Next Article in Journal
Metabolites Potentially Determine the High Antioxidant Properties of Limosilactobacillus fermentum U-21
Previous Article in Journal
Recent Genome-Editing Approaches toward Post-Implanted Fetuses in Mice
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Recent Developments in Protein Lactylation in PTSD and CVD: Novel Strategies and Targets

1
International Agency for Research on Cancer, World Health Organization (IARC/WHO), 69372 Lyon, France
2
Department of Psychiatry, Loma Linda University, Loma Linda, CA 92350, USA
3
Patton State Hospital, University of California, Riverside, CA 92521, USA
4
Napa State Hospital, Napa, CA 94558, USA
5
Department of Psychiatry, University of California, Riverside, CA 92521, USA
*
Author to whom correspondence should be addressed.
BioTech 2023, 12(2), 38; https://doi.org/10.3390/biotech12020038
Submission received: 18 February 2023 / Revised: 27 April 2023 / Accepted: 1 May 2023 / Published: 15 May 2023
(This article belongs to the Section Medical Biotechnology)

Abstract

:
In 1938, Corneille Heymans received the Nobel Prize in physiology for discovering that oxygen sensing in the aortic arch and carotid sinus was mediated by the nervous system. The genetics of this process remained unclear until 1991 when Gregg Semenza while studying erythropoietin, came upon hypoxia-inducible factor 1, for which he obtained the Nobel Prize in 2019. The same year, Yingming Zhao found protein lactylation, a posttranslational modification that can alter the function of hypoxia-inducible factor 1, the master regulator of cellular senescence, a pathology implicated in both post-traumatic stress disorder (PTSD) and cardiovascular disease (CVD). The genetic correlation between PTSD and CVD has been demonstrated by many studies, of which the most recent one utilizes large-scale genetics to estimate the risk factors for these conditions. This study focuses on the role of hypertension and dysfunctional interleukin 7 in PTSD and CVD, the former caused by stress-induced sympathetic arousal and elevated angiotensin II, while the latter links stress to premature endothelial cell senescence and early vascular aging. This review summarizes the recent developments and highlights several novel PTSD and CVD pharmacological targets. They include lactylation of histone and non-histone proteins, along with the related biomolecular actors such as hypoxia-inducible factor 1α, erythropoietin, acid-sensing ion channels, basigin, and Interleukin 7, as well as strategies to delay premature cellular senescence by telomere lengthening and resetting the epigenetic clock.
Key Contribution: Although identified as a pathology during WWI when PTSD was known as “shell shock”, today, there is a paucity of treatments for this condition. The cardiovascular involvement in PTSD was hinted at by the term “soldier's heart” used even earlier, during the American Civil War, to describe feelings of anxiety, tension, and being “on edge”. Although treated primarily by psychiatrists, PTSD is a systemic disease which has been associated with numerous medical problems, including chronic pain, hypertension, fatigue, muscle tension, headaches as well as metabolic syndrome and immune dysfunction. As systemic disorders call for systemic interventions, here we discuss epigenetic therapies to lower lactylation of histone proteins as new therapeutic strategies for PTSD and CVD.

1. Introduction

In response to stressors, cells undergo premature senescence, a state of proliferation arrest, glycolysis-fueled metabolism, and a detrimental secretome, known as senescence-associated secretory phenotype (SASP), which drives organismal aging by disseminating senescence to the neighboring healthy cells [1]. SASP has been implicated in cellular deregulation as it is the precursor of many pathologies, including cancer, while at the same time providing a fertile scientific ground for the discovery of potential new treatments [2]. For example, SASP has been associated with shortened telomere length (STL) and impaired DNA methylation, the epigenetic clock (EpiClock), hallmarks of posttraumatic stress disorder (PTSD), and cardiovascular disease (CVD) [3,4]. These features likely account for the shorter lifespan and early development of age-related diseases demonstrated in many patients with PTSD and CVD [5,6,7].
Hypoxia is another important cellular stressor as dysfunctional oxygenation and hypoxia-inducible factor 1 alpha (HIF-1α) lead to premature aging, a characteristic of PTSD and CVD [5,8,9,10]. Indeed, both hypoxia and hyperoxygenation have been associated with neuropsychiatric pathology, and several studies have shown that hyperbaric oxygen or treatment with erythropoietin (EPO) can be efficacious in several psychiatric disorders, including PTSD and traumatic brain disorder (TBI) [11,12,13,14,15,16] (NCT00525863).
At the subcellular level, hypoxia induces endoplasmic reticulum (ER) stress, a common PTSD and CVD pathology, suggesting that psychological and biological stressors can contribute to molecular stress [17,18,19,20,21]. In this regard, it is tempting to speculate that ER stress and the unfolded protein response (UPR) may mirror the organismal stress. Moreover, hypoxia upregulates oxytocin (OXT), while dysfunctional HIF-1α lowers the levels of this protective hormone, predisposing to PTSD and CVD [22,23].
A recent study has identified hypertension as the common genetic risk factor for PTSD and CVD, indicating that sympathetic arousal and elevated angiotensin II (ANG II) levels contribute to these disorders [3,24]. Moreover, the connection between high blood pressure and hypoxia has been well documented in pulmonary arterial hypertension (PAH), a condition marked by dysfunctional HIF-1α and premature endothelial aging [25,26,27]. Interestingly, about half of PAH patients also experience depression, anxiety, or panic disorder, indicating that this disease may be utilized as a working model of PTSD/CVD [28,29].
Under normal circumstances, endothelial cells (ECs) utilize aerobic glycolysis or the Warburg effect for their bioenergetic need, deriving over 80% of adenosine triphosphate (ATP) from lactate despite oxygen availability [30,31,32]. As senescent ECs upregulate lactate even more, excessive glycolysis likely promotes protein lactylation, predisposing to pathology, including PTSD and CVD [33,34,35]. For example, lactylation of HIF-1 α may disable this protein, causing premature EC senescence, which in turn increases vascular permeability, contributing to the pathogenesis of several cardiovascular and neuropsychiatric illnesses [36,37,38].
Dysfunctional interleukin 7 (IL-7), a cytokine known for shifting cellular metabolism from oxidative phosphorylation (OXPHOS) to aerobic glycolysis or Warburg effect, has been associated with PAH, indicating that upregulated lactate serves as a lactylation substrate [39,40]. Lactylation is a posttranslational modification (PTM) of histone and non-histone proteins which alters the physiological function of many biomolecules, including HIF-1α [38].
The association of IL-7 with lactylation is significant as, under normal circumstances, this cytokine opposes cellular senescence and lowers blood pressure [41], suggesting that its dysfunction may drive PTSD and CVD, validating the study of Seligovski et al. [24,39,42,43,44]. In this regard, impaired IL-7 signaling with IL-7 receptors (IL-7R) on ECs, likely contributes to PTSD and CVD by promoting early vascular aging (EVA) and premature ECs senescence [5,34,43,44,45,46,47,48,49]. In contrast, upregulated HIF-1α exerts cardioprotective and neuroprotective effects and can alleviate many PTSD symptoms [50,51,52]. For example, preclinical studies have shown that intermittent hypoxia may ameliorate both PTSD and CVD, further connecting these conditions to oxygen disturbances [53,54].
Several studies have reported that HIF-1α upregulates erythropoietin (EPO), a hormone with demonstrated antidepressant and anxiolytic properties, highlighting the cardio and neuroprotective effect of this protein [55,56,57,58]. Indeed, as EPO counteracts hypoxia by increasing the systemic oxygen-carrying capacity, it protects neuronal cells and cardiomyocytes from ischemia–reperfusion injury [50,51,52,53,54,55,56,57,58,59,60]. Conversely, impaired EPO signaling, documented in PTSD and CVD, points to this pathway as a druggable target for both conditions [55,61]. Under normal circumstances, IL-7 upregulates EPO, reducing the adverse effects of psychological stress, and further emphasizing the beneficial role of these molecules in PTSD and CVD [55,62]. Moreover, depleted IL-7 has been associated with the atrophy of the thymus gland, a characteristic of PTSD and CVD, suggesting that supplementation with IL-7 could ameliorate these pathologies [63,64,65,66]. Furthermore, photobiomodulation (PBM), a non-thermal red-light therapy, has been shown to reverse thymic involution, indicating potential benefits in PTSD and CVD [67].
Although poorly defined, the existence of an intra-thymic OXT system has been known for at least four decades, linking this gland to OXT depletion [68,69,70]. Indeed, intranasal OXT administration was found beneficial for neuropsychiatric pathologies, including PTSD and psychotic polydipsia, linking these conditions to IL-7 [71,72,73,74,75]. As IL-7 has also been implicated in neuromyelitis optica (NMO), a condition marked by antibodies against aquaporin 4 (AQP-4) water channels, IL-7 may protect not only from PTSD and CVD but also from NMO, and the related condition, multiple sclerosis (MS) [76,77,78,79].
This review summarizes the recent developments and highlights novel pharmacological targets for PTSD and CVD, including lactylation of histone and non-histone proteins, along with the related biomolecular actors, HIF-1α, EPO, ASC1 channels, CD147, and IL-7 (Figure 1). Potential interventions and new strategies are also discussed.

2. Human Stress Response: A Quick Reminder

Biological and psychological stressors of sufficient intensity may activate organismal responses which aim at restoring homeostasis by inducing physiological or behavioral changes [80]. For example, stressors, such as medical illnesses or social rejection, activate the sympathetic-adreno-medullar (SAM) system, inducing immediate adaptation as well as delayed hypothalamus-pituitary-adrenal (HPA) responses [81,82]. In the central nervous system (CNS), biological and psychological stressors are processed by different pathways, which are believed to converge at the level of the amygdala [83,84].
Amygdala, a sensor for both biological and psychological stressors, functions to activate the HPA axis and restore homeostasis [85]. Amygdala plays a major role in CVD, and the activity of this nucleus is directly correlated with the development of heart disease [86]. The basolateral nucleus of the amygdala (BLA) processes primarily psychological stressors, including aversive stimuli, while the central nucleus of the amygdala (CeA) responds to biological input, such as blood pressure or redox imbalance [87,88,89,90]. However, hypertension due to ANG II-mediated fear-behavior is also processed by the CeA, blurring the boundaries between the stressor types [91,92].
OXT is an established inhibitor of stress-related amygdalar activation, thus a negative regulator of PTSD and CVD, while ANG II has the opposite effect [86,93,94]. Human ECs express abundant OXT and AVP receptors which were implicated in vasodilation, thus benefiting patients with hypertension, a major risk factor for both PTSD and CVD [95].
OXT is produced primarily in the magnocellular neurosecretory cells of the paraventricular and supraoptic nuclei of the hypothalamus and is transported to the posterior pituitary, from where it migrates into the systemic circulation [96]. Smaller sources of OXT, such as the thymus and bone marrow, modulate immune responses, probably mediating the psychosomatic symptoms, such as the sickness behavior [69]. The magnocellular neurosecretory cells project to the amygdala, striatum, hypothalamus, hippocampus, and nucleus accumbens, areas previously implicated in PTSD and other neuropsychiatric illnesses [97,98,99,100,101]. The lateral nucleus of CeA expresses OXT receptors (OXTRs) which are activated by the inwardly rectifying K+ channels (Kir4.1), proteins involved in amygdalar inhibition [102]. Kir4.1 are co-localized with AQP4 water channels which play a major role in cardiovascular homeostasis and synaptic plasticity, probably contributing to PTSD-associated traumatic hypermnesia [103,104,105,106].

3. Protein Lactylation, the Warburg Effect 2.0

In the 1920s, Otto Warburg studied the metabolism of cancer cells and discovered that even in the presence of oxygen, malignancies satisfy their energetic requirement by fermenting glucose to lactate instead of carbon dioxide [107]. Lactate was believed to be a metabolic waste product; however, several studies have shown that it can function as a signaling molecule, participating in numerous physiological processes, ranging from immunity to inflammation and gene expression [108]. Lactate-induced epigenetic changes can promote or inhibit protein expression, potentially triggering pathology [109]. For example, the high mobility group box 1 (HMGB1) and HIF1α are lactylation substrates that have previously been associated with tumorigenesis, linking lactylation to malignant transformation [38,109,110]. The metabolic shift from OXPHOS to glycolysis, observed in both PTSD and CVD, may account for lactate-induced anxiety, a phenomenon known since the 1980s but poorly defined previously [111,112,113,114]. Indeed, the molecular underpinnings of PTSD activation by lactate infusion remained unclear until the discovery of acid-sensing ion channels (ASICs) and protein lactylation [109,112,115]. Upregulated lactate and HIF1α lactylation have been shown to promote premature EC senescence, a pathological driver of both CVD and PTSD [116,117]. Under physiological circumstances, HIF1α is a negative regulator of both EC senescence and lactylation; however, loss of this molecule may contribute to EVA and CVD [27,118]. Furthermore, preclinical studies have implicated lactylation in neuropsychiatric pathology, including anxiety, stress, fear, avoidant behavior, and social defeat, emphasizing a probable link to PTSD [35,116,119,120]. These findings are summarized in Figure 2.

3.1. Virus-Induced PTSD and CVD

Several pathogens, including SARS-CoV-2, have been associated with a high prevalence of PTSD and CVD, suggesting that a better understanding of virus-induced PTSD (viPTSD) may shed light on the pathogenesis of both conditions [121,122]. The COVID-19 pandemic has highlighted a novel phenomenon, viPTSD, an entity that can be more prevalent than the trauma-related syndrome encountered in special populations, such as combat veterans. For example, new epidemiological data show a PTSD prevalence of 30.2% in COVID-19 survivors, 39.5% in Ebola, and 34% in HIV, numbers that exceed the 8% found in the general population and 16% in war veterans [123,124,125].
We surmise that excessive lactylation may account for the high prevalence of viPTSD as pathogens often establish hospitable microenvironments in host cells by usurping HIF-1α and the Warburg effect [10,27,126]. Elevated lactate levels can also activate ASIC1 channels, triggering PTSD symptoms by a different mechanism [127]. In contrast, IL-7/IL-7R signaling opposes senescence by upregulating HIF-1α and activating the thymus [128].

3.2. Virus-Induced Senescence (VIS)

Several viruses, including SARS-CoV-2, have been known to promote premature cellular aging, known as virus-induced senescence (VIS), a phenotype indistinguishable from other forms of senescence [129,130]. Cellular senescence is an antitumor program marked by replication arrest, an active glycolysis-mediated metabolism, and a toxic SASP secretome.
In our previous work, we hypothesized that COVID-19 triggers EC senescence and EVA by blocking angiotensin-converting enzyme-2 (ACE-2), leading to ANG II accumulation [131]. This hypothesis was validated in vitro a few months later [132] and, at present, we hypothesize further that ANG II upregulation is also caused by deficient endoplasmic reticulum aminopeptidase 1 and 2 (ERAP1 and ERAP2), which break down ANG II into ANG III and ANG IV (Figure 3). In addition, others have shown that ANG II disrupts IL-7Rs, enabling viral thriving by inducing immune senescence [133].
ECs are major producers of brain-derived neurotrophic factor (BDNF), a nerotrophin depleted in PTSD, suggesting that senescent endothelia may predispose to this syndrome by depleting BDNF [134]. Furthermore, senescent ECs disrupt the blood–brain barrier (BBB), allowing molecules from the systemic circulation, such as ANG II and lactate, to activate ASIC1, inducing fear-related behaviors [131,135]. ASICs are lactate-activated channels that match the CNS work with energy requirements; however, excessive glycolysis and ASIC activation may trigger anxiety and PTSD [52,115,136,137].
Taken together, viPTSD operates by promoting premature vascular aging, which increases endothelial permeability, allowing circulatory molecules and toxins to access the brain, activating the stress-processing areas, and triggering PTSD.

