Next Article in Journal
Adult Langerhans Cell Histiocytosis and the Skeleton
Next Article in Special Issue
Is Poor Lithium Response in Individuals with Bipolar Disorder Associated with Increased Degradation of Tryptophan along the Kynurenine Pathway? Results of an Exploratory Study
Previous Article in Journal
Machine Learning for Screening Microvascular Complications in Type 2 Diabetic Patients Using Demographic, Clinical, and Laboratory Profiles
Previous Article in Special Issue
Ordering Knowledge in the Markers of Psychiatric/Mental Disorders
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Multiple Aspects of Inappropriate Action of Renin–Angiotensin, Vasopressin, and Oxytocin Systems in Neuropsychiatric and Neurodegenerative Diseases

by
Ewa Szczepanska-Sadowska
*,
Agnieszka Wsol
,
Agnieszka Cudnoch-Jedrzejewska
,
Katarzyna Czarzasta
and
Tymoteusz Żera
Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
*
Author to whom correspondence should be addressed.
Submission received: 30 December 2021 / Revised: 4 February 2022 / Accepted: 5 February 2022 / Published: 9 February 2022
(This article belongs to the Special Issue Advances in Markers of Psychiatric Disorders)

Abstract

:
The cardiovascular system and the central nervous system (CNS) closely cooperate in the regulation of primary vital functions. The autonomic nervous system and several compounds known as cardiovascular factors, especially those targeting the renin–angiotensin system (RAS), the vasopressin system (VPS), and the oxytocin system (OTS), are also efficient modulators of several other processes in the CNS. The components of the RAS, VPS, and OTS, regulating pain, emotions, learning, memory, and other cognitive processes, are present in the neurons, glial cells, and blood vessels of the CNS. Increasing evidence shows that the combined function of the RAS, VPS, and OTS is altered in neuropsychiatric/neurodegenerative diseases, and in particular in patients with depression, Alzheimer’s disease, Parkinson’s disease, autism, and schizophrenia. The altered function of the RAS may also contribute to CNS disorders in COVID-19. In this review, we present evidence that there are multiple causes for altered combined function of the RAS, VPS, and OTS in psychiatric and neurodegenerative disorders, such as genetic predispositions and the engagement of the RAS, VAS, and OTS in the processes underlying emotions, memory, and cognition. The neuroactive pharmaceuticals interfering with the synthesis or the action of angiotensins, vasopressin, and oxytocin can improve or worsen the effectiveness of treatment for neuropsychiatric/neurodegenerative diseases. Better knowledge of the multiple actions of the RAS, VPS, and OTS may facilitate programming the most efficient treatment for patients suffering from the comorbidity of neuropsychiatric/neurodegenerative and cardiovascular diseases.

1. Introduction

Cardiovascular diseases (CVDs), which are the leading causes of disability-adjusted life years (DALYs), morbidity, and premature death, are particularly challenging when they are associated with diseases affecting the central nervous system (CNS) [1,2,3]. Several studies point to the close cooperation of the CNS and the cardiovascular system (CVS) in the regulation of basic vital functions (Figure 1). The CNS requires a continuous supply of oxygen and nutrients, and the removal of metabolites by the circulating blood, whereas the blood flow needs to be precisely controlled by the autonomic nervous system and the cardiovascular brain regions of the CNS, located at several levels of the brain, including the cortex [4,5,6]. Neurons and glial cells release potent cardiovascular factors which regulate the function of neighboring cells and remote organs, while the heart, vessels, and kidneys, produce neuroactive factors, which are transported to the brain and modulate the function of the CNS [6,7,8,9,10,11]. Substantial evidence indicates that all classical neurotransmitters and several neuroactive peptides, which transmit information between the cardiovascular regions of the brain and the spinal cord, are also engaged in the regulation of cognition, emotions, pain, and behavior at the molecular level [12,13,14,15,16,17]. Moreover, it has been shown that brain diseases are often associated with cardiovascular disturbances [15,18,19,20,21].
Several neuropeptides, initially known as the cardiovascular agents, such as angiotensins, vasopressin, and oxytocin, have been identified as effective modulators of cognitive functions, pain, stress, and emotions [16,17,22,23,24]. Moreover, these compounds frequently interact with other neuroactive agents and their interaction can be altered in cardiovascular pathologies [25,26,27]. It has been found that the effectiveness of second-generation antipsychotics and antidepressive compounds may differ in patients with CVDs, but the reasons for these differences have not been satisfactorily determined [18,28].
The purpose of the present review is to draw attention to the role of cardiovascular neuropeptides in neuropsychiatric and neurodegenerative disorders. We focused on neuropeptides forming the renin–angiotensin (RAS), vasopressin (VPS), and oxytocin (OTS) systems for several reasons. First, these peptides are essential multifunctional molecules when it comes to tuning up the excitatory and inhibitory processes in the brain. Second, the processes of synthesis, release, and the action of these peptides in the CVS and CNS are relatively well recognized. Third, in many instances, the RAS, VPS, and OTS are activated jointly. Fourth, the release of angiotensins, vasopressin, and oxytocin, and the action of these peptides, are altered in CVDs, neuropsychiatric and neurodegenerative diseases. Fifth, some of the specific agonists and antagonists of the RAS, VPS, and OTS are commercially available and used for therapeutic purposes.

2. Renin–Angiotensin System

2.1. A Brief Overview of the RAS

The main components forming the RAS are shown in Figure 2. Circulating renin is mainly synthesized in the juxtaglomerular cells of the renal afferent arterioles. Renin is necessary to detach angiotensin I (Ang I) from angiotensinogen. Subsequently, Ang I is converted either to Ang II, a potent vasoconstrictive octapeptide, by angiotensin converting enzyme 1 (ACE), or to a vasodilatory peptide angiotensin-(1-7) [Ang-(1-7)] by angiotensin converting enzyme 2 (ACE2). An alternative pathway for the formation of Ang-(1-7) involves the cleavage of Ang-(1-9) from Ang I by ACE2 and the subsequent detachment of Ang-(1-7) by ACE. Ang II can also be metabolized to Ang III and then to Ang IV [29,30,31,32]. ACE is expressed mainly by the endothelial cells of the pulmonary vessels, but it is also present in the brain, heart, and other organs. In addition to the classical hormonal (tissue-to-tissue) RAS, the local paracrine/autocrine (cell-to-cell) RAS and the intracrine (intracellular/nuclear) RAS have both been identified in various organs, including the brain and the heart. The local RAS systems are activated by topical stimuli [11,29,32].
The actions of Ang II and Ang III are mediated by the AT1 receptors (AT1R) and the AT2 receptors (AT2R). Stimulation of AT1R causes the activation of the nicotinamide adenine dinucleotide phosphate (NADPH)-oxidase complex and may promote inflammatory processes [33,34]. Stimulation of AT2R is associated with the activation of phosphotyrosine phosphatases, especially serine/threonine phosphatase 2A, protein kinase phosphatase, and SHP-1 tyrosine phosphatase. It also causes the inactivation of mitogen activated protein kinases (MAPK), specifically p42 and p44 MAPK [35]. The most prominent expression of AT2R has been found in the kidney, heart, blood vessels, and brain, especially in the soma and dendrites of the paraventricular nucleus (PVN) [36]. AT1Rs and AT2Rs may form heteromers, and the blockade of one of the components of this heterodimer increases the interaction of Ang II with the other component [37]. Ang IV stimulates the AT4 receptors (AT4Rs), identified as insulin-regulated aminopeptidase (IRAP) [38], but, in high concentrations, it can also stimulate AT1R. It has been shown that the Ang IV/AT4R pathway in the brain interacts with the hepatocyte growth factor/c-Met receptor system [39]. Ang-(1-7) activates the Mas receptors (MasRs) and the ACE2/Ang-(1-7)/Mas axis [40,41,42].

2.2. Systemic RAS and Brain RAS

Active elements of the RAS have been found in the kidney, lungs, adrenal glands, heart, vessels, carotid glomeruli, brain, and the spinal cord [16,31,43,44,45]. Among the stimulators of the RAS are hypoxia, hypovolemia, hypotension, sympathetic stimulation, stress, pain, and specific neuroendocrine factors [7,16,17,31,43,44,46,47,48,49]. The RAS is activated by inflammatory processes associated with tissue injury and oxidative stress. On the other hand, the components of the RAS intensify the inflammatory processes through cooperation with proinflammatory cytokines [10,24,27,50,51,52,53,54]. Acting via the AT1R, Ang II stimulates the NADPH-oxidase complex and NADPH-dependent oxidases that generate inflammatory processes involved in tissue degeneration [34,55].
Prorenin, renin angiotensinogen, angiotensin converting enzyme (ACE), angiotensins I-IV, ACE2, Ang-(1-7) and their receptors (AT1R, AT2R, and MasR) are present in multiple brain regions regulating blood flow, the water-electrolyte balance, and cognitive and emotional processes [31,44,56,57,58].
In the forebrain, components of the RAS have been found in the cortex, the hypothalamus, and the circumventricular organs (organum vasculosum laminae terminalis, OVLT, and the subfornical organ, SFO) [36,59,60,61,62]. They have been detected in the midbrain and hindbrain, specifically in the periaqueductal gray (PAG), the substantia nigra (SN), the dorsal raphe nucleus (DRN), the rostral ventrolateral medulla (RVLM), the caudal ventrolateral medulla (CVLM), the nucleus of the solitary tract (NTS), the nucleus ambiguous (NcAmb), the dorsal motor nucleus of the vagus (DMVNc), and the area postrema (AP) [40,63,64,65,66,67,68,69,70]. MasR mRNA and its protein have been identified in the brain stem cardiovascular regions encompassing the dopaminergic neurons of the substantia nigra [40,71].
In the brain, the central components of the RAS mediate sympathetic stimulation, which is particularly intensive during hypoxia [72,73]. In addition, they regulate the release of several factors involved in the generation of the final cardiovascular response. For example, the local application of prorenin into the supraoptic nucleus (SON) increases membrane excitability and the firing responses of magnocellular neurons, in addition to elevating plasma vasopressin (AVP), with all of these effects being mediated by the inhibition of A-type potassium channels [74]. RAS components have been found in the cranial and spinal cord ganglia. In the spinal cord, AT1R and AT2R have been detected in the regions involved in the regulation of pain and sympathetic outflow (intermediolateral cell column, lamina X and V) [17,56,75].

2.3. RAS in Cardiovascular Disorders

The cardiovascular actions of Ang II are exerted by the stimulation of the central and peripheral angiotensin receptors and are partly mediated by the stimulation of neurons regulating the autonomic nervous system or the secretion of cardiovascular hormones, cytokines, and other vasoactive factors acting in the brain or on the systemic circulation [24,27,52,63,64,76,77,78,79,80,81,82,83,84]. Experimental studies have shown that the administration of Ang II directly into the brain exerts a pressor action, which is significantly potentiated in hypertension and cardiac failure, and which is mediated by AT1Rs [24,46,85,86,87]. It is essential to note that Ang II upregulates its own receptors in the brain, specifically in the hypothalamus, the SFO, and the RVLM, and it is likely that, under chronic conditions, it can potentiate its own effects [63,88,89,90,91]. The central administration of AT1R antagonists significantly reduces the cardiovascular effects of the sympathetic stimulation in heart failure and suppresses the activation of the hypothalamic–pituitary axis during isolation stress [22,83]. In the mouse model, the deletion of the AT1R gene in the paraventricular nucleus (PVN) has been shown to significantly reduce anxiety-like behavior, blood pressure elevation, heart rate variability, and, in addition, decreases the expression of proinflammatory cytokines in the hypothalamus during exposure to the elevated plus maze test [92]. In contrast, transgenic mice expressing the AT1R in the C1 neurons of the ventrolateral medulla manifested an exaggerated pressor response to aversive cage-switch stress [93]. There is evidence that the central pressor action of Ang II is potentiated by cytokines, and that AT1Rs are involved in the pressor action of interleukin 1 beta (IL-1β) and tumor necrosis factor α (TNF-α) [27,52,55,84]. In this context, it is worth noting that brain inflammation, induced by the systemic administration of lipopolysaccharide, elicits a significant release of the proinflammatory cytokines TNF-α, IL-1β, and interleukin 6 (IL-6) to the systemic circulation and increases the expression of TNF-α, IL-1β, and IL-6 mRNAs in the prefrontal cortex, the PVN, the SFO, the amygdala, and the hippocampus. The above effects can be significantly reduced by the systemic administration of centrally acting AT1R antagonist candesartan [94].
In many respects, the activation of the ACE2 → Ang (1-7) → MasR axis plays the opposite role in blood pressure regulation. An overexpression of ACE2 or the intracerebroventricular administration of Ang-(1-7) reduces the blood pressure in experimental models of hypertension and heart failure [56,70,95,96,97].

2.4. Inappropriate Function of RAS in Neuropsychiatric/Neurodegenerative Diseases

The inappropriate functional actions of renin–angiotensin system in selected neuropsychiatric and neurodegenerative disorders are summarized in Table 1.

2.4.1. RAS in Cognitive Disorders

Both experimental and clinical studies have shown that the excessive activation of Ang II receptors may impair cognitive processes [98,99], whereas the blockade of AT1 receptors by a specific AT1R antagonist exerts a neuroprotective action and improves cognitive functions [100,101,102]. In the caucasian population, exposure to ACE inhibitors protected carriers of the AA genotype of the GAG and the CC genotype of the M235T from mental decline [121]. In contrast, Ang IV and Ang (1-7) appear to exert positive effects on cognition and both of these peptides improve learning and memory capabilities [103,104,105,229]. In the brain, Ang-(1-7), Ang IV, and their receptors are present mainly in the neocortex, hippocampus, amygdala, and basal ganglia, and it is likely that these regions may be the main sites of their positive role in cognition [230,231].

2.4.2. RAS in Stress and Pain

Stress provoked by tissue injury, ischemia, hypoxia, inflammation, stroke, or myocardial infarction activates the RAS and increases expression of the AT1R in the brain, heart, and kidney [16,23,25,66,67,85,87,115]. Twenty-four hours isolation stress increases AT1R binding in the PVN and the adrenal medulla and these effects can be abolished by the central or systemic administration of the AT1R antagonist candesartan [232,233]. Experiments on animal models of chronic mild stress have provided evidence for the significant increase of AT1R mRNA expression in the septal/accumbal, diencephalic, medullary, and cerebellar regions of the brain, and in the renal medulla [67]. Furthermore, studies on rats have shown that the cardiovascular responses to chronic mild stress and acute restraint stress are modulated by the RAS and the stimulation of AT1 receptors [86,234]. The effects of microinjections of ACE inhibitors and AT1R and AT2R antagonists directly into the prelimbic cortex give rise to the assumption that Ang II enhances the pressor response to stress by means of the AT1R, while the tachycardic response to stress is enhanced by AT2R [116].
Pain, which is one of the stress-inducers and which is frequently experienced in cardiovascular and neuropsychiatric/neurodegenerative diseases, is a potent activator of the RAS. The specific components of the RAS play a complex role in the regulation of pain [17,235,236]. In the spinal cord, heart, and other organs, Ang II induces nociceptive action, whereas both stimulation of the AT1R and AT2R and the activation of the Ang-(1-7) → MasR pathway in the brain elicit analgesia [117,118].

2.4.3. RAS in Depression and Anxiety

The clinical picture of depression manifests significant heterogeneity. Inappropriate dimensions, the activity and metabolism of the cortex, the subcortical limbic regions, the basal ganglia, and the brain stem may suggest that the pathogenesis of depression is associated with the improper function of these structures [237,238]. The role of the dorsolateral prefrontal cortex, the anterior cingulate cortex, the orbital frontal cortex, and the insula, appears to be particularly interesting. These regions have multiple connections with the amygdala, the thalamus, the lateral and medial orbitofrontal cortex, and the medial prefrontal cortex, which are all involved in the regulation of emotions [239,240,241]. Magnetic resonance imaging studies performed in patients with depression revealed reduced dimensions of the frontal and orbitofrontal lobes, and decreased metabolism and blood flow in the dorsolateral prefrontal cortex and the ventral region of the anterior cingulate cortex [237,242,243]. In addition, it has been found that a deficit in facial disgust recognition correlates with reduced grey matter volume in the insula, which is engaged in the regulation of emotions and blood pressure regulation [244].
Earlier studies could not confirm any associations between ACE I/D genotypes and depression [119,245]; however, the significant association between depression and the AT1R A1166C CC genotype, found by Saab et al. [119], suggests that the inappropriate synthesis of AT1R may contribute to the development of depression in patients. More recently, an analysis of haplotype-tagging single nucleotide polymorphism of angiotensin AT1R revealed significant differences between the cohorts of depressed and nondepressed patients with rs10935724 and rs12721331 htSNPs. The authors found significant associations between AT1R htSNPs and the volumes of the prefrontal cortex and the hippocampus [246].
Experimental studies on transgenic rats [TGR(ASrAOGEN)680] have shown that rats with low brain angiotensinogen manifest anxiety-related behavior and depressive-like behavior, which can be reversed by the ICV application of Ang-(1-7) [120]. The authors suggest that anxiety and depression in these rats may be caused by a deficit of Ang-(1-7), which is one of the derivatives of angiotesinogen (Figure 2).

2.4.4. RAS in Alzheimer’s Disease

The extracellular deposition of aggregated amyloid beta (Aβ) plaques and the formation of neurofibrillary tangles of hyperphosphorylated tau protein as well as the suppressed function of the brain cholinergic system are characteristic features of Alzheimer’s disease (AD), but the mechanism of Aβ toxicity is not yet fully understood [247,248]. Elevated concentrations of ACE, angiotensin II, and AT1 receptors in the cerebral cortex of patients with AD suggest that enhanced activation of the RAS may inhibit the release of acetylcholine in the cortex and contribute to the development of Alzheimer’s dementia [106,107,108]. Losartan and valsartan decrease Aβ peptide oligomerization in primary neuronal cultures and reduce cognitive impairment in Tg2576 AD transgenic mice, expressing the human 695-aa isoform of the amyloid precursor protein gene (APP) [249]. In addition, captopril prevents Aβ-induced downregulation of some genes involved in neuronal regeneration and cognition [250]. Studies performed on animal models of AD and postmortem examinations of human brains has led investigators to suggest that the excessive activation of the brain AT1R and insufficient activation of AT2R may induce excessive generation of reactive oxygen species (ROS), and this may account for the prevalence of neurodegenerative processes over the neuroprotective processes in the brains of AD patients [109,110,111,112,113].
Studies exploring the effects of ACE inhibitors and AT1R blockers on the cognitive abilities of AD patients have not yielded uniform results. Symptoms of dementia tend to be lower in patients treated with ACE inhibitors; however, the differences are not significant [98]. Similarly, a lack of the beneficial effects of RAS targeting compounds on dementia and AD symptoms was reported in the ONTARGET and TRANSCEND clinical trials [251], and in a quantitative meta-analysis [252]. However, in another study, a significantly slower progression of AD was found in patients treated with ACE inhibitors crossing the blood–brain barrier (captopril, perindopril) [253]. More recently, the memory improving effects of ACE inhibitors and AT1R blockers were found in a meta-analysis of patients with AD and cognitive impairment of aging [254,255], as well as in a cohort study in which AT1R blockers were applied together with statins [256].
It has been found that the Ang-(1-7) level is significantly reduced in the cerebral cortex and hippocampus of the senescence-accelerated mouse prone 8 (SAMP8) model of Alzheimer’s disease and that the inhibition is associated with an inverse correlation between the Ang-(1-7) level and tau hyperphosphorylation. Therefore, it has been suggested that the inappropriate activation of the Ang-(1-7)/Mas axis may play a role in the pathogenesis of AD [114].

2.4.5. RAS in Parkinson’s Disease and Tardive Dyskinesia

Cardiac autonomic dysfunction, orthostatic hypotension, and ECG abnormalities belong to the most common non-motor symptoms of Parkinson’s disease (PD) and may even precede motoric disorders [19,257]. Parkinson’s disease is associated with a loss of dopaminergic neurons in the pars compacta of the substantia nigra. With the advancement of the disease, progressive degeneration and dysfunction occurs in other classical and nonclassical neurotransmitter systems, including the RAS [258]. The accumulation of α-synuclein, which has been proposed to be one of the cytotoxic PD factors inducing microglial activation, is associated with the increased expression of AT1Rs and NADPH oxidase activation. It has been shown that the blockade of AT1Rs by candesartan and telmisartan significantly reduces the negative effects of α-synuclein in microglia and dopaminergic neurons [51]. In the rat PD model produced by the administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), the application of AT1R antagonists or ACE inhibitors significantly reduces neuronal cell death. Similar effects were obtained in primary mesencephalic cell cultures. It has been suggested that endogenous Ang II potentiates the neurotoxic effect of MPTP on dopaminergic neurons, and that the antagonists of ACE or AT1Rs exert their beneficial effects through the inhibition of microglial and NADPH activation and the suppression of the prooxidative and proinflammatory effects mediated by cytokines [125,126,127]. Later studies have shown that the administration of candesartan does not influence motor efficiency nor the dopamine and serotonin levels in the striatum, but it increases the expression of dopamine D1 receptors and decreases the expression of dopamine D2 receptors. Studies on the rat model of Parkinson’s disease have indicated that the neuroprotective effect of chronic treatment with AT1R antagonists is associated with the formation of heterodimers of AT1R/AT2R [37,125]. It is worth noting that the administration of candesartan and lisinopril reduces the release of proinflammatory cytokines (IL-1β, TNF-α) and glutamate in the rat model of haloperidol-induced tardive dyskinesia, which is another dopaminergic disorder resulting from damage of the striatal neurons [128].
To date, meta-analyses on human subjects have not provided explicit evidence for the association between ACE gene I/D polymorphism and PD risk [259].

