MG,
renin-angiotensin system. Hypertension. 2000;35:480–483.
Forsythe DB. Selecting the appropriate rodent diet for endocrine disrup- 232. Foss JD, Fink GD, Osborn JW. Reversal of genetic salt-sensitive hyper-
tor research and testing studies. ILAR J. 2004;45:401–416. tension by targeted sympathetic ablation. Hypertension. 2013;61:806–
212. Chade AR, Mushin OP, Zhu X, Rodriguez-Porcel M, Grande JP, Textor 811. doi: 10.1161/HYPERTENSIONAHA.111.00474
SC, Lerman A, Lerman LO. Pathways of renal fibrosis and modulation 233. da Silva AA, do Carmo JM, Kanyicska B, Dubinion J, Brandon E,
of matrix turnover in experimental hypercholesterolemia. Hypertension. Hall JE. Endogenous melanocortin system activity contributes to
2005;46:772–779. doi: 10.1161/01.HYP.0000184250.37607.da the elevated arterial pressure in spontaneously hypertensive rats.
213. Oron-Herman M, Kamari Y, Grossman E, Yeger G, Peleg E, Shabtay Z, Hypertension. 2008;51:884–890. doi: 10.1161/HYPERTENSIONAHA.
Shamiss A, Sharabi Y. Metabolic syndrome: comparison of the two com- 107.100636
monly used animal models. Am J Hypertens. 2008;21:1018–1022. doi: 234. Maranon RO, Lima R, Mathbout M, do Carmo JM, Hall JE, Roman RJ,
10.1038/ajh.2008.218 Reckelhoff JF. Postmenopausal hypertension: role of the sympathetic
214. Tschöp M, Heiman ML. Rodent obesity models: an overview. Exp Clin nervous system in an animal model. Am J Physiol Regul Integr Comp
Endocrinol Diabetes. 2001;109:307–319. doi: 10.1055/s-2001-17297 Physiol. 2014;306:R248–R256. doi: 10.1152/ajpregu.00490.2013
215. Nagae A, Fujita M, Kawarazaki H, Matsui H, Ando K, Fujita T. Effect 235. Yanes LL, Romero DG, Iliescu R, Zhang H, Davis D, Reckelhoff JF.
of high fat loading in Dahl salt-sensitive rats. Clin Exp Hypertens. Postmenopausal hypertension: role of the renin-angiotensin system.
2009;31:451–461. Hypertension. 2010;56:359–363. doi: 10.1161/HYPERTENSIONAHA.
216. Zhang X, Li ZL, Woollard JR, Eirin A, Ebrahimi B, Crane JA, Zhu XY, 110.152975
Pawar AS, Krier JD, Jordan KL, Tang H, Textor SC, Lerman A, Lerman 236. Fortepiani LA, Reckelhoff JF. Role of oxidative stress in the sex differ-
LO. Obesity-metabolic derangement preserves hemodynamics but pro- ences in blood pressure in spontaneously hypertensive rats. J Hypertens.
motes intrarenal adiposity and macrophage infiltration in swine renovas- 2005;23:801–805.
cular disease. Am J Physiol Renal Physiol. 2013;305:F265–F276. 237. Sullivan JC, Sasser JM, Pollock JS. Sexual dimorphism in oxidant sta-
217. Li ZL, Ebrahimi B, Zhang X, Eirin A, Woollard JR, Tang H, Lerman A, tus in spontaneously hypertensive rats. Am J Physiol Regul Integr Comp
Wang SM, Lerman LO. Obesity-metabolic derangement exacerbates car- Physiol. 2007;292:R764–R768.
diomyocyte loss distal to moderate coronary artery stenosis in pigs with- 238. Sullivan JC, Pardieck JL, Hyndman KA, Pollock JS. Renal NOS activity,
out affecting global cardiac function. Am J Physiol Heart Circ Physiol. expression, and localization in male and female spontaneously hyperten-
2014;306:H1087–H1101. doi: 10.1152/ajpheart.00052.2013 sive rats. Am J Physiol Regul Integr Comp Physiol. 2010;298:R61–R69.
218. Ernsberger P, Ishizuka T, Liu S, Farrell CJ, Bedol D, Koletsky RJ, doi: 10.1152/ajpregu.00526.2009
Friedman JE. Mechanisms of antihyperglycemic effects of moxoni- 239. Fortepiani LA, Reckelhoff JF. Treatment with tetrahydrobiopterin

Downloaded from http://ahajournals.org by on March 14, 2019

dine in the obese spontaneously hypertensive Koletsky rat (SHROB). J reduces blood pressure in male SHR by reducing testosterone synthesis.
Pharmacol Exp Ther. 1999;288:139–147. Am J Physiol Regul Integr Comp Physiol. 2005;288:R733–R736.
219. Morrison RG, Carpenter AB, Moore SK, Mangiarua EI, Valentovic MA, 240. Su P, Kaushal KM, Kroetz DL. Inhibition of renal arachidonic acid
Walker EM Jr, Wehner PS, Rhoten WB, Touchon RC, McCumbee WD. omega-hydroxylase activity with ABT reduces blood pressure in the
Increased sensitivity of the obese Zucker rat to deoxycorticosterone-salt- SHR. Am J Physiol. 1998;275(pt 2):R426–R438.
induced hypertension. J Hypertens. 2002;20:2247–2255. 241. Tipton AJ, Baban B, Sullivan JC. Female spontaneously hypertensive
220. Sandberg K, Ji H. Sex differences in primary hypertension. Biol Sex rats have a compensatory increase in renal regulatory T cells in response
Differ. 2012;3:7. doi: 10.1186/2042-6410-3-7 to elevations in blood pressure. Hypertension. 2014;64:557–564. doi:
221. Evans RG, Stevenson KM, Bergström G, Denton KM, Madden AC, 10.1161/HYPERTENSIONAHA.114.03512
Gribben RL, Weekes SR, Anderson WP. Sex differences in pressure 242. Yanes LL, Romero DG, Cucchiarelli VE, Fortepiani LA, Gomez-