4. From Cellular to Organismal Stress

At the subcellular level, psychological and biological stressors likely merge on the endoplasmic reticulum (ER), inducing molecular stress by activating the unfolded protein response (UPR) [138,139]. For example, ER stress has been associated with major depressive disorder (MDD), PTSD, neurodegeneration, and CVD, bridging not only the gap between these pathologies but also the molecular and organismal stress [18,140,141]. For example, dysfunctional ANG II signaling with angiotensin 2 type 1 receptors (AT1Rs), documented in hypertension, induces ER stress, leading to premature EC senescence and EVA [140,142,143,144,145].
Under normal circumstances, unfolded or misfolded proteins are transported from the ER into the cytosol to be degraded by the proteasome. Accumulation of misfolded proteins and UPR activation in response to biological or psychological stressors may link molecular and organismal aging, while on the other hand, identifying these pathways as pharmacological targets [146]. Indeed, targeting the UPR comprises a novel strategy in the treatment of cardiovascular and neuropsychiatric illnesses, including PTSD [147,148,149,150,151] (see the section on potential interventions). Interestingly, ER stress and UPR activation upregulate IL-7, further connecting cellular stress with PTSD and CVD [152]. Table 1 provides a summary view of those observations.

5. The Mystery of ERAP2

The ER harbors ERAP1 and ERAP2, zinc-metalloproteases, which have been implicated in CVD and PTSD (via OXT depletion) [133,163,164]. Indeed, ERAP2 functions as an oxytocinase, breaking down OXN and terminating its action, suggesting that a dysfunction of this enzyme may predispose it to PTSD [133]. OXT is a prosocial hormone with cardio and neuroprotective properties, which under pathological circumstances, was associated with fibromyalgia, PTSD, and CVD [97,165,166,167]. Several studies have reported that ERAP1 and ERAP2 hydrolyze ANG II into ANG III and ANG IV, contributing to the rapid clearance of this toxic peptide (Figure 3). Interestingly, ERAP2 maintains two haplotypes, a long and a short one, of which the latter can be decayed, accounting for approximately 25% of the population which does not express this protein [133].
ERAP1 and ERAP2 single nucleotide polymorphisms (SNPs) have been connected to autoimmune disorders as well as to the susceptibility to bacterial and viral infections, including COVID-19 [168,169]. This may be significant since patients with autoimmune disorders are more susceptible to both PTSD and CVD [170,171]. Indeed, thymic ERAP2 has been associated with autoimmune pathology and susceptibility to PTSD and CVD [169]. Furthermore, ERAP2, an ANG IV agonist, has been shown to upregulate the amygdalar OXT, emphasizing another protective mechanism [172]. ERAPs also protect against hypertension and cellular senescence by promptly hydrolyzing ANG II, terminating its action [173] (summarized in Figure 3). As certain ERAP polymorphisms may lead to chronic, less severe infection, this mechanism may explain the beneficial effect of these proteins in SARS-CoV-2, influenza, and hepatitis C [174,175,176,177]. Interestingly, a new study has reported that ERAP2 likely decreased the vulnerability to Yersinia pestis during the Middle Ages epidemic in Europe, the Middle East, and Northern Africa, emphasizing the antimicrobial properties of this protein [178]. The same authors surmise that ERAP2 resilience to plague may have contributed to the development of modern diseases, including autoimmunity [133,179,180]. We surmise that ERAP2 protects against PTSD and CVD and that loss of this protein drives thymus-mediated autoimmunity, predisposing to these pathologies [181,182].
ERAP1 and ERAP2 proteins reside in the ER and are, therefore, susceptible to ER stress, further linking these enzymes to PTSD and CVD [18,183,184,185]. Indeed, ERAP2 activates autophagy and UPR, suggesting that loss of this protein may also predispose to PTSD [158,186]. For this reason, we believe that:
  • A recombinant ERAP2 may be beneficial for the treatment of PTSD and CVD;
  • ERAP2 may be a viable marker for PTSD vulnerability.
Together, these data highlight several mechanisms linking ERAP2 loss to PTSD and CVD, along with autoimmunity, ER stress, impaired autophagy, thymic involution, dysfunctional OXT processing, and loss of ANG IV.

6. Potential Interventions

In this section, we take a closer look at several novel and potentially druggable targets in PTSD and CVD.

6.1. OXT

In the CNS, OXT upregulates gamma-aminobutyric acid (GABA), dampening the amygdalar response to stress [187,188]. In addition, OXT modulates the mesocorticolimbic release of dopamine, a neurotransmitter implicated in reward and motivation [189]. Human and animal studies have shown that excessive social isolation lowers OXT levels, removing a major PTSD/CVD protective factor [190,191,192]. Furthermore, as OXT protects against COVID-19 and perhaps long COVID, socialization restrictions during the pandemic may have been counterproductive [193,194,195]. Interestingly, gut microbiota, including Lactobacillus reuteri upregulates OXT, suggesting that supplementation with this probiotic may be beneficial for patients with PTSD/CVD [196,197].

6.2. HIF-1α

Tissue hypoxia upregulates HIF-1α, a protective transcription factor that exerts antidepressant effects, lowers glycolysis, and upregulates OXPHOS [198]. Intermittent hypoxia and hyperbaric oxygen have shown promising results in PTSD and CVD, highlighting the role of dysfunctional oxygenation in these disorders [199,200,201].
Adenosine, acting via the A2B receptor (A2BR), upregulates HIF-1α even in the absence of hypoxia, suggesting a druggable PTSD and CVD target [202]. Indeed, PSB-603, an A2BR antagonist, has been found to reduce both obesity and the effects of aging, indicating potential beneficial effects for ECs senescence, EVA, and thymic involution, associated with PTSD and CVD [203,204]. In addition, A2BRs modulate EPO synthesis, a hormone endowed with antidepressant and anxiolytic properties of its own [205,206].

6.3. ASIC1 Blockers

ASIC channels, discovered in 1997, are expressed in the central and peripheral nervous systems, where they exert neuroprotective effects [207]. However, excessive ASIC1 opening in response to lactate may trigger pathology, including cancer, chronic pain, anxiety, and PTSD [208,209,210]. In humans, the ASIC1 ortholog, amiloride-sensitive cation channel 2 (ACCN2), is expressed abundantly in the amygdala and the dopaminergic neurons of substantia nigra, areas implicated in neuropathology [211,212]. Since ACCN2 risk alleles, rs685012 and rs10875995 have been associated with panic disorder, manipulation of these proteins may be beneficial for PTSD patients [213].
The inability to cross the BBB is a major disadvantage of ACCN2 blockers; however, a few compounds do enter the CNS. They are as follows:
-
Amiloride, a potassium-sparing diuretic, which exerts efficacy at the pH of 6.5 and lowers blood pressure, is an established PTSD/CVD risk factor [214]. In our previous work, we as well as other groups, have suggested that amiloride be tested for PTSD [215,216].
-
C5B, a synthetic ASIC1 inhibitor, crosses the BBB and protects against neuronal apoptosis, indicating potential benefits for PTSD and CVD [216].
-
Sevanol, a natural lignan extracted from Thymus armeniacus, inhibits ASIC3 and ASIC1, exhibiting potent analgesic and anti-inflammatory properties, indicating potential usefulness in PTSD and CVD [217,218].
-
The natural flavonoid, Epigallocatechin gallate is a potent inhibitor of ASIC3, implicated in anxiety, pain, and insulin resistance, suggesting potential PTSD effectiveness [219,220].

6.4. Lactylation

Lactate plays a major role in the acidification of the tumor microenvironment, while in PTSD, it opens ACCN2, likely exacerbating PTSD symptoms. Lactylation, a PTM which can epigenetically activate or silence gene expression, may play a key role in the pathogenesis of both PTSD and CVD via HIF1α lactylation and functional loss [221]. Aside from its role in PTSD and CVD, HIF1α lactylation was shown to facilitate angiogenesis, exacerbating the invasiveness of some cancers [38]. Moreover, lactylation of membrane-organizing extension spike protein (MOESIN), a protector of cardiomyocytes and neurons, has been associated with premature EC senescence, emphasizing a new PTSD/CVD target [222,223].
Lactylation inhibitors are compounds currently in clinical trials for cancer. However, a small number of these agents exert properties potentially beneficial for PTSD or CVD. Among these, the most significant are lactate dehydrogenase (LDH) inhibitors and demethylzeylasteral (DML).
LDH inhibitors: LDH catalyzes the reversible conversion of lactate to pyruvate and the reduction of NAD+ to NADH. Therefore, inhibition of this enzyme lowers the Warburg effect, likely compelling cells to rely on OXPHOS. Elevated LDH is a risk factor for both PTSD and CVD (measured by Framingham risk score) [224,225]. Indeed, the role of LDH in psychological stress, known for the past four decades, was confirmed by numerous studies; however, the inhibitors of this enzyme and adverse effects have been poorly characterized [226,227,228]. At present, several LDH inhibitors affecting the epigenome may be relevant for PTSD and CVD, including:
-
5-aminolevulinic acid (5-ALA), which exerts beneficial effects in CVD along with the amelioration of mood, fatigue, and sleep, suggesting potential usefulness in PTSD [229,230,231].
-
Oxamate has been found helpful for both CVD and psychological stress, indicating potential PTSD benefits [232].
-
Quinoline 3-sulfonamides, is a novel LDH inhibitor that reverses aerobic glycolysis in cancer cells, and various quinoline derivatives have been tested as anticancer agents [233,234]. Quinolines have been known for their antidepressant and antipsychotic properties, as aripiprazole is derived from these compounds [235]. Several studies have demonstrated the beneficial effect of aripiprazole on PTSD, suggesting that these drugs should be further interrogated [236,237].
-
Galloflavin is a newer LDH inhibitor that, to our knowledge, has not been assessed for CVD or PTSD, but its pharmacological profile suggests potential efficacy.
Demethylzeylasteral (DML): a compound isolated from the plant Tripterygium wilfordii, is a traditional Chinese herbal medicine that inhibits histone lactylation, suggesting beneficial PTSD effects [238]. In this regard, triptolide, an extract of this plant, was shown to lower chronic pain and slow atherosclerosis progression. In addition, triptolide possesses antidepressant properties, suggesting a beneficial effect on PTSD [239].
Glycolysis inhibitors: including WZB117, STF31, or BAY 876, block glucose transporter 1 (GLUT-1), suppressing the Warburg effect in cancer cells [240]. However, blocking GLUT-1 has been associated with severe adverse effects, including seizures, as seen in GlLUT-1 deficiency syndrome (GLUT-1-DS), rendering these agents unsuitable for PTSD or CVD [241]. A summary of the potential target for future interventions in PTSD and CVD is shown in Table 2.

6.5. Interleukin 7

IL-7 prolongs the lifespan of T-cells by inducing the activity of telomerase to prevent loss of telomere length. As premature senescence and EVA drive PTSD and CVD, IL-7 supplementation could be beneficial for these conditions [249]. Along this line, GX-17, a homodimeric IL-7, currently in Phase 1b/2 for lymphopenia and cancer, appears to possess properties suitable for PTSD and CVD (NCT02860715) (NCT03752723) [247,248].
A negative regulator of IL-7, aryl hydrocarbon receptor (Ahr), has recently been associated with PTSD and CVD, indicating a druggable target [250,251]. Moreover, the Ahr repressor (AHRR) gene was linked to both PTSD and CVD, suggesting that epigenetic interventions at this level could benefit both conditions [252,253].

6.6. Autophagy and UPR

Psychological and biological stressors seem to merge at the subcellular level, disrupting autophagy and UPR. Therefore, targeting autophagy could be a viable strategy for treating PTSD and CVD [254,255]. Indeed, many currently utilized antidepressant drugs enhance autophagy, and the rapid antidepressant action of ketamine has been attributed to this effect [242,243,244,245]. There are also other autophagy activators to be considered. For example, gastrodin induces lysosomal biogenesis and autophagy, suggesting beneficial effects for both PTSD and CVD [256,257,258]. Moreover, as ERAP2 functions as an oxytocinase, recombinant ERAP2 could be beneficial for patients with CVD and PTSD [246].
There are numerous natural and synthetic autophagy activators; however, here, we focus primarily on those preventing or reversing thymic involution and/or lactylation.
AC-73 is a small molecule, an inhibitor of basigin (BSG) or cluster of differentiation 147 (CD147). Basigin, a matrix metalloproteinase inducer, is expressed on the cell surface and mitochondrial inner membrane. During the COVID-19 pandemic, this protein drew the attention of researchers and clinicians as it was identified as an alternative entry portal for the SARS-CoV-2 virus as well as Plasmodium malariae [259,260]. In the mitochondrial inner membrane, CD147 is colocalized with LDH, and lactate monocarboxylate transporter 1 (MCT1), linking this protein to the Warburg effect [261]. It has been shown that psychological stress upregulates CD147, which in turn may contribute to cardiac hypertrophy [262,263]. In addition, tissue hypoxia-upregulated CD147 promotes thymic involution, triggering cellular and immune senescence [264,265] (Figure 4).
There is a significant knowledge gap regarding the cellular effects of potential PTSD/CVD agents described above; therefore, further work is required to evaluate the pharmacodynamics, pharmacokinetics, and safety levels of these compounds.

6.7. Significance for the Field and Novel Targets

Although identified as a pathology during WWI when PTSD was known as “shell shock”, today, there is a paucity of treatments for this condition [266]. The PTSD cardiovascular involvement was hinted at by the term “soldier’s heart”, used even earlier, during the American Civil War, to describe feelings of anxiety, tension, and being “on edge”.
We have discussed in other articles premature endothelial senescence in PTSD and advocated for different approaches than those utilized today that lack efficacy in most patients [267]. For this reason, we focus primarily on ECs as they derive 80% of their energy from aerobic glycolysis and rarely utilize oxidative phosphorylation (OXFOS) [267]. In contrast, other brain cells, such as resting microglia, rely on OXFOS, while activated microglia shift their metabolism to aerobic glycolysis [268]. Astrocytes are primarily glycolytic and generate lactate that is shuttled to the neurons for their energy needs [269]. Neuroimaging studies have demonstrated that activated areas of the brain upregulate aerobic glycolysis while aging brains decrease this metabolic modality [270]. In PTSD, due to premature cellular aging, there is less activation or even deactivation in the ventromedial prefrontal cortex during trauma-induced script-driven imagery [271].
Although treated primarily by psychiatrists, PTSD is a systemic disease that has been associated with numerous medical problems, including chronic pain, hypertension, fatigue, muscle tension, headaches as well as metabolic syndrome, and immune dysfunction [272,273,274]. As systemic disorders call for systemic interventions, epigenetic therapy to lower the lactylation of histone proteins seems to be an adequate approach.
The treatment targets, including lowering cellular senescence, reversing thymic involution, and enhancing EPO and IL-7, may lower lactylation without totally inhibiting lactate, a metabolic modality utilized by healthy brains. For example, senolytic drugs, including dasatinib, quercetin, and rapamycin, oppose cellular senescence, a phenotype known to upregulate lactylation, especially in ECs [275,276]. Other systemic approaches include ASIC1a and 3-phosphoinositide-dependent protein kinase 1 (PDK1) inhibitors, agents that can reverse cellular senescence and showed beneficial effects in CVD and PTSD [277].

7. Conclusions

Life on Earth depends on oxygen, and the brain and heart are the highest consumers of this gas. For this reason, the cardiovascular system and the neuraxis are extremely sensitive to dysfunctional oxygenation. Due to the high energy demand, hypoxia, a danger signal, activates glycolysis, an oxygen-sparing metabolic modality, likely altering cerebral and cardiac function. Lower oxygen concentration triggers anxiety, while chronic hypoxia could lead to persistent feelings of apprehension, which in patients with PTSD can activate the core symptoms.
In compensating for hypoxia, the HIF-1α-EPO axis upregulates the systemic oxygen-carrying capacity, attempting to restore homeostasis. To preserve oxygen, OXT is lowered (as it upregulates the respiratory rate), and cellular housekeeping functions, including autophagy and UPR, are placed on hold. Furthermore, to conserve energy, cellular replication is stopped by the activation of cellular senescence, contributing to premature organismal aging.
Lactylation hinders the HIF-1α compensatory efforts by utilizing this molecule as a substrate, leading to functional loss. Psychological stress, activation of CD147, cardiac hypertrophy, thymic involution, and premature endothelial senescence increase BBB permeability allowing peripheral molecules, including lactate, to access the brain and induce PTSD symptoms. In contrast, the IL-7-thymus axis, a protective system, opposes senescence by several mechanisms, including telomere elongation, the EpiClock adjustment, and thymus reactivation.
Various approaches have been utilized to improve cellular respiration and the associated symptoms, and here we have emphasized several potential strategies which we believe are worth further investigation.