2.4.6. RAS in Schizophrenia and Autism

Schizophrenia belongs to difficult neuropsychiatric disorders which affect human beings of different ages and sexes. Unfortunatelly, relatively little attention has been given to the regulation of the cardiovascular system in schizophrenic patients.
In comparison with the general population, patients with schizophrenia are more predisposed to CVDs and have a greater risk of adverse cardiac events, such as stroke and heart failure [260,261,262,263,264]. On the other hand, patients with schizophrenia and myocardial infarction who receive secondary preventive cardiovascular treatment have a similar mortality rate as the general population [265].
It is only in a few studies that attempts have been made to find out whether there are relations between mutations of RAS genes and schizophrenia, and the results are not uniform. Studies on cardiovascular genomics and cognitive function in patients with schizophrenia revealed some missense mutations of angiotensinogen (AGTM268T, AGT235T). The mutations involved replacement of valine by threonine and were associated with the decline of cognitive functions and lower verbal memory scores [121,122]. An association between ACE I/D (insertion/deletion) polymorphism and a disposition to schizophrenia was found in the study of Gadelha et al. [123], but not in the earlier report of Gard et al. [245], nor in the meta-analysis collecting data from European, Asian, and Turkish populations [266]. The alleviation of schizophrenia symptoms in patients with the disorder via the AT1R antagonist telmisartan was found by Fan et al. [124]. The beneficial effects of AT1 receptor blockade with irbesartan in schizophrenic patients with psychogenic polidypsia may suggest that the inappropriate activation of some of the components of RAS may account for polidypsia in patients with schizophrenia [267].
Strong associations between DD genotype of ACE I/D and the D allele has been found in autistic patients and the authors suggest that genetic diversity of RAS may enhance the risk of autism [268].

2.4.7. RAS in Coronavirus Infections

Growing evidence indicates that the inappropriate activation of RAS components may contribute to disturbances in brain function in coronavirus disease (COVID-19) and that survivors of COVID-19 manifest aggravated symptoms of neuropsychiatric disorders such as cognitive and attention deficits (i.e., brain fog), new-onset anxiety, depression, psychosis, seizures, and suicidal behavior [269]. A substantial body of evidence indicates that COVID-19 infection affects the RAS and that the imbalance of the local and systemic Ang II and Ang-(1-7) activities may play an essential role in the pathological processes developing in the lungs and other organs during SARS-CoV-2 infections [270,271]. Human ACE2 binds the virus S protein and plays a fundamental role in transmitting the original SARS-CoV and the new SARS-CoV-2 to the targeted cells [272]. Expression of ACE2 mRNA and its protein has been reported in the cortex, striatum, hippocampus, brain stem, and cerebrovascular endothelium of the rodent brain [273,274,275] and the human brain [276,277]. SARS-CoV-2 is expressed preferentially in cultured glial cells, specifically in astrocytes and radial glial progenitor cells [278,279]. Preclinical studies on transgenic mice suggest that SARS-CoV, very closely related to the SARS-CoV-2 coronavirus, can access the brain through the olfactory nerve and the olfactory bulb, and it may subsequently be transported transneuronally or spread via the Virchow–Robin spaces and along brain vessels causing extensive neuronal infection [280]. The SARS-CoV-2 spike protein, which readily crosses the blood–brain barrier, induces an inflammatory response within microvascular endothelial cells, leading to the dysfunction of the blood-brain barrier [276]. In the brains of infected patients, SARS-CoV has been detected almost exclusively in neurons [281,282]. The neuroinvasive capacity of the SARS-CoV-2 virus has also been reported. For example, autopsies of brains from patients who died of COVID-19 made it possible to detect SARS-CoV-2 in the cortical neurons [283].
It is likely that the inappropriate function of the RAS may contribute to the exaggeration of psychiatric symptoms in patients with COVID-19, especially when considering that the excessive stimulation of the AT1R influences microglial polarization and induces an active M2a proinflammatory state, thereby initiating neurodegenerative processes [42]. This assumption is highly feasible in view of the data showing a correlation between neuroinflammation, brain microvascular injury, and cognitive function impairment [129].
Some patients with COVID-19 infection may exhibit so-called “silent hypoxemia”, which is manifested by severe hypoxemia without dyspnea or tachypnea and suggests inadequate stimulation of the arterial chemoreceptors. Since carotid bodies possess local RAS elements, including ACE2, it has been hypothesized that the ACE2-mediated entry of SARS-CoV-2 into carotid bodies may contribute to the development of silent hypoxemia in COVID-19 infection [284,285,286,287]. This notion is further supported by evidence showing the expression of SARS-CoV-2 in carotid bodies from a patient with COVID-19 [288]. Altogether, it appears that the inappropriate activity of RAS should be taken into account as an effective cause of brain dysfunction occurring in COVID-19 disease.

3. Vasopressin and Oxytocin Systems

In many instances, vasopressin and oxytocin are released jointly and engage similar cellular mechanisms, acting either synergistically or antagonistically [289].

3.1. Overview of Systemic and Peripheral VPS and OTS

Vasopressin and oxytocin are synthesized mainly in the supraoptic, paraventricular, and suprachiasmatic nuclei of the hypothalamus and released to the blood in the neurohypophysis. They are also synthesized in some other cells of the central nervous system and in the peripheral organs [17,289]. The neuroregulatory and peripheral actions of vasopressin are mediated by three types of AVP receptors: V1a (V1aR), V1b (V1bR), and V2 (V2R) [289,290]. Oxytocin stimulates its own receptors (OXTR); however, in higher concentrations, it can also act by means of V1aR [291].
Current evidence indicates that cardiovascular and neuroregulatory processes are tuned up mainly by the V1 receptors [292,293,294,295,296]. AVP cooperates with the RAS through multiple actions exerted in the brain and in the peripheral organs, and its action is significantly altered in cardiovascular diseases [26,85,140,141,292,297,298,299]. The inappropriate functions of the VPS and OTS have been described in neuroregulatory disorders, and it has been suggested that they may play essential roles in the development of social, emotional, and cognitive dysfunctions, including dementia [300,301,302,303,304,305,306,307]. It has been shown that neurogenic stress provokes the release of AVP and oxytocin (OT) and that these two peptides are involved in the regulation of the cardiovascular, emotional, and behavioral responses, and in processing social information [26,140,141,142,143,144,145]. Studies on rodents indicate that the AVP → V1bR pathway plays an important role in the regulation of the hormonal and behavioral responses to stress and in the formation of social recognition memory. Experiments on Avpr1b-/-mice suggest that social memory is regulated by the V1bR located in the CA2 region of the hippocampus [154,300] and that the blockade of V1bR induces anxiolytic actions in various models of depression [146,147].

3.2. Vasopressin and Oxytocin in Depression

The inappropriate functional actions of VPS and OTS in selected neuropsychiatric and neurodegenerative disorders are summarized in Table 1.

3.2.1. Vasopressin and Depression

Post-mortem examinations of human brains revealed the elevated expression of AVP mRNA in the PVN and SON and a significantly greater number of AVP and OT neurons in the PVN of patients with MDD (major depressive disorder) [130]. In rodent models of anxiety and depression, the application of orally active V1bR antagonists reduced hyperemotionality and elicited anxiolytic- and antidepressant-like effects [137,138,139]. Furthermore, oral administration of a V1bR antagonist (TASPO390325) antagonized the elevation of adrenocorticotropic hormone levels induced by the joint application of corticotrophin-releasing hormone and vasopressin analog (desmopressin, dDAVP) [139]. In the rat model of depression, the blockade of the central V1 receptors abolished anhedonia elicited by chronic mild stress [140].
Indirect evidence for the engagement of the VPS in the pathogenesis of MDD and aggression comes from studies of the V1bR gene polymorphism [131,132]. The disposition to affective disorders was associated with the polymorphism of the V1bR gene, in particular, with the presence of the haplotype associated with A-T-C-A-G for the single nucleotide polymorphism (SNP) s1-s2-s3-s4-s5 allele. It appears that presence of this haplotype may protect from recurrent MDD [131]. A linkage between the V1bR genetic variation SNP rs33990840 and a predisposition to suicidal behavior was also reported [133], and clinical trials assessing the usefulness of V1bR antagonists for the treatment of MDD have recently started [134,135].
Earlier studies assessing the usefulness of blood AVP measurements in patients with depression or mania did not yield uniform results [308,309,310,311,312], and the meta-analyses did not find support to diagnose depression based on the estimations of AVP and OT levels in blood, saliva, urine, and the cerebrospinal fluid (CSF) [306,313]. However, measurements of copeptin, which is a surrogate marker of AVP, indicate that blood copeptin levels are elevated in patients who are insensitive to antidepressant treatment, and it appears that copeptin may be a useful biomarker for the early selection of non-responders to specific antidepressant treatments [136].

3.2.2. Oxytocin in Depression and Anxiety

A prevailing number of studies indicate a correlation between the altered function of the central oxytocinergic system and affective disorders, although the results of human studies are inconsistent. Most of the studies report lower levels of plasma OT in patients with MDD and bipolar affective disorder during depressive episodes than in the control subjects [184,187,188,189]; however, higher [190,191] or normal [192] levels were also found. It appears that plasma OT levels may be lower in MDD than in the control subjects [187,314] for women in particular, which may suggest the greater sensibility of the female OT system to disturbed signaling in affective disorders. Similarly, measurements of the OT level in the CSF did not give explicit results. Demitrack and Gold did not find significant differences in OT levels in the CSF between MDD patients and the control group [315]. Analogous results were obtained in other studies [316,317]. On the contrary, postmortem studies to assess the neuronal OT mRNA levels in MDD patients have indicated an elevated activation of the OT system in MDD patients. Namely, using immunocytochemical techniques on the post-mortem samples of the PVN collected from patients with MDD and bipolar affective depressive episode, Purba et al. [130] found significant elevation of OT-immunoreactivity in patients with depressive mood disorders. These results were supported by a case-control study, in which elevated OT-immunoreactivity in the PVN was found in MDD and bipolar disorder patients [185]. The increased expression of OT mRNA in the PVN was also reported in melancholic MDD patients [186]. Recently, post-mortem estimations of OT receptor (OTR) mRNA expression have shown that patients with MDD and bipolar disorder have significantly higher OTR mRNA levels in the dorsolateral prefrontal cortex, a brain structure implicated in the pathophysiology of numerous psychiatric disorders including MDD and bipolar disease [318]. Altogether, the available data indicate the increased activity of the central OT system in depression. However, it should be emphasized that the intracerebral and peripheral release of OT in depression may occur either in a coordinated or independent manner depending on the quality or the strength of the stimulus [319].
It should also be noted that plasma oxytocin levels positively correlate with help-seeking intentions and behavior in depressed patients [193]. Anderberg and Uvnäs-Moberg found a negative correlation between the plasma OT concentration and the scored symptoms of depression and anxiety [184]. They also reported a positive correlation between the estimation of happiness and the plasma oxytocin level. It has been suggested that plasma OT levels may help to predict whether a patient with chronic depression will respond to psychotherapy [194].
The involvement of OT in mechanisms underlying depression and anxiety has also been analyzed in studies based on genetic tests. In humans, a positive association between depression and separation anxiety was found for a single nucleotide polymorphism (SNP; rs53576) of the OTR gene [195]. Specifically, the GG genotype of this SNP has been linked to high levels of separation anxiety and insecure attachment in MDD patients [196]. Other authors found an interaction between another OTR SNP polymorphism (rs2254298) and the symptoms of depression and anxiety in adolescent girls, especially when the polymorphism occurred in association with an adverse parental environment [197]. More recent data indicate that variations in the OTR and G-protein (Gβ3 rs5443) genes are specifically associated with separation anxiety and depressive symptoms both in childhood and in adulthood [198]. Other authors reported the negative influence of the coincidence of the presence of the OTR rs53576 genotype A allele and environmental adversity in postnatal life (for example, maternal postpartum depression) on the mental health and social behavior of the child [199]. In addition, studies of a SNP of the OTR gene in depressed adults indicate the association of the A allele rs53576 with a history of suicide attempts [200].
Special attention should also be given to the correlation between the dysregulation of the OT system and postpartum depression (PPD). Data from animal models and human studies have suggested that disruptions in the activity of the OT system may account for the relationship between breastfeeding, stress coping, and mood [201,320]. It has been shown that responsiveness to stress is decreased in breastfeeding women (the stimulus for OT secretion) and that this is associated with a stress-induced rise in plasma cortisol [320]. Furthermore, it has been found that lowered plasma OT levels in the third trimester of pregnancy makes it possible to predict postpartum depressive symptoms [321]. In addition, OT levels were inversely correlated with depressive symptoms in mothers who intended to breastfeed in both the third trimester and at 8 weeks postpartum. During breastfeeding, OT release was lower in depressed mothers than in nondepressed mothers [202].
Despite promising results from experimental studies, clinical trials have yielded inconclusive results, with some reports supporting a significant positive effect and others suggesting a null effect in the treatment of anxiety and depression [322,323,324,325]. However, it has been found that oxytocin improves the therapeutic effects of other antidepressants, such as escitalopram [326]. Additionally, intranasally applied OT was shown to modify neural activity in the limbic regions of depressed patients [327]. In a group of Vietnam veterans suffering from post-traumatic stress, the intranasal administration of OT reduced physiological responses during personal combat imagery [328].

3.2.3. Vasopressin and Oxytocin in Alzheimer’s Disease

Patients suffering from Alzheimer’s disease have a lower level of AVP in the CSF [148], and post-mortem studies revealed a reduced expression of AVP immunoreactivity in the hippocampus, nucleus accumbens, and the internal portion of the globus pallidus of AD patients in comparison with controls [149]. Other post-mortem studies of human brains provided evidence for the reduced number of AVP expressing cells in the suprachiasmatic nucleus in senescence and AD patients [150]; however, the vasopressinergic innervation of the PVN, SON, and locus coeruleus in AD patients and non-demented controls did not differ [151,152].
Recently, experiments on the APP/PS1 mouse model of AD have shown that the intranasal application of an AVP derivative [AVP-(4-8)] markedly improves working memory and long-term memory in this experimental model of AD [153].
A number of studies have reported the impact of intranasally applied OT on human cognitive functions (see [178] for a review); however, most of the previous studies did not find significant changes in the activity of the central oxytocinergic system in different brain regions of patients with Alzheimer’s disease [151,179,180,181,182]. A trend for elevated hippocampal OT immunoreactivity in post-mortem brain samples of patients with AD was observed in one of the investigations [180]. More recent studies suggest that the OTS may be affected to some extent in AD patients. Lardenoije et al. found changes in the methylation of the OT gene in patients with AD [329]. Recently, an increase of the OT signal value and plasma OT concentration were correlated with the right parahippocampal gyrus volume in MRI images from the AD group but not in the images from the control group [183].

3.2.4. Vasopressin and Oxytocin in Autism

A growing body of evidence indicates that AVP and OT may play a role in the etiology of autism spectrum disorder (ASD) [172,176,177,330,331]. AVP concentrations in the CSF were found to be lower in children with autism, and AVP levels were associated with the severity of symptoms. Moreover, the AVP concentration in the CSF in neonates made it possible to predict a subsequent diagnosis of autism. No such associations were found for oxytocin; however, both OTR and V1aR mRNA levels were lower in autistic patients [169,170,171]. Studies on gene polymorphism of AVP receptors in autistic patients of the Korean population showed a significant association of ASD with SNP RS1 and SNP RS3 in the 5′flanking regions of the V1aR receptor [173,174,175]. The association between polymorphism of the V1aR and V1bR genes and autism was also studied in the North American population [172]. The authors reported that there may be a significant link between ASD and polymorphism of the AVP V1bR in SNP rs35369693 and SNP rs28632197. An improvement of social abilities and a reduction of anxiety symptoms after 4–5 weeks of intranasally applied AVP was reported by Parker et al. [176,177]. A systematic review and meta-analysis of the long-term intranasal application of OT in ASD showed that this type of treatment is well tolerated and safe [228]. The positive effects of AVP on social behavior in ASD have been shown in the studies of Hendaus et al. [332] and Parker et al. [177].
There is evidence for an association between OTR gene polymorphism and susceptibility to ASD. Based on a study encompassing 314 autism-affected families, Ylisaukko-Oja et al. suggested that the p24–26 region of chromosome 3 expressing the OTR gene may play a role in increasing susceptibility to the development of ASD [210]. In the Chinese population, polymorphism was present in the rs2254298 and rs53576 genes [211], in the Caucasian and Japanese populations, it was present in rs2254298 [212,213], in the European population, it was present in rs237887 [214], and in the North America population it was present in rs2268493, rs1042778, and rs7632287 [172,215]. Two meta-analyses, encompassing 8 studies [216] and 10 studies [217], reported associations between ASD and OTR SNP polymorphism in rs2254298, rs7632287, and rs2268491.
Lower plasma oxytocin levels have been observed in children with ASD [218,219,220,221]. Moreover, ASD subjects had lower levels of the bioactive amidated OT form and higher OT precursor levels, suggesting the altered processing of the OT peptide in the brains of children with autism [218]. Several studies have shown significant positive effects of OT application on autistic behavior. OT infusion was shown to ameliorate repetitive behavior in adults with ASD and Asperger’s disorder [222]. This finding was supported by a recent clinical trial based on a group of 106 ASD patients, in which intranasally applied OT reduced repetitive behavior and increased the time of gaze fixation on socially relevant regions [223]. In the same study, however, no improvement was found in the primary outcome, the Autism Diagnostic Observation Scheduled (ADOS)—a social reciprocity subscale with regard to the prevalence of adverse events [223]. Other randomized crossover trials performed on children with ASD showed that treatment with intranasal OT improved caregiver-rated social responsiveness [176,333]. In addition, intranasally applied OT enhanced learning in response to social targets and feedback, and this was correlated with the activation of the nucleus accumbens detected by functional MRI [224]. Other studies failed to find beneficial effects in relation to OT in ASD [225,226,227]. Although the present evidence is promising, further clinical studies are necessary to provide better insight into the role of oxytocin in the pathogenesis of ASD and the potential utility of this peptide in the treatment of ASD in humans.

3.2.5. Vasopressin and Oxytocin in Schizophrenia

Experimental and clinical studies indicate that the dysregulation of the VPS and OTS may play a role in the pathogenesis of schizophrenia [302,303,306,334]. Schizophrenia-like symptoms with impairment of social behavior were described in vasopressin-deficient (di/di) Brattleboro rats [165,166] and V1aR knockout mice [167]. In addition, significantly lower concentrations of AVP receptors in the prefrontal cortex and hypothalamus were found in the rat model of schizophrenia induced by prenatal exposure to methylazoxymethanol acetate (MAM) [168].
Post-mortem studies revealed lower AVP levels in the temporal cortex of schizophrenic patients [155]. Similarly, reduced AVP mRNA expression was found in the PVN of schizophrenic patients [156]. The blood AVP levels in patients with schizophrenia were either elevated or not altered [157,158,159]. Interestingly, a positive correlation was found between the blood AVP level and the severity of symptoms in female patients but not in male patients [159]. Some schizophrenic patients with inappropriately high blood AVP levels manifested polydipsia, hypoosmolality, and hyponatremia, which were further potentiated by antipsychotic treatment [160,161,162]. The intranasal application of DDAVP—a synthetic analog of AVP—increased the effectiveness of risperidone in reducing the negative symptoms of schizophrenia [163].
Significant associations between single nucleotide polymorphisms of AVP and OT genes and schizophrenia were detected in the chromosomal region 20p13, specifically in the loci of rs2740204 of the shared promoter of AVP and OT, in rs4813626 of the 5′promoter of OT, and in rs3011589 of the second intron of the AVP promoter [164].
Plasma OT concentrations were found to be lower in patients with schizophrenia than in healthy controls [158,203], and there was a negative correlation between OT levels and symptom severity in patients with schizophrenia [157]. Experiments on rats with the MAM model of schizophrenia revealed reduced concentrations of oxytocin and OTRs in the prefrontal cortex (PFC) and in the hypothalamus of the MAM schizophrenic model [168].
Positive effects related to the intranasal application of OT on social cognition and interpersonal reactivity have been reported in patients with schizophrenia [204,205]; however, clinical trials and meta-analyses have not provided evidence for a significant therapeutic effect of OT in relation to schizophrenia [206,207,208,209].

4. Conclusions

A survey of the literature shows that the RAS, VPS, and OTS, the classical endocrine systems regulating blood pressure and the water-electrolyte balance, are also potent regulators of other CNS processes through their actions on cerebral blood flow, the metabolism, and intercellular and intracellular signal transmission. Angiotensinogen, angiotensins, AVP, OT, and their respective receptors have been detected in the neurons, glial cells, and blood vessels of multiple brain regions regulating cardiovascular functions, pain, emotion, susceptibility to stress, as well as learning, memory, and cognitive processes. The RAS, VPS, and OTS innervate the same regions of the brain and in many instances are activated jointly and interact at the cellular level. The components of the RAS, VPS, and OTS have been identified in the forebrain (cortex, hypothalamus, circumventricular organs) and in the midbrain and hindbrain (PAG, DRN, RVLM, CVLM, NTS, NcAmb, DMVNc, AP). It has been shown that the RAS, VPS, and OTS act differently during stress, depression, and anxiety, as well as in neuropsychiatric and neurodegenerative diseases such as Alzheimer’s disease, Parkinson’s disease, autism, and schizophrenia (Table 1). Mutations of angiotensinogen, AT1Rs, and ACE genes were detected in some patients with depression and schizophrenia. In addition, polymorphism of V1bR and OTR genes was demonstrated in patients with depression and autism. It appears that the RAS, VPS, and OTS form a multifunctional cooperating triad which may have a significant impact on the efficacy of therapies of neurodegenerative diseases and psychiatric disorders. Clinical trials and meta-analyses indicate that specific compounds interfering with the action of the RAS, VPS, or OTS may improve the effectiveness of the treatment of neuropsychiatric and neurodegenerative diseases; however, further investigations are needed to establish the guidelines for their use for medical purposes. The present study does not address the putative role of these systems in other essential neurological and psychiatric disorders, such as bipolar disorder, ADHD, epilepsy, migraine headaaches, and additional resaerch in this field should be conducted. Better knowledge of the mechanisms of the actions of these compounds should be helpful in programming the most efficient individually-tailored treatment for patients suffering from the comorbidity of neuropsychiatric/neurodegenerative and cardiovascular diseases.

