Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Mar 1.
Published in final edited form as: Pharmacol Res. 2020 Feb 2;153:104677. doi: 10.1016/j.phrs.2020.104677

Gut microbiota and neuroinflammation in pathogenesis of hypertension: A potential role for hydrogen sulfide

Basak DONERTAS AYAZ a,b, Jasenka ZUBCEVIC a,*
PMCID: PMC7056572  NIHMSID: NIHMS1563008  PMID: 32023431

Abstract

Inflammation and gut dysbiosis are hallmarks of hypertension (HTN). Hydrogen sulfide (H2S) is an important freely diffusing molecule that modulates the function of neural, cardiovascular and immune systems, and circulating levels of H2S are reduced in animals and humans with HTN. While most research to date has focused on H2S produced endogenously by the host, H2S is also produced by the gut bacteria and may affect the host homeostasis. Here, we review an association between neuroinflammation and gut dysbiosis in HTN, with special emphasis on a potential role of H2S in this interplay.

Keywords: hypertension, neuroinflammation, gut microbiota, hydrogen sulfide

Graphical Abstract

graphic file with name nihms-1563008-f0001.jpg

1. Evidence of neuroinflammation in hypertension

Hypertension (HTN) is a major risk factor for cardiovascular and cerebrovascular events and chronic kidney disease leading to high levels of mortality [1, 2]. The estimated prevalence of HTN among U.S. adults aged ≥20 is reportedly 46% [3]. Approximately 13.7% of hypertensive patients remain resistant or refractory to available anti-hypertensive treatments [4], which may in part be due to neurogenic mechanisms reflecting in increased sympathetic activity and reduced vagal cardiac tone [5, 6]. However, the mechanisms contributing to treatment-resistant HTN are still not completely understood [7]. Dysregulation of renin-angiotensin system (RAS) [8] and presence of inflammation in autonomic brain regions are touted as important contributors to the sympathetic overactivity in HTN [9, 10].

Neuroinflammation is an inflammatory response of central nervous system (CNS) mediated by cytokines, chemokines, reactive oxygen species (ROS), and secondary messengers produced by the resident CNS glia, endothelial cells, and peripherally-derived immune cells [11]. Neuroinflammation has been reported in HTN, however, it is still not clear whether it is a cause or a consequence of HTN, as RAS overactivity, blood brain barrier (BBB) disruption, peripheral immune system (IS) activation, and oxidative stress among other factors are all implicated in pathogenesis of HTN [8]. Correlation of HTN with serious neurological disorders such as Alzheimer’s disease and Parkinson’s disease, both characterized by a neuroinflammatory phenotype and degrees of autonomic dysfunction and blood pressure (BP) abnormalities, highlights the importance of adequate anti-hypertensive treatments for overall brain health [1214].

Brain renin-angiotensin system, sympathetic outflow, neuroinflammation and hypertension

Numerous neurotransmitters and neuromodulators are involved in regulation of sympathetic outflow and neuroinflammation. A plethora of evidence implicates hyperactivity of brain RAS in development and maintenance of HTN in several experimental and genetic animal models including the spontaneously hypertensive rats (SHR) [15, 16], renin transgenic hypertensive rats [17, 18], Dahl salt-sensitive rats [1921], deoxycorticosterone acetate (DOCA) salt-treated rats [2224] and renal hypertensive rats [2528]. Indeed, angiotensin (Ang) II levels were found to be high in cerebrospinal fluid of SHR [29] and in dogs with renal HTN [30], as well as in hypertensive humans following sodium depletion [31].

Within the CNS, the Ang II precursor angiotensinogen is reportedly synthesized in neuronal cells, glial cells and astrocytes [3234]. Pro-hypertensive and pro-sympathetic action of brain Ang II is primarily mediated via the neuronal Ang type 1 receptor (AT1R) [35, 36]. The evidence for this is significant. For example, removal of AT1R in catecholaminergic neurons delayed the onset of Ang II-HTN and attenuated the sympathetic activation [37]. In addition, genetic inhibition of AT1R subtype in the paraventricular nucleus (PVN) of the hypothalamus decreased Ang II HTN [38], and decreased BP, sympathetic activity, and plasma Ang II and norepinephrine (NE) levels in the SHR [39]. AT1R is densely distributed in the ventrolateral medulla, nucleus tractus solitarii (NTS), lamina terminalis, supraoptic nuclei and the PVN, all known to involved in cardioregulation and regulation of body fluid and electrolyte balance [32]. Central AT1R-mediated actions of Ang II are reportedly mediated by phosphoinositide 3-kinase [40, 41], nicotinamide adenine dinucleotide phosphate (NADPH) oxidase [4246], catecholamines [4754], serotonin [54, 55], gamma-amino butyric acid (GABA) [5660], glutamate [61, 62], acetylcholine [63], substance P [6467], endothelial nitric oxide (NO) synthase and NO [68, 69], and vasopressin [7072], all reportedly involved in modulating sympathetic activity. In line with this, PVN infusion of Ang II increased glutamate and decreased GABA levels in PVN, while elevating the BP [73]. Microinjection of Ang II into the PVN increased renal sympathetic nerve discharge, BP [7476] and NO release [74], while microinjection of an AT1R antagonist Losartan reduced GABAA receptor-mediated increase in renal sympathetic nerve activity and BP [77]. Moreover, the increase in BP in response to AT1R activation in the PVN of hypertensive Dahl rats is also suggested to be mediated by enhanced local glutamate receptor activation [78], possibly the glutamate N-methyl-D-aspartate (NMDA) receptor activation [79].

In addition to direct neuronal activation, Ang II is shown to mediate overexpression of brain pro-inflammatory cytokines (PIC) which may also contribute to sympathetic overactivity in HTN [73]. Several studies have reported a mild inflammatory state in several cardioregulatory brain regions in animal models of HTN (Table 1). However, direct effects of neuroinflammation on autonomic nervous system and BP in HTN remain unexplored. Early studies in the NTS of SHR suggested that elevated expression of junctional adhesion molecule-1 mediated the neuroimmune interaction in regulation of BP [80]. Another study showed that an injection of PIC interleukin (IL)-1β into the PVN of the hypothalamus increased BP, while overexpression of the anti-inflammatory IL-10 within the PVN reduced BP in Ang II rodent HTN [81]. Moreover, PVN microinjection of tumor necrosis factor (TNF)-α and IL-1β increased BP and sympathetic outflow in the SHR [82] suggesting direct neuronal effects, while elevated levels of IL-1β and decreased levels of IL-10 and nuclear factor kappa B (NF-κB) activity in the PVN have also been linked to HTN-induced sympathetic hyperactivity in Dahl salt-sensitive hypertensive rats [83, 84].

Table 1.

Presence of inflammation in the cardioregulatory brain regions in animals with hypertension.

HTN model Species Region Reference
SHR rat PVN [8589]
RVLM [86, 9094]
NTS [9597]
2K1C rat PVN [98]
RVLM [99]
Ang II-induced HTN rat PVN [73, 85, 100104]
RVLM [93]
High salt HTN rat PVN [83, 84, 105110]
RVLM [111, 112]
Stress-induced HTN rat PVN [113]
RVLM [114]

Ang II: Angiotensin II; HTN: Hypertension; NTS: Nucleus tractus solitarii; PVN: Paraventricular nucleus; RVLM: Rostral ventrolateral medulla; SHR: Spontaneously hypertensive rat; 2K1C: Two-kidney one-clip

One potential source of increased PIC in the brain are the resident microglia, activation of which has been reported in HTN. Microglia are the resident immune cells of the brain involved in the maintenance of neural environment [115]. They are widely distributed but appear to have varied roles in specific brain regions [116, 117] playing an active role in both physiological and pathological states [118]. Microglia are activated upon various noxious stimuli and inflammatory states [119], upon which they undergo morphological changes [120] and release PIC [121] such as cytokines and prostaglandins, which can influence and modulate neuronal function during both physiological and pathological processes [122124]. Expression of AT1R in has also been reported in glial cells [125128]. It has been suggested that Ang II can increase the production and/or release of PIC from glia [7, 81, 100], which in turn increases the production of ROS that may increase neuronal discharge, thereby indirectly contributing to the sympathetic outflow and increased BP [129, 130]. Ang II binding to the AT1R can also stimulate the microglial NADPH oxidase, leading to oxidative stress and resulting in neuroinflammation [125, 126]. Activation of microglial RhoA/Rho kinase pathway is suggested to be involved in the Ang II-induced microglial activation [131133]. However, AT1R expression has only been reported in activated microglia in certain neurological conditions [134]. Moreover, microglial activation in rodent models of HTN in which Ang II has been infused systemically [7, 81, 100] raises an important question whether Ang II acts directly or indirectly on microglia in HTN. Thus, the exact mechanisms of how Ang II induces microglial activation need to be clarified.

Alongside microglia, astrocytes are involved in neuroinflammation and are capable of producing PIC in response to various pro-inflammatory stimuli [135, 136]. Astrocytes express AT1R and can mediate Ang II-dependent increase in BP and sympathetic outflow in the PVN via inhibition of the astrocyte glutamate transporters [137]. Moreover, Ang II stimulates the production of IL-6 in SHR astrocytes in culture via the AT1R/ NF-κB/ROS pathway [138], while application of Ang II to human astrocytes in vitro increased the translocation of cytosolic components of NADPH oxidase to the cell membrane [139]. In neurons co-cultured with astrocytes, the Ang II-induced neuronal damage was inhibited by an AT1R blocker Valsartan, suggesting that astrocytes mediated central effects of Ang II [140]. In addition, microglia-astroglia communication is enhanced following microglial activation, which can indirectly affect neuronal function through strengthening of gliotransmission mediated by astrocytes [141]. For instance, TNF-α released by activated microglia reportedly potentiates glutamate release from the neighboring astrocytes, which can modulate synaptic plasticity and even lead to neurotoxicity [142]. In addition, microglial adenosine triphosphate is also shown to induce glutamate release by astrocytes which can excite proximal neurons [143].

Microglia can reportedly support adaptive synaptic plasticity through release of neurotrophic factors such as the brain-derived neurotrophic factors [144], while also being able to phagocytose synapses during neurodevelopment [145]. In line with this, minocycline, a microglial inhibitor with antibiotic and anti-inflammatory properties that can cross the BBB, can inhibit microglia in the PVN, thus reducing PIC and ROS production and alleviating HTN [81, 85, 146149]. Concurrent central infusion of minocycline also attenuated increased expression of a microglial marker CD11b in lamina terminalis of rats receiving Ang II [150]. However, although microglial activation in the PVN was increased in Ang II HTN and in stroke-prone SHR, systemic or central administration of minocycline into the PVN failed to decrease BP [148, 151], possibly related to the animal model and dose/route of administration. Minocycline has also been shown to decrease BP in patients with treatment-resistant HTN, owing to its anti-inflammatory properties [152]. Moreover, central administration of a chemically modified tetracycline (CMT-3) with anti-inflammatory activity reduced microglial activation in the PVN in Ang II-induced rodent HTN [153]. Considering the above studies, agents with central anti-oxidant and anti-inflammatory properties may be beneficial in neurogenic HTN. In addition, centrally acting angiotensin converting enzyme (ACE) inhibitors [154156] and AT1R blockers [156160] may be useful in curbing neuroinflammation in HTN. However, the glial-neuronal communication in HTN is complex, and more sophisticated techniques are necessary to delineate the precise cellular and molecular mechanisms.

2. Correlation between gut dysbiosis and neuroinflammation in hypertension Gut microbiota

Gastrointestinal (GI) tract is a major interface between the host and external factors such as food, medication, alcohol and pathogens [161]. Mucosal surfaces of the GI tract are colonized by diverse microorganisms (bacteria, viruses and eukaryotes) collectively known as the ‘gut microbiota’, suggested to play significant roles in health and disease [161]. Gut microbiota is individually shaped by numerous factors such as the genetics, gender, age, IS, diet, drugs etc. [162]. The healthy adult gut microbiota consists of more than 100 trillion microorganisms [161], and Bacteroidetes and Firmicutes are the most abundant phyla [163].

Microbial colonization of the GI tract starts before birth and continues with the transfer of microorganisms during the delivery and breast-feeding, shaping the development of the host IS [164]. Gut microbiota has many beneficial functions in the body. It is involved in the conversion of food and indigestible components such as plant polysaccharides into nutrients such as short chain fatty acids (SCFA), which predominantly consist of acetic acid, butyric acid, and propionic acid, for the more efficient absorption and utilization by the host [165]; development of the intestinal epithelium and protection against tissue injury [166, 167]; regulation of fat storage [168]; modulation of enteric nervous system (ENS) [169]; development and maintenance of host IS [164]; and shape CNS development and function [170172].

Possible association between gut dysbiosis and neuroinflammatory responses in hypertension

Although components of the microbiota are often referred as commensals, interaction between microbiota and the host includes mutualistic, parasitic and commensal relationships [173]. Mounting evidence indicates that any alteration in the balance between gut microbiota and mucosal IS resulting from diet, medication, pathogens etc., results in changes in balance and composition of intestinal commensal communities i.e. gut dysbiosis, which has been associated with a plethora of diseases and conditions such as the inflammatory bowel disease, diabetes, obesity, arthritis, neurodegenerative diseases and HTN, among others. [146, 174, 175].

In recent years the concept of the “gut-brain axis” has been introduced. Communication between the gut and the brain is bidirectional and includes neural pathways via the ENS, autonomic and spinal nerves, and humoral pathways via cytokines, hormones, and various neuropeptide signaling molecules as well as metabolic factors such as the SCFA and tryptophan [176, 177]. Evidence suggests a role for gut dysbiosis in several neuroinflammatory diseases [178181]. Recently, gut dysbiosis has been reported in HTN [146, 182189] and played an important role in BP control. Administration of SCFA causes vasodilation in both rodents and humans [190193], and gut bacteria and/or their metabolites may affect the regulation of the circulatory system via ENS and the afferent sensory feedback to the brain that may modulate the activity of the cardioregulatory brain regions [194, 195]. In rodent models of HTN characterized by neuroinflammation, elevated BP is linked to increased gut permeability, heightened inflammatory state and increased sympathetic neuronal communication between the PVN and the gut [153, 188]. On the other hand, inhibition of microglial activation in the PVN decreased sympathetic activity, impacted selective gut microbial communities, attenuated gut wall pathology, and lowered BP in the Ang II-dependent rodent HTN [153]. Additionally, gut dysbosis may directly affect the IS and vice versa [196199], while SCFA can dampen glial inflammatory responses [200203] and in this way may be regulating BP [195, 204]. However, whether gut dysbiosis is a cause or a consequence of HTN, as well as the precise mechanisms that lead to development of gut dysbiosis in HTN and the impact of established gut dysbiosis on BP and neuroinflammation in HTN, is not known. Recent studies offered a clue to the possible mechanisms of gut dysbiosis-linked HTN, as the gut microbiota transplant from SHR to normotensive rats increased BP and sympathetic drive via activation of both systemic and central IS responses [197, 205, 206], suggesting that the microbiota-microglial crosstalk is important in regulation of BP.

Gut microbiota and microglia crosstalk

The gut microbiota can significantly affect microglia from before birth and throughout the adulthood, as the microbial-derived metabolites and neurotransmitters can regulate the inflammatory responses mediated by microglia in the CNS [200203, 207209]. The gut microbiota has been shown to affect microglia through microbial-derived metabolites and neurotransmitters [200203, 207209]. A study investigating interactions between the gut microbiota and microglia showed that germ-free mice displayed microglial immaturity and defects in microglial proportions, which are normalized by treatment with microbial SCFA [210]. In addition, an anti-inflammatory SCFA butyrate has been shown to reduce lipopolysaccharide (LPS)-induced responses in rat primary microglia [200203, 211] and dampen inflammatory mediators in SHR primary astrocytes [200]. Since animal [146, 212] and human studies [213] have shown a decrease in butyrate-producing bacterial population in HTN with reduced plasma butyrate levels [195, 213], which may contribute to microglial activation we see in HTN.

Gut dysbiosis may also contribute to neuroinflammatory responses by increasing both gut and BBB permeability thus allowing infiltration of peripheral immune cells and PIC from systemic circulation into the brain [214216]. Indeed, in rodent models of HTN, activation of peripheral IS is linked to microglial activation [85, 92], while elevated BP is marked by increased gut permeability, heightened inflammation, and increased sympathetic neuronal communication between the PVN and the gut [153, 188].

The vagus nerve is a crucial physiological component of gut-brain axis as well as the immune function [217, 218]. Efferent vagal branches innervate visceral tissues such as the liver, GI tract, thymus and spleen involved in regulation of immune and metabolic responses, while the afferent vagal branches report on the inflammatory and metabolic status in the periphery via their sensory afferent feedback to the NTS [219, 220]. In the gut, the vagus can sense the inflammatory mediators and relay these signals to the brain, and this may in turn activate vagal efferent responses to attenuate inflammation [221] in a process termed the vagal anti-inflammatory reflex. For example, intraperitoneal injection of microbial-derived LPS will cause a release of IL-1β which activates the vagal afferents in the periphery and, via the NTS, modulates efferent autonomic responses in order to curtail the inflammation in the periphery [222]. LPS has also been shown to cause glia-mediated neuroinflammation, while electric vagal nerve stimulation can dampen the LPS-induced activation of microglia and PIC production in brain [223]. In addition, gut bacterial-derived metabolites and neuroactive factors can directly activate the vagal afferents via receptors located on vagal nerve endings and on the cell soma [219, 224]. These studies support the role of the vagus nerve in gut-brain axis and regulation of neuroinflammatory responses.