Author Contributions

Conceptualization, A.S.; methodology, P.S.; validation, C.K., and Z.K.; formal analysis, investigation, P.S.; data curation, G.A.; writing—original draft preparation, G.A. and P.S.; writing—review and editing, A.S.; supervision, Z.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The study did not report any data.

Conflicts of Interest

The authors declare no conflict of interest. Where authors are identified as personnel of the International Agency for Research on Cancer/WHO, the authors alone are responsible for the views expressed in this article and they do not necessarily represent the decisions, policy or views of the International Agency for Research on Cancer/WHO.

References

  1. Tasdemir, N.; Lowe, S.W. Senescent cells spread the word: Non-cell autonomous propagation of cellular senescence. EMBO J. 2013, 32, 1975–1976. [Google Scholar] [CrossRef]
  2. Faget, D.V.; Ren, Q.; Stewart, S.A. Unmasking senescence: Context-dependent effects of SASP in cancer. Nat. Rev. Cancer 2019, 19, 439–453. [Google Scholar] [CrossRef]
  3. Wolf, E.J.; Schnurr, P.P. PTSD-Related Cardiovascular Disease and Accelerated Cellular Aging. Psychiatry Ann. 2016, 46, 527–532. [Google Scholar] [CrossRef] [PubMed]
  4. Kuan, P.F.; Waszczuk, M.A.; Kotov, R.; Marsit, C.J.; Guffanti, G.; Gonzalez, A.; Yang, X.; Koenen, K.; Bromet, E.; Luft, B.J. An epigenome-wide DNA methylation study of PTSD and depression in World Trade Center responders. Transl. Psychiatry 2017, 7, e1158. [Google Scholar] [CrossRef] [PubMed]
  5. Wu, H.; Zhang, Y. Reversing DNA methylation: Mechanisms, genomics, and biological functions. Cell 2014, 156, 45–68. [Google Scholar] [CrossRef]
  6. Katrinli, S.; Stevens, J.; Wani, A.H.; Lori, A.; Kilaru, V.; van Rooij, S.J.H.; Hinrichs, R.; Powers, A.; Gillespie, C.F.; Michopoulos, V.; et al. Evaluating the impact of trauma and PTSD on epigenetic prediction of lifespan and neural integrity. Neuropsychopharmacology 2020, 45, 1609–1616. [Google Scholar] [CrossRef]
  7. Li, D.; Wang, L.; Zhou, Z.; Song, L.; Chen, S.; Yang, Y.; Hu, Y.; Wang, Y.; Wu, S.; Tian, Y. Lifetime risk of cardiovascular disease and life expectancy with and without cardiovascular disease according to changes in metabolic syndrome status. Nutr. Metab. Cardiovasc. Dis. 2022, 32, 373–381. [Google Scholar] [CrossRef] [PubMed]
  8. Hamadneh, L.; Al-Lakkis, L.; Alhusban, A.A.; Tarawneh, S.; Abu-Irmaileh, B.; Albustanji, S.; Al-Bawab, A.Q. Changes in Lactate Production, Lactate Dehydrogenase Genes Expression and DNA Methylation in Response to Tamoxifen Resistance Development in MCF-7 Cell Line. Genes 2021, 12, 777. [Google Scholar] [CrossRef]
  9. Cimmino, F.; Avitabile, M.; Lasorsa, V.A.; Montella, A.; Pezone, L.; Cantalupo, S.; Capasso, M. HIF-1 transcription activity: HIF1A driven response in normoxia and in hypoxia. BMC Med. Genet. 2019, 20, 37. [Google Scholar] [CrossRef]
  10. Welford, S.M.; Bedogni, B.; Gradin, K.; Poellinger, L.; Broome Powell, M.; Giaccia, A.J. HIF1alpha delays premature senescence through the activation of MIF. Genes Dev. 2006, 20, 3366–3371. [Google Scholar] [CrossRef]
  11. Hopkins, R.O.; Weaver, L.K. Acute psychosis associated with diving. Undersea Hyperb. Med. 2001, 28, 145–148. [Google Scholar] [PubMed]
  12. Chiner, E.; Arriero, J.M.; Signes-Costa, J.; Marco, J. Acute psychosis after CPAP treatment in a schizophrenic patient with sleep apnoea-hypopnoea syndrome. Eur. Respir. J. 2001, 17, 313–315. [Google Scholar] [CrossRef] [PubMed]
  13. Burtscher, J.; Niedermeier, M.; Hüfner, K.; van den Burg, E.; Kopp, M.; Stoop, R.; Burtscher, M.; Gatterer, H.; Millet, G.P. The interplay of hypoxic and mental stress: Implications for anxiety and depressive disorders. Neurosci. Biobehav. Rev. 2022, 138, 104718. [Google Scholar] [CrossRef]
  14. Vijayarangan, A.; Deru, K.; Hebert, D. Hyperbaric oxygen for mTBI-associated PCS and PTSD: Pooled analysis of results from Department of Defense and other published studies. Undersea Hyperb. Med. 2019, 46, 353–383. [Google Scholar]
  15. Parr, N.J.; Anderson, J.; Veazie, S. Evidence. Brief: Hyperbaric Oxygen Therapy for Traumatic Brain Injury and/or Post-Traumatic Stress Disorder; Department of Veterans Affairs (US): Washington, DC, USA, 2021. [Google Scholar]
  16. Bloch, Y.; Applebaum, J.; Osher, Y.; Amar, S.; Azab, A.N.; Agam, G.; Belmaker, R.H.; Bersudsky, Y. Normobaric hyperoxia treatment of schizophrenia. J. Clin. Psychopharmacol. 2012, 32, 525–530. [Google Scholar] [CrossRef]
  17. Akman, M.; Belisario, D.C.; Salaroglio, I.C.; Kopecka, J.; Donadelli, M.; De Smaele, E.; Riganti, C. Hypoxia, endoplasmic reticulum stress and chemoresistance: Dangerous liaisons. J. Exp. Clin. Cancer Res. 2021, 40, 28. [Google Scholar] [CrossRef]
  18. Nevell, L.; Zhang, K.; Aiello, A.E.; Koenen, K.; Galea, S.; Soliven, R.; Zhang, C.; Wildman, D.E.; Uddin, M. Elevated systemic expression of ER stress related genes is associated with stress-related mental disorders in the Detroit Neighborhood Health Study. Psychoneuroendocrinology 2014, 43, 62–70. [Google Scholar] [CrossRef] [PubMed]
  19. Hong, J.; Kim, K.; Kim, J.H.; Park, Y. The Role of Endoplasmic Reticulum Stress in Cardiovascular Disease and Exercise. Int. J. Vasc. Med. 2017, 2017, 2049217. [Google Scholar] [CrossRef] [PubMed]
  20. Mao, J.; Hu, Y.; Ruan, L.; Ji, Y.; Lou, Z. Role of endoplasmic reticulum stress in depression (Review). Mol. Med. Rep. 2019, 20, 4774–4780. [Google Scholar] [CrossRef]
  21. Lin, J.H.; Walter, P.; Yen, T.S. Endoplasmic reticulum stress in disease pathogenesis. Annu. Rev. Pathol. 2008, 3, 399–425. [Google Scholar] [CrossRef]
  22. Chipurupalli, S.; Kannan, E.; Tergaonkar, V.; D’Andrea, R.; Robinson, N. Hypoxia Induced ER Stress Response as an Adaptive Mechanism in Cancer. Int. J. Mol. Sci. 2019, 20, 749. [Google Scholar] [CrossRef] [PubMed]
  23. Minamino, T.; Kitakaze, M. ER stress in cardiovascular disease. J. Mol. Cell Cardiol. 2010, 48, 1105–1110. [Google Scholar] [CrossRef] [PubMed]
  24. Seligowski, A.V.; Misganaw, B.; Duffy, L.A.; Ressler, K.J.; Guffanti, G. Leveraging Large-Scale Genetics of PTSD and Cardiovascular Disease to Demonstrate Robust Shared Risk and Improve Risk Prediction Accuracy. Am. J. Psychiatry 2022, 179, 814–823. [Google Scholar] [CrossRef]
  25. Pullamsetti, S.S.; Mamazhakypov, A.; Weissmann, N.; Seeger, W.; Savai, R. Hypoxia-inducible factor signaling in pulmonary hypertension. J. Clin. Invest. 2020, 130, 5638–5651. [Google Scholar] [CrossRef]
  26. Culley, M.K.; Chan, S.Y. Endothelial Senescence: A New Age in Pulmonary Hypertension. Circ. Res. 2022, 130, 928–941. [Google Scholar] [CrossRef]
  27. Alique, M.; Sánchez-López, E.; Bodega, G.; Giannarelli, C.; Carracedo, J.; Ramírez, R. Hypoxia-Inducible Factor-1α: The Master Regulator of Endothelial Cell Senescence in Vascular Aging. Cells 2020, 9, 195. [Google Scholar] [CrossRef]
  28. Bussotti, M.; Sommaruga, M. Anxiety and depression in patients with pulmonary hypertension: Impact and management challenges. Vasc. Health. Risk. Manag. 2018, 14, 349–360. [Google Scholar] [CrossRef] [PubMed]
  29. McDivitt, J.D.; Barstow, C. Cardiovascular Disease Update: Pulmonary Hypertension. FP Essent. 2017, 454, 24–28. [Google Scholar] [PubMed]
  30. Sabbatinelli, J.; Prattichizzo, F.; Olivieri, F.; Procopio, A.D.; Rippo, M.R.; Giuliani, A. Where Metabolism Meets Senescence: Focus on Endothelial Cells. Front. Physiol. 2019, 10, 1523. [Google Scholar] [CrossRef]
  31. Eelen, G.; de Zeeuw, P.; Treps, L.; Harjes, U.; Wong, B.W.; Carmeliet, P. Endothelial Cell Metabolism. Physiol. Rev. 2018, 98, 3–58. [Google Scholar] [CrossRef]
  32. Fitzgerald, G.; Soro-Arnaiz, I.; De Bock, K. The Warburg Effect in Endothelial Cells and its Potential as an Anti-angiogenic Target in Cancer. Front. Cell Dev. Biol. 2018, 6, 100. [Google Scholar] [CrossRef] [PubMed]
  33. Lorenzen, J.M.; Martino, F.; Thum, T. Epigenetic modifications in cardiovascular disease. Basic Res. Cardiol. 2012, 107, 245. [Google Scholar] [CrossRef] [PubMed]
  34. Hermann, R.; Lay, D.; Wahl, P.; Roth, W.T.; Petrowski, K. Effects of psychosocial and physical stress on lactate and anxiety levels. Stress 2019, 22, 664–669. [Google Scholar] [CrossRef]
  35. Hagihara, H.; Shoji, H.; Otabi, H.; Toyoda, A.; Katoh, K.; Namihira, M.; Miyakawa, T. Protein lactylation induced by neural excitation. Cell Rep. 2021, 37, 109820. [Google Scholar] [CrossRef] [PubMed]
  36. Sara, J.D.S.; Toya, T.; Ahmad, A.; Clark, M.M.; Gilliam, W.P.; Lerman, L.O.; Lerman, A. Mental Stress and Its Effects on Vascular Health. Mayo. Clin. Proc. 2022, 97, 951–990. [Google Scholar] [CrossRef]
  37. Chistiakov, D.A.; Orekhov, A.N.; Bobryshev, Y.V. Endothelial Barrier and Its Abnormalities in Cardiovascular Disease. Front. Physiol. 2015, 6, 365. [Google Scholar] [CrossRef] [PubMed]
  38. Luo, Y.; Yang, Z.; Yu, Y.; Zhang, P. HIF1α lactylation enhances KIAA1199 transcription to promote angiogenesis and vasculogenic mimicry in prostate cancer. Int. J. Biol. Macromol. 2022, 222, 2225–2243. [Google Scholar] [CrossRef]
  39. Diekmann, F.; Legchenko, E.; Chouvarine, P.; Lichtinghagen, R.; Bertram, H.; Happel, C.M.; Hansmann, G. Circulating Interleukin-7 in Human Pulmonary Arterial Hypertension. Front. Cardiovasc. Med. 2021, 8, 794549. [Google Scholar] [CrossRef]
  40. Mori, K.; Nakaya, Y.; Sakamoto, S.; Hayabuchi, Y.; Matsuoka, S.; Kuroda, Y. Lactate-induced vascular relaxation in porcine coronary arteries is mediated by Ca2+-activated K+ channels. J. Mol. Cell Cardiol. 1998, 30, 349–356. [Google Scholar] [CrossRef]
  41. Duś, D.; Krawczenko, A.; Załecki, P.; Paprocka, M.; Wiedłocha, A.; Goupille, C.; Kieda, C. IL-7 receptor is present on human microvascular endothelial cells. Immunol. Lett. 2003, 86, 163–168. [Google Scholar] [CrossRef]
  42. Jacobs, S.R.; Michalek, R.D.; Rathmell, J.C. IL-7 is essential for homeostatic control of T cell metabolism in vivo. J. Immunol. 2010, 184, 3461–3469. [Google Scholar] [CrossRef]
  43. Shive, C.L.; Clagett, B.; McCausland, M.R.; Mudd, J.C.; Funderburg, N.T.; Freeman, M.L.; Younes, S.A.; Ferrari, B.M.; Rodriguez, B.; McComsey, G.A.; et al. Inflammation Perturbs the IL-7 Axis, Promoting Senescence and Exhaustion that Broadly Characterize Immune Failure in Treated HIV Infection. J. Acquir. Immune Defic. Syndr. 2016, 71, 483–492. [Google Scholar] [CrossRef] [PubMed]
  44. Nguyen, V.; Mendelsohn, A.; Larrick, J.W. Interleukin-7 and Immunosenescence. J. Immunol. Res. 2017, 2017, 4807853. [Google Scholar] [CrossRef]
  45. Ahlgrim, C.; Baumstark, M.W.; Roecker, K. Clarifying the Link Between the Blood Lactate Concentration and Cardiovascular Risk. Int. J. Sports Med. 2022, 43, 1106–1112. [Google Scholar] [CrossRef] [PubMed]
  46. Unterluggauer, H.; Mazurek, S.; Lener, B.; Hütter, E.; Eigenbrodt, E.; Zwerschke, W.; Jansen-Dürr, P. Premature senescence of human endothelial cells induced by inhibition of glutaminase. Biogerontology 2008, 9, 247–259. [Google Scholar] [CrossRef]
  47. Nilsson, P.M. Early Vascular Aging in Hypertension. Front. Cardiovasc. Med. 2020, 7, 6. [Google Scholar] [CrossRef]
  48. Ramírez, R.; Ceprian, N.; Figuer, A.; Valera, G.; Bodega, G.; Alique, M.; Carracedo, J. Endothelial Senescence and the Chronic Vascular Diseases: Challenges and Therapeutic Opportunities in Atherosclerosis. J. Pers. Med. 2022, 12, 215. [Google Scholar] [CrossRef] [PubMed]
  49. Sher, L.D.; Geddie, H.; Olivier, L.; Cairns, M.; Truter, N.; Beselaar, L.; Essop, M.F. Chronic stress and endothelial dysfunction: Mechanisms, experimental challenges, and the way ahead. Am. J. Physiol. Heart Circ. Physiol. 2020, 319, H488–H506. [Google Scholar] [CrossRef] [PubMed]
  50. Tekin, D.; Dursun, A.D.; Xi, L. Hypoxia inducible factor 1 (HIF-1) and cardioprotection. Acta Pharmacol. Sin. 2010, 31, 1085–1094. [Google Scholar] [CrossRef]
  51. Mellon, S.H.; Bersani, F.S.; Lindqvist, D.; Hammamieh, R.; Donohue, D.; Dean, K.; Jett, M.; Yehuda, R.; Flory, J.; Reus, V.I.; et al. Metabolomic analysis of male combat veterans with post-traumatic stress disorder. PLoS ONE 2019, 14, e0213839. [Google Scholar] [CrossRef]