Author Contributions

E.S.-S. proposed the design of the review. E.S.-S., A.W., A.C.-J., K.C. and T.Ż. searched the literature and prepared the final design of the review. E.S.-S. and T.Ż. designed figures. A.W., K.C., T.Ż. and E.S.-S. prepared table. All authors wrote the first draft of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This review was prepared with the use of CePT infrastructure financed by the European Union—the European Regional Development Fund, and was supported by the governmental financial resources provided for the scientific activity of the Medical University of Warsaw. This review did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

The authors wish to express their gratitude to Marcin Kumosa for the preparation of the figures and to Edyta Lalik for secretarial cooperation.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

Aβ = amyloid beta, ACE = angiotensin converting enzyme 1, ACE2 = angiotensin converting enzyme 2, AD = Alzheimer’s disease, ADHD = attention deficit hyperactivity disorder, Agt = angiotensinogen, Ang = angiotensin, AP = area postrema, APA = aminopeptidase A, APB = aminopeptidase B, APN = aminopeptidase N, APP = amyloid precursor protein gene, ASD = autism spectrum disorder, AT1R = angiotensin receptor of type 1, AT2R = angiotensin receptor of type 2, AT4R = angiotensin receptor of type 4, AVP = arginine vasopressin, CNS = central nervous system, COVID-19 = coronavirus disease 2019, CSF = cerebrospinal fluid, CVDs = cardiovascular diseases, CVLM = caudal ventrolateral medulla, CVS = cardiovascular system, DALYs = disability adjusted life years, DDAVP = desmopressin, an analog of AVP, DMVNc = dorsal motor nucleus of the vagus, DRN = dorsal raphe nucleus, IL = interleukin, IRAP = insulin-regulated aminopeptidase, MAM = methylazoxymethanol acetate, MAPK = mitogen-activated protein kinase, MasR = MAS receptor for angiotensin-(1-7), MDD = major depressive disorder, MPTP = 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, NcAmb = nucleus ambiguous, NADPH = nicotinamide adenine dinucleotide phosphate, NEP = neutral endopeptidase, NTS = nucleus of the solitary tract, OTR = oxytocin receptor, OVLT = organum vasculosum laminae terminalis, OT = oxytocin, OTS = oxytocin system, PAG = periaqueductal gray, PD = Parkinson’s disease, PFC = prefrontal cortex, PVN = paraventricular nucleus, PPD = post-partum depression, RAS = renin–angiotensin system, ROS = reactive oxygen species, RVLM = rostral ventrolateral medulla, SARS-CoV = severe acute respiratory syndrome coronavirus, SARS-CoV-2 = severe acute respiratory syndrome associated coronavirus 2, SFO = subfornical organ, SON = supraoptic nucleus, SNP = single nucleotide polymorphism, TNF-α = tumor necrosis factor α, V1aR = vasopressin receptor of type 1a, V1bR = vasopressin receptor of type 1b, V2R = vasopressin receptor of type 2, VPS = vasopressin system.

