Next Article in Journal
MicroRNAs in Learning and Memory and Their Impact on Alzheimer’s Disease
Next Article in Special Issue
Elevated Vascular Sympathetic Neurotransmission and Remodelling Is a Common Feature in a Rat Model of Foetal Programming of Hypertension and SHR
Previous Article in Journal
Special Issue “Mitochondria and Brain Disease”
Previous Article in Special Issue
Sacubitril/Valsartan and Ivabradine Attenuate Left Ventricular Remodelling and Dysfunction in Spontaneously Hypertensive Rats: Different Interactions with the Renin–Angiotensin–Aldosterone System
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Genetic Modifications to Alter Blood Pressure Level

Department of Functional Pathology, Faculty of Medicine, Shimane University, Izumo 693-8501, Japan
*
Author to whom correspondence should be addressed.
Biomedicines 2022, 10(8), 1855; https://doi.org/10.3390/biomedicines10081855
Submission received: 30 June 2022 / Revised: 27 July 2022 / Accepted: 29 July 2022 / Published: 1 August 2022

Abstract

:
Genetic manipulation is one of the indispensable techniques to examine gene functions both in vitro and in vivo. In particular, cardiovascular phenotypes such as blood pressure cannot be evaluated in vitro system, necessitating the creation of transgenic or gene-targeted knock-out and knock-in experimental animals to understand the pathophysiological roles of specific genes on the disease conditions. Although genome-wide association studies (GWAS) in various human populations have identified multiple genetic variations associated with increased risk for hypertension and/or its complications, the causal links remain unresolved. Genome-editing technologies can be applied to many different types of cells and organisms for creation of knock-out/knock-in models. In the post-GWAS era, it may be more worthwhile to validate pathophysiological implications of the risk variants and/or candidate genes by creating genome-edited organisms.

1. Introduction

Hypertension is the leading preventable risk factor for cerebro-cardiovascular complications, including heart failure and stroke. Effective anti-hypertensive drugs with different pharmacological actions have been developed; nevertheless, it is deemed that there are 1.28 billion hypertensive patients globally and 0.7 billion or more patients are untreated [1]. Given the resulting mortality and disability as well as the high prevalence, hypertension is still a major public health burden in the world.
It is needless to say that gene-targeted knock-out (KO) and knock-in (KI) or transgenic rodent models have greatly contributed to understanding the pathophysiological basis of hypertension and its vascular complications. In particular, mice have been widely used as the best experimental animal since the gene engineering technique to create KO models was established for over 30 years ago. By contrast, it had been technically difficult to create KO rats for a long time because of the difficulty of rat ES cell culture. Recent advances in genome-editing technologies, however, have made it possible to easily create KO rats similar to mice [2,3]. Given that spontaneous cerebro-cardiovascular disease models, such as Dahl salt-sensitive (SS) and stroke-prone spontaneously hypertensive rats (SHRSP), have been commercially available, a genome-editing strategy using the rat disease models has much potential to clarify the novel pathogenesis of hypertension. In this review, we outlined recent advances in basic research for hypertension using KO and KI or transgenic rodent models to clarify the underlying mechanisms.

2. Mouse Models

Essential hypertension is a highly complex pathological condition that is formed by synergistic influences of multiple lifestyles, social, environmental, and genetic factors. Since blood pressure (BP) is collaboratively controlled by various organs and tissues, there are many studies that have investigated tissue (or cell)-specific roles of genes on BP regulation using conventional and conditional KO or transgenic mice. In contrast to rats, no spontaneous hypertensive mouse models have been established; accordingly, angiotensin II (Ang II)-infused models have been widely used to investigate the pathogenesis of Ang II-related hypertension. Deoxycorticosterone acetate (DOCA)-salt or high-salt diet (usually containing 4% or 8% NaCl) models have been also used to investigate the pathogenesis of salt-sensitive hypertension. In this section, we overview proposed mechanisms for controlling BP found in the phenotyping of KO or transgenic mouse models, especially focusing on the findings in the recent decade.

2.1. Kidney

The kidney plays pivotal roles in arterial BP regulation by controlling blood volume and plasma electrolyte balance. Activities of the renin–angiotensin–aldosterone system (RAAS) and mineral transporters (Na+/H+ exchanger; NHE, Na+–K+–Cl co-transporter; NKCC, Na+–Cl co-transporter; NCC, epithelial sodium channel; ENaC, etc.) distributed along with nephron are important for physiological BP regulation; thus, genes that may regulate those activities have been widely investigated (Table 1).
Ang II regulates BP via Ang II type 1 receptor (Agtr1a, AT1R). As BP lowering effects were observed in proximal tubules (PT) or collecting duct (CD)-specific KO mice [4,5], blockade of AT1R signaling in renal epithelial cells would be a pharmacological target for hypertension therapy. Of note, AT1R-associated protein (Agtrap), which is widely distributed along renal tubules, has been found to suppress AT1R signaling by facilitating internalization of AT1R resulting in decreased cell surface expression of AT1R [6,7], suggesting that activation of endogenous AGTRAP has potential to reduce BP. In fact, it has been reported that the renal-specific overexpression and conventional KO mice show lower and higher BP phenotype compared with the wild-type (WT) control, respectively [8,9,10,11]. In contrast to the results in mice, however, the deletion in Dahl SS rats exacerbated renal damage under a 4% NaCl diet condition with no change in BP [12]. Although AGTRAP may play double-edged roles in reno-cardiovascular functions in a context-specific manner, it is a potential candidate gene located in a GWAS loci for BP in humans [12].
Although Ang II is the most well-known bioactive peptide hormone in the RAAS, (pro)renin and Ang-(1-7) produced by angiotensin-converting enzyme 2 (ACE2) are also known to regulate BP via its specific receptors. The (Pro)renin receptor (PRR) that specifically recognizes both prorenin and renin was cloned by Nguyen et al. in 2002 [13]. In the kidney, PRR is mainly expressed in renal vasculature, PT and distal tubules (DT), and CD and enhances the catalytic activity of (pro)renin that converts Ang I to Ang II, resulting in an increase in Ang II production [14]. Consistent with the physiological function of PRR, decreases in BP elevation induced by Ang II infusion have been observed in both tubular- and CD-specific KO mice through inhibition of ENaC activation [15,16,17]. Ang-(1-7) generated by mainly ACE2 is a vasoactive peptide that induces a vasodilation response by binding to Mas receptor [18]. Therefore, ACE2-Ang-(1-7)-Mas axis exerts a counteracting effect on Ang II that causes BP elevation. Ni et al. reported that conventional double KO of both ACE2 and Mas receptor in mice caused greater Ang II-induced BP elevation when compared with the WT littermates [19]. In addition, they also showed that the dual deletion of ACE2 and Mas receptor worsened hypertensive nephropathy, suggesting that ACE2-Ang-(1-7)-Mas receptor axis has protective roles in both the development of hypertension and the resulting hypertensive kidney injury.
Tubuloglomerular feedback (TGF) is an important physiological system to regulate long-term BP by sensing blood volume and electrolyte balance at the level of juxtaglomerular apparatus in each nephron [20]. Accumulating evidence has shown that local activities of renal oxide synthases (NOS), which produce a major chemical vasodilator NO, play an important role in the regulation of the TGF system. NOS families are composed of three isoforms, i.e., neuronal NOS (nNOS, encoded by Nos1), inducible NOS (iNOS, Nos2), and endothelial NOS (eNOS, Nos3). Although all the three isoforms are expressed in the kidney, Nos1 and Nos3 are thought to be major isoforms that physiologically participate in the TGF because of low baseline expression of Nos2. Interestingly, Lu et al. showed that macula densa-specific deletion of Nos1 exacerbated a high-salt diet-induced BP elevation under a condition of Ang II infusion accompanied by reduced glomerular filtration rate (GFR) and Na+ excretion [21]. It was also reported that local NOS1 activity at the macula densa contributed to a sex difference in BP response to Ang II [22]. Moreover, Hyndman et al. and Gao et al. have investigated renal-specific roles of NOS1 and NOS3 on BP regulation using CD-specific and nephron-specific KO mice, respectively [23,24]. They suggested that deletion of the two isoforms caused greater high-salt-induced BP elevation by enhancing ENaC [25] and NCC activities in the tubular cells, respectively.
Pathophysiological roles of NEDD4-2 (encoded by Nedd4l) and with-no-lysine kinases 1 and 4 (Wnk1 and Wnk4) in (salt-sensitive) hypertension have been well-investigated in humans as well as in rodent models. NEDD4-2 is an E3 ubiquitin ligase that ubiquitylates ENaC to down-regulate its cell surface expression and activity [26]. Although NEDD4-2 was initially found as a ENaC-specific regulator in the kidney [25], Ronzaud et al. reported that NEED4-2 also regulated NCC activity and its renal tubule-specific deletion caused salt-dependent hypertension [27]. Consequently, NEED4-2 is involved in the pathogenesis of salt-sensitive hypertension through the two-independent pathways that controls renal Na+ homeostasis. WNK1 and WNK4 are known to be responsible genes of pseudohypoaldosteronism type 2 (PHA2) that is caused by large deletions in intron 1 of WNK1 or gain-of function mutations in WNK4 [28]. Mechanistically, WNKs phosphorylate SPAK/OSR1, thereby activating NCC in the DT and resulting in increased Na+ reabsorption and salt-sensitive hypertension [28,29]; however, the molecular network may be a little complicated as a paradoxical role of kidney-specific WNK1 lacking a kinase domain on the development of salt-sensitive hypertension was reported [30]. Moreover, Mu et al. suggested a unique pathway involving salt-sensitive hypertension caused by epigenetic down-regulation of WNK4 [31]. In this context, kelch-like protein 3 (KLHL3) and cullin 3 (CUL3), which are the E3 ubiquitin ligase complex to degrade WNK, have also received much attention as target molecules to prevent salt-sensitive hypertension [28,29].
Unlike the local mechanisms in the kidney described above, Pan et al. uniquely identified the liver–kidney and liver–adipocytes axis to control BP via a hepatocytes-producing hormone, fibroblast growth factor 21 (FGF21), which has pleiotropic effects on glucose and lipid metabolism [32]. They found that FGF21 augmented peroxisome proliferator-activated receptor γ (PPARγ)-mediated activation of ACE2 in both the kidney and adipocytes; thereby, an increase in Ang-(1-7) production reduced both BP and vascular injury. Because FGF21 production was stimulated by Ang II, the FGF21–ACE2 axis may counteract Ang II-induced hypertension and the vascular injury. This might be a key mechanism in obesity-related hypertension.
Besides the above, multiple mechanisms have been proposed such as by circadian clock- [33,34], osmotic stress- [35], and genome-wide association study (GWAS)-related genes [36,37] as well.
Table 1. Target molecules in kidney.
Table 1. Target molecules in kidney.
TargetsType of Genetic ModificationModelsPhenotypesReferences
AGTRAP (angiotensin II receptor-associated protein, Agtrap)Renal tubule-specific overexpressionAng II↓BP, ↓NCC and αENaC activitiesWakui et al. [8]
Conventional KOAng II↑BP, ↑ENaC activityOhsawa et al. [9]
Conventional KO5/6 nephrectomy↑BP, ↑plasma volume, ↑αENaC and TNF-α expressionKobayashi et al. [10]
Proximal tubule-specific KOAng IINo differences in basal BP, pressor response to Ang II, and cardiac hypertrophyKinguchi et al. [11]
PRR ((Pro)renin receptor, Atp6ap2)Tubular-specific KOAng II↓BP, ↓Na+ retention, ↓αENaC expressionRamkumar et al. [15]
Collecting duct-specific KOAng II↓BP, ↓urinary renin and αENaC activitiesPeng et al. [16]
Collecting duct-specific KOAng II↓BP (basal and Ang II), ↓α/γENaC activation, ↓urinary Ang II and renin levelsPrieto et al. [17]
ACE2 (angiotensin-converting enzyme-2, Ace2), Mas receptor (Mas1)Conventional double KOAng II↑BP, ↑ renal injury, ↑serum Cr, ↓Cr clearanceNi et al. [19]
NOS1 (NO synthase 1, Nos1)Macula densa-specific KOAng II + high-salt diet↑BP, ↑ tubuloglomerular feedback response, ↓GFR, urine flow, and N+ excretionLu et al. [21]
Macula densa-specific KOAng IIDiminished sex difference in Ang II-induced BP, tubuloglomerular feedback response, and natriuretic responseZhang et al. [22]
Collecting duct-specific KOHigh-salt diet↑BP, ↓urine output, ↓Na+, Cl, and NOx excretionHyndman et al. [23]
NOS3 (NO synthase 3, Nos3)Doxycycline-inducible nephron-specific KOHigh-salt diet↑BP, ↑Na+ retention, ↑NCC activationGao et al. [24]
NEDD4-2 (Nedd4l)Tetracycline-inducible tubule-specific KOHigh-salt diet↑BP, ↑β/γENaC and ROMK expression, ↑NCC activation, hypercalciuriaRonzaud et al. [27]
WNK1 (with-no-lysine kinase 1, Wnk1)Kidney-specific overexpression of the kidney-specific isoformNo treatment↓BP, ↑plasma Ang II and aldosterone, ↓NCC and NKCC2 activationLiu et al. [30]
Kidney-specific KO (targeted deletion of the first exon of the kidney-specific isoform)High-salt diet↑BP, ↑Na+ retention, ↑NCC and NKCC2 activationLiu et al. [30]
FGF21 (fibroblast growth factor 21, Fgf21)Conventional KOAng II↑BP, ↑vascular hypertrophy and fibrosis, ↓vascular relaxation, ↓plasma/adipose ACE2 and Ang-(1-7), ↑plasma/adipose Ang IIPan et al. [32]
BMAL1 (brain and muscle ARNT-like 1, Arntl)Kidney-specific KONo treatment (or K+-restricted diet)↑BP, ↓Na+ retention under K+-restricted dietCrislip et al. [33]
Per1 (period 1, Per1)Distal nephron-specific KODOCP-salt↑BP, ↑Na+ retention, ↑plasma aldosterone, ↑medullary endothelin-1Douma et al. [34]
NFAT5 (nuclear factor of activated T-cells 5, Nfat5)Doxycycline-inducible tubular cell-specific KOHigh-salt diet↑BP, hypernatremia, polyuria, ↓Na+ excretion, ↑ENaC expressionHiramatsu et al. [35]
HSD11β2 (11β-hydroxysteroid dehydrogenase, Hsd11b2)Kidney-specific KONo treatment↑BP, ↑αENaC and NCC activationUeda et al. [36]
NPR-C (natriuretic peptide receptor-C, Npr3)Conventional KOAng II↓BP, ↑diuretic and natriuretic response, ↓NCC activation via WNK4/SPAKShao et al. [37]
Tubule-specific KOAng II↓BP, ↓NCC activation via WNK4/SPAKShao et al. [37]

