Next Article in Journal
High Prevalence of Antibiotic Resistance in Iranian Helicobacter pylori Isolates: Importance of Functional and Mutational Analysis of Resistance Genes and Virulence Genotyping
Previous Article in Journal
Potential Utility of Neurosonology in Paroxysmal Atrial Fibrillation Detection in Patients with Cryptogenic Stroke
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Exploring a New Natural Treating Agent for Primary Hypertension: Recent Findings and Forthcoming Perspectives

1
Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Hualien 97401, Taiwan
2
Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei 110, Taiwan
3
Department of Anatomy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
4
Camillian Saint Mary’s Hospital Luodong,160 Zhongzheng S. Rd. Luodong, Yilan 26546, Taiwan
5
Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
6
Center for Infectious Disease and Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
7
Department of Basic Medical Science, Center for Transitional Medicine, Xiamen Medical College, Xiamen 361023, China
*
Authors to whom correspondence should be addressed.
These two authors contribute equally.
J. Clin. Med. 2019, 8(11), 2003; https://doi.org/10.3390/jcm8112003
Submission received: 18 October 2019 / Revised: 14 November 2019 / Accepted: 14 November 2019 / Published: 16 November 2019
(This article belongs to the Section Epidemiology & Public Health)

Abstract

:
Primary hypertension describes abnormally-high systolic/diastolic blood pressure in a resting condition caused by various genetic or environmental risk factors. Remarkably, severe complications, such as ischemic cardiovascular disease, stroke, and chronic renal disease have led to primary hypertension becoming a huge burden for almost one-third of the total population. Medication is the major regimen for treating primary hypertension; however, recent medications may have adverse effects that attenuate energy levels. Hence, the search for new hypotensive agents from folk or traditional medicine may be fruitful in the discovery and development of new drugs. This review assembles recent findings for natural antihypertensive agents, extracts, or decoctions published in PubMed, and provides insights into the search for new hypotensive compounds based on blood-pressure regulating mechanisms, including the renin-angiotensin-aldosterone system and the sympathetic/adrenergic receptor/calcium channel system.

1. Introduction

Hypertension is a homeostatic blood pressure shift from normal (<120/80 mmHg) to abnormal levels (>140/90 mmHg) which is categorized into the following four stages: normal pressure (<120/80 mmHg), elevated pressure (120–139/80–89 mmHg), stage 1 hypertension (140–159/90–99 mmHg), and stage 2 hypertension (>160/100 mmHg) [1,2,3]. Only 5–10% of hypertensive patients can be diagnosed from certain causes e.g., aldosteronism, gestation, or renal artery stenosis (secondary hypertension); however, the other 90–95% of hypertensive patients have unknown or multiple reasons for their condition [4,5]. Despite uncertain causes, some predisposing factors for primary hypertension, such as genetic variation, obesity, improper eating habits including excessive sodium and alcohol consumption, tobacco and marijuana use, and physical unfitness, are summarized [6,7,8].
About 31.1% of adults were hypertensive worldwide in 2010, with 28.5% of them living in high-income countries [9]. Notably, 32.5% of adults (17.3% of total citizens) are in various stages of hypertension in China [10]. Furthermore, the overall mortality of high systolic blood pressure is 20,526/100,000 people, and the age-standardized disability-adjusted life year rate (DALY) is 242.5/100,000 people [11,12]. These harsh statistics remind us that hypertension is highly prevalent, and is a burden on society in terms of the associated health expenditure. Generally, hypertensive patients endure various symptoms that are difficult to self-assess, such as headaches, dizziness, shortness of breath, chest pain, palpitations, and nosebleeds. However, common complications of hypertension are more severe, e.g., stroke, ischemic cardiovascular disease (ICD), and chronic kidney disease (CKD) (see Table 1) [13,14,15]. Therefore, the treatment and medical care of hypertension is a critical issue for elevating the well-being of mankind.
The clinical foci of hypertension treatment comprise ameliorating blood pressure and preventing complications. Generally speaking, by using a blood pressure lowering approach, medication is the first choice, while the management life style gives constructive support in enhancing adherence [3,16]. Numerous agents, e.g., verapamil, amlodipine, and captopril, have been applied in the treatment of hypertension; the antihypertensive effects of these drugs have been well established [3,17]. Beyond medication, patients and clinicians propose a variety of supports from pharmaceuticals, society, family, and patient’s personal pharmacogenomics for managing hypertension. Social isolation, low income, depression, or psychological distress have been ascertained as causes of withdrawal during treatment [3,16,18,19]. Statistical analyses from the investigation of African American women, Latino migrants, seasonal farmworkers, Chinese people, and people from south-west Nigeria have illustrated that social supports could promote patient adherence to treatment via sharing information and assisting in bringing about life style changes [20,21,22,23]. Surprisingly, about 10.3% of hypertension patients develop hypertensive resistance during the treatment regimen, which may be typified by a lower response to medications [24]. Nevertheless, several adverse effects—e.g., hyponatremia [25], hyperkalemia [26], and edema [27]—of recent medications do not facilitate the medical compliance of patients [28]. Accordingly, finding new blood pressure lowering agents with medical benefits for hypertensive patients, and to promoting the progress of new drug discovery, are increasingly important. Expectedly, cumulative research evidence has illustrated that natural compounds or herbal medicines are believed to be equipotent with fewer adverse effects compared to synthetic drugs [29]. In this review, we summarize recent findings pertaining to hypertension remedies from natural products and research approaches according the causes and shedding mechanism of hypertension that could provide new insights into perspective studies for the research and discovery of natural antihypertensive agents.

2. Physiological Regulation Mechanism in Blood-Pressure

Basically, the body undertakes the regulation of blood-pressure via two strategies: total blood volume and blood flow. Total blood volume is mainly mediated by renin-angiotensin-aldosterone systems (RAAS), while blood flow is predominantly regulated by neurogenic hypertensive system and nitric oxide (NO) [30,31]. Consequently, current hypertensive medication also targets the aforementioned mechanisms (RAAS and neurogenic hypertensive system) to alleviate high blood pressure [1]. The following section will individually address each blood-pressure-controlling mechanism to summarize our present knowledge of the regulation of blood pressure, medications, and screening tools for natural compounds.

2.1. Renin-Angiotensin-Aldosterone System (RAAS)

2.1.1. Mechanical Action of RAAS in Blood-Pressure Controlling

RAAS is long-term regulator of blood pressure composed of renin (an aspartic protease produced by the juxtaglomerular apparatus of kidney), angiotensin converting enzyme (ACE, a zinc-dependent dicarboxyprotease secreted from the lungs), angiotensinogen (a hormone precursor), and aldosterone (mineralocorticoid hormone from adrenal cortex, the endpoint effector). Angiotensinogen is converted into angiotensin II (Ang II) because it is sequentially cleaved by renin and ACE; subsequently, Ang II stimulates the adrenal gland cortex to produce aldosterone [32]. Aldosterone raises blood-pressure via two phases: (i) potentiating Na+/K+-ATPase activity on sodium and water reabsorption of renal tubular cells; and (ii) consequently managing (i.e., increasing or decreasing) blood volume (see Figure 1A), as well as vascular smooth muscle cells for vasoconstriction [33,34]. To understand the cause of elevating blood pressure, the consequence of a decrease of sympathetic input and an increase in parasympathetic input to the heart is observed. Second, if the baroreceptors detect that blood pressure is too high, the cardio-regulatory center of the medulla will also decrease the sympathetic input to the blood vessels. This causes vasodilation instead of vasoconstriction, which decreases the total peripheral resistance and concurrently decreases blood pressure (see Figure 1B). In this section, we will not discuss the sympathetic nerve regulating system; rather, we will mainly focus on the regulation of the RAAS system in order to present a comprehensive review of potential targets for lowering blood pressure.

2.1.2. Conventional RAAS Modulators in Hypertensive Medication

According to the physiological mechanism of RAAS, several targets have been identified: ACE, the angiotensin II type-1 receptor (AGTR1), the aldosterone receptor, and Na+/K+-ATPase. ACE inhibitors (e.g., Captopril, Ramipril, and Fosinopril) and Ang II receptor blockers (e.g., Valsartan, Irbesartan, Candesartan, and Olmesartan) directly suppress Ang II and aldosterone production in the adrenal gland [35,36]. Aldosterone receptor inhibitors (also known as K+-sparing diuretics, e.g., Spironolactone, Pironolactone, and Eplerenone) allosterically inhibit the interactions of aldosterone with its receptor. Additionally, Na+/K+-ATPase blockers (thiazides such as benzylhydrochlorothiazide and indapamide) lead to reduced Na+/K+-ATPase activity and reduced water reabsorption [37]. Moreover, RAAS modulators can improve hypertension through directly diminishing the total blood volume. Hence, RAAS modulators play a major role in primary hypertension treatments [3]. However, RAAS modulators tend to preserve K+ and eliminate Na+, which would increase the risks of edema, hyperkalemia, and hyponatremia [25,26,37]. This adverse effect needs to be taken into account when using these modulators.

2.1.3. Methods for Screening RAAS Modulators

Numerous available models are used in the search for new RAAS modulators through in silico, in vitro, and in vivo approaches. Chemical induction, renal artery ligation, and genetic modification are common methods with which to generate the in vivo hypertensive model [38]. Of note, chemical induction is conducted by administrating 11-deoxycorticosterone acetate (DOCA) or Ang II, which results in transient hypertension [39,40]. Furthermore, transient hypertension transforms into persistent hypertension during chemical induction, which occurs when combined with a high-salt diet [40]. In general, the intravenous (i.v) or subcutaneous (s.c) administration of Ang II is a common model [41,42]. Ang II infusion could keep high levels of Ang II in serum; however, in experiments, animals needed to be anesthetized for more than 7 days [41,42]. Therefore, this technique is rarely used in drug screening but the risk of hypertensive complications, e.g., aneurysm, nephropathy, preeclampsia, and cognitive impairment [42,43,44,45]. Renal artery ligation is the most reported animal model for simulating reno-vascular hypertension, the most common cause of secondary hypertension [46]. Renal artery stenosis can be simulated by partial ligation on the left side or on both sides of the renal artery, leading to reno-vascular hypertension in animal models [47]. The genetic hypertension model is particular to spontaneous hypertension and includes over half of the hypertension population [38]. A well-known spontaneous hypertensive rodent model mainly used in the study of RAAS-induced hypertension is the spontaneous hypertensive rat (SHR) [48]. SHR is originally derived from inbreeding the Wistar–Kyoto (WKY) rats with hypertension, and is suitable for studying either hypertension or renal disease and left ventricular diastolic dysfunction [49,50]. Despite spontaneous hypertension, another primary cause of primary hypertension is obesity, which accounts for 60–75% of primary hypertension cases [51]. To study obesity-induced hypertension, the New Zealand Obese strain (NZO) is appropriate because of its spontaneously polygenic obesity and insulin resistance [52]. In diet-induced hypertension, a high-fat and high-salt diet could be alternatively applied to obtain the desired model [53]. A high-fat diet is another choice for obesity-induced hypertension mice model, and is usually coupled with type-2 diabetes [54]. When mice are fed with a high-salt diet for 4–8 weeks, glucocorticoid resistance could be found in animals, eventually resulting in hypercortisolemia, which is an appropriate model for the diuretic screening of hypernatremia, a cause of hypertension [55,56]. Currently, newly-bred, spontaneous Phase-1 hypertension (P1-HT) rats were successfully generated and used to confirm Plantago asiatica seed extract as a potentially antihypertensive herb when compared with SHR [57].
ACE inhibitors can also be tested by in silico to in vitro modeling. In in silico modeling, compound modules and ACE structural modules are employed to simulate ligand/receptor interactions. Higher molecular interaction strengths could demonstrate a greater possibility of being potent inhibitors [58]. By comparing interacting patterns, different ligand types might interact with different amino acid residues in ACE. For small molecules, Gln281, Lys511, and Tyr520 of ACE are essential for binding, forming a hydrogen bond with the ligand [59]. In the search of ACE peptide inhibitors, Val518, His383, His387, and His51, are the most important, forming electrostatic and hydrophobic interactions with peptide inhibitors and Zn2+ [60]. In the enzymatic approach, ACE activity could be determined by interacting with peptide derivatives that are composed of peptides and colorimetric indicators. The colorimetric assays for ACE are very sensitive, fast, and easy to use [61]. However, in vitro enzymatic conditions will not ever be equivalent to in vivo conditions, due to the highly complex composition of body fluids [62]. The foreseeable gap between enzymatic screening and in vivo or in vitro analyses needs more studies to shorten the distance.
Other RAAS targets, including AGTR1, aldosterone receptor, and Na+/K+-ATPase, could not be tested by colorimetric or chemical assay. Recently, cell membrane chromatography has used cell membranes as a stationary phase, allowing the molecules to interact with receptors on the cell membrane [63]. This technique could theoretically overcome the limitation of enzymatic assays for which this principle is not suitable for evaluations of ligand/receptor interactions [64]. Nonetheless, the interaction between cell membranes and ligand candidates could not distinguished whether they were activators or inhibitors. On the other hand, human kidney proximal tubule epithelial cells HK-2 have been identified as carriers of mineral corticoid receptor, angiotensin receptor type 2, and Na+/K+ ATPase expression [65,66,67]. Moreover, aldosterone is shown to be an apoptotic inducer in proximal tubule cells [68]. This evidence shows that HK-2 could be an ideal cellular platform for the screening of diuretics, be it Na+/K+ ATPase inhibitor or aldosterone receptor inhibitor. Hou et al. reported on the anti-hypertensive efficacy of malate and aspartate via NO and L-arginine generation in HK-2 cells [69]. Using HK-2 cells, they demonstrated that interleukin-17A raises blood pressure through upregulating Na+/Cl cotransporter and Na+/H+ exchanger 3, which are also the main characters of sodium reabsorption in the proximal and distal convoluted tubules [70]. These studies have confirmed that HK-2 is a promising model for the screening of anti-hypertensive agents.