References

  1. Roth, G.A.; Mensah, G.A.; Johnson, C.O.; Addolorato, G.; Ammirati, E.; Baddour, L.M.; Barengo, N.C.; GBD-NHLBI-JACC Global Burden of Cardiovascular Diseases Writing Group. Global Burden of Cardiovascular Diseases and Risk Factors, 1990–2019: Update From the GBD 2019 Study. J. Am. Coll. Cardiol. 2020, 76, 2982–3021. [Google Scholar] [CrossRef] [PubMed]
  2. Musunuru, K.; Ingelsson, E.; Fornage, M.; Liu, P.; Murphy, A.M.; Newby, L.K.; Newton Cheh, C.; Perez, M.V.; Voora, D.; Woo, D.; et al. Expressed Genome in Cardiovascular Diseases and Stroke: Refinement, Diagnosis, and Prediction: A Scientific Statement From the American Heart Association. Circ. Cardiovasc. Genet. 2017, 10, e000037. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Correll, C.U.; Solmi, M.; Veronese, N.; Bortolato, B.; Rosson, S.; Santonastaso, P.; Thapa Chhetri, N.; Fornaro, M.; Gallicchio, D.; Collantoni, E.; et al. Prevalence, incidence and mortality from cardiovascular disease in patients with pooled and specific severe mental illness: A large-scale meta-analysis of 3,211,768 patients and 113,383,368 controls. World Psychiatry 2017, 16, 163–180. [Google Scholar] [CrossRef] [Green Version]
  4. Ardell, J.L.; Andresen, M.C.; Armour, J.A.; Billman, G.E.; Chen, P.S.; Foreman, R.D.; Herring, N.; O’Leary, D.S.; Sabbah, H.N.; Schultz, H.D.; et al. Translational neurocardiology: Preclinical models and cardioneural integrative aspects. J. Physiol. 2016, 594, 3877–3909. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Dimsdale, J.E. Psychological stress and cardiovascular disease. J. Am. Coll. Cardiol. 2008, 51, 1237–1246. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Lang, U.E.; Borgwardt, S. Molecular mechanisms of depression: Perspectives on new treatment strategies. Cell Physiol. Biochem. 2013, 31, 761–777. [Google Scholar] [CrossRef] [PubMed]
  7. El Abdellati, K.; De Picker, L.; Morrens, M. Antipsychotic Treatment Failure: A Systematic Review on Risk Factors and Interventions for Treatment Adherence in Psychosis. Front. Neurosci. 2020, 14, 531763. [Google Scholar] [CrossRef]
  8. Imboden, H.; Patil, J.; Nussberger, J.; Nicoud, F.; Hess, B.; Ahmed, N.; Schaffner, T.; Wellner, M.; Müller, D.; Inagami, T.; et al. Endogenous angiotensinergic system in neurons of rat and human trigeminal ganglia. Regul. Pept. 2009, 154, 23–31. [Google Scholar] [CrossRef] [Green Version]
  9. Scigliano, G.; Ronchetti, G. Antipsychotic-induced metabolic and cardiovascular side effects in schizophrenia: A novel mechanistic hypothesis. CNS Drugs 2013, 27, 249–257. [Google Scholar] [CrossRef] [Green Version]
  10. Szczepanska-Sadowska, E.; Cudnoch-Jedrzejewska, A.; Ufnal, M.; Zera, T. Brain and cardiovascular diseases: Common neurogenic background of cardiovascular, metabolic and inflammatory diseases. J. Physiol. Pharmacol. 2010, 61, 509–521. [Google Scholar]
  11. Yang, T.; Richards, E.M.; Pepine, C.J.; Raizada, M.K. The gut microbiota and the brain-gut-kidney axis in hypertension and chronic kidney disease. Nat. Rev. Nephrol. 2018, 14, 442–456. [Google Scholar] [CrossRef] [PubMed]
  12. Critchley, H.D. The human cortex responds to an interoceptive challenge. Proc. Natl. Acad. Sci. USA 2004, 101, 6333–6334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Ikemoto, S. Brain reward circuitry beyond the mesolimbic dopamine system: A neurobiological theory. Neurosci. Biobehav. Rev. 2010, 35, 129–150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. McKlveen, J.M.; Myers, B.; Herman, J.P. The Medial Prefrontal Cortex: Coordinator of Autonomic, Neuroendocrine, and Behavioral Responses to Stress. J. Neuroendocrinol. 2015, 27, 446–456. [Google Scholar] [CrossRef] [Green Version]
  15. Rossi, M.A.; Stuber, G.D. Overlapping brain circuits for homeostatic and hedonic feeding. Cell Metab. 2018, 27, 42–56. [Google Scholar] [CrossRef]
  16. Szczepanska-Sadowska, E.; Czarzasta, K.; Cudnoch-Jedrzejewska, A. Dysregulation of the renin-angiotensin system and the vasopressinergic system; interactions in cardiovascular disorders. Curr. Hypertens. Rep. 2018, 20, 19. [Google Scholar] [CrossRef] [Green Version]
  17. Szczepanska-Sadowska, E.; Cudnoch-Jedrzejewska, A.; Sadowski, B. Differential role of specific cardiovascular neuropeptides in pain regulation: Relevance to cardiovascular diseases. Neuropeptides 2020, 81, 102046. [Google Scholar] [CrossRef]
  18. Correll, C.U.; Joffe, B.I.; Rosen, L.M.; Sullivan, T.B.; Joffe, R.T. Cardiovascular and cerebrovascular risk factors and events associated with second-generation antipsychotic compared to antidepressant use in a non-elderly adult sample: Results from a claims-based inception cohort study. World Psychiatry 2015, 14, 56–63. [Google Scholar] [CrossRef] [Green Version]
  19. Jain, S.; Goldstein, D.S. Cardiovascular dysautonomia in Parkinson Disease: From pathophysiology to pathogenesis. Neurobiol. Dis. 2012, 46, 572–580. [Google Scholar] [CrossRef] [Green Version]
  20. Wood, J.D. Enteric nervous system neuropathy: Repair and restoration. Curr. Opin. Gastroenterol. 2011, 27, 106–111. [Google Scholar] [CrossRef]
  21. Wood, J.D. Enteric neurobiology: Discoveries and directions. Adv. Exp. Med. Biol. 2016, 891, 175–191. [Google Scholar] [CrossRef] [PubMed]
  22. Pavel, J.; Benicky, J.; Murakami, Y.; Sanchez-Lemus, E.; Saavedra, J.M. Peripherally administered angiotensin II AT1 receptor antagonists are anti-stress compounds in vivo. Ann. N. Y. Acad. Sci. 2008, 1148, 360–366. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Saavedra, J.M.; Benicky, J. Brain and peripheral angiotensin II play a major role in stress. Stress 2007, 10, 185–193. [Google Scholar] [CrossRef]
  24. Saavedra, J.M.; Sánchez-Lemus, E.; Benicky, J. Blockade of brain angiotensin II AT1 receptors ameliorates stress, anxiety, brain inflammation and ischemia: Therapeutic implications. Psychoneuroendocrinology 2011, 36, 1–18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Cudnoch-Jedrzejewska, A.; Szczepanska-Sadowska, E.; Dobruch, J.; Puchalska, L.; Ufnal, M.; Kowalewski, S.; Wsół, A. Differential sensitisation to central cardiovascular effects of angiotensin II in rats with a myocardial infarct: Relevance to stress and interaction with vasopressin. Stress 2008, 11, 290–301. [Google Scholar] [CrossRef] [PubMed]
  26. Dobruch, J.; Cudnoch-Jedrzejewska, A.; Szczepanska-Sadowska, E. Enhanced involvement of brain vasopressin V1 receptors in cardiovascular responses to stress in rats with myocardial infarction. Stress 2005, 8, 273–284. [Google Scholar] [CrossRef] [PubMed]
  27. Ufnal, M.; Dudek, M.; Zera, T.; Szczepańska-Sadowska, E. Centrally administered interleukin-1 beta sensitizes to the central pressor action of angiotensin II. Brain Res. 2006, 1100, 64–72. [Google Scholar] [CrossRef]
  28. Kreyenbuhl, J.; Dickerson, F.B.; Medoff, D.R.; Brown, C.H.; Goldberg, R.W.; Fang, L.; Wohlheiter, K.; Mittal, L.P.; Dixon, L.B. Extent and management of cardiovascular risk factors in patients with type 2 diabetes and serious mental illness. J. Nerv. Ment. Dis. 2006, 194, 404–410. [Google Scholar] [CrossRef] [Green Version]
  29. Cosarderelioglu, C.; Nidadavolu, L.S.; George, C.J.; Oh, E.S.; Bennett, D.A.; Walston, J.D.; Abadir, P.M. Brain Renin-Angiotensin System at the Intersect of Physical and Cognitive Frailty. Front. Neurosci. 2020, 14, 586314. [Google Scholar] [CrossRef]
  30. Feng, Y.; Xia, H.; Santos, R.A.; Speth, R.; Lazartigues, E. Angiotensin-converting enzyme 2: A new target for neurogenic hypertension. Exp. Physiol. 2010, 95, 601–606. [Google Scholar] [CrossRef] [Green Version]
  31. McKinley, M.J.; Albiston, A.L.; Allen, A.M.; Mathai, M.L.; May, C.N.; McAllen, R.M.; Oldfield, B.J.; Mendelsohn, F.A.; Chai, S.Y. The brain renin-angiotensin system: Location and physiological roles. Int. J. Biochem. Cell Biol. 2003, 35, 901–918. [Google Scholar] [CrossRef]
  32. Paul, M.; Poyan Mehr, A.; Kreutz, R. Physiology of local renin-angiotensin systems. Physiol. Rev. 2006, 86, 747–803. [Google Scholar] [CrossRef] [PubMed]
  33. Labandeira-Garcia, J.L.; Rodríguez-Perez, A.I.; Garrido-Gil, P.; Rodriguez-Pallares, J.; Lanciego, J.L.; Guerra, M.J. Brain renin-angiotensin System and microglial polarization: Implications for aging and neurodegeneration. Front. Aging Neurosci. 2017, 9, 129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Zalba, G.; San José, G.; Moreno, M.U.; Fortuño, M.A.; Fortuño, A.; Beaumont, F.J.; Díez, J. Oxidative stress in arterial hypertension: Role of NAD(P)H oxidase. Hypertension 2001, 38, 1395–1399. [Google Scholar] [CrossRef] [PubMed]
  35. Horiuchi, M.; Akishita, M.; Dzau, V.J. Recent progress in angiotensin II type 2 receptor research in the cardiovascular system. Hypertension 1999, 33, 613–621. [Google Scholar] [CrossRef] [Green Version]
  36. Coleman, C.G.; Anrather, J.; Iadecola, C.; Pickel, V.M. Angiotensin II type 2 receptors have a major somatodendritic distribution in vasopressin-containing neurons in the mouse hypothalamic paraventricular nucleus. Neuroscience 2009, 63, 129–142. [Google Scholar] [CrossRef] [Green Version]
  37. Rivas-Santisteban, R.; Rodriguez-Perez, A.I.; Muñoz, A.; Reyes-Resina, I.; Labandeira-García, J.L.; Navarro, G.; Franco, R. Angiotensin AT1 and AT2 receptor heteromer expression in the hemilesioned rat model of Parkinson’s disease that increases with levodopa-induced dyskinesia. J. Neuroinflamm. 2020, 17, 243. [Google Scholar] [CrossRef]
  38. Albiston, A.L.; McDowall, S.G.; Matsacos, D.; Sim, P.; Clune, E.; Mustafa, T.; Lee, J.; Mendelsohn, F.A.; Simpson, R.J.; Connolly, L.M.; et al. Evidence that the angiotensin IV (AT(4)) receptor is the enzyme insulin-regulated aminopeptidase. J. Biol. Chem. 2001, 276, 48623–48626. [Google Scholar] [CrossRef] [Green Version]
  39. Benoist, C.C.; Kawas, L.H.; Zhu, M.; Tyson, K.A.; Stillmaker, L.; Appleyard, S.M.; Wright, J.W.; Wayman, G.A.; Harding, J.W. The procognitive and synaptogenic effects of angiotensin IV-derived peptides are dependent on activation of the hepatocyte growth factor/c-met system. J. Pharmacol. Exp. Ther. 2014, 351, 390–402. [Google Scholar] [CrossRef] [Green Version]
  40. Costa-Besada, M.A.; Valenzuela, R.; Garrido-Gil, P.; Villar-Cheda, B.; Parga, J.A.; Lanciego, J.L.; Labandeira-Garcia, J.L. Paracrine and intracrine angiotensin 1-7/Mas receptor axis in the substantia nigra of rodents, monkeys, and humans. Mol. Neurobiol. 2018, 55, 5847–5867. [Google Scholar] [CrossRef]
  41. Karamyan, V.T.; Stockmeier, C.A.; Speth, R.C. Human brain contains a novel non-AT1, non-AT2 binding site for active angiotensin peptides. Life Sci. 2008, 83, 421–425. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Labandeira-Garcia, J.L.; Valenzuela, R.; Costa-Besada, M.A.; Villar-Cheda, B.; Rodriguez-Perez, A.I. The intracellular renin-angiotensin system: Friend or foe. Some light from the dopaminergic neurons. Prog. Neurobiol. 2020, 8, 101919. [Google Scholar] [CrossRef] [PubMed]
  43. Lam, S.Y.; Liu, Y.; Ng, K.M.; Liong, E.C.; Tipoe, G.L.; Leung, P.S.; Fung, M.L. Upregulation of a local renin-angiotensin system in the rat carotid body during chronic intermittent hypoxia. Exp. Physiol. 2014, 99, 220–231. [Google Scholar] [CrossRef] [PubMed]
  44. Phillips, M.I.; de Oliveira, E.M. Brain renin angiotensin in disease. J. Mol. Med. 2008, 86, 715–722. [Google Scholar] [CrossRef]
  45. von Bohlen, O.; Halbach, O.; Albrecht, D. The CNS renin-angiotensin system. Cell Tissue Res. 2006, 326, 599–616. [Google Scholar] [CrossRef]
  46. Bunnemann, B.; Fuxe, K.; Ganten, D. The renin-angiotensin system in the brain: An update 1993. Regul. Pept. 1993, 46, 487–509. [Google Scholar] [CrossRef]
  47. Shan, Z.; Shi, P.; Cuadra, A.E.; Dong, Y.; Lamont, G.J.; Li, Q.; Seth, D.M.; Navar, L.G.; Katovich, M.J.; Sumners, C.; et al. Involvement of the brain (pro)renin receptor in cardiovascular homeostasis. Circ. Res. 2010, 107, 934–938. [Google Scholar] [CrossRef]
  48. Wang, W.-Z.; Gao, L.; Wang, H.-J.; Zucker, I.H.; Wang, W. Interaction between cardiac sympathetic afferent reflex and chemoreflex is mediated by the NTS AT1 receptors in heart failure. Am. J. Physiol. Heart Circ. Physiol. 2008, 295, H1216–H1226. [Google Scholar] [CrossRef] [Green Version]
  49. Rivas-Santisteban, R.; Lillo, J.; Muñoz, A.; Rodríguez-Pérez, A.I.; Labandeira-García, J.L.; Navarro, G.; Franco, R. Novel Interactions Involving the Mas Receptor Show Potential of the Renin-Angiotensin System in the Regulation of Microglia Activation: Altered Expression in Parkinsonism and Dyskinesia. Neurotherapeutics 2021, 18, 998–1016. [Google Scholar] [CrossRef]
  50. Rodriguez-Perez, A.I.; Sucunza, D.; Pedrosa, M.A.; Garrido-Gil, P.; Kulisevsky, J.; Lanciego, J.L.; Labandeira-Garcia, J.L. Angiotensin type 1 receptor antagonists protect against alpha-synuclein-induced neuroinflammation and dopaminergic neuron death. Neurotherapeutics 2018, 15, 1063–1081. [Google Scholar] [CrossRef] [Green Version]
  51. Rodriguez-Perez, A.I.; Garrido-Gil, P.; Pedrosa, M.A.; Garcia-Garrote, M.; Valenzuela, R.; Navarro, G.; Franco, R.; Labandeira-Garcia, J.L. Angiotensin type 2 receptors: Role in aging and neuroinflammation in the substantia nigra. Brain Behav. Immun. 2020, 87, 256–271. [Google Scholar] [CrossRef] [PubMed]
  52. Ufnal, M.; Zera, T.; Szczepańska-Sadowska, E. Blockade of angiotensin II AT1 receptors inhibits pressor action of centrally administered interleukin-1beta in Sprague Dawley rats. Neuropeptides 2005, 39, 581–585. [Google Scholar] [CrossRef] [PubMed]
  53. Ufnal, M.; Sikora, M.; Szczepanska-Sadowska, E. Interleukin-1 receptor antagonist reduces the magnitude of the pressor response to acute stress. Neurosci. Lett. 2008, 448, 47–51. [Google Scholar] [CrossRef] [PubMed]
  54. Zera, T.; Ufnal, M.; Szczepanska-Sadowska, E. Central TNF-alpha elevates blood pressure and sensitizes to central pressor action of angiotensin II in the infarcted rats. J. Physiol. Pharmacol. 2008, 59 (Suppl. 8), 117–121. [Google Scholar] [PubMed]
  55. Griendling, K.K.; Sorescu, D.; Ushio-Fukai, M. NAD(P)H oxidase: Role in cardiovascular biology and disease. Circ. Res. 2000, 86, 494–501. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. De Morais, S.D.B.; Shanks, J.; Zucker, I.H. Integrative Physiological Aspects of Brain RAS in Hypertension. Curr. Hypertens. Rep. 2018, 20, 10. [Google Scholar] [CrossRef]
  57. Thomas, W.G.; Sernia, C. Immunocytochemical localization of angiotensinogen in the rat brain. Neuroscience 1988, 25, 319–341. [Google Scholar] [CrossRef]
  58. Yang, G.; Gray, T.S.; Sigmund, C.D.; Cassell, M.D. The angiotensinogen gene is expressed in both astrocytes and neurons in murine central nervous system. Brain Res. 1999, 817, 123–131. [Google Scholar] [CrossRef]
  59. Huang, B.S.; Chen, A.; Ahmad, M.; Wang, H.W.; Leenen, F.H. Mineralocorticoid and AT1 receptors in the paraventricular nucleus contribute to sympathetic hyperactivity and cardiac dysfunction in rats post myocardial infarct. J. Physiol. 2014, 592, 3273–3286. [Google Scholar] [CrossRef] [Green Version]
  60. Kakar, S.S.; Riel, K.K.; Neill, J.D. Differential expression of angiotensin II receptor subtype mRNAs (AT-1A and AT-1B) in the brain. Biochem. Biophys. Res. Commun. 1992, 185, 688–692. [Google Scholar] [CrossRef]
  61. Lenkei, Z.; Corvol, P.; Llorens-Cortes, C. Comparative expression of vasopressin and angiotensin type-1 receptor mRNA in rat hypothalamic nuclei: A double in situ hybridization study. Brain Res. Mol. Brain Res. 1995, 34, 135–142. [Google Scholar] [CrossRef]
  62. Von Bohlen und Halbach, O.; Albrecht, D. Mapping of angiotensin AT1 receptors in the rat limbic system. Regul. Pept. 1998, 78, 51–56. [Google Scholar] [CrossRef]
  63. Gao, L.; Wang, W.; Li, Y.L.; Schultz, H.D.; Liu, D.; Cornish, K.G.; Zucker, I.H. Sympathoexcitation by central ANG II: Roles for AT1 receptor upregulation and NAD(P)H oxidase in RVLM. Am. J. Physiol. Heart Circ. Physiol. 2005, 288, H2271–H2279. [Google Scholar] [CrossRef] [PubMed]
  64. Gao, L.; Wang, W.Z.; Wang, W.; Zucker, I.H. Imbalance of angiotensin type 1 receptor and angiotensin II type 2 receptor in the rostral ventrolateral medulla: Potential mechanism for sympathetic overactivity in heart failure. Hypertension 2008, 52, 708–714. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Huang, J.; Hara, Y.; Anrather, J.; Speth, R.C.; Iadecola, C.; Pickel, V.M. Angiotensin II subtype 1A (AT1A) receptors in the rat sensory vagal complex: Subcellular localization and association with endogenous angiotensin. Neuroscience 2003, 122, 21–36. [Google Scholar] [CrossRef]
  66. Milik, E.; Szczepanska-Sadowska, E.; Dobruch, J.; Cudnoch-Jedrzejewska, A.; Maslinski, W. Altered expression of V1a receptors mRNA in the brain and kidney after myocardial infarction and chronic stress. Neuropeptides 2014, 48, 257–266. [Google Scholar] [CrossRef]
  67. Milik, E.; Cudnoch-Jedrzejewska, A.; Szczepanska-Sadowska, E. Effect of chronic mild stress on AT1 receptor messenger RNA expression in the brain and kidney of rats. Psychosom. Med. 2016, 78, 208–220. [Google Scholar] [CrossRef]
  68. Porcari, C.Y.; Araujo, I.G.; Urzedo-Rodrigues, L.; De Luca, L.A., Jr.; Menani, J.V.; Caeiro, X.E.; Imboden, H.; Antunes-Rodrigues, J.; Reis, L.C.; Vivas, L.; et al. Whole body sodium depletion modifies AT1 mRNA expression and serotonin content in the dorsal raphe nucleus. J. Neuroendocrinol. 2019, 31, e12703. [Google Scholar] [CrossRef]
  69. Valenzuela, R.; Costa-Besada, M.A.; Iglesias-Gonzalez, J.; Perez-Costas, E.; Villar-Cheda, B.; Garrido-Gil, P.; Melendez-Ferro, M.; Soto-Otero, R.; Lanciego, J.L.; Henrion, D.; et al. Mitochondrial angiotensin receptors in dopaminergic neurons. Role in cell protection and aging-related vulnerability to neurodegeneration. Cell Death Dis. 2016, 7, e2427. [Google Scholar] [CrossRef] [Green Version]
  70. Yamazato, M.; Yamazato, Y.; Sun, C.; Diez-Freire, C.; Raizada, M.K. Overexpression of angiotensin-converting enzyme 2 in the rostral ventrolateral medulla causes long-term decrease in blood pressure in the spontaneously hypertensive rats. Hypertension 2007, 49, 926–931. [Google Scholar] [CrossRef] [Green Version]
  71. Becker, L.K.; Etelvino, G.M.; Walther, T.; Santos, R.A.S.; Campagnole-Santos, M.J. Immunofluorescence localization of the receptor Mas in cardiovascular-related areas of the rat brain. Am. J. Physiol. Circ. Physiol. 2007, 293, H1416–H1424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Ferguson, A.V. Angiotensinergic regulation of autonomic and neuroendocrine outputs: Critical roles for the subfornical organ and paraventricular nucleus. Neuroendocrinology 2009, 89, 370–376. [Google Scholar] [CrossRef] [PubMed]
  73. Gaudet, E.A.; Godwin, S.J.; Lukoshkova, E.; Head, G.A. Effect of central endogenous angiotensin II on sympathetic activation induced by hypoxia. Clin. Exp. Hypertens. 1997, 19, 913–923. [Google Scholar] [CrossRef] [PubMed]
  74. Pitra, S.; Worker, C.J.; Feng, Y.; Stern, J.E. Exacerbated effects of prorenin on hypothalamic magnocellular neuronal activity and vasopressin plasma levels during salt-sensitive hypertension. Am. J. Physiol. Heart Circ. Physiol. 2019, 317, H496–H504. [Google Scholar] [CrossRef] [PubMed]
  75. Ahmad, Z.; Milligan, C.J.; Paton, J.F.; Deuchars, J. Angiotensin type 1 receptor immunoreactivity in the thoracic spinal cord. Brain Res. 2003, 985, 21–31. [Google Scholar] [CrossRef]
  76. Geerling, J.C.; Loewy, A.D. Aldosterone in the brain. Am. J. Physiol.-Renal Physiol. 2009, 297, F559–F576. [Google Scholar] [CrossRef]
  77. Huang, B.S.; White, R.A.; Ahmad, M.; Leenen, F.H. Role of brain corticosterone and aldosterone in central angiotensin II-induced hypertension. Hypertension 2013, 62, 564–571. [Google Scholar] [CrossRef] [Green Version]
  78. Łoń, S.; Szczepańska-Sadowska, E.; Szczypaczewska, M. Evidence that centrally released arginine vasopressin is involved in central pressor action of angiotensin II. Am. J. Physiol. 1996, 270 (Pt 2), H167–H173. [Google Scholar] [CrossRef]
  79. Paczwa, P.; Szczepańska-Sadowska, E.; Loń, S.; Ganten, S.L.; Ganten, D. Role of central AT1 and V1 receptors in cardiovascular adaptation to hemorrhage in SD and renin TGR rats. Am. J. Physiol. 1999, 276, H1918–H1926. [Google Scholar] [CrossRef] [Green Version]
  80. Sztechman, D.; Czarzasta, K.; Cudnoch-Jedrzejewska, A.; Szczepanska-Sadowska, E.; Zera, T. Aldosterone and mineralocorticoid receptors in regulation of the cardiovascular system and pathological remodelling of the heart and arteries. J. Physiol. Pharmacol. 2018, 69, 829–845. [Google Scholar] [CrossRef]