  52. Yang, J.; Liu, C.; Du, X.; Liu, M.; Ji, X.; Du, H.; Zhao, H. Hypoxia Inducible Factor 1α Plays a Key Role in Remote Ischemic Preconditioning Against Stroke by Modulating Inflammatory Responses in Rats. J. Am. Heart Assoc. 2018, 7, e007589. [Google Scholar] [CrossRef]
  53. Manukhina, E.B.; Tseilikman, V.E.; Karpenko, M.N.; Pestereva, N.S.; Tseilikman, O.B.; Komelkova, M.V.; Kondashevskaya, M.V.; Goryacheva, A.V.; Lapshin, M.S.; Platkovskii, P.O.; et al. Intermittent Hypoxic Conditioning Alleviates Post-Traumatic Stress Disorder-Induced Damage and Dysfunction of Rat Visceral Organs and Brain. Int. J. Mol. Sci. 2020, 21, 345. [Google Scholar] [CrossRef] [PubMed]
  54. Serebrovskaya, T.V.; Xi, L. Intermittent hypoxia training as non-pharmacologic therapy for cardiovascular diseases: Practical analysis on methods and equipment. Exp. Biol. Med. 2016, 241, 1708–1723. [Google Scholar] [CrossRef]
  55. Fahy, G.M.; Brooke, R.T.; Watson, J.P.; Good, Z.; Vasanawala, S.S.; Maecker, H.; Leipold, M.D.; Lin, D.T.S.; Kobor, M.S.; Horvath, S. Reversal of epigenetic aging and immunosenescent trends in humans. Aging Cell 2019, 18, e13028. [Google Scholar] [CrossRef] [PubMed]
  56. Rey, F.; Balsari, A.; Giallongo, T.; Ottolenghi, S.; Di Giulio, A.M.; Samaja, M.; Carelli, S. Erythropoietin as a Neuroprotective Molecule: An Overview of Its Therapeutic Potential in Neurodegenerative Diseases. ASN Neuro. 2019, 11, 1759091419871420. [Google Scholar] [CrossRef]
  57. Burger, D.; Xenocostas, A.; Feng, Q.P. Molecular basis of cardioprotection by erythropoietin. Curr. Mol. Pharmacol. 2009, 2, 56–69. [Google Scholar] [CrossRef] [PubMed]
  58. Osborn, M.; Rustom, N.; Clarke, M.; Litteljohn, D.; Rudyk, C.; Anisman, H.; Hayley, S. Antidepressant-like effects of erythropoietin: A focus on behavioural and hippocampal processes. PLoS ONE 2013, 8, e72813. [Google Scholar] [CrossRef]
  59. Cantarelli, C.; Angeletti, A.; Cravedi, P. Erythropoietin, a multifaceted protein with innate and adaptive immune modulatory activity. Am. J. Transplant. 2019, 19, 2407–2414. [Google Scholar] [CrossRef]
  60. Sakanaka, M.; Wen, T.C.; Matsuda, S.; Masuda, S.; Morishita, E.; Nagao, M.; Sasaki, R. In vivo evidence that erythropoietin protects neurons from ischemic damage. Proc. Natl. Acad. Sci. USA 1998, 95, 4635–4640. [Google Scholar] [CrossRef]
  61. van der Meer, P.; Voors, A.A.; Lipsic, E.; van Gilst, W.H.; van Veldhuisen, D.J. Erythropoietin in cardiovascular diseases. Eur. Heart J. 2004, 25, 285–291. [Google Scholar] [CrossRef]
  62. Aiello, F.B.; Keller, J.R.; Klarmann, K.D.; Dranoff, G.; Mazzucchelli, R.; Durum, S.K. IL-7 induces myelopoiesis and erythropoiesis. J. Immunol. 2007, 178, 1553–1563. [Google Scholar] [CrossRef]
  63. Andrew, D.; Aspinall, R. Age-associated thymic atrophy is linked to a decline in IL-7 production. Exp. Gerontol. 2002, 37, 455–463. [Google Scholar] [CrossRef] [PubMed]
  64. Dai, X.; Zhang, D.; Wang, C.; Wu, Z.; Liang, C. The Pivotal Role of Thymus in Atherosclerosis Mediated by Immune and Inflammatory Response. Int. J. Med. Sci. 2018, 15, 1555–1563. [Google Scholar] [CrossRef] [PubMed]
  65. Fukunaga, T.; Mizoi, Y.; Yamashita, A.; Yamada, M.; Yamamoto, Y.; Tatsuno, Y.; Nishi, K. Thymus of abused/neglected children. Forensic. Sci. Int. 1992, 53, 69–79. [Google Scholar] [CrossRef]
  66. Gruver, A.L.; Sempowski, G.D. Cytokines, leptin, and stress-induced thymic atrophy. J. Leukoc. Biol. 2008, 84, 915–923. [Google Scholar] [CrossRef]
  67. Odinokov, D.; Hamblin, M.R. Aging of lymphoid organs: Can photobiomodulation reverse age-associated thymic involution via stimulation of extrapineal melatonin synthesis and bone marrow stem cells? J. Biophotonics. 2018, 11, e201700282. [Google Scholar] [CrossRef] [PubMed]
  68. Geenen, V.; Legros, J.J.; Franchimont, P.; Baudrihaye, M.; Defresne, M.P.; Boniver, J. The neuroendocrine thymus: Coexistence of oxytocin and neurophysin in the human thymus. Science 1986, 232, 508–511. [Google Scholar] [CrossRef] [PubMed]
  69. Moll, U.M. Functional histology of the neuroendocrine thymus. Microsc. Res. Tech. 1997, 38, 300–310. [Google Scholar] [CrossRef]
  70. Tani-ichi, S.; Shimba, A.; Wagatsuma, K.; Miyachi, H.; Kitano, S.; Imai, K.; Hara, T.; Ikuta, K. Interleukin-7 receptor controls development and maturation of late stages of thymocyte subpopulations. Proc. Natl. Acad. Sci. USA 2013, 110, 612–617. [Google Scholar] [CrossRef] [PubMed]
  71. van Zuiden, M.; Frijling, J.L.; Nawijn, L.; Koch, S.B.J.; Goslings, J.C.; Luitse, J.S.; Biesheuvel, T.H.; Honig, A.; Veltman, D.J.; Olff, M. Intranasal Oxytocin to Prevent Posttraumatic Stress Disorder Symptoms: A Randomized Controlled Trial in Emergency Department Patients. Biol. Psychiatry 2017, 81, 1030–1040. [Google Scholar] [CrossRef]
  72. Petersson, M. Cardiovascular effects of oxytocin. Prog. Brain Res. 2002, 139, 281–288. [Google Scholar] [CrossRef] [PubMed]
  73. Tirko, N.N.; Eyring, K.W.; Carcea, I.; Mitre, M.; Chao, M.V.; Froemke, R.C.; Tsien, R.W. Oxytocin Transforms Firing Mode of CA2 Hippocampal Neurons. Neuron 2018, 100, 593–608.e3. [Google Scholar] [CrossRef]
  74. Jankowski, M.; Broderick, T.L.; Gutkowska, J. The Role of Oxytocin in Cardiovascular Protection. Front. Psychol. 2020, 11, 2139. [Google Scholar] [CrossRef] [PubMed]
  75. 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] [PubMed]
  76. Genel, O.; Pariante, C.M.; Borsini, A. The role of AQP4 in the pathogenesis of depression, and possible related mechanisms. Brain Behav. Immun. 2021, 98, 366–377. [Google Scholar] [CrossRef]
  77. Rutkovskiy, A.; Stensløkken, K.O.; Mariero, L.H.; Skrbic, B.; Amiry-Moghaddam, M.; Hillestad, V.; Valen, G.; Perreault, M.C.; Ottersen, O.P.; Gullestad, L.; et al. Aquaporin-4 in the heart: Expression, regulation and functional role in ischemia. Basic Res. Cardiol. 2012, 107, 280. [Google Scholar] [CrossRef] [PubMed]
  78. Zhuang, J.C.; Wu, L.; Qian, M.Z.; Cai, P.P.; Liu, Q.B.; Zhao, G.X.; Li, Z.X.; Wu, Z.Y. Variants of Interleukin-7/Interleukin-7 Receptor Alpha are Associated with Both Neuromyelitis Optica and Multiple Sclerosis Among Chinese Han Population in Southeastern China. Chin. Med. J. 2015, 128, 3062–3068. [Google Scholar] [CrossRef]
  79. Wei, F.; Song, J.; Zhang, C.; Lin, J.; Xue, R.; Shan, L.D.; Gong, S.; Zhang, G.X.; Qin, Z.H.; Xu, G.Y.; et al. Chronic stress impairs the aquaporin-4-mediated glymphatic transport through glucocorticoid signaling. Psychopharmacology 2019, 236, 1367–1384. [Google Scholar] [CrossRef]
  80. Russell, G.; Lightman, S. The human stress response. Nat. Rev. Endocrinol. 2019, 15, 525–534. [Google Scholar] [CrossRef]
  81. Godoy, L.D.; Rossignoli, M.T.; Delfino-Pereira, P.; Garcia-Cairasco, N.; de Lima Umeoka, E.H. A Comprehensive Overview on Stress Neurobiology: Basic Concepts and Clinical Implications. Front. Behav. Neurosci. 2018, 12, 127. [Google Scholar] [CrossRef]
  82. Schneiderman, N.; Ironson, G.; Siegel, S.D. Stress and health: Psychological, behavioral, and biological determinants. Annu. Rev. Clin. Psychol. 2005, 1, 607–628. [Google Scholar] [CrossRef]
  83. Muscatell, K.A.; Merritt, C.C.; Cohen, J.R.; Chang, L.; Lindquist, K.A. The Stressed Brain: Neural Underpinnings of Social Stress Processing in Humans. Curr. Top. Behav. Neurosci. 2022, 54, 373–392. [Google Scholar] [CrossRef] [PubMed]
  84. Bremner, J.D. Traumatic stress: Effects on the brain. Dialogues Clin. Neurosci. 2006, 8, 445–461. [Google Scholar] [CrossRef]
  85. Janak, P.H.; Tye, K.M. From circuits to behaviour in the amygdala. Nature 2015, 517, 284–292. [Google Scholar] [CrossRef] [PubMed]
  86. Tawakol, A.; Ishai, A.; Takx, R.A.; Figueroa, A.L.; Ali, A.; Kaiser, Y.; Truong, Q.A.; Solomon, C.J.; Calcagno, C.; Mani, V.; et al. Relation between resting amygdalar activity and cardiovascular events: A longitudinal and cohort study. Lancet 2017, 389, 834–845. [Google Scholar] [CrossRef] [PubMed]
  87. Hostinar, C.E.; Sullivan, R.M.; Gunnar, M.R. Psychobiological mechanisms underlying the social buffering of the hypothalamic–pituitary–adrenocortical axis: A review of animal models and human studies across development. Psychol. Bull. 2014, 140, 256–282. [Google Scholar] [CrossRef] [PubMed]
  88. Saha, S. Role of the central nucleus of the amygdala in the control of blood pressure: Descending pathways to medullary cardiovascular nuclei. Clin. Exp. Pharmacol. Physiol. 2005, 32, 450–456. [Google Scholar] [CrossRef] [PubMed]
  89. Wellman, L.L.; Forcelli, P.A.; Aguilar, B.L.; Malkova, L. Bidirectional Control of Social Behavior by Activity within Basolateral and Central Amygdala of Primates. J. Neurosci. 2016, 36, 8746–8756. [Google Scholar] [CrossRef]
  90. Jackson, K.L.; Palma-Rigo, K.; Nguyen-Huu, T.P.; Davern, P.J.; Head, G.A. Major contribution of the medial amygdala to hypertension in BPH/2J genetically hypertensive mice. Hypertension 2014, 63, 811–818. [Google Scholar] [CrossRef]
  91. Marinzalda, M.L.; Pérez, P.A.; Gargiulo, P.A.; Casarsa, B.S.; Bregonzio, C.; Baiardi, G. Fear-potentiated behaviour is modulated by central amygdala angiotensin II AT1 receptors stimulation. Biomed. Res. Int. 2014, 2014, 183248. [Google Scholar] [CrossRef]
  92. Wiaderkiewicz, J.; Speth, R.C.; Park, J.; Marvar, P.J. Angiotensin II Type 2 Receptor-Expressing Neurons in the Central Amygdala Influence Fear-Related Behavior. Biol. Psychiatry 2019, 86, 899–909. [Google Scholar] [CrossRef]
  93. Labuschagne, I.; Phan, K.L.; Wood, A.; Angstadt, M.; Chua, P.; Heinrichs, M.; Stout, J.C.; Nathan, P.J. Oxytocin attenuates amygdala reactivity to fear in generalized social anxiety disorder. Neuropsychopharmacol 2010, 35, 2403–2413. [Google Scholar] [CrossRef] [PubMed]
  94. Ebner, K.; Bosch, O.J.; Krömer, S.A.; Singewald, N.; Neumann, I.D. Release of Oxytocin in the Rat Central Amygdala Modulates Stress-Coping Behavior and the Release of Excitatory Amino Acids. Neuropsychopharmacology 2005, 30, 223–230. [Google Scholar] [CrossRef] [PubMed]
  95. Thibonnier, M.; Conarty, D.M.; Preston, J.A.; Plesnicher, C.L.; Dweik, R.A.; Erzurum, S.C. Human vascular endothelial cells express oxytocin receptors. Endocrinology 1999, 140, 1301–1309. [Google Scholar] [CrossRef] [PubMed]
  96. Ishunina, T.A.; Swaab, D.F. Vasopressin and oxytocin neurons of the human supraoptic and paraventricular nucleus: Size changes in relation to age and sex. J. Clin. Endocrinol. Metab. 1999, 84, 4637–4644. [Google Scholar] [CrossRef]
  97. Wang, P.; Wang, S.C.; Liu, X.; Jia, S.; Wang, X.; Li, T.; Yu, J.; Parpura, V.; Wang, Y.F. Neural Functions of Hypothalamic Oxytocin and its Regulation. ASN Neuro. 2022, 14, 17590914221100706. [Google Scholar] [CrossRef]
  98. Sambuco, N.; Bradley, M.M.; Lang, P.J. Trauma-related dysfunction in the fronto-striatal reward circuit. J. Affect. Disord. 2021, 287, 359–366. [Google Scholar] [CrossRef]
  99. Taghva, A.; Oluigbo, C.; Corrigan, J.; Rezai, A.R. Posttraumatic stress disorder: Neurocircuitry and implications for potential deep brain stimulation. Stereotact. Funct. Neurosurg. 2013, 91, 207–219. [Google Scholar] [CrossRef]
  100. Logue, M.W.; van Rooij, S.J.H.; Dennis, E.L.; Davis, S.L.; Hayes, J.P.; Stevens, J.S.; Densmore, M.; Haswell, C.C.; Ipser, J.; Koch, S.B.J.; et al. Smaller Hippocampal Volume in Posttraumatic Stress Disorder: A Multisite ENIGMA-PGC Study: Subcortical Volumetry Results from Posttraumatic Stress Disorder Consortia. Biol. Psychiatry 2018, 83, 244–253. [Google Scholar] [CrossRef]
  101. Sailer, U.; Robinson, S.; Fischmeister, F.P.; König, D.; Oppenauer, C.; Lueger-Schuster, B.; Moser, E.; Kryspin-Exner, I.; Bauer, H. Altered reward processing in the nucleus accumbens and mesial prefrontal cortex of patients with posttraumatic stress disorder. Neuropsychologia 2008, 46, 2836–2844. [Google Scholar] [CrossRef]
  102. Hu, B.; Boyle, C.A.; Lei, S. Oxytocin receptors excite lateral nucleus of central amygdala by phospholipase Cβ- and protein kinase C-dependent depression of inwardly rectifying K+ channels. J. Physiol. 2020, 598, 3501–3520. [Google Scholar] [CrossRef] [PubMed]
  103. Hubbard, J.A.; Szu, J.I.; Binder, D.K. The role of aquaporin-4 in synaptic plasticity, memory and disease. Brain Res. Bull. 2018, 136, 118–129. [Google Scholar] [CrossRef] [PubMed]
  104. Verkerk, A.O.; Lodder, E.M.; Wilders, R. Aquaporin Channels in the Heart-Physiology and Pathophysiology. Int. J. Mol. Sci. 2019, 20, 2039. [Google Scholar] [CrossRef]