diuresis/natriuresis in rabbits. Acta Physiol Scand. 2000;169:309–316. Sanchez CE, Santacruz F, Reckelhoff JF. Role of endothelin in mediat-
doi: 10.1046/j.1365-201x.2000.00749.x ing postmenopausal hypertension in a rat model. Am J Physiol Regul
222. Sartori-Valinotti JC, Iliescu R, Yanes LL, Dorsett-Martin W, Reckelhoff Integr Comp Physiol. 2005;288:R229–R233. doi: 10.1152/ajpregu.
JF. Sex differences in the pressor response to angiotensin II when 00697.2003
the endogenous renin-angiotensin system is blocked. Hypertension. 243. Gupte M, Thatcher SE, Boustany-Kari CM, Shoemaker R, Yiannikouris
2008;51:1170–1176. doi: 10.1161/HYPERTENSIONAHA.107.106922 F, Zhang X, Karounos M, Cassis LA. Angiotensin converting enzyme 2
223. Veiras LC, Girardi ACC, Curry J, Pei L, Ralph DL, Tran A, Castelo- contributes to sex differences in the development of obesity hypertension
Branco RC, Pastor-Soler N, Arranz CT, Yu ASL, McDonough AA. Sexual in C57BL/6 mice. Arterioscler Thromb Vasc Biol. 2012;32:1392–1399.
dimorphic pattern of renal transporters and electrolyte homeostasis. J Am doi: 10.1161/ATVBAHA.112.248559
Soc Nephrol. 2017;28:3504–3517. doi: 10.1681/ASN.2017030295 244. Riazi S, Madala-Halagappa VK, Dantas AP, Hu X, Ecelbarger CA. Sex
224. Cohen JA, Lindsey SH, Pirro NT, Brosnihan KB, Gallagher PE, Chappell differences in renal nitric oxide synthase, NAD(P)H oxidase, and blood
MC. Influence of estrogen depletion and salt loading on renal angioten- pressure in obese Zucker rats. Gend Med. 2007;4:214–229.
sinogen expression in the mRen(2).Lewis strain. Am J Physiol Renal 245. Doursout MF, Chelly JE, Wouters P, Lawrence C, Liang YY, Buckley JP.
Physiol. 2010;299:F35–F42. doi: 10.1152/ajprenal.00138.2010 Effect of gender in centrally induced angiotensin II hypertension in dogs.
225. Brinson KN, Rafikova O, Sullivan JC. Female sex hormones protect Hypertension. 1990;15(suppl I):I117–I120.
against salt-sensitive hypertension but not essential hypertension. Am J 246. Shi X, Bai Y, Ke Y, Chen R, Lin X, Chen L, Hong H. Ageing-related aorta
Physiol Regul Integr Comp Physiol. 2014;307:R149–R157. remodelling and calcification occur earlier and progress more severely in
226. Xue B, Pamidimukkala J, Hay M. Sex differences in the develop-
rats with spontaneous hypertension. Histol Histopathol. 2018;33:727–
ment of angiotensin II-induced hypertension in conscious mice. 736. doi: 10.14670/HH-11-971
Am J Physiol Heart Circ Physiol. 2005;288:H2177–H2184. doi: 247. Park JB, Schiffrin EL. Small artery remodeling is the most prevalent
10.1152/ajpheart.00969.2004 (earliest?) form of target organ damage in mild essential hypertension. J
227. Zimmerman MA, Baban B, Tipton AJ, O’Connor PM, Sullivan JC. Hypertens. 2001;19:921–930.
Chronic ANG II infusion induces sex-specific increases in renal T cells 248. Baumbach GL, Heistad DD. Remodeling of cerebral arterioles in chronic
in Sprague-Dawley rats. Am J Physiol Renal Physiol. 2015;308:F706– hypertension. Hypertension. 1989;13(pt 2):968–972.
F712. doi: 10.1152/ajprenal.00446.2014 249. Boudier HA. Arteriolar and capillary remodelling in hypertension.

228. Fortepiani LA, Zhang H, Racusen L, Roberts LJ 2nd, Reckelhoff JF. Drugs. 1999;58(spec No 1):37–40.
Characterization of an animal model of postmenopausal hypertension in 250. Mulvany MJ, Baumbach GL, Aalkjaer C, Heagerty AM, Korsgaard

spontaneously hypertensive rats. Hypertension. 2003;41(pt 2):640–645. N, Schiffrin EL, Heistad DD. Vascular remodeling. Hypertension.
doi: 10.1161/01.HYP.0000046924.94886.EF 1996;28:505–506.
e32  Hypertension   TBD 2019

251.
Schiffrin EL. Vascular remodeling in hypertension: mecha- similar to old spontaneously hypertensive rats. J Vasc Res. 2012;49:309–
nisms and treatment. Hypertension. 2012;59:367–374. doi: 318. doi: 10.1159/000337470
10.1161/HYPERTENSIONAHA.111.187021 270. Arnet UA, Novosel D, Barton M, Noll G, Ganten D, Lüscher TF.