2.2. Vasculatures

Table 2 summarizes target genes in vasculatures and the representative phenotypes described below. Peripheral vascular tone is one of the primary factors to control BP. Two primary cell types, i.e., endothelial cells (ECs) and vascular smooth muscle cells (VSMCs), play major roles in the regulation of the vascular tone mainly through production of vasodilators (NO, etc.) or vasoconstrictors (endothelin-1, etc.) and sympathetic vasoconstriction, respectively. Endothelial NOS (eNOS, NOS3) predominantly generates NO from L-arginine in ECs; thereby, the released NO activates NO-sensitive guanylyl cyclase (NO-GC) in VSMCs to increase cytosolic cGMP, then the activated cGMP-dependent protein kinase (PKG) induces smooth muscle relaxation. The NO-GC/cGMP/PKG signaling is indispensable for NO-dependent BP regulation as spontaneous BP elevation was found in VSMC-specific NO-GC deficient mice [38].
In addition, NO-independent pathways that stimulate cGMP/PKG also exist. Natriuretic peptides, which are composed of A- (atrial; ANP), B- (brain; BNP), and C-type (CNP), are well-studied vasoactive peptides that exert vasodilation via direct activation of the transmembrane receptor NPR1 (natriuretic peptide receptor 1, also known as guanylyl cyclase-A; GC-A) or NPR2 (also known as GC-B). Among the three members, CNP is secreted from ECs and specifically bind to NPR2, whereas ANP and BNP are cardiac peptides targeting NPR1 [39]. In addition, CNP is thought to be an autocrine/paracrine factor in the circulation system because of the relatively low plasma concentration compared with ANP and BNP [40]. Several recent reports have uncovered the detailed mechanisms of NPR1/2-mediated vasodilation.
Nakao et al. showed that EC-specific CNP KO mice had higher BP compared with WT control independently of NO production, whereas VSMC-specific NPR2 KO had unaltered BP [41]. On the other hand, Špiranec et al. thereafter reported that the deletion in ‘precapillary arteriole SMCs and capillary pericytes’ caused BP elevation in mice accompanied by an impaired CNP-induced vasodilatory response [42]. Collectively, these results indicate that EC-derived CNP acts on precapillary arteriole SMCs and capillary pericytes as well as ECs to lower peripheral vascular resistance and BP through an NO-independent manner. It is of note that a CNP-induced vasodilatory response in mesenteric arteries was also impaired in VSMC-specific KO mice by Nakao et al.; nevertheless, BP of the KO mice was compatible with that of WT [41]. Conflicting results for the BP phenotype between the two VSMC-specific KO models may be partly due to the difference in promoters driving Cre expression (sm22 [41] or Pdgf-rb [42] promoter) to create the conditional NRP2 KO mice. Furthermore, it was very recently reported that EC-specific, but not VSMC-specific, deletion of NPR1 diminished BP reduction by intravenous ANP administration [43].
Intracellular Ca2+ mobilization is a key modulator to induce both NOS-mediated vascular relaxation by EC and VSMC contraction; thus, cell-type specific genetic modification is necessary to clarify functional implications of the target molecules on vascular responses. Stromal interaction molecule 1 (Stim1) is an endoplasmic reticulum (ER) resident transmembrane protein that senses Ca2+ store in ER lumen via its N-terminal EF hand motif. When the Ca2+ store is depleted, STIM1 moves toward the cytoplasmic membrane and opens the target Ca2+ channels, ORAI1, and transient receptor potential families (TRPs) to elicit Ca2+ entry into the cytosol (store-operated Ca2+ entry; SOCE) [44]. STIM1 is expressed in broad cell types including ECs and VSMCs and plays key roles in the maintenance of intracellular Ca2+ homeostasis [43]. Kassan et al. revealed that VSMC-specific deletion of Stim1 ameliorated Ang II-induced hypertension with decreased vascular ER stress [45]. By contrast, a significant increase in nighttime BP was observed in EC-specific Stim1 KO mice that showed decreased NO production and an EC-dependent vasodilation phenotype in vitro [46]. These studies suggest that STIM1 exerts an opposite role in the regulation of vascular tone in the two different cells. Interestingly, we found that the stroke-prone spontaneously hypertensive rat (SHRSP) had a premature stop codon in this gene that caused the expression of truncated STIM1 with decreased SOCE activity [47,48]. Although the recovery of STIM1 function in SHRSP by CRISPR-Cas9-mediated gene KI did not alter the BP [49], systemic impairment of SOCE activity would have important implications for the pathogenesis of hypertensive end-organ damage in SHRSP independently of the BP phenotype. Phenotyping of the KI rat model is currently in progress; the findings will be described elsewhere.
Fluid shear stress is an important mechanical stimulus that physiologically enhances NO production by ECs to maintain vascular integrity. Increasing evidence has shown that a mechanosensitive cation channel PIEZO1 on ECs mediates laminal flow-dependent activation of purinergic P2Y2 receptor, thereby activating PI3K/Akt signaling to phosphorylate NOS3 [50]. Recently, an alternative pathway mediated by the PIEZO1adrenomedullin (ADM) axis was reported [51]. ADM is a circulatory vasodilator and diuretic and natriuretic peptide that is mainly produced by ECs [52]. Iring et al. showed that PIEZO1 enhanced endothelial ADM secretion, then the secreted ADM bound to calcitonin receptor-like receptor (CALCRL) on ECs by an autocrine/paracrine fashion. ADM-CALCRL complex activates its adjacent adenylyl cyclase; thereby, cAMP-dependent protein kinase (PKA) phosphorylates and activates NOS3, resulting in NO-dependent vasorelaxation [50]. Actually, it is of interest that all EC-specific single KO of ADM, CALCRL, and Gαs, which is the downstream G protein of CALCRL, in mice caused apparent BP elevation. This finding indicates an essential role of PIEZO1-ADM signaling on controlling vascular tone and BP at a resting condition.
Prostaglandins (PGs) are endogenous lipid mediators that have multiple bioactivities such as uterine contraction, platelet aggregation, and bronchodilation and is generated from arachidonic acid by catalytic activities of cyclooxygenases (COXs). PGs are also involved in BP regulation as nonsteroidal anti-inflammatory drugs (NSAIDs), which block COX activity, have hypertensive side effects [53]. Among the known PGs, PGE2 is a major prostanoid that affects BP both positively and negatively via its specific receptor EP1-4 [54,55]. Thus far, it was shown that EP1 and EP3 mediate vasoconstrictive response, while EP2 and EP4 lead to vasodilation [53]. The diverse effects of PGE2 on vascular functions may be due to the characteristic of tissue distribution of the receptors. Recently, Xu et al. reported that EC-specific KO and overexpression of EP4 resulted in higher and lower BP compared with control mice, respectively [56], under both basal and high-salt diet conditions. Physiological roles of PGs on BP regulation may be still controversial; however, the recent report clearly indicates a hypotensive potential of PGE2-EP4 signaling via enhancing NO production in ECs.
Table 2. Target molecules in vasculatures.
Table 2. Target molecules in vasculatures.
Targets (Official Symbols)Type of Genetic ModificationModelsPhenotypesReferences
NO-GC (NO-sensitive guanylyl cyclase, Gucy1b1)Tamoxifen-inducible VSMC-specific KONo treatment↑BP, ↓NO-induced vasorelaxationGroneberg et al. [38]
CNP (C-type natriuretic peptide, Nppc)EC-specific KONo treatment↑BP, ↓acetylcholine- and endothelium-dependent relaxation, ↑Endothelin-1 and Ace expression in ECsNakao et al. [41]
NPR2 (natriuretic peptide receptor 2, Npr2)/Guanylyl cyclase-B (GC-B)VSMC-specific KONo treatmentNo difference in BP, ↓CNP-induced relaxation in mesenteric arteriesNakao et al. [41]
Tamoxifen-inducible EC-specific KONo treatment↑BP, ↓cGMP productionŠpiranec et al. [41]
NPR1 (natriuretic peptide receptor 1, Npr1)EC-specific KONo treatmentLoss of EC-dependent BP reduction by ANP, unaltered NO production, K+ channel-mediated hyperpolarization in ECTokudome et al. [43]
STIM1 (stromal interaction molecule 1, Stim1)VSMC-specific KOAng II↓BP, ↓cardiac hypertrophy, ↓perivascular fibrosis, ↓endothelial dysfunctionKassan et al. [45]
EC-specific KONo treatment↑BP (nighttime), ↓NO production, ↓endothelium-dependent relaxationNishimoto et al. [46]
ADM (adrenomedullin, Adm), CALCRL (calcitonin receptor-like, Calcrl), Gαs (GNAS (guanine nucleotide binding protein, alpha stimulating) complex locus, Gnas)Tamoxifen-inducible EC-specific KONo treatment↑BP, ↓eNOS activation, ↓flow-induced vasorelaxationIring et al. [51]
EP4 (prostaglandin E2 receptor, Ptger4)EC-specific KONo treatment, high-salt diet, Ang II↑BP, ↓NO production, ↓vasorelaxation responseXu et al. [56]
EC-specific overexpressionNo treatment, high-salt diet↓BP, ↑eNOS activation, ↑NO productionXu et al. [56]