  105. Li, Y.K.; Wang, F.; Wang, W.; Luo, Y.; Wu, P.F.; Xiao, J.L.; Hu, Z.L.; Jin, Y.; Hu, G.; Chen, J.G. Aquaporin-4 deficiency impairs synaptic plasticity and associative fear memory in the lateral amygdala: Involvement of downregulation of glutamate transporter-1 expression. Neuropsychopharmacology 2012, 37, 1867–1878. [Google Scholar] [CrossRef]
  106. Kalsi, A.S.; Greenwood, K.; Wilkin, G.; Butt, A.M. Kir4.1 expression by astrocytes and oligodendrocytes in CNS white matter: A developmental study in the rat optic nerve. J. Anat. 2004, 204, 475–485. [Google Scholar] [CrossRef]
  107. Liberti, M.V.; Locasale, J.W. The Warburg Effect: How Does it Benefit Cancer Cells? Trends Biochem. Sci. 2016, 41, 211–218, Erratum in: Trends. Biochem. Sci. 2016, 41, 287. [Google Scholar] [CrossRef]
  108. Lee, T.Y. Lactate: A multifunctional signaling molecule. Yeungnam Univ. J. Med. 2021, 38, 183–193. [Google Scholar] [CrossRef] [PubMed]
  109. Zhang, D.; Tang, Z.; Huang, H.; Zhou, G.; Cui, C.; Weng, Y.; Liu, W.; Kim, S.; Lee, S.; Perez-Neut, M.; et al. Metabolic regulation of gene expression by histone lactylation. Nature 2019, 574, 575–580. [Google Scholar] [CrossRef]
  110. Yang, K.; Fan, M.; Wang, X.; Xu, J.; Wang, Y.; Tu, F.; Gill, P.S.; Ha, T.; Liu, L.; Williams, D.L.; et al. Lactate promotes macrophage HMGB1 lactylation, acetylation, and exosomal release in polymicrobial sepsis. Cell Death Differ. 2022, 29, 133–146. [Google Scholar] [CrossRef]
  111. Liebowitz, M.R.; Gorman, J.M.; Fyer, A.J.; Levitt, M.; Dillon, D.; Levy, G.; Klein, D.F. Lactate Provocation of Panic Attacks: II. Biochemical and Physiological Findings. Arch. Gen. Psychiatry 1985, 42, 709–719. [Google Scholar] [CrossRef]
  112. Jensen, C.F.; Keller, T.W.; Peskind, E.R.; McFall, M.E.; Veith, R.C.; Martin, D.; Wilkinson, C.W.; Raskind, M.A. Behavioral and neuroendocrine responses to sodium lactate infusion in subjects with posttraumatic stress disorder. Am. J. Psychiatry 1997, 154, 266–268. [Google Scholar] [CrossRef]
  113. Matsushita, K.; Williams, E.K.; Mongraw-Chaffin, M.L.; Coresh, J.; Schmidt, M.I.; Brancati, F.L.; Hoogeveen, R.C.; Ballantyne, C.M.; Young, J.H. The association of plasma lactate with incident cardiovascular outcomes: The ARIC Study. Am. J. Epidemiol. 2013, 178, 401–409. [Google Scholar] [CrossRef]
  114. Chen, Z.; Liu, M.; Li, L.; Chen, L. Involvement of the Warburg effect in non-tumor diseases processes. J. Cell Physiol. 2018, 233, 2839–2849. [Google Scholar] [CrossRef]
  115. Azoulay, I.S.; Qi, X.; Rozenfeld, M.; Liu, F.; Hu, Q.; Ben Kasus Nissim, T.; Stavsky, A.; Zhu, M.X.; Xu, T.L.; Sekler, I. ASIC1a senses lactate uptake to regulate metabolism in neurons. Redox. Biol. 2022, 51, 102253. [Google Scholar] [CrossRef]
  116. Chen, L.; Huang, L.; Gu, Y.; Cang, W.; Sun, P.; Xiang, Y. Lactate-Lactylation Hands between Metabolic Reprogramming and Immunosuppression. Int. J. Mol. Sci. 2022, 23, 11943. [Google Scholar] [CrossRef] [PubMed]
  117. Kozlov, A.M.; Lone, A.; Betts, D.H.; Cumming, R.C. Lactate preconditioning promotes a HIF-1α-mediated metabolic shift from OXPHOS to glycolysis in normal human diploid fibroblasts. Sci. Rep. 2020, 10, 8388. [Google Scholar] [CrossRef]
  118. Semenza, G.L. Hypoxia-inducible factor 1 and cardiovascular disease. Annu. Rev. Physiol. 2014, 76, 39–56. [Google Scholar] [CrossRef]
  119. Pan, R.Y.; He, L.; Zhang, J.; Liu, X.; Liao, Y.; Gao, J.; Liao, Y.; Yan, Y.; Li, Q.; Zhou, X.; et al. Positive feedback regulation of microglial glucose metabolism by histone H4 lysine 12 lactylation in Alzheimer’s disease. Cell Metab. 2022, 34, 634–648.e6. [Google Scholar] [CrossRef] [PubMed]
  120. Xie, Y.; Hu, H.; Liu, M.; Zhou, T.; Cheng, X.; Huang, W.; Cao, L. The role and mechanism of histone lactylation in health and diseases. Front. Genet. 2022, 13, 949252. [Google Scholar] [CrossRef] [PubMed]
  121. Yuan, K.; Gong, Y.M.; Liu, L.; Sun, Y.K.; Tian, S.S.; Wang, Y.J.; Zhong, Y.; Zhang, A.Y.; Su, S.Z.; Liu, X.X.; et al. Prevalence of posttraumatic stress disorder after infectious disease pandemics in the twenty-first century, including COVID-19: A meta-analysis and systematic review. Mol. Psychiatry 2021, 26, 4982–4998. [Google Scholar] [CrossRef]
  122. Sfera, A.; Osorio, C.; Rahman, L.; Zapata-Martín Del Campo, C.M.; Maldonado, J.C.; Jafri, N.; Cummings, M.A.; Maurer, S.; Kozlakidis, Z. PTSD as an Endothelial Disease: Insights From COVID-19. Front. Cell Neurosci. 2021, 15, 770387. [Google Scholar] [CrossRef]
  123. Israelski, D.M.; Prentiss, D.E.; Lubega, S.; Balmas, G.; Garcia, P.; Muhammad, M.; Cummings, S.; Koopman, C. Psychiatric co-morbidity in vulnerable populations receiving primary care for HIV/AIDS. AIDS Care 2007, 19, 220–225. [Google Scholar] [CrossRef] [PubMed]
  124. Bah, A.J.; James, P.B.; Bah, N.; Sesay, A.B.; Sevalie, S.; Kanu, J.S. Prevalence of anxiety, depression and post-traumatic stress disorder among Ebola survivors in northern Sierra Leone: A cross-sectional study. BMC Public Health 2020, 20, 1391. [Google Scholar] [CrossRef]
  125. Janiri, D.; Carfì, A.; Kotzalidis, G.D.; Bernabei, R.; Landi, F.; Sani, G.; Gemelli Against COVID-19 Post-Acute Care Study Group. Posttraumatic Stress Disorder in Patients After Severe COVID-19 Infection. JAMA Psychiatry 2021, 78, 567–569. [Google Scholar] [CrossRef]
  126. Icard, P.; Lincet, H.; Wu, Z.; Coquerel, A.; Forgez, P.; Alifano, M.; Fournel, L. The key role of Warburg effect in SARS-CoV-2 replication and associated inflammatory response. Biochimie 2021, 180, 169–177. [Google Scholar] [CrossRef]
  127. Quagliato, L.A.; Freire, R.C.; Nardi, A.E. The role of acid-sensitive ion channels in panic disorder: A systematic review of animal studies and meta-analysis of human studies. Transl. Psychiatry 2018, 8, 185. [Google Scholar] [CrossRef] [PubMed]
  128. Hollenbeck, A.; Weber, S.; Händschke, K.; Necke, M.; Opalka, B.; Dührsen, U.; Göthert, J.R. Activation of the Hypoxia-Inducible Factor Pathway Expands Early Thymic Progenitors. Blood 2014, 124, 2896. [Google Scholar] [CrossRef]
  129. Tayeri, K.; Asadollahi, K.; Madani, N.; Haghjooy Javanmard, S. Does COVID-19 Escalate Aging Process? A Possible Concern. Adv. Biomed. Res. 2022, 11, 106. [Google Scholar] [CrossRef]
  130. Cao, X.; Li, W.; Wang, T.; Ran, D.; Davalos, V.; Planas-Serra, L.; Pujol, A.; Esteller, M.; Wang, X.; Yu, H. Accelerated biological aging in COVID-19 patients. Nat. Commun. 2022, 13, 2135. [Google Scholar] [CrossRef]
  131. Sfera, A.; Osorio, C.; Jafri, N.; Diaz, E.L.; Campo Maldonado, J.E. Intoxication with Endogenous Angiotensin II: A COVID-19 Hypothesis. Front. Immunol. 2020, 11, 1472. [Google Scholar] [CrossRef]
  132. Lee, S.; Yu, Y.; Trimpert, J.; Benthani, F.; Mairhofer, M.; Richter-Pechanska, P.; Wyler, E.; Belenki, D.; Kaltenbrunner, S.; Pammer, M.; et al. Virus-induced senescence is a driver and therapeutic target in COVID-19. Nature 2021, 599, 283–289. [Google Scholar] [CrossRef] [PubMed]
  133. Mattorre, B.; Tedeschi, V.; Paldino, G.; Fiorillo, M.T.; Paladini, F.; Sorrentino, R. The emerging multifunctional roles of ERAP1, ERAP2 and IRAP between antigen processing and renin-angiotensin system modulation. Front. Immunol. 2022, 13, 1002375. [Google Scholar] [CrossRef]
  134. Marie, C.; Pedard, M.; Quirié, A.; Tessier, A.; Garnier, P.; Totoson, P.; Demougeot, C. Brain-derived neurotrophic factor secreted by the cerebral endothelium: A new actor of brain function? J. Cereb. Blood Flow Metab. 2018, 38, 935–949. [Google Scholar] [CrossRef] [PubMed]
  135. Taugher, R.J.; Lu, Y.; Fan, R.; Ghobbeh, A.; Kreple, C.J.; Faraci, F.M.; Wemmie, J.A. ASIC1A in neurons is critical for fear-related behaviors. Genes Brain Behav. 2017, 16, 745–755. [Google Scholar] [CrossRef] [PubMed]
  136. Yingjun, G.; Xun, Q. Acid-sensing ion channels under hypoxia. Channels 2013, 7, 231–237. [Google Scholar] [CrossRef] [PubMed]
  137. Dienel, G.A. Brain Glucose Metabolism: Integration of Energetics with Function. Physiol. Rev. 2019, 99, 949–1045. [Google Scholar] [CrossRef]
  138. Hayashi, T. Conversion of psychological stress into cellular stress response: Roles of the sigma-1 receptor in the process. Psychiatry Clin. Neurosci. 2015, 69, 179–191. [Google Scholar] [CrossRef]
  139. Hetz, C. The unfolded protein response: Controlling cell fate decisions under ER stress and beyond. Nat. Rev. Mol. Cell. Biol. 2012, 13, 89–102. [Google Scholar] [CrossRef]
  140. Mahendra, A.N.; Putra, I.N.A.J. Hippocampal Endoplasmic Reticulum Stress: Novel Target in PTSD Pharmacotherapy? Biomed. Pharmacol. J. 2018, 11, 1269–1274. [Google Scholar] [CrossRef]
  141. Ghemrawi, R.; Khair, M. Endoplasmic Reticulum Stress and Unfolded Protein Response in Neurodegenerative Diseases. Int. J. Mol. Sci. 2020, 21, 6127. [Google Scholar] [CrossRef]
  142. Díaz-Hung, M.L.; Martínez, G.; Hetz, C. Emerging roles of the unfolded protein response (UPR) in the nervous system: A link with adaptive behavior to environmental stress? Int. Rev. Cell Mol. Biol. 2020, 350, 29–61. [Google Scholar] [CrossRef] [PubMed]
  143. Ramalingam, L.; Sopontammarak, B.; Menikdiwela, K.R.; Moustaid-Moussa, N. Endoplasmic Reticulum (ER) Stress in Part Mediates Effects of Angiotensin II in Pancreatic Beta Cells. Diabetes Metab. Syndr. Obes. 2020, 13, 2843–2853. [Google Scholar] [CrossRef]
  144. Cicalese, S.; Cooper, H.; Preston, K.; Kawai, T.; Scalia, R.; Eguchi, S. Angiotensin II-induced ER stress response contributes to profibrotic VSMC phenotype and subsequent hypertensive arterial stiffness. J. Hypertens. 2021, 39, e40. [Google Scholar] [CrossRef]
  145. Su, J.B. Different cross-talk sites between the renin-angiotensin and the kallikrein-kinin systems. J. Renin. Angiotensin Aldosterone Syst. 2014, 15, 319–328. [Google Scholar] [CrossRef]
  146. Uddin, M.S.; Tewari, D.; Sharma, G.; Kabir, M.T.; Barreto, G.E.; Bin-Jumah, M.N.; Perveen, A.; Abdel-Daim, M.M.; Ashraf, G.M. Molecular Mechanisms of ER Stress and UPR in the Pathogenesis of Alzheimer’s Disease. Mol. Neurobiol. 2020, 57, 2902–2919. [Google Scholar] [CrossRef] [PubMed]
  147. Zhang, G.; Wang, X.; Gillette, T.G.; Deng, Y.; Wang, Z.V. Unfolded Protein Response as a Therapeutic Target in Cardiovascular Disease. Curr. Top. Med. Chem. 2019, 19, 1902–1917. [Google Scholar] [CrossRef]
  148. Kim, P.; Scott, M.R.; Meador-Woodruff, J.H. Dysregulation of the unfolded protein response (UPR) in the dorsolateral prefrontal cortex in elderly patients with schizophrenia. Mol. Psychiatry 2021, 26, 1321–1331. [Google Scholar] [CrossRef]
  149. Marciniak, S.J.; Chambers, J.E.; Ron, D. Pharmacological targeting of endoplasmic reticulum stress in disease. Nat. Rev. Drug. Discov. 2022, 21, 115–140. [Google Scholar] [CrossRef] [PubMed]
  150. Suliman, M.; Schmidtke, M.W.; Greenberg, M.L. The Role of the UPR Pathway in the Pathophysiology and Treatment of Bipolar Disorder. Front. Cell Neurosci. 2021, 15, 735622. [Google Scholar] [CrossRef]
  151. Srinivasan, V.; Korhonen, L.; Lindholm, D. The Unfolded Protein Response and Autophagy as Drug Targets in Neuropsychiatric Disorders. Front. Cell Neurosci. 2020, 14, 554548. [Google Scholar] [CrossRef]
  152. Kudo, K.; Ohta, M.; Yang, L.; Wakasa, Y.; Takahashi, S.; Takaiwa, F. ER stress response induced by the production of human IL-7 in rice endosperm cells. Plant. Mol. Biol. 2013, 81, 461–475. [Google Scholar] [CrossRef] [PubMed]
  153. Chehtane, M.; Khaled, A.R. Interleukin-7 mediates glucose utilization in lymphocytes through transcriptional regulation of the hexokinase II gene. Am. J. Physiol. Cell Physiol. 2010, 298, C1560–C1571. [Google Scholar] [CrossRef] [PubMed]
  154. Bekele, Y.; Sui, Y. Berzofsky JA. IL-7 in SARS-CoV-2 Infection and as a Potential Vaccine Adjuvant. Front. Immunol. 2021, 12, 737406. [Google Scholar] [CrossRef]
  155. Tang, C.; Goldsamt, L.; Meng, J.; Xiao, X.; Zhang, L.; Williams, A.B.; Wang, H. Global estimate of the prevalence of post-traumatic stress disorder among adults living with HIV: A systematic review and meta-analysis. BMJ Open 2020, 10, e032435. [Google Scholar] [CrossRef]
  156. Kaputu-Kalala-Malu, C.; Musalu, E.M.; Walker, T.; Ntumba-Tshitenge, O.; Ahuka-Mundeke, S. PTSD, depression and anxiety in Ebola virus disease survivors in Beni town, Democratic Republic of the Congo. BMC Psychiatry 2021, 21, 342. [Google Scholar] [CrossRef]