252. Sharifi AM, Schiffrin EL. Apoptosis in vasculature of spontaneously Endothelial dysfunction in the aorta of transgenic rats harboring the
hypertensive rats: effect of an angiotensin converting enzyme inhibitor mouse Ren-2 gene. Endothelium. 1999;6:175–184.
and a calcium channel antagonist. Am J Hypertens. 1998;11:1108–1116. 271. Tochitani T, Mori M, Matsuda K, Kouchi M, Fujii Y, Matsumoto I.
253. Geng J, Zhao Z, Yang L, Zhang M, Liu X. Protein kinase D was involved Histopathological characteristics of renal changes in human renin-angio-
in vascular remodeling in spontaneously hypertensive rats. Clin Exp tensinogen double transgenic rats. J Toxicol Pathol. 2016;29:125–129.
Hypertens. 2018:1–8. doi: 10.1080/10641963.2018.1469647 doi: 10.1293/tox.2015-0055
254. Rizzoni D, Muiesan ML, Porteri E, De Ciuceis C, Boari GE, Salvetti 272. Montezano AC, Nguyen Dinh Cat A, Rios FJ, Touyz RM. Angiotensin
M, Paini A, Rosei EA. Vascular remodeling, macro- and microves- II and vascular injury. Curr Hypertens Rep. 2014;16:431. doi:
sels: therapeutic implications. Blood Press. 2009;18:242–246. doi: 10.1007/s11906-014-0431-2
10.3109/08037050903254923 273. Antunes TT, Callera GE, He Y, Yogi A, Ryazanov AG, Ryazanova

255. Jiang J, Zheng JP, Li Y, Gan Z, Jiang Y, Huang D, Li H, Liu Z, Ke Y. LV, Zhai A, Stewart DJ, Shrier A, Touyz RM. Transient receptor
Differential contribution of endothelium-derived relaxing factors to potential melastatin 7 cation channel kinase: new player in angioten-
vascular reactivity in conduit and resistance arteries from normoten- sin II-induced hypertension. Hypertension. 2016;67:763–773. doi:
sive and hypertensive rats. Clin Exp Hypertens. 2016;38:393–398. doi: 10.1161/HYPERTENSIONAHA.115.07021
10.3109/10641963.2016.1148155 274. Schiffrin EL. Mechanisms of remodelling of small arteries, antihyper-
256. Deng LY, Schiffrin EL. Effects of endothelin-1 and vasopressin on
tensive therapy and the immune system in hypertension. Clin Invest Med.
resistance arteries of spontaneously hypertensive rats. Am J Hypertens. 2015;38:E394–E402.
1992;5:817–822. 275. Zhang L, Wu JH, Huang TQ, Nepliouev I, Brian L, Zhang Z, Wertman
257. Harvey AP, Montezano AC, Hood KY, Lopes RA, Rios F, Ceravolo V, Rudemiller NP, McMahon TJ, Shenoy SK, Miller FJ, Crowley SD,
G, Graham D, Touyz RM. Vascular dysfunction and fibrosis in Freedman NJ, Stiber JA. Drebrin regulates angiotensin II-induced
stroke-prone spontaneously hypertensive rats: the aldosterone-min- aortic remodelling. Cardiovasc Res. 2018;114:1806–1815. doi:
eralocorticoid receptor-Nox1 axis. Life Sci. 2017;179:110–119. doi: 10.1093/cvr/cvy151
10.1016/j.lfs.2017.05.002 276. Avendaño MS, Martínez-Revelles S, Aguado A, Simões MR,

258. Pires PW, Jackson WF, Dorrance AM. Regulation of myogenic tone González-Amor M, Palacios R, Guillem-Llobat P, Vassallo DV, Vila
and structure of parenchymal arterioles by hypertension and the min- L, García-Puig J, Beltrán LM, Alonso MJ, Cachofeiro MV, Salaices M,
eralocorticoid receptor. Am J Physiol Heart Circ Physiol. 2015;309: Briones AM. Role of COX-2-derived PGE2 on vascular stiffness and
H127–H136. function in hypertension. Br J Pharmacol. 2016;173:1541–1555. doi:
259. Ledingham JM, Laverty R. Basilar artery remodelling in the geneti- 10.1111/bph.13457
cally hypertensive rat: effects of nitric oxide synthase inhibition and 277. Avendaño MS, García-Redondo AB, Zalba G, González-Amor M,

treatment with valsartan and enalapril. Clin Exp Pharmacol Physiol. Aguado A, Martínez-Revelles S, Beltrán LM, Camacho M, Cachofeiro
2000;27:642–646. V, Alonso MJ, Salaices M, Briones AM. mPGES-1 (microsomal pros-
260. Rizzoni D, Porteri E, Castellano M, Bettoni G, Muiesan ML, Muiesan taglandin E synthase-1) mediates vascular dysfunction in hyperten-
P, Giulini SM, Agabiti-Rosei E. Vascular hypertrophy and remodeling in sion through oxidative stress. Hypertension. 2018;72:492–502. doi:
secondary hypertension. Hypertension. 1996;28:785–790. 10.1161/HYPERTENSIONAHA.118.10833
Downloaded from http://ahajournals.org by on March 14, 2019