2.3. Immunity

A growing body of evidence has emerged in the last decade suggesting the pathogenic aspects of innate and adaptive immune responses on the development and progression of hypertension and hypertensive end-organ damages. The possible mechanisms have been well reviewed [57,58,59]; herein, we shortly highlight recent findings on this topic (Table 3). Among various subpopulations of immune cells, previous reports indicate that T cells especially have diverse contributions to the etiology [56]. In particular, CD4+- and regulatory T cell (Treg)-mediated pathological cascades have been raised in several recent studies.
Hydrogen sulfide (H2S) is a cardioprotective endogenous gaseous mediator that is generated by three major enzymes: cystathionine beta synthase (CBS), cystathionine gamma lyase (CSE), or 3-mercaptopyruvate sulfurtransferase [60]. Although it was reported that a conventional CSE KO mice showed an age-dependent increase in BP [61], Cui et al. revealed that CD4+ T cell-specific deletion of CSE was sufficient to induce greater BP in mice under both physiological and Ang II-treated conditions [62]. Mechanistically, they suggest that CSE-derived H2S activates liver kinase B1 (LKB1)-PKA signaling and the resulting activation of Treg attenuates vascular and renal inflammation, thereby preventing BP elevation. In addition, Sun et al. reported that mineralocorticoid receptor (MR) deficiency in CD4+ T cells ameliorated Ang II-induced BP elevation and vascular and renal damage in mice [63]. In contrast to the KO model, the MR overexpression exacerbated the increase in BP after Ang II infusion; however, IFN-γ-neutralizing antibodies could abolish the deleterious effect, suggesting that IFN-γ produced by infiltrated T cells was a key cytokine link between MR signaling in CD4+ T cells and the resulting hypertension.
For Treg, a detailed pathological mechanism caused by a microRNA function has been proposed. MicroRNAs (miRs) are involved in numerous (patho)physiological conditions by controlling gene expression mainly at a translational level, and among the identified miRs, miR-31 has multifaceted roles in regulation of immune responses [64]. Interestingly, Li et al. reported that Ang II-induced BP elevation and vascular and renal damage were reduced in mice lacking miR-31 in Treg compared with control mice, which were accompanied by increased Treg differentiation [65]. Furthermore, the opposite phenotypes were observed in mice with Treg-specific deletion of protein phosphatase 6c (Ppp6c), a direct target of miR-31, suggesting that Ppp6c had potential to improve Ang II-induced hypertension [64]. This may be a novel posttranslational mechanism that worsens hypertensive phenotypes through an overexpression of a specific microRNA that regulates Treg functions.
AT1R, an Ang II receptor, is widely expressed in immune cells [66]. It has been shown that deletion of AT1R on T lymphocytes or macrophages do not affect BP even under an Ang II-infused condition [67,68]. In contrast to the previous findings, Lu et al. revealed that the deletion on CD11c+ myeloid cells (dendritic cells; DCs) in mice with chronic Ang II infusion resulted in increased BP, renal infiltration of inflammatory cells (memory T, CD40+ DCs), and Na+ retention with greater β/γENaC expression [69]. It is of interest that AT1R on DCs exerts a cardioprotective role in spite of harmful effects of Ang II on renal and cardiovascular functions. Moreover, Sag et al. showed that mice with myeloid cell-, but not endothelial cell-, specific deletion of NADPH oxidase 2 (NOX2), which is a superoxide-generating enzyme, had lower basal BP compared with control mice [70].
Beside the above, the pathophysiological actions of C-C motif chemokine receptor 7 (CCR7) [71], toll-like receptor 3/4 (TLR3/4) [72], placental growth factor (PlGF) [73], complement C3a/C5a receptors (C3aR/C5aR) [74], T cell receptor delta chain (TCRδ) [75], and interleukin-1 receptor type 1 (IL-1R1) [76] have been also proposed using conventional KO mouse models. Overall, accumulated evidence has commonly suggested pathophysiological connections between immune responses and renal dysfunction on the development of hypertensive conditions. Clinical perspectives of anti-inflammatory therapies targeting specific cytokines were also discussed [77].
Table 3. Target molecules in immune system.
Table 3. Target molecules in immune system.
Targets (Official Symbols)Type of Genetic ModificationModelsPhenotypesReferences
CSE (Cystathionine γ lyase, Cth)CD4+ T cell-specific KOAng II↑BP, ↑blood and renal Treg, ↑renal and peripheral adipose tissue CD4+/CD8+ TCui et al. [62]
MR (mineralocorticoid receptor)/nuclear receptor subfamily 3, group C, member 2 (Nr3c2)CD4+ T cell-specific KOAng II↓BP, ↓renal/vascular damage, ↓IFNγ-producing T cellSun et al. [63]
CD4+ T cell-specific overexpressionAng II↑BPSun et al. [63]
MicroRNA-31 (miR-31, Mir31)Conventional and Treg-specific KOAng II↓BP, ↑Treg differentiation,↓renal and vascular injuryLi et al. [65]
Ppp6c (protein phosphatase 6c, Ppp6c)Treg-specific KOAng II↑BP, ↓Treg differentiation,↑renal injuryLi et al. [65]
AT1R (angiotensin II receptor type 1, Agtr1a)CD11c+ cell-specific KOAng II↑BP, ↑renal memory T and CD40+ DC, ↑ENaCLu et al. [69]
NOX2 (NADPH oxidase 2, Cybb)Myeloid cells-specific KONo treatment↓BP, ↑NO bioavailabilitySag et al. [70]
Ang IINo effect on BPSag et al. [70]
CCR7 (C-C motif chemokine receptor 7, Ccr7)Conventional KOAng II↓BP, ↑renal CD8+ T, ↓renal draining lymph node CD4+ T and CD8+ TWen et al. [71]
TLR3/4 (toll-like receptor 3/4, Tlr3/4)Conventional KOAng II↓BP and cardiac hypertrophy in TLR3 KO, ↓cardiac hypertrophy in TLR4 KOSingh et al. [72]
PIGF (placental growth factor, Pgf)Conventional KODOCA-salt↓BP, ↓renal damage and T cell infiltrationPerrotta et al. [73]
C3aR/C5aR (complement 3a and 5a receptors, C3ar1/C5ar1)Conventional double KOAng II↓BP, ↑renal Treg, ↓renal/vascular remodelingChen et al. [74]
TCRδ (T cell receptor delta chain, Tcrd)Conventional KOAng II↓BP, ↓endothelial dysfunctionCaillon et al. [75]
IL-1R1 (IL-1 receptor type 1, Il1r1)Conventional KOAng II↓BP, ↓NKCC2 activityZhang et al. [76]

2.4. Other Organs and Tissues (Brain, Adipocyte, and Adrenal Gland)

Table 4 summarizes target genes in other major organs and tissues and the representative phenotypes. Pathological implications of brain RAAS on hypertension have been well investigated [78]. Based on the distribution of RAAS components in the brain, it has been verified that brain RAAS activity can induce BP elevation independently of renal RAAS function by using cell type-specific transgenic mice targeting AGT and/or renin [79,80,81]. Consistent results were observed in DOCA-salt mice with neuron-specific deletion of PRR that exhibited decreases in BP and brain Ang II production [82].
Pathological relationships between salt intake and hypertension have been long suggested in humas as well as in rodents; however, the precise mechanism remains elusive. In this context, it is noteworthy that Nomura et al. reported that the Nax channel expressed in specific glial cells in the organum vasculosum lamina terminalis (OVLT) functioned as the brain sensor detecting [Na+] increase in the body and that deletion of Nax diminished salt-induced hypertensive phenotype [83]. Concerning this, neuronal 11β-hydroxysteroid dehydrogenase type2 (Hsd11b2), which encodes a corticosterone-producing enzyme, and PRR have been proposed to be involved in both the development of salt-sensitive hypertension and sodium appetite [84,85]. In addition, PRR deficiency in adipocytes was pathologically implicated in a high-fat diet-induced BP increase in male mice but not in female mice [86].
The adrenal gland is a major endocrine organ that plays a pivotal role in BP regulation and fluid and electrolyte homeostasis via production of steroid hormones and catecholamines. The two-pore domain K+ channels (TASKs) expressed in zona glomerulosa (zG) cells down-regulate the production of aldosterone in the cells [87]. Guagliardo et al. showed that zG cell-specific deletion of TASK-1 and -3 caused autonomous hyperaldosteronism and chronic BP elevation in mice [88]. In addition, Mathar et al. reported that mice lacking transient receptor potential melastatin 4 (TRPM4) had chronically increased BP with exaggerated sympathetic tone [89]. TRP families are non-selective cation channels that are involved in many physiological processes and are regarded as potential targets for drug design for various diseases [50]. According to the report by Mathar et al., TRPM4 deficiency increases catecholamine release from chromaffin cells and thereby augments sympathetic tone resulting in a continuous BP elevation.
It is generally known that plasma concentrations of adrenal gland-derived steroid hormones are controlled by the physiological circadian rhythm of adrenocorticotropic hormone (ACTH) secretion. Circadian clock genes, cryptochrome-1 and -2 (Cry-1 and -2), play key roles in this mechanism by direct regulation of Hsd3b6 expression encoding an aldosterone-producing enzyme, 3β-hydroxysteroid dehydrogenase-isomerase (3β-HSD). Therefore, Cry-1 and Cry-2 KO mice exhibited salt-sensitive hypertension under a high-salt diet condition due to constitutive activation of 3β-HSD such asthe DOCA-salt model [90].
Multiple genetic and physiological mechanisms as thus far described are complicatedly involved in the pathogenesis of hypertension. Furthermore, the pathogenic roles of epigenetic modifications [91,92,93], microbiota/metabolome [94,95,96], and sympathetic overactivity [97,98,99] in cardiovascular disease have been also discussed.
Table 4. Target molecules in brain, adipocyte, and adrenal gland.
Table 4. Target molecules in brain, adipocyte, and adrenal gland.
Organs, TissuesTargets (Official Symbols)Type of Genetic ModificationModelsPhenotypesReferences
BrainHuman AGT (angiotensinogen, AGT)Glial-specific overexpressionNo treatment↑BP,↑salt preferenceMorimoto et al. [79]
Human REN (renin, REN)Glial- and neuron-specific overexpressionNo treatment↑BP, ↑salt preferenceMorimoto et al. [80]
Human AGT (angiotensinogen, AGT)Glial-specific KONo treatment↓BPSherrod et al. [81]
PRR ((Pro)renin receptor, Atp6ap2)Neuron-specific KODOCA-salt↓BP, ↓brain Ang II production, ↓cardiac and vasomotor sympathetic toneLi et al. [82]
Nax (sodium channel, voltage-gated, type VII, alpha, Scn7a)Conventional KOHigh-salt diet↑BPNomura et al. [83]
HSD11β2 (11β-hydroxysteroid dehydrogenase, Hsd11b2)Neuron-specific KOHigh-salt water↑BP, ↑salt preferenceEvans et al. [84]
AdipocytesPRR ((Pro)renin receptor, Atp6ap2)Adipocyte-specific KOHigh-fat diet↑BP (basal and high fat diet-induced), ↑glucose tolerance, ↓diet-induced obesityWu et al. [86]
Adrenal glandTASK-1/3 (potassium channel, subfamily K, member 3/9, Kcnk3/9)Zona glomerulosa cells-specific KONo treatment↑BPGuagliardo et al. [88]
TRPM4 (transient receptor potential cation channel, subfamily M, member 4, Trpm4)Conventional KONo treatment↑BP, ↑plasma epinephrine, ↑urinary catecholamine metabolitesMathar et al. [89]
Cry1/2 (cryptochrome-1/2, Cry1/2)Conventional KOHigh-salt diet↑BP, ↑increased expression and activity of 3β-HSDDoi et al. [90]

3. Rat Models

Rats are the generally used experimental animal the same as mice and have some advantages compared with mice such as large body and tissue size and physiological properties similar to those in humans. Despite the advantages, mice have been more frequently used than rats; this is probably due to, except for higher experimental costs than mice, the technical difficulty of creating KO/KI rats. However, genome-editing technologies, i.e., zinc finger nuclease (ZFN), transcription activator-like effector nuclease (TALEN), and clustered regularly interspaced short palindromic repeats (CRISPRs)-associated proteins 9 (CRISPR-Cas9), made it possible to also create KO/KI rats easily. In 2009, Geurts et al. first reported the creation of KO rats by ZFN [100]. Thereafter, a growing body of literature has emerged in the last decade reporting phenotypes of KO/KI rats including genetic hypertensive models as below [101,102].