  81. Ufnal, M.; Sikora, M.; Zera, T.; Szczepanska-Sadowska, E. Simvastatin reduces pressor response to centrally administered angiotensin II. Am. J. Hypertens. 2010, 23, 956–959. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Zhang, Z.H.; Yu, Y.; Kang, Y.M.; Wei, S.G.; Felder, R.B. Aldosterone acts centrally to increase brain renin-angiotensin system activity and oxidative stress in normal rats. Am. J. Physiol. Heart Circ. Physiol. 2008, 294, H1067–H1074. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Zucker, I.H.; Schultz, H.D.; Patel, K.P.; Wang, W.; Gao, L. Regulation of central angiotensin type 1 receptors and sympathetic outflow in heart failure. Am. J. Physiol. Heart Circ. Physiol. 2009, 297, H1557–H1566. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Żera, T.; Ufnal, M.; Szczepańska-Sadowska, E. TNF and angiotensin type 1 receptors interact in the brain control of blood pressure in heart failure. Cytokine 2015, 71, 272–277. [Google Scholar] [CrossRef] [PubMed]
  85. Cudnoch-Jedrzejewska, A.; Dobruch, J.; Puchalska, L.; Szczepańska-Sadowska, E. Interaction of AT1 receptors and V1a receptors-mediated effects in the central cardiovascular control during the post-infarct state. Regul. Pept. 2007, 142, 86–94. [Google Scholar] [CrossRef]
  86. Cudnoch-Jedrzejewska, A.; Czarzasta, K.; Puchalska, L.; Dobruch, J.; Borowik, O.; Pachucki, J.; Szczepanska-Sadowska, E. Angiotensin converting enzyme inhibition reduces cardiovascular responses to acute stress in myocardially infarcted and chronically stressed rats. Biomed. Res. Int. 2014, 2014, 385082. [Google Scholar] [CrossRef]
  87. Huang, B.S.; Ahmad, M.; White, R.A.; Marc, Y.; Llorens-Cortes, C.; Leenen, F.H. Inhibition of brain angiotensin III attenuates sympathetic hyperactivity and cardiac dysfunction in rats post-myocardial infarction. Cardiovasc. Res. 2013, 97, 424–431. [Google Scholar] [CrossRef] [Green Version]
  88. Macova, M.; Pavel, J.; Saavedra, J.M. A peripherally administered, centrally acting angiotensin II AT2 antagonist selectively increases brain AT1 receptors and decreases brain tyrosine hydroxylase transcription, pituitary vasopressin and ACTH. Brain Res. 2009, 1250, 130–140. [Google Scholar] [CrossRef] [Green Version]
  89. Mitra, A.K.; Gao, L.; Zucker, I.H. Angiotensin II-induced upregulation of AT(1) receptor expression: Sequential activation of NF-kappaB and Elk-1 in neurons. Am. J. Physiol. Cell Physiol. 2010, 299, C561–C569. [Google Scholar] [CrossRef] [Green Version]
  90. Nunes, F.C.; Braga, V.A. Chronic angiotensin II infusion modulates angiotensin II type I receptor expression in the subfornical organ and the rostral ventrolateral medulla in hypertensive rats. J. Renin-Angiotensin-Aldosterone Syst. 2011, 12, 440–445. [Google Scholar] [CrossRef] [Green Version]
  91. Wei, S.G.; Yu, Y.; Zhang, Z.H.; Weiss, R.M.; Felder, R.B. Mitogen-activated protein kinases mediate upregulation of hypothalamic AT1 receptors in heart failure rats. Hypertension 2008, 52, 679–686. [Google Scholar] [CrossRef] [PubMed]
  92. Wang, L.; Hiller, H.; Smith, J.A.; de Kloet, A.D.; Krause, E.G. Angiotensin type 1a receptors in the paraventricular nucleus of the hypothalamus control cardiovascular reactivity and anxiety-like behavior in male mice. Physiol. Genomics 2016, 48, 667–676. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Chen, D.; Jancovski, N.; Bassi, J.K.; Nguyen-Huu, T.P.; Choong, Y.T.; Palma-Rigo, K.; Davern, P.J.; Gurley, S.B.; Thomas, W.G.; Head, G.A.; et al. Angiotensin type 1A receptors in C1 neurons of the rostral ventrolateral medulla modulate the pressor response to aversive stress. J. Neurosci. 2012, 32, 2051–2061. [Google Scholar] [CrossRef] [Green Version]
  94. Benicky, J.; Sánchez-Lemus, E.; Honda, M.; Pang, T.; Orecna, M.; Wang, J.; Leng, Y.; Chuang, D.M.; Saavedra, J.M. Angiotensin II AT1 Receptor Blockade Ameliorates Brain Inflammation. Neuropsychopharmacology 2011, 36, 857–870. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Dobruch, J.; Paczwa, P.; Łoń, S.; Khosla, M.C.; Szczepańska-Sadowska, E. Hypotensive function of the brain angiotensin-(1-7) in Sprague Dawley and renin transgenic rats. J. Physiol. Pharmacol. 2003, 54, 371–381. [Google Scholar] [PubMed]
  96. Xia, H.; Suda, S.; Bindom, S.; Feng, Y.; Gurley, S.B.; Seth, D.; Navar, L.G.; Lazartigues, E. ACE2-mediated reduction of oxidative stress in the central nervous system is associated with improvement of autonomic function. PLoS ONE 2011, 6, e22682. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Xia, H.; de Queiroz, T.M.; Sriramula, S.; Feng, Y.; Johnson, T.; Mungrue, I.N.; Lazartigues, E. Brain ACE2 overexpression reduces DOCA-salt hypertension independently of endoplasmic reticulum stress. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2015, 308, R370–R378. [Google Scholar] [CrossRef] [Green Version]
  98. Sink, K.M.; Leng, X.; Williamson, J.; Kritchevsky, S.B.; Yaffe, K.; Kulle, L.; Yasar, S.; Atkinson, H.; Robbins, M.; Psaty, B.; et al. Angiotensin-converting enzyme inhibitors and cognitive decline in older adults with hypertension: Results from the Cardiovascular Health Study. Arch. Intern. Med. 2009, 169, 1195–1202. [Google Scholar] [CrossRef]
  99. Yasar, S.; Moored, K.D.; Adam, A.; Zabel, F.; Chuang, Y.F.; Varma, V.R.; Carlson, M.C. Angiotensin II Blood Levels Are Associated with Smaller Hippocampal and Cortical Volumes in Cognitively Normal Older Adults. J. Alzheimers Dis. 2020, 75, 521–529. [Google Scholar] [CrossRef]
  100. Li, Z.; Cao, Y.; Li, L.; Liang, Y.; Tian, X.; Mo, N.; Liu, Y.; Li, M.; Chui, D.; Guo, X. Prophylactic angiotensin type 1 receptor antagonism confers neuroprotection in an aged rat model of postoperative cognitive dysfunction. Biochem. Biophys. Res. Commun. 2014, 449, 74–80. [Google Scholar] [CrossRef]
  101. Wilms, H.; Rosenstiel, P.; Unger, T.; Deuschl, G.; Lucius, R. Neuroprotection with angiotensin receptor antagonists: A review of the evidence and potential mechanisms. Am. J. Cardiovasc. Drugs 2005, 5, 245–253. [Google Scholar] [CrossRef] [PubMed]
  102. Wincewicz, D.; Braszko, J.J. Telmisartan attenuates cognitive impairment caused by chronic stress in rats. Pharmacol. Rep. 2014, 66, 436–441. [Google Scholar] [CrossRef] [PubMed]
  103. Braszko, J.J.; Walesiuk, A.; Wielgat, P. Cognitive effects attributed to angiotensin II may result from its conversion to angiotensin IV. J. Renin-Angiotensin-Aldosterone Syst. 2006, 7, 168–174. [Google Scholar] [CrossRef] [PubMed]
  104. Cao, C.; Hasegawa, Y.; Hayashi, K.; Takemoto, Y.; Kim-Mitsuyama, S. Chronic Angiotensin 1-7 Infusion Prevents Angiotensin-II-Induced Cognitive Dysfunction and Skeletal Muscle Injury in a Mouse Model of Alzheimer’s Disease. J. Alzheimers Dis. 2019, 69, 297–309. [Google Scholar] [CrossRef] [PubMed]
  105. Hellner, K.; Walther, T.; Schubert, M.; Albrecht, D. Angiotensin-(1-7) enhances LTP in the hippocampus through the G-protein-coupled receptor Mas. Mol. Cell. Neurosci. 2005, 29, 427–435. [Google Scholar] [CrossRef]
  106. Barnes, J.M.; Barnes, N.M.; Costall, B.; Horovitz, Z.P.; Ironside, J.W.; Naylor, R.J.; Williams, T.J. Angiotensin II inhibits cortical cholinergic function: Implications for cognition. J. Cardiovasc. Pharmacol. 1990, 16, 234–238. [Google Scholar] [CrossRef]
  107. Barnes, N.M.; Cheng, C.H.; Costall, B.; Naylor, R.J.; Williams, T.J.; Wischik, C.M. Angiotensin converting enzyme density is increased in temporal cortex from patients with Alzheimer’s disease. Eur. J. Pharmacol. 1991, 200, 289–292. [Google Scholar] [CrossRef]
  108. Savaskan, E.; Hock, C.; Olivieri, G.; Bruttel, S.; Rosenberg, C.; Hulette, C.; Müller-Spahn, F. Cortical alterations of angiotensin converting enzyme, angiotensin II and AT1 receptor in Alzheimer’s dementia. Neurobiol. Aging 2001, 22, 541–546. [Google Scholar] [CrossRef]
  109. Butterfield, D.A.; Swomley, A.M.; Sultana, R. Amyloid β-peptide (1-42)-induced oxidative stress in Alzheimer disease: Importance in disease pathogenesis and progression. Antioxid. Redox Signal. 2013, 19, 823–835. [Google Scholar] [CrossRef] [Green Version]
  110. Guimond, M.O.; Gallo-Payet, N. How does angiotensin AT(2) receptor activation help neuronal differentiation and improve neuronal pathological situations? Front. Endocrinol. 2012, 3, 164. [Google Scholar] [CrossRef] [Green Version]
  111. Masters, C.L.; Simms, G.; Weinman, N.A.; Multhaup, G.; McDonald, B.L.; Beyreuther, K. Amyloid plaque core protein in Alzheimer disease and Down syndrome. Proc. Natl. Acad. Sci. USA 1985, 82, 4245–4249. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Quitterer, U.; AbdAlla, S. Improvements of symptoms of Alzheimer’s disease by inhibition of the angiotensin system. Pharmacol. Res. 2020, 154, 104230. [Google Scholar] [CrossRef] [PubMed]
  113. Takeda, S.; Sato, N.; Takeuchi, D.; Kurinami, H.; Shinohara, M.; Niisato, K.; Kano, M.; Ogihara, T.; Rakugi, H.; Morishita, R. Angiotensin receptor blocker prevented beta amyloid-induced cognitive impairment associated with recovery of neurovascular coupling. Hypertension 2009, 54, 1345–1352. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Jiang, T.; Zhang, Y.-D.; Zhou, J.-S.; Zhu, X.-C.; Tian, Y.-Y.; Zhao, H.D.; Lu, H.; Gao, Q.; Tan, L.; Yu, J.T. Angiotensin-(1-7) is Reduced and Inversely Correlates with Tau Hyperphosphorylation in Animal Models of Alzheimer’s Disease. Mol. Neurobiol. 2016, 53, 2489. [Google Scholar] [CrossRef]
  115. Aguilera, G.; Kiss, A.; Luo, X. Increased expression of type 1 angiotensin II receptors in the hypothalamic paraventricular nucleus following stress and glucocorticoid administration. J. Neuroendocrinol. 1995, 7, 775–783. [Google Scholar] [CrossRef]
  116. Brasil, T.F.S.; Fassini, A.; Corrêa, F.M. AT1 and AT2 receptors in the prelimbic cortex modulate the cardiovascular response evoked by acute exposure to restraint stress in rats. Cell. Mol. Neurobiol. 2018, 38, 305–316. [Google Scholar] [CrossRef]
  117. Costa, A.C.O.; Becker, L.K.; Moraes, É.R.; Romero, T.R.L.; Guzzo, L.; Santos, R.A.S.; Duarte, I.D.G. Angiotensin-(1–7) induces peripheral antinociception through Mas receptor activation in an opioid-independent pathway. Pharmacology 2012, 89, 137–144. [Google Scholar] [CrossRef]
  118. Zhao, Y.; Qin, Y.; Liu, T.; Hao, D. Chronic nerve injury-induced Mas receptor expression in dorsal root ganglion neurons alleviates neuropathic pain. Exp. Ther. Med. 2015, 10, 2384–2388. [Google Scholar] [CrossRef] [Green Version]
  119. Saab, Y.B.; Gard, P.R.; Yeoman, M.S.; Mfarrej, B.; El-Moalem, H.; Ingram, M.J. Renin-angiotensin-system gene polymorphisms and depression. Prog. Neuropsychopharmacol. Biol. Psychiatry 2007, 31, 1113–1118. [Google Scholar] [CrossRef]
  120. Kangussu, L.M.; Almeida-Santos, A.F.; Bader, M.; Alenina, N.; Fontes, M.A.P.; Santos, R.A.S.; Aguiar, D.C.; Campagnole-Santos, M.J. Angiotensin-(1-7) attenuates the anxiety and depression-like behaviors in transgenic rats with low brain angiotensinogen. Behav. Brain Res. 2013, 257, 25–30. [Google Scholar] [CrossRef]
  121. Hajjar, I.; Kritchevsky, S.; Newman, A.B.; Li, R.; Yaffe, K.; Simonsick, E.M.; Lipsitz, L.A.; Health, Aging and Body Composition Study. Renin angiotensin system gene polymorphisms modify angiotensin-converting enzyme inhibitors’ effect on cognitive function: The health, aging and body composition study. J. Am. Geriatr. Soc. 2010, 58, 1035–1042. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Ward, K.M.; Kraal, A.Z.; Flowers, S.A.; Ellingrod, V.L. Cardiovascular Pharmacogenomics and Cognitive Function in Patients with Schizophrenia. Pharmacotherapy 2017, 37, 1122–1130. [Google Scholar] [CrossRef] [PubMed]
  123. Gadelha, A.; Vendramini, A.M.; Yonamine, C.M.; Nering, M.; Berberian, A.; Suiama, M.A.; Oliveira, V.; Lima-Landman, M.T.; Breen, G.; Bressan, R.A.; et al. Convergent evidences from human and animal studies implicate angiotensin I-converting enzyme activity in cognitive performance in schizophrenia. Transl. Psychiatry 2015, 5, e691. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Fan, X.; Song, X.; Zhao, M.; Jarskog, L.F.; Natarajan, R.; Shukair, N.; Freudenreich, O.; Henderson, D.C.; Goff, D.C. The effect of adjunctive telmisartan treatment on psychopathology and cognition in patients with schizophrenia. Acta Psychiatr. Scand. 2017, 136, 465–472. [Google Scholar] [CrossRef]
  125. Dominguez-Meijide, A.; Villar-Cheda, B.; Garrido-Gil, P.; Sierrra-Paredes, G.; Guerra, M.J.; Labandeira-Garcia, J.L. Effect of chronic treatment with angiotensin type 1 receptor antagonists on striatal dopamine levels in normal rats and in a rat model of Parkinson’s disease treated with L-DOPA. Neuropharmacology 2014, 76, 156–168. [Google Scholar] [CrossRef]
  126. Joglar, B.; Rodriguez-Pallares, J.; Rodriguez-Perez, A.I.; Rey, P.; Guerra, M.J.; Labandeira-Garcia, J.L. The inflammatory response in the MPTP model of Parkinson’s disease is mediated by brain angiotensin: Relevance to progression of the disease. J. Neurochem. 2009, 109, 656–669. [Google Scholar] [CrossRef]
  127. Sonsalla, P.K.; Coleman, C.; Wong, L.Y.; Harris, S.L.; Richardson, J.R.; Gadad, B.S.; Li, W.; German, D.C. The angiotensin converting enzyme inhibitor captopril protects nigrostriatal dopamine neurons in animal models of parkinsonism. Exp. Neurol. 2013, 250, 376–383. [Google Scholar] [CrossRef] [Green Version]
  128. Thakur, K.S.; Prakash, A.; Bisht, R.; Bansal, P.K. Beneficial effect of candesartan and lisinopril against haloperidol-induced tardive dyskinesia in rat. J. Renin-Angiotensin-Aldosterone Syst. 2015, 16, 917–929. [Google Scholar] [CrossRef] [Green Version]
  129. Zhou, Y.; Xu, J.; Hou, Y.; Leverenz, J.B.; Kallianpur, A.; Mehra, R.; Liu, Y.; Yu, H.; Pieper, A.A.; Jehi, L.; et al. Network medicine links SARS-CoV-2/COVID-19 infection to brain microvascular injury and neuroinflammation in dementia-like cognitive impairment. Alzheimer’s Res. Ther. 2021, 13, 110. [Google Scholar] [CrossRef]
  130. Purba, J.S.; Hoogendijk, W.J.; Hofman, M.A.; Swaab, D.F. Increased number of vasopressin- and oxytocin-expressing neurons in the paraventricular nucleus of the hypothalamus in depression. Arch. Gen. Psychiatry 1996, 53, 137–143. [Google Scholar] [CrossRef]
  131. Van West, D.; Del-Favero, J.; Aulchenko, Y.; Oswald, P.; Souery, D.; Forsgren, T.; Sluijs, S.; Bel-Kacem, S.; Adolfsson, R.; Mendlewicz, J.; et al. A major SNP haplotype of the arginine vasopressin 1B receptor protects against recurrent major depression. Mol. Psychiatry 2004, 9, 287–292. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Zai, C.C.; Muir, K.E.; Nowrouzi, B.; Shaikh, S.A.; Choi, E.; Berall, L.; Trépanier, M.O.; Beitchman, J.H.; Kennedy, J.L. Possible genetic association between vasopressin receptor 1B and child aggression. Psychiatry Res. 2012, 200, 784–788. [Google Scholar] [CrossRef] [PubMed]
  133. Ben-Efraim, Y.J.; Wasserman, D.; Wasserman, J.; Sokolowski, M.V. Family-based study of AVPR1B association and interaction with stressful life events on depression and anxiety in suicide attempts. Neuropsychopharmacology 2013, 38, 1504–1511. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Kamiya, M.; Sabia, H.D.; Marella, J.; Fava, M.; Nemeroff, C.B.; Umeuchi, H.; Iijima, M.; Chaki, S.; Nishino, I. Efficacy and safety of TS-121, a novel vasopressin V(1B) receptor antagonist, as adjunctive treatment for patients with major depressive disorder: A randomized, double-blind, placebo-controlled study. J. Psychiatr. Res. 2020, 128, 43–51. [Google Scholar] [CrossRef]
  135. Katz, D.A.; Locke, C.; Greco, N.; Liu, W.; Tracy, K.A. Hypothalamic-pituitary-adrenal axis and depression symptom effects of an arginine vasopressin type 1B receptor antagonist in a one-week randomized Phase 1b trial. Brain Behav. 2017, 7, e00628. [Google Scholar] [CrossRef] [Green Version]
  136. Agorastos, A.; Sommer, A.; Heinig, A.; Wiedemann, K.; Demiralay, C. Vasopressin Surrogate Marker Copeptin as a Potential Novel Endocrine Biomarker for Antidepressant Treatment Response in Major Depression: A Pilot Study. Front. Psychiatry 2020, 11, 453. [Google Scholar] [CrossRef]
  137. Griebel, G.; Simiand, J.; Serradeil-Le Gal, C.; Wagnon, J.; Pascal, M.; Scatton, B.; Maffrand, J.P.; Soubrie, P. Anxiolytic- and antidepressant-like effects of the non-peptide vasopressin V1b receptor antagonist, SSR149415, suggest an innovative approach for the treatment of stress-related disorders. Proc. Natl. Acad. Sci. USA 2002, 99, 6370–6375. [Google Scholar] [CrossRef] [Green Version]
  138. Iijima, M.; Chaki, S. An arginine vasopressin V1b antagonist, SSR149415 elicits antidepressant-like effects in an olfactory bulbectomy model. Prog. Neuropsychopharmacol. Biol. Psychiatry 2007, 31, 622–627. [Google Scholar] [CrossRef]
  139. Iijima, M.; Yoshimizu, T.; Shimazaki, T.; Tokugawa, K.; Fukumoto, K.; Kurosu, S.; Kuwada, T.; Sekiguchi, Y.; Chaki, S. Antidepressant and anxiolytic profiles of newly synthesized arginine vasopressin V1B receptor antagonists: TASP0233278 and TASP0390325. Br. J. Pharmacol. 2014, 171, 3511–3525. [Google Scholar] [CrossRef] [Green Version]
  140. Cudnoch-Jedrzejewska, A.; Puchalska, L.; Szczepanska-Sadowska, E.; Wsol, A.; Kowalewski, S.; Czarzasta, K. The effect of blockade of the central V1 vasopressin receptors on anhedonia in chronically stressed infarcted and non-infarcted rats. Physiol. Behav. 2014, 135, 208–214. [Google Scholar] [CrossRef]
  141. Cudnoch-Jedrzejewska, A.; Szczepanska-Sadowska, E.; Dobruch, J.; Gomolka, R.; Puchalska, L. Brain vasopressin V(1) receptors contribute to enhanced cardiovascular responses to acute stress in chronically stressed rats and rats with myocardial infarction. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2010, 298, R672–R680. [Google Scholar] [CrossRef] [PubMed]
  142. Cilz, N.I.; Cymerblit-Sabba, A.; Young, W.S. Oxytocin and vasopressin in the rodent hippocampus. Genes Brain Behav. 2019, 18, e12535. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Wsol, A.; Cudnoch-Jedrzejewska, A.; Szczepanska-Sadowska, E.; Kowalewski, S.; Dobruch, J. Central oxytocin modulation of acute stress-induced cardiovascular responses after myocardial infarction in the rat. Stress 2009, 12, 517–525. [Google Scholar] [CrossRef] [PubMed]
  144. Wsol, A.; Szczepanska-Sadowska, E.; Kowalewski, S.; Puchalska, L.; Cudnoch-Jedrzejewska, A. Oxytocin differently regulates pressor responses to stress in WKY and SHR rats: The role of central oxytocin and V1a receptors. Stress 2014, 17, 117–125. [Google Scholar] [CrossRef]
  145. Wsol, A.; Wojno, O.; Puchalska, L.; Wrzesien, R.; Szczepanska-Sadowska, E.; Cudnoch-Jedrzejewska, A. Impaired hypotensive effects of centrally acting oxytocin in SHR and WKY rats exposed to chronic mild stress. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2020, 318, R160–R172. [Google Scholar] [CrossRef]
  146. Breuer, M.E.; van Gaalen, M.M.; Wernet, W.; Claessens, S.E.; Oosting, R.S.; Behl, B.; Korte, S.M.; Schoemaker, H.; Gross, G.; Olivier, B.; et al. SSR149415, a non-peptide vasopressin V1b receptor antagonist, has long-lasting antidepressant effects in the olfactory bulbectomy-induced hyperactivity depression model. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2009, 379, 101–106. [Google Scholar] [CrossRef]
  147. Serradeil-Le Gal, C.; Derick, S.; Brossard, G.; Manning, M.; Simiand, J.; Gaillard, R.; Griebel, G.; Guillon, G. Functional and pharmacological characterization of the first specific agonist and antagonist for the V1b receptor in mammals. Stress 2003, 6, 199–206. [Google Scholar] [CrossRef]
  148. Raskind, M.A.; Peskind, E.R.; Lampe, T.H.; Risse, S.C.; Taborsky, G.J., Jr.; Dorsa, D. Cerebrospinal fluid vasopressin, oxytocin, somatostatin, and beta-endorphin in Alzheimer’s disease. Arch. Gen Psychiatry 1986, 43, 382–388. [Google Scholar] [CrossRef]
  149. Mazurek, M.F.; Beal, M.F.; Bird, E.D.; Martin, J.B. Vasopressin in Alzheimer’s disease: A study of postmortem brain concentrations. Ann. Neurol. 1986, 20, 665–670. [Google Scholar] [CrossRef]
  150. Goudsmit, E.; Filers, E.; Swaab, D.F. Changes in vasopressin neurons and fibers in aging and Alzheimer’s disease: Reversibility in the rat. Prog. Clin. Biol. Res. 1989, 317, 1193–1208. [Google Scholar]
  151. Meynen, G.; Unmehopa, U.A.; Hofman, M.A.; Swaab, D.F.; Hoogendijk, W.J. Hypothalamic vasopressin and oxytocin mRNA expression in relation to depressive state in Alzheimer’s disease: A difference with major depressive disorder. J. Neuroendocrinol. 2009, 21, 722–729. [Google Scholar] [CrossRef] [PubMed]
  152. Van Zwieten, E.J.; Ravid, R.; Hoogendijk, W.J.; Swaab, D.F. Stable vasopressin innervation in the degenerating human locus coeruleus in Alzheimer’s disease. Brain Res. 1994, 649, 329–333. [Google Scholar] [CrossRef] [Green Version]