261. d’Uscio LV, Barton M, Shaw S, Moreau P, Lüscher TF. Structure


278. Leloup AJA, De Moudt S, Van Hove CE, Dugaucquier L, Vermeulen Z,
and function of small arteries in salt-induced hypertension: effects Segers VFM, De Keulenaer GW, Fransen P. Short-term angiotensin II
of chronic endothelin-subtype-A-receptor blockade. Hypertension. treatment affects large artery biomechanics and function in the absence
1997;30:905–911. of small artery alterations in mice. Front Physiol. 2018;9:582. doi:
262. Fan F, Geurts AM, Murphy SR, Pabbidi MR, Jacob HJ, Roman RJ. 10.3389/fphys.2018.00582
Impaired myogenic response and autoregulation of cerebral blood 279. Brouwers-Ceiler DL, Nelissen-Vrancken HJ, Smits JF, De Mey JG. The
flow is rescued in CYP4A1 transgenic Dahl salt-sensitive rat. Am influence of angiotensin II-induced increase in aortic wall mass on com-
J Physiol Regul Integr Comp Physiol. 2015;308:R379–R390. doi: pliance in rats in vivo. Cardiovasc Res. 1997;33:478–484.
10.1152/ajpregu.00256.2014 280. Zhou X, Wang J, Fa Y, Ye H. Signature microRNA expression profile is
263. Sharifi AM, Schiffrin EL. Apoptosis in aorta of deoxycorticosterone associated with spontaneous hypertension in African green monkey. Clin
acetate-salt hypertensive rats: effect of endothelin receptor antagonism. J Exp Hypertens. 2018:1–5. doi: 10.1080/10641963.2018.1469646
Hypertens. 1997;15(pt 1):1441–1448. 281. Zhang X, Li ZL, Eirin A, Ebrahimi B, Pawar AS, Zhu XY,

264. Li JS, Knafo L, Turgeon A, Garcia R, Schiffrin EL. Effect of endothe- Lerman A, Lerman LO. Cardiac metabolic alterations in hyper-
lin antagonism on blood pressure and vascular structure in renovascu- tensive obese pigs. Hypertension. 2015; 66:430–436. doi:
lar hypertensive rats. Am J Physiol. 1996;271(pt 2):H88–H93. doi: 10.1161/HYPERTENSIONAHA.115.05478
10.1152/ajpheart.1996.271.1.H88 282. Yongming P, Zhaowei C, Yichao M, Keyan Z, Liang C, Fangming C,
265. Paulis L, Becker ST, Lucht K, Schwengel K, Slavic S, Kaschina
Xiaoping X, Quanxin M, Minli C. Involvement of peroxisome prolif-
E, Thöne-Reineke C, Dahlöf B, Baulmann J, Unger T, Steckelings erator-activated receptors in cardiac and vascular remodeling in a novel
UM. Direct angiotensin II type 2 receptor stimulation in Nω-nitro-L- minipig model of insulin resistance and atherosclerosis induced by con-
arginine-methyl ester-induced hypertension: the effect on pulse wave sumption of a high-fat/cholesterol diet. Cardiovasc Diabetol. 2015;14:6.
velocity and aortic remodeling. Hypertension. 2012;59:485–492. doi: doi: 10.1186/s12933-014-0165-0
10.1161/HYPERTENSIONAHA.111.185496 283. del Campo L, Ferrer M. Wire myography to study vascular tone and
266. Ferreira-Melo SE, Yugar-Toledo JC, Coelho OR, De Luca IM, Tanus- vascular structure of isolated mouse arteries. Methods Mol Biol.
Santos JE, Hyslop S, Irigoyen MC, Moreno H Jr. Sildenafil reduces car- 2015;1339:255–276. doi: 10.1007/978-1-4939-2929-0_18
diovascular remodeling associated with hypertensive cardiomyopathy in 284. Schjørring OL, Carlsson R, Simonsen U. Pressure myography to study
NOS inhibitor-treated rats. Eur J Pharmacol. 2006;542:141–147. doi: the function and structure of isolated small arteries. Methods Mol Biol.
10.1016/j.ejphar.2006.04.039 2015;1339:277–295. doi: 10.1007/978-1-4939-2929-0_19
267. Hsieh NK, Wang JY, Liu JC, Lee WH, Chen HI. Structural changes 285. Zhang Y, Carreras D, de Bold AJ. Discoordinate re-expression of cardiac
in cerebral arteries following nitric oxide deprivation: a compari- fetal genes in N(omega)-nitro-L-arginine methyl ester (L-NAME) hyper-
son between normotensive and hypertensive rats. Thromb Haemost. tension. Cardiovasc Res. 2003;57:158–167.
2004;92:162–170. doi: 10.1160/TH03-10-0610 286. Kvakan H, Kleinewietfeld M, Qadri F, Park JK, Fischer R, Schwarz
268. Pistea A, Bakker EN, Spaan JA, Hardeman MR, van Rooijen N,
I, Rahn HP, Plehm R, Wellner M, Elitok S, Gratze P, Dechend R,
VanBavel E. Small artery remodeling and erythrocyte deformability in Luft FC, Muller DN. Regulatory T cells ameliorate angiotensin
L-NAME-induced hypertension: role of transglutaminases. J Vasc Res. II-induced cardiac damage. Circulation. 2009;119:2904–2912. doi:
2008;45:10–18. doi: 10.1159/000109073 10.1161/CIRCULATIONAHA.108.832782
269. Isabelle M, Simonet S, Ragonnet C, Sansilvestri-Morel P, Clavreul N, 287. Griffin KA. Hypertensive kidney injury and the progression of

Vayssettes-Courchay C, Verbeuren TJ. Chronic reduction of nitric oxide chronic kidney disease. Hypertension. 2017;70:687–694. doi:
level in adult spontaneously hypertensive rats induces aortic stiffness 10.1161/HYPERTENSIONAHA.117.08314
Lerman et al   Animal Models of Hypertension   e33