3.1. KO Models of SHR and SHRSP

SHR (spontaneously hypertensive rat) is a representative genetically hypertensive model that was established by selective breeding of rats with relatively high blood pressure in an outbred colony of Wistar rats that had been maintained in Kyoto University. SHRSP (stroke-prone SHR) is a substrain of SHR that genetically develops more severe hypertension and stroke. Despite the fact that both strains have been widely used for clarifying the responsible genes and the underlying mechanisms of hypertension and its complications [103,104,105], the literature evaluating cardiovascular phenotypes by using KO/KI models are still scant (Table 5).
SHR is a useful model for hypertensive cardiac hypertrophy [106]. A quantitative trait locus (QTL) related to the left ventricular hypertrophy was previously mapped on chromosome (Chr) 8 by phenotyping of congenic strains between SHR and normotensive Brown Norway (BN) rats [107]. Liška et al. identified promyelocytic leukemia zinc finger (Plzf) as a candidate gene on the cardiac QTL and showed that the deletion in SHR did not alter the BP but ameliorated cardiac hypertrophy and fibrosis [108].
Complement 3 (C3) that is overexpressed in aortic smooth muscle cells of SHR has been proposed as a candidate gene responsible for the development of hypertension in this model [109]. Mechanistically, C3-C3a receptor signaling accelerates a change in the characteristic of VSMC and glomerular mesangial cells from contractile to synthetic phenotype via activation of Krüppel-like factor 5 (KLF5) that is a transcription factor to induce the synthetic phenotype of mesenchymal cells [110,111]. Negishi et al. revealed that the C3 deficiency mitigated a salt-sensitive BP elevation and renal injury with decreased renal Ang II level and urinary catecholamine excretion [112].
Rubattu et al. previously identified a QTL on Chr 1 responsible for the susceptibility to salt-induced stroke by a linkage analysis F2 cross between SHR and SHRSP [113]. They identified NADH dehydrogenase (ubiquinone) 1 subunit C2 (Ndufc2), encoding a component of the electron transport chain, as a plausible candidate gene in the stroke QTL, then proved that the heterozygous deletion in SHR by ZFN strongly exacerbated the stroke susceptibility with increased oxidative stress and inflammation both in vitro and in vivo [114].
Besides the above, we recently created peroxiredoxin 2 (Prdx2) KO SHR to investigate whether the deletion of an antioxidant gene exacerbates cerebro-cardiovascular phenotypes of SHR [115]. Consequently, Prdx2 KO SHR had greater basal BP compared with WT SHR. Furthermore, the lifespan of Prdx2 KO SHR under a salt loading condition was shorter than that of WT SHR despite no difference in BP after salt loading between the KO and the WT. No apparent inter-strain differences were found in histopathological evaluation for brain, heart, and kidney lesions, and therefore, the reason for the short life span of Prdx2 KO SHR under the salt loading condition remains fully unknown.
Lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) is an endothelial scavenger receptor that is closely involved in the pathogenesis of atherosclerosis [116]. Recently, Liang et al. reported that LOX-1 deficiency had a protective role in spontaneous brain damage in SHRSP with no significant change of BP [117].
We previously found a QTL on Chr1 that affected exaggerated sympathetic responses to the stress of SHRSP by genetic analysis of congenic lines between SHRSP and normotensive Wistar-Kyoto rat (WKY) [118]. Among the genes in the QTL region, stromal interaction molecule 1 (Stim1) with a nonsense mutation in SHRSP was identified as a promising candidate ([47], see also Section 2.2 Vasculatures). As STIM1 plays a key role in Ca2+ homeostasis in the body, we expected that the Stim1 mutation was a genetic determinant responsible for cerebro-cardiovascular traits; however, no significant differences were observed in the sympathetic stress responses as well as age-dependent changes in BP between Stim1 KI SHRSP and SHRSP, i.e., with WT and mutant allele for Stim1, respectively [49]. Phenotyping of the Stim1 KI SHRSP is currently in progress, and the results will be described elsewhere.

3.2. KO Models of Dahl SS

Dahl salt-sensitive (SS) rats originate from a closed colony of Sprague-Dawley (SD) rats and are widely used as a salt-sensitive hypertension model that develop severe hypertension (>200 mmHg) and the complications such as hypertensive kidney injury and heart failure when fed high-salt diets [119]. SS/Jr and DSS/N strains have been separately established by Rapp and Iwai, respectively. Compared with SHR and SHRSP, multiple KO/KI models with SS/Jr genetic backgrounds have been actively created (Table 5).
In 2011, Moreno et al. first reported the phenotype of renin KO SS/Jr, in which a severe decrease in basal BP and abnormal kidney morphologies were observed [120]. Thereafter, a growing literature has shown pathophysiological implications of multiple genes on cardiorenal disease traits in SS/Jr [121,122,123,124,125,126,127,128,129,130,131,132,133]. Among them, pleckstrin homology domain containing family A member 7 (Plekha7) is a plausible candidate gene for essential hypertension identified by GWAS. A risk variation on Plekha7, encoding an adherence junction protein [134], for elevated systolic BP has been found in multiple human populations [135,136,137,138,139]. In this context, Endres et al. created SS/Jr lacking the functional domain of Plekha7 by ZFN and revealed that the Plekha7 functional KO SS/Jr had significantly lower BP and renal and cardiac damage under the 8% high-salt diet condition [123]. This is a meaningful study that verified a direct effect of the GWAS gene on the hypertensive phenotype in a genetic rat model with salt sensitivity.
Table 5. Target molecules in rat models.
Table 5. Target molecules in rat models.
StrainsTargets (Official Symbols)MethodsPhenotypesReferences
SHR/OlaIpcvPlzf (promyelocytic leukemia zinc finger, Zbtb16)TALEN↑cardiomyocyte hypertrophy and fibrosisLiška et al. [108]
SHR/NCrlNdufc2 (NADH dehydrogenase (ubiquinone) 1 subunit C2, Ndufc2)ZFNNo effect on BP, ↑salt-induced stroke susceptibility, oxidative stress, and inflammatory signalingRubattu et al. [113]
SHR/IzmC3 (complement 3, C3)ZFN↓salt-induced BP, ↓renal Ang II level, ↓urinary catecholamine excretionNegishi et al. [112]
Prdx2 (peroxiredoxin 2, Prdx2)CRISPR-Cas9↓basal BP, ↑life span under salt loading conditionMahal et al. [115]
SHRSP/IzmLOX-1 (lectin-like oxidized low-density lipoprotein receptor-1, Olr1)ZFN↓stroke susceptibility independently of BPLiang et al. [117]
SS/JrHsdMcwiRenin (Ren)ZFN↓BP, abnormal kidney morphologyMoreno et al. [120]
Rag1 (recombination activating 1, Rag1)ZFN↓BP, ↓renal injuryMattson et al. [121]
ROMK (renal outer medullary potassium channel, Kcnj1)ZFN↓BPZhou et al. [122]
Plekha7 (pleckstrin homology domain containing family A member 7, Plekha7)ZFN↓BP, ↓renal injury, ↓cardiac fibrosisEndres et al. [123]
HV1 (voltage-gated H+ channel, Hvcn1)ZFN↓BP, ↓renal injury, ↓oxidative stressJin et al. [124]
CD247 (Cd247)ZFN↓BP, ↓CD3+ T cells, ↓renal injuryRudemiller et al. [125]
BNP (B-type natriuretic peptide, Nppb)ZFN↑BP, ↑cardiac hypertrophy and fibrosis, ↑renal injuryHolditch et al. [126]
Nr2f2 (nuclear receptor subfamily 2 group F member 2, Nr2f2)ZFN↓BP, ↑left ventricular/vascular functions, ↑urinary proteinKumarasamy et al. [127]
Adora2b (A2B adenosine receptor, Adora2b)ZFN↑BP, ↑body weight, ↓glucose clearanceNayak et al. [128]
Nox4 (NADPH oxidase 4, Nox4)ZFN↓BP, ↓renal injury, ↓oxidative stressCowley et al. [129]
Rffl-lnc1 (a novel long-noncoding RNA)CRISPR-Cas9↑BP, shorter QT intervalsCheng et al. [130]
Resp18 (regulated endocrine-specific protein 18, Resp18)ZFN↑BP, ↑renal injury, ↓survival timeKumarasamy et al. [131]
Gper1 (G protein-coupled estrogen receptor 1, Gper1)CRISPR-Cas9↓BP, ↑vascular relaxation, ↓microbiotal dysbiosisWaghulde et al. [132]
p67phox (neutrophil cytosolic factor 2, Ncf2)ZFN↓BP, ↓renal injury, ↓renal immune cell infiltrationAbais-Battad et al. [133]

4. Conclusions

Hypertension is a multifactorial disease; nevertheless, the majority of previous research has focused on monogenic effects under inducible hypertensive conditions such as Ang II infusion and DOCA-salt in mice. Recent advances in genome-editing techniques, however, have made it possible to create knock-out and knock-in animals more easily, efficiently, and rapidly in rats as well as in mice [140,141,142,143]. Accordingly, it is necessary to create knock-out and knock-in models with multiple mutations in different loci to mimic complex genetic backgrounds of hypertensive patients and to uncover how the genetic interactions cause hypertension. As in the case of Dahl SS [144], translation of the findings in the experimental model into human hypertension remains highly challenging. However, translational approaches to bridge the gap between humans and rodent models would be necessary for understanding genetic and molecular mechanisms of essential hypertension in the post-GWAS era. A goal of basic hypertension research using experimental models may reconstruct ‘genetically hypertensive mice/rats’ from normotensive strains, and vice versa.