  153. Zhang, X.; Zhao, F.; Wang, C.; Zhang, J.; Bai, Y.; Zhou, F.; Wang, Z.; Guo, J.; Qi, J. AVP(4-8) Improves Cognitive Behaviors and Hippocampal Synaptic Plasticity in the APP/PS1 Mouse Model of Alzheimer’s Disease. Neurosci. Bull. 2020, 36, 254–262. [Google Scholar] [CrossRef] [PubMed]
  154. Smith, A.S.; Williams Avram, S.K.; Cymerblit-Sabba, A.; Song, J.; Young, W.S. Targeted activation of the hippocampal CA2 area strongly enhances social memory. Mol. Psychiatry 2016, 21, 1137–1144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Frederiksen, S.O.; Ekman, R.; Gottfries, C.G.; Widerlöv, E.; Jonsson, S. Reduced concentrations of galanin, arginine vasopressin, neuropeptide Y and peptide YY in the temporal cortex but not in the hypothalamus of brains from schizophrenics. Acta Psychiatr. Scand. 1991, 83, 273–277. [Google Scholar] [CrossRef] [PubMed]
  156. Busch, J.R.; Jacobsen, C.; Lynnerup, N.; Banner, J.; Møller, M. Expression of vasopressin mRNA in the hypothalamus of individuals with a diagnosis of schizophrenia. Brain Behav. 2019, 9, e01355. [Google Scholar] [CrossRef] [Green Version]
  157. Guzel, D.; Yazici, A.B.; Pek, T.M.; Doganay, S.; Simsek, A.B.S.; Saglam, K.; Turan, C.; Yazici, E. Atrial natriuretic peptide and posterior pituitary neurohormone changes in patients with acute schizophrenia. Neuropsychiatr. Dis. Treat. 2018, 14, 1855–1860. [Google Scholar] [CrossRef] [Green Version]
  158. Liu, Y.; Tao, H.; Yang, X.; Huang, K.; Zhang, X.; Li, C. Decreased Serum Oxytocin and Increased Homocysteine in First-Episode Schizophrenia Patients. Front. Psychiatry 2019, 10, 217. [Google Scholar] [CrossRef] [Green Version]
  159. Rubin, L.H.; Carter, C.S.; Bishop, J.R.; Pournajafi-Nazarloo, H.; Harris, M.S.; Hill, S.K.; Reilly, J.L.; Sweeney, J.A. Peripheral vasopressin but not oxytocin relates to severity of acute psychosis in women with acutely-ill untreated first-episode psychosis. Schizophr. Res. 2013, 146, 138–143. [Google Scholar] [CrossRef] [Green Version]
  160. Goldman, M.B. The mechanism of life-threatening water imbalance in schizophrenia and its relationship to the underlying psychiatric illness. Brain Res. Rev. 2009, 61, 210–220. [Google Scholar] [CrossRef] [Green Version]
  161. Goldman, M.B.; Robertson, G.L.; Luchins, D.J.; Hedeker, D.; Pandey, G.N. Psychotic exacerbations and enhanced vasopressin secretion in schizophrenic patients with hyponatremia and polydipsia. Arch. Gen. Psychiatry 1997, 54, 443–449. [Google Scholar] [CrossRef] [PubMed]
  162. Ohsawa, H.; Kishimoto, T.; Shimayoshi, N.; Matsumura, K.; Tahara, K.; Kitera, K.; Higashiura, N.; Noriyama, Y.; Matsumoto, H.; Hirai, M.; et al. Atrial natriuretic peptide and arginine vasopressin secretion in schizophrenic patients. Acta Psychiatr. Scand. 1993, 88, 130–134. [Google Scholar] [CrossRef] [PubMed]
  163. Hosseini, S.M.; Farokhnia, M.; Rezaei, F.; Gougol, A.; Yekehtaz, H.; Iranpour, N.; Salehi, B.; Tabrizi, M.; Tajdini, M.; Ghaleiha, A.; et al. Intranasal desmopressin as an adjunct to risperidone for negative symptoms of schizophrenia: A randomized, double-blind, placebo-controlled, clinical trial. Eur. Neuropsychopharmacol. 2014, 24, 846–855. [Google Scholar] [CrossRef] [PubMed]
  164. Teltsh, O.; Kanyas-Sarner, K.; Rigbi, A.; Greenbaum, L.; Lerer, B.; Kohn, Y. Oxytocin and vasopressin genes are significantly associated with schizophrenia in a large Arab-Israeli pedigree. Int. J. Neuropsychopharmacol. 2012, 15, 309–319. [Google Scholar] [CrossRef] [Green Version]
  165. Cilia, J.; Gartlon, J.E.; Shilliam, C.; Dawson, L.A.; Moore, S.H.; Jones, D.N.C. Further neurochemical and behavioural investigation of Brattleboro rats as a putative model of schizophrenia. J. Psychopharmacol. 2010, 24, 407–419. [Google Scholar] [CrossRef]
  166. Demeter, K.; Török, B.; Fodor, A.; Varga, J.; Ferenczi, S.; Kovács, K.J.; Eszik, I.; Szegedi, V.; Zelena, D. Possible contribution of epigenetic changes in the development of schizophrenia-like behavior in vasopressin-deficient Brattleboro rats. Behav. Brain Res. 2016, 300, 123–134. [Google Scholar] [CrossRef] [Green Version]
  167. Egashira, N.; Tanoue, A.; Matsuda, T.; Koushi, E.; Harada, S.; Takano, Y.; Tsujimoto, G.; Mishima, K.; Iwasaki, K.; Fujiwara, M. Impaired social interaction and reduced anxiety-related behavior in vasopressin V1a receptor knockout mice. Behav. Brain Res. 2007, 178, 123–127. [Google Scholar] [CrossRef]
  168. Potasiewicz, A.; Holuj, M.; Litwa, E.; Gzielo, K.; Socha, L.; Popik, P.; Nikiforuk, A. Social dysfunction in the neurodevelopmental model of schizophrenia in male and female rats: Behavioural and biochemical studies. Neuropharmacology 2020, 170, 108040. [Google Scholar] [CrossRef]
  169. Oztan, O.; Garner, J.P.; Partap, S.; Sherr, E.H.; Hardan, A.Y.; Farmer, C.; Thurm, A.; Swedo, S.E.; Parker, K.J. Cerebrospinal fluid vasopressin and symptom severity in children with autism. Ann. Neurol. 2018, 84, 611–615. [Google Scholar] [CrossRef]
  170. Oztan, O.; Garner, J.P.; Constantino, J.N.; Parker, K.J. Neonatal CSF vasopressin concentration predicts later medical record diagnoses of autism spectrum disorder. Proc. Natl. Acad. Sci. USA 2020, 117, 10609–10613. [Google Scholar] [CrossRef]
  171. Oztan, O.; Jackson, L.P.; Libove, R.A.; Sumiyoshi, R.D.; Phillips, J.M.; Garner, J.P.; Hardan, A.Y.; Parker, K.J. Biomarker discovery for disease status and symptom severity in children with autism. Psychoneuroendocrinology 2018, 89, 39–45. [Google Scholar] [CrossRef] [PubMed]
  172. Francis, S.M.; Kim, S.J.; Kistner-Griffin, E.; Guter, S.; Cook, E.H.; Jacob, S. ASD and Genetic Associations with Receptors for Oxytocin and Vasopressin-AVPR1A, AVPR1B, and OXTR. Front. Neurosci. 2016, 10, 516. [Google Scholar] [CrossRef] [PubMed]
  173. Cataldo, I.; Azhari, A.; Esposito, G. A Review of Oxytocin and Arginine-Vasopressin Receptors and Their Modulation of Autism Spectrum Disorder. Front. Mol. Neurosci. 2018, 11, 27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Yang, S.Y.; Cho, S.C.; Yoo, H.J.; Cho, I.H.; Park, M.; Kim, B.N.; Kim, J.W.; Shin, M.S.; Park, T.W.; Son, J.W.; et al. Association study between single nucleotide polymorphisms in promoter region of AVPR1A and Korean autism spectrum disorders. Neurosci. Lett. 2010, 479, 197–200. [Google Scholar] [CrossRef] [PubMed]
  175. Yang, S.Y.; Cho, S.C.; Yoo, H.J.; Cho, I.H.; Park, M.; Yoe, J.; Kim, S.A. Family-based association study of microsatellites in the 5′ flanking region of AVPR1A with autism spectrum disorder in the Korean population. Psychiatry Res. 2010, 178, 199–201. [Google Scholar] [CrossRef]
  176. Parker, K.J.; Oztan, O.; Libove, R.A.; Sumiyoshi, R.D.; Jackson, L.P.; Karhson, D.S.; Summers, J.E.; Hinman, K.E.; Motonaga, K.S.; Phillips, J.M.; et al. Intranasal oxytocin treatment for social deficits and biomarkers of response in children with autism. Proc. Natl. Acad. Sci. USA 2017, 114, 8119–8124. [Google Scholar] [CrossRef] [Green Version]
  177. Parker, K.J.; Oztan, O.; Libove, R.A.; Mohsin, N.; Karhson, D.S.; Sumiyoshi, R.D.; Summers, J.E.; Hinman, K.E.; Motonaga, K.S.; Phillips, J.M.; et al. A randomized placebo-controlled pilot trial shows that intranasal vasopressin improves social deficits in children with autism. Sci. Transl. Med. 2019, 11, eaau7356. [Google Scholar] [CrossRef]
  178. Brambilla, M.; Manenti, R.; de Girolamo, G.; Adenzato, M.; Bocchio-Chiavetto, L.; Cotelli, M. Effects of Intranasal Oxytocin on Long-Term Memory in Healthy Humans: A Systematic Review. Drug Dev. Res. 2016, 77, 479–488. [Google Scholar] [CrossRef] [Green Version]
  179. Lucassen, P.J.; Tilders, F.J.; Salehi, A.; Swaab, D.F. Neuropeptides vasopressin (AVP), oxytocin (OXT) and corticotropin-releasing hormone (CRH) in the human hypothalamus: Activity changes in aging, Alzheimer’s disease and depression. Aging 1997, 9 (Suppl. 4), 48–50. [Google Scholar] [CrossRef]
  180. Mazurek, M.F.; Beal, M.F.; Bird, E.D.; Martin, J.B. Oxytocin in Alzheimer’s disease: Postmortem brain levels. Neurology 1987, 37, 1001–1003. [Google Scholar] [CrossRef]
  181. Van Zwieten, E.J.; Ravid, R.; Swaab, D.F. Differential vasopressin and oxytocin innervation of the human parabrachial nucleus: No changes in Alzheimer’s disease. Brain Res. 1996, 711, 146–152. [Google Scholar] [CrossRef] [Green Version]
  182. Wierda, M.; Goudsmit, E.; van der Woude, P.F.; Purba, J.S.; Hofman, M.A.; Bogte, H.; Swaab, D.F. Oxytocin cell number in the human paraventricular nucleus remains constant with aging and in Alzheimer’s disease. Neurobiol. Aging 1991, 12, 511–516. [Google Scholar] [CrossRef] [Green Version]
  183. Petekkaya, E.; Burakgazi, G.; Kuş, B.; Melek, İ.M.; Arpacı, A. Comparative study of the volume of the temporal lobe sections and neuropeptide effect in Alzheimer’s patients and healthy persons. Int. J. Neurosci. 2021, 131, 725–734. [Google Scholar] [CrossRef]
  184. Anderberg, U.M.; Uvnäs-Moberg, K. Plasma oxytocin levels in female fibromyalgia syndrome patients. Z. Rheumatol. 2000, 59, 373–379. [Google Scholar] [CrossRef] [PubMed]
  185. Dai, D.; Li, Q.C.; Zhu, Q.B.; Hu, S.H.; Balesar, R.; Swaab, D.; Bao, A.M. Direct Involvement of Androgen Receptor in Oxytocin Gene Expression: Possible Relevance for Mood Disorders. Neuropsychopharmacology 2017, 42, 2064–2071. [Google Scholar] [CrossRef] [PubMed]
  186. Meynen, G.; Unmehopa, U.A.; Hofman, M.A.; Swaab, D.F.; Hoogendijk, W.J. Hypothalamic oxytocin mRNA expression and melancholic depression. Mol. Psychiatry 2007, 12, 118–119. [Google Scholar] [CrossRef] [Green Version]
  187. Ozsoy, S.; Esel, E.; Kula, M. Serum oxytocin levels in patients with depression and the effects of gender and antidepressant treatment. Psychiatry Res. 2009, 169, 249–252. [Google Scholar] [CrossRef]
  188. Scantamburlo, G.; Hansenne, M.; Fuchs, S.; Pitchot, W.; Maréchal, P.; Pequeux, C.; Ansseau, M.; Legros, J.J. Plasma oxytocin levels and anxiety in patients with major depression. Psychoneuroendocrinology 2007, 32, 407–410. [Google Scholar] [CrossRef] [Green Version]
  189. Tsai, T.Y.; Tseng, H.H.; Chi, M.H.; Chang, H.H.; Wu, C.K.; Yang, Y.K.; Chen, P.S. The Interaction of Oxytocin and Social Support, Loneliness, and Cortisol Level in Major Depression. Clin. Psychopharmacol. Neurosci. 2019, 17, 487–494. [Google Scholar] [CrossRef] [Green Version]
  190. Holt-Lunstad, J.; Birmingham, W.; Light, K.C. The influence of depressive symptomatology and perceived stress on plasma and salivary oxytocin before, during and after a support enhancement intervention. Psychoneuroendocrinology 2011, 36, 1249–1256. [Google Scholar] [CrossRef]
  191. Parker, K.J.; Kenna, H.A.; Zeitzer, J.M.; Keller, J.; Blasey, C.M.; Amico, J.A.; Schatzberg, A.F. Preliminary evidence that plasma oxytocin levels are elevated in major depression. Psychiatry Res. 2010, 178, 359–362. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Warrener, C.D.; Valentin, E.M.; Gallin, C.; Richey, L.; Ross, D.B.; Hood, C.J.; Lori, A.; Cubells, J.; Rauch, S.A.M.; Rilling, J.K. The role of oxytocin signaling in depression and suicidality in returning war veterans. Psychoneuroendocrinology 2021, 126, 105085. [Google Scholar] [CrossRef] [PubMed]
  193. Thomas, S.; Larkin, T. Plasma cortisol and oxytocin levels predict help-seeking intentions for depressive symptoms. Psychoneuroendocrinology 2018, 87, 159–165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Jobst, A.; Sabaß, L.; Hall, D.; Brücklmeier, B.; Buchheim, A.; Hall, J.; Sarubin, N.; Zill, P.; Falkai, P.; Brakemeier, E.L.; et al. Oxytocin plasma levels predict the outcome of psychotherapy: A pilot study in chronic depression. J. Affect. Disord. 2018, 227, 206–213. [Google Scholar] [CrossRef]
  195. Costa, B.; Pini, S.; Martini, C.; Abelli, M.; Gabelloni, P.; Ciampi, O.; Muti, M.; Gesi, C.; Lari, L.; Cardini, A.; et al. Mutation analysis of oxytocin gene in individuals with adult separation anxiety. Psychiatry Res. 2009, 168, 87–93. [Google Scholar] [CrossRef]
  196. Costa, B.; Pini, S.; Gabelloni, P.; Abelli, M.; Lari, L.; Cardini, A.; Muti, M.; Gesi, C.; Landi, S.; Galderisi, S.; et al. Oxytocin receptor polymorphisms and adult attachment style in patients with depression. Psychoneuroendocrinology 2009, 34, 1506–1514. [Google Scholar] [CrossRef]
  197. Thompson, R.J.; Parker, K.J.; Hallmayer, J.F.; Waugh, C.E.; Gotlib, I.H. Oxytocin receptor gene polymorphism (rs2254298) interacts with familial risk for psychopathology to predict symptoms of depression and anxiety in adolescent girls. Psychoneuroendocrinology 2011, 36, 144–147. [Google Scholar] [CrossRef] [Green Version]
  198. Costa, B.; Pini, S.; Baldwin, D.S.; Silove, D.; Manicavasagar, V.; Abelli, M.; Coppedè, F.; Martini, C. Oxytocin receptor and G-protein polymorphisms in patients with depression and separation anxiety. J. Affect. Disord. 2017, 218, 365–373. [Google Scholar] [CrossRef] [Green Version]
  199. Choi, D.; Tsuchiya, K.J.; Takei, N. Interaction effect of oxytocin receptor (OXTR) rs53576 genotype and maternal postpartum depression on child behavioural problems. Sci. Rep. 2019, 9, 7685. [Google Scholar] [CrossRef]
  200. Parris, M.S.; Grunebaum, M.F.; Galfalvy, H.C.; Andronikashvili, A.; Burke, A.K.; Yin, H.; Min, E.; Huang, Y.Y.; Mann, J.J. Attempted suicide and oxytocin-related gene polymorphisms. J. Affect. Disord. 2018, 238, 62–68. [Google Scholar] [CrossRef]
  201. Neumann, I.D.; Torner, L.; Wigger, A. Brain oxytocin: Differential inhibition of neuroendocrine stress responses and anxiety-related behaviour in virgin, pregnant and lactating rats. Neuroscience 2000, 95, 567–575. [Google Scholar] [CrossRef]
  202. Stuebe, A.M.; Grewen, K.; Meltzer-Brody, S. Association between maternal mood and oxytocin response to breastfeeding. J. Women’s Health 2013, 22, 352–361. [Google Scholar] [CrossRef] [PubMed]
  203. Jobst, A.; Dehning, S.; Ruf, S.; Notz, T.; Buchheim, A.; Henning-Fast, K.; Meißner, D.; Meyer, S.; Bondy, B.; Müller, N.; et al. Oxytocin and vasopressin levels are decreased in the plasma of male schizophrenia patients. Acta Neuropsychiatr. 2014, 26, 347–355. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Halverson, T.; Jarskog, L.F.; Pedersen, C.; Penn, D. Effects of oxytocin on empathy, introspective accuracy, and social symptoms in schizophrenia: A 12-week twice-daily randomized controlled trial. Schizophr. Res. 2019, 204, 178–182. [Google Scholar] [CrossRef]
  205. Woolley, J.D.; Chuang, B.; Lam, O.; Lai, W.; O’Donovan, A.; Rankin, K.P.; Mathalon, D.H.; Vinogradov, S. Oxytocin administration enhances controlled social cognition in patients with schizophrenia. Psychoneuroendocrinology 2014, 47, 116–125. [Google Scholar] [CrossRef] [Green Version]
  206. Dagani, J.; Sisti, D.; Abelli, M.; Di Paolo, L.; Pini, S.; Raimondi, S.; Rocchi, M.B.; Saviotti, F.M.; Scocco, P.; Totaro, S.; et al. Do we need oxytocin to treat schizophrenia? A randomized clinical trial. Schizophr. Res. 2016, 172, 158–164. [Google Scholar] [CrossRef]
  207. Jarskog, L.F.; Pedersen, C.A.; Johnson, J.L.; Hamer, R.M.; Rau, S.W.; Elliott, T.; Penn, D.L. A 12-week randomized controlled trial of twice-daily intranasal oxytocin for social cognitive deficits in people with schizophrenia. Schizophr. Res. 2017, 185, 88–95. [Google Scholar] [CrossRef]
  208. Oya, K.; Matsuda, Y.; Matsunaga, S.; Kishi, T.; Iwata, N. Efficacy and safety of oxytocin augmentation therapy for schizophrenia: An updated systematic review and meta-analysis of randomized, placebo-controlled trials. Eur. Arch. Psychiatry Clin. Neurosci. 2016, 266, 439–450. [Google Scholar] [CrossRef]
  209. Williams, D.R.; Bürkner, P.C. Effects of intranasal oxytocin on symptoms of schizophrenia: A multivariate Bayesian meta-analysis. Psychoneuroendocrinology 2017, 75, 141–151. [Google Scholar] [CrossRef]
  210. Ylisaukko-oja, T.; Alarcón, M.; Cantor, R.M.; Auranen, M.; Vanhala, R.; Kempas, E.; von Wendt, L.; Järvelä, I.; Geschwind, D.H.; Peltonen, L. Search for autism loci by combined analysis of Autism Genetic Resource Exchange and Finnish families. Ann. Neurol. 2006, 59, 145–155. [Google Scholar] [CrossRef]
  211. Wu, S.; Jia, M.; Ruan, Y.; Liu, J.; Guo, Y.; Shuang, M.; Gong, X.; Zhang, Y.; Yang, X.; Zhang, D. Positive association of the oxytocin receptor gene (OXTR) with autism in the Chinese Han population. Biol. Psychiatry 2005, 58, 74–77. [Google Scholar] [CrossRef] [PubMed]
  212. Jacob, S.; Brune, C.W.; Carter, C.S.; Leventhal, B.L.; Lord, C.; Cook, E.H., Jr. Association of the oxytocin receptor gene (OXTR) in Caucasian children and adolescents with autism. Neurosci. Lett. 2007, 417, 6–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Liu, X.; Kawamura, Y.; Shimada, T.; Otowa, T.; Koishi, S.; Sugiyama, T.; Nishida, H.; Hashimoto, O.; Nakagami, R.; Tochigi, M.; et al. Association of the oxytocin receptor (OXTR) gene polymorphisms with autism spectrum disorder (ASD) in the Japanese population. J. Hum. Genet. 2010, 55, 137–141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Skuse, D.H.; Lori, A.; Cubells, J.F.; Lee, I.; Conneely, K.N.; Puura, K.; Lehtimäki, T.; Binder, E.B.; Young, L.J. Common polymorphism in the oxytocin receptor gene (OXTR) is associated with human social recognition skills. Proc. Natl. Acad. Sci. USA 2014, 111, 1987–1992. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  215. Campbell, D.B.; Datta, D.; Jones, S.T.; Batey Lee, E.; Sutcliffe, J.S.; Hammock, E.A.; Levitt, P. Association of oxytocin receptor (OXTR) gene variants with multiple phenotype domains of autism spectrum disorder. J. Neurodev. Disord. 2011, 3, 101–112. [Google Scholar] [CrossRef] [Green Version]
  216. LoParo, D.; Waldman, I.D. The oxytocin receptor gene (OXTR) is associated with autism spectrum disorder: A meta-analysis. Mol. Psychiatry 2015, 20, 640–646. [Google Scholar] [CrossRef] [PubMed]
  217. Kranz, T.M.; Kopp, M.; Waltes, R.; Sachse, M.; Duketis, E.; Jarczok, T.A.; Degenhardt, F.; Görgen, K.; Meyer, J.; Freitag, C.M.; et al. Meta-analysis and association of two common polymorphisms of the human oxytocin receptor gene in autism spectrum disorder. Autism Res. 2016, 9, 1036–1045. [Google Scholar] [CrossRef]
  218. Green, L.; Fein, D.; Modahl, C.; Feinstein, C.; Waterhouse, L.; Morris, M. Oxytocin and autistic disorder: Alterations in peptide forms. Biol. Psychiatry 2001, 50, 609–613. [Google Scholar] [CrossRef]
  219. Husarova, V.M.; Lakatosova, S.; Pivovarciova, A.; Babinska, K.; Bakos, J.; Durdiakova, J.; Kubranska, A.; Ondrejka, I.; Ostatnikova, D. Plasma Oxytocin in Children with Autism and Its Correlations with Behavioral Parameters in Children and Parents. Psychiatry Investig. 2016, 13, 174–183. [Google Scholar] [CrossRef]
  220. John, S.; Jaeggi, A.V. Oxytocin levels tend to be lower in autistic children: A meta-analysis of 31 studies. Autism 2021, 25, 2152–2161. [Google Scholar] [CrossRef]
  221. Modahl, C.; Green, L.; Fein, D.; Morris, M.; Waterhouse, L.; Feinstein, C.; Levin, H. Plasma oxytocin levels in autistic children. Biol. Psychiatry 1998, 43, 270–277. [Google Scholar] [CrossRef]
  222. Hollander, E.; Novotny, S.; Hanratty, M.; Yaffe, R.; DeCaria, C.M.; Aronowitz, B.R.; Mosovich, S. Oxytocin infusion reduces repetitive behaviors in adults with autistic and Asperger’s disorders. Neuropsychopharmacology 2003, 28, 193–198. [Google Scholar] [CrossRef] [PubMed]
  223. Yamasue, H.; Okada, T.; Munesue, T.; Kuroda, M.; Fujioka, T.; Uno, Y.; Matsumoto, K.; Kuwabara, H.; Mori, D.; Okamoto, Y.; et al. Effect of intranasal oxytocin on the core social symptoms of autism spectrum disorder: A randomized clinical trial. Mol. Psychiatry 2020, 25, 1849–1858. [Google Scholar] [CrossRef] [PubMed]
  224. Kruppa, J.A.; Gossen, A.; Weiß, E.O.; Kohls, G.; Großheinrich, N.; Cholemkery, H.; Freitag, C.M.; Karges, W.; Wölfle, E.; Sinzig, J.; et al. Neural modulation of social reinforcement learning by intranasal oxytocin in male adults with high-functioning autism spectrum disorder: A randomized trial. Neuropsychopharmacology 2019, 44, 749–756. [Google Scholar] [CrossRef] [PubMed]
  225. Bernaerts, S.; Boets, B.; Bosmans, G.; Steyaert, J.; Alaerts, K. Behavioral effects of multiple-dose oxytocin treatment in autism: A randomized, placebo-controlled trial with long-term follow-up. Mol. Autism 2020, 11, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  226. Dadds, M.R.; MacDonald, E.; Cauchi, A.; Williams, K.; Levy, F.; Brennan, J. Nasal oxytocin for social deficits in childhood autism: A randomized controlled trial. J. Autism Dev. Disord. 2014, 44, 521–531. [Google Scholar] [CrossRef]
  227. Henneberry, E.; Lamy, M.; Dominick, K.C.; Erickson, C.A. Decades of Progress in the Psychopharmacology of Autism Spectrum Disorder. J. Autism Dev. Disord. 2021, 51, 4370–4394. [Google Scholar] [CrossRef]
  228. Cai, Q.; Feng, L.; Yap, K.Z. Systematic review and meta-analysis of reported adverse events of long-term intranasal oxytocin treatment for autism spectrum disorder. Psychiatry Clin. Neurosci. 2018, 72, 140–151. [Google Scholar] [CrossRef]