2.1.4. RAAS Modulators Identified from Nature

Numerous RAAS modulators have been identified in nature that can be classified as ACE and AGT1R inhibitors (Table 2). The identified ACE inhibitors include polyphenolics, alkaloids, and short-chain polypeptides (Table 2). Particularly in chalcones, these widespread flavonoids serve as calcium flux or ACE inhibitors [71,72]. Some Traditional Chinese medicines such as Dohaekseunggi-tang and Huatuo exhibit anti-hypertensive activity in benchside experiments, and are worthy of further investigation (Table 2). Some proteins digested and isolated from bonito, milk, sardines, seaweeds, or royal jelly show antihypertensive activity via inhibiting ACE activity. These bioactive peptides are currently available commercially [73,74].
To screen AGT1R inhibitors, two parallel approaches, i.e., directly blocking receptor activity and reducing receptor expression, will be performed. Table 2 shows natural compounds that could block AGT1R activity. As far as we know, only five plants, i.e., Alisma orientale Juzepczuk, Cynodon dactylon, Eucommia ulmoides Oliver, Astragalus membranaceus, and Salvia miltiorrhizae, have traditionally been used as herbal medicines and contain identified natural AGT1R inhibitors (see Table 2). Two traditional Chinese medicines, i.e., the injection of DanHong and Liuwei Dihuang taken orally have also been proven to inhibit AGT1R activity in an in vitro assay (Table 2). On the other hand, Yiqi Huaju formula and Songling xuemaikang capsules have been found to attenuate hypertension in vivo through down-regulating AGT1R expression (Table 2). Tanshinone IIA, a flavonoid from S. miltiorrhizae, directly blocks AGT1R activity and reduces AGT1R expression via TGF-1β/Smads signaling pathway in vivo (Table 2). These reports provide the potential for insights into finding new RAAS modulators from natural sources.
Despite the data presented in Table 2, several common foods have also shown antihypertensive potency in multiple phases. Probiotics can diminish high blood pressure by mediating immunomodulation and adjusting sympathetic nerve activity [125]. Bioactive components of avocado (Persea americana) contain alkaloids, unsaturated fatty acids, and flavonoids that lessen either high arterial pressure or body weight [126]. Tomato (Solanum lycopersicum), a common vegetable, is another example of a natural ACE inhibitor. During the past two years, five clinical trials studying the health impact of tomato extracts on a high cardiovascular risk population pointed to a reduced risk of hypertension or other cardiovascular diseases [127,128,129,130,131]. By exploring the detailed mechanisms, the major bioactive component of tomato, lycopene, was identified as an ACE inhibitor and antioxidant that might be the main substance responsible for its antihypertensive efficacy [132]. Additionally, coffee and tea are well known as antihypertensive foods [133]. Tea and coffee have antihypertensive potential through diuresis [134,135] and changes in emotional states [136,137]. The main components of tea and coffee [138,139], caffeine, theophylline, chlorogenic acid, and epigallocatechin, are known as antihypertensive compounds through their ability to modulate salt reabsorption and immunomodulation [137,139,140,141,142]. However, the antihypertensive efficacy of coffee and tea has been challenged by the latest epidemiological studies in different regions and on both genders. According to a statistical analysis of US adults and a meta-analysis from PubMed, coffee intake reduces the likelihood of hypertension [143,144]. In Singapore, antihypertensive potential versus daily coffee intake exhibits a U shape, and tea consumption slightly increases the risk of hypertension occurrence [145]. Through further investigation by gender, three epidemiological studies and meta-analyses reveal that only women could alleviate hypertensive risk through the consumption of coffee, except in the case of postmenopausal women [146,147,148]. These controversial results remind us of the discrepancy between recent knowledge and our common sense concerning coffee/tea consumption and lowering hypertensive risk.

2.2. Neurogenic Hypertensive System

2.2.1. Action Mechanism of Neurogenic Hypertensive System in Blood Pressure Managing

Unlike RAAS, which regulates blood pressure by controlling total blood volume, sympathetic nerves govern blood pressure by adjusting heart rate and vascular diameter. Sympathetic nerves send neurotransmitters (mainly epinephrine and norepinephrine) as signals to the sinus node and smooth muscle cells within blood vessels [149]. Once target cells receive signals via binding to adrenergic receptors (ARs), intracellular Ca2+ flow forms via the L-type Ca2+ channel, causing smooth muscle cell contraction [149]. ARs are divided into two types: α-type and β-type, for which α-AR mainly induces smooth muscle contraction and β-AR excites the sinus node and relaxes smooth muscle cells (see Figure 1B). This balancing pathway plays a critical role in mediating blood pressure.

2.2.2. Conventional Drugs for Modulating the Neurogenic Hypertensive System

The main actors for the neurogenic hypertensive system are ARs and the Ca2+ channel, so major modulators in this system are predominantly focused on ARs and Ca2+ channel. α-blockers (e.g., Doxazosin and Terazosin) block signals that are sympathetic toward α-AR, which induces vasocontraction [150]. β-blockers (e.g., Propranolol and Bisprolol) can hinder heart rates [151]. Critically, calcium blockers (e.g., Nifedipine and Diltiazem) act at the end of the neurogenic hypertensive system by inhibiting both heart rate and vasoconstriction [152]. Ordinarily, sympathetic hyperactivity is an important cause of primary hypertension in youth [153,154]. Hence, AR and calcium blockers are the first line of hypertension medication, especially primary hypertension in the young- or middle-aged population [155,156]. However, a rapid decrease of blood pressure might sometimes trigger ischemic heart disease [152]. Also, ARs are reported to induce sexual dysfunction like erectile dysfunction, ejaculatory disorder, and reduced sexual desire [157,158]. These adverse effects might diminish willingness to follow medical advice.

2.2.3. Approaches to Screening Controllers for Neurogenic Hypertensive System

The screening methods for controllers of neurogenic hypertensive systems are mainly in vitro, ex vivo, and in vivo models. In vitro assays are performed only at the cellular or tissue levels due to the absence of enzymes in the neurogenic hypertensive system. Chinese hamster ovary cells (CHO), vascular smooth muscle cells (VSMC, isolated from thoracic aorta or the commercial cell line, A7r5), and the cardiomyocyte cell line h9c2 are used to evaluate the overall inhibiting activity of AR or calcium. CHO cell derivatives like CHO-K1 and CHO-S are used as carriers for expressing α-AR or β-AR and for measuring the downstream cAMP change after the induction of the neurotransmitter [159,160]. These cell models exert a searching approach for a potential ligand. VSMC directly responds to signals from norepinephrine or external calcium supplements [121,161]. β-ARs and the Ca2+ channel have demonstrated their expression in the h9c2 cell line [162,163]. Hence, the h9c2 cell has been applied in the study of pathological or cardioprotective efficacy in ischemic heart disease or cardiac hypertrophy for years [164,165]. However, the application of h9c2 is not widely used in screening β-blockers or calcium blockers in drug development; this weakness needs further exploration. An ex vivo model usually comprises 4 mm lengths of rat aortic rings isolated from the thoracic arteries and immersed in Krebs solution for treatment [166]. This explant model could directly observe the diameter changes of the aortic rings after treatment with a neurotransmitter or Ca2+ [167].
The two most frequently-used animals in in vivo models for screening neurogenic hypertensive systems are environmental stimulation and spontaneous hypertensive mice [168]. The principle of environmental induction involves using environmental stimulus to trigger psycho-emotional stress, and subsequently arterial hypertension, through sympathetic nerve hyperactivation [169]. Common environmental stimuli include flashing lights, loud noises, restraints, cages, and high/low temperatures [170]. Therefore, continuous stimulation is also useful for investigating neurogenic hypertensive systems. Inherited hypertensive mice, BPH/2J (Schlager), are derived from the hypertensive parental strain BPH/1J, whose hypertensive symptoms are milder than those of BPH/2J [171]. BPH/2J exhibits spontaneously-sympathetic hyperactivity and low RAAS activity that results in a high heart rate and high blood pressure [172,173]. Due to these different causes of hypertension, BPH/2J and SHR represent completely different approaches.
Despite the existence of the wet-lab model, one in silico model is also provided for neurogenic hypertensive systems, i.e., β2-AR [174]. By evaluating ligand-binding properties, the essential ligand binding sites of β2-AR are identified, i.e., Asp113 and Asn312 [175]. Also, polar amino acid residues in transmembrane domain 5 of β2-AR, and especially Ser203 and Ser207, play an important role in agonist-binding stabilization [175].

2.2.4. Natural ARs and Calcium Blockers

Compared with AGT1R, blocking the AR and L-type calcium channel is more common for maintaining normal-tension. The AR and calcium channel can directly control blood vessel diameter and vasoconstriction; this activation lessens the blood vessel diameter and causes an increase in blood pressure [152]. Comparing adrenergic blockers and calcium blockers, calcium blockers also affect the heart rate; thereby, calcium blockers are used in arrythmia more frequently than in hypertension [152]. For the treatment of hypertension, Table 3 shows known β-blockers and calcium blockers from natural sources. For α-blockers, tempol (a food in South-East Asia made from fermented soybean) and fermented Lactobacillus rhamnosus GR-1 could treat preeclampsia and ameliorate cardiac hypertrophy by inhibiting α-AR [176,177]. These two studies have demonstrated the anti-hypertensive potential of probiotics instead of RAAS. In a review about the pathogenetic characteristics of gut microbiota in hypertension published by Kang and Cai, a possible underlying mechanism affects hypertensive progress through three phases: inflammatory regulation, ACE inhibition, and sympathetic activity adjustment [178]; this area needs further investigation. Again, three coumarins (paeruptorin A, B, and C) isolated from Peucedanum praeruptorum Dunn and one alkaloid ((+)-nantenine) from Nandina domestica have also been shown to have α-AR blocker activities in vitro and in vivo [179]. These results further revealed that finding adrenergic blockers and calcium blockers from natural sources is a promising avenue of research in new drug discovery and development.

3. Conclusions and Remarks

In this article, we have collected recent research and developments of antihypertensive agents from natural sources according to the two main blood-pressure-regulating mechanisms: RAAS and the neurogenic hypertensive system. The implementation of American Heart Association (AHA)/European Society of Cardiology (ESC) guidelines for the management of hypertensive patients, as well as the public health and cost implications, need to be reflected upon [17]. However, drug resistance and poor adherence are urgent issues associated with the treatment of hypertension; these could be a niche for drug development, especially from natural sources [204]. Additionally, folk medicines, medicinal plants, and herbs may be incorporated into the regimen of anti-hypertension treatments according to their experimental efficacy in preclinical trials. Figure 2 summarizes such substances. To accelerate the screening rates, molecular docking is a potential tool for use before any wet-lab validation, including enzymatic tests or cellular assays. However, the absence of target protein modules is an emergent application for molecular docking in antihypertensive compound discovery. Also, recently-developed cellular or enzymatic analyzing methods are not particularly well-suited for the in vivo or in silico discovery, validation, or evaluation of the regulating mechanisms of blood pressure. Nevertheless, numerous compounds or decoctions have been found to have hypotensive efficacy. Moreover, some studies have found other enzymes that could also become antihypertensive targets, such as endothelial NO synthase (eNOS) [123,205,206,207,208,209,210], endothelin-1 receptor [211], and G-protein-coupled receptor kinase 4 (GRK4) [212].
Additionally, the characteristics of the gut–brain axis in blood pressure regulation has received much attention in recent decades. To our knowledge, gut microbiota and the brain could cross talk through various routes, e.g., small microbial metabolites such as sodium butyrate and vitamin D, autonomic nerves system, and intestinal immunocytes [213,214,215,216,217]. A comparison of gut microbiota between healthy and hypertensive people reveals compositional differences, particularly in Firmicutes and Bacteroidetes; this difference might be attributed to the cause of hypertension, at least partially [218]. Accordingly, some studies have focused on the treatment of fluctuating gut microbiota. Probiotic products, such as kefir and lactulose, have been shown to negate hypertensive symptoms in a rat model [125,219,220]. This evidence also offers an insights into the potential of applying natural-source gut-microbiota modulators or traditional Chinese herbal medicine in hypertensive regimens [221]. The exploration of new targets for drug screening to accelerate anti-hypertension drug discovery and development can be more profitable and more effective than other recent methods.