3. Hydrogen sulfide in hypertension and neuroinflammation

Hydrogen sulfide (H2S), traditionally known as a toxic gas with the smell of rotten eggs, is an endogenously produced signaling molecule in bacteria, plants, and animals including the mammals [225, 226]. Studies in animals and humans have shown the H2S to be involved in diverse physiological and pathophysiological processes, including immune conditions, HTN, neurodegenerative diseases and metabolic disorders [227].

Hydrogen sulfide in human and rodent hypertension

H2S is considered an endogenous vasoactive factor [222] that can elicit concentration-dependent vasorelaxation [223226]. Several mechanisms have been proposed to contribute to this effect. H2S can induce vascular smooth muscle relaxation through the activation of KATP channels leading to membrane hyperpolarization [228]. Moreover, H2S has been suggested to dilate blood vessels [229] by promoting the release of NO [230], while NO can also enhance H2S production from vascular tissues [228]. H2S can also produce vasodilation by increasing intracellular cyclic guanosine monophosphate levels via inhibition of the phosphodiesterase activity [231]. In addition, H2S can reportedly attenuate vascular inflammation [232], reduce ROS production [233], and inhibit the synthesis and release of renin [234] as well as lower ACE activity [235]. In this way, H2S can produce BP-lowering effects by modulation of neural and immune activity. However, the full extent of H2S BP-lowering effects remains unknown.

Mounting evidence suggests that H2S may play a role in development of rodent and human HTN (Table 2). In the SHR, low plasma H2S and diminished activity and expression of H2S-producing enzyme cystathionine γ-lyase (CSE) has been reported, whereas intraperitoneal administration of sodium hydrosulfide (NaHS), an H2S donor, alleviated HTN in the SHR by upregulation of the diminished H2S synthase activity [236, 237] by increasing renal H2S production and NO bioavailability and inhibiting renal RAS [238]. In the 2K1C (2kidney-1clip) hypertensive rats, plasma H2S levels, CSE expression levels and BP were all normalized by the NaHS treatment [239, 240]. In Dahl salt-sensitive rats, high salt intake reduced H2S in serum and the kidney and inhibited renal expression of H2S-producing enzyme cystathionine β-synthase (CBS), while administration of NaHS normalized H2S and inhibited renal RAS, thus preventing salt-sensitive HTN in Dahl rats [241]. In rats with hypoxic pulmonary HTN, low plasma H2S levels were linked with decreased CSE activity in the lung, which was reversed by administration of an H2S donor [242]. In the CSE knock-out mice, the developing HTN is linked with low serum H2S levels, and injection of NaHS lowered BP in the 10-week-old CSE knock-out mice [243]. Chronic Ang II infusion increased BP, lowered plasma H2S levels and downregulated kidney expression of CBS and CSE in mice, which were all reversed upon treatment with an H2S donor GYY4137 [244]. Chronic administration of CSE and CBS enzyme inhibitors to normotensive rats resulted in a decrease in urinary excretion rate of sulfate, which is considered an indicator for endogenous H2S production. Decrease in the excretion rate of sulfate was also associated with increased mean arterial pressure following dual enzyme inhibition [245].

Table 2.

Plasma/serum hydrogen sulfide levels in animals and patients with hypertension.

Animal studies Species H2S levels Reference
Animal model SHR rat Low [236, 237]
Diabetic SHR rat Low [252]
2K1C rat Low [239, 240]
Dahl rat Low [241]
Hypoxic pulmonary HTN rat Low [242]
Load induced HTN rat Low [251]
CSE knock-out mice Low [243]
Ang II induced HTN mice Low [244]
Human studies H2S levels Reference
Type of HTN Essential Low [240, 246, 247]
Portal Low [248]
Pulmonary Low [249]
Preeclampsia Low [250]
Lead-induced HTN Low [251]

Ang II: Angiotensin II; HTN: Hypertension; H2S: Hydrogen sulfide; CSE: cystathionine γ-lyase; SHR: Spontaneously hypertensive rat; 2K1C: Two-kidney one-clip

In hypertensive adults and children, plasma H2S levels [240, 246, 247] and expression of CSE were constantly low [240]. In addition, plasma H2S levels are also reportedly low in patients with portal HTN [248], pulmonary HTN [249], women with preeclampsia [250], and lead-induced HTN [251]. However, although H2S are generally low in HTN, underlying mechanism(s) of this remain to be fully revealed.

Hydrogen sulfide in neuroinflammation

H2S is a neuromodulatory and neuroprotective molecule [253]. As it can freely cross the cell membrane, it can regulate various intracellular signaling processes [253]. H2S can act as an endogenous neuromodulator via enhancing NMDA receptor-mediated responses [254, 255] and by modulating intracellular pH [256] and Ca2+ levels in neurons [257], astrocytes [258] and microglia [259]. In addition, H2S donors have shown beneficial effects on glia-mediated neuroinflammation in various neurodegenerative conditions [260264] and HTN [108, 265] via their anti-inflammatory [108, 260, 261, 263, 264], anti-oxidant [108, 265] and anti-apoptotic effects [261]. Thus, at physiological levels, H2S exerts largely beneficial effects on the brain. However, if the brain concentration of H2S deviates from its physiological range, this can lead to pathology and/or toxicity [229, 274, 276].

Restoration of diminished H2S levels or the H2S-producing enzyme activity in the brain is beneficial in various neurodegenerative conditions [264, 266268]. For example, decreased CBS expression has been reported in the rostral ventrolateral medulla (RVLM) of SHR, and microinjection of NaHS or CBS agonist S-adenosyl-l-methionine into the RVLM decreased BP by decreasing the NADPH oxidase activity. On the contrary, microinjection of CBS inhibitor hydroxylamine hydrochloride increased BP [265]. One study suggested that decreased CBS expression and H2S production in the RVLM of SHR might be a compensatory mechanism of HTN involving the H2S-NO cross talk [269], but this remains to be confirmed. In addition, decreased CBS expression and H2S production was observed in the PVN of high salt-fed hypertensive rats, and microinjection of H2S donor GYY4137 into the PVN restored CBS expression and H2S production while reducing the sympathetic activity and BP in these rats [108]. A decreased CBS activity and H2S production were also reported in astrocytes of stroke-prone SHR compared with the SHR [270]. H2S has also been shown to protect BBB integrity following experimental cerebral ischemia by suppressing local inflammation and ROS generation [271], or during cardiac arrest by suppressing matrix metalloproteinase-9 and vascular endothelial growth factor expression while increasing Ang-I expression and tight junction expression [272, 273], thus preventing inflammatory factors from crossing the BBB and causing neuroinflammation. Taken together, these findings indicate that disruption of endogenous H2S generation in the brain may be involved in pathogenesis of HTN. However, there remains a need to understand the precise underlying mechanisms causing this disruption.

Hydrogen sulfide in the gut

Biosynthesis of hydrogen sulfide in the gut

H2S in the GI tract is produced through enzymatic reaction in host epithelial cells [274] and by the gut microbiota [275]. Enzymatically produced H2S is primarily derived from sulfur-containing amino acids methionine, homocysteine and cysteine [276] by CBS and CSE, and cysteine aminotransferase in conjunction with mercaptopyruvate sulfurtransferase (3-MST) [277, 278]. Expression of these enzymes differs in tissues. CBS is mainly expressed in the liver, colon and CNS, CSE is mainly found in smooth muscle cells in the cardiovascular system and in the GI tract, and 3-MST is expressed mainly in the brain [279, 280].

Several different genera of colonic bacteria are capable of producing the H2S. Gut bacteria produce H2S by enzymatic degradation in the large intestine while utilizing the sulfur-containing amino acids, inorganic sulfate, sulfited additives and sulfated polysaccharides such as sulfomucins [275]. In this way, Enterococci, Enterobacteria and Clostridia including Escherichia coli produce H2S by cysteine desulfhydrase activity [275]. Metabolism of sulfite by sulfite reductase is another mechanism of H2S production. Sulfate-reducing bacteria (SRB) are a heterogeneous group of microorganisms which use sulfate as an electron acceptor in the process of dissimilatory sulfate reduction resulting in the production of H2S [281, 282]. SRB may be divided into four groups based on rRNA sequence analysis: Gram-negative mesophilic SRB (within the delta subdivision of the Proteobacteria: Desulfobulbus, Desulfomicrobium, Desulfomonas, Desulfovibrio, Desulfobacter, Desulfobacterium, Desulfococcus, Desulfomonile, Desulfonema, Desulfosarcina); Gram-positive spore forming SRB (Desulfotomaculum); thermophilic bacterial SRB (Thermodesulfobacterium); and thermophilic archaeal SRB (Archaeoglobus) [283]. For sulfate reduction, SRB need exogenous electron donors, including hydrogen (H2), methanol, ethanol, acetate, lactate, propionate, butyrate and sugar [284]. A study with germ-free mice showed reduced plasma, cecum and colon H2S levels compared to the conventional mice, concluding that gut microbiota contributes to circulating and GI H2S levels. Moreover, absence of the gut microbiota in germ-free mice was associated with reduced CSE activity in several tissues including the cecum, colon, small intestine, kidney, liver, aorta, heart and brain, suggesting that gut microbiota may also influence CSE activity or expression in the host tissues [285].

Multiple biological roles of hydrogen sulfide in the gastrointestinal tract

In the GI tract, H2S is an important contributor to the maintenance of the mucosal barrier. For example, H2S can protect against gastric mucosal injury by increasing gastric mucosal blood flow and preventing leukocyte adherence to the endothelium of mesenteric vasculature [279], suggesting vascular and immune targets in the GI tract. H2S can protect mucosal integrity by inducing bicarbonate secretion, possibly via stimulating the capsaicin-sensitive afferent neurons resulting in NO and prostaglandin E2 release [286, 287], suggesting effects on the peripheral nervous system within the GI tract. Indeed, H2S has a neuromodulatory role in the gut by acting on the transient receptor potential vanilloid 1 receptors on primary afferent terminals in the GI tract [288], and causing excitation of the sensory nerves by activating Ca(v)3.2 T-type Ca(2+) channels considered to be protective in rats with colitis [289]. H2S also acts as an energy source for the GI epithelial cells [290], in addition to its anti-oxidant role and a role in regulation of the cell cycle [291].

4. A potential role for hydrogen sulfide in gut-brain axis in hypertension

Evidence thus far indicates that (i) gut microbiota and neuroinflammation are involved in pathogenesis of HTN; (ii) bidirectional communication exists between gut and brain; and (iii) H2S levels are reduced in HTN, while studies to date have largely focused on the enzymatic production and activity in circulation and the host tissues. Thus, the contribution of gut-derived H2S in the pathogenesis of HTN is not known. The following questions remain unanswered: Is there a link between gut microbiota-derived H2S and HTN? What is the role of H2S-producing bacteria in HTN and how may the modulation of H2S-producing bacteria impact HTN? Can we alleviate neuroinflammation by exogenous H2S administration in HTN? Answers to these questions may aid in the development of dietary or pharmaceutical gut-brain targeted approaches to prevent or control high BP.

Hydrogen sulfide and gut dysbiosis in hypertension

Recently, experimental intracolonic administration of H2S donors lowered BP, suggesting gut derived-H2S may contribute to BP control [292, 293]. As mentioned before, gut dysbiosis is present in HTN; however, the evidence of contribution of the H2S-producing bacteria to the hypertensive phenotype is scarce. Desulfovibrio, the predominant SRB genus, shows higher abundance in patients with HTN [184, 185], while in the SHR the abundance of Desulfovibrio does not correlate with BP [294]. As discussed above, circulating levels of H2S are reportedly reduced in HTN including the SHR (Table 2). We measured fecal H2S using the modified methylene blue method [295], and our new preliminary data show that fecal H2S levels are also reduced in the SHR (Figure 1) [unpublished data]. Thus, fecal H2S levels could be an important and direct biomarker of H2S production by the gut bacteria as well as an indicator of circulating H2S in HTN.

Figure 1.

Figure 1.

A) Schematic representation of the currently available methods of hydrogen sulfide (H2S) detection in biological samples, including colorimetric methods [296298], gas chromatography [299, 300], high performance liquid chromatography (HPLC) [301303], headspace-gas chromatography-mass spectrometry (HS-GC-MS) [304], liquid chromatography-mass spectrometry (LC-MS/MS) [305307], polarographic sensors [308], fluorescent probes [309, 310], enzyme-linked immunosorbent assay (ELISA) [311, 312]. These detection methods have several limitations, including detection sensitivity, expense, sample type etc. [313, 314]. Accurate measurement of biologically active H2S is challenging since H2S is a volatile gas and exists in the body in various forms, including free, acid-labile, and sulfane sulfur bound [315]. B) Methylene blue method [295] was used for the measurement of fecal H2S in spontaneously hypertensive rats (SHR). Fecal H2S levels were significantly lower in the adult male SHR compared to the age- and sex-matched normotensive Wistar-Kyoto rats (SHR: 0±0.01703 Arbitrary Unit (AU) vs. WKY: 0.094±0.03385 AU, n=5 per strain; *p<0.05 by T-test, mean ± SEM).

Indirect but supporting evidence to our current findings are that plasma levels of sulfur-containing amino acids (taurine, serine, methionine and threonine) are reportedly decreased in patients with essential HTN [316]. This may have resulted from impairment in sulfur amino acid metabolism; however, the underlying mechanism is not yet known. Interestingly, Desulfovibrio is elevated in a small group of hypertensive patients [185] and patients with inflammatory GI disorders such as ulcerative colitis and irritable bowel disease [317319] and neurodegenerative diseases [320322]. This increase in SRB Desulfovibrio may be related to inflammation of the gut present in the GI disorders [317319]. SRB can reduce disulfide bonds thereby causing mucin denaturation and microbial access to the mucus layer [323]. Thus, high non-physiologic levels of H2S could disrupt the gut barrier function and cause detrimental effects such as colonic pain, gastrointestinal discomfort, inflammatory bowel disease, colonic nociception, and colorectal cancer [324327]. Further studies employing genomic sequencing of SRB and investigation of fecal and host H2S levels in rodents and humans with HTN are needed to clarify this matter.

HTN-associated gut dysbiosis is characterized by an imbalance in specific microbial populations and their corresponding metabolites, specifically decreased acetate and butyrate-producing bacteria and increased lactate-producing bacteria [146]. SRB can utilize these organic compounds as electron donors for sulfate reduction [328]. In this way, NaHS can inhibit n-butyrate oxidation by inhibiting short chain acyl-CoA dehydrogenase, which is important in maintaining colonic mucosal integrity [329, 330]. In addition, reduced Desulfovibrio was beneficial in mice with phenylketonuria as it increased cecal levels of acetate, butyrate, and propionate while reducing plasma PIC levels, suggesting improved intestinal barrier function [331]. Moreover, SRB competition for organic compounds such as lactate can stimulate sulphide formation by SRB [332]. Interestingly, both SRB and lactate-producing bacteria are increased in HTN. Consequently, reducing the abundance of SRB may be beneficial for the protection of gut-integrity. However, there remains a need to clarify cause-effect relationships between SRB and HTN.

Supplementation and dietary modification as anti-hypertensive treatment options

Experimental and clinical studies suggest that H2S donors or dietary supplementation with sulfur-containing amino acids, the precursors of H2S, may be beneficial for HTN and neuroinflammation. For instance, injection of H2S donors such as NaHS and STS reduces BP in Ang II-dependent HTN and the SHR [233, 252, 333, 334], while D-cysteine or L-cysteine supplementation via gastric gavage results in decreased BP and reduces kidney damage induced by high-salt diet in the SHR [335]. Moreover, garlic contains a number of active sulfur compounds and has been shown to reduce BP in hypertensive individuals [336, 337], which may be partly mediated via increase in H2S production [338]. In pre-hypertensive and borderline HTN humans, chronic taurine supplementation, a sulfur-containing amino acid, has been shown to lower BP, which was correlated with an increase in plasma H2S levels and upregulated vascular H2S-producing enzymes [339, 340]. Taurine supplementation has also been shown to reduce glial activation and levels of inflammatory mediators while increasing H2S levels and CBS expression in rats with intracerebral hemorrhage [341, 342]. Methionine is another sulfur-containing essential amino acid. However, the effects of dietary methionine intake on BP remain tenuous. Some studies linked elevated dietary methionine intake with increase in BP in normotensive rats [343345], whereas it reduced BP in the SHR [346] and DOCA rats [347]. Methionine overloading has also been shown to attenuate development of HTN in female SHR [348]. On the other hand, restriction of sulfur amino acids methionine and cysteine can enhance endogenous CSE activity resulting in increased H2S production and protection from hepatic ischemia reperfusion injury in mice [349]. Thus, the effects of dietary sulfur-containing amino acids may differ in physiological and pathological conditions and could involve the interaction of various compensatory mechanisms. These studies predominantly focus on the enzymatic production of H2S by the host, while the gut microbiota can also contribute to circulating H2S levels. Absence of the gut microbiota in germ-free mice was associated with reduced CSE activity in several tissues including the cecum, colon, small intestine, kidney, liver, aorta, heart and brain, suggesting that gut microbiota may also influence CSE activity and/or expression in host tissues [285]. H2S production in the gut also depends on bacterial substrates. For instance, sulfur-amino acid cysteine is reportedly more powerful and more reliable in stimulation of H2S production compared to the inorganic sulfur [350, 351]. Thus, these factors should also be considered while attempting to manage H2S production in the host.

In addition to the influence of dietary sulfur-containing amino acids on H2S production, other types of diets can also affect this process. For instance, in the high fat Western diet-fed rat model, L-cysteine-induced aortic vasorelaxation is significantly inhibited [352]. Moreover, diets high in protein can increase SRB abundance and consequently the fecal sulfide levels [353]. Although diet can impact endogenous H2S production and H2S-generating bacteria, the dietary requirements and bacterial manipulations needed for restoring the H2S deficiency in HTN are currently unknown. Thus, the potential contribution of H2S-producing bacteria in dietary H2S production in HTN needs to be clarified in future studies.