Author Contributions

H.O.: conceptualized and drafted the manuscript; H.O. and T.N.: revised the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was partly supported by JSPS KAKENHI (Grant Number 21H03374) to T.N.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Zhou, B.; Carrillo-Larco, R.M.; Danaei, G.; Riley, L.M.; Paciorek, C.J.; Stevens, G.A.; Gregg, E.W.; Bennett, J.E.; Solomon, B.; Singleton, R.K.; et al. Worldwide trends in hypertension prevalence and progress in treatment and control from 1990 to 2019: A pooled analysis of 1201 population-representative studies with 104 million participants. Lancet 2021, 398, 957–980. [Google Scholar] [CrossRef]
  2. Mashimo, T. Gene targeting technologies in rats: Zinc finger nucleases, transcription activator-like effector nucleases, and clustered regularly interspaced short palindromic repeats. Dev. Growth Differ. 2014, 56, 46–52. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Kaneko, T.; Mashimo, T. Simple Genome Editing of Rodent Intact Embryos by Electroporation. PLoS ONE 2015, 10, e0142755. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Stegbauer, J.; Chen, D.; Herrera, M.; Sparks, M.A.; Yang, T.; Konigshausen, E.; Gurley, S.B.; Coffman, T.M. Resistance to hypertension mediated by intercalated cells of the collecting duct. JCI Insight 2017, 2, e92720. [Google Scholar] [CrossRef] [Green Version]
  5. Li, X.C.; Leite, A.P.O.; Zheng, X.; Zhao, C.; Chen, X.; Zhang, L.; Zhou, X.; Rubera, I.; Tauc, M.; Zhuo, J.L. Proximal Tubule-Specific Deletion of Angiotensin II Type 1a Receptors in the Kidney Attenuates Circulating and Intratubular Angiotensin II-Induced Hypertension in PT-Agtr1a−/− Mice. Hypertension 2021, 77, 1285–1298. [Google Scholar] [CrossRef]
  6. Cui, T.; Nakagami, H.; Iwai, M.; Takeda, Y.; Shiuchi, T.; Tamura, K.; Daviet, L.; Horiuchi, M. ATRAP, novel AT1 receptor associated protein, enhances internalization of AT1 receptor and inhibits vascular smooth muscle cell growth. Biochem. Biophys. Res. Commun. 2000, 279, 938–941. [Google Scholar] [CrossRef]
  7. Tsurumi, Y.; Tamura, K.; Tanaka, Y.; Koide, Y.; Sakai, M.; Yabana, M.; Noda, Y.; Hashimoto, T.; Kihara, M.; Hirawa, N.; et al. Interacting molecule of AT1 receptor, ATRAP, is colocalized with AT1 receptor in the mouse renal tubules. Kidney Int. 2006, 69, 488–494. [Google Scholar] [CrossRef] [Green Version]
  8. Wakui, H.; Tamura, K.; Masuda, S.-I.; Tsurumi-Ikeya, Y.; Fujita, M.; Maeda, A.; Ohsawa, M.; Azushima, K.; Uneda, K.; Matsuda, M.; et al. Enhanced Angiotensin Receptor-Associated Protein in Renal Tubule Suppresses Angiotensin-Dependent Hypertension. Hypertension 2013, 61, 1203–1210. [Google Scholar] [CrossRef] [Green Version]
  9. Ohsawa, M.; Tamura, K.; Wakui, H.; Maeda, A.; Dejima, T.; Kanaoka, T.; Azushima, K.; Uneda, K.; Tsurumi-Ikeya, Y.; Kobayashi, R.; et al. Deletion of the angiotensin II type 1 receptor–associated protein enhances renal sodium reabsorption and exacerbates angiotensin II–mediated hypertension. Kidney Int. 2014, 86, 570–581. [Google Scholar] [CrossRef] [Green Version]
  10. Kobayashi, R.; Wakui, H.; Azushima, K.; Uneda, K.; Haku, S.; Ohki, K.; Haruhara, K.; Kinguchi, S.; Matsuda, M.; Ohsawa, M.; et al. An angiotensin II type 1 receptor binding molecule has a critical role in hypertension in a chronic kidney disease model. Kidney Int. 2017, 91, 1115–1125. [Google Scholar] [CrossRef]
  11. Kinguchi, S.; Wakui, H.; Azushima, K.; Haruhara, K.; Koguchi, T.; Ohki, K.; Uneda, K.; Matsuda, M.; Haku, S.; Yamaji, T.; et al. Effects of ATRAP in Renal Proximal Tubules on Angiotensin-Dependent Hypertension. J. Am. Heart Assoc. 2019, 8, e012395. [Google Scholar] [CrossRef]
  12. Flister, M.J.; Tsaih, S.W.; O’Meara, C.C.; Endres, B.; Hoffman, M.J.; Geurts, A.M.; Dwinell, M.R.; Lazar, J.; Jacob, H.J.; Moreno, C. Identifying multiple causative genes at a single GWAS locus. Genome Res. 2013, 23, 1996–2002. [Google Scholar] [CrossRef] [Green Version]
  13. Nguyen, G.; Delarue, F.; Burcklé, C.; Bouzhir, L.; Giller, T.; Sraer, J.D. Pivotal role of the renin/prorenin receptor in angiotensin II production and cellular responses to renin. J. Clin. Investig. 2002, 109, 1417–1427. [Google Scholar] [CrossRef]
  14. Ramkumar, N.; Kohan, D.E. The nephron (pro)renin receptor: Function and significance. Am. J. Physiol. Renal Physiol. 2016, 311, F1145–F1148. [Google Scholar] [CrossRef] [Green Version]
  15. Ramkumar, N.; Stuart, D.; Mironova, E.; Bugay, V.; Wang, S.; Abraham, N.; Ichihara, A.; Stockand, J.D.; Kohan, D.E. Renal tubular epithelial cell prorenin receptor regulates blood pressure and sodium transport. Am. J. Physiol. Renal Physiol. 2016, 311, F186–F194. [Google Scholar] [CrossRef] [Green Version]
  16. Peng, K.; Lu, X.; Wang, F.; Nau, A.; Chen, R.; Zhou, S.F.; Yang, T. Collecting duct (pro)renin receptor targets ENaC to mediate angiotensin II-induced hypertension. Am. J. Physiol. Renal Physiol. 2017, 312, F245–F253. [Google Scholar] [CrossRef]
  17. Prieto, M.C.; Reverte, V.; Mamenko, M.; Kuczeriszka, M.; Veiras, L.C.; Rosales, C.B.; McLellan, M.; Gentile, O.; Jensen, V.B.; Ichihara, A.; et al. Collecting duct prorenin receptor knockout reduces renal function, increases sodium excretion, and mitigates renal responses in ANG II-induced hypertensive mice. Am. J. Physiol. Renal Physiol. 2017, 313, F1243–F1253. [Google Scholar] [CrossRef] [Green Version]
  18. Mendoza-Torres, E.; Oyarzún, A.; Mondaca-Ruff, D.; Azocar, A.; Castro, P.F.; Jalil, J.E.; Chiong, M.; Lavandero, S.; Ocaranza, M.P. ACE2 and vasoactive peptides: Novel players in cardiovascular/renal remodeling and hypertension. Ther. Adv. Cardiovasc. Dis. 2015, 9, 217–237. [Google Scholar] [CrossRef]
  19. Ni, J.; Yang, F.; Huang, X.R.; Meng, J.; Chen, J.; Bader, M.; Penninger, J.M.; Fung, E.; Yu, X.Q.; Lan, H.Y. Dual deficiency of angiotensin-converting enzyme-2 and Mas receptor enhances angiotensin II-induced hypertension and hypertensive nephropathy. J. Cell. Mol. Med. 2020, 24, 13093–13103. [Google Scholar] [CrossRef]
  20. Ushiogi, Y.; Takabatake, T.; Häberle, D.A. Blood pressure and tubuloglomerular feedback mechanism in chronically salt-loaded spontaneously hypertensive rats. Kidney Int. 1991, 39, 1184–1192. [Google Scholar] [CrossRef] [Green Version]
  21. Lu, Y.; Wei, J.; Stec, D.E.; Roman, R.J.; Ge, Y.; Cheng, L.; Liu, E.Y.; Zhang, J.; Hansen, P.B.L.; Fan, F.; et al. Macula Densa Nitric Oxide Synthase 1β Protects against Salt-Sensitive Hypertension. J. Am. Soc. Nephrol. 2016, 27, 2346–2356. [Google Scholar] [CrossRef] [Green Version]
  22. Zhang, J.; Qu, L.; Wei, J.; Jiang, S.; Xu, L.; Wang, L.; Cheng, F.; Jiang, K.; Buggs, J.; Liu, R. A new mechanism for the sex differences in angiotensin II-induced hypertension: The role of macula densa NOS1beta-mediated tubuloglomerular feedback. Am. J. Physiol. Renal Physiol. 2020, 319, F908–F919. [Google Scholar] [CrossRef]
  23. Hyndman, K.A.; Boesen, E.I.; Elmarakby, A.A.A.; Brands, M.W.; Huang, P.; Kohan, D.E.; Pollock, D.M.; Pollock, J.S. Renal collecting duct NOS1 maintains fluid-electrolyte homeostasis and blood pressure. Hypertension 2013, 62, 91–98. [Google Scholar] [CrossRef] [Green Version]
  24. Gao, Y.; Stuart, D.; Takahishi, T.; Kohan, D.E. Nephron-Specific Disruption of Nitric Oxide Synthase 3 Causes Hypertension and Impaired Salt Excretion. J. Am. Heart Assoc. 2018, 7, e009236. [Google Scholar] [CrossRef] [Green Version]
  25. Hyndman, K.A.; Bugaj, V.; Mironova, E.; Stockand, J.D.; Pollock, J.S. NOS1-dependent negative feedback regulation of the epithelial sodium channel in the collecting duct. Am. J. Physiol. Renal Physiol. 2015, 308, F244–F251. [Google Scholar] [CrossRef] [Green Version]
  26. Raikwar, N.S.; Thomas, C.P. Nedd4-2 isoforms ubiquitinate individual epithelial sodium channel subunits and reduce surface expression and function of the epithelial sodium channel. Am. J. Physiol. Renal Physiol. 2008, 294, F1157–F1165. [Google Scholar] [CrossRef]
  27. Ronzaud, C.; Loffing-Cueni, D.; Hausel, P.; Debonneville, A.; Malsure, S.R.; Fowler-Jaeger, N.; Boase, N.A.; Perrier, R.; Maillard, M.; Yang, B.; et al. Renal tubular NEDD4-2 deficiency causes NCC-mediated salt-dependent hypertension. J. Clin. Investig. 2013, 123, 657–665. [Google Scholar] [CrossRef] [Green Version]
  28. Furusho, T.; Uchida, S.; Sohara, E. The WNK signaling pathway and salt-sensitive hypertension. Hypertens. Res. 2020, 43, 733–743. [Google Scholar] [CrossRef]
  29. Sohara, E.; Uchida, S. Kelch-like 3/Cullin 3 ubiquitin ligase complex and WNK signaling in salt-sensitive hypertension and electrolyte disorder. Nephrol. Dial. Transplant. 2016, 31, 1417–1424. [Google Scholar] [CrossRef] [Green Version]
  30. Liu, Z.; Xie, J.; Wu, T.; Truong, T.; Auchus, R.J.; Huang, C.-L. Downregulation of NCC and NKCC2 cotransporters by kidney-specific WNK1 revealed by gene disruption and transgenic mouse models. Hum. Mol. Genet. 2011, 20, 855–866. [Google Scholar] [CrossRef] [Green Version]
  31. Mu, S.; Shimosawa, T.; Ogura, S.; Wang, H.; Uetake, Y.; Kawakami-Mori, F.; Marumo, T.; Yatomi, Y.; Geller, D.S.; Tanaka, H.; et al. Epigenetic modulation of the renal β-adrenergic–WNK4 pathway in salt-sensitive hypertension. Nat. Med. 2011, 17, 573–580. [Google Scholar] [CrossRef] [PubMed]
  32. Pan, X.; Shao, Y.; Wu, F.; Wang, Y.; Xiong, R.; Zheng, J.; Tian, H.; Wang, B.; Wang, Y.; Zhang, Y.; et al. FGF21 Prevents Angiotensin II-Induced Hypertension and Vascular Dysfunction by Activation of ACE2/Angiotensin-(1-7) Axis in Mice. Cell Metab. 2018, 27, 1323–1337.e1325. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Crislip, G.R.; Douma, L.G.; Masten, S.H.; Cheng, K.Y.; Lynch, I.J.; Johnston, J.G.; Barral, D.; Glasford, K.B.; Holzworth, M.R.; Verlander, J.W.; et al. Differences in renal BMAL1 contribution to Na+ homeostasis and blood pressure control in male and female mice. Am. J. Physiol. Renal Physiol. 2020, 318, F1463–F1477. [Google Scholar] [CrossRef] [PubMed]
  34. Douma, L.G.; Costello, H.M.; Crislip, G.R.; Cheng, K.Y.; Lynch, I.J.; Juffre, A.; Barral, D.; Masten, S.; Roig, E.; Beguiristain, K.; et al. Kidney-specific KO of the circadian clock protein PER1 alters renal Na+ handling, aldosterone levels, and kidney/adrenal gene expression. Am. J. Physiol. Renal Physiol. 2022, 322, F449–F459. [Google Scholar] [CrossRef] [PubMed]