Author Contributions

Conceptualization, S.-R.L. and C.-F.W.; validation, S.-Y.L., Y.-S.F., and C.-F.W.; investigation, C.-C.C. and S.-R.L.; writing—original draft preparation, S.-Y.L. and C.-C.C.; writing—review and editing, Y.-S.F. and C.-F.W.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. James, P.A.; Oparil, S.; Carter, B.L.; Cushman, W.C.; Dennison-Himmelfarb, C.; Handler, J.; Lackland, D.T.; LeFevre, M.L.; MacKenzie, T.D.; Ogedegbe, O.; et al. 2014 evidence-based guideline for the management of high blood pressure in adults: Report from the panel members appointed to the eighth Joint National Committee (JNC 8). JAMA 2014, 311, 507–520. [Google Scholar] [CrossRef]
  2. Zhang, W.; Li, N. Prevalence, risk factors, and management of prehypertension. Int. J. Hypertens. 2011, 2011, 605359. [Google Scholar] [CrossRef] [PubMed]
  3. Whelton, P.K.; Carey, R.M.; Aronow, W.S.; Casey, D.E., Jr.; Collins, K.J.; Dennison Himmelfarb, C.; DePalma, S.M.; Gidding, S.; Jamerson, K.A.; Jones, D.W.; et al. 2017 ACC/AHA/AAPA/ABC/ACPM/AGS/APhA/ASH/ASPC/NMA/PCNA guideline for the prevention, detection, evaluation, and management of high blood pressure in adults: A report of the american college of cardiology/american heart association task force on clinical practice guidelines. J. Am. Coll. Cardiol. 2018, 71, e127–e248. [Google Scholar] [CrossRef] [PubMed]
  4. Bolivar, J.J. Essential hypertension: An approach to its etiology and neurogenic pathophysiology. Int. J. Hypertens. 2013, 2013, 547809. [Google Scholar] [CrossRef] [PubMed]
  5. Puar, T.H.; Mok, Y.; Debajyoti, R.; Khoo, J.; How, C.H.; Ng, A.K. Secondary hypertension in adults. Singap. Med. J. 2016, 57, 228–232. [Google Scholar] [CrossRef] [PubMed]
  6. Ji, L.D.; Li, J.Y.; Yao, B.B.; Cai, X.B.; Shen, Q.J.; Xu, J. Are genetic polymorphisms in the renin-angiotensin-aldosterone system associated with essential hypertension? Evidence from genome-wide association studies. J. Hum. Hypertens. 2017, 31, 695–698. [Google Scholar] [CrossRef] [PubMed]
  7. Alshaarawy, O.; Elbaz, H.A. Cannabis use and blood pressure levels: United States national health and nutrition examination survey, 2005–2012. J. Hypertens. 2016, 34, 1507–1512. [Google Scholar] [CrossRef]
  8. Andriolo, V.; Dietrich, S.; Knüppel, S.; Bernigau, W.; Boeing, H. Traditional risk factors for essential hypertension: Analysis of their specific combinations in the EPIC-Potsdam cohort. Sci. Rep. 2019, 9, 1501. [Google Scholar] [CrossRef]
  9. Mills, K.T.; Bundy, J.D.; Kelly, T.N.; Reed, J.E.; Kearney, P.M.; Reynolds, K.; Chen, J.; He, J. Global disparities of hypertension prevalence and control: A systematic analysis of population-based studies from 90 countries. Circulation 2016, 134, 441–450. [Google Scholar] [CrossRef]
  10. Lewington, S.; Lacey, B.; Clarke, R.; Guo, Y.; Kong, X.L.; Yang, L.; Chen, Y.; Bian, Z.; Chen, J.; Meng, J.; et al. The burden of hypertension and associated risk for cardiovascular mortality in China. JAMA Intern. Med. 2016, 176, 524–532. [Google Scholar] [CrossRef]
  11. Forouzanfar, M.H.; Liu, P.; Roth, G.A.; Ng, M.; Biryukov, S.; Marczak, L.; Alexander, L.; Estep, K.; Hassen Abate, K.; Akinyemiju, T.F.; et al. Global burden of hypertension and systolic blood pressure of at least 110 to 115 mm Hg, 1990–2015. JAMA 2017, 317, 165–182. [Google Scholar] [CrossRef] [PubMed]
  12. Hay, S.I.; Abajobir, A.A.; Abate, K.H.; Abbafati, C.; Abbas, K.M.; Abd-Allah, F.; Abdulkader, R.S.; Abdulle, A.M.; Abebo, T.A.; Abera, S.F.; et al. Global, regional, and national disability-adjusted life-years (DALYs) for 333 diseases and injuries and healthy life expectancy (HALE) for 195 countries and territories, 1990–2016: A systematic analysis for the global burden of disease study 2016. Lancet 2017, 390, 1260–1344. [Google Scholar] [CrossRef]
  13. Muxfeldt, E.S.; de Souza, F.; Margallo, V.S.; Salles, G.F. Cardiovascular and renal complications in patients with resistant hypertension. Curr. Hypertens. Rep. 2014, 16, 471. [Google Scholar] [CrossRef] [PubMed]
  14. Walker, K.A.; Power, M.C.; Gottesman, R.F. Defining the relationship between hypertension, cognitive decline, and dementia: A review. Curr. Hypertens. Rep. 2017, 19, 24. [Google Scholar] [CrossRef] [PubMed]
  15. Van den Born, B.H.; Lip, G.Y.H.; Brguljan-Hitij, J.; Cremer, A.; Segura, J.; Morales, E.; Mahfoud, F.; Amraoui, F.; Persu, A.; Kahan, T.; et al. ESC Council on hypertension position document on the management of hypertensive emergencies. Eur. Heart J. Cardiovasc. Pharmacother. 2019, 5, 37–46. [Google Scholar] [CrossRef] [PubMed]
  16. Villalva, C.M.; Alvarez-Muino, X.L.L.; Mondelo, T.G.; Fachado, A.A.; Fernandez, J.C. Adherence to treatment in hypertension. Adv. Exp. Med. Biol. 2017, 956, 129–147. [Google Scholar] [CrossRef]
  17. Williams, B.; Mancia, G.; Spiering, W.; Agabiti Rosei, E.; Azizi, M.; Burnier, M.; Clement, D.L.; Coca, A.; de Simone, G.; Dominiczak, A.; et al. 2018 ESC/ESH Guidelines for the management of arterial hypertension. Eur. Heart J. 2018, 39, 3021–3104. [Google Scholar] [CrossRef]
  18. Cooper-DeHoff, R.M.; Johnson, J.A. Hypertension pharmacogenomics: In search of personalized treatment approaches. Nat. Rev. Nephrol. 2016, 12, 110–122. [Google Scholar] [CrossRef]
  19. Nakaya, N.; Nakamura, T.; Tsuchiya, N.; Narita, A.; Tsuji, I.; Hozawa, A.; Tomita, H. Psychological distress and the risk of withdrawing from hypertension treatment after an earthquake disaster. Disaster Med. Public Health Prep. 2017, 11, 179–182. [Google Scholar] [CrossRef]
  20. Hu, H.H.; Li, G.; Arao, T. The association of family social support, depression, anxiety and self-efficacy with specific hypertension self-care behaviours in Chinese local community. J. Hum. Hypertens. 2015, 29, 198–203. [Google Scholar] [CrossRef]
  21. Osamor, P.E. Social support and management of hypertension in South-west Nigeria. Cardiovasc. J. Afr. 2015, 26, 29–33. [Google Scholar] [CrossRef] [PubMed]
  22. Hall, E.; Lee, S.Y.; Clark, P.C.; Perilla, J. Social ecology of adherence to hypertension treatment in latino migrant and seasonal farmworkers. J. Transcult. Nurs. 2016, 27, 33–41. [Google Scholar] [CrossRef] [PubMed]
  23. Fongwa, M.N.; Dela Cruz, F.A.; Hays, R.D. African American women’s perceptions of the meaning of support groups for improving adherence to hypertension treatment: A conceptual model. Nurs. Open 2019, 6, 860–870. [Google Scholar] [CrossRef] [PubMed]
  24. Noubiap, J.J.; Nansseu, J.R.; Nyaga, U.F.; Sime, P.S.; Francis, I.; Bigna, J.J. Global prevalence of resistant hypertension: A meta-analysis of data from 3.2 million patients. Heart 2019, 105, 98–105. [Google Scholar] [CrossRef] [PubMed]
  25. Nadal, J.; Channavajjhala, S.K.; Jia, W.; Clayton, J.; Hall, I.P.; Glover, M. Clinical and molecular features of thiazide-induced hyponatremia. Curr. Hypertens. Rep. 2018, 20, 31. [Google Scholar] [CrossRef] [PubMed]
  26. Bandak, G.; Sang, Y.; Gasparini, A.; Chang, A.R.; Ballew, S.H.; Evans, M.; Arnlov, J.; Lund, L.H.; Inker, L.A.; Coresh, J.; et al. Hyperkalemia after initiating renin-angiotensin system blockade: The Stockholm creatinine measurements (SCREAM) project. J. Am. Heart Assoc. 2017, 6, e005428. [Google Scholar] [CrossRef]
  27. Hoover, T.; Lippmann, M.; Grouzmann, E.; Marceau, F.; Herscu, P. Angiotensin converting enzyme inhibitor induced angio-oedema: A review of the pathophysiology and risk factors. Clin. Exp. Allergy 2010, 40, 50–61. [Google Scholar] [CrossRef]
  28. Tedla, Y.G.; Bautista, L.E. drug side effect symptoms and adherence to antihypertensive medication. Am. J. Hypertens. 2016, 29, 772–779. [Google Scholar] [CrossRef]
  29. Lin, S.R.; Fu, Y.S.; Tsai, M.J.; Cheng, H.; Weng, C.F. Natural compounds from herbs that can potentially execute as autophagy inducers for cancer therapy. Int. J. Mol. Sci. 2017, 18, 1412. [Google Scholar] [CrossRef]
  30. Rabkin, S.W. Considerations in understanding the coronary blood flow- left ventricular mass relationship in patients with hypertension. Curr. Cardiol. Rev. 2017, 13, 75–83. [Google Scholar] [CrossRef]
  31. Hsu, C.N.; Tain, Y.L. Regulation of nitric oxide production in the developmental programming of hypertension and kidney disease. Int. J. Mol. Sci. 2019, 20, 681. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Bernstein, K.E.; Khan, Z.; Giani, J.F.; Cao, D.Y.; Bernstein, E.A.; Shen, X.Z. Angiotensin-converting enzyme in innate and adaptive immunity. Nat. Rev. Nephrol. 2018, 14, 325–336. [Google Scholar] [CrossRef] [PubMed]
  33. Feraille, E.; Dizin, E. Coordinated control of ENaC and Na+,K+-ATPase in renal Collecting DUCT. J. Am. Soc. Nephrol. 2016, 27, 2554–2563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Michel, M.C.; Brunner, H.R.; Foster, C.; Huo, Y. Angiotensin II type 1 receptor antagonists in animal models of vascular, cardiac, metabolic and renal disease. Pharmacol. Ther. 2016, 164, 1–81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Salvador, G.L.; Marmentini, V.M.; Cosmo, W.R.; Junior, E.L. Angiotensin-converting enzyme inhibitors reduce mortality compared to angiotensin receptor blockers: Systematic review and meta-analysis. Eur. J. Prev. Cardiol. 2017, 24, 1914–1924. [Google Scholar] [CrossRef] [PubMed]
  36. Satoh, M.; Haga, T.; Hosaka, M.; Obara, T.; Metoki, H.; Murakami, T.; Kikuya, M.; Inoue, R.; Asayama, K.; Mano, N.; et al. The velocity of antihypertensive effects of seven angiotensin II receptor blockers determined by home blood pressure measurements. J. Hypertens. 2016, 34, 1218–1223. [Google Scholar] [CrossRef]
  37. Roush, G.C.; Sica, D.A. Diuretics for Hypertension: A Review and Update. Am. J. Hypertens. 2016, 29, 1130–1137. [Google Scholar] [CrossRef] [Green Version]
  38. Lin, H.Y.; Lee, Y.T.; Chan, Y.W.; Tse, G. Animal models for the study of primary and secondary hypertension in humans. Biomed. Rep. 2016, 5, 653–659. [Google Scholar] [CrossRef] [Green Version]
  39. Sadegh Vishkaei, M.; Ebrahimpour, A.; Abdul-Hamid, A.; Ismail, A.; Saari, N. Angiotensin-I converting enzyme (ACE) inhibitory and anti-hypertensive effect of protein hydrolysate from Actinopyga lecanora (Sea Cucumber) in rats. Mar. Drugs 2016, 14, 176. [Google Scholar] [CrossRef] [Green Version]
  40. Basting, T.; Lazartigues, E. DOCA-salt hypertension: An update. Curr. Hypertens. Rep. 2017, 19, 32. [Google Scholar] [CrossRef]
  41. Bressan, A.F.; Fonseca, G.A.; Tostes, R.C.; Webb, R.C.; Lima, V.V.; Giachini, F.R. Interleukin-10 negatively modulates extracellular signal-regulated kinases 1 and 2 in aorta from hypertensive mouse induced by angiotensin II infusion. Fundam. Clin. Pharmacol. 2019, 33, 31–40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Lu, H.; Howatt, D.A.; Balakrishnan, A.; Moorleghen, J.J.; Rateri, D.L.; Cassis, L.A.; Daugherty, A. Subcutaneous angiotensin II infusion using osmotic pumps induces aortic aneurysms in mice. J. Vis. Exp. 2015, 103, e53191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Liu, Z.; Huang, X.R.; Chen, H.Y.; Fung, E.; Liu, J.; Lan, H.Y. Deletion of angiotensin-converting enzyme-2 promotes hypertensive nephropathy by targeting Smad7 for ubiquitin degradation. Hypertension 2017, 70, 822–830. [Google Scholar] [CrossRef] [PubMed]