4. Conclusion and future prospects

With reported neuromodulatory, anti-inflammatory and anti-hypertensive properties, both host- and gut bacterial-derived H2S may be affected in HTN. Low plasma H2S levels are reported in human hypertensives and rodent models of HTN, and our new preliminary data show that this is also reflected in reduced fecal H2S in the SHR. However, contribution of gut dysbiosis and gut microbiota-derived H2S to the pathology of HTN remains unknown. Future studies should aim to provide detailed understanding of how the levels of H2S may be modified with dietary manipulations including application of exogenous H2S donors in order to manipulate bacterial and host production of H2S as a potential therapy for HTN.

Abbreviations:

Ang

Angiotensin

H2S

Hydrogen sulfide

PNA

Parasympathetic nerve activity

SNA

Sympathetic nerve activity

ROS

Reactive oxygen species

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Conflict of Interest

The authors declare no conflict of interest.

5. References

  • [1].Kjeldsen SE, Hypertension and cardiovascular risk: General aspects, Pharmacol Res 129 (2018) 95–99. [DOI] [PubMed] [Google Scholar]
  • [2].Franklin SS, Wong ND, Hypertension and cardiovascular disease: contributions of the framingham heart study, Glob Heart 8(1) (2013) 49–57. [DOI] [PubMed] [Google Scholar]
  • [3].Benjamin EJ, Muntner P, Alonso A, Bittencourt MS, Callaway CW, Carson AP, Chamberlain AM, Chang AR, Cheng S, Das SR, Delling FN, Djousse L, Elkind MSV, Ferguson JF, Fornage M, Jordan LC, Khan SS, Kissela BM, Knutson KL, Kwan TW, Lackland DT, Lewis TT, Lichtman JH, Longenecker CT, Loop MS, Lutsey PL, Martin SS, Matsushita K, Moran AE, Mussolino ME, O’Flaherty M, Pandey A, Perak AM, Rosamond WD, Roth GA, Sampson UKA, Satou GM, Schroeder EB, Shah SH, Spartano NL, Stokes A, Tirschwell DL, Tsao CW, Turakhia MP, VanWagner LB, Wilkins JT, Wong SS, Virani SS, E. American Heart Association Council on, C. Prevention Statistics, S. Stroke Statistics, Heart Disease and Stroke Statistics-2019 Update: A Report From the American Heart Association, Circulation 139(10) (2019) e56–e528. [DOI] [PubMed] [Google Scholar]
  • [4].Achelrod D, Wenzel U, Frey S, Systematic review and meta-analysis of the prevalence of resistant hypertension in treated hypertensive populations, Am J Hypertens 28(3) (2015) 355–61. [DOI] [PubMed] [Google Scholar]
  • [5].Yang T, Richards EM, Pepine CJ, Raizada MK, The gut microbiota and the brain-gut-kidney axis in hypertension and chronic kidney disease, Nat Rev Nephrol 14(7) (2018) 442–456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Esler M, Lambert E, Schlaich M, Point: Chronic activation of the sympathetic nervous system is the dominant contributor to systemic hypertension, J Appl Physiol (1985) 109(6) (2010) 1996–8; discussion 2016. [DOI] [PubMed] [Google Scholar]
  • [7].Shen XZ, Li Y, Li L, Shah KH, Bernstein KE, Lyden P, Shi P, Microglia participate in neurogenic regulation of hypertension, Hypertension 66(2) (2015) 309–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [8].Mowry FE, Biancardi VC, Neuroinflammation in hypertension: the renin-angiotensin system versus pro-resolution pathways, Pharmacol Res 144 (2019) 279–291. [DOI] [PubMed] [Google Scholar]
  • [9].Paton JF, Waki H, Is neurogenic hypertension related to vascular inflammation of the brainstem?, Neurosci Biobehav Rev 33(2) (2009) 89–94. [DOI] [PubMed] [Google Scholar]
  • [10].Winklewski PJ, Radkowski M, Wszedybyl-Winklewska M, Demkow U, Brain inflammation and hypertension: the chicken or the egg?, J Neuroinflammation 12 (2015) 85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].DiSabato DJ, Quan N, Godbout JP, Neuroinflammation: the devil is in the details, J Neurochem 139 Suppl 2 (2016) 136–153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Carnevale D, Mascio G, Ajmone-Cat MA, D’Andrea I, Cifelli G, Madonna M, Cocozza G, Frati A, Carullo P, Carnevale L, Alleva E, Branchi I, Lembo G, Minghetti L, Role of neuroinflammation in hypertension-induced brain amyloid pathology, Neurobiol Aging 33(1) (2012) 205 e19–29. [DOI] [PubMed] [Google Scholar]
  • [13].Tsukamoto T, Kitano Y, Kuno S, Blood pressure fluctuation and hypertension in patients with Parkinson’s disease, Brain Behav 3(6) (2013) 710–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Labandeira-Garcia JL, Rodriguez-Pallares J, Villar-Cheda B, Rodriguez-Perez AI, Garrido-Gil P, Guerra MJ, Aging, Angiotensin system and dopaminergic degeneration in the substantia nigra, Aging Dis 2(3) (2011) 257–74. [PMC free article] [PubMed] [Google Scholar]
  • [15].Tamura K, Umemura S, Nyui N, Yamakawa T, Yamaguchi S, Ishigami T, Tanaka S, Tanimoto K, Takagi N, Sekihara H, Murakami K, Ishii M, Tissue-specific regulation of angiotensinogen gene expression in spontaneously hypertensive rats, Hypertension 27(6) (1996) 1216–23. [DOI] [PubMed] [Google Scholar]
  • [16].Hermann K, McDonald W, Unger T, Lang RE, Ganten D, Angiotensin biosynthesis and concentrations in brain of normotensive and hypertensive rats, J Physiol (Paris) 79(6) (1984) 471–80. [PubMed] [Google Scholar]
  • [17].Szczepanska-Sadowska E, Paczwa P, Lon S, Ganten D, Increased pressor function of central vasopressinergic system in hypertensive renin transgenic rats, J Hypertens 16(10) (1998) 1505–14. [DOI] [PubMed] [Google Scholar]
  • [18].Senanayake PD, Moriguchi A, Kumagai H, Ganten D, Ferrario CM, Brosnihan KB, Increased expression of angiotensin peptides in the brain of transgenic hypertensive rats, Peptides 15(5) (1994) 919–26. [DOI] [PubMed] [Google Scholar]
  • [19].Teruya H, Muratani H, Takishita S, Sesoko S, Matayoshi R, Fukiyama K, Brain angiotensin II contributes to the development of hypertension in Dahl-Iwai salt-sensitive rats, J Hypertens 13(8) (1995) 883–90. [DOI] [PubMed] [Google Scholar]
  • [20].Zhao X, White R, Huang BS, Van Huysse J, Leenen FH, High salt intake and the brain renin--angiotensin system in Dahl salt-sensitive rats, J Hypertens 19(1) (2001) 89–98. [DOI] [PubMed] [Google Scholar]
  • [21].Wang JM, Veerasingham SJ, Tan J, Leenen FH, Effects of high salt intake on brain AT1 receptor densities in Dahl rats, Am J Physiol Heart Circ Physiol 285(5) (2003) H1949–55. [DOI] [PubMed] [Google Scholar]
  • [22].Park CG, Leenen FH, Effects of centrally administered losartan on deoxycorticosterone-salt hypertension rats, J Korean Med Sci 16(5) (2001) 553–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [23].Itaya Y, Suzuki H, Matsukawa S, Kondo K, Saruta T, Central renin-angiotensin system and the pathogenesis of DOCA-salt hypertension in rats, Am J Physiol 251(2 Pt 2) (1986) H261–8. [DOI] [PubMed] [Google Scholar]
  • [24].Schenk J, McNeill JH, The pathogenesis of DOCA-salt hypertension, J Pharmacol Toxicol Methods 27(3) (1992) 161–70. [DOI] [PubMed] [Google Scholar]
  • [25].Faber JE, Brody MJ, Central nervous system action of angiotensin during onset of renal hypertension in awake rats, Am J Physiol 247(3 Pt 2) (1984) H349–60. [DOI] [PubMed] [Google Scholar]
  • [26].Sun HJ, Li P, Chen WW, Xiong XQ, Han Y, Angiotensin II and angiotensin-(1–7) in paraventricular nucleus modulate cardiac sympathetic afferent reflex in renovascular hypertensive rats, PLoS One 7(12) (2012) e52557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Li P, Zhang F, Sun HJ, Zhang F, Han Y, Angiotensin-(1–7) enhances the effects of angiotensin II on the cardiac sympathetic afferent reflex and sympathetic activity in rostral ventrolateral medulla in renovascular hypertensive rats, J Am Soc Hypertens 9(11) (2015) 865–77. [DOI] [PubMed] [Google Scholar]
  • [28].Chen AD, Zhang SJ, Yuan N, Xu Y, De W, Gao XY, Zhu GQ, Angiotensin AT1 receptors in paraventricular nucleus contribute to sympathetic activation and enhanced cardiac sympathetic afferent reflex in renovascular hypertensive rats, Exp Physiol 96(2) (2011) 94–103. [DOI] [PubMed] [Google Scholar]
  • [29].Ganten D, Hutchinson JS, Schelling P, The intrinsic brain iso-renin--angiotensin system in the rat: its possible role in central mechanisms of blood pressure regulation, Clin Sci Mol Med Suppl 2 (1975) 265s–268s. [DOI] [PubMed] [Google Scholar]
  • [30].Suzuki H, Ferrario CM, Speth RC, Brosnihan KB, Smeby RR, de Silva P, Alterations in plasma and cerebrospinal fluid norepinephrine and angiotensin II during the development of renal hypertension in conscious dogs, Hypertension 5(2 Pt 2) (1983) I139–48. [DOI] [PubMed] [Google Scholar]
  • [31].Kawamura M, Kawano Y, Yoshida K, Imanishi M, Akabane S, Matsushima Y, Kuramochi M, Shimamoto K, Ito K, Omae T, Alterations in cerebrospinal fluid angiotensin II by sodium intake in patients with essential hypertension, Clin Sci (Lond) 77(4) (1989) 389–94. [DOI] [PubMed] [Google Scholar]
  • [32].McKinley MJ, Albiston AL, Allen AM, Mathai ML, May CN, McAllen RM, Oldfield BJ, Mendelsohn FA, Chai SY, The brain renin-angiotensin system: location and physiological roles, Int J Biochem Cell Biol 35(6) (2003) 901–18. [DOI] [PubMed] [Google Scholar]
  • [33].Hermann K, Phillips MI, Hilgenfeldt U, Raizada MK, Biosynthesis of angiotensinogen and angiotensins by brain cells in primary culture, J Neurochem 51(2) (1988) 398–405. [DOI] [PubMed] [Google Scholar]
  • [34].Stornetta RL, Hawelu-Johnson CL, Guyenet PG, Lynch KR, Astrocytes synthesize angiotensinogen in brain, Science 242(4884) (1988) 1444–6. [DOI] [PubMed] [Google Scholar]
  • [35].Llorens-Cortes C, Mendelsohn FA, Organisation and functional role of the brain angiotensin system, J Renin Angiotensin Aldosterone Syst 3 Suppl 1 (2002) S39–48. [DOI] [PubMed] [Google Scholar]
  • [36].Gorbea-Oppliger VJ, Fink GD, Cerebroventricular injection of angiotensin II antagonist: effects on blood pressure responses to central and systemic angiotensin II, J Pharmacol Exp Ther 273(2) (1995) 611–6. [PubMed] [Google Scholar]
  • [37].Jancovski N, Bassi JK, Carter DA, Choong YT, Connelly A, Nguyen TP, Chen D, Lukoshkova EV, Menuet C, Head GA, Allen AM, Stimulation of angiotensin type 1A receptors on catecholaminergic cells contributes to angiotensin-dependent hypertension, Hypertension 62(5) (2013) 866–71. [DOI] [PubMed] [Google Scholar]
  • [38].Northcott CA, Watts S, Chen Y, Morris M, Chen A, Haywood JR, Adenoviral inhibition of AT1a receptors in the paraventricular nucleus inhibits acute increases in mean arterial blood pressure in the rat, Am J Physiol Regul Integr Comp Physiol 299(5) (2010) R1202–11. [DOI] [PubMed] [Google Scholar]
  • [39].Fan ZD, Zhang L, Shi Z, Gan XB, Gao XY, Zhu GQ, Artificial microRNA interference targeting AT(1a) receptors in paraventricular nucleus attenuates hypertension in rats, Gene Ther 19(8) (2012) 810–7. [DOI] [PubMed] [Google Scholar]
  • [40].Yang H, Raizada MK, Role of phosphatidylinositol 3-kinase in angiotensin II regulation of norepinephrine neuromodulation in brain neurons of the spontaneously hypertensive rat, J Neurosci 19(7) (1999) 2413–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Buttler L, Ribeiro IM, Ferreira-Neto HC, Antunes VR, Angiotensin II acting on PVN induces sympathoexcitation and pressor responses via the PI3K-dependent pathway, Auton Neurosci 198 (2016) 54–8. [DOI] [PubMed] [Google Scholar]
  • [42].Zhong MK, Gao J, Zhang F, Xu B, Fan ZD, Wang W, Zhu GQ, Reactive oxygen species in rostral ventrolateral medulla modulate cardiac sympathetic afferent reflex in rats, Acta Physiol (Oxf) 197(4) (2009) 297–304. [DOI] [PubMed] [Google Scholar]
  • [43].Li P, Zhang F, Zhou YB, Cui BP, Han Y, Superoxide anions modulate the effects of angiotensin-(1–7) in the rostral ventrolateral medulla on cardiac sympathetic afferent reflex and sympathetic activity in rats, Neuroscience 223 (2012) 388–98. [DOI] [PubMed] [Google Scholar]
  • [44].Zhang Y, Yu Y, Zhang F, Zhong MK, Shi Z, Gao XY, Wang W, Zhu GQ, NAD(P)H oxidase in paraventricular nucleus contributes to the effect of angiotensin II on cardiac sympathetic afferent reflex, Brain Res 1082(1) (2006) 132–41. [DOI] [PubMed] [Google Scholar]
  • [45].Gao L, Wang W, Li YL, Schultz HD, Liu D, Cornish KG, Zucker IH, Sympathoexcitation by central ANG II: roles for AT1 receptor upregulation and NAD(P)H oxidase in RVLM, Am J Physiol Heart Circ Physiol 288(5) (2005) H2271–9. [DOI] [PubMed] [Google Scholar]
  • [46].Wang G, Anrather J, Huang J, Speth RC, Pickel VM, Iadecola C, NADPH oxidase contributes to angiotensin II signaling in the nucleus tractus solitarius, J Neurosci 24(24) (2004) 5516–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Nakamura K, Shimizu T, Yanagita T, Nemoto T, Taniuchi K, Shimizu S, Dimitriadis F, Yawata T, Higashi Y, Ueba T, Saito M, Angiotensin II acting on brain AT1 receptors induces adrenaline secretion and pressor responses in the rat, Sci Rep 4 (2014) 7248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [48].Stadler T, Veltmar A, Qadri F, Unger T, Angiotensin II evokes noradrenaline release from the paraventricular nucleus in conscious rats, Brain Res 569(1) (1992) 117–22. [DOI] [PubMed] [Google Scholar]
  • [49].Qadri F, Badoer E, Stadler T, Unger T, Angiotensin II-induced noradrenaline release from anterior hypothalamus in conscious rats: a brain microdialysis study, Brain Res 563(1–2) (1991) 137–41. [DOI] [PubMed] [Google Scholar]
  • [50].Brown DC, Steward LJ, Ge J, Barnes NM, Ability of angiotensin II to modulate striatal dopamine release via the AT1 receptor in vitro and in vivo, Br J Pharmacol 118(2) (1996) 414–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Jenkins TA, Allen AM, Chai SY, Mendelsohn FA, Interactions of angiotensin II with central catecholamines, Clin Exp Hypertens 17(1–2) (1995) 267–80. [DOI] [PubMed] [Google Scholar]
  • [52].Jenkins TA, Allen AM, Chai SY, MacGregor DP, Paxinos G, Mendelsohn FA, Interactions of angiotensin II with central dopamine, Adv Exp Med Biol 396 (1996) 93–103. [DOI] [PubMed] [Google Scholar]
  • [53].Pawlak R, Napiorkowska-Pawlak D, Takada Y, Urano T, Nagai N, Ihara H, Takada A, The differential effect of angiotensin II and angiotensin 1–7 on norepinephrine, epinephrine, and dopamine concentrations in rat hypothalamus: the involvement of angiotensin receptors, Brain Res Bull 54(6) (2001) 689–94. [DOI] [PubMed] [Google Scholar]
  • [54].Mendelsohn FA, Jenkins TA, Berkovic SF, Effects of angiotensin II on dopamine and serotonin turnover in the striatum of conscious rats, Brain Res 613(2) (1993) 221–9. [DOI] [PubMed] [Google Scholar]
  • [55].Nahmod VE, Finkielman S, Benarroch EE, Pirola CJ, Angiotensin regulates release and synthesis of serotonin in brain, Science 202(4372) (1978) 1091–3. [DOI] [PubMed] [Google Scholar]
  • [56].Li DP, Chen SR, Pan HL, Angiotensin II stimulates spinally projecting paraventricular neurons through presynaptic disinhibition, J Neurosci 23(12) (2003) 5041–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [57].Li DP, Pan HL, Angiotensin II attenuates synaptic GABA release and excites paraventricular-rostral ventrolateral medulla output neurons, J Pharmacol Exp Ther 313(3) (2005) 1035–45. [DOI] [PubMed] [Google Scholar]