  157. Silva, S.L.; Albuquerque, A.S.; Matoso, P.; Charmeteau-de-Muylder, B.; Cheynier, R.; Ligeiro, D.; Abecasis, M.; Anjos, R.; Barata, J.T.; Victorino, R.M.; et al. IL-7-Induced Proliferation of Human Naive CD4 T-Cells Relies on Continued Thymic Activity. Front. Immunol. 2017, 8, 20. [Google Scholar] [CrossRef]
  158. Zheng, S.; Han, F.; Shi, Y.; Wen, L.; Han, D. Single-Prolonged-Stress-Induced Changes in Autophagy-Related Proteins Beclin-1, LC3, and p62 in the Medial Prefrontal Cortex of Rats with Post-traumatic Stress Disorder. J. Mol. Neurosci. 2017, 62, 43–54. [Google Scholar] [CrossRef] [PubMed]
  159. Zhu, Y.; Jiang, X.; Ding, Z.; Ming, J. Interleukin 7 inhibit autophagy via P53 regulated AMPK/mTOR signaling pathway in non-small cell lung cancer. Sci. Rep. 2022, 12, 11208. [Google Scholar] [CrossRef] [PubMed]
  160. Shehata, M.; Abdou, K.; Choko, K.; Matsuo, M.; Nishizono, H.; Inokuchi, K. Autophagy Enhances Memory Erasure through Synaptic Destabilization. J. Neurosci. 2018, 38, 3809–3822. [Google Scholar] [CrossRef]
  161. Abdellatif, M.; Ljubojevic-Holzer, S.; Madeo, F.; Sedej, S. Autophagy in cardiovascular health and disease. Prog. Mol. Biol. Transl. Sci. 2020, 172, 87–106. [Google Scholar] [CrossRef]
  162. Vranova, M.; Friess, M.C.; Haghayegh Jahromi, N.; Collado-Diaz, V.; Vallone, A.; Hagedorn, O.; Halin, C. Opposing roles of endothelial and leukocyte-expressed IL-7Rα in the regulation of psoriasis-like skin inflammation. Sci. Rep. 2019, 9, 11714. [Google Scholar] [CrossRef]
  163. Saric, T.; Chang, S.C.; Hattori, A.; York, I.A.; Markant, S.; Rock, K.L.; Tsujimoto, M.; Goldberg, A.L. An IFN-gamma-induced aminopeptidase in the ER, ERAP1, trims precursors to MHC class I-presented peptides. Nat. Immunol. 2002, 3, 1169–1176. [Google Scholar] [CrossRef]
  164. Zee, R.Y.L.; Rivera, A.; Inostroza, Y.; Ridker, P.M.; Chasman, D.I.; Romero, J.R. Gene Variation of Endoplasmic Reticulum Aminopeptidases 1 and 2, and Risk of Blood Pressure Progression and Incident Hypertension among 17,255 Initially Healthy Women. Int. J. Genom. 2018, 2018, 2308585. [Google Scholar] [CrossRef] [PubMed]
  165. Cochran, D.M.; Fallon, D.; Hill, M.; Frazier, J.A. The role of oxytocin in psychiatric disorders: A review of biological and therapeutic research findings. Harv. Rev. Psychiatry 2013, 21, 219–247. [Google Scholar] [CrossRef] [PubMed]
  166. Frijling, J.L. Preventing PTSD with oxytocin: Effects of oxytocin administration on fear neurocircuitry and PTSD symptom development in recently trauma-exposed individuals. Eur. J. Psychotraumatol. 2017, 8, 1302652. [Google Scholar] [CrossRef]
  167. Martínez-Martos, J.M.; Correa-Rodríguez, M.; Rus, A.; Molina, F.; Ramírez-Expósito, M.J.; Aguilar-Ferrandiz, M.E. Altered Serum Oxytocinase and Enkephalin-Degrading aminopeptidase activities in patients with Fibromyalgia. Biol. Res. Nurs. 2019, 21, 431–439. [Google Scholar] [CrossRef] [PubMed]
  168. López de Castro, J.A.; Alvarez-Navarro, C.; Brito, A.; Guasp, P.; Martín-Esteban, A.; Sanz-Bravo, A. Molecular and pathogenic effects of endoplasmic reticulum aminopeptidases ERAP1 and ERAP2 in MHC-i-associated inflammatory disorders: Towards a unifying view. Mol. Immunol. 2016, 77, 193–204. [Google Scholar] [CrossRef]
  169. Gabrielsen, I.; Viken, M.; Amundsen, S.S.; Helgeland, H.; Holm, K.; Flåm, S.T.; Lie, B.A. Autoimmune risk variants in ERAP2 are associated with gene-expression levels in thymus. Genes Immun. 2016, 17, 406–411. [Google Scholar] [CrossRef]
  170. Liew, J.; Lucas Williams, J.; Dobscha, S.; Barton, J.L. Posttraumatic stress disorder and correlates of disease activity among veterans with ankylosing spondylitis. Rheumatol. Int. 2017, 37, 1765–1769. [Google Scholar] [CrossRef]
  171. Qureshi, S.U.; Pyne, J.M.; Magruder, K.M.; Schulz, P.E.; Kunik, M.E. The link between post-traumatic stress disorder and physical comorbidities: A systematic review. Psychiatry Q. 2009, 80, 87–97. [Google Scholar] [CrossRef]
  172. Beyer, C.E.; Dwyer, J.M.; Platt, B.J.; Neal, S.; Luo, B.; Ling, H.P.; Lin, Q.; Mark, R.J.; Rosenzweig-Lipson, S.; Schechter, L.E. Angiotensin IV elevates oxytocin levels in the rat amygdala and produces anxiolytic-like activity through subsequent oxytocin receptor activation. Psychopharmacology 2010, 209, 303–311. [Google Scholar] [CrossRef] [PubMed]
  173. D’Amico, S.; Tempora, P.; Lucarini, V.; Melaiu, O.; Gaspari, S.; Algeri, M.; Fruci, D. ERAP1 and ERAP2 Enzymes: A Protective Shield for RAS against COVID-19? Int. J. Mol. Sci. 2021, 22, 1705. [Google Scholar] [CrossRef] [PubMed]
  174. Saulle, I.; Vanetti, C.; Goglia, S.; Vicentini, C.; Tombetti, E.; Garziano, M.; Clerici, M.; Biasin, M. A New ERAP2/Iso3 Isoform Expression Is Triggered by Different Microbial Stimuli in Human Cells. Could It Play a Role in the Modulation of SARS-CoV-2 Infection? Cells 2020, 9, 1951. [Google Scholar] [CrossRef] [PubMed]
  175. Ye, C.J.; Chen, J.; Villani, A.C.; Gate, R.E.; Subramaniam, M.; Bhangale, T.; Lee, M.N.; Raj, T.; Raychowdhury, R.; Li, W.; et al. Genetic analysis of isoform usage in the human anti-viral response reveals influenza-specific regulation of ERAP2 transcripts under balancing selection. Genome Res. 2018, 28, 1812–1825. [Google Scholar] [CrossRef]
  176. Gracia-Ramos, A.E.; Martin-Nares, E.; Hernández-Molina, G. New Onset of Autoimmune Diseases Following COVID-19 Diagnosis. Cells 2021, 10, 3592. [Google Scholar] [CrossRef]
  177. Liu, S.; Cao, D.; Shen, Y.; Li, Y.; Li, Y.; Shi, L.; Yu, J.; Li, C.; Zhang, X.; Sun, M.; et al. The ERAP gene is associated with HCV chronic infection in a Chinese Han population. Hum. Immunol. 2017, 78, 731–738. [Google Scholar] [CrossRef]
  178. Klunk, J.; Vilgalys, T.P.; Demeure, C.E. Evolution of immune genes is associated with the Black Death. Nature 2022, 611, 312–319. [Google Scholar] [CrossRef]
  179. Chang, Y.; Wei, W. Angiotensin II in inflammation, immunity and rheumatoid arthritis. Clin. Exp. Immunol. 2015, 179, 137–145. [Google Scholar] [CrossRef]
  180. Ferreira, L.C.; Gomes, C.E.M.; Duggal, P.; De Paula Holanda, I.; de Lima, A.S.; do Nascimento, P.R.P.; Jeronimo, S.M.B. Genetic association of ERAP1 and ERAP2 with eclampsia and preeclampsia in northeastern Brazilian women. Sci. Rep. 2021, 11, 6764. [Google Scholar] [CrossRef]
  181. Gelfand, J.M.; Wang, S.; Garshick, M.S. Autoimmune diseases and cardiovascular risk. Trends Mol. Med. 2022, 28, 1025–1027. [Google Scholar] [CrossRef]
  182. Song, H.; Fang, F.; Tomasson, G.; Arnberg, F.K.; Mataix-Cols, D.; Fernández de la Cruz, L.; Almqvist, C.; Fall, K.; Valdimarsdóttir, U.A. Association of Stress-Related Disorders with Subsequent Autoimmune Disease. JAMA 2018, 319, 2388–2400. [Google Scholar] [CrossRef]
  183. Blake, M.K.; O’Connell, P.; Pepelyayeva, Y.; Godbehere, S.; Aldhamen, Y.A.; Amalfitano, A. ERAP1 is a critical regulator of inflammasome-mediated proinflammatory and ER stress responses. BMC Immunol. 2022, 23, 9. [Google Scholar] [CrossRef] [PubMed]
  184. Guan, W.; Nakata, K.; Sagara, A.; Iwamoto, C.; Endo, S.; Matsuda, R.; Matsumoto, S.; Ikenaga, N.; Shindo, K.; Moriyama, T.; et al. ERAP2 is a novel target involved in autophagy and activation of pancreatic stellate cells via UPR signaling pathway. Pancreatology 2022, 22, 9–19. [Google Scholar] [CrossRef]
  185. Ren, J.; Bi, Y.; Sowers, J.R.; Hetz, C.; Zhang, Y. Endoplasmic reticulum stress and unfolded protein response in cardiovascular diseases. Nat. Rev. Cardiol. 2021, 18, 499–521. [Google Scholar] [CrossRef] [PubMed]
  186. Lei, X.; Yuan, Y.; Zou, Q. The role and mechanism of gastrodin in the medial prefrontal cortex autophagy of PTSD rats. Int. J. Clin. Exp. Pathol. 2020, 13, 989–994. [Google Scholar] [PubMed]
  187. Martin-Du Pan, R.C. L’ocytocine: Hormone de l’amour, de la confiance et du lien conjugal et social [Oxytocin: The hormone of love, trust and social bond. Clinical use in autism and social phobia]. Rev. Med. Suisse 2012, 8, 627–630. (In French) [Google Scholar]
  188. Qi, J.; Han, W.Y.; Yang, J.Y.; Wang, L.H.; Dong, Y.X.; Wang, F.; Song, M.; Wu, C.F. Oxytocin regulates changes of extracellular glutamate and GABA levels induced by methamphetamine in the mouse brain. Addict. Biol. 2012, 17, 758–769. [Google Scholar] [CrossRef]
  189. Love, T.M. Oxytocin, motivation and the role of dopamine. Pharmacol. Biochem. Behav. 2014, 119, 49–60. [Google Scholar] [CrossRef] [PubMed]
  190. Krimberg, J.S.; Lumertz, F.S.; Orso, R.; Viola, T.W.; de Almeida, R.M.M. Impact of social isolation on the oxytocinergic system: A systematic review and meta-analysis of rodent data. Neurosci. Biobehav. Rev. 2022, 134, 104549. [Google Scholar] [CrossRef]
  191. Engel, S.; Klusmann, H.; Laufer, S.; Pfeifer, A.C.; Ditzen, B.; van Zuiden, M.; Knaevelsrud, C.; Schumacher, S. Trauma exposure, posttraumatic stress disorder and oxytocin: A meta-analytic investigation of endogenous concentrations and receptor genotype. Neurosci. Biobehav. Rev. 2019, 107, 560–601. [Google Scholar] [CrossRef]
  192. Valtorta, N.K.; Kanaan, M.; Gilbody, S.; Hanratty, B. Loneliness, social isolation and risk of cardiovascular disease in the English Longitudinal Study of Ageing. Eur. J. Prev. Cardiol. 2018, 25, 1387–1396. [Google Scholar] [CrossRef] [PubMed]
  193. Golaszewski, N.M.; LaCroix, A.Z.; Godino, J.G.; Allison, M.A.; Manson, J.E.; King, J.J.; Weitlauf, J.C.; Bea, J.W.; Garcia, L.; Kroenke, C.H.; et al. Evaluation of Social Isolation, Loneliness, and Cardiovascular Disease Among Older Women in the US. JAMA Netw. Open 2022, 5, e2146461. [Google Scholar] [CrossRef]
  194. Wang, C.; Pan, R.; Wan, X.; Tan, Y.; Xu, L.; McIntyre, R.S.; Choo, F.N.; Tran, B.; Ho, R.; Sharma, V.K.; et al. A longitudinal study on the mental health of general population during the COVID-19 epidemic in China. Brain Behav. Immun. 2020, 87, 40–48. [Google Scholar] [CrossRef] [PubMed]
  195. Gryksa, K.; Neumann, I.D. Consequences of pandemic-associated social restrictions: Role of social support and the oxytocin system. Psychoneuroendocrinology 2022, 135, 105601. [Google Scholar] [CrossRef] [PubMed]
  196. Erdman, S.E.; Poutahidis, T. Microbes and Oxytocin: Benefits for Host Physiology and Behavior. Int. Rev. Neurobiol. 2016, 131, 91–126. [Google Scholar] [CrossRef] [PubMed]
  197. Varian, B.J.; Poutahidis, T.; DiBenedictis, B.T.; Levkovich, T.; Ibrahim, Y.; Didyk, E.; Shikhman, L.; Cheung, H.K.; Hardas, A.; Ricciardi, C.E.; et al. Microbial lysate upregulates host oxytocin. Brain Behav. Immun. 2017, 61, 36–49. [Google Scholar] [CrossRef] [PubMed]
  198. Kang, I.; Kondo, D.; Kim, J.; Lyoo, I.K.; Yurgelun-Todd, D.; Hwang, J.; Renshaw, P.F. Elevating the level of hypoxia inducible factor may be a new potential target for the treatment of depression. Med. Hypotheses 2021, 146, 110398. [Google Scholar] [CrossRef]
  199. Doenyas-Barak, K.; Catalogna, M.; Kutz, I.; Levi, G.; Hadanny, A.; Tal, S.; Daphna-Tekoha, S.; Sasson, E.; Shechter, Y.; Efrati, S. Hyperbaric oxygen therapy improves symptoms, brain’s microstructure and functionality in veterans with treatment resistant post-traumatic stress disorder: A prospective, randomized, controlled trial. PLoS ONE 2022, 17, e0264161. [Google Scholar] [CrossRef]
  200. Ding, F.S.; Cheng, X.; Zhao, T.; Zhao, Y.Q.; Zhang, G.B.; Wu, H.T.; Wu, K.W. Intermittent hypoxic preconditioning relieves fear and anxiety behavior in post-traumatic stress model mice. Sheng Li Xue Bao Acta Physiol. Sin. 2019, 71, 537–546. [Google Scholar]
  201. Aparci, M.; Kardesoglu, E.; Suleymanoglu, S.; Uzun, G.; Onem, Y.; Uz, O.; Kucukardali, Y.; Ozkan, S. Hyperbaric oxygen therapy improves myocardial diastolic function in diabetic patients. Tohoku. J. Exp. Med. 2008, 214, 281–289. [Google Scholar] [CrossRef]
  202. Hesse, J.; Groterath, W.; Owenier, C.; Steinhausen, J.; Ding, Z.; Steckel, B.; Czekelius, C.; Alter, C.; Marzoq, A.; Schrader, J. Normoxic induction of HIF-1α by adenosine-A2B R signaling in epicardial stromal cells formed after myocardial infarction. FASEB J. 2021, 35, e21517. [Google Scholar] [CrossRef]