  44. Morgan, H.L.; Butler, E.; Ritchie, S.; Herse, F.; Dechend, R.; Beattie, E.; McBride, M.W.; Graham, D. Modeling superimposed preeclampsia using ang II (Angiotensin II) infusion in pregnant stroke-prone spontaneously hypertensive rats. Hypertension 2018, 72, 208–218. [Google Scholar] [CrossRef] [PubMed]
  45. Foulquier, S.; Namsolleck, P.; Van Hagen, B.T.; Milanova, I.; Post, M.J.; Blankesteijn, W.M.; Rutten, B.P.; Prickaerts, J.; Van Oostenbrugge, R.J.; Unger, T. Hypertension-induced cognitive impairment: Insights from prolonged angiotensin II infusion in mice. Hypertens. Res. 2018, 41, 817–827. [Google Scholar] [CrossRef] [PubMed]
  46. Herrmann, S.M.; Textor, S.C. Current concepts in the treatment of renovascular hypertension. Am. J. Hypertens. 2018, 31, 139–149. [Google Scholar] [CrossRef] [PubMed]
  47. Grossman, R.C. Experimental models of renal disease and the cardiovascular system. Open Cardiovasc. Med. J. 2010, 4, 257–264. [Google Scholar] [CrossRef]
  48. Carrive, P. Orexin, stress and central cardiovascular control. A link with hypertension? Neurosci. Biobehav. Rev. 2017, 74, 376–392. [Google Scholar] [CrossRef]
  49. Mohammed-Ali, Z.; Carlisle, R.E.; Nademi, S.; Dickhout, J.G. Animal models of kidney disease. In Animal Models for the Study of Human Disease; Academic Press: Cambridge, MA, USA, 2017; pp. 379–417. [Google Scholar] [CrossRef]
  50. Dupont, S.; Maizel, J.; Mentaverri, R.; Chillon, J.M.; Six, I.; Giummelly, P.; Brazier, M.; Choukroun, G.; Tribouilloy, C.; Massy, Z.A.; et al. The onset of left ventricular diastolic dysfunction in SHR rats is not related to hypertrophy or hypertension. Am. J. Physiol. Heart Circ. Physiol. 2012, 302, H1524–H1532. [Google Scholar] [CrossRef] [Green Version]
  51. Hall, J.E.; do Carmo, J.M.; da Silva, A.A.; Wang, Z.; Hall, M.E. Obesity-induced hypertension: Interaction of neurohumoral and renal mechanisms. Circ. Res. 2015, 116, 991–1006. [Google Scholar] [CrossRef] [Green Version]
  52. Lawson, H.A. Chapter 11—Animal models of metabolic syndrome. In Animal Models for the Study of Human Disease; Conn, P.M., Ed.; Academic Press: Boston, MA, USA, 2013; pp. 243–264. [Google Scholar] [CrossRef]
  53. Bazzano, L.A.; Green, T.; Harrison, T.N.; Reynolds, K. Dietary approaches to prevent hypertension. Curr. Hypertens. Rep. 2013, 15, 694–702. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Wang, C.Y.; Liao, J.K. A mouse model of diet-induced obesity and insulin resistance. Methods Mol. Biol. 2012, 821, 421–433. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Drenjancevic-Peric, I.; Jelakovic, B.; Lombard, J.H.; Kunert, M.P.; Kibel, A.; Gros, M. High-salt diet and hypertension: Focus on the renin-angiotensin system. Kidney Blood Press. Res. 2011, 34, 1–11. [Google Scholar] [CrossRef] [PubMed]
  56. Ivy, J.R.; Evans, L.C.; Moorhouse, R.; Richardson, R.V.; Al-Dujaili, E.A.S.; Flatman, P.W.; Kenyon, C.J.; Chapman, K.E.; Bailey, M.A. Renal and blood pressure response to a high-salt diet in mice with reduced global expression of the glucocorticoid receptor. Front. Physiol. 2018, 9, 848. [Google Scholar] [CrossRef] [PubMed]
  57. Fu, Y.S.; Lue, S.I.; Lin, S.Y.; Luo, C.L.; Chou, C.C.; Weng, C.F. Plantago asiatica seed extracts alleviated blood pressure in phase I(-)Spontaneous hypertension rats. Molecules 2019, 24, 1734. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Banegas-Luna, A.J.; Ceron-Carrasco, J.P.; Perez-Sanchez, H. A review of ligand-based virtual screening web tools and screening algorithms in large molecular databases in the age of big data. Future Med. Chem. 2018, 10, 2641–2658. [Google Scholar] [CrossRef]
  59. Ke, Z.; Su, Z.; Zhang, X.; Cao, Z.; Ding, Y.; Cao, L.; Ding, G.; Wang, Z.; Liu, H.; Xiao, W. Discovery of a potent angiotensin converting enzyme inhibitor via virtual screening. Bioorg. Med. Chem. Lett. 2017, 27, 3688–3692. [Google Scholar] [CrossRef]
  60. Qiao, L.; Li, B.; Chen, Y.; Li, L.; Chen, X.; Wang, L.; Lu, F.; Luo, G.; Li, G.; Zhang, Y. Discovery of anti-hypertensive oligopeptides from adlay based on in silico proteolysis and virtual screening. Int. J. Mol. Sci. 2016, 17, 2099. [Google Scholar] [CrossRef] [Green Version]
  61. Ahmad, I.; Yanuar, A.; Mulia, K.; Mun’im, A. Review of angiotensin-converting enzyme inhibitory assay: Rapid method in drug discovery of herbal plants. Pharmacogn. Rev. 2017, 11, 1–7. [Google Scholar] [CrossRef] [Green Version]
  62. Garcia-Contreras, R.; Vos, P.; Westerhoff, H.V.; Boogerd, F.C. Why in vivo may not equal in vitro—New effectors revealed by measurement of enzymatic activities under the same in vivo-like assay conditions. FEBS J. 2012, 279, 4145–4159. [Google Scholar] [CrossRef]
  63. Han, S.; Lv, Y.; Wei, F.; Fu, J.; Hu, Q.; Wang, S. Screening of bioactive components from traditional Chinese medicines using cell membrane chromatography coupled with mass spectrometry. Phytochem. Anal. 2018, 29, 341–350. [Google Scholar] [CrossRef] [PubMed]
  64. Hou, X.; Wang, S.; Zhang, T.; Ma, J.; Zhang, J.; Zhang, Y.; Lu, W.; He, H.; He, L. Recent advances in cell membrane chromatography for traditional Chinese medicines analysis. J. Pharm. Biomed. Anal. 2014, 101, 141–150. [Google Scholar] [CrossRef] [PubMed]
  65. Zhang, Y.R.; Yuan, Z.Y. Dopamine-mediated inhibition of renal Na+/K+-ATPase in HK-2 cells is reduced by ouabain. Clin. Exp. Pharmacol. Physiol. 2010, 37, 613–618. [Google Scholar] [CrossRef] [PubMed]
  66. Wei, J.; Li, Z.; Ma, C.; Zhan, F.; Wu, W.; Han, H.; Huang, Y.; Li, W.; Chen, D.; Peng, Y. Rho kinase pathway is likely responsible for the profibrotic actions of aldosterone in renal epithelial cells via inducing epithelial-mesenchymal transition and extracellular matrix excretion. Cell Biol. Int. 2013, 37, 725–730. [Google Scholar] [CrossRef]
  67. Saleem, M.; Pokkunuri, I.; Asghar, M. Superoxide increases angiotensin II AT1 receptor function in human kidney-2 cells. FEBS Open Bio 2016, 6, 1273–1284. [Google Scholar] [CrossRef]
  68. Kitada, K.; Nakano, D.; Hitomi, H.; Kobori, H.; Deguchi, K.; Mori, H.; Masaki, T.; Nishiyama, A. Aldosterone induces p21-regulated apoptosis via increased synthesis and secretion of tumour necrosis factor-alpha in human proximal tubular cells. Clin. Exp. Pharmacol. Physiol. 2012, 39, 858–863. [Google Scholar] [CrossRef]
  69. Hou, E.; Sun, N.; Zhang, F.; Zhao, C.; Usa, K.; Liang, M.; Tian, Z. Malate and aspartate increase l-arginine and nitric oxide and attenuate hypertension. Cell Rep. 2017, 19, 1631–1639. [Google Scholar] [CrossRef] [Green Version]
  70. Norlander, A.E.; Saleh, M.A.; Kamat, N.V.; Ko, B.; Gnecco, J.; Zhu, L.; Dale, B.L.; Iwakura, Y.; Hoover, R.S.; McDonough, A.A.; et al. Interleukin-17A regulates renal sodium transporters and renal injury in angiotensin II-induced hypertension. Hypertension 2016, 68, 167–174. [Google Scholar] [CrossRef] [Green Version]
  71. Mahapatra, D.K.; Bharti, S.K. Therapeutic potential of chalcones as cardiovascular agents. Life Sci. 2016, 148, 154–172. [Google Scholar] [CrossRef]
  72. Zhuang, C.; Zhang, W.; Sheng, C.; Zhang, W.; Xing, C.; Miao, Z. Chalcone: A privileged structure in medicinal chemistry. Chem. Rev. 2017, 117, 7762–7810. [Google Scholar] [CrossRef]
  73. Seca, A.M.L.; Pinto, D. Overview on the antihypertensive and anti-obesity effects of secondary metabolites from seaweeds. Mar. Drugs 2018, 16, 237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Bhat, Z.F.; Kumar, S.; Bhat, H.F. Antihypertensive peptides of animal origin: A review. Crit. Rev. Food Sci. Nutr. 2017, 57, 566–578. [Google Scholar] [CrossRef] [PubMed]
  75. Sung, Y.Y.; Kim, D.S.; Choi, G.; Kim, S.H.; Kim, H.K. Dohaekseunggi-tang extract inhibits obesity, hyperlipidemia, and hypertension in high-fat diet-induced obese mice. BMC Complement. Altern. Med. 2014, 14, 372. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Wu, K.Z.; Jiang, Y.G.; Zuo, Y.; Li, A.X. Establishment of the method for screening the potential targets and effective components of huatuo reconstruction pill. Interdiscip. Sci. 2014, 6, 125–132. [Google Scholar] [CrossRef] [PubMed]
  77. Basi, Z.; Turkoglu, V. Purification of angiotensin-converting enzyme from human plasma and investigation of the effect of some active ingredients isolated from Nigella sativa L. extract on the enzyme activity. Biomed. Chromatogr. 2018, 32, e4175. [Google Scholar] [CrossRef] [PubMed]
  78. Kolsi, R.B.A.; Salah, H.B.; Saidi, S.A.; Allouche, N.; Belghith, H.; Belghith, K. Evaluation of nutritional value, characteristics, functional properties of Cymodocea nodosa and its benefits on health diseases. Lipids Health Dis. 2017, 16, 238. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Oboh, G.; Ademiluyi, A.O.; Oyeleye, S.I.; Olasehinde, T.A.; Boligon, A.A. Modulation of some markers of erectile dysfunction and malonaldehyde levels in isolated rat penile tissue with unripe and ripe plantain peels: Identification of the constituents of the plants using HPLC. Pharm. Biol. 2017, 55, 1920–1926. [Google Scholar] [CrossRef] [Green Version]
  80. Khan, M.Y.; Kumar, V. Mechanism of antihypertensive effect of Mucuna pruriens L. seed extract and its isolated compounds. J. Complement. Integr. Med. 2017, 14. [Google Scholar] [CrossRef]
  81. Syama, H.P.; Arya, A.D.; Dhanya, R.; Nisha, P.; Sundaresan, A.; Jacob, E.; Jayamurthy, P. Quantification of phenolics in Syzygium cumini seed and their modulatory role on tertiary butyl-hydrogen peroxide-induced oxidative stress in H9c2 cell lines and key enzymes in cardioprotection. J. Food Sci. Technol. 2017, 54, 2115–2125. [Google Scholar] [CrossRef]
  82. Paiva, L.; Lima, E.; Neto, A.I.; Baptista, J. Angiotensin I-converting enzyme (ACE) inhibitory activity of Fucus spiralis macroalgae and influence of the extracts storage temperature—A short report. J. Pharm. Biomed. Anal. 2016, 131, 503–507. [Google Scholar] [CrossRef]
  83. Gamboa-Gomez, C.I.; Gonzalez-Laredo, R.F.; Gallegos-Infante, J.A.; Perez, M.D.; Moreno-Jimenez, M.R.; Flores-Rueda, A.G.; Rocha-Guzman, N.E. Antioxidant and angiotensin-converting enzyme inhibitory activity of eucalyptus camaldulensis and Litsea glaucescens infusions fermented with kombucha consortium. Food Technol. Biotechnol. 2016, 54, 367–374. [Google Scholar] [CrossRef] [PubMed]