  35. Hiramatsu, A.; Izumi, Y.; Eguchi, K.; Matsuo, N.; Deng, Q.; Inoue, H.; Nakayama, Y.; Nonoguchi, H.; Aramburu, J.; Lopez-Rodriguez, C.; et al. Salt-Sensitive Hypertension of the Renal Tubular Cell-Specific NFAT5 (Nuclear Factor of Activated T-Cells 5) Knockout Mice. Hypertension 2021, 78, 1335–1346. [Google Scholar] [CrossRef] [PubMed]
  36. Ueda, K.; Nishimoto, M.; Hirohama, D.; Ayuzawa, N.; Kawarazaki, W.; Watanabe, A.; Shimosawa, T.; Loffing, J.; Zhang, M.Z.; Marumo, T.; et al. Renal Dysfunction Induced by Kidney-Specific Gene Deletion of Hsd11b2 as a Primary Cause of Salt-Dependent Hypertension. Hypertension 2017, 70, 111–118. [Google Scholar] [CrossRef] [Green Version]
  37. Shao, S.; Li, X.D.; Lu, Y.Y.; Li, S.J.; Chen, X.H.; Zhou, H.D.; He, S.; Guo, Y.T.; Lu, X.; Gao, P.J.; et al. Renal Natriuretic Peptide Receptor-C Deficiency Attenuates NaCl Cotransporter Activity in Angiotensin II-Induced Hypertension. Hypertension 2021, 77, 868–881. [Google Scholar] [CrossRef]
  38. Groneberg, D.; KöNig, P.; Wirth, A.; Offermanns, S.; Koesling, D.; Friebe, A. Smooth Muscle–Specific Deletion of Nitric Oxide–Sensitive Guanylyl Cyclase Is Sufficient to Induce Hypertension in Mice. Circulation 2010, 121, 401–409. [Google Scholar] [CrossRef] [Green Version]
  39. Kuhn, M. Molecular Physiology of Membrane Guanylyl Cyclase Receptors. Physiol. Rev. 2016, 96, 751–804. [Google Scholar] [CrossRef]
  40. Moyes, A.J.; Hobbs, A.J. C-type Natriuretic Peptide: A Multifaceted Paracrine Regulator in the Heart and Vasculature. Int. J. Mol. Sci. 2019, 20, 2281. [Google Scholar] [CrossRef] [Green Version]
  41. Nakao, K.; Kuwahara, K.; Nishikimi, T.; Nakagawa, Y.; Kinoshita, H.; Minami, T.; Kuwabara, Y.; Yamada, C.; Yamada, Y.; Tokudome, T.; et al. Endothelium-Derived C-Type Natriuretic Peptide Contributes to Blood Pressure Regulation by Maintaining Endothelial Integrity. Hypertension 2017, 69, 286–296. [Google Scholar] [CrossRef]
  42. Špiranec, K.; Chen, W.; Werner, F.; Nikolaev, V.O.; Naruke, T.; Koch, F.; Werner, A.; Eder-Negrin, P.; Diéguez-Hurtado, R.; Adams, R.H.; et al. Endothelial C-Type Natriuretic Peptide Acts on Pericytes to Regulate Microcirculatory Flow and Blood Pressure. Circulation 2018, 138, 494–508. [Google Scholar] [CrossRef]
  43. Tokudome, T.; Otani, K.; Mao, Y.; Jensen, L.J.; Arai, Y.; Miyazaki, T.; Sonobe, T.; Pearson, J.T.; Osaki, T.; Minamino, N.; et al. Endothelial Natriuretic Peptide Receptor 1 Play Crucial Role for Acute and Chronic Blood Pressure Regulation by Atrial Natriuretic Peptide. Hypertension 2022, 79, 1409–1422. [Google Scholar] [CrossRef]
  44. Hooper, R.; Samakai, E.; Kedra, J.; Soboloff, J. Multifaceted roles of STIM proteins. Pflügers Arch.-Eur. J. Physiol. 2013, 465, 1383–1396. [Google Scholar] [CrossRef] [Green Version]
  45. Kassan, M.; Ait-Aissa, K.; Radwan, E.; Mali, V.; Haddox, S.; Gabani, M.; Zhang, W.; Belmadani, S.; Irani, K.; Trebak, M.; et al. Essential Role of Smooth Muscle STIM1 in Hypertension and Cardiovascular Dysfunction. Arterioscler. Thromb. Vasc. Biol. 2016, 36, 1900–1909. [Google Scholar] [CrossRef] [Green Version]
  46. Nishimoto, M.; Mizuno, R.; Fujita, T.; Isshiki, M. Stromal interaction molecule 1 modulates blood pressure via NO production in vascular endothelial cells. Hypertens. Res. 2018, 41, 506–514. [Google Scholar] [CrossRef]
  47. Ferdaus, M.Z.; Xiao, B.; Ohara, H.; Nemoto, K.; Harada, Y.; Saar, K.; Hübner, N.; Isomura, M.; Nabika, T. Identification of Stim1 as a candidate gene for exaggerated sympathetic response to stress in the stroke-prone spontaneously hypertensive rat. PLoS ONE 2014, 9, e95091. [Google Scholar] [CrossRef] [Green Version]
  48. Ohara, H.; Nabika, T. A nonsense mutation of Stim1 identified in stroke-prone spontaneously hypertensive rats decreased the store-operated calcium entry in astrocytes. Biochem. Biophys. Res. Commun. 2016, 476, 406–411. [Google Scholar] [CrossRef]
  49. Odongoo, B.; Ohara, H.; Ngarashi, D.; Kaneko, T.; Kunihiro, Y.; Mashimo, T.; Nabika, T. Pathophysiological significance of Stim1 mutation in sympathetic response to stress and cardiovascular phenotypes in SHRSP/Izm: In vivo evaluation by creation of a novel gene knock-in rat using CRISPR/Cas9. Clin. Exp. Hypertens. 2021, 43, 34–41. [Google Scholar] [CrossRef]
  50. Fallah, H.P.; Ahuja, E.; Lin, H.; Qi, J.; He, Q.; Gao, S.; An, H.; Zhang, J.; Xie, Y.; Liang, D. A Review on the Role of TRP Channels and Their Potential as Drug Targets_An Insight Into the TRP Channel Drug Discovery Methodologies. Front. Pharmacol. 2022, 13, 914499. [Google Scholar] [CrossRef]
  51. Iring, A.; Jin, Y.-J.; Albarrán-Juárez, J.; Siragusa, M.; Wang, S.; Dancs, P.T.; Nakayama, A.; Tonack, S.; Chen, M.; Künne, C.; et al. Shear stress–induced endothelial adrenomedullin signaling regulates vascular tone and blood pressure. J. Clin. Investig. 2019, 129, 2775–2791. [Google Scholar] [CrossRef] [Green Version]
  52. Kita, T.; Kitamura, K. Translational studies of adrenomedullin and related peptides regarding cardiovascular diseases. Hypertens. Res. 2022, 45, 389–400. [Google Scholar] [CrossRef]
  53. Smyth, E.M.; Grosser, T.; Wang, M.; Yu, Y.; FitzGerald, G.A. Prostanoids in health and disease. J. Lipid Res. 2009, 50, S423–S428. [Google Scholar] [CrossRef] [Green Version]
  54. Yang, T.; Du, Y. Distinct roles of central and peripheral prostaglandin E2 and EP subtypes in blood pressure regulation. Am. J. Hypertens. 2012, 25, 1042–1049. [Google Scholar] [CrossRef] [Green Version]
  55. Bryson, T.D.; Harding, P. Prostaglandin E2 EP receptors in cardiovascular disease: An update. Biochem. Pharmacol. 2022, 195, 114858. [Google Scholar] [CrossRef]
  56. Xu, H.; Fang, B.; Du, S.; Wang, S.; Li, Q.; Jia, X.; Bao, C.; Ye, L.; Sui, X.; Qian, L.; et al. Endothelial cell prostaglandin E2 receptor EP4 is essential for blood pressure homeostasis. JCI Insight 2020, 5, e138505. [Google Scholar] [CrossRef]
  57. Caillon, A.; Paradis, P.; Schiffrin, E.L. Role of immune cells in hypertension. Br. J. Pharmacol 2019, 176, 1818–1828. [Google Scholar] [CrossRef]
  58. Lu, X.; Crowley, S.D. The Immune System in Hypertension: A Lost Shaker of Salt 2021 Lewis K. Dahl Memorial Lecture. Hypertension 2022, 79, 1339–1347. [Google Scholar] [CrossRef]
  59. Rizzoni, D.; De Ciuceis, C.; Szczepaniak, P.; Paradis, P.; Schiffrin, E.L.; Guzik, T.J. Immune System and Microvascular Remodeling in Humans. Hypertension 2022, 79, 691–705. [Google Scholar] [CrossRef]
  60. Shen, Y.; Shen, Z.; Luo, S.; Guo, W.; Zhu, Y.Z. The Cardioprotective Effects of Hydrogen Sulfide in Heart Diseases: From Molecular Mechanisms to Therapeutic Potential. Oxid. Med. Cell. Longev. 2015, 2015, 925167. [Google Scholar] [CrossRef]
  61. Yang, G.; Wu, L.; Jiang, B.; Yang, W.; Qi, J.; Cao, K.; Meng, Q.; Mustafa, A.K.; Mu, W.; Zhang, S.; et al. H2S as a physiologic vasorelaxant: Hypertension in mice with deletion of cystathionine gamma-lyase. Science 2008, 322, 587–590. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Cui, C.; Fan, J.; Zeng, Q.; Cai, J.; Chen, Y.; Chen, Z.; Wang, W.; Li, S.Y.; Cui, Q.; Yang, J.; et al. CD4+ T-Cell Endogenous Cystathionine gamma Lyase-Hydrogen Sulfide Attenuates Hypertension by Sulfhydrating Liver Kinase B1 to Promote T Regulatory Cell Differentiation and Proliferation. Circulation 2020, 142, 1752–1769. [Google Scholar] [CrossRef] [PubMed]
  63. Sun, X.N.; Li, C.; Liu, Y.; Du, L.J.; Zeng, M.R.; Zheng, X.J.; Zhang, W.C.; Liu, Y.; Zhu, M.; Kong, D.; et al. T-Cell Mineralocorticoid Receptor Controls Blood Pressure by Regulating Interferon-Gamma. Circ. Res. 2017, 120, 1584–1597. [Google Scholar] [CrossRef] [PubMed]
  64. Stepicheva, N.A.; Song, J.L. Function and regulation of microRNA-31 in development and disease. Mol. Reprod. Dev. 2016, 83, 654–674. [Google Scholar] [CrossRef] [PubMed]
  65. Li, X.; Cai, W.; Xi, W.; Sun, W.; Shen, W.; Wei, T.; Chen, X.; Sun, L.; Zhou, H.; Sun, Y.; et al. MicroRNA-31 Regulates Immunosuppression in Ang II (Angiotensin II)-induced Hypertension by Targeting Ppp6C (Protein Phosphatase 6c). Hypertension 2019, 73, e14–e24. [Google Scholar] [CrossRef] [PubMed]
  66. Crowley, S.D.; Rudemiller, N.P. Immunologic Effects of the Renin-Angiotensin System. J. Am. Soc. Nephrol. 2017, 28, 1350–1361. [Google Scholar] [CrossRef] [Green Version]
  67. Zhang, J.-D.; Patel, M.B.; Song, Y.-S.; Griffiths, R.; Burchette, J.; Ruiz, P.; Sparks, M.A.; Yan, M.; Howell, D.N.; Gomez, J.A.; et al. A Novel Role for Type 1 Angiotensin Receptors on T Lymphocytes to Limit Target Organ Damage in Hypertension. Circ. Res. 2012, 110, 1604–1617. [Google Scholar] [CrossRef] [Green Version]
  68. Zhang, J.D.; Patel, M.B.; Griffiths, R.; Dolber, P.C.; Ruiz, P.; Sparks, M.A.; Stegbauer, J.; Jin, H.; Gomez, J.A.; Buckley, A.F.; et al. Type 1 angiotensin receptors on macrophages ameliorate IL-1 receptor-mediated kidney fibrosis. J. Clin. Investig. 2014, 124, 2198–2203. [Google Scholar] [CrossRef] [Green Version]
  69. Lu, X.; Zhang, J.; Wen, Y.; Ren, J.; Griffiths, R.; Rudemiller, N.P.; Ide, S.; Souma, T.; Crowley, S.D. Type 1 Angiotensin Receptors on CD11c-Expressing Cells Protect Against Hypertension by Regulating Dendritic Cell-Mediated T Cell Activation. Hypertension 2022, 79, 1227–1236. [Google Scholar] [CrossRef]
  70. Sag, C.M.; Schnelle, M.; Zhang, J.; Murdoch, C.E.; Kossmann, S.; Protti, A.; Santos, C.X.C.; Sawyer, G.; Zhang, X.; Mongue-Din, H.; et al. Distinct Regulatory Effects of Myeloid Cell and Endothelial Cell NAPDH Oxidase 2 on Blood Pressure. Circulation 2017, 135, 2163–2177. [Google Scholar] [CrossRef] [Green Version]
  71. Wen, Y.; Rudemiller, N.P.; Zhang, J.; Lu, X.; Ren, J.; Privratsky, J.R.; Griffiths, R.; Zhang, J.J.; Hammer, G.E.; Crowley, S.D. C-C Motif Chemokine Receptor 7 Exacerbates Hypertension through Effects on T Lymphocyte Trafficking. Hypertension 2020, 75, 869–876. [Google Scholar] [CrossRef]
  72. Singh, M.V.; Cicha, M.Z.; Nunez, S.; Meyerholz, D.K.; Chapleau, M.W.; Abboud, F.M. Angiotensin II-induced hypertension and cardiac hypertrophy are differentially mediated by TLR3- and TLR4-dependent pathways. Am. J. Physiol. Heart Circ. Physiol. 2019, 316, H1027–H1038. [Google Scholar] [CrossRef]