  • [58].Legat L, Smolders I, Dupont AG, AT1 Receptor Mediated Hypertensive Response to Ang II in the Nucleus Tractus Solitarii of Normotensive Rats Involves NO Dependent Local GABA Release, Front Pharmacol 10 (2019) 460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Zhang Q, Yao F, O’Rourke ST, Qian SY, Sun C, Angiotensin II enhances GABA(B) receptor-mediated responses and expression in nucleus tractus solitarii of rats, Am J Physiol Heart Circ Physiol 297(5) (2009) H1837–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Yao F, Sumners C, O’Rourke ST, Sun C, Angiotensin II increases GABAB receptor expression in nucleus tractus solitarii of rats, Am J Physiol Heart Circ Physiol 294(6) (2008) H2712–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [61].Averill DB, Tsuchihashi T, Khosla MC, Ferrario CM, Losartan, nonpeptide angiotensin II-type 1 (AT1) receptor antagonist, attenuates pressor and sympathoexcitatory responses evoked by angiotensin II and L-glutamate in rostral ventrolateral medulla, Brain Res 665(2) (1994) 245–52. [DOI] [PubMed] [Google Scholar]
  • [62].Vieira AA, Colombari E, De Luca LA Jr., Colombari DS, De Paula PM, Menani JV, Importance of angiotensinergic mechanisms for the pressor response to l-glutamate into the rostral ventrolateral medulla, Brain Res 1322 (2010) 72–80. [DOI] [PubMed] [Google Scholar]
  • [63].Barnes JM, Barnes NM, Costall B, Horovitz ZP, Ironside JW, Naylor RJ, Williams TJ, Angiotensin II inhibits acetylcholine release from human temporal cortex: implications for cognition, Brain Res 507(2) (1990) 341–3. [DOI] [PubMed] [Google Scholar]
  • [64].Qu L, McQueeney AJ, Barnes KL, Presynaptic or postsynaptic location of receptors for angiotensin II and substance P in the medial solitary tract nucleus, J Neurophysiol 75(6) (1996) 2220–8. [DOI] [PubMed] [Google Scholar]
  • [65].Barnes KL, Diz DI, Ferrario CM, Functional interactions between angiotensin II and substance P in the dorsal medulla, Hypertension 17(6 Pt 2) (1991) 1121–6. [DOI] [PubMed] [Google Scholar]
  • [66].Diz DI, Fantz DL, Benter IF, Bosch SM, Acute depressor actions of angiotensin II in the nucleus of the solitary tract are mediated by substance P, Am J Physiol 273(1 Pt 2) (1997) R28–34. [DOI] [PubMed] [Google Scholar]
  • [67].Diz DI, Pirro NT, Differential actions of angiotensin II and angiotensin-(1–7) on transmitter release, Hypertension 19(2 Suppl) (1992) II41–8. [DOI] [PubMed] [Google Scholar]
  • [68].Paton JF, Lonergan T, Deuchars J, James PE, Kasparov S, Detection of angiotensin II mediated nitric oxide release within the nucleus of the solitary tract using electron-paramagnetic resonance (EPR) spectroscopy, Auton Neurosci 126–127 (2006) 193–201. [DOI] [PubMed] [Google Scholar]
  • [69].Paton JF, Deuchars J, Ahmad Z, Wong LF, Murphy D, Kasparov S, Adenoviral vector demonstrates that angiotensin II-induced depression of the cardiac baroreflex is mediated by endothelial nitric oxide synthase in the nucleus tractus solitarii of the rat, J Physiol 531(Pt 2) (2001) 445–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].Veltmar A, Culman J, Qadri F, Rascher W, Unger T, Involvement of adrenergic and angiotensinergic receptors in the paraventricular nucleus in the angiotensin II-induced vasopressin release, J Pharmacol Exp Ther 263(3) (1992) 1253–60. [PubMed] [Google Scholar]
  • [71].Hogarty DC, Speakman EA, Puig V, Phillips MI, The role of angiotensin, AT1 and AT2 receptors in the pressor, drinking and vasopressin responses to central angiotensin, Brain Res 586(2) (1992) 289–94. [DOI] [PubMed] [Google Scholar]
  • [72].Qadri F, Culman J, Veltmar A, Maas K, Rascher W, Unger T, Angiotensin II-induced vasopressin release is mediated through alpha-1 adrenoceptors and angiotensin II AT1 receptors in the supraoptic nucleus, J Pharmacol Exp Ther 267(2) (1993) 567–74. [PubMed] [Google Scholar]
  • [73].Qi J, Zhang DM, Suo YP, Song XA, Yu XJ, Elks C, Lin YX, Xu YY, Zang WJ, Zhu Z, Kang YM, Renin-angiotensin system modulates neurotransmitters in the paraventricular nucleus and contributes to angiotensin II-induced hypertensive response, Cardiovasc Toxicol 13(1) (2013) 48–54. [DOI] [PubMed] [Google Scholar]
  • [74].Li YF, Wang W, Mayhan WG, Patel KP, Angiotensin-mediated increase in renal sympathetic nerve discharge within the PVN: role of nitric oxide, Am J Physiol Regul Integr Comp Physiol 290(4) (2006) R1035–43. [DOI] [PubMed] [Google Scholar]
  • [75].Zhu GQ, Patel KP, Zucker IH, Wang W, Microinjection of ANG II into paraventricular nucleus enhances cardiac sympathetic afferent reflex in rats, Am J Physiol Heart Circ Physiol 282(6) (2002) H2039–45. [DOI] [PubMed] [Google Scholar]
  • [76].Zhu GQ, Gao L, Patel KP, Zucker IH, Wang W, ANG II in the paraventricular nucleus potentiates the cardiac sympathetic afferent reflex in rats with heart failure, J Appl Physiol (1985) 97(5) (2004) 1746–54. [DOI] [PubMed] [Google Scholar]
  • [77].Chen QH, Toney GM, Responses to GABA-A receptor blockade in the hypothalamic PVN are attenuated by local AT1 receptor antagonism, Am J Physiol Regul Integr Comp Physiol 285(5) (2003) R1231–9. [DOI] [PubMed] [Google Scholar]
  • [78].Gabor A, Leenen FH, Cardiovascular effects of angiotensin II and glutamate in the PVN of Dahl salt-sensitive rats, Brain Res 1447 (2012) 28–37. [DOI] [PubMed] [Google Scholar]
  • [79].Ma H, Chen SR, Chen H, Li L, Li DP, Zhou JJ, Pan HL, alpha2delta-1 Is Essential for Sympathetic Output and NMDA Receptor Activity Potentiated by Angiotensin II in the Hypothalamus, J Neurosci 38(28) (2018) 6388–6398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Waki H, Liu B, Miyake M, Katahira K, Murphy D, Kasparov S, Paton JF, Junctional adhesion molecule-1 is upregulated in spontaneously hypertensive rats: evidence for a prohypertensive role within the brain stem, Hypertension 49(6) (2007) 1321–7. [DOI] [PubMed] [Google Scholar]
  • [81].Shi P, Diez-Freire C, Jun JY, Qi Y, Katovich MJ, Li Q, Sriramula S, Francis J, Sumners C, Raizada MK, Brain microglial cytokines in neurogenic hypertension, Hypertension 56(2) (2010) 297–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [82].Shi Z, Jiang SJ, Wang GH, Xu AL, Guo L, Pro-inflammatory cytokines in paraventricular nucleus mediate the cardiac sympathetic afferent reflex in hypertension, Auton Neurosci 186 (2014) 54–61. [DOI] [PubMed] [Google Scholar]
  • [83].Qi J, Zhao XF, Yu XJ, Yi QY, Shi XL, Tan H, Fan XY, Gao HL, Yue LY, Feng ZP, Kang YM, Targeting Interleukin-1 beta to Suppress Sympathoexcitation in Hypothalamic Paraventricular Nucleus in Dahl Salt-Sensitive Hypertensive Rats, Cardiovasc Toxicol 16(3) (2016) 298–306. [DOI] [PubMed] [Google Scholar]
  • [84].Qi J, Yu XJ, Shi XL, Gao HL, Yi QY, Tan H, Fan XY, Zhang Y, Song XA, Cui W, Liu JJ, Kang YM, NF-kappaB Blockade in Hypothalamic Paraventricular Nucleus Inhibits High-Salt-Induced Hypertension Through NLRP3 and Caspase-1, Cardiovasc Toxicol 16(4) (2016) 345–54. [DOI] [PubMed] [Google Scholar]
  • [85].Santisteban MM, Ahmari N, Carvajal JM, Zingler MB, Qi Y, Kim S, Joseph J, Garcia-Pereira F, Johnson RD, Shenoy V, Raizada MK, Zubcevic J, Involvement of bone marrow cells and neuroinflammation in hypertension, Circ Res 117(2) (2015) 178–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [86].Agarwal D, Welsch MA, Keller JN, Francis J, Chronic exercise modulates RAS components and improves balance between pro- and anti-inflammatory cytokines in the brain of SHR, Basic Res Cardiol 106(6) (2011) 1069–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [87].Masson GS, Nair AR, Silva Soares PP, Michelini LC, Francis J, Aerobic training normalizes autonomic dysfunction, HMGB1 content, microglia activation and inflammation in hypothalamic paraventricular nucleus of SHR, Am J Physiol Heart Circ Physiol 309(7) (2015) H1115–22. [DOI] [PubMed] [Google Scholar]
  • [88].Masson GS, Costa TS, Yshii L, Fernandes DC, Soares PP, Laurindo FR, Scavone C, Michelini LC, Time-dependent effects of training on cardiovascular control in spontaneously hypertensive rats: role for brain oxidative stress and inflammation and baroreflex sensitivity, PLoS One 9(5) (2014) e94927. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].Dange RB, Agarwal D, Teruyama R, Francis J, Toll-like receptor 4 inhibition within the paraventricular nucleus attenuates blood pressure and inflammatory response in a genetic model of hypertension, J Neuroinflammation 12 (2015) 31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [90].Kishi T, Hirooka Y, Kimura Y, Ito K, Shimokawa H, Takeshita A, Increased reactive oxygen species in rostral ventrolateral medulla contribute to neural mechanisms of hypertension in stroke-prone spontaneously hypertensive rats, Circulation 109(19) (2004) 2357–62. [DOI] [PubMed] [Google Scholar]
  • [91].Koga Y, Hirooka Y, Araki S, Nozoe M, Kishi T, Sunagawa K, High salt intake enhances blood pressure increase during development of hypertension via oxidative stress in rostral ventrolateral medulla of spontaneously hypertensive rats, Hypertens Res 31(11) (2008) 2075–83. [DOI] [PubMed] [Google Scholar]
  • [92].Wu KL, Chan SH, Chan JY, Neuroinflammation and oxidative stress in rostral ventrolateral medulla contribute to neurogenic hypertension induced by systemic inflammation, J Neuroinflammation 9 (2012) 212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [93].Tan X, Jiao PL, Wang YK, Wu ZT, Zeng XR, Li ML, Wang WZ, The phosphoinositide-3 kinase signaling is involved in neuroinflammation in hypertensive rats, CNS Neurosci Ther 23(4) (2017) 350–359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [94].Cohen EM, Mohammed S, Kavurma M, Nedoboy PE, Cartland S, Farnham MMJ, Pilowsky PM, Microglia in the RVLM of SHR have reduced P2Y12R and CX3CR1 expression, shorter processes, and lower cell density, Auton Neurosci 216 (2019) 9–16. [DOI] [PubMed] [Google Scholar]
  • [95].Waki H, Gouraud SS, Maeda M, Paton JF, Gene expression profiles of major cytokines in the nucleus tractus solitarii of the spontaneously hypertensive rat, Auton Neurosci 142(1–2) (2008) 40–4. [DOI] [PubMed] [Google Scholar]
  • [96].Waki H, Gouraud SS, Maeda M, Paton JF, Evidence of specific inflammatory condition in nucleus tractus solitarii of spontaneously hypertensive rats, Exp Physiol 95(5) (2010) 595–600. [DOI] [PubMed] [Google Scholar]
  • [97].Waki H, Gouraud SS, Maeda M, Paton JF, Specific inflammatory condition in nucleus tractus solitarii of the SHR: novel insight for neurogenic hypertension?, Auton Neurosci 142(1–2) (2008) 25–31. [DOI] [PubMed] [Google Scholar]
  • [98].Yi QY, Qi J, Yu XJ, Li HB, Zhang Y, Su Q, Shi T, Zhang DM, Guo J, Feng ZP, Wang ML, Zhu GQ, Liu JJ, Shi XL, Kang YM, Paraventricular Nucleus Infusion of Epigallocatechin-3-O-Gallate Improves Renovascular Hypertension, Cardiovasc Toxicol 16(3) (2016) 276–85. [DOI] [PubMed] [Google Scholar]
  • [99].Li HB, Qin DN, Ma L, Miao YW, Zhang DM, Lu Y, Song XA, Zhu GQ, Kang YM, Chronic infusion of lisinopril into hypothalamic paraventricular nucleus modulates cytokines and attenuates oxidative stress in rostral ventrolateral medulla in hypertension, Toxicol Appl Pharmacol 279(2) (2014) 141–9. [DOI] [PubMed] [Google Scholar]
  • [100].Jun JY, Zubcevic J, Qi Y, Afzal A, Carvajal JM, Thinschmidt JS, Grant MB, Mocco J, Raizada MK, Brain-mediated dysregulation of the bone marrow activity in angiotensin II-induced hypertension, Hypertension 60(5) (2012) 1316–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [101].Cardinale JP, Sriramula S, Mariappan N, Agarwal D, Francis J, Angiotensin II-induced hypertension is modulated by nuclear factor-kappaBin the paraventricular nucleus, Hypertension 59(1) (2012) 113–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [102].Su Q, Qin DN, Wang FX, Ren J, Li HB, Zhang M, Yang Q, Miao YW, Yu XJ, Qi J, Zhu Z, Zhu GQ, Kang YM, Inhibition of reactive oxygen species in hypothalamic paraventricular nucleus attenuates the renin-angiotensin system and proinflammatory cytokines in hypertension, Toxicol Appl Pharmacol 276(2) (2014) 115–20. [DOI] [PubMed] [Google Scholar]
  • [103].Kang YM, Zhang DM, Yu XJ, Yang Q, Qi J, Su Q, Suo YP, Yue LY, Zhu GQ, Qin DN, Chronic infusion of enalaprilat into hypothalamic paraventricular nucleus attenuates angiotensin II-induced hypertension and cardiac hypertrophy by restoring neurotransmitters and cytokines, Toxicol Appl Pharmacol 274(3) (2014) 436–44. [DOI] [PubMed] [Google Scholar]
  • [104].Kang YM, Ma Y, Zheng JP, Elks C, Sriramula S, Yang ZM, Francis J, Brain nuclear factor-kappa B activation contributes to neurohumoral excitation in angiotensin II-induced hypertension, Cardiovasc Res 82(3) (2009) 503–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [105].Zhang M, Qin DN, Suo YP, Su Q, Li HB, Miao YW, Guo J, Feng ZP, Qi J, Gao HL, Mu JJ, Zhu GQ, Kang YM, Endogenous hydrogen peroxide in the hypothalamic paraventricular nucleus regulates neurohormonal excitation in high salt-induced hypertension, Toxicol Lett 235(3) (2015) 206–15. [DOI] [PubMed] [Google Scholar]
  • [106].Su Q, Liu JJ, Cui W, Shi XL, Guo J, Li HB, Huo CJ, Miao YW, Zhang M, Yang Q, Kang YM, Alpha lipoic acid supplementation attenuates reactive oxygen species in hypothalamic paraventricular nucleus and sympathoexcitation in high salt-induced hypertension, Toxicol Lett 241 (2016) 152–8. [DOI] [PubMed] [Google Scholar]
  • [107].Yu XJ, Miao YW, Li HB, Su Q, Liu KL, Fu LY, Hou YK, Shi XL, Li Y, Mu JJ, Chen WS, Cui W, Zhu GQ, Ebenezer PJ, Francis J, Kang YM, Blockade of Endogenous Angiotensin-(1–7) in Hypothalamic Paraventricular Nucleus Attenuates High Salt-Induced Sympathoexcitation and Hypertension, Neurosci Bull 35(1) (2019) 47–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [108].Liang YF, Zhang DD, Yu XJ, Gao HL, Liu KL, Qi J, Li HB, Yi QY, Chen WS, Cui W, Zhu GQ, Kang YM, Hydrogen sulfide in paraventricular nucleus attenuates blood pressure by regulating oxidative stress and inflammatory cytokines in high salt-induced hypertension, Toxicol Lett 270 (2017) 62–71. [DOI] [PubMed] [Google Scholar]
  • [109].Bai J, Yu XJ, Liu KL, Wang FF, Li HB, Shi XL, Zhang Y, Huo CJ, Li X, Gao HL, Qi J, Liu JJ, Zhu GQ, Chen WS, Cui W, Kang YM, Tert-butylhydroquinone attenuates oxidative stress and inflammation in hypothalamic paraventricular nucleus in high salt-induced hypertension, Toxicol Lett 281 (2017) 1–9. [DOI] [PubMed] [Google Scholar]
  • [110].Zhang DD, Liang YF, Qi J, Kang KB, Yu XJ, Gao HL, Liu KL, Chen YM, Shi XL, Xin GR, Fu LY, Kang YM, Cui W, Carbon Monoxide Attenuates High Salt-Induced Hypertension While Reducing Pro-inflammatory Cytokines and Oxidative Stress in the Paraventricular Nucleus, Cardiovasc Toxicol (2019). [DOI] [PubMed] [Google Scholar]
  • [111].Huang YP, Jin HY, Yu HP, Inhibitory effects of alpha-lipoic acid on oxidative stress in the rostral ventrolateral medulla in rats with salt-induced hypertension, Int J Mol Med 39(2) (2017) 430–436. [DOI] [PubMed] [Google Scholar]
  • [112].Li HB, Huo CJ, Su Q, Li X, Bai J, Zhu GQ, Kang YM, Exercise Training Attenuates Proinflammatory Cytokines, Oxidative Stress and Modulates Neurotransmitters in the Rostral Ventrolateral Medulla of Salt-Induced Hypertensive Rats, Cell Physiol Biochem 48(3) (2018) 1369–1381. [DOI] [PubMed] [Google Scholar]