  84. Penas, E.; Limon, R.I.; Martinez-Villaluenga, C.; Restani, P.; Pihlanto, A.; Frias, J. Impact of elicitation on antioxidant and potential antihypertensive properties of Lentil Sprouts. Plant Foods Hum. Nutr. 2015, 70, 401–407. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Li, S.; Shah, N.P. Effects of Pleurotus eryngii polysaccharides on bacterial growth, texture properties, proteolytic capacity, and angiotensin-I-converting enzyme-inhibitory activities of fermented milk. J. Dairy Sci. 2015, 98, 2949–2961. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Limon, R.I.; Penas, E.; Torino, M.I.; Martinez-Villaluenga, C.; Duenas, M.; Frias, J. Fermentation enhances the content of bioactive compounds in kidney bean extracts. Food Chem. 2015, 172, 343–352. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Sila, A.; Bayar, N.; Ghazala, I.; Bougatef, A.; Ellouz-Ghorbel, R.; Ellouz-Chaabouni, S. Water-soluble polysaccharides from agro-industrial by-products: Functional and biological properties. Int. J. Biol. Macromol. 2014, 69, 236–243. [Google Scholar] [CrossRef]
  88. Kancabas, A.; Karakaya, S. Angiotensin-converting enzyme (ACE)-inhibitory activity of boza, a traditional fermented beverage. J. Sci. Food Agric. 2013, 93, 641–645. [Google Scholar] [CrossRef]
  89. Bakri, M.; Yi, Y.; Chen, L.D.; Aisa, H.A.; Wang, M.H. Alkaloids of Nitraria sibirica Pall. decrease hypertension and albuminuria in angiotensin II-salt hypertension. Chin. J. Nat. Med. 2014, 12, 266–272. [Google Scholar] [CrossRef]
  90. Ramesar, S.; Baijnath, H.; Govender, T.; Mackraj, I. Angiotensin I-converting enzyme inhibitor activity of nutritive plants in KwaZulu-Natal. J. Med. Food 2008, 11, 331–336. [Google Scholar] [CrossRef]
  91. Kang, D.G.; Oh, H.; Cho, D.K.; Kwon, E.K.; Han, J.H.; Lee, H.S. Effects of bulb of Fritillaria ussuriensis maxim. on angiotensin converting enzyme and vascular release of NO/cGMP in rats. J. Ethnopharmacol. 2002, 81, 49–55. [Google Scholar] [CrossRef]
  92. Lau, Y.S.; Kwan, C.Y.; Ku, T.C.; Hsieh, W.T.; Wang, H.D.; Nishibe, S.; Dharmani, M.; Mustafa, M.R. Apocynum venetum leaf extract, an antihypertensive herb, inhibits rat aortic contraction induced by angiotensin II: A nitric oxide and superoxide connection. J. Ethnopharmacol. 2012, 143, 565–571. [Google Scholar] [CrossRef]
  93. Zhang, M.Y.; Guo, F.F.; Wu, H.W.; Yu, Y.Y.; Wei, J.Y.; Wang, S.F.; Zhang, Y.X.; Xian, M.H.; Wu, Q.H.; Zhao, B.C.; et al. DanHong injection targets endothelin receptor type B and angiotensin II receptor type 1 in protection against cardiac hypertrophy. Oncotarget 2017, 8, 103393–103409. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Zhang, Y.; Qian, X.; Sun, X.; Lin, C.; Jing, Y.; Yao, Y.; Ma, Z.; Kuai, M.; Lu, Y.; Kong, X.; et al. Liuwei Dihuang, a traditional Chinese medicinal formula, inhibits proliferation and migration of vascular smooth muscle cells via modulation of estrogen receptors. Int. J. Mol. Med. 2018, 42, 31–40. [Google Scholar] [CrossRef] [PubMed]
  95. He, Y.M.; Yang, H.J.; Yang, Q.; Cui, J.G.; Wang, T.Z.; Chen, Y.; Wang, P.W.; Zhang, T.; Wang, W.J. Yiqi Huaju formula, a Chinese herbal medicine, reduces arterial pressure in saltsensitive hypertension by inhibiting reninangiotensin system activation. Mol. Med. Rep. 2015, 12, 5321–5327. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Zhao, Y.Q.; Liu, W.; Cai, X.Y.; Xu, Q.; Shi, H.; Wang, W.; Wu, C.Y.; Li, J.; Wang, R.H.; Jiang, H.T. The regulatory mechanism of songling xuemaikang capsule on PPARgamma in spontaneously hypertensive rats: An experimental study. Zhongguo Zhong Xi Yi Jie He Za Zhi 2013, 33, 1236–1241. [Google Scholar]
  97. Su, P.S.; Doerksen, R.J.; Chen, S.H.; Sung, W.C.; Juan, C.C.; Rawendra, R.D.; Chen, C.R.; Li, J.W.; Aisha; Huang, T.C.; et al. Screening and profiling stilbene-type natural products with angiotensin-converting enzyme inhibitory activity from Ampelopsis brevipedunculata var. hancei (Planch.) Rehder. J. Pharm. Biomed. Anal. 2015, 108, 70–77. [Google Scholar] [CrossRef]
  98. Nakabayashi, R.; Yang, Z.; Nishizawa, T.; Mori, T.; Saito, K. Top-down targeted metabolomics reveals a sulfur-containing metabolite with inhibitory activity against angiotensin-converting enzyme in Asparagus officinalis. J. Nat. Prod. 2015, 78, 1179–1183. [Google Scholar] [CrossRef]
  99. Bleakley, S.; Hayes, M.; O’Shea, N.; Gallagher, E.; Lafarga, T. Predicted release and analysis of novel ACE-I, renin, and DPP-IV inhibitory peptides from common oat (Avena sativa) protein hydrolysates using in silico analysis. Foods 2017, 6, 108. [Google Scholar] [CrossRef] [Green Version]
  100. Morais, K.L.; Ianzer, D.; Miranda, J.R.; Melo, R.L.; Guerreiro, J.R.; Santos, R.A.; Ulrich, H.; Lameu, C. Proline rich-oligopeptides: Diverse mechanisms for antihypertensive action. Peptides 2013, 48, 124–133. [Google Scholar] [CrossRef]
  101. Siemerink, M.; Schebb, N.H.; Liesener, A.; Perchuc, A.M.; Schoni, R.; Wilmer, M.; Hayen, H.; Karst, U.; Vogel, M. Development of a fast liquid chromatography/mass spectrometry screening method for angiotensin-converting enzyme (ACE) inhibitors in complex natural mixtures like snake venom. Rapid Commun. Mass Spectrom. 2010, 24, 687–697. [Google Scholar] [CrossRef]
  102. Nawaz, K.A.A.; David, S.M.; Murugesh, E.; Thandeeswaran, M.; Kiran, K.G.; Mahendran, R.; Palaniswamy, M.; Angayarkanni, J. Identification and in silico characterization of a novel peptide inhibitor of angiotensin converting enzyme from pigeon pea (Cajanus cajan). Phytomedicine 2017, 36, 1–7. [Google Scholar] [CrossRef]
  103. Liu, J.C.; Hsu, F.L.; Tsai, J.C.; Chan, P.; Liu, J.Y.; Thomas, G.N.; Tomlinson, B.; Lo, M.Y.; Lin, J.Y. Antihypertensive effects of tannins isolated from traditional Chinese herbs as non-specific inhibitors of angiontensin converting enzyme. Life Sci. 2003, 73, 1543–1555. [Google Scholar] [CrossRef]
  104. Vijayakumar, B.; Parasuraman, S.; Raveendran, R.; Velmurugan, D. Identification of natural inhibitors against angiotensin I converting enzyme for cardiac safety using induced fit docking and MM-GBSA studies. Pharmacogn. Mag. 2014, 10, S639–S644. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Kang, D.G.; Lee, Y.S.; Kim, H.J.; Lee, Y.M.; Lee, H.S. Angiotensin converting enzyme inhibitory phenylpropanoid glycosides from Clerodendron trichotomum. J. Ethnopharmacol. 2003, 89, 151–154. [Google Scholar] [CrossRef]
  106. Li, B.; Qiao, L.; Li, L.; Zhang, Y.; Li, K.; Wang, L.; Qiao, Y. A novel antihypertensive derived from adlay (Coix larchryma-jobi L. var. ma-yuen Stapf) glutelin. Molecules 2017, 22, 123. [Google Scholar] [CrossRef] [Green Version]
  107. Parichatikanond, W.; Pinthong, D.; Mangmool, S. Blockade of the renin-angiotensin system with delphinidin, cyanin, and quercetin. Planta Med. 2012, 78, 1626–1632. [Google Scholar] [CrossRef] [Green Version]
  108. Zhang, Y.Q.; Luo, J.G.; Han, C.; Xu, J.F.; Kong, L.Y. Bioassay-guided preparative separation of angiotensin-converting enzyme inhibitory C-flavone glycosides from Desmodium styracifolium by recycling complexation high-speed counter-current chromatography. J. Pharm. Biomed. Anal. 2015, 102, 276–281. [Google Scholar] [CrossRef]
  109. Nagai, T.; Nagashima, T. Functional properties of dioscorin, a soluble viscous protein from Japanese yam (Dioscorea opposita thunb.) tuber mucilage Tororo. Z. Naturforsch. C. J. Biosci. 2006, 61, 792–798. [Google Scholar] [CrossRef]
  110. Eckert, E.; Zambrowicz, A.; Pokora, M.; Setner, B.; Dabrowska, A.; Szoltysik, M.; Szewczuk, Z.; Polanowski, A.; Trziszka, T.; Chrzanowska, J. Egg-yolk protein by-product as a source of ACE-inhibitory peptides obtained with using unconventional proteinase from Asian pumpkin (Cucurbita ficifolia). J. Proteom. 2014, 110, 107–116. [Google Scholar] [CrossRef]
  111. Yan, J.; Shi, X.; Donkor, P.O.; Zhu, H.; Gao, X.; Ding, L.; Qiu, F. Nine pairs of megastigmane enantiomers from the leaves of Eucommia ulmoides oliver. J. Nat. Med. 2017, 71, 780–790. [Google Scholar] [CrossRef]
  112. Jenis, J.; Kim, J.Y.; Uddin, Z.; Song, Y.H.; Lee, H.H.; Park, K.H. Phytochemical profile and angiotensin I converting enzyme (ACE) inhibitory activity of Limonium michelsonii Lincz. J. Nat. Med. 2017, 71, 650–658. [Google Scholar] [CrossRef]
  113. Geng, F.; He, Y.; Yang, L.; Wang, Z. A rapid assay for angiotensin-converting enzyme activity using ultra-performance liquid chromatography-mass spectrometry. Biomed. Chromatogr. 2010, 24, 312–317. [Google Scholar] [CrossRef] [PubMed]
  114. San Pablo-Osorio, B.; Mojica, L.; Urias-Silvas, J.E. Chia Seed (Salvia hispanica L.) pepsin hydrolysates inhibit angiotensin-converting enzyme by interacting with its catalytic site. J. Food Sci. 2019, 84, 1170–1179. [Google Scholar] [CrossRef] [PubMed]
  115. Guo, S.; Li, P.; Fu, B.; Chuo, W.; Gao, K.; Zhang, W.; Wang, J.; Chen, J.; Wang, W. Systems-biology dissection of mechanisms and chemical basis of herbal formula in treating chronic myocardial ischemia. Pharmacol. Res. 2016, 114, 196–208. [Google Scholar] [CrossRef] [PubMed]
  116. Gao, X.P.; Xu, D.Y.; Deng, Y.L.; Zhang, Y. Screening of angiotensin converting enzyme inhibitors from Salvia miltiorrhizae. Zhongguo Zhong Yao Za Zhi 2004, 29, 359–362. [Google Scholar]
  117. Maneesh, A.; Chakraborty, K. Previously undescribed antioxidative O-heterocyclic angiotensin converting enzyme inhibitors from the intertidal seaweed Sargassum wightii as potential antihypertensives. Food Res. Int. 2018, 113, 474–486. [Google Scholar] [CrossRef]
  118. Xing, Y.; Liao, J.; Tang, Y.; Zhang, P.; Tan, C.; Ni, H.; Wu, X.; Li, N.; Jia, X. ACE and platelet aggregation inhibitors from Tamarix hohenackeri Bunge (host plant of Herba Cistanches) growing in Xinjiang. Pharmacogn. Mag. 2014, 10, 111–117. [Google Scholar] [CrossRef]
  119. Hou, D.; Yousaf, L.; Xue, Y.; Hu, J.; Wu, J.; Hu, X.; Feng, N.; Shen, Q. Mung bean (Vigna radiata L.): Bioactive polyphenols, polysaccharides, peptides, and health benefits. Nutrients 2019, 11, 1238. [Google Scholar] [CrossRef] [Green Version]
  120. Makino, B.; Kobayashi, M.; Kimura, K.; Ishimatsu, M.; Sakakibara, I.; Higuchi, M.; Kubo, M.; Sasaki, H.; Okada, M. Local variation in the content of angiotensin II and arginine vasopressin receptor antagonistic terpenoids in the rhizomes of Alisma orientale. Planta Med. 2002, 68, 226–231. [Google Scholar] [CrossRef]
  121. Lu, Y.; Li, S.; Wu, H.; Bian, Z.; Xu, J.; Gu, C.; Chen, X.; Yang, D. Beneficial effects of astragaloside IV against angiotensin II-induced mitochondrial dysfunction in rat vascular smooth muscle cells. Int. J. Mol. Med. 2015, 36, 1223–1232. [Google Scholar] [CrossRef] [Green Version]
  122. Jananie, R.K.; Priya, V.; Vijayalakshmi, K. Secondary metabolites of Cynodon dactylon as an antagonist to angiotensin II type1 receptor: Novel in silico drug targeting approach for diabetic retinopathy. J. Pharmacol. Pharmacother. 2012, 3, 20–25. [Google Scholar] [CrossRef] [Green Version]