288. Griffin KA, Churchill PC, Picken M, Webb RC, Kurtz TW, Bidani AK. 308. Zhang X, Yeung PK, McAlonan GM, Chung SS, Chung SK. Transgenic
Differential salt-sensitivity in the pathogenesis of renal damage in SHR mice over-expressing endothelial endothelin-1 show cognitive defi-
and stroke prone SHR. Am J Hypertens. 2001;14(pt 1):311–320. cit with blood-brain barrier breakdown after transient ischemia with
289. Griffin KA, Polichnowski A, Litbarg N, Picken M, Venkatachalam MA, long-term reperfusion. Neurobiol Learn Mem. 2013;101:46–54. doi:
Bidani AK. Critical blood pressure threshold dependence of hypertensive 10.1016/j.nlm.2013.01.002
injury and repair in a malignant nephrosclerosis model. Hypertension. 309. Baumbach GL, Sigmund CD, Faraci FM. Structure of cerebral

2014;64:801–807. doi: 10.1161/HYPERTENSIONAHA.114.03609 arterioles in mice deficient in expression of the gene for endo-
290. Griffin KA, Picken M, Bidani AK. Method of renal mass reduction is a thelial nitric oxide synthase. Circ Res. 2004;95:822–829. doi:
critical modulator of subsequent hypertension and glomerular injury. J 10.1161/01.RES.0000146279.11923.14
Am Soc Nephrol. 1994;4:2023–2031. 310. Faraci FM, Sigmund CD, Shesely EG, Maeda N, Heistad DD. Responses
291. Gangadhariah MH, Luther JM, Garcia V, Paueksakon P, Zhang MZ, of carotid artery in mice deficient in expression of the gene for endothe-
Hayward SW, Love HD, Falck JR, Manthati VL, Imig JD, Schwartzman lial NO synthase. Am J Physiol. 1998;274(pt 2):H564–H570.
ML, Zent R, Capdevila JH, Pozzi A. Hypertension is a major con- 311. Girouard H, Park L, Anrather J, Zhou P, Iadecola C. Cerebrovascular
tributor to 20-hydroxyeicosatetraenoic acid-mediated kidney injury nitrosative stress mediates neurovascular and endothelial dysfunction
in diabetic nephropathy. J Am Soc Nephrol. 2015;26:597–610. doi: induced by angiotensin II. Arterioscler Thromb Vasc Biol. 2007;27:303–
10.1681/ASN.2013090980 309. doi: 10.1161/01.ATV.0000253885.41509.25
292. Iadecola C. The pathobiology of vascular dementia. Neuron.
312. Huang Z, Huang PL, Ma J, Meng W, Ayata C, Fishman MC, Moskowitz
2013;80:844–866. doi: 10.1016/j.neuron.2013.10.008 MA. Enlarged infarcts in endothelial nitric oxide synthase knockout
293. Iadecola C. The neurovascular unit coming of age: a journey through mice are attenuated by nitro-L-arginine. J Cereb Blood Flow Metab.
neurovascular coupling in health and disease. Neuron. 2017;96:17–42. 1996;16:981–987. doi: 10.1097/00004647-199609000-00023
doi: 10.1016/j.neuron.2017.07.030 313. Capone C, Faraco G, Coleman C, Young CN, Pickel VM, Anrather J,
294. Toth P, Tucsek Z, Sosnowska D, Gautam T, Mitschelen M, Tarantini Davisson RL, Iadecola C. Endothelin 1-dependent neurovascular dys-
S, Deak F, Koller A, Sonntag WE, Csiszar A, Ungvari Z. Age-related function in chronic intermittent hypoxia. Hypertension. 2012;60:106–
autoregulatory dysfunction and cerebromicrovascular injury in mice 113. doi: 10.1161/HYPERTENSIONAHA.112.193672
with angiotensin II-induced hypertension. J Cereb Blood Flow Metab. 314. Jackman KA, Zhou P, Faraco G, Peixoto PM, Coleman C, Voss HU,
2013;33:1732–1742. doi: 10.1038/jcbfm.2013.143 Pickel V, Manfredi G, Iadecola C. Dichotomous effects of chronic inter-
295. Faraco G, Sugiyama Y, Lane D, Garcia-Bonilla L, Chang H, Santisteban mittent hypoxia on focal cerebral ischemic injury. Stroke. 2014;45:1460–
MM, Racchumi G, Murphy M, Van Rooijen N, Anrather J, Iadecola C. 1467. doi: 10.1161/STROKEAHA.114.004816
Perivascular macrophages mediate the neurovascular and cognitive dys- 315. Durgan DJ, Crossland RF, Lloyd EE, Phillips SC, Bryan RM. Increased
function associated with hypertension. J Clin Invest. 2016;126:4674– cerebrovascular sensitivity to endothelin-1 in a rat model of obstructive
4689. doi: 10.1172/JCI86950 sleep apnea: a role for endothelin receptor B. J Cereb Blood Flow Metab.
296. Capone C, Faraco G, Peterson JR, Coleman C, Anrather J, Milner TA, 2015;35:402–411. doi: 10.1038/jcbfm.2014.214
Pickel VM, Davisson RL, Iadecola C. Central cardiovascular circuits con- 316. Crossland RF, Durgan DJ, Lloyd EE, Phillips SC, Reddy AK, Marrelli
tribute to the neurovascular dysfunction in angiotensin II hypertension. J SP, Bryan RM Jr. A new rodent model for obstructive sleep apnea: effects
Neurosci. 2012;32:4878–4886. doi: 10.1523/JNEUROSCI.6262-11.2012 on ATP-mediated dilations in cerebral arteries. Am J Physiol Regul Integr
297. Duchemin S, Belanger E, Wu R, Ferland G, Girouard H. Chronic perfu- Comp Physiol. 2013;305:R334–R342. doi: 10.1152/ajpregu.00244.2013
sion of angiotensin II causes cognitive dysfunctions and anxiety in mice. 317. Phillips SA, Olson EB, Morgan BJ, Lombard JH. Chronic intermittent
Downloaded from http://ahajournals.org by on March 14, 2019