  • [113].Mi Y, Wu Q, Yuan W, Chen F, Du D, Role of microglia M1/M2 polarisation in the paraventricular nucleus: New insight into the development of stress-induced hypertension in rats, Auton Neurosci 213 (2018) 71–80. [DOI] [PubMed] [Google Scholar]
  • [114].Du D, Hu L, Wu J, Wu Q, Cheng W, Guo Y, Guan R, Wang Y, Chen X, Yan X, Zhu D, Wang J, Zhang S, Guo Y, Xia C, Neuroinflammation contributes to autophagy flux blockage in the neurons of rostral ventrolateral medulla in stress-induced hypertension rats, J Neuroinflammation 14(1) (2017) 169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [115].Kraft AD, Harry GJ, Features of microglia and neuroinflammation relevant to environmental exposure and neurotoxicity, Int J Environ Res Public Health 8(7) (2011) 2980–3018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [116].Grabert K, Michoel T, Karavolos MH, Clohisey S, Baillie JK, Stevens MP, Freeman TC, Summers KM, McColl BW, Microglial brain region-dependent diversity and selective regional sensitivities to aging, Nat Neurosci 19(3) (2016) 504–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [117].Gosselin D, Link VM, Romanoski CE, Fonseca GJ, Eichenfield DZ, Spann NJ, Stender JD, Chun HB, Garner H, Geissmann F, Glass CK, Environment drives selection and function of enhancers controlling tissue-specific macrophage identities, Cell 159(6) (2014) 1327–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [118].Bechade C, Cantaut-Belarif Y, Bessis A, Microglial control of neuronal activity, Front Cell Neurosci 7 (2013) 32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [119].Dheen ST, Kaur C, Ling EA, Microglial activation and its implications in the brain diseases, Curr Med Chem 14(11) (2007) 1189–97. [DOI] [PubMed] [Google Scholar]
  • [120].Kreutzberg GW, Microglia: a sensor for pathological events in the CNS, Trends Neurosci 19(8) (1996) 312–8. [DOI] [PubMed] [Google Scholar]
  • [121].Takeuchi H, Jin S, Wang J, Zhang G, Kawanokuchi J, Kuno R, Sonobe Y, Mizuno T, Suzumura A, Tumor necrosis factor-alpha induces neurotoxicity via glutamate release from hemichannels of activated microglia in an autocrine manner, J Biol Chem 281(30) (2006) 21362–8. [DOI] [PubMed] [Google Scholar]
  • [122].Stellwagen D, Malenka RC, Synaptic scaling mediated by glial TNF-alpha, Nature 440(7087) (2006) 1054–9. [DOI] [PubMed] [Google Scholar]
  • [123].Delpech JC, Madore C, Nadjar A, Joffre C, Wohleb ES, Laye S, Microglia in neuronal plasticity: Influence of stress, Neuropharmacology 96(Pt A) (2015) 19–28. [DOI] [PubMed] [Google Scholar]
  • [124].Yirmiya R, Goshen I, Immune modulation of learning, memory, neural plasticity and neurogenesis, Brain Behav Immun 25(2) (2011) 181–213. [DOI] [PubMed] [Google Scholar]
  • [125].Joglar B, Rodriguez-Pallares J, Rodriguez-Perez AI, Rey P, Guerra MJ, Labandeira-Garcia JL, The inflammatory response in the MPTP model of Parkinson’s disease is mediated by brain angiotensin: relevance to progression of the disease, J Neurochem 109(2) (2009) 656–69. [DOI] [PubMed] [Google Scholar]
  • [126].Rodriguez-Pallares J, Rey P, Parga JA, Munoz A, Guerra MJ, Labandeira-Garcia JL, Brain angiotensin enhances dopaminergic cell death via microglial activation and NADPH-derived ROS, Neurobiol Dis 31(1) (2008) 58–73. [DOI] [PubMed] [Google Scholar]
  • [127].Borrajo A, Rodriguez-Perez AI, Diaz-Ruiz C, Guerra MJ, Labandeira-Garcia JL, Microglial TNF-alpha mediates enhancement of dopaminergic degeneration by brain angiotensin, Glia 62(1) (2014) 145–57. [DOI] [PubMed] [Google Scholar]
  • [128].Rodriguez-Perez AI, Borrajo A, Rodriguez-Pallares J, Guerra MJ, Labandeira-Garcia JL, Interaction between NADPH-oxidase and Rho-kinase in angiotensin II-induced microglial activation, Glia 63(3) (2015) 466–82. [DOI] [PubMed] [Google Scholar]
  • [129].Zubcevic J, Waki H, Raizada MK, Paton JF, Autonomic-immune-vascular interaction: an emerging concept for neurogenic hypertension, Hypertension 57(6) (2011) 1026–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [130].Shi P, Raizada MK, Sumners C, Brain cytokines as neuromodulators in cardiovascular control, Clin Exp Pharmacol Physiol 37(2) (2010) e52–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [131].Villar-Cheda B, Dominguez-Meijide A, Joglar B, Rodriguez-Perez AI, Guerra MJ, Labandeira-Garcia JL, Involvement of microglial RhoA/Rho-kinase pathway activation in the dopaminergic neuron death. Role of angiotensin via angiotensin type 1 receptors, Neurobiol Dis 47(2) (2012) 268–79. [DOI] [PubMed] [Google Scholar]
  • [132].Rodriguez-Perez AI, Dominguez-Meijide A, Lanciego JL, Guerra MJ, Labandeira-Garcia JL, Inhibition of Rho kinase mediates the neuroprotective effects of estrogen in the MPTP model of Parkinson’s disease, Neurobiol Dis 58 (2013) 209–19. [DOI] [PubMed] [Google Scholar]
  • [133].Tonges L, Frank T, Tatenhorst L, Saal KA, Koch JC, Szego EM, Bahr M, Weishaupt JH, Lingor P, Inhibition of rho kinase enhances survival of dopaminergic neurons and attenuates axonal loss in a mouse model of Parkinson’s disease, Brain 135(Pt 11) (2012) 3355–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [134].Sun H, Wu H, Yu X, Zhang G, Zhang R, Zhan S, Wang H, Bu N, Ma X, Li Y, Angiotensin II and its receptor in activated microglia enhanced neuronal loss and cognitive impairment following pilocarpine-induced status epilepticus, Mol Cell Neurosci 65 (2015) 58–67. [DOI] [PubMed] [Google Scholar]
  • [135].Rothhammer V, Quintana FJ, Control of autoimmune CNS inflammation by astrocytes, Semin Immunopathol 37(6) (2015) 625–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [136].Colombo E, Farina C, Astrocytes: Key Regulators of Neuroinflammation, Trends Immunol 37(9) (2016) 608–620. [DOI] [PubMed] [Google Scholar]
  • [137].Stern JE, Son S, Biancardi VC, Zheng H, Sharma N, Patel KP, Astrocytes Contribute to Angiotensin II Stimulation of Hypothalamic Neuronal Activity and Sympathetic Outflow, Hypertension 68(6) (2016) 1483–1493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [138].Gowrisankar YV, Clark MA, Angiotensin II induces interleukin-6 expression in astrocytes: Role of reactive oxygen species and NF-kappaB, Mol Cell Endocrinol 437 (2016) 130–141. [DOI] [PubMed] [Google Scholar]
  • [139].Liu G, Hosomi N, Hitomi H, Pelisch N, Fu H, Masugata H, Murao K, Ueno M, Matsumoto M, Nishiyama A, Angiotensin II induces human astrocyte senescence through reactive oxygen species production, Hypertens Res 34(4) (2011) 479–83. [DOI] [PubMed] [Google Scholar]
  • [140].Min LJ, Mogi M, Iwanami J, Sakata A, Jing F, Tsukuda K, Ohshima K, Horiuchi M, Angiotensin II and aldosterone-induced neuronal damage in neurons through an astrocyte-dependent mechanism, Hypertens Res 34(6) (2011) 773–8. [DOI] [PubMed] [Google Scholar]
  • [141].Santello M, Volterra A, TNFalpha in synaptic function: switching gears, Trends Neurosci 35(10) (2012) 638–47. [DOI] [PubMed] [Google Scholar]
  • [142].Bezzi P, Domercq M, Brambilla L, Galli R, Schols D, De Clercq E, Vescovi A, Bagetta G, Kollias G, Meldolesi J, Volterra A, CXCR4-activated astrocyte glutamate release via TNFalpha: amplification by microglia triggers neurotoxicity, Nat Neurosci 4(7) (2001) 702–10. [DOI] [PubMed] [Google Scholar]
  • [143].Pascual O, Ben Achour S, Rostaing P, Triller A, Bessis A, Microglia activation triggers astrocyte-mediated modulation of excitatory neurotransmission, Proc Natl Acad Sci U S A 109(4) (2012) E197–205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [144].Parkhurst CN, Yang G, Ninan I, Savas JN, Yates JR 3rd, Lafaille JJ, Hempstead BL, Littman DR, Gan WB, Microglia promote learning-dependent synapse formation through brain-derived neurotrophic factor, Cell 155(7) (2013) 1596–609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [145].Stevens B, Allen NJ, Vazquez LE, Howell GR, Christopherson KS, Nouri N, Micheva KD, Mehalow AK, Huberman AD, Stafford B, Sher A, Litke AM, Lambris JD, Smith SJ, John SW, Barres BA, The classical complement cascade mediates CNS synapse elimination, Cell 131(6) (2007) 1164–78. [DOI] [PubMed] [Google Scholar]
  • [146].Yang T, Santisteban MM, Rodriguez V, Li E, Ahmari N, Carvajal JM, Zadeh M, Gong M, Qi Y, Zubcevic J, Sahay B, Pepine CJ, Raizada MK, Mohamadzadeh M, Gut dysbiosis is linked to hypertension, Hypertension 65(6) (2015) 1331–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [147].Sharma RK, Oliveira AC, Kim S, Rigatto K, Zubcevic J, Rathinasabapathy A, Kumar A, Lebowitz JJ, Khoshbouei H, Lobaton G, Aquino V, Richards EM, Katovich MJ, Shenoy V, Raizada MK, Involvement of Neuroinflammation in the Pathogenesis of Monocrotaline-Induced Pulmonary Hypertension, Hypertension 71(6) (2018) 1156–1163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [148].Takesue K, Kishi T, Hirooka Y, Sunagawa K, Activation of microglia within paraventricular nucleus of hypothalamus is NOT involved in maintenance of established hypertension, J Cardiol 69(1) (2017) 84–88. [DOI] [PubMed] [Google Scholar]
  • [149].Biancardi VC, Stranahan AM, Krause EG, de Kloet AD, Stern JE, Cross talk between AT1 receptors and Toll-like receptor 4 in microglia contributes to angiotensin II-derived ROS production in the hypothalamic paraventricular nucleus, Am J Physiol Heart Circ Physiol 310(3) (2016) H404–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [150].Xue B, Thunhorst RL, Yu Y, Guo F, Beltz TG, Felder RB, Johnson AK, Central Renin-Angiotensin System Activation and Inflammation Induced by High-Fat Diet Sensitize Angiotensin II-Elicited Hypertension, Hypertension 67(1) (2016) 163–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [151].Bardgett ME, Holbein WW, Herrera-Rosales M, Toney GM, Ang II-salt hypertension depends on neuronal activity in the hypothalamic paraventricular nucleus but not on local actions of tumor necrosis factor-alpha, Hypertension 63(3) (2014) 527–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [152].YanFei Qi SK, Leach Dana D, Long Sarah J, Handberg Eileen M, Rodriguez Vermali, Mandloi Avinash, Raizada Mohan K, Pepine Carl J, Antihypertensive Effects of Minocycline Are Associated With Improvement of Inflammatory Status in Patients With Treatment Resistant Hypertension, Circulation 134 (2016) A15921–A15921. [Google Scholar]
  • [153].Sharma RK, Yang T, Oliveira AC, Lobaton GO, Aquino V, Kim S, Richards EM, Pepine CJ, Sumners C, Raizada MK, Microglial Cells Impact Gut Microbiota and Gut Pathology in Angiotensin II-Induced Hypertension, Circ Res 124(5) (2019) 727–736. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [154].Asraf K, Torika N, Apte RN, Fleisher-Berkovich S, Microglial Activation Is Modulated by Captopril: in Vitro and in Vivo Studies, Front Cell Neurosci 12 (2018) 116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [155].Torika N, Asraf K, Roasso E, Danon A, Fleisher-Berkovich S, Angiotensin Converting Enzyme Inhibitors Ameliorate Brain Inflammation Associated with Microglial Activation: Possible Implications for Alzheimer’s Disease, J Neuroimmune Pharmacol 11(4) (2016) 774–785. [DOI] [PubMed] [Google Scholar]
  • [156].Bhat SA, Goel R, Shukla R, Hanif K, Angiotensin Receptor Blockade Modulates NFkappaB and STAT3 Signaling and Inhibits Glial Activation and Neuroinflammation Better than Angiotensin-Converting Enzyme Inhibition, Mol Neurobiol 53(10) (2016) 6950–6967. [DOI] [PubMed] [Google Scholar]
  • [157].Torika N, Asraf K, Danon A, Apte RN, Fleisher-Berkovich S, Telmisartan Modulates Glial Activation: In Vitro and In Vivo Studies, PLoS One 11(5) (2016) e0155823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [158].Benicky J, Sanchez-Lemus E, Honda M, Pang T, Orecna M, Wang J, Leng Y, Chuang DM, Saavedra JM, Angiotensin II AT1 receptor blockade ameliorates brain inflammation, Neuropsychopharmacology 36(4) (2011) 857–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [159].Saavedra JM, Angiotensin II AT(1) receptor blockers as treatments for inflammatory brain disorders, Clin Sci (Lond) 123(10) (2012) 567–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [160].Torika N, Asraf K, Apte RN, Fleisher-Berkovich S, Candesartan ameliorates brain inflammation associated with Alzheimer’s disease, CNS Neurosci Ther 24(3) (2018) 231–242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [161].Thursby E, Juge N, Introduction to the human gut microbiota, Biochem J 474(11) (2017) 1823–1836. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [162].Mohajeri MH, Brummer RJM, Rastall RA, Weersma RK, Harmsen HJM, Faas M, Eggersdorfer M, The role of the microbiome for human health: from basic science to clinical applications, Eur J Nutr 57(Suppl 1) (2018) 1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [163].C. Human Microbiome Project, Structure, function and diversity of the healthy human microbiome, Nature 486(7402) (2012) 207–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [164].Lazar V, Ditu LM, Pircalabioru GG, Gheorghe I, Curutiu C, Holban AM, Picu A, Petcu L, Chifiriuc MC, Aspects of Gut Microbiota and Immune System Interactions in Infectious Diseases, Immunopathology, and Cancer, Front Immunol 9 (2018) 1830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [165].Hooper LV, Midtvedt T, Gordon JI, How host-microbial interactions shape the nutrient environment of the mammalian intestine, Annu Rev Nutr 22 (2002) 283–307. [DOI] [PubMed] [Google Scholar]
  • [166].Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F, Edberg S, Medzhitov R, Recognition of commensal microflora by toll-like receptors is required for intestinal homeostasis, Cell 118(2) (2004) 229–41. [DOI] [PubMed] [Google Scholar]
  • [167].Hooper LV, Wong MH, Thelin A, Hansson L, Falk PG, Gordon JI, Molecular analysis of commensal host-microbial relationships in the intestine, Science 291(5505) (2001) 881–4. [DOI] [PubMed] [Google Scholar]
  • [168].Backhed F, Ding H, Wang T, Hooper LV, Koh GY, Nagy A, Semenkovich CF, Gordon JI, The gut microbiota as an environmental factor that regulates fat storage, Proc Natl Acad Sci U S A 101(44) (2004) 15718–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [169].De Vadder F, Grasset E, Manneras Holm L, Karsenty G, Macpherson AJ, Olofsson LE, Backhed F, Gut microbiota regulates maturation of the adult enteric nervous system via enteric serotonin networks, Proc Natl Acad Sci U S A 115(25) (2018) 6458–6463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [170].Cryan JF, Dinan TG, Mind-altering microorganisms: the impact of the gut microbiota on brain and behaviour, Nat Rev Neurosci 13(10) (2012) 701–12. [DOI] [PubMed] [Google Scholar]
  • [171].Borre YE, O’Keeffe GW, Clarke G, Stanton C, Dinan TG, Cryan JF, Microbiota and neurodevelopmental windows: implications for brain disorders, Trends Mol Med 20(9) (2014) 509–18. [DOI] [PubMed] [Google Scholar]
  • [172].Desbonnet L, Clarke G, Traplin A, O’Sullivan O, Crispie F, Moloney RD, Cotter PD, Dinan TG, Cryan JF, Gut microbiota depletion from early adolescence in mice: Implications for brain and behaviour, Brain Behav Immun 48 (2015) 165–73. [DOI] [PubMed] [Google Scholar]