  123. Ling, W.C.; Liu, J.; Lau, C.W.; Murugan, D.D.; Mustafa, M.R.; Huang, Y. Treatment with salvianolic acid B restores endothelial function in angiotensin II-induced hypertensive mice. Biochem. Pharmacol. 2017, 136, 76–85. [Google Scholar] [CrossRef] [PubMed]
  124. Zhang, N.; Dong, M.; Luo, Y.; Zhao, F.; Li, Y. Danshensu prevents hypoxic pulmonary hypertension in rats by inhibiting the proliferation of pulmonary artery smooth muscle cells via TGF-beta-smad3-associated pathway. Eur. J. Pharmacol. 2018, 820, 1–7. [Google Scholar] [CrossRef] [PubMed]
  125. Robles-Vera, I.; Toral, M.; Romero, M.; Jimenez, R.; Sanchez, M.; Perez-Vizcaino, F.; Duarte, J. Antihypertensive effects of probiotics. Curr. Hypertens. Rep. 2017, 19, 26. [Google Scholar] [CrossRef] [PubMed]
  126. Tabeshpour, J.; Razavi, B.M.; Hosseinzadeh, H. Effects of Avocado (Persea americana) on metabolic syndrome: A comprehensive systematic review. Phytother. Res. 2017, 31, 819–837. [Google Scholar] [CrossRef]
  127. Osinska, A.N.; Begier-Krasinska, B.; Rzymski, P.; Krasinska, A.; Tykarski, A.; Krasinski, Z. The influence of adding tomato extract and acetylsalicylic acid to hypotensive therapy on the daily blood pressure profiles of patients with arterial hypertension and high cardiovascular risk. Kardiochir. Torakochirurgia Pol. 2017, 14, 245–252. [Google Scholar] [CrossRef] [Green Version]
  128. Krasinska, B.; Osinska, A.; Osinski, M.; Krasinska, A.; Rzymski, P.; Tykarski, A.; Krasinski, Z. Standardised tomato extract as an alternative to acetylsalicylic acid in patients with primary hypertension and high cardiovascular risk—A randomised, controlled trial. Arch. Med. Sci. 2018, 14, 773–780. [Google Scholar] [CrossRef]
  129. Krasinska, B.; Osinska, A.; Krasinska, A.; Osinski, M.; Rzymski, P.; Tykarski, A.; Krasinski, Z. Favourable hypotensive effect after standardised tomato extract treatment in hypertensive subjects at high cardiovascular risk: A randomised controlled trial. Kardiol. Pol. 2018, 76, 388–395. [Google Scholar] [CrossRef] [Green Version]
  130. Michalickova, D.; Belovic, M.; Ilic, N.; Kotur-Stevuljevic, J.; Slanar, O.; Sobajic, S. Comparison of polyphenol-enriched tomato juice and standard tomato juice for cardiovascular benefits in subjects with stage 1 hypertension: A randomized controlled study. Plant Foods Hum. Nutr. 2019, 74, 122–127. [Google Scholar] [CrossRef]
  131. Wolak, T.; Sharoni, Y.; Levy, J.; Linnewiel-Hermoni, K.; Stepensky, D.; Paran, E. Effect of tomato nutrient complex on blood pressure: A double blind, randomized dose(-)response study. Nutrients 2019, 11, 950. [Google Scholar] [CrossRef] [Green Version]
  132. Khan, N.I.; Noori, S.; Mahboob, T. Efficacy of lycopene on modulation of renal antioxidant enzymes, ACE and ACE gene expression in hyperlipidaemic rats. J. Renin Angiotensin Aldosterone Syst. 2016, 17, 1–7. [Google Scholar] [CrossRef] [Green Version]
  133. Di Lorenzo, A.; Curti, V.; Tenore, G.C.; Nabavi, S.M.; Daglia, M. Effects of tea and coffee consumption on cardiovascular diseases and relative risk factors: An update. Curr. Pharm. Des. 2017, 23, 2474–2487. [Google Scholar] [CrossRef] [PubMed]
  134. Zhang, Y.; Coca, A.; Casa, D.J.; Antonio, J.; Green, J.M.; Bishop, P.A. Caffeine and diuresis during rest and exercise: A meta-analysis. J. Sci. Med. Sport 2015, 18, 569–574. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Abeywickrama, K.R.; Ratnasooriya, W.D.; Amarakoon, A.M. Oral diuretic activity of hot water infusion of Sri Lankan black tea (Camellia sinensis L.) in rats. Pharmacogn. Mag. 2010, 6, 271–277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Mancini, E.; Beglinger, C.; Drewe, J.; Zanchi, D.; Lang, U.E.; Borgwardt, S. Green tea effects on cognition, mood and human brain function: A systematic review. Phytomedicine 2017, 34, 26–37. [Google Scholar] [CrossRef] [Green Version]
  137. Guessous, I.; Eap, C.B.; Bochud, M. Blood pressure in relation to coffee and caffeine consumption. Curr. Hypertens. Rep. 2014, 16, 468. [Google Scholar] [CrossRef]
  138. Ye, N.S. A minireview of analytical methods for the geographical origin analysis of teas (Camellia sinensis). Crit. Rev. Food Sci. Nutr. 2012, 52, 775–780. [Google Scholar] [CrossRef]
  139. Baspinar, B.; Eskici, G.; Ozcelik, A.O. How coffee affects metabolic syndrome and its components. Food Funct. 2017, 8, 2089–2101. [Google Scholar] [CrossRef]
  140. Agunloye, O.M.; Oboh, G.; Ademiluyi, A.O.; Ademosun, A.O.; Akindahunsi, A.A.; Oyagbemi, A.A.; Omobowale, T.O.; Ajibade, T.O.; Adedapo, A.A. Cardio-protective and antioxidant properties of caffeic acid and chlorogenic acid: Mechanistic role of angiotensin converting enzyme, cholinesterase and arginase activities in cyclosporine induced hypertensive rats. Biomed. Pharmacother. 2019, 109, 450–458. [Google Scholar] [CrossRef]
  141. Hussein, R.M.; Elsirafy, O.M.; Wahba, Y.S.; Kawy, H.S.; Hasanin, A.H.; Hamam, G.G. Theophylline, an old drug with multi-faceted effects: Its potential benefits in immunological liver injury in rats. Life Sci. 2015, 136, 100–107. [Google Scholar] [CrossRef]
  142. Legeay, S.; Rodier, M.; Fillon, L.; Faure, S.; Clere, N. Epigallocatechin gallate: A review of its beneficial properties to prevent metabolic syndrome. Nutrients 2015, 7, 5443–5468. [Google Scholar] [CrossRef] [Green Version]
  143. Ngueta, G. Caffeine and caffeine metabolites in relation to hypertension in U.S. adults. Eur. J. Clin. Nutr. 2019, 1. [Google Scholar] [CrossRef] [PubMed]
  144. Xie, C.; Cui, L.; Zhu, J.; Wang, K.; Sun, N.; Sun, C. Coffee consumption and risk of hypertension: A systematic review and dose-response meta-analysis of cohort studies. J. Hum. Hypertens. 2018, 32, 83–93. [Google Scholar] [CrossRef] [PubMed]
  145. Chei, C.L.; Loh, J.K.; Soh, A.; Yuan, J.M.; Koh, W.P. Coffee, tea, caffeine, and risk of hypertension: The Singapore Chinese health study. Eur. J. Nutr. 2018, 57, 1333–1342. [Google Scholar] [CrossRef] [PubMed]
  146. Grosso, G.; Micek, A.; Godos, J.; Pajak, A.; Sciacca, S.; Bes-Rastrollo, M.; Galvano, F.; Martinez-Gonzalez, M.A. Long-term coffee consumption is associated with decreased incidence of new-onset hypertension: A dose-response meta-analysis. Nutrients 2017, 9, 890. [Google Scholar] [CrossRef] [PubMed]
  147. Navarro, A.M.; Martinez-Gonzalez, M.A.; Gea, A.; Ramallal, R.; Ruiz-Canela, M.; Toledo, E. Coffee consumption and risk of hypertension in the SUN project. Clin. Nutr. 2019, 38, 389–397. [Google Scholar] [CrossRef]
  148. Rhee, J.J.; Qin, F.; Hedlin, H.K.; Chang, T.I.; Bird, C.E.; Zaslavsky, O.; Manson, J.E.; Stefanick, M.L.; Winkelmayer, W.C. Coffee and caffeine consumption and the risk of hypertension in postmenopausal women. Am. J. Clin. Nutr. 2016, 103, 210–217. [Google Scholar] [CrossRef] [Green Version]
  149. Tobe, S.W. beta-adrenergic receptor blockers in hypertension. Can. J. Cardiol. 2014, 30, S1–S2. [Google Scholar] [CrossRef] [Green Version]
  150. Heran, B.S.; Galm, B.P.; Wright, J.M. Blood pressure lowering efficacy of alpha blockers for primary hypertension. Cochrane Database Syst. Rev. 2012, CD004643. [Google Scholar] [CrossRef]
  151. Wiysonge, C.S.; Bradley, H.A.; Volmink, J.; Mayosi, B.M.; Opie, L.H. Beta-blockers for hypertension. Cochrane Database Syst. Rev. 2017, 1, CD002003. [Google Scholar] [CrossRef] [Green Version]
  152. Godfraind, T. Discovery and development of calcium channel blockers. Front. Pharmacol. 2017, 8, 286. [Google Scholar] [CrossRef] [Green Version]
  153. Voora, R.; Hinderliter, A.L. Modulation of sympathetic overactivity to treat resistant hypertension. Curr. Hypertens. Rep. 2018, 20, 92. [Google Scholar] [CrossRef] [PubMed]
  154. Saladini, F.; Palatini, P. Isolated systolic hypertension in young individuals: Pathophysiological mechanisms, prognostic significance, and clinical implications. High Blood Press. Cardiovasc. Prev. 2017, 24, 133–139. [Google Scholar] [CrossRef] [PubMed]
  155. Cruickshank, J.M. The role of beta-blockers in the treatment of hypertension. Adv. Exp. Med. Biol. 2017, 956, 149–166. [Google Scholar] [CrossRef] [PubMed]
  156. Wright, J.M.; Musini, V.M.; Gill, R. First-line drugs for hypertension. Cochrane Database Syst. Rev. 2018, 4, CD001841. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. La Torre, A.; Giupponi, G.; Duffy, D.; Conca, A.; Cai, T.; Scardigli, A. Sexual dysfunction related to drugs: A critical review. Part V: Alpha-blocker and 5-ARI drugs. Pharmacopsychiatry 2016, 49, 3–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Sharp, R.P.; Gales, B.J. Nebivolol versus other beta blockers in patients with hypertension and erectile dysfunction. Ther. Adv. Urol. 2017, 9, 59–63. [Google Scholar] [CrossRef] [Green Version]
  159. Yue, Y.; Xue, H.; Wang, X.; Yang, Q.; Song, Y.; Li, X. High-expression β(1) adrenergic receptor/cell membrane chromatography method based on a target receptor to screen active ingredients from traditional Chinese medicines. J. Sep. Sci. 2014, 37, 244–249. [Google Scholar] [CrossRef]
  160. Tubio, M.R.; Fernandez, N.; Fitzsimons, C.P.; Copsel, S.; Santiago, S.; Shayo, C.; Davio, C.; Monczor, F. Expression of a G protein-coupled receptor (GPCR) leads to attenuation of signaling by other GPCRs: Experimental evidence for a spontaneous GPCR constitutive inactive form. J. Biol. Chem. 2010, 285, 14990–14998. [Google Scholar] [CrossRef] [Green Version]
  161. Valero, M.S.; Olivan-Viguera, A.; Garrido, I.; Langa, E.; Berzosa, C.; Lopez, V.; Gomez-Rincon, C.; Murillo, M.D.; Kohler, R. Rock Tea extract (Jasonia glutinosa) relaxes rat aortic smooth muscle by inhibition of L-type Ca2+ channels. J. Physiol. Biochem. 2015, 71, 785–793. [Google Scholar] [CrossRef]
  162. Parichatikanond, W.; Nishimura, A.; Nishida, M.; Mangmool, S. Prolonged stimulation of beta2-adrenergic receptor with beta2-agonists impairs insulin actions in H9c2 cells. J. Pharmacol. Sci. 2018, 138, 184–191. [Google Scholar] [CrossRef]
  163. Ren, G.; Qiao, H.X.; Yang, J.; Zhou, C.X. Protective effects of steroids from Allium chinense against H2O2-induced oxidative stress in rat cardiac H9C2 cells. Phytother. Res. 2010, 24, 404–409. [Google Scholar] [CrossRef] [PubMed]
  164. Kuo, S.C.; Li, Y.; Cheng, Y.Z.; Lee, W.J.; Cheng, J.T.; Cheng, K.C. Molecular mechanisms regarding potassium bromateinduced cardiac hypertrophy without apoptosis in H9c2 cells. Mol. Med. Rep. 2018, 18, 4700–4708. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Tsai, F.J.; Ho, T.J.; Cheng, C.F.; Shiao, Y.T.; Chien, W.K.; Chen, J.H.; Liu, X.; Tsang, H.; Lin, T.H.; Liao, C.C.; et al. Characteristics of Chinese herbal medicine usage in ischemic heart disease patients among type 2 diabetes and their protection against hydrogen peroxide-mediated apoptosis in H9C2 cardiomyoblasts. Oncotarget 2017, 8, 15470–15489. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Do, K.H.; Kim, M.S.; Kim, J.H.; Rhim, B.Y.; Lee, W.S.; Kim, C.D.; Bae, S.S. Angiotensin II-induced aortic ring constriction is mediated by phosphatidylinositol 3-kinase/L-type calcium channel signaling pathway. Exp. Mol. Med. 2009, 41, 569–576. [Google Scholar] [CrossRef] [Green Version]