  73. Perrotta, M.; Lori, A.; Carnevale, L.; Fardella, S.; Cifelli, G.; Iacobucci, R.; Mastroiacovo, F.; Iodice, D.; Pallante, F.; Storto, M.; et al. Deoxycorticosterone acetate-salt hypertension activates placental growth factor in the spleen to couple sympathetic drive and immune system activation. Cardiovasc. Res. 2018, 114, 456–467. [Google Scholar] [CrossRef]
  74. Chen, X.H.; Ruan, C.C.; Ge, Q.; Ma, Y.; Xu, J.Z.; Zhang, Z.B.; Lin, J.R.; Chen, D.R.; Zhu, D.L.; Gao, P.J. Deficiency of Complement C3a and C5a Receptors Prevents Angiotensin II-Induced Hypertension via Regulatory T Cells. Circ. Res. 2018, 122, 970–983. [Google Scholar] [CrossRef]
  75. Caillon, A.; Mian, M.O.R.; Fraulob-Aquino, J.C.; Huo, K.G.; Barhoumi, T.; Ouerd, S.; Sinnaeve, P.R.; Paradis, P.; Schiffrin, E.L. gammadelta T Cells Mediate Angiotensin II-Induced Hypertension and Vascular Injury. Circulation 2017, 135, 2155–2162. [Google Scholar] [CrossRef]
  76. Zhang, J.; Rudemiller, N.P.; Patel, M.B.; Karlovich, N.S.; Wu, M.; McDonough, A.A.; Griffiths, R.; Sparks, M.A.; Jeffs, A.D.; Crowley, S.D. Interleukin-1 Receptor Activation Potentiates Salt Reabsorption in Angiotensin II-Induced Hypertension via the NKCC2 Co-transporter in the Nephron. Cell Metab. 2016, 23, 360–368. [Google Scholar] [CrossRef] [Green Version]
  77. Van Beusecum, J.P.; Moreno, H.; Harrison, D.G. Innate immunity and clinical hypertension. J. Hum. Hypertens. 2021, 36, 503–509. [Google Scholar] [CrossRef]
  78. Su, C.; Xue, J.; Ye, C.; Chen, A. Role of the central renin-angiotensin system in hypertension (Review). Int. J. Mol. Med. 2021, 47, 95. [Google Scholar] [CrossRef]
  79. Morimoto, S.; Cassell, M.D.; Beltz, T.G.; Johnson, A.K.; Davisson, R.L.; Sigmund, C.D. Elevated Blood Pressure in Transgenic Mice With Brain-Specific Expression of Human Angiotensinogen Driven by the Glial Fibrillary Acidic Protein Promoter. Circ. Res. 2001, 89, 365–372. [Google Scholar] [CrossRef] [Green Version]
  80. Morimoto, S.; Cassell, M.D.; Sigmund, C.D. Glia- and Neuron-specific Expression of the Renin-Angiotensin System in Brain Alters Blood Pressure, Water Intake, and Salt Preference. J. Biol. Chem. 2002, 277, 33235–33241. [Google Scholar] [CrossRef] [Green Version]
  81. Sherrod, M.; Davis, D.R.; Zhou, X.; Cassell, M.D.; Sigmund, C.D. Glial-specific ablation of angiotensinogen lowers arterial pressure in renin and angiotensinogen transgenic mice. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2005, 289, R1763–R1769. [Google Scholar] [CrossRef] [Green Version]
  82. Li, W.; Peng, H.; Mehaffey, E.P.; Kimball, C.D.; Grobe, J.L.; Van Gool, J.M.G.; Sullivan, M.N.; Earley, S.; Danser, A.H.J.; Ichihara, A.; et al. Neuron-Specific (Pro)renin Receptor Knockout Prevents the Development of Salt-Sensitive Hypertension. Hypertension 2014, 63, 316–323. [Google Scholar] [CrossRef] [Green Version]
  83. Nomura, K.; Hiyama, T.Y.; Sakuta, H.; Matsuda, T.; Lin, C.-H.; Kobayashi, K.; Kobayashi, K.; Kuwaki, T.; Takahashi, K.; Matsui, S.; et al. [Na+] Increases in Body Fluids Sensed by Central Nax Induce Sympathetically Mediated Blood Pressure Elevations via H+-Dependent Activation of ASIC1a. Neuron 2019, 101, 60–75.e66. [Google Scholar] [CrossRef] [Green Version]
  84. Evans, L.C.; Ivy, J.R.; Wyrwoll, C.; McNairn, J.A.; Menzies, R.I.; Christensen, T.H.; Al-Dujaili, E.A.; Kenyon, C.J.; Mullins, J.J.; Seckl, J.R.; et al. Conditional Deletion of Hsd11b2 in the Brain Causes Salt Appetite and Hypertension. Circulation 2016, 133, 1360–1370. [Google Scholar] [CrossRef]
  85. Trebak, F.; Li, W.; Feng, Y. Neuronal (pro)renin receptor regulates deoxycorticosterone-induced sodium intake. Physiological Genomics 2018, 50, 904–912. [Google Scholar] [CrossRef]
  86. Wu, C.H.; Mohammadmoradi, S.; Thompson, J.; Su, W.; Gong, M.; Nguyen, G.; Yiannikouris, F. Adipocyte (Pro)Renin-Receptor Deficiency Induces Lipodystrophy, Liver Steatosis and Increases Blood Pressure in Male Mice. Hypertension 2016, 68, 213–219. [Google Scholar] [CrossRef] [Green Version]
  87. Davies, L.A.; Hu, C.; Guagliardo, N.A.; Sen, N.; Chen, X.; Talley, E.M.; Carey, R.M.; Bayliss, D.A.; Barrett, P.Q. TASK channel deletion in mice causes primary hyperaldosteronism. Proc. Natl. Acad. Sci. USA 2008, 105, 2203–2208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Guagliardo, N.A.; Yao, J.; Stipes, E.J.; Cechova, S.; Le, T.H.; Bayliss, D.A.; Breault, D.T.; Barrett, P.Q. Adrenal Tissue-Specific Deletion of TASK Channels Causes Aldosterone-Driven Angiotensin II–Independent Hypertension. Hypertension 2019, 73, 407–414. [Google Scholar] [CrossRef] [PubMed]
  89. Mathar, I.; Vennekens, R.; Meissner, M.; Kees, F.; Van Der Mieren, G.; Camacho Londoño, J.E.; Uhl, S.; Voets, T.; Hummel, B.; Van Den Bergh, A.; et al. Increased catecholamine secretion contributes to hypertension in TRPM4-deficient mice. J. Clin. Investig. 2010, 120, 3267–3279. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Doi, M.; Takahashi, Y.; Komatsu, R.; Yamazaki, F.; Yamada, H.; Haraguchi, S.; Emoto, N.; Okuno, Y.; Tsujimoto, G.; Kanematsu, A.; et al. Salt-sensitive hypertension in circadian clock–deficient Cry-null mice involves dysregulated adrenal Hsd3b6. Nat. Med. 2010, 16, 67–74. [Google Scholar] [CrossRef]
  91. Wise, I.A.; Charchar, F.J. Epigenetic Modifications in Essential Hypertension. Int. J. Mol. Sci. 2016, 17, 451. [Google Scholar] [CrossRef] [Green Version]
  92. Arif, M.; Sadayappan, S.; Becker, R.C.; Martin, L.J.; Urbina, E.M. Epigenetic modification: A regulatory mechanism in essential hypertension. Hypertens. Res. 2019, 42, 1099–1113. [Google Scholar] [CrossRef]
  93. Chaudhary, M. Novel methylation mark and essential hypertension. J. Genet. Eng. Biotechnol. 2022, 20, 11. [Google Scholar] [CrossRef]
  94. Chakraborty, S.; Mandal, J.; Yang, T.; Cheng, X.; Yeo, J.Y.; McCarthy, C.G.; Wenceslau, C.F.; Koch, L.G.; Hill, J.W.; Vijay-Kumar, M.; et al. Metabolites and Hypertension: Insights into Hypertension as a Metabolic Disorder: 2019 Harriet Dustan Award. Hypertension 2020, 75, 1386–1396. [Google Scholar] [CrossRef]
  95. Louca, P.; Menni, C.; Padmanabhan, S. Genomic Determinants of Hypertension With a Focus on Metabolomics and the Gut Microbiome. Am. J. Hypertens. 2020, 33, 473–481. [Google Scholar] [CrossRef]
  96. Mishima, E.; Abe, T. Role of the microbiota in hypertension and antihypertensive drug metabolism. Hypertens. Res. 2022, 45, 246–253. [Google Scholar] [CrossRef]
  97. Johnson, A.K.; Xue, B. Central nervous system neuroplasticity and the sensitization of hypertension. Nat. Rev. Nephrol. 2018, 14, 750–766. [Google Scholar] [CrossRef]
  98. Mann, S.J. Neurogenic hypertension: Pathophysiology, diagnosis and management. Clin. Auton. Res. 2018, 28, 363–374. [Google Scholar] [CrossRef]
  99. Ahmari, N.; Hayward, L.F.; Zubcevic, J. The importance of bone marrow and the immune system in driving increases in blood pressure and sympathetic nerve activity in hypertension. Exp. Physiol. 2020, 105, 1815–1826. [Google Scholar] [CrossRef]
  100. Geurts, A.M.; Cost, G.J.; Freyvert, Y.; Zeitler, B.; Miller, J.C.; Choi, V.M.; Jenkins, S.S.; Wood, A.; Cui, X.; Meng, X.; et al. Knockout Rats via Embryo Microinjection of Zinc-Finger Nucleases. Science 2009, 325, 433. [Google Scholar] [CrossRef] [Green Version]
  101. Szpirer, C. Rat models of human diseases and related phenotypes: A systematic inventory of the causative genes. J. Biomed. Sci. 2020, 27, 84. [Google Scholar] [CrossRef]
  102. Sato, M.; Nakamura, S.; Inada, E.; Takabayashi, S. Recent Advances in the Production of Genome-Edited Rats. Int. J. Mol. Sci. 2022, 23, 2548. [Google Scholar] [CrossRef]
  103. Nabika, T.; Ohara, H.; Kato, N.; Isomura, M. The stroke-prone spontaneously hypertensive rat: Still a useful model for post-GWAS genetic studies? Hypertens. Res. 2012, 35, 477–484. [Google Scholar] [CrossRef]
  104. Schulz, A.; Kreutz, R. Mapping genetic determinants of kidney damage in rat models. Hypertens. Res. 2012, 35, 675–694. [Google Scholar] [CrossRef] [Green Version]
  105. Yang, Y.; Kimura-Ohba, S.; Thompson, J.; Rosenberg, G.A. Rodent Models of Vascular Cognitive Impairment. Transl. Stroke Res. 2016, 7, 407–414. [Google Scholar] [CrossRef]
  106. Doggrell, S.A.; Brown, L. Rat models of hypertension, cardiac hypertrophy and failure. Cardiovasc. Res. 1998, 39, 89–105. [Google Scholar] [CrossRef]
  107. Liška, F.; Mancini, M.; Krupková, M.; Chylíková, B.; Křenová, D.; Šeda, O.; Šilhavý, J.; Mlejnek, P.; Landa, V.; Zídek, V.; et al. Plzf as a candidate gene predisposing the spontaneously hypertensive rat to hypertension, left ventricular hypertrophy, and interstitial fibrosis. Am. J. Hypertens. 2014, 27, 99–106. [Google Scholar] [CrossRef] [Green Version]
  108. Liška, F.; Landa, V.; Zídek, V.; Mlejnek, P.; Šilhavý, J.; Šimáková, M.; Strnad, H.; Trnovská, J.; Škop, V.; Kazdová, L.; et al. Downregulation of Plzf Gene Ameliorates Metabolic and Cardiac Traits in the Spontaneously Hypertensive Rat. Hypertension 2017, 69, 1084–1091. [Google Scholar] [CrossRef] [PubMed]
  109. Lin, Z.-H.; Fukuda, N.; Jin, X.-Q.; Yao, E.-H.; Ueno, T.; Endo, M.; Saito, S.; Matsumoto, K.; Mugishima, H. Complement 3 Is Involved in the Synthetic Phenotype and Exaggerated Growth of Vascular Smooth Muscle Cells From Spontaneously Hypertensive Rats. Hypertension 2004, 44, 42–47. [Google Scholar] [CrossRef] [PubMed]
  110. Han, Y.; Fukuda, N.; Ueno, T.; Endo, M.; Ikeda, K.; Xueli, Z.; Matsumoto, T.; Soma, M.; Matsumoto, K. Role of Complement 3a in the Synthetic Phenotype and Angiotensin II-Production in Vascular Smooth Muscle Cells from Spontaneously Hypertensive Rats. Am. J. Hypertens. 2012, 25, 284–289. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Ikeda, K.; Fukuda, N.; Ueno, T.; Endo, M.; Kobayashi, N.; Soma, M.; Matsumoto, K. Role of complement 3a in the growth of mesangial cells from stroke-prone spontaneously hypertensive rats. Clin. Exp. Hypertens. 2014, 36, 58–63. [Google Scholar] [CrossRef]