  • [173].Belkaid Y, Hand TW, Role of the microbiota in immunity and inflammation, Cell 157(1) (2014) 121–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [174].Carding S, Verbeke K, Vipond DT, Corfe BM, Owen LJ, Dysbiosis of the gut microbiota in disease, Microb Ecol Health Dis 26 (2015) 26191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [175].Roy Sarkar S, Banerjee S, Gut microbiota in neurodegenerative disorders, J Neuroimmunol 328 (2019) 98–104. [DOI] [PubMed] [Google Scholar]
  • [176].Bonaz B, Bazin T, Pellissier S, The Vagus Nerve at the Interface of the Microbiota-Gut-Brain Axis, Front Neurosci 12 (2018) 49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [177].Cryan JF, O’Mahony SM, The microbiome-gut-brain axis: from bowel to behavior, Neurogastroenterol Motil 23(3) (2011) 187–92. [DOI] [PubMed] [Google Scholar]
  • [178].Wen SW, Wong CHY, An unexplored brain-gut microbiota axis in stroke, Gut Microbes 8(6) (2017) 601–606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [179].Hasegawa S, Goto S, Tsuji H, Okuno T, Asahara T, Nomoto K, Shibata A, Fujisawa Y, Minato T, Okamoto A, Ohno K, Hirayama M, Intestinal Dysbiosis and Lowered Serum Lipopolysaccharide-Binding Protein in Parkinson’s Disease, PLoS One 10(11) (2015) e0142164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [180].Sundman MH, Chen NK, Subbian V, Chou YH, The bidirectional gut-brain-microbiota axis as a potential nexus between traumatic brain injury, inflammation, and disease, Brain Behav Immun 66 (2017) 31–44. [DOI] [PubMed] [Google Scholar]
  • [181].Lin L, Zheng LJ, Zhang LJ, Neuroinflammation, Gut Microbiome, and Alzheimer’s Disease, Mol Neurobiol 55(11) (2018) 8243–8250. [DOI] [PubMed] [Google Scholar]
  • [182].Lv LJ, Li SH, Li SC, Zhong ZC, Duan HL, Tian C, Li H, He W, Chen MC, He TW, Wang YN, Zhou X, Yao L, Yin AH, Early-Onset Preeclampsia Is Associated With Gut Microbial Alterations in Antepartum and Postpartum Women, Front Cell Infect Microbiol 9 (2019) 224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [183].Gomez-Arango LF, Barrett HL, McIntyre HD, Callaway LK, Morrison M, Dekker Nitert M, Group ST, Increased Systolic and Diastolic Blood Pressure Is Associated With Altered Gut Microbiota Composition and Butyrate Production in Early Pregnancy, Hypertension 68(4) (2016) 974–81. [DOI] [PubMed] [Google Scholar]
  • [184].Dan X, Mushi Z, Baili W, Han L, Enqi W, Huanhu Z, Shuchun L, Differential Analysis of Hypertension-Associated Intestinal Microbiota, Int J Med Sci 16(6) (2019) 872–881. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [185].Li J, Zhao F, Wang Y, Chen J, Tao J, Tian G, Wu S, Liu W, Cui Q, Geng B, Zhang W, Weldon R, Auguste K, Yang L, Liu X, Chen L, Yang X, Zhu B, Cai J, Gut microbiota dysbiosis contributes to the development of hypertension, Microbiome 5(1) (2017) 14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [186].Richards EM, Pepine CJ, Raizada MK, Kim S, The Gut, Its Microbiome, and Hypertension, Curr Hypertens Rep 19(4) (2017) 36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [187].Pevsner-Fischer M, Blacher E, Tatirovsky E, Ben-Dov IZ, Elinav E, The gut microbiome and hypertension, Curr Opin Nephrol Hypertens 26(1) (2017) 1–8. [DOI] [PubMed] [Google Scholar]
  • [188].Santisteban MM, Qi Y, Zubcevic J, Kim S, Yang T, Shenoy V, Cole-Jeffrey CT, Lobaton GO, Stewart DC, Rubiano A, Simmons CS, Garcia-Pereira F, Johnson RD, Pepine CJ, Raizada MK, Hypertension-Linked Pathophysiological Alterations in the Gut, Circ Res 120(2) (2017) 312–323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [189].Yan Q, Gu Y, Li X, Yang W, Jia L, Chen C, Han X, Huang Y, Zhao L, Li P, Fang Z, Zhou J, Guan X, Ding Y, Wang S, Khan M, Xin Y, Li S, Ma Y, Alterations of the Gut Microbiome in Hypertension, Front Cell Infect Microbiol 7 (2017) 381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [190].Nutting CW, Islam S, Daugirdas JT, Vasorelaxant effects of short chain fatty acid salts in rat caudal artery, Am J Physiol 261(2 Pt 2) (1991) H561–7. [DOI] [PubMed] [Google Scholar]
  • [191].Mortensen FV, Nielsen H, Mulvany MJ, Hessov I, Short chain fatty acids dilate isolated human colonic resistance arteries, Gut 31(12) (1990) 1391–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [192].Pluznick JL, Protzko RJ, Gevorgyan H, Peterlin Z, Sipos A, Han J, Brunet I, Wan LX, Rey F, Wang T, Firestein SJ, Yanagisawa M, Gordon JI, Eichmann A, Peti-Peterdi J, Caplan MJ, Olfactory receptor responding to gut microbiota-derived signals plays a role in renin secretion and blood pressure regulation, Proc Natl Acad Sci U S A 110(11) (2013) 4410–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [193].Ganesh BP, Nelson JW, Eskew JR, Ganesan A, Ajami NJ, Petrosino JF, Bryan RM Jr., Durgan DJ, Prebiotics, Probiotics, and Acetate Supplementation Prevent Hypertension in a Model of Obstructive Sleep Apnea, Hypertension 72(5) (2018) 1141–1150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [194].Yang T, Zubcevic J, Gut-Brain Axis in Regulation of Blood Pressure, Front Physiol 8 (2017) 845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [195].Yang T, Magee KL, Colon-Perez LM, Larkin R, Liao YS, Balazic E, Cowart JR, Arocha R, Redler T, Febo M, Vickroy T, Martyniuk CJ, Reznikov LR, Zubcevic J, 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 (Oxf) 226(2) (2019) e13256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [196].Faraco G, Brea D, Garcia-Bonilla L, Wang G, Racchumi G, Chang H, Buendia I, Santisteban MM, Segarra SG, Koizumi K, Sugiyama Y, Murphy M, Voss H, Anrather J, Iadecola C, Dietary salt promotes neurovascular and cognitive dysfunction through a gut-initiated TH17 response, Nat Neurosci 21(2) (2018) 240–249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [197].Toral M, Robles-Vera I, de la Visitacion N, Romero M, Sanchez M, Gomez-Guzman M, Rodriguez-Nogales A, Yang T, Jimenez R, Algieri F, Galvez J, Raizada MK, Duarte J, Role of the immune system in vascular function and blood pressure control induced by faecal microbiota transplantation in rats, Acta Physiol (Oxf) (2019) e13285. [DOI] [PubMed] [Google Scholar]
  • [198].Bartley A, Yang T, Arocha R, Malphurs WL, Larkin R, Magee KL, Vickroy TW, Zubcevic J, Increased Abundance of Lactobacillales in the Colon of Beta-Adrenergic Receptor Knock Out Mouse Is Associated With Increased Gut Bacterial Production of Short Chain Fatty Acids and Reduced IL17 Expression in Circulating CD4(+) Immune Cells, Front Physiol 9 (2018) 1593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [199].Yang T, Ahmari N, Schmidt JT, Redler T, Arocha R, Pacholec K, Magee KL, Malphurs W, Owen JL, Krane GA, Li E, Wang GP, Vickroy TW, Raizada MK, Martyniuk CJ, Zubcevic J, Shifts in the Gut Microbiota Composition Due to Depleted Bone Marrow Beta Adrenergic Signaling Are Associated with Suppressed Inflammatory Transcriptional Networks in the Mouse Colon, Front Physiol 8 (2017) 220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [200].Yang T, Rodriguez V, Malphurs WL, Schmidt JT, Ahmari N, Sumners C, Martyniuk CJ, Zubcevic J, Butyrate regulates inflammatory cytokine expression without affecting oxidative respiration in primary astrocytes from spontaneously hypertensive rats, Physiol Rep 6(14) (2018) e13732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [201].Patnala R, Arumugam TV, Gupta N, Dheen ST, HDAC Inhibitor Sodium Butyrate-Mediated Epigenetic Regulation Enhances Neuroprotective Function of Microglia During Ischemic Stroke, Mol Neurobiol 54(8) (2017) 6391–6411. [DOI] [PubMed] [Google Scholar]
  • [202].Yamawaki Y, Yoshioka N, Nozaki K, Ito H, Oda K, Harada K, Shirawachi S, Asano S, Aizawa H, Yamawaki S, Kanematsu T, Akagi H, Sodium butyrate abolishes lipopolysaccharide-induced depression-like behaviors and hippocampal microglial activation in mice, Brain Res 1680 (2018) 13–38. [DOI] [PubMed] [Google Scholar]
  • [203].Matt SM, Allen JM, Lawson MA, Mailing LJ, Woods JA, Johnson RW, Butyrate and Dietary Soluble Fiber Improve Neuroinflammation Associated With Aging in Mice, Front Immunol 9 (2018) 1832. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [204].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 35(9) (2017) 1899–1908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [205].Adnan S, Nelson JW, Ajami NJ, Venna VR, Petrosino JF, Bryan RM Jr., Durgan DJ, Alterations in the gut microbiota can elicit hypertension in rats, Physiol Genomics 49(2) (2017) 96–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [206].Toral M, Robles-Vera I, de la Visitacion N, Romero M, Yang T, Sanchez M, Gomez-Guzman M, Jimenez R, Raizada MK, Duarte J, Critical Role of the Interaction Gut Microbiota - Sympathetic Nervous System in the Regulation of Blood Pressure, Front Physiol 10 (2019) 231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [207].Wang Y, Wang Z, Wang Y, Li F, Jia J, Song X, Qin S, Wang R, Jin F, Kitazato K, Wang Y, The Gut-Microglia Connection: Implications for Central Nervous System Diseases, Front Immunol 9 (2018) 2325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [208].Abdel-Haq R, Schlachetzki JCM, Glass CK, Mazmanian SK, Microbiome-microglia connections via the gut-brain axis, J Exp Med 216(1) (2019) 41–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [209].Brown DG, Soto R, Yandamuri S, Stone C, Dickey L, Gomes-Neto JC, Pastuzyn ED, Bell R, Petersen C, Buhrke K, Fujinami RS, O’Connell RM, Stephens WZ, Shepherd JD, Lane TE, Round JL, The microbiota protects from viral-induced neurologic damage through microglia-intrinsic TLR signaling, Elife 8 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [210].Erny D, Hrabe de Angelis AL, Jaitin D, Wieghofer P, Staszewski O, David E, Keren-Shaul H, Mahlakoiv T, Jakobshagen K, Buch T, Schwierzeck V, Utermohlen O, Chun E, Garrett WS, McCoy KD, Diefenbach A, Staeheli P, Stecher B, Amit I, Prinz M, Host microbiota constantly control maturation and function of microglia in the CNS, Nat Neurosci 18(7) (2015) 965–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [211].Huuskonen J, Suuronen T, Nuutinen T, Kyrylenko S, Salminen A, Regulation of microglial inflammatory response by sodium butyrate and short-chain fatty acids, Br J Pharmacol 141(5) (2004) 874–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [212].Durgan DJ, Ganesh BP, Cope JL, Ajami NJ, Phillips SC, Petrosino JF, Hollister EB, Bryan RM Jr., Role of the Gut Microbiome in Obstructive Sleep Apnea-Induced Hypertension, Hypertension 67(2) (2016) 469–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [213].Kim S, Goel R, Kumar A, Qi Y, Lobaton G, Hosaka K, Mohammed M, Handberg EM, Richards EM, Pepine CJ, Raizada MK, Imbalance of gut microbiome and intestinal epithelial barrier dysfunction in patients with high blood pressure, Clin Sci (Lond) 132(6) (2018) 701–718. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [214].Hoyles L, Snelling T, Umlai UK, Nicholson JK, Carding SR, Glen RC, McArthur S, Microbiome-host systems interactions: protective effects of propionate upon the blood-brain barrier, Microbiome 6(1) (2018) 55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [215].Braniste V, Al-Asmakh M, Kowal C, Anuar F, Abbaspour A, Toth M, Korecka A, Bakocevic N, Ng LG, Kundu P, Gulyas B, Halldin C, Hultenby K, Nilsson H, Hebert H, Volpe BT, Diamond B, Pettersson S, The gut microbiota influences blood-brain barrier permeability in mice, Sci Transl Med 6(263) (2014) 263ra158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [216].Touyz RM, Camargo LL, Microglia, the Missing Link in the Brain-Gut-Hypertension Axis, Circ Res 124(5) (2019) 671–673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [217].Pavlov VA, Tracey KJ, Neural circuitry and immunity, Immunol Res 63(1–3) (2015) 38–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [218].Pavlov VA, Tracey KJ, The vagus nerve and the inflammatory reflex--linking immunity and metabolism, Nat Rev Endocrinol 8(12) (2012) 743–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [219].Forsythe P, Bienenstock J, Kunze WA, Vagal pathways for microbiome-brain-gut axis communication, Adv Exp Med Biol 817 (2014) 115–33. [DOI] [PubMed] [Google Scholar]
  • [220].Olofsson PS, Rosas-Ballina M, Levine YA, Tracey KJ, Rethinking inflammation: neural circuits in the regulation of immunity, Immunol Rev 248(1) (2012) 188–204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [221].Miao FJ, Green PG, Levine JD, Mechanosensitive duodenal afferents contribute to vagal modulation of inflammation in the rat, J Physiol 554(Pt 1) (2004) 227–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [222].Goehler LE, Gaykema RP, Nguyen KT, Lee JE, Tilders FJ, Maier SF, Watkins LR, Interleukin-1beta in immune cells of the abdominal vagus nerve: a link between the immune and nervous systems?, J Neurosci 19(7) (1999) 2799–806. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [223].Meneses G, Bautista M, Florentino A, Diaz G, Acero G, Besedovsky H, Meneses D, Fleury A, Del Rey A, Gevorkian G, Fragoso G, Sciutto E, Electric stimulation of the vagus nerve reduced mouse neuroinflammation induced by lipopolysaccharide, J Inflamm (Lond) 13 (2016) 33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [224].Raybould HE, Gut chemosensing: interactions between gut endocrine cells and visceral afferents, Auton Neurosci 153(1–2) (2010) 41–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [225].Bouillaud F, Blachier F, Mitochondria and sulfide: a very old story of poisoning, feeding, and signaling?, Antioxid Redox Signal 15(2) (2011) 379–91. [DOI] [PubMed] [Google Scholar]
  • [226].Olson KR, Mitochondrial adaptations to utilize hydrogen sulfide for energy and signaling, J Comp Physiol B 182(7) (2012) 881–97. [DOI] [PubMed] [Google Scholar]
  • [227].Szabo C, Hydrogen sulphide and its therapeutic potential, Nat Rev Drug Discov 6(11) (2007) 917–35. [DOI] [PubMed] [Google Scholar]
  • [228].Zhao W, Zhang J, Lu Y, Wang R, The vasorelaxant effect of H(2)S as a novel endogenous gaseous K(ATP) channel opener, EMBO J 20(21) (2001) 6008–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [229].Hosoki R, Matsuki N, Kimura H, The possible role of hydrogen sulfide as an endogenous smooth muscle relaxant in synergy with nitric oxide, Biochem Biophys Res Commun 237(3) (1997) 527–31. [DOI] [PubMed] [Google Scholar]
  • [230].Rodriguez J, Maloney RE, Rassaf T, Bryan NS, Feelisch M, Chemical nature of nitric oxide storage forms in rat vascular tissue, Proc Natl Acad Sci U S A 100(1) (2003) 336–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [231].Bucci M, Papapetropoulos A, Vellecco V, Zhou Z, Pyriochou A, Roussos C, Roviezzo F, Brancaleone V, Cirino G, Hydrogen sulfide is an endogenous inhibitor of phosphodiesterase activity, Arterioscler Thromb Vasc Biol 30(10) (2010) 1998–2004. [DOI] [PubMed] [Google Scholar]
  • [232].Pan LL, Liu XH, Gong QH, Wu D, Zhu YZ, Hydrogen sulfide attenuated tumor necrosis factor-alpha-induced inflammatory signaling and dysfunction in vascular endothelial cells, PLoS One 6(5) (2011) e19766. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • [233].Al-Magableh MR, Kemp-Harper BK, Hart JL, Hydrogen sulfide treatment reduces blood pressure and oxidative stress in angiotensin II-induced hypertensive mice, Hypertens Res 38(1) (2015) 13–20. [DOI] [PubMed] [Google Scholar]