  167. Mao, L.M.; Qi, X.W.; Hao, J.H.; Liu, H.F.; Xu, Q.H.; Bu, P.L. In vitro, ex vivo and in vivo anti-hypertensive activity of Chrysophyllum cainito L. extract. Int. J. Clin. Exp. Med. 2015, 8, 17912–17921. [Google Scholar]
  168. Sviglerova, J.; Kuncova, J.; Stengl, M. Chapter 9—Cardiovascular models: Heart secondarily affected by disease. In Animal Models for the Study of Human Disease; Conn, P.M., Ed.; Academic Press: Boston, MA, USA, 2013; pp. 195–220. [Google Scholar] [CrossRef]
  169. Redina, O.E.; Markel, A.L. Stress, genes, and hypertension. Contribution of the ISIAH rat strain study. Curr. Hypertens. Rep. 2018, 20, 66. [Google Scholar] [CrossRef]
  170. Leong, X.F.; Ng, C.Y.; Jaarin, K. Animal models in cardiovascular research: Hypertension and atherosclerosis. Biomed. Res. Int. 2015, 2015, 528757. [Google Scholar] [CrossRef] [Green Version]
  171. Davern, P.J.; Jackson, K.L.; Nguyen-Huu, T.P.; La Greca, L.; Head, G.A. Cardiovascular responses to aversive and nonaversive stressors in Schlager genetically hypertensive mice. Am. J. Hypertens. 2010, 23, 838–844. [Google Scholar] [CrossRef] [Green Version]
  172. Palma-Rigo, K.; Jackson, K.L.; Davern, P.J.; Nguyen-Huu, T.P.; Elghozi, J.L.; Head, G.A. Renin-angiotensin and sympathetic nervous system contribution to high blood pressure in Schlager mice. J. Hypertens. 2011, 29, 2156–2166. [Google Scholar] [CrossRef]
  173. Jackson, K.L.; Dampney, B.W.; Moretti, J.L.; Stevenson, E.R.; Davern, P.J.; Carrive, P.; Head, G.A. Contribution of orexin to the neurogenic hypertension in BPH/2J mice. Hypertension 2016, 67, 959–969. [Google Scholar] [CrossRef]
  174. The UniProt, C. UniProt: The universal protein knowledgebase. Nucleic Acids Res. 2017, 45, D158–D169. [Google Scholar] [CrossRef]
  175. Chan, H.C.; Filipek, S.; Yuan, S. The Principles of Ligand Specificity on beta-2-adrenergic receptor. Sci. Rep. 2016, 6, 34736. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Talebianpoor, M.S.; Mirkhani, H. The effect of tempol administration on the aortic contractile responses in rat preeclampsia model. ISRN Pharmacol. 2012, 2012, 187208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Ettinger, G.; Burton, J.P.; Gloor, G.B.; Reid, G. Lactobacillus rhamnosus GR-1 attenuates induction of hypertrophy in cardiomyocytes but not through secreted protein MSP-1 (p75). PLoS ONE 2017, 12, e0168622. [Google Scholar] [CrossRef] [PubMed]
  178. Kang, Y.; Cai, Y. Gut microbiota and hypertension: From pathogenesis to new therapeutic strategies. Clin. Res. Hepatol. Gastroenterol. 2018, 42, 110–117. [Google Scholar] [CrossRef] [PubMed]
  179. Han, S.; Li, C.; Huang, J.; Wei, F.; Zhang, Y.; Wang, S. Cell membrane chromatography coupled with UHPLC-ESI-MS/MS method to screen target components from Peucedanum praeruptorum Dunn acting on alpha1A adrenergic receptor. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 2016, 1011, 158–162. [Google Scholar] [CrossRef] [PubMed]
  180. Tan, C.S.; Loh, Y.C.; Ng, C.H.; Ch’ng, Y.S.; Asmawi, M.Z.; Ahmad, M.; Yam, M.F. Anti-hypertensive and vasodilatory effects of amended Banxia Baizhu Tianma Tang. Biomed. Pharmacother. 2018, 97, 985–994. [Google Scholar] [CrossRef]
  181. Liu, C.; Zhang, X.; Jing, H.; Zhang, J.; Miao, Y.; Zhai, X.; Chen, S. Using open-tubular capillary electrochromatography with part-coating column for binding constants determination of beta2 -adrenergic receptor with seven drugs. Electrophoresis 2019, 40, 289–295. [Google Scholar] [CrossRef]
  182. Liu, C.; Zhang, X.; Jing, H.; Miao, Y.; Zhao, L.; Han, Y.; Cui, C. Research on drug-receptor interactions and prediction of drug activity via oriented immobilized receptor capillary electrophoresis. Electrophoresis 2015, 36, 2433–2441. [Google Scholar] [CrossRef]
  183. Feng, T.S.; Yuan, Z.Y.; Yang, R.Q.; Zhao, S.; Lei, F.; Xiao, X.Y.; Xing, D.M.; Wang, W.H.; Ding, Y.; Du, L.J. Purgative components in rhubarbs: Adrenergic receptor inhibitors linked with glucose carriers. Fitoterapia 2013, 91, 236–246. [Google Scholar] [CrossRef]
  184. Endale, M.; Song, J.C.; Rhee, M.H.; Liu, K.H.; Kim, T.K.; Kwon, J.G.; Park, K.S.; Chung, K.M.; Kim, T.W. Inhibitory effect of Suaeda asparagoides (Miq.) extract on the motility of rat gastric antrum is mediated by beta-adrenoceptor. Lab. Anim. Res. 2011, 27, 317–325. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Saqib, F.; Janbaz, K.H. Rationalizing ethnopharmacological uses of Alternanthera sessilis: A folk medicinal plant of Pakistan to manage diarrhea, asthma and hypertension. J. Ethnopharmacol. 2016, 182, 110–121. [Google Scholar] [CrossRef] [PubMed]
  186. Shah, A.J.; Gilani, A.H. Blood pressure lowering effect of the extract of aerial parts of Capparis aphylla is mediated through endothelium-dependent and independent mechanisms. Clin. Exp. Hypertens. 2011, 33, 470–477. [Google Scholar] [CrossRef] [PubMed]
  187. Sun, Y.H.; Zhao, J.; Jin, H.T.; Cao, Y.; Ming, T.; Zhang, L.L.; Hu, M.Y.; Hamlati, H.; Pang, S.B.; Ma, X.P. Vasorelaxant effects of the extracts and some flavonoids from the buds of Coreopsis tinctoria. Pharm. Biol. 2013, 51, 1158–1164. [Google Scholar] [CrossRef] [PubMed]
  188. Khan, A.U.; Mustafa, M.R.; Khan, A.U.; Murugan, D.D. Hypotensive effect of Gentiana floribunda is mediated through Ca++ antagonism pathway. BMC Complement. Altern. Med. 2012, 12, 121. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  189. Khan, M.; Khan, A.U.; Najeeb ur, R.; Zafar, M.A.; Hazrat, A.; Gilani, A.H. Cardiovascular effects of Juniperus excelsa are mediated through multiple pathways. Clin. Exp. Hypertens. 2012, 34, 209–216. [Google Scholar] [CrossRef]
  190. Shah, A.J.; Rasheed, M.; Jabeen, Q.; Ahmed, A.; Tareen, R.B.; Gilani, A.H.; Nadir, M.; Ahmad, V.U. Chemical analysis and calcium channel blocking activity of the essential oil of Perovskia abrotanoides. Nat. Prod. Commun. 2013, 8, 1633–1636. [Google Scholar] [CrossRef] [Green Version]
  191. El-Mosallamy, A.E.; Sleem, A.A.; Abdel-Salam, O.M.; Shaffie, N.; Kenawy, S.A. Antihypertensive and cardioprotective effects of pumpkin seed oil. J. Med. Food 2012, 15, 180–189. [Google Scholar] [CrossRef]
  192. Liu, S.J.; Liu, Y.; Zhang, X.; Yang, Z.C.; Liu, W.Z.; Tan, Y.Z. Effect of TGRJ on blood press, NO and Ang II in renal hypertensive rats. Zhong Yao Cai 2012, 35, 953–957. [Google Scholar]
  193. Yang, Q.; Li, Y.; Weng, X.; Chen, Y.; Ruan, C.; Zhu, X. Effect of Tiangou Jiangya capsule on rabbit aortic strip contraction. Zhongguo Zhong Yao Za Zhi 2011, 36, 3349–3352. [Google Scholar]
  194. Siddiqi, H.S.; Mehmood, M.H.; Rehman, N.U.; Gilani, A.H. Studies on the antihypertensive and antidyslipidemic activities of Viola odorata leaves extract. Lipids Health Dis. 2012, 11, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Kato, E.; Inagaki, Y.; Kawabata, J. Higenamine 4′-O-beta-d-glucoside in the lotus plumule induces glucose uptake of L6 cells through beta2-adrenergic receptor. Bioorg. Med. Chem. 2015, 23, 3317–3321. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Kang, N.H.; Mukherjee, S.; Min, T.; Kang, S.C.; Yun, J.W. Trans-anethole ameliorates obesity via induction of browning in white adipocytes and activation of brown adipocytes. Biochimie 2018, 151, 1–13. [Google Scholar] [CrossRef] [PubMed]
  197. Carreyre, H.; Carre, G.; Ouedraogo, M.; Vandebrouck, C.; Bescond, J.; Supuran, C.T.; Thibaudeau, S. Bioactive natural product and superacid chemistry for lead compound identification: A case study of selective hCA III and L-Type Ca2+ current inhibitors for hypotensive agent discovery. Molecules 2017, 22, 915. [Google Scholar] [CrossRef] [Green Version]
  198. Cuong, N.M.; Khanh, P.N.; Huyen, P.T.; Duc, H.V.; Huong, T.T.; Ha, V.T.; Durante, M.; Sgaragli, G.; Fusi, F. Vascular L-type Ca2+ channel blocking activity of sulfur-containing indole alkaloids from Glycosmis petelotii. J. Nat. Prod. 2014, 77, 1586–1593. [Google Scholar] [CrossRef]
  199. Wenjie, W.; Houqing, L.; Liming, S.; Ping, Z.; Gengyun, S. Effects of praeruptorin C on blood pressure and expression of phospholamban in spontaneously hypertensive rats. Phytomedicine 2014, 21, 195–198. [Google Scholar] [CrossRef]
  200. Lim, K.M.; Kwon, J.H.; Kim, K.; Noh, J.Y.; Kang, S.; Park, J.M.; Lee, M.Y.; Bae, O.N.; Chung, J.H. Emodin inhibits tonic tension through suppressing PKCdelta-mediated inhibition of myosin phosphatase in rat isolated thoracic aorta. Br. J. Pharmacol. 2014, 171, 4300–4310. [Google Scholar] [CrossRef] [Green Version]
  201. Park, B.G.; Shin, W.S.; Oh, S.; Park, G.M.; Kim, N.I.; Lee, S. A novel antihypertension agent, sargachromenol D from marine brown algae, Sargassum siliquastrum, exerts dual action as an L-type Ca2+ channel blocker and endothelin A/B2 receptor antagonist. Bioorg. Med. Chem. 2017, 25, 4649–4655. [Google Scholar] [CrossRef]
  202. Huang, Y.L.; Cui, S.Y.; Cui, X.Y.; Cao, Q.; Ding, H.; Song, J.Z.; Hu, X.; Ye, H.; Yu, B.; Sheng, Z.F.; et al. Tetrandrine, an alkaloid from S. tetrandra exhibits anti-hypertensive and sleep-enhancing effects in SHR via different mechanisms. Phytomedicine 2016, 23, 1821–1829. [Google Scholar] [CrossRef]
  203. Onakpoya, I.J.; Heneghan, C.J. Effect of the natural sweetener, steviol glycoside, on cardiovascular risk factors: A systematic review and meta-analysis of randomised clinical trials. Eur. J. Prev. Cardiol. 2015, 22, 1575–1587. [Google Scholar] [CrossRef]
  204. Petit, G.; Berra, E.; Georges, C.M.G.; Capron, A.; Huang, Q.F.; Lopez-Sublet, M.; Rabbia, F.; Staessen, J.A.; Wallemacq, P.; de Timary, P.; et al. Impact of psychological profile on drug adherence and drug resistance in patients with apparently treatment-resistant hypertension. Blood Press. 2018, 27, 358–367. [Google Scholar] [CrossRef] [PubMed]
  205. Lee, J.; Bae, E.H.; Ma, S.K.; Kim, S.W. Altered nitric oxide system in cardiovascular and renal diseases. Chonnam Med. J. 2016, 52, 81–90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Rattmann, Y.D.; Anselm, E.; Kim, J.H.; Dal-Ros, S.; Auger, C.; Miguel, O.G.; Santos, A.R.; Chataigneau, T.; Schini-Kerth, V.B. Natural product extract of Dicksonia sellowiana induces endothelium-dependent relaxations by a redox-sensitive Src- and Akt-dependent activation of eNOS in porcine coronary arteries. J. Vasc. Res. 2012, 49, 284–298. [Google Scholar] [CrossRef] [PubMed]
  207. Ye, B.H.; Lee, S.J.; Choi, Y.W.; Park, S.Y.; Kim, C.D. Preventive effect of gomisin J from Schisandra chinensis on angiotensin II-induced hypertension via an increased nitric oxide bioavailability. Hypertens. Res. 2015, 38, 169–177. [Google Scholar] [CrossRef]