Physiol Behav. 2013;109:63–68. doi: 10.1016/j.physbeh.2012.10.005 hypoxia impairs endothelium-dependent dilation in rat cerebral and
298. Faraco G, Park L, Zhou P, Luo W, Paul SM, Anrather J, Iadecola C. skeletal muscle resistance arteries. Am J Physiol Heart Circ Physiol.
Hypertension enhances Aβ-induced neurovascular dysfunction, promotes 2004;286:H388–H393. doi: 10.1152/ajpheart.00683.2003
β-secretase activity, and leads to amyloidogenic processing of APP. J 318. Hartman RE, Kamper JE, Goyal R, Stewart JM, Longo LD. Motor and
Cereb Blood Flow Metab. 2016;36:241–252. doi: 10.1038/jcbfm.2015.79 cognitive deficits in mice bred to have low or high blood pressure. Physiol
299. Wakisaka Y, Chu Y, Miller JD, Rosenberg GA, Heistad DD. Spontaneous Behav. 2012;105:1092–1097. doi: 10.1016/j.physbeh.2011.11.022
intracerebral hemorrhage during acute and chronic hypertension in mice. 319. Takahashi S, Hamai Y, Okamoto K. Fluorescein fundus angiography of
J Cereb Blood Flow Metab. 2010;30:56–69. doi: 10.1038/jcbfm.2009.183 stroke-prone SHR. Jpn Heart J. 1978;19:646–647.
300. Meissner A, Minnerup J, Soria G, Planas AM. Structural and functional 320. Takahashi S, Hamai Y, Okamoto K. On the findings of stroke-prone SHR.
brain alterations in a murine model of angiotensin II-induced hyperten- Jpn Heart J. 1978;19:645.
sion. J Neurochem. 2017;140:509–521. doi: 10.1111/jnc.13905 321. Tamaki K, Sadoshima S, Baumbach GL, Iadecola C, Reis DJ, Heistad
301. Dunn WR, Gardiner SM. Differential alteration in vascular structure DD. Evidence that disruption of the blood-brain barrier precedes reduc-
of resistance arteries isolated from the cerebral and mesenteric vascu- tion in cerebral blood flow in hypertensive encephalopathy. Hypertension.
lar beds of transgenic [(mRen-2)27], hypertensive rats. Hypertension. 1984;6(pt 2):I75–I81.
1997;29:1140–1147. 322. Spratt NJ, Fernandez J, Chen M, Rewell S, Cox S, van Raay L, Hogan
302. Baumbach GL, Sigmund CD, Faraci FM. Cerebral arteriolar structure in L, Howells DW. Modification of the method of thread manufacture
mice overexpressing human renin and angiotensinogen. Hypertension. improves stroke induction rate and reduces mortality after thread-
2003;41:50–55. occlusion of the middle cerebral artery in young or aged rats. J Neurosci
303. Inaba S, Iwai M, Tomono Y, Senba I, Furuno M, Kanno H, Okayama H, Methods. 2006;155:285–290. doi: 10.1016/j.jneumeth.2006.01.020
Mogi M, Higaki J, Horiuchi M. Exaggeration of focal cerebral ischemia in 323. Barone FC, Price WJ, White RF, Willette RN, Feuerstein GZ. Genetic
transgenic mice carrying human renin and human angiotensinogen genes. hypertension and increased susceptibility to cerebral ischemia. Neurosci
Stroke. 2009;40:597–603. doi: 10.1161/STROKEAHA.108.519801 Biobehav Rev. 1992;16:219–233.
304. Inaba S, Iwai M, Furuno M, Tomono Y, Kanno H, Senba I, Okayama 324. Jalal FY, Yang Y, Thompson J, Lopez AC, Rosenberg GA. Myelin

H, Mogi M, Higaki J, Horiuchi M. Continuous activation of renin- loss associated with neuroinflammation in hypertensive rats. Stroke.
angiotensin system impairs cognitive function in renin/angioten- 2012;43:1115–1122. doi: 10.1161/STROKEAHA.111.643080
sinogen transgenic mice. Hypertension. 2009;53:356–362. doi: 325. Kalani A, Pushpakumar SB, Vacek JC, Tyagi SC, Tyagi N. Inhibition
10.1161/HYPERTENSIONAHA.108.123612 of MMP-9 attenuates hypertensive cerebrovascular dysfunction in
305. Faraci FM, Lamping KG, Modrick ML, Ryan MJ, Sigmund CD, Didion Dahl salt-sensitive rats. Mol Cell Biochem. 2016;413:25–35. doi:
SP. Cerebral vascular effects of angiotensin II: new insights from 10.1007/s11010-015-2623-8
genetic models. J Cereb Blood Flow Metab. 2006;26:449–455. doi: 326. Smeda JS, Payne GW. Alterations in autoregulatory and myogenic