  • [234].Lu M, Liu YH, Goh HS, Wang JJ, Yong QC, Wang R, Bian JS, Hydrogen sulfide inhibits plasma renin activity, J Am Soc Nephrol 21(6) (2010) 993–1002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [235].Laggner H, Hermann M, Esterbauer H, Muellner MK, Exner M, Gmeiner BM, Kapiotis S, The novel gaseous vasorelaxant hydrogen sulfide inhibits angiotensin-converting enzyme activity of endothelial cells, J Hypertens 25(10) (2007) 2100–4. [DOI] [PubMed] [Google Scholar]
  • [236].Yan H, Du JB, Tang CS, [Changes and significance of hydrogen sulfide/cystathionine gamma-lyase system in hypertension: an experimental study with rats], Zhonghua Yi Xue Za Zhi 84(13) (2004) 1114–7. [PubMed] [Google Scholar]
  • [237].Yan H, Du J, Tang C, The possible role of hydrogen sulfide on the pathogenesis of spontaneous hypertension in rats, Biochem Biophys Res Commun 313(1) (2004) 22–7. [DOI] [PubMed] [Google Scholar]
  • [238].Tain YL, Hsu CN, Lu PC, Early short-term treatment with exogenous hydrogen sulfide postpones the transition from prehypertension to hypertension in spontaneously hypertensive rat, Clin Exp Hypertens 40(1) (2018) 58–64. [DOI] [PubMed] [Google Scholar]
  • [239].Xiao L, Dong JH, Jin S, Xue HM, Guo Q, Teng X, Wu YM, Hydrogen Sulfide Improves Endothelial Dysfunction via Downregulating BMP4/COX-2 Pathway in Rats with Hypertension, Oxid Med Cell Longev 2016 (2016) 8128957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [240].Xiao L, Dong JH, Teng X, Jin S, Xue HM, Liu SY, Guo Q, Shen W, Ni XC, Wu YM, Hydrogen sulfide improves endothelial dysfunction in hypertension by activating peroxisome proliferator-activated receptor delta/endothelial nitric oxide synthase signaling, J Hypertens 36(3) (2018) 651–665. [DOI] [PubMed] [Google Scholar]
  • [241].Huang P, Chen S, Wang Y, Liu J, Yao Q, Huang Y, Li H, Zhu M, Wang S, Li L, Tang C, Tao Y, Yang G, Du J, Jin H, Down-regulated CBS/H2S pathway is involved in high-salt-induced hypertension in Dahl rats, Nitric Oxide 46 (2015) 192–203. [DOI] [PubMed] [Google Scholar]
  • [242].Chunyu Z, Junbao D, Dingfang B, Hui Y, Xiuying T, Chaoshu T, The regulatory effect of hydrogen sulfide on hypoxic pulmonary hypertension in rats, Biochem Biophys Res Commun 302(4) (2003) 810–6. [DOI] [PubMed] [Google Scholar]
  • [243].Yang G, Wu L, Jiang B, Yang W, Qi J, Cao K, Meng Q, Mustafa AK, Mu W, Zhang S, Snyder SH, Wang R, H2S as a physiologic vasorelaxant: hypertension in mice with deletion of cystathionine gamma-lyase, Science 322(5901) (2008) 587–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [244].Weber GJ, Pushpakumar SB, Sen U, Hydrogen sulfide alleviates hypertensive kidney dysfunction through an epigenetic mechanism, Am J Physiol Heart Circ Physiol 312(5) (2017) H874–H885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [245].Roy A, Khan AH, Islam MT, Prieto MC, Majid DS, Interdependency of cystathione gamma-lyase and cystathione beta-synthase in hydrogen sulfide-induced blood pressure regulation in rats, Am J Hypertens 25(1) (2012) 74–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [246].Chen L, Ingrid S, Ding YG, Liu Y, Qi JG, Tang CS, Du JB, Imbalance of endogenous homocysteine and hydrogen sulfide metabolic pathway in essential hypertensive children, Chin Med J (Engl) 120(5) (2007) 389–93. [PubMed] [Google Scholar]
  • [247].Sun NL, Xi Y, Yang SN, Ma Z, Tang CS, [Plasma hydrogen sulfide and homocysteine levels in hypertensive patients with different blood pressure levels and complications], Zhonghua Xin Xue Guan Bing Za Zhi 35(12) (2007) 1145–8. [PubMed] [Google Scholar]
  • [248].Wang C, Han J, Xiao L, Jin CE, Li DJ, Yang Z, Role of hydrogen sulfide in portal hypertension and esophagogastric junction vascular disease, World J Gastroenterol 20(4) (2014) 1079–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [249].Sun L, Sun S, Li Y, Pan W, Xie Y, Wang S, Zhang Z, Potential biomarkers predicting risk of pulmonary hypertension in congenital heart disease: the role of homocysteine and hydrogen sulfide, Chin Med J (Engl) 127(5) (2014) 893–9. [PubMed] [Google Scholar]
  • [250].Wang K, Ahmad S, Cai M, Rennie J, Fujisawa T, Crispi F, Baily J, Miller MR, Cudmore M, Hadoke PW, Wang R, Gratacos E, Buhimschi IA, Buhimschi CS, Ahmed A, Dysregulation of hydrogen sulfide producing enzyme cystathionine gamma-lyase contributes to maternal hypertension and placental abnormalities in preeclampsia, Circulation 127(25) (2013) 2514–22. [DOI] [PubMed] [Google Scholar]
  • [251].Possomato-Vieira JS, Goncalves-Rizzi VH, do Nascimento RA, Wandekin RR, Caldeira-Dias M, Chimini JS, da Silva MLS, Dias-Junior CA, Clinical and Experimental Evidences of Hydrogen Sulfide Involvement in Lead-Induced Hypertension, Biomed Res Int 2018 (2018) 4627391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [252].Ahmad FU, Sattar MA, Rathore HA, Abdullah MH, Tan S, Abdullah NA, Johns EJ, Exogenous hydrogen sulfide (H2S) reduces blood pressure and prevents the progression of diabetic nephropathy in spontaneously hypertensive rats, Ren Fail 34(2) (2012) 203–10. [DOI] [PubMed] [Google Scholar]
  • [253].Zhang X, Bian JS, Hydrogen sulfide: a neuromodulator and neuroprotectant in the central nervous system, ACS Chem Neurosci 5(10) (2014) 876–83. [DOI] [PubMed] [Google Scholar]
  • [254].Abe K, Kimura H, The possible role of hydrogen sulfide as an endogenous neuromodulator, J Neurosci 16(3) (1996) 1066–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [255].Kimura H, Physiological role of hydrogen sulfide and polysulfide in the central nervous system, Neurochem Int 63(5) (2013) 492–7. [DOI] [PubMed] [Google Scholar]
  • [256].Lu M, Choo CH, Hu LF, Tan BH, Hu G, Bian JS, Hydrogen sulfide regulates intracellular pH in rat primary cultured glia cells, Neurosci Res 66(1) (2010) 92–8. [DOI] [PubMed] [Google Scholar]
  • [257].Yong QC, Choo CH, Tan BH, Low CM, Bian JS, Effect of hydrogen sulfide on intracellular calcium homeostasis in neuronal cells, Neurochem Int 56(3) (2010) 508–15. [DOI] [PubMed] [Google Scholar]
  • [258].Nagai Y, Tsugane M, Oka J, Kimura H, Hydrogen sulfide induces calcium waves in astrocytes, FASEB J 18(3) (2004) 557–9. [DOI] [PubMed] [Google Scholar]
  • [259].Lee SW, Hu YS, Hu LF, Lu Q, Dawe GS, Moore PK, Wong PT, Bian JS, Hydrogen sulphide regulates calcium homeostasis in microglial cells, Glia 54(2) (2006) 116–24. [DOI] [PubMed] [Google Scholar]
  • [260].Lee M, McGeer EG, McGeer PL, Sodium thiosulfate attenuates glial-mediated neuroinflammation in degenerative neurological diseases, J Neuroinflammation 13 (2016) 32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [261].Xuan A, Long D, Li J, Ji W, Zhang M, Hong L, Liu J, Hydrogen sulfide attenuates spatial memory impairment and hippocampal neuroinflammation in beta-amyloid rat model of Alzheimer’s disease, J Neuroinflammation 9 (2012) 202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [262].Lee M, McGeer E, Kodela R, Kashfi K, McGeer PL, NOSH-aspirin (NBS-1120), a novel nitric oxide and hydrogen sulfide releasing hybrid, attenuates neuroinflammation induced by microglial and astrocytic activation: a new candidate for treatment of neurodegenerative disorders, Glia 61(10) (2013) 1724–34. [DOI] [PubMed] [Google Scholar]
  • [263].Tu F, Li J, Wang J, Li Q, Chu W, Hydrogen sulfide protects against cognitive impairment induced by hepatic ischemia and reperfusion via attenuating neuroinflammation, Exp Biol Med (Maywood) 241(6) (2016) 636–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [264].Hu LF, Lu M, Tiong CX, Dawe GS, Hu G, Bian JS, Neuroprotective effects of hydrogen sulfide on Parkinson’s disease rat models, Aging Cell 9(2) (2010) 135–46. [DOI] [PubMed] [Google Scholar]
  • [265].Yu H, Xu H, Liu X, Zhang N, He A, Yu J, Lu N, Superoxide Mediates Depressive Effects Induced by Hydrogen Sulfide in Rostral Ventrolateral Medulla of Spontaneously Hypertensive Rats, Oxid Med Cell Longev 2015 (2015) 927686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [266].Eto K, Asada T, Arima K, Makifuchi T, Kimura H, Brain hydrogen sulfide is severely decreased in Alzheimer’s disease, Biochem Biophys Res Commun 293(5) (2002) 1485–8. [DOI] [PubMed] [Google Scholar]
  • [267].Beyer K, Lao JI, Carrato C, Rodriguez-Vila A, Latorre P, Mataro M, Llopis MA, Mate JL, Ariza A, Cystathionine beta synthase as a risk factor for Alzheimer disease, Curr Alzheimer Res 1(2) (2004) 127–33. [DOI] [PubMed] [Google Scholar]
  • [268].Vandini E, Ottani A, Zaffe D, Calevro A, Canalini F, Cavallini GM, Rossi R, Guarini S, Giuliani D, Mechanisms of Hydrogen Sulfide against the Progression of Severe Alzheimer’s Disease in Transgenic Mice at Different Ages, Pharmacology 103(1–2) (2019) 50–60. [DOI] [PubMed] [Google Scholar]
  • [269].Duan XC, Liu SY, Guo R, Xiao L, Xue HM, Guo Q, Jin S, Wu YM, Cystathionine-beta-Synthase Gene Transfer Into Rostral Ventrolateral Medulla Exacerbates Hypertension via Nitric Oxide in Spontaneously Hypertensive Rats, Am J Hypertens 28(9) (2015) 1106–13. [DOI] [PubMed] [Google Scholar]
  • [270].Juman S, Nara Y, Yasui N, Negishi H, Okuda H, Takado N, Miki T, Reduced Production of Hydrogen Sulfide and Sulfane Sulfur Due to Low Cystathionine beta-Synthase Levels in Brain Astrocytes of Stroke-Prone Spontaneously Hypertensive Rats, Biol Pharm Bull 39(12) (2016) 1932–1938. [DOI] [PubMed] [Google Scholar]
  • [271].Wang Y, Jia J, Ao G, Hu L, Liu H, Xiao Y, Du H, Alkayed NJ, Liu CF, Cheng J, Hydrogen sulfide protects blood-brain barrier integrity following cerebral ischemia, J Neurochem 129(5) (2014) 827–38. [DOI] [PubMed] [Google Scholar]
  • [272].Geng Y, Li E, Mu Q, Zhang Y, Wei X, Li H, Cheng L, Zhang B, Hydrogen sulfide inhalation decreases early blood-brain barrier permeability and brain edema induced by cardiac arrest and resuscitation, J Cereb Blood Flow Metab 35(3) (2015) 494–500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [273].Li H, Zhu L, Feng J, Hu X, Li C, Zhang B, Hydrogen Sulfide Decreases Blood-Brain Barrier Damage via Regulating Protein Kinase C and Tight Junction After Cardiac Arrest in Rats, Cell Physiol Biochem 47(3) (2018) 994–1006. [DOI] [PubMed] [Google Scholar]
  • [274].Martin GR, McKnight GW, Dicay MS, Coffin CS, Ferraz JG, Wallace JL, Hydrogen sulphide synthesis in the rat and mouse gastrointestinal tract, Dig Liver Dis 42(2) (2010) 103–9. [DOI] [PubMed] [Google Scholar]
  • [275].Blachier F, Davila AM, Mimoun S, Benetti PH, Atanasiu C, Andriamihaja M, Benamouzig R, Bouillaud F, Tome D, Luminal sulfide and large intestine mucosa: friend or foe?, Amino Acids 39(2) (2010) 335–47. [DOI] [PubMed] [Google Scholar]
  • [276].Kabil O, Vitvitsky V, Banerjee R, Sulfur as a signaling nutrient through hydrogen sulfide, Annu Rev Nutr 34 (2014) 171–205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [277].Liu YH, Lu M, Hu LF, Wong PT, Webb GD, Bian JS, Hydrogen sulfide in the mammalian cardiovascular system, Antioxid Redox Signal 17(1) (2012) 141–85. [DOI] [PubMed] [Google Scholar]
  • [278].Singh S, Banerjee R, PLP-dependent H(2)S biogenesis, Biochim Biophys Acta 1814(11) (2011) 1518–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [279].Fiorucci S, Antonelli E, Distrutti E, Rizzo G, Mencarelli A, Orlandi S, Zanardo R, Renga B, Di Sante M, Morelli A, Cirino G, Wallace JL, Inhibition of hydrogen sulfide generation contributes to gastric injury caused by anti-inflammatory nonsteroidal drugs, Gastroenterology 129(4) (2005) 1210–24. [DOI] [PubMed] [Google Scholar]
  • [280].Hu LF, Lu M, Hon Wong PT, Bian JS, Hydrogen sulfide: neurophysiology and neuropathology, Antioxid Redox Signal 15(2) (2011) 405–19. [DOI] [PubMed] [Google Scholar]
  • [281].Linden DR, Hydrogen sulfide signaling in the gastrointestinal tract, Antioxid Redox Signal 20(5) (2014) 818–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [282].Barton LL, Fauque GD, Biochemistry, physiology and biotechnology of sulfate-reducing bacteria, Adv Appl Microbiol 68 (2009) 41–98. [DOI] [PubMed] [Google Scholar]
  • [283].Castro HF, Williams NH, Ogram A, Phylogeny of sulfate-reducing bacteria(1), FEMS Microbiol Ecol 31(1) (2000) 1–9. [DOI] [PubMed] [Google Scholar]
  • [284].Liamleam W, Annachhatre AP, Electron donors for biological sulfate reduction, Biotechnol Adv 25(5) (2007) 452–63. [DOI] [PubMed] [Google Scholar]
  • [285].Shen X, Carlstrom M, Borniquel S, Jadert C, Kevil CG, Lundberg JO, Microbial regulation of host hydrogen sulfide bioavailability and metabolism, Free Radic Biol Med 60 (2013) 195–200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [286].Ise F, Takasuka H, Hayashi S, Takahashi K, Koyama M, Aihara E, Takeuchi K, Stimulation of duodenal HCO(3)(−) secretion by hydrogen sulphide in rats: relation to prostaglandins, nitric oxide and sensory neurones, Acta Physiol (Oxf) 201(1) (2011) 117–26. [DOI] [PubMed] [Google Scholar]
  • [287].Takeuchi K, Ise F, Takahashi K, Aihara E, Hayashi S, H2S-induced HCO3- secretion in the rat stomach--involvement of nitric oxide, prostaglandins, and capsaicin-sensitive sensory neurons, Nitric Oxide 46 (2015) 157–64. [DOI] [PubMed] [Google Scholar]
  • [288].Schicho R, Krueger D, Zeller F, Von Weyhern CW, Frieling T, Kimura H, Ishii I, De Giorgio R, Campi B, Schemann M, Hydrogen sulfide is a novel prosecretory neuromodulator in the Guinea-pig and human colon, Gastroenterology 131(5) (2006) 1542–52. [DOI] [PubMed] [Google Scholar]
  • [289].Matsunami M, Kirishi S, Okui T, Kawabata A, Hydrogen sulfide-induced colonic mucosal cytoprotection involves T-type calcium channel-dependent neuronal excitation in rats, J Physiol Pharmacol 63(1) (2012) 61–8. [PubMed] [Google Scholar]
  • [290].Goubern M, Andriamihaja M, Nubel T, Blachier F, Bouillaud F, Sulfide, the first inorganic substrate for human cells, FASEB J 21(8) (2007) 1699–706. [DOI] [PubMed] [Google Scholar]
  • [291].Pimentel M, Mathur R, Chang C, Gas and the microbiome, Curr Gastroenterol Rep 15(12) (2013) 356. [DOI] [PubMed] [Google Scholar]
  • [292].Tomasova L, Dobrowolski L, Jurkowska H, Wrobel M, Huc T, Ondrias K, Ostaszewski R, Ufnal M, Intracolonic hydrogen sulfide lowers blood pressure in rats, Nitric Oxide 60 (2016) 50–58. [DOI] [PubMed] [Google Scholar]
  • [293].Huc T, Jurkowska H, Wrobel M, Jaworska K, Onyszkiewicz M, Ufnal M, Colonic hydrogen sulfide produces portal hypertension and systemic hypotension in rats, Exp Biol Med (Maywood) 243(1) (2018) 96–106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [294].Galla S, Chakraborty S, Cheng X, Yeo J, Mell B, Zhang H, Mathew AV, Vijay-Kumar M, Joe B, Disparate effects of antibiotics on hypertension, Physiol Genomics 50(10) (2018) 837–845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [295].Strocchi A, Furne JK, Levitt MD, A modification of the methylene blue method to measure bacterial sulfide production in feces, Journal of Microbiological Methods 15(2) (1992) 75–82. [Google Scholar]