  229. Wincewicz, D.; Braszko, J.J. Validation of Brain Angiotensin System Blockade as a Novel Drug Target in Pharmacological Treatment of Neuropsychiatric Disorders. Pharmacopsychiatry 2017, 50, 233–247. [Google Scholar] [CrossRef]
  230. Royea, J.; Hamel, E. Brain angiotensin II and angiotensin IV receptors as potential Alzheimer’s disease therapeutic targets. Geroscience 2020, 42, 1237–1256. [Google Scholar] [CrossRef]
  231. Thomas, W.G.; Mendelsohn, F.A. Angiotensin receptors: Form and function and distribution. Int. J. Biochem. Cell Biol. 2003, 35, 774–779. [Google Scholar] [CrossRef]
  232. Armando, I.; Carranza, A.; Nishimura, Y.; Barontini, M.; Ito, T.; Saavedra, J.M. Candesartan decreases the sympatho-adrenal and hormonal response to isolation stress. J. Renin-Angiotensin-Aldosterone Syst. 2001, 2 (Suppl. 1), S130–S135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  233. Armando, I.; Carranza, A.; Nishimura, Y.; Hoe, K.L.; Barontini, M.; Terrón, J.A.; Falcón-Neri, A.; Ito, T.; Juorio, A.V.; Saavedra, J.M. Peripheral administration of an angiotensin II AT(1) receptor antagonist decreases the hypothalamic-pituitary-adrenal response to isolation stress. Endocrinology 2001, 142, 3880–3889. [Google Scholar] [CrossRef] [PubMed]
  234. Costa-Ferreira, W.; Vieira, J.O.; Almeida, J.; Gomes-de-Souza, L.; Crestani, C.C. Involvement of Type 1 Angiontensin II Receptor (AT1) in Cardiovascular Changes Induced by Chronic Emotional Stress: Comparison between Homotypic and Heterotypic Stressors. Front. Pharmacol. 2016, 7, 262. [Google Scholar] [CrossRef] [Green Version]
  235. Pelegrini-da-Silva, A.; Martins, A.R.; Prado, W.A. A new role for the renin-angiotensin system in the rat periaqueductal gray matter: Angiotensin receptor-mediated modulation of nociception. Neuroscience 2005, 132, 453–463. [Google Scholar] [CrossRef]
  236. Raghavendra, V.; Chopra, K.; Kulkarni, S.K. Brain renin angiotensin system (RAS) in stress-induced analgesia and impaired retention. Peptides 1999, 20, 335–342. [Google Scholar] [CrossRef]
  237. Kumar, A.; Bilker, W.; Lavretsky, H.; Gottlieb, G. Volumetric asymmetries in late-onset mood disorders: An attenuation of frontal asymmetry with depression severity. Psychiatry Res. 2000, 100, 41–47. [Google Scholar] [CrossRef]
  238. Pandya, M.; Altinay, M.; Malone, D.A., Jr.; Anand, A. Where in the brain is depression? Curr. Psychiatry Rep. 2012, 14, 634–642. [Google Scholar] [CrossRef] [Green Version]
  239. Drevets, W.C.; Price, J.L.; Simpson, J.R., Jr.; Todd, R.D.; Reich, T.; Vannier, M.; Raichle, M.E. Subgenual prefrontal cortex abnormalities in mood disorders. Nature 1997, 386, 824–827. [Google Scholar] [CrossRef]
  240. Mayberg, H.S.; Liotti, M.; Brannan, S.K.; McGinnis, S.; Mahurin, R.K.; Jerabek, P.A.; Silva, J.A.; Tekell, J.L.; Martin, C.C.; Lancaster, J.L.; et al. Reciprocal limbic-cortical function and negative mood: Converging PET findings in depression and normal sadness. Am. J. Psychiatry 1999, 156, 675–682. [Google Scholar] [CrossRef]
  241. Vogt, B.A. Midcingulate cortex: Structure, connections, homologies, functions and diseases. J. Chem. Neuroanat. 2016, 74, 28–46. [Google Scholar] [CrossRef] [PubMed]
  242. Coffey, C.E.; Wilkinson, W.E.; Weiner, R.D.; Parashos, I.A.; Djang, W.T.; Webb, M.C.; Figiel, G.S.; Spritzer, C.E. Quantitative cerebral anatomy in depression. A controlled magnetic resonance imaging study. Arch. Gen. Psychiatry 1993, 50, 7–16. [Google Scholar] [CrossRef] [PubMed]
  243. Schweitzer, I.; Tuckwell, V.; Ames, D.; O’Brien, J. Structural neuroimaging studies in late-life depression: A review. World J. Biol. Psychiatry 2001, 2, 83–88. [Google Scholar] [CrossRef] [PubMed]
  244. Sprengelmeyer, R.; Steele, J.D.; Mwangi, B.; Kumar, P.; Christmas, D.; Milders, M.; Matthews, K. The insular cortex and the neuroanatomy of major depression. J. Affect. Disord. 2011, 133, 120–127. [Google Scholar] [CrossRef]
  245. Gard, P.R. Implications of the angiotensin converting enzyme gene insertion/deletion polymorphism in health and disease: A snapshot review. Int. J. Mol. Epidemiol. Genet. 2010, 1, 145–157. [Google Scholar]
  246. Zannas, A.S.; McQuoid, D.R.; Payne, M.E.; MacFall, J.R.; Ashley-Koch, A.; Steffens, D.C.; Potter, G.G.; Taylor, W.D. Association of gene variants of the renin-angiotensin system with accelerated hippocampal volume loss and cognitive decline in old age. Am. J. Psychiatry 2014, 171, 1214–1221. [Google Scholar] [CrossRef] [Green Version]
  247. Ferreira-Vieira, T.H.; Guimaraes, I.M.; Silva, F.R.; Ribeiro, F.M. Alzheimer’s disease: Targeting the Cholinergic System. Curr. Neuropharmacol. 2016, 14, 101–115. [Google Scholar] [CrossRef] [Green Version]
  248. Huang, Y.; Mucke, L. Alzheimer mechanisms and therapeutic strategies. Cell 2012, 148, 1204–1222. [Google Scholar] [CrossRef] [Green Version]
  249. Wang, J.; Ho, L.; Chen, L.; Zhao, Z.; Zhao, W.; Qian, X.; Humala, N.; Seror, I.; Bartholomew, S.; Rosendorff, C.; et al. Valsartan lowers brain beta-amyloid protein levels and improves spatial learning in a mouse model of Alzheimer disease. J. Clin. Investig. 2007, 117, 3393–3402. [Google Scholar] [CrossRef]
  250. AbdAlla, S.; Langer, A.; Fu, X.; Quitterer, U. ACE inhibition with captopril retards the development of signs of neurodegeneration in an animal model of Alzheimer’s disease. Int. J. Mol. Sci. 2013, 14, 16917–16942. [Google Scholar] [CrossRef]
  251. Anderson, C.; Teo, K.; Gao, P.; Arima, H.; Dans, A.; Unger, T.; Commerford, P.; Dyal, L.; Schumacher, H.; Pogue, J.; et al. Renin-angiotensin system blockade and cognitive function in patients at high risk of cardiovascular disease: Analysis of data from the ONTARGET and TRANSCEND studies. Lancet Neurol. 2011, 10, 43–53. [Google Scholar] [CrossRef]
  252. Chang-Quan, H.; Hui, W.; Chao-Min, W.; Zheng-Rong, W.; Jun-Wen, G.; Yong-Hong, L.; Yan-You, L.; Qing-Xiu, L. The association of antihypertensive medication use with risk of cognitive decline and dementia: A meta-analysis of longitudinal studies. Int. J. Clin. Pract. 2011, 65, 1295–1305. [Google Scholar] [CrossRef] [PubMed]
  253. Ohrui, T.; Tomita, N.; Sato-Nakagawa, T.; Matsui, T.; Maruyama, M.; Niwa, K.; Arai, H.; Sasaki, H. Effects of brain-penetrating ACE inhibitors on Alzheimer disease progression. Neurology 2004, 63, 1324–1325. [Google Scholar] [CrossRef] [PubMed]
  254. Tedesco, M.A.; Ratti, G.; Di Salvo, G.; Natale, F. Does the angiotensin II receptor antagonist losartan improve cognitive function? Drugs Aging 2002, 19, 723–732. [Google Scholar] [CrossRef] [PubMed]
  255. Zhuang, S.; Wang, H.F.; Wang, X.; Li, J.; Xing, C.M. The association of renin-angiotensin system blockade use with the risks of cognitive impairment of aging and Alzheimer’s disease: A meta-analysis. J. Clin. Neurosci. 2016, 33, 32–38. [Google Scholar] [CrossRef]
  256. Barthold, D.; Joyce, G.; Diaz Brinton, R.; Wharton, W.; Kehoe, P.G.; Zissimopoulos, J. Association of combination statin and antihypertensive therapy with reduced Alzheimer’s disease and related dementia risk. PLoS ONE 2020, 15, e0229541. [Google Scholar] [CrossRef] [Green Version]
  257. Valappil, R.A.; Black, J.E.; Broderick, M.J.; Carrillo, O.; Frenette, E.; Sullivan, S.S.; Goldman, S.M.; Tanner, C.M.; Langston, J.W. Exploring the electrocardiogram as a potential tool to screen for premotor Parkinson’s disease. Mov. Disord. 2010, 25, 2296–2303. [Google Scholar] [CrossRef]
  258. Wright, J.W.; Harding, J.W. Importance of the brain Angiotensin system in Parkinson’s disease. Parkinson’s Dis. 2012, 2012, 860923. [Google Scholar] [CrossRef] [Green Version]
  259. Su, G.; Dou, H.; Zhao, L.; Wang, H.; Liu, G.; Huang, B.; Peng, B. The angiotensin-converting enzyme (ACE) I/D polymorphism in Parkinson’s disease. J. Renin-Angiotensin-Aldosterone Syst. 2015, 16, 428–433. [Google Scholar] [CrossRef] [Green Version]
  260. Attar, R.; Wester, A.; Koul, S.; Eggert, S.; Polcwiartek, C.; Jernberg, T.; Erlinge, D.; Andell, P. Higher risk of major adverse cardiac events after acute myocardial infarction in patients with schizophrenia. Open Heart 2020, 7, e001286. [Google Scholar] [CrossRef]
  261. Castillejos, M.C.; Martín-Pérez, C.; García-Ruiz, A.; Mayoral-Cleries, F.; Moreno-Küstner, B. Recording of cardiovascular risk factors by general practitioners in patients with schizophrenia. Ann. Gen. Psychiatry 2020, 19, 34. [Google Scholar] [CrossRef] [PubMed]
  262. Daumit, G.L.; Goff, D.C.; Meyer, J.M.; Davis, V.G.; Nasrallah, H.A.; McEvoy, J.P.; Rosenheck, R.; Davis, S.M.; Hsiao, J.K.; Stroup, T.S.; et al. Antipsychotic effects on estimated 10-year coronary heart disease risk in the CATIE schizophrenia study. Schizophr. Res. 2008, 105, 175–187. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  263. Norman, S.M.; Sullivan, K.M.; Liu, F.; DiPaula, B.A.; Jose, P.A.; Kitchen, C.A.; Feldman, S.M.; Kelly, D.L. Blood Pressure and Heart Rate Changes during Clozapine Treatment. Psychiatr. Q. 2017, 88, 545–552. [Google Scholar] [CrossRef] [PubMed]
  264. Oh, S.J.; Fan, X. The Possible Role of the Angiotensin System in the Pathophysiology of Schizophrenia: Implications for Pharmacotherapy. CNS Drugs 2019, 33, 539–547. [Google Scholar] [CrossRef] [PubMed]
  265. Kugathasan, P.; Horsdal, H.T.; Aagaard, J.; Jensen, S.E.; Laursen, T.M.; Nielsen, R.E. Association of Secondary Preventive Cardiovascular Treatment after Myocardial Infarction with Mortality among Patients with Schizophrenia. JAMA Psychiatry 2018, 75, 1234–1240. [Google Scholar] [CrossRef] [Green Version]
  266. Song, G.G.; Lee, Y.H. The insertion/deletion polymorphism in the angiotensin-converting enzyme and susceptibility to schizophrenia or Parkinson’s disease: A meta-analysis. J. Renin-Angiotensin-Aldosterone Syst. 2015, 16, 434–442. [Google Scholar] [CrossRef]
  267. Kruse, D.; Pantelis, C.; Rudd, R.; Quek, J.; Herbert, P.; McKinley, M. Treatment of psychogenic polydipsia: Comparison of risperidone and olanzapine, and the effects of an adjunctive angiotensin-II receptor blocking drug (irbesartan). Aust. N. Z. J. Psychiatry 2001, 35, 65–68. [Google Scholar] [CrossRef]
  268. Firouzabadi, N.; Ghazanfari, N.; Alavi Shoushtari, A.; Erfani, N.; Fathi, F.; Bazrafkan, M.; Bahramali, E. Genetic Variants of Angiotensin-Converting Enzyme Are Linked to Autism: A Case-Control Study. PLoS ONE 2016, 11, e0153667. [Google Scholar] [CrossRef] [Green Version]
  269. Welcome, M.O.; Mastorakis, N.E. Neuropathophysiology of coronavirus disease 2019: Neuroinflammation and blood brain barrier disruption are critical pathophysiological processes that contribute to the clinical symptoms of SARS-CoV-2 infection. Inflammopharmacology 2021, 29, 939–963. [Google Scholar] [CrossRef]
  270. Magalhaes, G.S.; Rodrigues-Machado, M.D.G.; Motta-Santos, D.; Campagnole-Santos, M.J.; Santos, R.A.S. Activation of Ang-(1-7)/Mas Receptor Is a Possible Strategy to Treat Coronavirus (SARS-CoV-2) Infection. Front. Physiol. 2020, 11, 730. [Google Scholar] [CrossRef]
  271. Zamai, L. The Yin and Yang of ACE/ACE2 pathways: The rationale for the use of renin-angiotensin system inhibitors in COVID-19 patients. Cells 2000, 9, 1704. [Google Scholar] [CrossRef] [PubMed]
  272. Yan, R.; Zhang, Y.; Li, Y.; Xia, L.; Guo, Y.; Zhou, Q. Structural basis for the recognition of SARS-CoV-2 by full-length human ACE2. Science 2020, 367, 1444–1448. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  273. Doobay, M.F.; Talman, L.S.; Obr, T.D.; Tian, X.; Davisson, R.L.; Lazartigues, E. Differential expression of neuronal ACE2 in transgenic mice with overexpression of the brain renin-angiotensin system. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2007, 292, R373–R381. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  274. Gallagher, P.E.; Chappell, M.C.; Ferrario, C.M.; Tallant, E.A. Distinct roles for ANG II and ANG-(1-7) in the regulation of angiotensin-converting enzyme 2 in rat astrocytes. Am. J. Physiol. Cell. Physiol. 2006, 290, C420–C426. [Google Scholar] [CrossRef] [PubMed]
  275. Sakima, A.; Averill, D.B.; Gallagher, P.E.; Kasper, S.O.; Tommasi, E.N.; Ferrario, C.M.; Diz, D.I. Impaired heart rate baroreflex in older rats: Role of endogenous angiotensin-(1-7) at the nucleus tractus solitarii. Hypertension 2005, 46, 333–340. [Google Scholar] [CrossRef] [Green Version]
  276. Buzhdygan, T.P.; DeOre, B.J.; Baldwin-Leclair, A.; Bullock, T.A.; McGary, H.M.; Khan, J.A.; Razmpour, R.; Hale, J.F.; Galie, P.A.; Potula, R.; et al. The SARS-CoV-2 spike protein alters barrier function in 2D static and 3D microfluidic in-vitro models of the human blood-brain barrier. Neurobiol. Dis. 2020, 146, 105131. [Google Scholar] [CrossRef]
  277. Kehoe, P.G.; Wong, S.; Al Mulhim, N.; Palmer, L.E.; Miners, J.S. Angiotensin-converting enzyme 2 is reduced in Alzheimer’s disease in association with increasing amyloid-β and tau pathology. Alzheimer’s Res. Ther. 2016, 8, 50. [Google Scholar] [CrossRef] [Green Version]
  278. Chen, R.; Wang, K.; Yu, J.; Howard, D.; French, L.; Chen, Z.; Wen, C.; Xu, Z. The Spatial and Cell-Type Distribution of SARS-CoV-2 Receptor ACE2 in the Human and Mouse Brains. Front. Neurol. 2021, 11, 573095. [Google Scholar] [CrossRef]
  279. McMahon, C.L.; Staples, H.; Gazi, M.; Carrion, R.; Hsieh, J. SARS-CoV-2 targets glial cells in human cortical organoids. Stem Cell Rep. 2021, 16, 1156–1164. [Google Scholar] [CrossRef]
  280. Netland, J.; Meyerholz, D.K.; Moore, S.; Cassell, M.; Perlman, S. Severe acute respiratory syndrome coronavirus infection causes neuronal death in the absence of encephalitis in mice transgenic for human ACE2. J. Virol. 2008, 82, 7264–7275. [Google Scholar] [CrossRef] [Green Version]
  281. Ding, Y.; He, L.; Zhang, Q.; Huang, Z.; Che, X.; Hou, J.; Wang, H.; Shen, H.; Qiu, L.; Li, Z.; et al. Organ distribution of severe acute respiratory syndrome (SARS) associated coronavirus (SARS-CoV) in SARS patients: Implications for pathogenesis and virus transmission pathways. J. Pathol. 2004, 203, 622–630. [Google Scholar] [CrossRef] [PubMed]
  282. Xu, J.; Zhong, S.; Liu, J.; Li, L.; Li, Y.; Wu, X.; Li, Z.; Deng, P.; Zhang, J.; Zhong, N.; et al. Detection of severe acute respiratory syndrome coronavirus in the brain: Potential role of the chemokine mig in pathogenesis. Clin. Infect. Dis. 2005, 41, 1089–1096. [Google Scholar] [CrossRef] [Green Version]
  283. Song, E.; Zhang, C.; Israelow, B.; Lu-Culligan, A.; Prado, A.V.; Skriabine, S.; Lu, P.; Weizman, O.E.; Liu, F.; Dai, Y.; et al. Neuroinvasion of SARS-CoV-2 in human and mouse brain. J. Exp. Med. 2021, 218, e20202135. [Google Scholar] [CrossRef] [PubMed]
  284. Brognara, F.; Felippe, I.S.A.; Salgado, H.C.; Paton, J.F.R. Autonomic innervation of the carotid body as a determinant of its sensitivity: Implications for cardiovascular physiology and pathology. Cardiovasc. Res. 2021, 117, 1015–1032. [Google Scholar] [CrossRef] [PubMed]
  285. Machado, B.H.; Paton, J.F.R. Relevance of carotid bodies in COVID-19: A hypothetical viewpoint. Auton Neurosci. 2021, 233, 102810. [Google Scholar] [CrossRef] [PubMed]
  286. Porzionato, A.; Emmi, A.; Stocco, E.; Barbon, S.; Boscolo-Berto, R.; Macchi, V.; De Caro, R. The potential role of the carotid body in COVID-19. Am. J. Physiol. Lung Cell. Mol. Physiol. 2020, 319, L620–L626. [Google Scholar] [CrossRef] [PubMed]
  287. Villadiego, J.; Ramírez-Lorca, R.; Cala, F.; Labandeira-García, J.L.; Esteban, M.; Toledo-Aral, J.J.; López-Barneo, J. Is Carotid Body Infection Responsible for Silent Hypoxemia in COVID-19 Patients? Function 2020, 2, zqaa032. [Google Scholar] [CrossRef]
  288. Lambermont, B.; Davenne, E.; Maclot, F.; Delvenne, P. SARS-CoV-2 in carotid body. Intensive Care Med. 2021, 47, 342–343. [Google Scholar] [CrossRef]
  289. Szczepanska-Sadowska, E.; Wsol, A.; Cudnoch-Jedrzejewska, A.; Żera, T. Complementary role of oxytocin and vasopressin in cardiovascular regulation. Int. J. Mol. Sci. 2021, 22, 11465. [Google Scholar] [CrossRef]
  290. Szczepanska-Sadowska, E.; Zera, T.; Sosnowski, P.; Cudnoch-Jedrzejewska, A.; Puszko, A.; Misicka, A. Vasopressin and related peptides; potential value in diagnosis, prognosis and treatment of clinical disorders. Curr. Drug Metab. 2017, 18, 306–345. [Google Scholar] [CrossRef]
  291. Gimpl, G.; Fahrenholz, F. The oxytocin receptor system: Structure, function, and regulation. Physiol. Rev. 2001, 81, 629–683. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  292. Góźdź, A.; Szczepańska-Sadowska, E.; Maśliński, W.; Kumosa, M.; Szczepańska, K.; Dobruch, J. Differential expression of vasopressin V1a and V1b receptors mRNA in the brain of renin transgenic TGR (mRen2) 27 and Sprague–Dawley rats. Brain Res. Bull. 2003, 59, 399–3403. [Google Scholar] [CrossRef]
  293. Hallbeck, M.; Blomqvist, A. Spinal cord-projecting vasopressinergic neurons in the rat paraventricular hypothalamus. J. Comp. Neurol. 1999, 411, 201–211. [Google Scholar] [CrossRef]
  294. Hallbeck, M.; Hermanson, O.; Blomqvist, A. Distribution of preprovasopressin mRNA in the rat central nervous system. J. Comp. Neurol. 1999, 411, 181–200. [Google Scholar] [CrossRef]
  295. Phillips, P.A.; Abrahams, J.M.; Kelly, J.; Paxinos, G.; Grzonka, Z.; Mendelsohn, F.A.; Johnston, C.I. Localization of vasopressin binding sites in rat brain by in vitro autoradiography using a radioiodinated V1 receptor antagonist. Neuroscience 1988, 27, 749–761. [Google Scholar] [CrossRef]
  296. Young, L.; Toloczko, D.; Insel, R. Localization of vasopressin (V1a) receptor binding and mRNA in the rhesus monkey brain. J. Neuroendocrinol. 1999, 11, 291–298. [Google Scholar] [CrossRef]
  297. Czarzasta, K.; Wojno, O.; Zera, T.; Puchalska, L.; Dobruch, J.; Cudnoch-Jedrzejewska, A. The influence of post-infarct heart failure and high fat diet on the expression of apelin APJ and vasopressin V1a and V1b receptors. Neuropeptides 2019, 78, 101975. [Google Scholar] [CrossRef]
  298. Mathai, M.L.; Evered, M.D.; McKinley, M.J. Central losartan blocks natriuretic, vasopressin, and pressor responses to central hypertonic NaCl in sheep. Am. J. Physiol. 1998, 275, R548–R554. [Google Scholar] [CrossRef]
  299. Schrier, R.W. Interactions between angiotensin II and arginine vasopressin in water homeostasis. Kidney Int. 2009, 76, 137–139. [Google Scholar] [CrossRef] [Green Version]
  300. Caldwell, H.K. Oxytocin and Vasopressin: Powerful Regulators of Social Behavior. Neuroscientist 2017, 23, 517–528. [Google Scholar] [CrossRef]
  301. Caldwell, H.K.; Aulino, E.A.; Rodriguez, K.M.; Witchey, S.K.; Yaw, A.M. Social Context, Stress, Neuropsychiatric Disorders, and the Vasopressin 1b Receptor. Front. Neurosci. 2017, 11, 567. [Google Scholar] [CrossRef] [PubMed]
  302. De Wied, D. Neuropeptides as psychotropic drugs. Acta Neuropsychiatr. 1992, 4, 1–7. [Google Scholar] [CrossRef] [PubMed]
  303. De Wied, D.; Sigling, H.O. Neuropeptides involved in the pathophysiology of schizophrenia and major depression. Neurotox. Res. 2002, 4, 453–468. [Google Scholar] [CrossRef] [PubMed]
  304. Dwyer, J.B.; Aftab, A.; Radhakrishnan, R.; Widge, A.; Rodriguez, C.I.; Carpenter, L.L.; Nemeroff, C.B.; McDonald, W.M.; Kalin, N.H.; APA Council of Research Task Force on Novel Biomarkers and Treatments. Hormonal Treatments for Major Depressive Disorder: State of the Art. Am. J. Psychiatry 2020, 177, 686–705. [Google Scholar] [CrossRef]
  305. Frank, E.; Landgraf, R. The vasopressin system-from antidiuresis to psychopathology. Eur. J. Pharmacol. 2008, 583, 226–242. [Google Scholar] [CrossRef]
  306. Iovino, M.; Messana, T.; De Pergola, G.; Iovino, E.; Dicuonzo, F.; Guastamacchia, E.; Giagulli, V.A.; Triggiani, V. The Role of Neurohypophyseal Hormones Vasopressin and Oxytocin in Neuropsychiatric Disorders. Endocr. Metab. Immune Disord. Drug Targets Actions 2018, 18, 341–347. [Google Scholar] [CrossRef] [PubMed]
  307. Kunitake, Y.; Imamura, Y.; Mizoguchi, Y.; Matsushima, J.; Tateishi, H.; Murakawa-Hirachi, T.; Nabeta, H.; Kawashima, T.; Kojima, N.; Yamada, S.; et al. Serum Oxytocin Levels and Logical Memory in Older People in Rural Japan. J. Geriatr. Psychiatry Neurol. 2021, 34, 156–161. [Google Scholar] [CrossRef]
  308. De Winter, R.F.; van Hemert, A.M.; De Rijk, R.H.; Zwinderman, K.H.; Frankhuijzen-Sierevogel, A.C.; Wiegant, V.M.; Goekoop, J.G. Anxious-retarded depression: Relation with plasma vasopressin and cortisol. Neuropsychopharmacology 2003, 28, 140–147. [Google Scholar] [CrossRef] [Green Version]
  309. Gjerris, A.; Hammer, M.; Vendsborg, P.; Christensen, N.J.; Rafaelsen, O.J. Cerebrospinal fluid vasopressin-changes in depression. Br. J. Psychiatry 1985, 147, 696–701. [Google Scholar] [CrossRef]