10.1038/sj.jcbfm.9600204 function in the cerebrovasculature of Dahl salt-sensitive rats. Stroke.
306. Iida S, Baumbach GL, Lavoie JL, Faraci FM, Sigmund CD, Heistad DD. 2003;34:1484–1490.
Spontaneous stroke in a genetic model of hypertension in mice. Stroke. 327. Rodrigues SF, Granger DN. Cerebral microvascular inflammation in
2005;36:1253–1258. doi: 10.1161/01.str.0000167694.58419.a2 DOCA salt-induced hypertension: role of angiotensin II and mitochon-
307. Leung JW, Ho MC, Lo AC, Chung SS, Chung SK. Endothelial cell-spe- drial superoxide. J Cereb Blood Flow Metab. 2012;32:368–375. doi:
cific over-expression of endothelin-1 leads to more severe cerebral dam- 10.1038/jcbfm.2011.139
age following transient middle cerebral artery occlusion. J Cardiovasc 328. Tada Y, Wada K, Shimada K, Makino H, Liang EI, Murakami S, Kudo
Pharmacol. 2004;44(suppl 1):S293–S300. M, Kitazato KT, Nagahiro S, Hashimoto T. Roles of hypertension in
e34  Hypertension   TBD 2019

the rupture of intracranial aneurysms. Stroke. 2014;45:579–586. doi: 344. Hainsworth AH, Allan SM, Boltze J, Cunningham C, Farris C, Head
10.1161/STROKEAHA.113.003072 E, Ihara M, Isaacs JD, Kalaria RN, Lesnik Oberstein SA, Moss
329. Jabaris SS, Sumathy H, Girish R, Narayanan S, Sugumar M, Saravana MB, Nitzsche B, Rosenberg GA, Rutten JW, Salkovic-Petrisic M,
Babu C, Thanikachalam S, Thanikachalam M. Phosphodiesterase-4 Troen AM. Translational models for vascular cognitive impair-
inhibitors ameliorates cognitive deficits in deoxycorticosterone acetate ment: a review including larger species. BMC Med. 2017;15:16. doi:
induced hypertensive rats via cAMP/CREB signaling system. Brain Res. 10.1186/s12916-017-0793-9
2015;1622:279–291. doi: 10.1016/j.brainres.2015.07.003 345. Friso S, Pizzolo F, Choi SW, Guarini P, Castagna A, Ravagnani V,
330. Dorrance AM, Rupp NC, Nogueira EF. Mineralocorticoid receptor
Carletto A, Pattini P, Corrocher R, Olivieri O. Epigenetic control of
activation causes cerebral vessel remodeling and exacerbates the dam- 11 beta-hydroxysteroid dehydrogenase 2 gene promoter is related
age caused by cerebral ischemia. Hypertension. 2006;47:590–595. doi: to human hypertension. Atherosclerosis. 2008;199:323–327. doi:
10.1161/01.HYP.0000196945.73586.0d 10.1016/j.atherosclerosis.2007.11.029
331. Nuki Y, Tsou TL, Kurihara C, Kanematsu M, Kanematsu Y, Hashimoto 346. Lee HA, Baek I, Seok YM, Yang E, Cho HM, Lee DY, Hong SH, Kim
T. Elastase-induced intracranial aneurysms in hypertensive mice. IK. Promoter hypomethylation upregulates Na+-K+-2Cl- cotransporter
Hypertension. 2009;54:1337–1344. doi: 10.1161/HYPERTENSIONAHA. 1 in spontaneously hypertensive rats. Biochem Biophys Res Commun.
109.138297 2010;396:252–257. doi: 10.1016/j.bbrc.2010.04.074
332. Hosaka K, Downes DP, Nowicki KW, Hoh BL. Modified murine
347. Cheng X, Joe B. Circular RNAs in rat models of cardiovascu-

intracranial aneurysm model: aneurysm formation and rupture by lar and renal diseases. Physiol Genomics. 2017;49:484–490. doi:
elastase and hypertension. J Neurointerv Surg. 2014;6:474–479. doi: 10.1152/physiolgenomics.00064.2017
10.1136/neurintsurg-2013-010788 348. Gopalakrishnan K, Kumarasamy S, Mell B, Joe B. Genome-wide