  203. Gnad, T.; Navarro, G.; Lahesmaa, M.; Reverte-Salisa, L.; Copperi, F.; Cordomi, A.; Naumann, J.; Hochhäuser, A.; Haufs-Brusberg, S.; Wenzel, D.; et al. Adenosine/A2B Receptor Signaling Ameliorates the Effects of Aging and Counteracts Obesity. Cell Metab. 2020, 32, 56–70.e7. [Google Scholar] [CrossRef] [PubMed]
  204. Kotańska, M.; Dziubina, A.; Szafarz, M.; Mika, K.; Bednarski, M.; Nicosia, N.; Temirak, A.; Müller, C.E.; Kieć-Kononowicz, K. Preliminary Evidence of the Potent and Selective Adenosine A2B Receptor Antagonist PSB-603 in Reducing Obesity and Some of Its Associated Metabolic Disorders in Mice. Int. J. Mol. Sci. 2022, 23, 13439. [Google Scholar] [CrossRef]
  205. Fisher, J.W.; Brookins, J. Adenosine A(2A) and A(2B) receptor activation of erythropoietin production. Am. J. Physiol. Renal. Physiol. 2001, 281, F826–F832. [Google Scholar] [CrossRef] [PubMed]
  206. Sampath, D.; McWhirt, J.; Sathyanesan, M.; Newton, S.S. Carbamoylated erythropoietin produces antidepressant-like effects in male and female mice. Prog. Neuropsychopharmacol. Biol. Psychiatry 2020, 96, 109754. [Google Scholar] [CrossRef] [PubMed]
  207. Stankowska, D.L.; Mueller, B.H., 2nd; Oku, H.; Ikeda, T.; Dibas, A. Neuroprotective effects of inhibitors of Acid-Sensing ion channels (ASICs) in optic nerve crush model in rodents. Curr. Eye. Res. 2018, 43, 84–95. [Google Scholar] [CrossRef] [PubMed]
  208. Zhang, L.; Zheng, L.; Yang, X.; Yao, S.; Wang, H.; An, J.; Jin, H.; Wen, G.; Tuo, B. Pathology and physiology of acid-sensitive ion channels in the digestive system (Review). Int. J. Mol. Med. 2022, 50, 94. [Google Scholar] [CrossRef]
  209. Dwyer, J.M.; Rizzo, S.J.; Neal, S.J.; Lin, Q.; Jow, F.; Arias, R.L.; Rosenzweig-Lipson, S.; Dunlop, J.; Beyer, C.E. Acid sensing ion channel (ASIC) inhibitors exhibit anxiolytic-like activity in preclinical pharmacological models. Psychopharmacology 2009, 203, 41–52. [Google Scholar] [CrossRef]
  210. Gu, Q.; Lee, L.Y. Acid-Sensing Ion Channels and Pain. Pharmaceuticals 2010, 3, 1411–1425. [Google Scholar] [CrossRef]
  211. Arias, R.L.; Sung, M.L.; Vasylyev, D.; Zhang, M.Y.; Albinson, K.; Kubek, K.; Kagan, N.; Beyer, C.; Lin, Q.; Dwyer, J.M.; et al. Amiloride is neuroprotective in an MPTP model of Parkinson’s disease. Neurobiol. Dis. 2008, 31, 334–341. [Google Scholar] [CrossRef]
  212. Ziemann, A.E.; Allen, J.E.; Dahdaleh, N.S.; Drebot, I.I.; Coryell, M.W.; Wunsch, A.M.; Lynch, C.M.; Faraci, F.M.; Howard, M.A., 3rd; Welsh, M.J.; et al. The amygdala is a chemosensor that detects carbon dioxide and acidosis to elicit fear behavior. Cell 2009, 139, 1012–1021. [Google Scholar] [CrossRef] [PubMed]
  213. Smoller, J.W.; Gallagher, P.J.; Duncan, L.E.; McGrath, L.M.; Haddad, S.A.; Holmes, A.J.; Wolf, A.B.; Hilker, S.; Block, S.R.; Weill, S.; et al. The human ortholog of acid-sensing ion channel gene ASIC1a is associated with panic disorder and amygdala structure and function. Biol. Psychiatry 2014, 76, 902–910. [Google Scholar] [CrossRef] [PubMed]
  214. Bhagatwala, J.; Harris, R.A.; Parikh, S.J.; Zhu, H.; Huang, Y.; Kotak, I.; Seigler, N.; Pierce, G.L.; Egan, B.M.; Dong, Y. Epithelial sodium channel inhibition by amiloride on blood pressure and cardiovascular disease risk in young prehypertensives. J. Clin. Hypertens. 2014, 16, 47–53. [Google Scholar] [CrossRef] [PubMed]
  215. Battaglia, M.; Rossignol, O.; Bachand, K.; D’Amato, F.R.; De Koninck, Y. Amiloride modulation of carbon dioxide hypersensitivity and thermal nociceptive hypersensitivity induced by interference with early maternal environment. J. Psychopharmacol. 2019, 33, 101–108. [Google Scholar] [CrossRef]
  216. Qi, X.; Lu, J.F.; Huang, Z.Y.; Liu, Y.J.; Cai, L.B.; Wen, X.L.; Song, X.L.; Xiong, J.; Sun, P.Y.; Zhang, H.; et al. Pharmacological Validation of ASIC1a as a Druggable Target for Neuroprotection in Cerebral Ischemia Using an Intravenously Available Small Molecule Inhibitor. Front. Pharmacol. 2022, 13, 849498. [Google Scholar] [CrossRef]
  217. Osmakov, D.I.; Koshelev, S.G.; Belozerova, O.A.; Kublitski, V.S.; Andreev, Y.A.; Grishin, E.V.; Kozlov, S.A. The Biological Activity of the Sevanol and Its Analogues. Bioorg. Khim. 2015, 41, 606–611, In Russian. [Google Scholar] [CrossRef]
  218. Belozerova, O.A.; Osmakov, D.I.; Vladimirov, A.; Koshelev, S.G.; Chugunov, A.O.; Andreev, Y.A.; Palikov, V.A.; Palikova, Y.A.; Shaykhutdinova, E.R.; Gvozd, A.N.; et al. Sevanol and Its Analogues: Chemical Synthesis, Biological Effects and Molecular Docking. Pharmaceuticals 2020, 13, 163. [Google Scholar] [CrossRef] [PubMed]
  219. Wu, W.L.; Cheng, C.F.; Sun, W.H.; Wong, C.W.; Chen, C.C. Targeting ASIC3 for pain, anxiety, and insulin resistance. Pharmacol. Ther. 2012, 134, 127–138. [Google Scholar] [CrossRef]
  220. Yan, X.G.; Li, W.G.; Qi, X.; Zhu, J.J.; Huang, C.; Han, S.L.; Jiang, Q.; Xu, T.L.; Liu, J.H. Subtype-selective inhibition of acid-sensing ion channel 3 by a natural flavonoid. CNS Neurosci. Ther. 2019, 25, 47–56. [Google Scholar] [CrossRef]
  221. Gu, J.; Zhou, J.; Chen, Q.; Xu, X.; Gao, J.; Li, X.; Shao, Q.; Zhou, B.; Zhou, H.; Wei, S.; et al. Tumor metabolite lactate promotes tumorigenesis by modulating MOESIN lactylation and enhancing TGF-β signaling in regulatory T cells. Cell Rep. 2022, 39, 110986. [Google Scholar] [CrossRef] [PubMed]
  222. Miyawaki, A.; Mitsuhara, Y.; Orimoto, A.; Nakayasu, Y.; Tsunoda, S.; Obana, M.; Maeda, M.; Nakayama, H.; Yoshioka, Y.; Tsutsumi, Y.; et al. Moesin is activated in cardiomyocytes in experimental autoimmune myocarditis and mediates cytoskeletal reorganization with protrusion formation. Am. J. Physiol. Heart. Circ. Physiol. 2016, 311, H476–H486. [Google Scholar] [CrossRef]
  223. Freymuth, P.S.; Fitzsimons, H.L. The ERM protein Moesin is essential for neuronal morphogenesis and long-term memory in Drosophila. Mol. Brain 2017, 10, 41. [Google Scholar] [CrossRef] [PubMed]
  224. Swartz, C.M.; Breen, K.J. Multiple muscle enzyme release with psychiatric illness. J. Nerv. Ment. Dis. 1990, 178, 755–759. [Google Scholar] [CrossRef] [PubMed]
  225. Zhu, W.; Ma, Y.; Guo, W.; Lu, J.; Li, X.; Wu, J.; Qin, P.; Zhu, C.; Zhang, Q. Serum Level of Lactate Dehydrogenase is Associated with Cardiovascular Disease Risk as Determined by the Framingham Risk Score and Arterial Stiffness in a Health-Examined Population in China. Int. J. Gen. Med. 2022, 15, 11–17. [Google Scholar] [CrossRef]
  226. Ivanova, T.M.; Belova, T.I. Effect of emotional stress on lactate dehydrogenase isozyme spectrum in the rat reticular formation. Bull. Exp. Biol. Med. 1986, 102, 1331–1334. [Google Scholar] [CrossRef]
  227. Kato, A.; Sakakibara, H.; Tsuboi, H.; Tatsumi, A.; Akimoto, M.; Shimoi, K.; Ohashi, N. Depressive symptoms of female nursing staff working in stressful environments and their association with serum creatine kinase and lactate dehydrogenase—A preliminary study. BioPsychoSocial Med. 2014, 8, 21. [Google Scholar] [CrossRef] [PubMed]
  228. Padoli, P.; Suwito, J.; Hariyanto, T. Self-Affirmation Reduces the Anxiety, LDH and Troponin I in the Clients with Coronary Heart Disease (CHD). J. Ners. 2020, 14, 310–315. [Google Scholar] [CrossRef]
  229. Higashikawa, F.; Kanno, K.; Ogata, A.; Sugiyama, M. Reduction of fatigue and anger-hostility by the oral administration of 5-aminolevulinic acid phosphate: A randomized, double-blind, placebo-controlled, parallel study. Sci. Rep. 2020, 10, 16004. [Google Scholar] [CrossRef]
  230. Perez, M.; Shintani, T.; Rodriguez, B.; Davis, J.; Harrigan, R. The Role of 5-Aminolevulinic Acid (5-ALA) and Sleep. Int. J. Clin. Med. 2013, 4, 1–7. [Google Scholar] [CrossRef]
  231. Hagisawa, K.; Ayaori, M.; Ikewaki, K.; Nakajima, M.; Morimoto, Y. 5-Aminolevulinic Acid Attenuates Atherosclerotic Plaque Progression in Low-Density Lipoprotein Receptor-Deficient Mice by Heme Oxygenase-1 Induction. Circ. Rep. 2019, 2, 60–68. [Google Scholar] [CrossRef]
  232. Mohammed Abdul, K.S.; Jovanović, S.; Du, Q.; Sukhodub, A.; Jovanović, A. Mild hypoxia in vivo regulates cardioprotective SUR2A: A role for Akt and LDH. Biochim. Biophys. Acta 2015, 1852, 709–719. [Google Scholar] [CrossRef]
  233. Jafary, F.; Ganjalikhany, M.R.; Moradi, A.; Hemati, M.; Jafari, S. Novel Peptide Inhibitors for Lactate Dehydrogenase A (LDHA): A Survey to Inhibit LDHA Activity via Disruption of Protein-Protein Interaction. Sci. Rep. 2019, 9, 4686. [Google Scholar] [CrossRef] [PubMed]
  234. Matada, B.S.; Pattanashettar, R.; Yernale, N.G. A comprehensive review on the biological interest of quinoline and its derivatives. Bioorg. Med. Chem. 2021, 32, 115973. [Google Scholar] [CrossRef] [PubMed]
  235. Zajdel, P.; Marciniec, K.; Maślankiewicz, A.; Grychowska, K.; Satała, G.; Duszyńska, B.; Lenda, T.; Siwek, A.; Nowak, G.; Partyka, A.; et al. Antidepressant and antipsychotic activity of new quinoline- and isoquinoline-sulfonamide analogs of aripiprazole targeting serotonin 5-HT1A/5-HT2A/5-HT7 and dopamine D2/D3 receptors. Eur. J. Med. Chem. 2013, 60, 42–50. [Google Scholar] [CrossRef] [PubMed]
  236. Britnell, S.R.; Jackson, A.D.; Brown, J.N.; Capehart, B.P. Aripiprazole for Post-traumatic Stress Disorder: A Systematic Review. Clin. Neuropharmacol. 2017, 40, 273–278. [Google Scholar] [CrossRef] [PubMed]
  237. Richardson, J.D.; Fikretoglu, D.; Liu, A.; McIntosh, D. Aripiprazole augmentation in the treatment of military-related PTSD with major depression: A retrospective chart review. BMC Psychiatry 2011, 11, 86. [Google Scholar] [CrossRef]
  238. Pan, L.; Feng, F.; Wu, J.; Fan, S.; Han, J.; Wang, S.; Yang, L.; Liu, W.; Wang, C.; Xu, K. Demethylzeylasteral targets lactate by inhibiting histone lactylation to suppress the tumorigenicity of liver cancer stem cells. Pharmacol. Res. 2022, 181, 106270. [Google Scholar] [CrossRef] [PubMed]
  239. Hu, X.; Dong, Y.; Jin, X.; Zhang, C.; Zhang, T.; Zhao, J.; Shi, J.; Li, J. The novel and potent anti-depressive action of triptolide and its influences on hippocampal neuroinflammation in a rat model of depression comorbidity of chronic pain. Brain Behav. Immun. 2017, 64, 180–194. [Google Scholar] [CrossRef] [PubMed]
  240. Liu, Y.; Cao, Y.; Zhang, W.; Bergmeier, S.; Qian, Y.; Akbar, H.; Colvin, R.; Ding, J.; Tong, L.; Wu, S.; et al. A small-molecule inhibitor of glucose transporter 1 downregulates glycolysis, induces cell-cycle arrest, and inhibits cancer cell growth in vitro and in vivo. Mol. Cancer. Ther. 2012, 11, 1672–1682. [Google Scholar] [CrossRef] [PubMed]
  241. Koch, H.; Weber, Y.G. The glucose transporter type 1 (Glut1) syndromes. Epilepsy. Behav. 2019, 91, 90–93. [Google Scholar] [CrossRef]
  242. Gulbins, A.; Schumacher, F.; Becker, K.A.; Wilker, B.; Soddemann, M.; Boldrin, F.; Müller, C.P.; Edwards, M.J.; Goodman, M.; Caldwell, C.C.; et al. Antidepressants act by inducing autophagy controlled by sphingomyelin-ceramide. Mol. Psychiatry 2018, 23, 2324–2346. [Google Scholar] [CrossRef] [PubMed]
  243. Gassen, N.C.; Rein, T. Is There a Role of Autophagy in Depression and Antidepressant Action? Front. Psychiatry 2019, 10, 337. [Google Scholar] [CrossRef] [PubMed]
  244. Alcocer-Gómez, E.; Casas-Barquero, N.; Williams, M.R.; Romero-Guillena, S.L.; Cañadas-Lozano, D.; Bullón, P.; Sánchez-Alcazar, J.A.; Navarro-Pando, J.M.; Cordero, M.D. Antidepressants induce autophagy dependent-NLRP3-inflammasome inhibition in Major depressive disorder. Pharmacol. Res. 2017, 121, 114–121. [Google Scholar] [CrossRef]
  245. Lyu, D.; Wang, F.; Zhang, M.; Yang, W.; Huang, H.; Huang, Q.; Wu, C.; Qian, N.; Wang, M.; Zhang, H.; et al. Ketamine induces rapid antidepressant effects via the autophagy-NLRP3 inflammasome pathway. Psychopharmacology 2022, 239, 3201–3212. [Google Scholar] [CrossRef]
  246. Hill, L.D.; Hilliard, D.D.; York, T.P.; Srinivas, S.; Kusanovic, J.P.; Gomez, R.; Strauss, J.F. Fetal ERAP2 variation is associated with preeclampsia in African Americans in a case-control study. BMC Med. Genet. 2011, 12, 64. [Google Scholar] [CrossRef] [PubMed]
  247. Wolfarth, A.A.; Dhar, S.; Goon, J.B.; Ezeanya, U.I.; Ferrando-Martínez, S.; Lee, B.H. Advancements of Common Gamma-Chain Family Cytokines in Cancer Immunotherapy. Immune. Netw. 2022, 22, e5. [Google Scholar] [CrossRef]
  248. Lee, S.W.; Choi, D.; Heo, M.; Shin, E.C.; Park, S.H.; Kim, S.J.; Oh, Y.K.; Lee, B.H.; Yang, S.H.; Sung, Y.C.; et al. hIL-7-hyFc, A Long-Acting IL-7, Increased Absolute Lymphocyte Count in Healthy Subjects. Clin. Transl. Sci. 2020, 13, 1161–1169. [Google Scholar] [CrossRef]