  310. Rubin, L.H.; Carter, C.S.; Bishop, J.R.; Pournajafi-Nazarloo, H.; Drogos, L.L.; Hill, S.K.; Ruocco, A.C.; Keedy, S.K.; Reilly, J.L.; Keshavan, M.S.; et al. Reduced levels of vasopressin and reduced behavioral modulation of oxytocin in psychotic disorders. Schizophr. Bull. 2014, 40, 1374–1384. [Google Scholar] [CrossRef] [Green Version]
  311. Surget, A.; Belzung, C. Involvement of vasopressin in affective disorders. Eur. J. Pharmacol. 2008, 583, 340–349. [Google Scholar] [CrossRef] [PubMed]
  312. Van Londen, L.; Goekoop, J.G.; van Kempen, G.M.; Frankhuijzen-Sierevogel, A.C.; Wiegant, V.M.; van der Velde, E.A.; De Wied, D. Plasma levels of arginine vasopressin elevated in patients with major depression. Neuropsychopharmacology 1997, 17, 284–292. [Google Scholar] [CrossRef] [Green Version]
  313. Rutigliano, G.; Rocchetti, M.; Paloyelis, Y.; Gilleen, J.; Sardella, A.; Cappucciati, M.; Palombini, E.; Dell’Osso, L.; Caverzasi, E.; Politi, P.; et al. Peripheral oxytocin and vasopressin: Biomarkers of psychiatric disorders? A comprehensive systematic review and preliminary meta-analysis. Psychiatry Res. 2016, 241, 207–220. [Google Scholar] [CrossRef] [PubMed]
  314. Barhale, C.; Raju, M.S.V.K.; Pawar, A.V.; Kadam, K.; De Sousa, A.; Andrade, C. Serum Oxytocin Concentration in Patients Receiving Electroconvulsive Therapy: An Exploratory Study and Review of Literature. J. ECT 2017, 33, 122–125. [Google Scholar] [CrossRef] [PubMed]
  315. Demitrack, M.A.; Gold, P.W. Oxytocin: Neurobiologic considerations and their implications for affective illness. Prog. Neuropsychopharmacol. Biol. Psychiatry 1988, 12 (Suppl. 1), S23–S51. [Google Scholar] [CrossRef]
  316. Sasayama, D.; Hattori, K.; Teraishi, T.; Hori, H.; Ota, M.; Yoshida, S.; Arima, K.; Higuchi, T.; Amano, N.; Kunugi, H. Negative correlation between cerebrospinal fluid oxytocin levels and negative symptoms of male patients with schizophrenia. Schizophr. Res. 2012, 139, 201–206. [Google Scholar] [CrossRef] [Green Version]
  317. Pitts, A.F.; Samuelson, S.D.; Meller, W.H.; Bissette, G.; Nemeroff, C.B.; Kathol, R.G. Cerebrospinal fluid corticotropin-releasing hormone, vasopressin, and oxytocin concentrations in treated patients with major depression and controls. Biol. Psychiatry 1995, 38, 330–335. [Google Scholar] [CrossRef]
  318. Lee, M.R.; Sheskier, M.B.; Farokhnia, M.; Feng, N.; Marenco, S.; Lipska, B.K.; Leggio, L. Oxytocin receptor mRNA expression in dorsolateral prefrontal cortex in major psychiatric disorders: A human post-mortem study. Psychoneuroendocrinology 2018, 96, 143–147. [Google Scholar] [CrossRef]
  319. Landgraf, R.; Neumann, I.D. Vasopressin and oxytocin release within the brain: A dynamic concept of multiple and variable modes of neuropeptide communication. Front. Neuroendocrinol. 2004, 25, 150–176. [Google Scholar] [CrossRef]
  320. Heinrichs, M.; Meinlschmidt, G.; Neumann, I.; Wagner, S.; Kirschbaum, C.; Ehlert, U.; Hellhammer, D.H. Effects of suckling on hypothalamic-pituitary-adrenal axis responses to psychosocial stress in postpartum lactating women. J. Clin. Endocrinol. Metab. 2001, 86, 4798–4804. [Google Scholar] [CrossRef]
  321. Skrundz, M.; Bolten, M.; Nast, I.; Hellhammer, D.H.; Meinlschmidt, G. Plasma oxytocin concentration during pregnancy is associated with development of postpartum depression. Neuropsychopharmacology 2011, 36, 1886–1893. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  322. De Cagna, F.; Fusar-Poli, L.; Damiani, S.; Rocchetti, M.; Giovanna, G.; Mori, A.; Politi, P.; Brondino, N. The Role of Intranasal Oxytocin in Anxiety and Depressive Disorders: A Systematic Review of Randomized Controlled Trials. Clin. Psychopharmacol. Neurosci. 2019, 17, 1–11. [Google Scholar] [CrossRef] [PubMed]
  323. Engel, S.; Laufer, S.; Knaevelsrud, C.; Schumacher, S. The endogenous oxytocin system in depressive disorders: A systematic review and meta-analysis. Psychoneuroendocrinology 2019, 101, 138–149. [Google Scholar] [CrossRef] [PubMed]
  324. Kim, S.; Soeken, T.A.; Cromer, S.J.; Martinez, S.R.; Hardy, L.R.; Strathearn, L. Oxytocin and postpartum depression: Delivering on what’s known and what’s not. Brain Res. 2014, 1580, 219–232. [Google Scholar] [CrossRef] [Green Version]
  325. Moura, D.; Canavarro, M.C.; Figueiredo-Braga, M. Oxytocin and depression in the perinatal period-a systematic review. Arch. Women’s Ment. Health 2016, 19, 561–570. [Google Scholar] [CrossRef]
  326. Scantamburlo, G.; Hansenne, M.; Geenen, V.; Legros, J.J.; Ansseau, M. Additional intranasal oxytocin to escitalopram improves depressive symptoms in resistant depression: An open trial. Eur. Psychiatry 2015, 30, 65–68. [Google Scholar] [CrossRef]
  327. Pincus, D.; Kose, S.; Arana, A.; Johnson, K.; Morgan, P.S.; Borckardt, J.; Herbsman, T.; Hardaway, F.; George, M.S.; Panksepp, J.; et al. Inverse effects of oxytocin on attributing mental activity to others in depressed and healthy subjects: A double-blind placebo controlled FMRI study. Front. Psychiatry 2010, 1, 134. [Google Scholar] [CrossRef] [Green Version]
  328. Pitman, R.K.; Orr, S.P.; Lasko, N.B. Effects of intranasal vasopressin and oxytocin on physiologic responding during personal combat imagery in Vietnam veterans with posttraumatic stress disorder. Psychiatry Res. 1993, 48, 107–117. [Google Scholar] [CrossRef]
  329. Lardenoije, R.; Roubroeks, J.A.Y.; Pishva, E.; Leber, M.; Wagner, H.; Iatrou, A.; Smith, A.R.; Smith, R.G.; Eijssen, L.M.T.; Kleineidam, L.; et al. Alzheimer’s disease-associated (hydroxy)methylomic changes in the brain and blood. Clin. Epigenetics 2019, 11, 164. [Google Scholar] [CrossRef]
  330. Green, J.J.; Hollander, E. Autism and oxytocin: New developments in translational approaches to therapeutics. Neurotherapeutics 2010, 7, 250–257. [Google Scholar] [CrossRef] [Green Version]
  331. Zhang, H.F.; Dai, Y.C.; Wu, J.; Jia, N.X.; Zhang, J.S.; Shou, X.J.; Han, S.P.; Zhang, R.; Han, J.S. Plasma oxytocin and arginine-vasopressin levels in childre with autism spectrum disorders in China: Association with symptoms. Neurosci. Bull. 2016, 32, 423–432. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  332. Hendaus, M.; Jomha, F.A.; Alhammadi, A.H. Vasopressin in the Amelioration of Social Functioning in Autism Spectrum Disorder. J. Clin. Med. 2019, 8, 1061. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  333. Yatawara, C.J.; Einfeld, S.L.; Hickie, I.B.; Davenport, T.A.; Guastella, A.J. The effect of oxytocin nasal spray on social interaction deficits observed in young children with autism: A randomized clinical crossover trial. Mol. Psychiatry 2016, 21, 1225–1231. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  334. Rich, M.E.; Caldwell, H.K. A Role for Oxytocin in the Etiology and Treatment of Schizophrenia. Front. Endocrinol. 2015, 6, 90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. The brain structures involved in the regulation of the cardiovascular, cognitive, emotional, and behavioral functions through actions exerted by the renin–angiotensin system (RAS), and the vasopressin (AVP) and oxytocin (OT) systems. Abbreviations: Ang II—angiotensin II; Ang-(1-7)—angiotensin-(1-7); AP—area postrema; CVLM—caudal ventrolateral medulla, CVOs—circumventricular organs; IML—intermediolateral column; NTS—nucleus of the solitary tract; OVLT—organum vasculosum laminae terminalis; PAG—periaqueductal gray; PFC—prefrontal cortex; PVN—paraventricular nucleus; RVLM—rostral ventrolateral medulla; SFO—subfornical organ; 3rdV—third ventricle.
Figure 1. The brain structures involved in the regulation of the cardiovascular, cognitive, emotional, and behavioral functions through actions exerted by the renin–angiotensin system (RAS), and the vasopressin (AVP) and oxytocin (OT) systems. Abbreviations: Ang II—angiotensin II; Ang-(1-7)—angiotensin-(1-7); AP—area postrema; CVLM—caudal ventrolateral medulla, CVOs—circumventricular organs; IML—intermediolateral column; NTS—nucleus of the solitary tract; OVLT—organum vasculosum laminae terminalis; PAG—periaqueductal gray; PFC—prefrontal cortex; PVN—paraventricular nucleus; RVLM—rostral ventrolateral medulla; SFO—subfornical organ; 3rdV—third ventricle.
Jcm 11 00908 g001
Figure 2. The main components of the renin–angiotensin system (RAS) engaged in the regulation of cardiovascular, cognitive, emotional, and behavioral functions. Abbreviations: ACE—angiotensin converting enzyme; ACEI—inhibitor of ACE; Ang—angiotensin; APA, APB, and APN—aminopeptidases A, B, and N; AT1R, AT2R—angiotensin receptors; AVP—arginine vasopressin; CNS—central nervous system; IRAP—insulin-regulated aminopeptidase; MasR—Mas receptor of Ang-(1-7); NEP—neutral endopeptidase; OT—oxytocin; OTR—oxytocin receptor; V1aR, V1bR, V2R—vasopressin receptors. See also refrences [16,17].
Figure 2. The main components of the renin–angiotensin system (RAS) engaged in the regulation of cardiovascular, cognitive, emotional, and behavioral functions. Abbreviations: ACE—angiotensin converting enzyme; ACEI—inhibitor of ACE; Ang—angiotensin; APA, APB, and APN—aminopeptidases A, B, and N; AT1R, AT2R—angiotensin receptors; AVP—arginine vasopressin; CNS—central nervous system; IRAP—insulin-regulated aminopeptidase; MasR—Mas receptor of Ang-(1-7); NEP—neutral endopeptidase; OT—oxytocin; OTR—oxytocin receptor; V1aR, V1bR, V2R—vasopressin receptors. See also refrences [16,17].
Jcm 11 00908 g002
Table 1. Summary of the inappropriate functional actions of renin–angiotensin, vasopressin, and oxytocin systems in selected neuropsychiatric and neurodegenerative disorders.
Table 1. Summary of the inappropriate functional actions of renin–angiotensin, vasopressin, and oxytocin systems in selected neuropsychiatric and neurodegenerative disorders.
Neuropsychiatric/Neurodegenrative DisorderFunctional ActionReferences
Renin–angiotensin system (RAS)
Cognitive disordersHuman and rodent studies:
  • Ang II may impair cognitive processes, probably via AT1R;
[98,99,100,101,102]
Rodent studies:
  • Ang IV and Ang-(1-7) may improve learning and memory;
[103,104,105]
Alzheimer’s diseaseHuman studies:
  • The enhanced activation of the RAS may inhibit acetylcholine release in the cortex and contribute to the development of AD dementia;
[106,107,108]
Human and rodent studies:
  • The excessive activation of the brain AT1R and insufficient activation of AT2R may induce excessive generation of ROS, and this may account for the prevalence of neurodegenerative processes over neuroprotective processes in the brains of AD patients;
[109,110,111,112,113]
Rodent studies:
  • The inappropriate activation of the Ang-(1-7)/Mas axis may play a role in the pathogenesis of AD;
[114]
Stress and painHuman and rodent studies:
  • Stress provoked by tissue injury, ischemia, hypoxia, inflammation, stroke, or myocardial infarction, as well as chronic mild stress, activates the RAS and increases expression of AT1R in the brain, heart, and kidney;
[16,23,25,66,67,85,87,115]
Rodent studies:
  • Ang II enhances the pressor response to stress by AT1R, while the tachycardic response to stress is enhanced by AT2R;
[116]
  • The stimulation of AT1R and AT2R, and the activation of the Ang-(1-7) MasR pathway in the brain reduces pain;
[117,118]
Affective disorders Human studies:
  • A significant association between depression and the AT1R A1166C CC genotype;
[119]
  • Ang (1-7) has an antidepressant effect;
[120]
SchizophreniaHuman studies:
  • In patients with schizophrenia, missense mutations of angiotensinogen (AGTM268T, AGT235T) with replacement of valine by threonine are associated with the decline of cognitive functions and lower verbal memory scores;
[121,122]
  • An association between ACE I/D (insertion/deletion) polymorphism and a disposition to schizophrenia was found;
[123]
  • The AT1R antagonist telmisartan can alleviate the symptoms of schizophrenia;
[124]
Parkinson’s diseaseRodent studies:
  • Increased expression of AT1R and NADPH oxidase activation;
[51]
  • Endogenous Ang II potentiates the neurotoxic effect of MPTP on dopaminergic neurons, whereas ACE or AT1Rs antagonists exert their beneficial effects through the inhibition of microglial NADPH activation and the suppression of prooxidative and proinflammatory effects mediated by cytokines;
[125,126,127]
  • Chronic treatment with AT1R antagonists is associated with the formation of heterodimers of AT1R/AT2R;
[37,125]
Tardive dyskinesiaRodent studies:
  • The administration of candesartan and lisinopril reduces the release of proinflammatory cytokines (IL-1β, TNF-α) and glutamate in the rat model of haloperidol-induced tardive dyskinesia;
[128]
Psychiatric symptoms in COVID 19Human studies:
  • The inappropriate function of the RAS may contribute to the exaggeration of psychiatric symptoms in patients with COVID-19
[129]
  • The excessive stimulation of AT1R influences microglial polarization and induces an active M2a proinflammatory state and may thereby initiate neurodegenerative processes;
Vasopressin system (VPS)
Affective disordersHuman studies:
  • The increased expression of AVP mRNA in the PVN/SON in brains of patients with MDD;
[130]
  • The association of V1bR gene polymorphism (haplotype associated with A-T-C-A-G for the single nucleotide polymorphism (SNP) s1-s2-s3-s4-s5 allele) with a protective effect for recurrent MDD;
[131]
  • The association of the V1bR SNPs (rs28676508, rs35369693) with child aggression;
[132]
  • The linkage of the V1bR genetic variation SNP rs33990840 with suicidal behavior;
[133]
  • Elevated copeptin (surrogate marker of AVP) in patients resistant to antidepressant pharmacotherapy;
[134,135,136]
  • V1bR antagonists are currently being trialed for the treatment of MDD;
[134,135]
Rodent studies:
  • In rodent models of anxiety and depression, the antagonists of V1bR show anxiolytic- and antidepressant-like effects;
[137,138,139]
  • The blockade of the central V1 receptors abolished anhedonia induced by chronic mild stress;
[140]
  • The activation of brain VPS in stress;
[26,140,141,142,143,144,145]
  • The blockade of V1bR induces anxiolytic actions in various models of depression;
[146,147]
Alzheimer’s diseaseHuman studies:
  • Low concentration of AVP in the CSF;
[148]
  • The reduced expression of AVP immunoreactivity in the hippocampus, nucleus accumbens, and the internal portion of the globus pallidus of AD patients in comparison with controls (post-mortem studies);
[149]
  • A reduced number of AVP expressing cells in the suprachiasmatic nucleus in senescence and AD patients;
[150]
  • Vasopressinergic innervation of the PVN, SON, and locus coeruleus in AD patients and non-demented controls do not differ;
[151,152]
Rodent studies:
  • The improvement of working memory and long-term memory in APP/PS1 mouse model of AD after the intranasal application of AVP-(4-8);
[153]
  • The improvement of social memory is enhanced by the stimulation of V1bR in the hippocampus in mice;
[154]
SchizophreniaHuman studies:
  • Lower AVP levels in the temporal cortex of schizophrenic patients (post-mortem studies);
[155]
  • Reduced AVP mRNA in the PVN of schizophrenic patients (post-mortem studies);
[156]
  • In patients with schizophrenia, blood AVP levels are either elevated or not altered;
[157,158,159]
  • A positive correlation between blood AVP level and severity of symptoms is found in female but not in male schizophrenia patients.
[159]
  • Increased blood AVP levels, polydipsia, hypoosmolality, and hyponatremia are found in some patients with schizophrenia
[160,161,162]
  • The intranasal application of DDAVP (synthetic analog of AVP) increases the effectiveness of risperidone in reducing the negative symptoms of schizophrenia;
[163]
  • The associations between SNPs of the AVP gene and schizophrenia (chromosomal region 20p13, loci rs2740204 and rs3011589);
[164]
Rodent studies:
  • Schizophrenia-like symptoms with impairment of social behavior in AVP-deficient (di/di) Brattleboro rats and V1aR knockout mice;
[165,166,167]
  • The lower expression of AVP receptors in the prefrontal cortex and hypothalamus in the MAM model of schizophrenia in rats;
[168]
Autism spectrum disorderHuman studies:
  • Lower AVP concentrations in CSF of children with autism, and AVP levels were associated with the severity of symptoms;
[169,170,171]
  • AVP concentration in the CSF in neonates predicts a subsequent diagnosis of autism;
[170]
  • A significant association between ASD with polymorphism of the V1aR and V1bR genes and autism (SNP rs35369693 and rs28632197);
[172,173,174,175]
  • Intranasally applied AVP improves social abilities and reduces anxiety symptoms in children with ASD;
[176,177]
Oxytocin system (OTS)
Alzheimer’s diseaseHuman studies:
  • In AD patients, intranasally applied OT does not influence the activity of the brain regions affected by AD;
[151,178,179,180,181,182]
  • In AD patients, magnetic resonance images show that the plasma OT concentration correlates with the right parahippocampal gyrus volume;
[183]
Affective disorders Human studies:
  • The increased activity of the central OTS in depressive mood disorders;
[130,184,185,186]
  • Inconsistent data on the correlation between plasma OT levels and depression;
[184,187,188,189,190,191,192]
  • Plasma OT levels positively correlate with help-seeking intentions, behavior, and estimation of happiness in patients with depression or anxiety;
[184,193,194]
  • The positive associations between depression, MDD, and separation anxiety and single nucleotide polymorphism (rs53576; rs2254298; rs53576 genotype A allele) of the OTR gene and with G-protein genes (Gβ3 rs5443);
[195,196,197,198,199,200]
  • Low plasma OT levels in the third trimester of pregnancy may predict postpartum depressive symptoms;
[201]
  • The level of blood oxytocin is lower in mothers with post-partum depression than in nondepressed mothers;
[202]
Rodent studies:
  • Endogenous OTS decreases anxiety behavior in pregnant and lactating rats;
[201]
Schizophrenia
  • Human studies:
  • The association of schizophrenia with single nucleotide polymorphisms of the OT gene in chromosomal region 20p13 (rs4813626);
[164]
  • Lower plasma OT concentrations in patients with schizophrenia;
[158,203]
  • The negative correlation between OT levels and the severity of symptoms in patients with schizophrenia;
[157]
  • The positive effects of the intranasally applied OT on social cognition in patients with schizophrenia;
[204,205]
  • Clinical trials and meta-analyses do not support the significant therapeutic effect of OT in schizophrenia;
[206,207,208,209]
Rodent studies:
  • Reduced concentrations of OT and OTRs in the prefrontal cortex and in the hypothalamus of rats in the experimental MAM model of schizophrenia;
[168]
Autism spectrum disorderHuman studies:
  • A strong association between OTR gene polymorphism (rs2254298, rs2268491, rs53576, rs237887, rs2268493, rs1042778 and rs7632287) and susceptibility to ASD;
[210,211,212,213,214,215,216,217]
  • Lower plasma oxytocin levels in children with ASD;
[218,219,220,221]
  • Intravenously infused or intranasally applied OT ameliorates repetitive behavior in adults with ASD and Asperger’s disorder;
[222,223]
  • Randomized crossover trials show that in children with ASD, treatment with intranasal OT improves caregiver-rated social responsiveness and enhances learning in response to social targets and feedback;
[176,224]
  • The lack of beneficial effects of OT in ASD;
[225,226,227]
  • A systematic review and meta-analysis of tolerance of long-term intranasal application of OT in ASD.
[228]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Szczepanska-Sadowska, E.; Wsol, A.; Cudnoch-Jedrzejewska, A.; Czarzasta, K.; Żera, T. Multiple Aspects of Inappropriate Action of Renin–Angiotensin, Vasopressin, and Oxytocin Systems in Neuropsychiatric and Neurodegenerative Diseases. J. Clin. Med. 2022, 11, 908. https://0-doi-org.brum.beds.ac.uk/10.3390/jcm11040908

AMA Style

Szczepanska-Sadowska E, Wsol A, Cudnoch-Jedrzejewska A, Czarzasta K, Żera T. Multiple Aspects of Inappropriate Action of Renin–Angiotensin, Vasopressin, and Oxytocin Systems in Neuropsychiatric and Neurodegenerative Diseases. Journal of Clinical Medicine. 2022; 11(4):908. https://0-doi-org.brum.beds.ac.uk/10.3390/jcm11040908

Chicago/Turabian Style

Szczepanska-Sadowska, Ewa, Agnieszka Wsol, Agnieszka Cudnoch-Jedrzejewska, Katarzyna Czarzasta, and Tymoteusz Żera. 2022. "Multiple Aspects of Inappropriate Action of Renin–Angiotensin, Vasopressin, and Oxytocin Systems in Neuropsychiatric and Neurodegenerative Diseases" Journal of Clinical Medicine 11, no. 4: 908. https://0-doi-org.brum.beds.ac.uk/10.3390/jcm11040908

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

Article Metrics

Back to TopTop