333. Zeng J, Zhang Y, Mo J, Su Z, Huang R. Two-kidney, two clip reno- identification of long noncoding RNAs in rat models of cardio-
vascular hypertensive rats can be used as stroke-prone rats. Stroke. vascular and renal disease. Hypertension. 2015;65:200–210. doi:
1998;29:1708–1713. 10.1161/HYPERTENSIONAHA.114.04498
334. Ménard B, Chazalviel L, Roussel S, Bernaudin M, Touzani O. Two- 349. Shi L, Liao J, Liu B, Zeng F, Zhang L. Mechanisms and therapeu-
kidney one-clip is a pertinent approach to integrate arterial hypertension tic potential of microRNAs in hypertension. Drug Discov Today.
in animal models of stroke: serial magnetic resonance imaging studies of 2015;20:1188–1204. doi: 10.1016/j.drudis.2015.05.007
brain lesions before and during cerebral ischemia. J Cereb Blood Flow 350. Zhu XY, Ebrahimi B, Eirin A, Woollard JR, Tang H, Jordan KL, Ofori
Metab 2018;38:1769–1780. doi: 10.1177/0271678X17715813 M, Saad A, Herrmann SM, Dietz AB, Textor SC, Lerman A, Lerman LO.
335. Chan SL, Baumbach GL. Nox2 deficiency prevents hypertension-
Renal vein levels of microRNA-26a are lower in the poststenotic kidney.
induced vascular dysfunction and hypertrophy in cerebral arterioles. Int J Am Soc Nephrol. 2015;26:1378–1388. doi: 10.1681/ASN.2014030248
J Hypertens. 2013;2013:793630. doi: 10.1155/2013/793630 351. Mell B, Jala VR, Mathew AV, Byun J, Waghulde H, Zhang Y, Haribabu
336. Carnevale D, Mascio G, D’Andrea I, Fardella V, Bell RD, Branchi I, B, Vijay-Kumar M, Pennathur S, Joe B. Evidence for a link between
Pallante F, Zlokovic B, Yan SS, Lembo G. Hypertension induces brain gut microbiota and hypertension in the Dahl rat. Physiol Genomics.
β-amyloid accumulation, cognitive impairment, and memory dete- 2015;47:187–197. doi: 10.1152/physiolgenomics.00136.2014
rioration through activation of receptor for advanced glycation end 352. Yang T, Santisteban MM, Rodriguez V, Li E, Ahmari N, Carvajal JM,
products in brain vasculature. Hypertension. 2012;60:188–197. doi: Zadeh M, Gong M, Qi Y, Zubcevic J, Sahay B, Pepine CJ, Raizada MK,
10.1161/HYPERTENSIONAHA.112.195511 Mohamadzadeh M. Gut dysbiosis is linked to hypertension. Hypertension.
337. Hu JJ, Fossum TW, Miller MW, Xu H, Liu JC, Humphrey JD.
2015;65:1331–1340. doi: 10.1161/HYPERTENSIONAHA.115.05315
Downloaded from http://ahajournals.org by on March 14, 2019

Biomechanics of the porcine basilar artery in hypertension. Ann Biomed 353. Jose PA, Raj D. Gut microbiota in hypertension. Curr Opin Nephrol
Eng. 2007;35:19–29. doi: 10.1007/s10439-006-9186-5 Hypertens. 2015;24:403–409. doi: 10.1097/MNH.0000000000000149
338. Olver TD, Klakotskaia D, Ferguson BS, Hiemstra JA, Schachtman TR, 354. Pevsner-Fischer M, Blacher E, Tatirovsky E, Ben-Dov IZ, Elinav E.
Laughlin MH, Emter CA. Carotid artery vascular mechanics serve as The gut microbiome and hypertension. Curr Opin Nephrol Hypertens.
biomarkers of cognitive dysfunction in aortic-banded miniature swine 2017;26:1–8. doi: 10.1097/MNH.0000000000000293
that can be treated with an exercise intervention. J Am Heart Assoc. 355. Wilck N, Matus MG, Kearney SM, Olesen SW, Forslund K, Bartolomaeus
2016.5:e003248. doi: 10.1161/JAHA.116.003248 H, Haase S, Mähler A, Balogh A, Markó L, Vvedenskaya O, Kleiner
339. Kemper T, Moss MB, Hollander W, Prusty S. Microinfarction as a result FH, Tsvetkov D, Klug L, Costea PI, Sunagawa S, Maier L, Rakova
of hypertension in a primate model of cerebrovascular disease. Acta N, Schatz V, Neubert P, Frätzer C, Krannich A, Gollasch M, Grohme
Neuropathol. 1999;98:295–303. DA, Côrte-Real BF, Gerlach RG, Basic M, Typas A, Wu C, Titze JM,
340. Prusty S, Kemper T, Moss MB, Hollander W. Occurrence of stroke Jantsch J, Boschmann M, Dechend R, Kleinewietfeld M, Kempa S,
in a nonhuman primate model of cerebrovascular disease. Stroke. Bork P, Linker RA, Alm EJ, Müller DN. Salt-responsive gut commen-
1988;19:84–90. sal modulates TH17 axis and disease. Nature. 2017;551:585–589. doi:
341. Moss MB, Jonak E. Cerebrovascular disease and dementia: a pri-
10.1038/nature24628
mate model of hypertension and cognition. Alzheimers Dement. 356. Wanjek C. Systems biology as defined by NIH: an intellectual resource
2007;3(suppl):S6–S15. doi: 10.1016/j.jalz.2007.01.002 for integrative biology. The NIH Catalyst. 2011. https://irp.nih.gov/cata-
342. Lecrux C, Nicole O, Chazalviel L, Catone C, Chuquet J, MacKenzie lyst/v19i6/systems-biology-as-defined-by-nih. Accessed November 1,
ET, Touzani O. Spontaneously hypertensive rats are highly vulner- 2017.
able to AMPA-induced brain lesions. Stroke. 2007;38:3007–3015. doi: 357. Kirschner MW. The meaning of systems biology. Cell. 2005;121:503–
10.1161/STROKEAHA.107.491126 504. doi: 10.1016/j.cell.2005.05.005
343. Jeffs B, Clark JS, Anderson NH, Gratton J, Brosnan MJ, Gauguier D, 358. Begley CG, Ellis LM. Drug development: raise standards for preclinical
Reid JL, Macrae IM, Dominiczak AF. Sensitivity to cerebral ischaemic cancer research. Nature. 2012;483:531–533. doi: 10.1038/483531a
insult in a rat model of stroke is determined by a single genetic locus. Nat 359. Perrin S. Preclinical research: make mouse studies work. Nature.

Genet. 1997;16:364–367. doi: 10.1038/ng0897-364 2014;507:423–425. doi: 10.1038/507423a

You might also like