  249. Yang, Y.; An, J.; Weng, N.P. Telomerase is involved in IL-7-mediated differential survival of naive and memory CD4+ T cells. J. Immunol. 2008, 180, 3775–3781. [Google Scholar] [CrossRef]
  250. Li, H.; Hong, W.; Jin, X.; Li, G.; Zhou, G.; Fan, L. The aryl hydrocarbon receptor is a novel negative regulator of interleukin-17-mediated signaling and inflammation in vitro. FEBS Lett. 2019, 593, 952–961. [Google Scholar] [CrossRef]
  251. Rothhammer, V.; Quintana, F.J. The aryl hydrocarbon receptor: An environmental sensor integrating immune responses in health and disease. Nat. Rev. Immunol. 2019, 19, 184–197. [Google Scholar] [CrossRef]
  252. Cole, J.W.; Xu, H. Aryl Hydrocarbon Receptor Repressor Methylation: A Link Between Smoking and Atherosclerosis. Circ. Cardiovasc. Genet. 2015, 8, 640–642. [Google Scholar] [CrossRef]
  253. Smith, A.K.; Ratanatharathorn, A.; Maihofer, A.X.; Naviaux, R.K.; Aiello, A.E.; Amstadter, A.B.; Ashley-Koch, A.E.; Baker, D.G.; Beckham, J.C.; Boks, M.P.; et al. Epigenome-wide meta-analysis of PTSD across 10 military and civilian cohorts identifies methylation changes in AHRR. Nat. Commun. 2020, 11, 5965. [Google Scholar] [CrossRef]
  254. Sakai, M.; Yu, Z.; Hirayama, R.; Nakasato, M.; Kikuchi, Y.; Ono, C.; Komatsu, H.; Nakanishi, M.; Yoshii, H.; Stellwagen, D.; et al. Deficient Autophagy in Microglia Aggravates Repeated Social Defeat Stress-Induced Social Avoidance. Neural. Plast. 2022, 2022, 7503553. [Google Scholar] [CrossRef] [PubMed]
  255. Mei, Y.; Thompson, M.D.; Cohen, R.A.; Tong, X. Autophagy and oxidative stress in cardiovascular diseases. Biochim. Biophys. Acta. 2015, 1852, 243–251. [Google Scholar] [CrossRef]
  256. Lee, B.; Sur, B.; Yeom, M.; Shim, I.; Lee, H.; Hahm, D.H. Gastrodin reversed the traumatic stress-induced depressed-like symptoms in rats. J. Nat. Med. 2016, 70, 749–759. [Google Scholar] [CrossRef] [PubMed]
  257. Liu, W.; Wang, L.; Yu, J.; Asare, P.F.; Zhao, Y.Q. Gastrodin Reduces Blood Pressure by Intervening with RAAS and PPARγ in SHRs. Evid. Based. Complement. Alternat. Med. 2015, 2015, 828427. [Google Scholar] [CrossRef] [PubMed]
  258. Tao, J.; Yang, P.; Xie, L.; Pu, Y.; Guo, J.; Jiao, J.; Sun, L.; Lu, D. Gastrodin induces lysosomal biogenesis and autophagy to prevent the formation of foam cells via AMPK-FoxO1-TFEB signalling axis. J. Cell Mol. Med. 2021, 25, 5769–5781. [Google Scholar] [CrossRef]
  259. Fenizia, C.; Galbiati, S.; Vanetti, C.; Vago, R.; Clerici, M.; Tacchetti, C.; Daniele, T. SARS-CoV-2 Entry: At the Crossroads of CD147 and ACE2. Cells 2021, 10, 1434. [Google Scholar] [CrossRef]
  260. van Ooij, C. Basigin opens the door to malaria. Nat. Rev. Microbiol. 2012, 10, 3. [Google Scholar] [CrossRef]
  261. Hashimoto, T.; Hussien, R.; Brooks, G.A. Colocalization of MCT1, CD147, and LDH in mitochondrial inner membrane of L6 muscle cells: Evidence of a mitochondrial lactate oxidation complex. Am. J. Physiol. Endocrinol. Metab. 2006, 290, E1237–E1244. [Google Scholar] [CrossRef]
  262. Wang, P.; Wang, Z.; Yan, Y.; Xiao, L.; Tian, W.; Qu, M.; Meng, A.; Sun, F.; Li, G.; Dong, J. Psychological Stress Up-Regulates CD147 Expression Through Beta-Arrestin1/ERK to Promote Proliferation and Invasiveness of Glioma Cells. Front. Oncol. 2020, 10, 571181. [Google Scholar] [CrossRef]
  263. Seizer, P.; Gawaz, M.; May, A.E. Cyclophilin A and EMMPRIN (CD147) in cardiovascular diseases. Cardiovasc. Res. 2014, 102, 17–23. [Google Scholar] [CrossRef]
  264. Chen, R.; Wang, K.; Feng, Z.; Zhang, M.Y.; Wu, J.; Geng, J.J.; Chen, Z.N. CD147 deficiency in T cells prevents thymic involution by inhibiting the EMT process in TECs in the presence of TGFβ. Cell Mol. Immunol. 2021, 18, 171–181. [Google Scholar] [CrossRef] [PubMed]
  265. Ke, X.; Fei, F.; Chen, Y.; Xu, L.; Zhang, Z.; Huang, Q.; Zhang, H.; Yang, H.; Chen, Z.; Xing, J. Hypoxia upregulates CD147 through a combined effect of HIF-1α and Sp1 to promote glycolysis and tumor progression in epithelial solid tumors. Carcinogenesis 2012, 33, 1598–1607. [Google Scholar] [CrossRef] [PubMed]
  266. McKenzie, A.G. Anaesthetic and other treatments of shell shock: World War I and beyond. J. R Army Med. Corps. 2012, 158, 29–33. [Google Scholar] [CrossRef]
  267. Du, W.; Ren, L.; Hamblin, M.H.; Fan, Y. Endothelial Cell Glucose Metabolism and Angiogenesis. Biomedicines 2021, 9, 147. [Google Scholar] [CrossRef] [PubMed]
  268. Cheng, J.; Zhang, R.; Xu, Z.; Ke, Y.; Sun, R.; Yang, H.; Zheng, L.T. Early glycolytic reprogramming controls microglial inflammatory activation. J. Neuroinflamm. 2021, 18, 129. [Google Scholar] [CrossRef] [PubMed]
  269. Mason, S. Lactate Shuttles in Neuroenergetics-Homeostasis, Allostasis and Beyond. Front. Neurosci. 2017, 11, 43. [Google Scholar] [CrossRef]
  270. Goyal, M.S.; Vlassenko, A.G.; Blazey, T.M.; Su, Y.; Couture, L.E.; Durbin, T.J.; Bateman, R.J.; Benzinger, T.L.; Morris, J.C.; Raichle, M.E. Loss of Brain Aerobic Glycolysis in Normal Human Aging. Cell Metab. 2017, 26, 353–360.e3. [Google Scholar] [CrossRef]
  271. Hughes, K.C.; Shin, L.M. Functional neuroimaging studies of post-traumatic stress disorder. Expert Rev. Neurother. 2011, 11, 275–285. [Google Scholar] [CrossRef]
  272. Michopoulos, V.; Vester, A.; Neigh, G. Posttraumatic stress disorder: A metabolic disorder in disguise? Exp. Neurol. 2016, 284, 220–229. [Google Scholar] [CrossRef] [PubMed]
  273. Wang, Z.; Caughron, B.; Young, M.R.I. Posttraumatic Stress Disorder: An Immunological Disorder? Front. Psychiatry 2017, 8, 222. [Google Scholar] [CrossRef]
  274. Boscarino, J.A. Posttraumatic stress disorder and physical illness: Results from clinical and epidemiologic studies. Ann. N. Y. Acad. Sci. 2004, 1032, 141–153. [Google Scholar] [CrossRef] [PubMed]
  275. Wróbel-Biedrawa, D.; Grabowska, K.; Galanty, A.; Sobolewska, D.; Podolak, I. A Flavonoid on the Brain: Quercetin as a Potential Therapeutic Agent in Central Nervous System Disorders. Life 2022, 12, 591. [Google Scholar] [CrossRef] [PubMed]
  276. Dookun, E.; Passos, J.F.; Arthur, H.M.; Richardson, G.D. Therapeutic Potential of Senolytics in Cardiovascular Disease. Cardiovasc. Drugs Ther. 2022, 36, 187–196. [Google Scholar] [CrossRef]
  277. Barile, E.; De, S.K.; Pellecchia, M. PDK1 inhibitors. Pharm. Pat. Anal. 2012, 1, 145–163. [Google Scholar] [CrossRef]
Figure 1. Common pathophysiological mechanisms of PTSD and CVD. Upregulated glycolysis in senescent endothelial cells generate excessive lactate, which serves as a lactylation substrate, triggering a downstream molecular cascade. Lactylation with loss of HIF-1 α induces additional senescence and glycolysis, disrupting autophagy, OXT, and EPO. In addition, it promotes premature aging, thymic involution, ER stress, and ANG II upregulation, engendering a vicious circle of hypoxia-cellular senescence-lactate—and further senescence.
Figure 1. Common pathophysiological mechanisms of PTSD and CVD. Upregulated glycolysis in senescent endothelial cells generate excessive lactate, which serves as a lactylation substrate, triggering a downstream molecular cascade. Lactylation with loss of HIF-1 α induces additional senescence and glycolysis, disrupting autophagy, OXT, and EPO. In addition, it promotes premature aging, thymic involution, ER stress, and ANG II upregulation, engendering a vicious circle of hypoxia-cellular senescence-lactate—and further senescence.
Biotech 12 00038 g001
Figure 2. A closer look at the PTSD/CVD connection: overwhelming psychological stressors activate the stress-processing brain areas, promoting glycolysis and excessive lactate. Lactate serves as a lactylation substrate, altering the function of many proteins, including HIF-1 α. Loss of HIF-1 α promotes hypoxia with subsequent premature endothelial senescence, depression, and anxiety. Premature endothelial senescence, stress-mediated release of catecholamines, and ANG II may trigger CVD.
Figure 2. A closer look at the PTSD/CVD connection: overwhelming psychological stressors activate the stress-processing brain areas, promoting glycolysis and excessive lactate. Lactate serves as a lactylation substrate, altering the function of many proteins, including HIF-1 α. Loss of HIF-1 α promotes hypoxia with subsequent premature endothelial senescence, depression, and anxiety. Premature endothelial senescence, stress-mediated release of catecholamines, and ANG II may trigger CVD.
Biotech 12 00038 g002
Figure 3. The SARS-CoV-2 virus attachment to ACE-2 disables this enzyme, leading to ANG II accumulation. ERAP1 And ERAP2 are complementary to ACE-2 as they hydrolyze ANG II into the protective ANG III and ANG IV. Loss of ERAP2 (in approximately 25% of the population) slows the processing of ANG II and OXT, increasing the susceptibility to some pathologies, such as hypertension, autoimmunity, PTSD, and CVD.
Figure 3. The SARS-CoV-2 virus attachment to ACE-2 disables this enzyme, leading to ANG II accumulation. ERAP1 And ERAP2 are complementary to ACE-2 as they hydrolyze ANG II into the protective ANG III and ANG IV. Loss of ERAP2 (in approximately 25% of the population) slows the processing of ANG II and OXT, increasing the susceptibility to some pathologies, such as hypertension, autoimmunity, PTSD, and CVD.
Biotech 12 00038 g003
Figure 4. ac-73 is a basigin (cd147) inhibitor that could be beneficial to patients with ptsd and cvd. hypoxia, thymic involution, and autoimmune inflammation upregulate cd47. In addition, psychological stress upregulates CD147, promoting CVD and senesce-related pathology.
Figure 4. ac-73 is a basigin (cd147) inhibitor that could be beneficial to patients with ptsd and cvd. hypoxia, thymic involution, and autoimmune inflammation upregulate cd47. In addition, psychological stress upregulates CD147, promoting CVD and senesce-related pathology.
Biotech 12 00038 g004
Table 1. Physiological and pathological Il-7 signaling.
Table 1. Physiological and pathological Il-7 signaling.
IL-7 SignalingPTSD/CVDReferences
(Adapted from)
Upregulate Warburg effectUpregulate Warburg effect[153]
Lower SARS-CoV-2 riskHigher SARS-CoV-2 risk[154,155,156]
Thymic activationThymic autophagy[65,157]
Upregulated autophagyDownregulated autophagy[158,159,160,161]
Lower endothelial/immune senescenceIncreased endothelial/immune senescence[44,162]
Table 2. Potential interventions for PTSD and CVD.
Table 2. Potential interventions for PTSD and CVD.
Therapeutic AgentMechanismReferences
(Adapted from)
OXT
Intranasal OXTSubstitution therapy
Lactobacillus reuteriSynthesizes OXT[196,197]
HIF-1α
Intermittent hypoxiaUpregulates HIF-1α[8,9,10]
Hyperbaric oxygenLowers hypoxia[11,12,13,14,15,16]
A2B blockers
PSB-603Decrease obesity and aging[203,204]
ASIC blockers
AmyloridePotassium-sparing diuretic[214,215]
C5BNeuroprotective[216]
SevanolASIC1 and ASIC3 inhibitor[219,220]
Epigallocatechin gallateASIC3 inhibitor
LDH inhibitors
5-ALAImproves mood, fatigue, sleep[229,230,231]
Quinoline 3-sulfonamidesReverses glycolysis[233,234]
GalloflavinInhibits lactate production; decreases ATP synthesis[217]
Autophagy activators
Antidepressants/ketamineIncrease autophagic markers LC3II/I[242,243,244,245]
Recombinant ERAP2Lower ER stress[246]
Lactylation inhibitors
Demethylzeylasteral (DML)Lowers depression, atherosclerosis, pain[238]
IL-7
IL-7 recombinant GX-17Prevents cellular senescence[247,248]
Thymic activators
PhotobiomodulationRestores thymic function[67]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kozlakidis, Z.; Shi, P.; Abarbanel, G.; Klein, C.; Sfera, A. Recent Developments in Protein Lactylation in PTSD and CVD: Novel Strategies and Targets. BioTech 2023, 12, 38. https://doi.org/10.3390/biotech12020038

AMA Style

Kozlakidis Z, Shi P, Abarbanel G, Klein C, Sfera A. Recent Developments in Protein Lactylation in PTSD and CVD: Novel Strategies and Targets. BioTech. 2023; 12(2):38. https://doi.org/10.3390/biotech12020038

Chicago/Turabian Style

Kozlakidis, Zisis, Patricia Shi, Ganna Abarbanel, Carolina Klein, and Adonis Sfera. 2023. "Recent Developments in Protein Lactylation in PTSD and CVD: Novel Strategies and Targets" BioTech 12, no. 2: 38. https://doi.org/10.3390/biotech12020038

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