  208. Abdallah, H.M.; El-Bassossy, H.M.; Mohamed, G.A.; El-Halawany, A.M.; Alshali, K.Z.; Banjar, Z.M. Phenolics from Garcinia mangostana alleviate exaggerated vasoconstriction in metabolic syndrome through direct vasodilatation and nitric oxide generation. BMC Complement. Altern. Med. 2016, 16, 359. [Google Scholar] [CrossRef]
  209. Je, H.D.; Kim, H.D.; La, H.O. The inhibitory effect of apigenin on the agonist-induced regulation of vascular contractility via calcium desensitization-related pathways. Biomol. Ther. 2014, 22, 100–105. [Google Scholar] [CrossRef] [Green Version]
  210. Xu, Z.; Wang, X.; Dai, Y.; Kong, L.; Wang, F.; Xu, H.; Lu, D.; Song, J.; Hou, Z. (+/−)-Praeruptorin A enantiomers exert distinct relaxant effects on isolated rat aorta rings dependent on endothelium and nitric oxide synthesis. Chem. Biol. Interact. 2010, 186, 239–246. [Google Scholar] [CrossRef]
  211. Chen, C.Y.; Ho, L.T.; Yang, F.Y.; Juan, C.C.; Au, L.C. Prunellae spica extract contains antagonists for human endothelin receptors. Am. J. Chin. Med. 2013, 41, 85–98. [Google Scholar] [CrossRef]
  212. Rayner, B.; Ramesar, R. The importance of G protein-coupled receptor kinase 4 (GRK4) in pathogenesis of salt sensitivity, salt sensitive hypertension and response to antihypertensive treatment. Int. J. Mol. Sci. 2015, 16, 5741–5749. [Google Scholar] [CrossRef] [Green Version]
  213. Yang, T.; Zubcevic, J. Gut-brain axis in regulation of blood pressure. Front. Physiol. 2017, 8, 845. [Google Scholar] [CrossRef] [Green Version]
  214. Vaidya, A.; Forman, J.P. Vitamin D and hypertension: Current evidence and future directions. Hypertension 2010, 56, 774–779. [Google Scholar] [CrossRef] [PubMed]
  215. Yang, T.; Magee, K.L.; Colon-Perez, L.M.; Larkin, R.; Liao, Y.S.; Balazic, E.; Cowart, J.R.; Arocha, R.; Redler, T.; Febo, M.; et al. Impaired butyrate absorption in the proximal colon, low serum butyrate and diminished central effects of butyrate on blood pressure in spontaneously hypertensive rats. Acta Physiol. 2019, 226, e13256. [Google Scholar] [CrossRef] [PubMed]
  216. Wang, L.; Zhu, Q.; Lu, A.; Liu, X.; Zhang, L.; Xu, C.; Liu, X.; Li, H.; Yang, T. Sodium butyrate suppresses angiotensin II-induced hypertension by inhibition of renal (pro)renin receptor and intrarenal renin-angiotensin system. J. Hypertens. 2017, 35, 1899–1908. [Google Scholar] [CrossRef] [PubMed]
  217. Zuo, K.; Li, J.; Xu, Q.; Hu, C.; Gao, Y.; Chen, M.; Hu, R.; Liu, Y.; Chi, H.; Yin, Q.; et al. Dysbiotic gut microbes may contribute to hypertension by limiting vitamin D production. Clin. Cardiol. 2019, 42, 710–719. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  218. Mushtaq, N.; Hussain, S.; Zhang, S.; Yuan, L.; Li, H.; Ullah, S.; Wang, Y.; Xu, J. Molecular characterization of alterations in the intestinal microbiota of patients with grade 3 hypertension. Int. J. Mol. Med. 2019, 44, 513–522. [Google Scholar] [CrossRef] [Green Version]
  219. Pimenta, F.S.; Luaces-Regueira, M.; Ton, A.M.; Campagnaro, B.P.; Campos-Toimil, M.; Pereira, T.M.; Vasquez, E.C. Mechanisms of action of kefir in chronic cardiovascular and metabolic diseases. Cell. Physiol. Biochem. 2018, 48, 1901–1914. [Google Scholar] [CrossRef]
  220. Zhang, Z.; Zhao, J.; Tian, C.; Chen, X.; Li, H.; Wei, X.; Lin, W.; Zheng, N.; Jiang, A.; Feng, R.; et al. Targeting the gut microbiota to investigate the mechanism of lactulose in negating the effects of a high-salt diet on hypertension. Mol. Nutr. Food Res. 2019, 63, e1800941. [Google Scholar] [CrossRef]
  221. Yang, M.; Lao, L. Emerging applications of metabolomics in traditional chinese medicine treating hypertension: Biomarkers, pathways and more. Front. Pharmacol. 2019, 10, 158. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Regulating mechanism for blood pressure. (A) RASS; (B) Sympathetic nerve regulating system. RAAS: renin-angiotensin-aldosterone systems; ACE I: angiotensin converting enzyme inhibitor.
Figure 1. Regulating mechanism for blood pressure. (A) RASS; (B) Sympathetic nerve regulating system. RAAS: renin-angiotensin-aldosterone systems; ACE I: angiotensin converting enzyme inhibitor.
Jcm 08 02003 g001
Figure 2. Natural compounds act on blood pressure control.
Figure 2. Natural compounds act on blood pressure control.
Jcm 08 02003 g002
Table 1. Complications of hypertension.
Table 1. Complications of hypertension.
Affect OrganComplications
Cardiovascular systemCardiac hypertrophy, heart failure, angina pectoris, coronary heart disease, myocardial infarction.
Artery Atherosclerosis, aneurysms.
BrainStroke (ischemic or hemorrhagic), hypertensive encephalopathy, cognitive decline, dementia.
EyeRetinopathy.
KidneyHypertensive nephropathy, chronic kidney disease.
Table 2. RAAS modulators from natural sources.
Table 2. RAAS modulators from natural sources.
SourceCompound/ExtractionsEvaluated MethodReference
Crude extract/decoction for modulating ACE
Dohaekseunggi-tang in vivo[75]
Huatuo reconstruction pill in silico[76]
Nigella sativa enzymatic[77]
Cymodocea Nodosa +[78]
Plantago majorSeeds extract+[79]
Mucuna pruriens +[80]
Syzygium cuminiSeeds extractin vitro[81]
Fucus spiralis enzymatic[82]
Eucalyptus camaldulensis
Litsea glaucescens
Ferment+[83]
Lens culinarisSprouted extract+[84]
Pleurotus eryngiipolysaccharide+[85]
Phaseolus vulgarisFerment+[86]
Prunus amygdalus
Pistacia vera
juice byproduct+[87]
Bozaprotein extract+[88]
Nitraria sibiricaalkaloidsin vivo[89]
Amaranthus dubius
Amaranthus hybridus
Asystasia gangetica
Galinsoga parviflora
Justicia flava
Oxygonum sinuatum
Physalis viscosa
enzymatic[90]
Tulbaghia violacea in vivo[90]
Bulbus Fritillaria in vivo[91]
Crude extracts or decoction for AGT1R
Apocynum venetum leaf extract in vitro[92]
DanHong injection +[93]
Liuwei Dihuang formula +[94]
Yiqi Huaju formula in vivo[95]
Songling xuemaikang capsule +[96]
ACE inhibitors
Ampelopsis Brevipedunculata(+)-Hopeaphenol; (+)-Vitisin Aenzymatic[97]
Asparagus officinalisAsparaptine+[98]
Avena sativaWWK, WCY, FLLA+[99]
Bothrops jararacaEWPRQIPP, EARPPHPPIPP, EWGRPPGPPIPP, EGGWPRPGP(Glu)IPP,in vitro[100]
Bothrops moojeni(Pyro)EKWPPGGKVPP, (Pyro)EKPRPGPEIPP, (Pyro)ENWPWPGPEIPPenzymatic[101]
Cajanus cajanVVSLSIPR+[102]
Camellia sinensisEpigallocatechin gallate+[59]
Chinese HerbPentagalloylglucose, Isochlorogenic acid B, Methyl 3,4-di-O-caffeoylquinate, (−)-Epigallocatechin gallate, Epigallocatechin-3-O-Methylgallatein vivo[103]
Cleistanthus collinusCleistanthins A, Bin silico[104]
Clerodendron trichotomumActeoside, Isomartynoside, Leucosceptoside A, Martynosideenzymatic[105]
Coptis chinensisBerberinein vitro[91]
Coix larchryma-jobiGAAGGAF, NPATYin vivo[60,106]
Delphinium sp.Cyanin, Delphinidinenzymatic[107]
Desmodium styracifoliumCarlinoside, Schaftoside, Vicenin 1–3+[108]
Dioscorea opposita Thunb.Diascorin+[109]
Egg YorkLapslpgkpkpd+[110]
Eucommia ulmoidesMegastigmane Enantiomersin silico[111]
Glycyrrhiza glabraLicochalcone Aenzymatic[59]
Glycyrrhiza uralensisEchinatin+[59]
Limonium michelsoniiIsolates+[112]
Mucuna PruriensGenistein+[80]
MultisourceCaffeic Acid, Caffeoyl Acetate, Chlorogenic Acid, Ferulic Acid+[113]
Salvia hispanica L.LIVSPLAGRL+[114]
Salvia miltiorrhizaeSalvianolic Acid B+[115,116]
Lithospermic Acid B+[105]
Sargassum wightiiO-Heterocyclic Analoguesin silico[117]
Tamarix hohenackeriChrysoeriol, Quercetin, Isoferulic acid, Methyl-4-O-methylgallate, Gallic acid, Methyl gallateenzymatic[118]
Toona sinensisQuercetin, Resveratrol+[107]
Vigna radiataKDYRL, VTPALR, KLPAGTLF+[119]
Xestospongia Cf. VansoestiSalsolinol+[115]
AGT1R inhibitors
Alisma orientale23-O-acetylalisol B, Alismol, Alisols A, Alisols Benzymatic[120]
Astragalus membranaceusAstragaloside IVin vitro[121]
Cynodon dactylonLinoleoylchloride, Diazoprogesteron, Didodecyl Phthalateenzymatic[122]
Eucommia ulmoidesMegastigmane Enantiomersin silico[111]
Salvia miltiorrhizaeSalvianolic acid Bin vivo[123]
+Tanshinone IIAin vivo[124]
+, as above; WWK, (Trp)2-Lys; WCY, Trp-Cys-Tyr; FLLA, Phe-(Leu)2-Ala EWPRQIPP, Glu-Trp-Pro-Arg-Pro-Gln-Ile-(Pro)2; EARPPHPPIPP, Glu-Ala-Arg-(Pro)2-His-(Pro)2-Ile-(Pro)2; EWGRPPGPPIPP, Glu-Trp-Gly-Arg-(Pro)2-Gly-(Pro)2-Ile-(Pro)2; EGGWPRPGP(Glu)IPP, Glu-(Gly)2-Trp-Pro-Arg-Pro-Gly-Pro-GluIle-(Pro)2; (Pyro)EKWPPGGKVPP, PyroGlu-Lys-Trp-(Pro)2-(Gly)2-Lys-Val-(Pro)2; (Pyro)EKPRPGPEIPP, PyroGlu-Lys-Pro-Arg-Pro-Gly-Pro-Glu-Ile-(Pro)2; (Pyro)ENWPWPGPEIPP, PyroGlu-Asn-(Trp-Pro)2-Gly-Pro-Glu-Ile-(Pro)2; VVSLSIPR, (Val)2-Ser-Leu-Ser-Ile-Pro-Arg; GAAGGAF, Gly-(Ala)2-(Gly)2-Ala-Phe; NPATY, Asn-Pro-Ala-Thr-Tyr; LIVSPLAGRL, Leu-Ile-Val-Ser-Pro-Leu-Ala-Gly-Arg-Leu; KDYRL, Lys-Asp-Tyr-Arg-Leu; VTPALR, Val-Thr-Pro-Ala-Leu-Arg; KLPAGTLF, Lys-Leu-Pro-Ala-Gly-Thr-Leu-Phe.
Table 3. Natural source β blockers and Ca2+ blockers.
Table 3. Natural source β blockers and Ca2+ blockers.
SourceCompoundsTest MethodReference
Crude extract/decoction
β-adrenergic receptor inhibitor
Banxia Baizhu Tianma Tang in vitro[180]
Paeoniae Rubra in vitro[181,182]
Rhubarbs in silico[183]
Suaeda asparagoides in vivo[184]
Ca2+ channel inhibitor
Alternanthera sessilis in vitro[185]
Capparis aphylla in vivo[186]
Coreopsis tinctoria in vitro[187]
Gentiana floribunda in vivo[188]
Jasonia glutinosa in vitro[161]
Juniperus excelsa in vitro[189]
Perovskia abrotanoides essential oil in vitro[190]
Pumpkin seed oil in vivo[191]
Ranunculus japoniucus in vivo[192]
Tiangou Jiangya in vitro[193]
Viola odorata in vivo[194]
Pure compounds
β-adrenergic receptor inhibitor
Nelumbo nuciferaHigenamine 4′-O-β-d-glucosidein vitro[195]
Notopterygium incisumIsoimperatorinenzymatic[159]
Pimpinella anisumTrans-anetholein silico[196]
Ca2+ channel inhibitor
Agelanthus dodoneifoliusDodoneinein vitro[197]
Glycosmis petelotiiN-demethylglypetelotine, Glypetelotinein vitro[198]
Peucedanum praeruptorumPraeruptorin C in vivo[199]
Polygonum multiflorumEmodinin vitro[200]
Sargassum siliquastrumSargachromenol Din vitro[201]
Stephaniae tetrandraeTetrandrine, Fangchinolinein vivo[202]
Stevia rebaudianaSteviosidein vivo[203]

Share and Cite

MDPI and ACS Style

Lin, S.-R.; Lin, S.-Y.; Chen, C.-C.; Fu, Y.-S.; Weng, C.-F. Exploring a New Natural Treating Agent for Primary Hypertension: Recent Findings and Forthcoming Perspectives. J. Clin. Med. 2019, 8, 2003. https://doi.org/10.3390/jcm8112003

AMA Style

Lin S-R, Lin S-Y, Chen C-C, Fu Y-S, Weng C-F. Exploring a New Natural Treating Agent for Primary Hypertension: Recent Findings and Forthcoming Perspectives. Journal of Clinical Medicine. 2019; 8(11):2003. https://doi.org/10.3390/jcm8112003

Chicago/Turabian Style

Lin, Shian-Ren, Shiuan-Yea Lin, Ching-Cheng Chen, Yaw-Syan Fu, and Ching-Feng Weng. 2019. "Exploring a New Natural Treating Agent for Primary Hypertension: Recent Findings and Forthcoming Perspectives" Journal of Clinical Medicine 8, no. 11: 2003. https://doi.org/10.3390/jcm8112003

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