  • [296].Sugahara S, Suzuki M, Kamiya H, Yamamuro M, Semura H, Senga Y, Egawa M, Seike Y, Colorimetric Determination of Sulfide in Microsamples, Anal Sci 32(10) (2016) 1129–1131. [DOI] [PubMed] [Google Scholar]
  • [297].Jarosz AP, Yep T, Mutus B, Microplate-based colorimetric detection of free hydrogen sulfide, Anal Chem 85(7) (2013) 3638–43. [DOI] [PubMed] [Google Scholar]
  • [298].Ahn YJ, Lee YJ, Lee J, Lee D, Park HK, Lee GJ, Colorimetric detection of endogenous hydrogen sulfide production in living cells, Spectrochim Acta A Mol Biomol Spectrosc 177 (2017) 118–124. [DOI] [PubMed] [Google Scholar]
  • [299].Vitvitsky V, Banerjee R, H2S analysis in biological samples using gas chromatography with sulfur chemiluminescence detection, Methods Enzymol 554 (2015) 111–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [300].Fume JK, Springfield J, Koenig T, Suarez F, Levitt MD, Measurement of fecal sulfide using gas chromatography and a sulfur chemiluminescence detector, J Chromatogr B Biomed Sci Appl 754(1) (2001) 253–8. [DOI] [PubMed] [Google Scholar]
  • [301].Shen X, Peter EA, Bir S, Wang R, Kevil CG, Analytical measurement of discrete hydrogen sulfide pools in biological specimens, Free Radic Biol Med 52(11–12) (2012) 2276–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [302].Wintner EA, Deckwerth TL, Langston W, Bengtsson A, Leviten D, Hill P, Insko MA, Dumpit R, VandenEkart E, Toombs CF, Szabo C, A monobromobimane-based assay to measure the pharmacokinetic profile of reactive sulphide species in blood, Br J Pharmacol 160(4) (2010) 941–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [303].Koike S, Kawamura K, Kimura Y, Shibuya N, Kimura H, Ogasawara Y, Analysis of endogenous H2S and H2Sn in mouse brain by high-performance liquid chromatography with fluorescence and tandem mass spectrometric detection, Free Radic Biol Med 113 (2017) 355–362. [DOI] [PubMed] [Google Scholar]
  • [304].Varlet V, Giuliani N, Palmiere C, Maujean G, Augsburger M, Hydrogen sulfide measurement by headspace-gas chromatography-mass spectrometry (HS-GC-MS): application to gaseous samples and gas dissolved in muscle, J Anal Toxicol 39(1) (2015) 52–7. [DOI] [PubMed] [Google Scholar]
  • [305].Tan B, Jin S, Sun J, Gu Z, Sun X, Zhu Y, Huo K, Cao Z, Yang P, Xin X, Liu X, Pan L, Qiu F, Jiang J, Jia Y, Ye F, Xie Y, Zhu YZ, New method for quantification of gasotransmitter hydrogen sulfide in biological matrices by LC-MS/MS, Sci Rep 7 (2017) 46278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [306].Maseda C, Hayakawa A, Okuda K, Asari M, Tanaka H, Yamada H, Jin S, Horioka K, Matoba K, Shiono H, Matsubara K, Shimizu K, Liquid chromatography-tandem mass spectrometry method for the determination of thiosulfate in human blood and urine as an indicator of hydrogen sulfide poisoning, Leg Med (Tokyo) 24 (2017) 67–74. [DOI] [PubMed] [Google Scholar]
  • [307].Jin S, Hyodoh H, Matoba K, Feng F, Hayakawa A, Okuda K, Shimizu K, Haga S, Ozaki M, Terazawa K, Development for the measurement of serum thiosulfate using LC-MS/MS in forensic diagnosis of H2S poisoning, Leg Med (Tokyo) 22 (2016) 18–22. [DOI] [PubMed] [Google Scholar]
  • [308].Doeller JE, Isbell TS, Benavides G, Koenitzer J, Patel H, Patel RP, Lancaster JR Jr., Darley-Usmar VM, Kraus DW, Polarographic measurement of hydrogen sulfide production and consumption by mammalian tissues, Anal Biochem 341(1) (2005) 40–51. [DOI] [PubMed] [Google Scholar]
  • [309].Huo FJ, Kang J, Yin C, Chao J, Zhang Y, Highly selective fluorescent and colorimetric probe for live-cell monitoring of sulphide based on bioorthogonal reaction, Sci Rep 5 (2015) 8969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [310].Shimamoto K, Hanaoka K, Fluorescent probes for hydrogen sulfide (H2S) and sulfane sulfur and their applications to biological studies, Nitric Oxide 46 (2015) 72–9. [DOI] [PubMed] [Google Scholar]
  • [311].Zheng D, Chen Z, Chen J, Zhuang X, Feng J, Li J, Exogenous hydrogen sulfide exerts proliferation, anti-apoptosis, migration effects and accelerates cell cycle progression in multiple myeloma cells via activating the Akt pathway, Oncol Rep 36(4) (2016) 1909–16. [DOI] [PubMed] [Google Scholar]
  • [312].Lin J, Zhao H, Jiao F, Ma L, Ma L, Lymphocyte hydrogen sulfide production predicts intravenous immunoglobulin resistance in children with Kawasaki disease: A preliminary, single-center, case-control study, Medicine (Baltimore) 97(47) (2018) e13069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [313].Ugliano M, Henschke PA, Comparison of three methods for accurate quantification of hydrogen sulfide during fermentation, Anal Chim Acta 660(1–2) (2010) 87–91. [DOI] [PubMed] [Google Scholar]
  • [314].Kolluru GK, Shen X, Bir SC, Kevil CG, Hydrogen sulfide chemical biology: pathophysiological roles and detection, Nitric Oxide 35 (2013) 5–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [315].Wang R, Physiological implications of hydrogen sulfide: a whiff exploration that blossomed, Physiol Rev 92(2) (2012) 791–896. [DOI] [PubMed] [Google Scholar]
  • [316].Ogawa M, Takahara A, Ishijima M, Tazaki S, Decrease of plasma sulfur amino acids in essential hypertension, Jpn Circ J 49(12) (1985) 1217–24. [DOI] [PubMed] [Google Scholar]
  • [317].Loubinoux J, Bronowicki JP, Pereira IA, Mougenel JL, Faou AE, Sulfate-reducing bacteria in human feces and their association with inflammatory bowel diseases, FEMS Microbiol Ecol 40(2) (2002) 107–12. [DOI] [PubMed] [Google Scholar]
  • [318].Figliuolo VR, Dos Santos LM, Abalo A, Nanini H, Santos A, Brittes NM, Bernardazzi C, de Souza HSP, Vieira LQ, Coutinho-Silva R, Coutinho C, Sulfate-reducing bacteria stimulate gut immune responses and contribute to inflammation in experimental colitis, Life Sci 189 (2017) 29–38. [DOI] [PubMed] [Google Scholar]
  • [319].Lennon G, Balfe A, Bambury N, Lavelle A, Maguire A, Docherty NG, Coffey JC, Winter DC, Sheahan K, O’Connell PR, Correlations between colonic crypt mucin chemotype, inflammatory grade and Desulfovibrio species in ulcerative colitis, Colorectal Dis 16(5) (2014) O161–9. [DOI] [PubMed] [Google Scholar]
  • [320].Haran JP, Bhattarai SK, Foley SE, Dutta P, Ward DV, Bucci V, McCormick BA, Alzheimer’s Disease Microbiome Is Associated with Dysregulation of the Anti-Inflammatory P-Glycoprotein Pathway, MBio 10(3) (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [321].Lin CH, Chen CC, Chiang HL, Liou JM, Chang CM, Lu TP, Chuang EY, Tai YC, Cheng C, Lin HY, Wu MS, Altered gut microbiota and inflammatory cytokine responses in patients with Parkinson’s disease, J Neuroinflammation 16(1) (2019) 129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [322].Tremlett H, Fadrosh DW, Faruqi AA, Zhu F, Hart J, Roalstad S, Graves J, Lynch S, Waubant E, U.S.N.o.P.M. Centers, Gut microbiota in early pediatric multiple sclerosis: a case-control study, Eur J Neurol 23(8) (2016) 1308–1321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [323].Ijssennagger N, Belzer C, Hooiveld GJ, Dekker J, van Mil SW, Muller M, Kleerebezem M, van der Meer R, Gut microbiota facilitates dietary heme-induced epithelial hyperproliferation by opening the mucus barrier in colon, Proc Natl Acad Sci U S A 112(32) (2015) 10038–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [324].Tsubota-Matsunami M, Noguchi Y, Okawa Y, Sekiguchi F, Kawabata A, Colonic hydrogen sulfide-induced visceral pain and referred hyperalgesia involve activation of both Ca(v)3.2 and TRPA1 channels in mice, J Pharmacol Sci 119(3) (2012) 293–6. [DOI] [PubMed] [Google Scholar]
  • [325].Chassard C, Dapoigny M, Scott KP, Crouzet L, Del’homme C, Marquet P, Martin JC, Pickering G, Ardid D, Eschalier A, Dubray C, Flint HJ, Bernalier-Donadille A, Functional dysbiosis within the gut microbiota of patients with constipated-irritable bowel syndrome, Aliment Pharmacol Ther 35(7) (2012) 828–38. [DOI] [PubMed] [Google Scholar]
  • [326].Rowan FE, Docherty NG, Coffey JC, O’Connell PR, Sulphate-reducing bacteria and hydrogen sulphide in the aetiology of ulcerative colitis, Br J Surg 96(2) (2009) 151–8. [DOI] [PubMed] [Google Scholar]
  • [327].Medani M, Collins D, Docherty NG, Baird AW, O’Connell PR, Winter DC, Emerging role of hydrogen sulfide in colonic physiology and pathophysiology, Inflamm Bowel Dis 17(7) (2011) 1620–5. [DOI] [PubMed] [Google Scholar]
  • [328].Finke N, Vandieken V, Jorgensen BB, Acetate, lactate, propionate, and isobutyrate as electron donors for iron and sulfate reduction in Arctic marine sediments, Svalbard, FEMS Microbiol Ecol 59(1) (2007) 10–22. [DOI] [PubMed] [Google Scholar]
  • [329].Moore JW, Babidge W, Millard S, Roediger WE, Effect of sulphide on short chain acyl-CoA metabolism in rat colonocytes, Gut 41(1) (1997) 77–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [330].Babidge W, Millard S, Roediger W, Sulfides impair short chain fatty acid beta-oxidation at acyl-CoA dehydrogenase level in colonocytes: implications for ulcerative colitis, Mol Cell Biochem 181(1–2) (1998) 117–24. [DOI] [PubMed] [Google Scholar]
  • [331].Sawin EA, De Wolfe TJ, Aktas B, Stroup BM, Murali SG, Steele JL, Ney DM, Glycomacropeptide is a prebiotic that reduces Desulfovibrio bacteria, increases cecal short-chain fatty acids, and is anti-inflammatory in mice, Am J Physiol Gastrointest Liver Physiol 309(7) (2015) G590–601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [332].Marquet P, Duncan SH, Chassard C, Bernalier-Donadille A, Flint HJ, Lactate has the potential to promote hydrogen sulphide formation in the human colon, FEMS Microbiol Lett 299(2) (2009) 128–34. [DOI] [PubMed] [Google Scholar]
  • [333].Snijder PM, Frenay AR, de Boer RA, Pasch A, Hillebrands JL, Leuvenink HG, van Goor H, Exogenous administration of thiosulfate, a donor of hydrogen sulfide, attenuates angiotensin II-induced hypertensive heart disease in rats, Br J Pharmacol 172(6) (2015) 1494–504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [334].Snijder PM, Frenay AR, Koning AM, Bachtler M, Pasch A, Kwakernaak AJ, van den Berg E, Bos EM, Hillebrands JL, Navis G, Leuvenink HG, van Goor H, Sodium thiosulfate attenuates angiotensin II-induced hypertension, proteinuria and renal damage, Nitric Oxide 42 (2014) 87–98. [DOI] [PubMed] [Google Scholar]
  • [335].Hsu CN, Lin YJ, Lu PC, Tain YL, Early Supplementation of d-Cysteine or l-Cysteine Prevents Hypertension and Kidney Damage in Spontaneously Hypertensive Rats Exposed to High-Salt Intake, Mol Nutr Food Res 62(2) (2018). [DOI] [PubMed] [Google Scholar]
  • [336].Ried K, Garlic Lowers Blood Pressure in Hypertensive Individuals, Regulates Serum Cholesterol, and Stimulates Immunity: An Updated Meta-analysis and Review, J Nutr 146(2) (2016) 389S–396S. [DOI] [PubMed] [Google Scholar]
  • [337].Ried K, Frank OR, Stocks NP, Fakler P, Sullivan T, Effect of garlic on blood pressure: a systematic review and meta-analysis, BMC Cardiovasc Disord 8 (2008) 13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [338].Benavides GA, Squadrito GL, Mills RW, Patel HD, Isbell TS, Patel RP, Darley-Usmar VM, Doeller JE, Kraus DW, Hydrogen sulfide mediates the vasoactivity of garlic, Proc Natl Acad Sci U S A 104(46) (2007) 17977–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [339].Sun Q, Wang B, Li Y, Sun F, Li P, Xia W, Zhou X, Li Q, Wang X, Chen J, Zeng X, Zhao Z, He H, Liu D, Zhu Z, Taurine Supplementation Lowers Blood Pressure and Improves Vascular Function in Prehypertension: Randomized, Double-Blind, Placebo-Controlled Study, Hypertension 67(3) (2016) 541–9. [DOI] [PubMed] [Google Scholar]
  • [340].Fujita T, Ando K, Noda H, Ito Y, Sato Y, Effects of increased adrenomedullary activity and taurine in young patients with borderline hypertension, Circulation 75(3) (1987) 525–32. [DOI] [PubMed] [Google Scholar]
  • [341].Zhao H, Qu J, Li Q, Cui M, Wang J, Zhang K, Liu X, Feng H, Chen Y, Taurine supplementation reduces neuroinflammation and protects against white matter injury after intracerebral hemorrhage in rats, Amino Acids 50(3–4) (2018) 439–451. [DOI] [PubMed] [Google Scholar]
  • [342].Hou L, Che Y, Sun F, Wang Q, Taurine protects noradrenergic locus coeruleus neurons in a mouse Parkinson’s disease model by inhibiting microglial M1 polarization, Amino Acids 50(5) (2018) 547–556. [DOI] [PubMed] [Google Scholar]
  • [343].de Moraes AC, Bel-Serrat S, Manios Y, Molnar D, Kafatos A, Cuenca-Garcia M, Huybrechts I, Sette S, Widhalm K, Stehle P, Jimenez-Pavon D, Carvalho HB, Moreno LA, Dietary protein and amino acids intake and its relationship with blood pressure in adolescents: the HELENA STUDY, Eur J Public Health 25(3) (2015) 450–6. [DOI] [PubMed] [Google Scholar]
  • [344].Tuttle KR, Milton JE, Packard DP, Shuler LA, Short RA, Dietary amino acids and blood pressure: a cohort study of patients with cardiovascular disease, Am J Kidney Dis 59(6) (2012) 803–9. [DOI] [PubMed] [Google Scholar]
  • [345].Zhou Y, Zhao L, Zhang Z, Lu X, Protective Effect of Enalapril against Methionine-Enriched Diet-Induced Hypertension: Role of Endoplasmic Reticulum and Oxidative Stress, Biomed Res Int 2015 (2015) 724876. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [346].Robin S, Maupoil V, Groubatch F, Laurant P, Jacqueson A, Berthelot A, Effect of a methionine-supplemented diet on the blood pressure of Wistar-Kyoto and spontaneously hypertensive rats, Br J Nutr 89(4) (2003) 539–48. [DOI] [PubMed] [Google Scholar]
  • [347].Robin S, Maupoil V, Laurant P, Jacqueson A, Berthelot A, Effect of a methionine-supplemented diet on the blood pressure of Sprague-Dawley and deoxycorticosterone acetate-salt hypertensive rats, Br J Nutr 91(6) (2004) 857–65. [DOI] [PubMed] [Google Scholar]
  • [348].Yen CH, Lau YT, Vascular responses in male and female hypertensive rats with hyperhomocysteinemia, Hypertension 40(3) (2002) 322–8. [DOI] [PubMed] [Google Scholar]
  • [349].Hine C, Harputlugil E, Zhang Y, Ruckenstuhl C, Lee BC, Brace L, Longchamp A, Trevino-Villarreal JH, Mejia P, Ozaki CK, Wang R, Gladyshev VN, Madeo F, Mair WB, Mitchell JR, Endogenous hydrogen sulfide production is essential for dietary restriction benefits, Cell 160(1–2) (2015) 132–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [350].Yao CK, Rotbart A, Ou JZ, Kalantar-Zadeh K, Muir JG, Gibson PR, Modulation of colonic hydrogen sulfide production by diet and mesalazine utilizing a novel gas-profiling technology, Gut Microbes 9(6) (2018) 510–522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [351].Florin TH, Hydrogen sulphide and total acid-volatile sulphide in faeces, determined with a direct spectrophotometric method, Clin Chim Acta 196(2–3) (1991) 127–34. [DOI] [PubMed] [Google Scholar]
  • [352].Jenkins TA, Nguyen JC, Hart JL, Decreased vascular H2S production is associated with vascular oxidative stress in rats fed a high-fat western diet, Naunyn Schmiedebergs Arch Pharmacol 389(7) (2016) 783–90. [DOI] [PubMed] [Google Scholar]
  • [353].Mu C, Yang Y, Luo Z, Guan L, Zhu W, The Colonic Microbiome and Epithelial Transcriptome Are Altered in Rats Fed a High-Protein Diet Compared with a Normal-Protein Diet, J Nutr 146(3) (2016) 474–83. [DOI] [PubMed] [Google Scholar]

RESOURCES