  112. Negishi, E.; Fukuda, N.; Otsuki, T.; Katakawa, M.; Komatsu, K.; Chen, L.; Tanaka, S.; Kobayashi, H.; Hatanaka, Y.; Ueno, T.; et al. Involvement of complement 3 in the salt-sensitive hypertension by activation of renal renin-angiotensin system in spontaneously hypertensive rats. Am. J. Physiol.-Ren. Physiol. 2018, 315, F1747–F1758. [Google Scholar] [CrossRef]
  113. Rubattu, S.; Volpe, M.; Kreutz, R.; Ganten, U.; Ganten, D.; Lindpaintner, K. Chromosomal mapping of quantitative trait loci contributing to stroke in a rat model of complex human disease. Nat. Genet. 1996, 13, 429–434. [Google Scholar] [CrossRef]
  114. Rubattu, S.; Di Castro, S.; Schulz, H.; Geurts, A.M.; Cotugno, M.; Bianchi, F.; Maatz, H.; Hummel, O.; Falak, S.; Stanzione, R.; et al. Ndufc2 Gene Inhibition Is Associated with Mitochondrial Dysfunction and Increased Stroke Susceptibility in an Animal Model of Complex Human Disease. J. Am. Heart Assoc. 2016, 5, e002701. [Google Scholar] [CrossRef] [Green Version]
  115. Mahal, Z.; Fujikawa, K.; Matsuo, H.; Zahid, H.M.; Koike, M.; Misumi, M.; Kaneko, T.; Mashimo, T.; Ohara, H.; Nabika, T. Effects of the Prdx2 depletion on blood pressure and life span in spontaneously hypertensive rats. Hypertens. Res. 2019, 42, 610–617. [Google Scholar] [CrossRef]
  116. Kattoor, A.J.; Goel, A.; Mehta, J.L. LOX-1: Regulation, Signaling and Its Role in Atherosclerosis. Antioxidants 2019, 8, 218. [Google Scholar] [CrossRef] [Green Version]
  117. Liang, Y.-Q.; Kakino, A.; Matsuzaka, Y.; Mashimo, T.; Isono, M.; Akamatsu, T.; Shimizu, H.; Tajima, M.; Kaneko, T.; Li, L.; et al. LOX-1 (Lectin-Like Oxidized Low-Density Lipoprotein Receptor-1) Deletion Has Protective Effects on Stroke in the Genetic Background of Stroke-Prone Spontaneously Hypertensive Rat. Stroke 2020, 51, 1835–1843. [Google Scholar] [CrossRef]
  118. Xiao, B.; Harada, Y.; Kawakami, K.; Nabika, T. A 1.8-Mbp fragment on chromosome 1 affects sympathetic response to stress: Evaluation in reciprocal congenic strains between stroke-prone spontaneously hypertensive rat and Wistar-Kyoto rat. J. Hypertens. 2011, 29, 257–265. [Google Scholar] [CrossRef]
  119. Rapp, J.P.; Garrett, M.R. Will the real Dahl S rat please stand up? Am. J. Physiol. Renal Physiol. 2019, 317, F1231–F1240. [Google Scholar] [CrossRef]
  120. Moreno, C.; Hoffman, M.; Stodola, T.J.; Didier, D.N.; Lazar, J.; Geurts, A.M.; North, P.E.; Jacob, H.J.; Greene, A.S. Creation and characterization of a renin knockout rat. Hypertension 2011, 57, 614–619. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Mattson, D.L.; Lund, H.; Guo, C.; Rudemiller, N.; Geurts, A.M.; Jacob, H. Genetic mutation of recombination activating gene 1 in Dahl salt-sensitive rats attenuates hypertension and renal damage. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2013, 304, R407–R414. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Zhou, X.; Zhang, Z.; Shin, M.K.; Horwitz, S.B.; Levorse, J.M.; Zhu, L.; Sharif-Rodriguez, W.; Streltsov, D.Y.; Dajee, M.; Hernandez, M.; et al. Heterozygous disruption of renal outer medullary potassium channel in rats is associated with reduced blood pressure. Hypertension 2013, 62, 288–294. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Endres, B.T.; Priestley, J.R.; Palygin, O.; Flister, M.J.; Hoffman, M.J.; Weinberg, B.D.; Grzybowski, M.; Lombard, J.H.; Staruschenko, A.; Moreno, C.; et al. Mutation of Plekha7 attenuates salt-sensitive hypertension in the rat. Proc. Natl. Acad. Sci. USA 2014, 111, 12817–12822. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Jin, C.; Sun, J.; Stilphen, C.A.; Smith, S.M.; Ocasio, H.; Bermingham, B.; Darji, S.; Guha, A.; Patel, R.; Geurts, A.M.; et al. HV1 acts as a sodium sensor and promotes superoxide production in medullary thick ascending limb of Dahl salt-sensitive rats. Hypertension 2014, 64, 541–550. [Google Scholar] [CrossRef] [Green Version]
  125. Rudemiller, N.; Lund, H.; Jacob, H.J.; Geurts, A.A.M.; Mattson, D.L. CD247 modulates blood pressure by altering T-lymphocyte infiltration in the kidney. Hypertension 2014, 63, 559–564. [Google Scholar] [CrossRef] [Green Version]
  126. Holditch, S.J.; Schreiber, C.A.; Nini, R.; Tonne, J.M.; Peng, K.-W.; Geurts, A.; Jacob, H.J.; Burnett, J.C.; Cataliotti, A.; Ikeda, Y. B-Type Natriuretic Peptide Deletion Leads to Progressive Hypertension, Associated Organ Damage, and Reduced Survival. Hypertension 2015, 66, 199–210. [Google Scholar] [CrossRef] [Green Version]
  127. Kumarasamy, S.; Waghulde, H.; Gopalakrishnan, K.; Mell, B.; Morgan, E.; Joe, B. Mutation within the hinge region of the transcription factor Nr2f2 attenuates salt-sensitive hypertension. Nat. Commun. 2015, 6, 6252. [Google Scholar] [CrossRef] [Green Version]
  128. Nayak, S.; Khan, M.A.; Wan, T.C.; Pei, H.; Linden, J.; Dwinell, M.R.; Geurts, A.M.; Imig, J.D.; Auchampach, J.A. Characterization of Dahl salt-sensitive rats with genetic disruption of the A2B adenosine receptor gene: Implications for A2B adenosine receptor signaling during hypertension. Purinergic Signal. 2015, 11, 519–531. [Google Scholar] [CrossRef] [Green Version]
  129. Cowley, A.W., Jr.; Yang, C.; Zheleznova, N.N.; Staruschenko, A.; Kurth, T.; Rein, L.; Kumar, V.; Sadovnikov, K.; Dayton, A.; Hoffman, M.; et al. Evidence of the Importance of Nox4 in Production of Hypertension in Dahl Salt-Sensitive Rats. Hypertension 2016, 67, 440–450. [Google Scholar] [CrossRef] [Green Version]
  130. Cheng, X.; Waghulde, H.; Mell, B.; Morgan, E.E.; Pruett-Miller, S.M.; Joe, B. Positional cloning of quantitative trait nucleotides for blood pressure and cardiac QT-interval by targeted CRISPR/Cas9 editing of a novel long non-coding RNA. PLoS Genet. 2017, 13, e1006961. [Google Scholar] [CrossRef]
  131. Kumarasamy, S.; Waghulde, H.; Cheng, X.; Haller, S.T.; Mell, B.; Abhijith, B.; Ashraf, U.M.; Atari, E.; Joe, B. Targeted disruption of regulated endocrine-specific protein (Resp18) in Dahl SS/Mcw rats aggravates salt-induced hypertension and renal injury. Physiol. Genom. 2018, 50, 369–375. [Google Scholar] [CrossRef]
  132. Waghulde, H.; Cheng, X.; Galla, S.; Mell, B.; Cai, J.; Pruett-Miller, S.M.; Vazquez, G.; Patterson, A.; Vijay Kumar, M.; Joe, B. Attenuation of Microbiotal Dysbiosis and Hypertension in a CRISPR/Cas9 Gene Ablation Rat Model of GPER1. Hypertension 2018, 72, 1125–1132. [Google Scholar] [CrossRef]
  133. Abais-Battad, J.M.; Lund, H.; Dasinger, J.H.; Fehrenbach, D.J.; Cowley, A.W., Jr.; Mattson, D.L. NOX2-derived reactive oxygen species in immune cells exacerbates salt-sensitive hypertension. Free Radic. Biol. Med. 2020, 146, 333–339. [Google Scholar] [CrossRef]
  134. Pulimeno, P.; Bauer, C.; Stutz, J.; Citi, S. PLEKHA7 is an adherens junction protein with a tissue distribution and subcellular localization distinct from ZO-1 and E-cadherin. PLoS ONE 2010, 5, e12207. [Google Scholar] [CrossRef]
  135. Levy, D.; Ehret, G.B.; Rice, K.; Verwoert, G.C.; Launer, L.J.; Dehghan, A.; Glazer, N.L.; Morrison, A.C.; Johnson, A.D.; Aspelund, T.; et al. Genome-wide association study of blood pressure and hypertension. Nat. Genet. 2009, 41, 677–687. [Google Scholar] [CrossRef] [Green Version]
  136. Hong, K.W.; Jin, H.S.; Lim, J.E.; Kim, S.; Go, M.J.; Oh, B. Recapitulation of two genomewide association studies on blood pressure and essential hypertension in the Korean population. J. Hum. Genet. 2010, 55, 336–341. [Google Scholar] [CrossRef] [Green Version]
  137. Fox, E.R.; Young, J.H.; Li, Y.; Dreisbach, A.W.; Keating, B.J.; Musani, S.K.; Liu, K.; Morrison, A.C.; Ganesh, S.; Kutlar, A.; et al. Association of genetic variation with systolic and diastolic blood pressure among African Americans: The Candidate Gene Association Resource study. Hum. Mol. Genet. 2011, 20, 2273–2284. [Google Scholar] [CrossRef] [Green Version]
  138. Ho, J.E.; Levy, D.; Rose, L.; Johnson, A.D.; Ridker, P.M.; Chasman, D.I. Discovery and replication of novel blood pressure genetic loci in the Women’s Genome Health Study. J. Hypertens. 2011, 29, 62–69. [Google Scholar] [CrossRef] [Green Version]
  139. Lin, Y.; Lai, X.; Chen, B.; Xu, Y.; Huang, B.; Chen, Z.; Zhu, S.; Yao, J.; Jiang, Q.; Huang, H.; et al. Genetic variations in CYP17A1, CACNB2 and PLEKHA7 are associated with blood pressure and/or hypertension in She ethnic minority of China. Atherosclerosis 2011, 219, 709–714. [Google Scholar] [CrossRef]
  140. Kobayashi, T.; Namba, M.; Koyano, T.; Fukushima, M.; Sato, M.; Ohtsuka, M.; Matsuyama, M. Successful production of genome-edited rats by the rGONAD method. BMC Biotechnol. 2018, 18, 19. [Google Scholar] [CrossRef] [Green Version]
  141. Miyasaka, Y.; Uno, Y.; Yoshimi, K.; Kunihiro, Y.; Yoshimura, T.; Tanaka, T.; Ishikubo, H.; Hiraoka, Y.; Takemoto, N.; Tanaka, T.; et al. CLICK: One-step generation of conditional knockout mice. BMC Genom. 2018, 19, 318. [Google Scholar] [CrossRef] [Green Version]
  142. Ohtsuka, M.; Sato, M.; Miura, H.; Takabayashi, S.; Matsuyama, M.; Koyano, T.; Arifin, N.; Nakamura, S.; Wada, K.; Gurumurthy, C.B. i-GONAD: A robust method for in situ germline genome engineering using CRISPR nucleases. Genome Biol. 2018, 19, 25. [Google Scholar] [CrossRef] [Green Version]
  143. Morisaka, H.; Yoshimi, K.; Okuzaki, Y.; Gee, P.; Kunihiro, Y.; Sonpho, E.; Xu, H.; Sasakawa, N.; Naito, Y.; Nakada, S.; et al. CRISPR-Cas3 induces broad and unidirectional genome editing in human cells. Nat. Commun. 2019, 10, 5302. [Google Scholar] [CrossRef]
  144. Padmanabhan, S.; Joe, B. Towards Precision Medicine for Hypertension: A Review of Genomic, Epigenomic, and Microbiomic Effects on Blood Pressure in Experimental Rat Models and Humans. Physiol. Rev. 2017, 97, 1469–1528. [Google Scholar] [CrossRef]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ohara, H.; Nabika, T. Genetic Modifications to Alter Blood Pressure Level. Biomedicines 2022, 10, 1855. https://doi.org/10.3390/biomedicines10081855

AMA Style

Ohara H, Nabika T. Genetic Modifications to Alter Blood Pressure Level. Biomedicines. 2022; 10(8):1855. https://doi.org/10.3390/biomedicines10081855

Chicago/Turabian Style

Ohara, Hiroki, and Toru Nabika. 2022. "Genetic Modifications to Alter Blood Pressure Level" Biomedicines 10, no. 8: 1855. https://doi.org/10.3390/biomedicines10081855

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop