OBM Neurobiology

(ISSN 2573-4407)

OBM Neurobiology is an international peer-reviewed Open Access journal published quarterly online by LIDSEN Publishing Inc. By design, the scope of OBM Neurobiology is broad, so as to reflect the multidisciplinary nature of the field of Neurobiology that interfaces biology with the fundamental and clinical neurosciences. As such, OBM Neurobiology embraces rigorous multidisciplinary investigations into the form and function of neurons and glia that make up the nervous system, either individually or in ensemble, in health or disease. OBM Neurobiology welcomes original contributions that employ a combination of molecular, cellular, systems and behavioral approaches to report novel neuroanatomical, neuropharmacological, neurophysiological and neurobehavioral findings related to the following aspects of the nervous system: Signal Transduction and Neurotransmission; Neural Circuits and Systems Neurobiology; Nervous System Development and Aging; Neurobiology of Nervous System Diseases (e.g., Developmental Brain Disorders; Neurodegenerative Disorders).

OBM Neurobiology publishes a variety of article types (Original Research, Review, Communication, Opinion, Comment, Conference Report, Technical Note, Book Review, etc.). Although the OBM Neurobiology Editorial Board encourages authors to be succinct, there is no restriction on the length of the papers. Authors should present their results in as much detail as possible, as reviewers are encouraged to emphasize scientific rigor and reproducibility.

Publication Speed (median values for papers published in 2024): Submission to First Decision: 7.6 weeks; Submission to Acceptance: 13.6 weeks; Acceptance to Publication: 6 days (1-2 days of FREE language polishing included)

Open Access Review

Love in the Time of COVID: Psychiatric Outcomes Related to Oxytocin and the “Endocrine Organ”

Adonis Sfera 1,*, Sabine Hazan 2, Jacob Anton 1, Ioana Ciuperca 3, Carolina Klein 4, Karina G. Thomas 1

  1. Patton State Hospital, Patton, CA, USA

  2. ProgenaBiome, Ventura, CA, USA

  3. UCLA, Department of Psychiatry, Los Angeles, CA, USA

  4. Department of State Hospitals, Sacramento, CA, USA

Correspondence: Adonis Sfera

Academic Editor: Bart Ellenbroek

Special Issue:  Time to Talk Outcomes: New Models and Strategies for Chronic Psychotic and Depressive Disorders

Received: February 09, 2025 | Accepted: April 29, 2025 | Published: May 09, 2025

OBM Neurobiology 2025, Volume 9, Issue 2, doi:10.21926/obm.neurobiol.2502287

Recommended citation: Sfera A, Hazan S, Anton J, Ciuperca I, Klein C, Thomas KG. Love in the Time of COVID: Psychiatric Outcomes Related to Oxytocin and the “Endocrine Organ”. OBM Neurobiology 2025; 9(2): 287; doi:10.21926/obm.neurobiol.2502287.

© 2025 by the authors. This is an open access article distributed under the conditions of the Creative Commons by Attribution License, which permits unrestricted use, distribution, and reproduction in any medium or format, provided the original work is correctly cited.

Abstract

The COVID-19 pandemic was an eye-opener for many medical disciplines. It highlighted viral exploitation of physiological cellular processes, including endocytosis and cellular senescence. These pathways play an essential role in cancer, neurodegenerative disorders, and schizophrenia (SCZ). Oxytocin, commonly called the “love hormone,” is produced in the posterior hypothalamus and is crucial for various physiological processes, including social intelligence, sexual activity, and metabolism. Many viruses, including SARS-CoV-2, have been shown to inhibit the release of oxytocin from intestinal epithelial cells, thus hastening gut barrier senescence. Premature molecular aging at this level enables microbial migration outside the intestinal lumen, triggering inflammation and immunogenicity. The gut microbial community is immunologically tolerated within the gastrointestinal tract but can activate host immunity upon translocation. Immune responses to displaced commensals and/or their components may contribute to neuroinflammation and gray matter volume reduction, a hallmark of severe mental illness. Oxytocin opposes microbial translocation into the systemic circulation through several mechanisms, including anti-inflammatory, tight junctions-upregulation, and suppression of senescence-associated secretory phenotype—the toxic secretome produced by senescent cells. Limosilactobacillus reuterii, a commensal microbe known for producing oxytocin, shows antiviral and anti-translocation effects, implying a beneficial role in schizophrenia. This condition has been linked to a dysfunctional gut barrier and increased microbial migration outside the intestinal lumen, suggesting that oxytocin replacement therapy could benefit patients with this mental illness. This review article summarizes the current understanding of oxytocin's role in schizophrenia and discusses natural and synthetic compounds that promote gut barrier homeostasis.

Graphical abstract

Click to view original image

Keywords

Oxitocin; neuroinflammation; gut barrier; blood-brain barrier; schizophrenia

1. Introduction

Viruses, including SARS-CoV-2, exploit other signaling pathways, including hormones and neurotransmitters, for their benefit. For example, the Japanese Encephalitis Virus (JEV) usurps dopamine (DA) receptors to enter host cells, therefore disrupting dopaminergic signaling [1]. Oxytocin (OXT) signaling with OXT receptors (OXRs) is essential for fertility, integrity of biological barriers, and emotional and self-referential processing in the central nervous system (CNS) [2,3,4]. Interestingly, patients with SCZ exhibit disruption of the same functions, reflected in fewer offspring, increased microbial translocation, and impaired cognitive-emotional processing [5,6]. Moreover, COVID-19 was associated with decreased birth rates and upregulated translocation markers and cases of new-onset SCZ, suggesting the intertwinement of underlying biological pathways [7,8,9].

The connection between viruses and severe mental illness has been documented for several decades. For example, women who were pregnant during the US rubella epidemic of 1964 gave birth to offspring that developed autism spectrum disorders (ASDs) or schizophrenia (SCZ) more often than the population at large, suggesting that many viruses, probably including COVID-19, may be associated with similar sequelae [10]. Other studies found that dormant viruses, such as human endogenous retroviruses (HERVs), could promote SCZ when activated by exogenous viral infections [11].

The gut microbes are known to produce most neurotransmitters and hormones generated by the host, so the microbiome is often conceptualized as the largest endocrine organ [12,13]. Limosilactobacillus reuteri (L. reuteri), one of the OXT-producing gut microbes, has been less abundant in the human microbiome over the past two or three decades, probably accounting for the increased prevalence of autoimmune disorders, conditions linked to microbial translocation [14]. L. reuteri is found in the GI tract, urinary tract, skin, and breast milk and exerts anti-inflammatory, antimicrobial, and anxiolytic properties [15,16].

OXT is a peptide hormone synthesized in the hypothalamic paraventricular (PVN) and supraoptic (SON) nuclei and stored in the posterior pituitary. In addition to L. reuteri, intestinal epithelial cells (IECs) produce OXT when exposed to the GI tract hormone secretin [16]. This may explain the beneficial effect of secretin in refractory SCZ observed by earlier studies [17,18]. OXT, implicated in social intelligence, signals with its receptors (OXRs) expressed throughout the brain [19,20,21].

The prosocial role of OXT and the fact that OXTRs are abundantly expressed in the insular cortex (IC) and anterior cingulate cortex (ACC) have led many researchers and clinicians to see a connection between OXT and the von Economo neurons (VENs) present in these areas [22,23]. VENs are part of the salience network (SN), a neuronal assembly that shifts attention to relevant stimuli or from exteroception to interoception [24]. This is significant as several studies linked the central action of OXT to an enhanced gut barrier [25].

Aside from the role of OXT in maintaining the homeostasis of the gut barrier, this review examines novel SCZ interventions based on the microbial translocation hypothesis, including aryl hydrocarbon receptor (AhR) antagonists, senolytic agents, and recombinant human interleukin 22 (Il-22).

2. Cellular Senescence

The common denominator between virus-induced pathology and severe mental illness (SMI) is premature cellular senescence. A physiological or pathological response to stress, cellular senescence is a phenotype characterized by proliferation arrest, metabolic restructuring, and the release of a toxic secretome known as the senescence-associated secretory phenotype (SASP). This toxin, composed of cytokines, chemokines, growth factors, and enzymes such as matrix metalloprotease-9 (MMP-9), is known to disrupt tight junctions (TJs), particularly occludins and claudin-5, which maintain the physiological permeability of the gut barrier [26] (Figure 1).

Click to view original image

Figure 1 Senescent brain cells release SASP, a toxic secretome containing MMP-9. MMP-9 degrades tight junctions (TJs), increasing the gut and blood-brain barrier (BBB) permeability. This enables microbial translocation from the gut lumen into the systemic circulation, eventually reaching the brain. TJs, occludins, and claudins act like molecular Velcro, holding cells of biological barriers together. In SCZ and certain viral infections, MMP-9 is upregulated, facilitating microbial translocation.

Virus-induced senescence (VIS) refers to viral infections that can trigger premature cellular senescence in human tissues by damaging the genome or hijacking various signaling pathways, including MMP-9 [27].

Under physiological circumstances, somatic cells replicate 40-60 times, after which they undergo senescence. Exceptions to this rule are cancer cells and probably stem cells, which can divide indefinitely. Aside from replicative senescence, cells can activate the senescence program when in danger of malignant transformation or when they contain damaged DNA.

Accumulation of senescent cells is believed to contribute to organismal aging, as SASP can spread the senescent phenotype locally and systemically. For example, aging endothelial cells (ECs) release SASP directly into the systemic circulation, disseminating senescence throughout the body. Several studies have shown that although senescence itself protects against malignant transformation, SASP maintains a degree of low-grade inflammation that may paradoxically predispose to cancer.

In patients with severe mental illness, the clearance of senescent cells is defective, leading to premature aging and low-grade inflammatory responses. Indeed, SCZ patients live on average 15-20 years shorter than the general population and develop age-related illnesses earlier in life, leading some researchers to conceptualize this disorder as a “segmental progeria” [28]. Along this line, telomeres are shorter in patients with SCZ, and senescent markers, such as p21, p16, and senescence-associated (SA)-β-galactosidase activity, are frequently elevated. On the other hand, OXT has been shown to improve social interaction and communication in children with autism spectrum disorders (ASD), highlighting a novel therapeutic strategy for this condition. Besides eliciting pro-social behavior, OXT exerts direct antiviral effects, particularly against the SARS-CoV-2 virus, as reported by several studies. Indeed, this action brought OXT into the research spotlight during the COVID-19 pandemic. OXT opposes cellular senescence, reduces SASP, and prevents the premature aging observed during the pandemic lockdowns [29]. OXT upregulates Claudin 5 and occludins, and suppressesMMP-9, reducing gut permeability [30,31].

OXT receptors are abundantly expressed in the myenteric plexus of the GI tract and are involved in gut motility, anti-inflammatory actions, and barrier homeostasis. This suggests that cholinergic neurotransmission contributes to OXT release, likely explained by the activation of signal transducer and activator of transcription 3 (STAT3) [32]. In addition, as OXT upregulates interleukin (IL)-10, an anti-inflammatory cytokine that shares its receptor with IL-22, preventing translocation [33] (Figure 2). Moreover, like IL-22, OXT mediates wound healing, further substantiating the beneficial effect of this hormone on biological barriers.

Click to view original image

Figure 2 IL-10 and IL-22 utilize the same receptors (IL-10 receptor beta and IL-22 receptor 1). The binding of IL-22 to these receptors activates STAT3 and Mitogen-Activated Protein Kinase (MAPK). Reduced brain-derived neurotrophic factor (BDNF) enhances the cholinergic system-linked STAT3, especially in the insular and anterior cingulate cortex (IC) (ACC). BDNF binding to its receptor, TrkB, mitigates inflammation. Low BDNF levels elevate pro-inflammatory cytokines such as TNFα and interleukin 1 beta (IL-1β).

Due to SASP, cellular senescence maintains inflammation and neuroinflammation that drives gray matter volume (GMV) reduction, a hallmark of SCZ demonstrated by numerous neuroimaging studies (Table 1). GMV loss is in line with SCZ outcome studies, which show that only a minority of patients attain complete recovery after the first psychotic episode, suggesting that brain volume atrophy may progress despite the use of antipsychotic drugs [34]. White matter is also depleted in SCZ, although less than the gray matter, probably reflecting myelin damage.

Table 1 The molecular underpinnings of idiopathic (exemplified by SCZ) and virus-induced senescence (columns 1 and 2). Premature molecular aging leads to a dysfunctional gut barrier and BBB, which contribute to GMV reduction. Column 3 shows the opposing effects of OXT.

GMV reduction in SCZ (occurring in medicated and unmedicated patients) has been associated with aggression and violence [63,64,65]. Conversely, OXT treatment may improve SCZ outcomes by preserving the GMV as documented in traumatic brain injury (TBI) [66,67]. At present, SCZ outcomes (measured by sustained recovery after the first psychotic episode) are below 15%. In this regard, 33% of patients relapse during the first 12 months after an initial psychotic episode, 26% remain homeless at two-year follow-up, while five years after the first psychotic outbreak, only 10% are employed [68,69,70]. These findings suggest that incorporating OXT into the standard SCZ treatment may enhance the overall outcome by preventing GMV reduction.

3. Senescent Microglia, Neuronophagy, and GMV Depletion

Premature microglial senescence may lead to aberrant activation of these cells and paradoxical neuronophagy (elimination of healthy neurons) [71,72]. Microglia are brain-resident macrophages, vigilant cells that constantly scan the brain parenchyma and repair the damaged tissue, maintaining cerebral homeostasis [73].

Under physiological circumstances, senescent microglia can be adaptive, participating in tissue remodeling and repair. Both young and aged microglia respond to injuries, apparent molecular debris-induced inflammation, and damaged or senescent cells [74]. Dysfunctional senescent microglia may release pro-inflammatory cytokines and chemokines, contributing to chronic inflammation. These findings underscore the importance of more research on microglial phenotypes to develop novel therapeutic strategies for neuropsychiatric disorders. Indeed, aberrant neuronophagy by senescent microglia may play a significant role in developing age-associated neurodegenerative disorders and SCZ [75,76]. For this reason, targeting senescent microglia and/or restoring the homeostasis of these cells could reduce neuroinflammation, neuronophagy, and GMV loss, potentially leading to improved outcomes in SCZ and neurodegenerative disorders.

In summary, the interplay between senescent microglia and neuronophagy is crucial for understanding the pathogenesis of several neuropsychiatric conditions and developing new treatment strategies.

4. The Role of Iron in Early Microglial Aging

The connection between senescence and iron accumulation is well-established. For example, intracellular iron levels 40 times higher were documented in senescent cells, linking aberrant neuronophagy to excess iron [77].

Furthermore, since reduced GMV has been directly correlated with aggressive behavior in neuropsychiatric disorders, abnormal iron levels in brain cells may drive both atrophy and violence. Indeed, like neuronophagy, ferroptosis, iron-induced cell death in the absence of lipid-repairing antioxidants, is a form of neuronal death without the involvement of glial cells [78]. On the other hand, iron accumulation in microglia, activation of these cells, and subsequent neuronophagy can also result in aggressive and violent behaviors [79].

A growing body of evidence supports that iron accumulation in senescent microglia activates these cells, triggering neurotoxicity, a phenotype known for eliminating viable neurons and synapses [80,81]. Indeed, neurotoxic microglia engage in the phagocytosis of live neurons and convert astrocytes into neurotoxic cells, further contributing to GMV reduction.

Lysine lactylation (Kla) in histone proteins, a lactate-linked posttranslational modification, was demonstrated to play a key role in behavioral regulation [82,83]. Lactate modulates iron metabolism via hepcidin, a liver hormone in charge of iron egress from hepatocytes and neurons [84]. In addition, a novel iron-histone pathway was recently described, further linking this biometal to lactylation, and aggression [84]. Lactylation appears to play a significant role in disseminating neurotoxicity from senescent microglia to astrocytes, bringing these brain macrophages into the arena of forensic psychiatry [85]. For example, iron as well as lactylation of histone 3 lysine 18 (H3K18) can trigger astrocytic neurotoxicity and subsequent reduction in GMV and emergence of violent behaviors [86]. Cellular senescence and SASP have been shown to promote cortical thinning, linking senescence to aggressive disturbances. In a previous article, we discussed ferosenescence, a phenomenon that is the opposite of ferroptosis [87]. In the brain, senescent neurons can undergo ferroptosis if astrocytes are damaged or ferrosenescence if astrocytes are viable and capable of supplying the neurons with the necessary antioxidants, such as glutathione peroxidase 4 (GPX4), ferritin, and “fresh” organelles, including mitochondria or lysosomes. The transfer of protective biomolecules from astrocytes to neurons through tunneling nanotubules (TNT) or extracellular vesicles (EVs) may prevent ferroptosis and enable the cell to thrive in ferrosenescenescence.

Recent studies have reported that antidepressant drugs, selective serotonin reuptake inhibitors (SSRIs) facilitate mitochondrial trafficking from astrocytes to neurons, linking major depressive disorder to low mitochondrial count [88] (Figure 3).

Click to view original image

Figure 3 Among other supportive functions, astrocytes assist neurons by transferring fresh antioxidants like GPX4 and mitochondria. A recently discovered transfer modality, tunneling nanotubules, has been used to transport mitochondria and lysosomes from astrocytes to neurons.

5. Aryl Hydrocarbon Receptor, Iron, and Nutritional Immunity

The aryl hydrocarbon receptor (AhR) was initially known as the dioxin receptor. However, many ligands at this protein have been identified over the years, some of which are relevant to neuropathology. For example, serotonin (5-HT), melatonin, and dopamine (DA) are upstream AhR ligands, bringing this protein into the neuropsychiatric arena. In addition, AhR is the master regulator of cellular senescence as it can sense endogenous and exogenous metabolites, xenobiotics, and toxicants.

AhR negatively regulates lactate and its posttranslational modification, lactylation. Consequently, dysfunctional AhR may drive premature neuronal and glial aging and the excessive lactylation documented in SCZ and other psychotic disorders [89].

Translocated microbes can trigger iron sequestration in host macrophages, a phenomenon known as nutritional immunity. Nutritional immunity aims to withhold iron from the invading pathogens, decreasing infectivity [90]. However, low circulatory iron often causes bacteria in host tissues, including the brain, to adopt a dormant phenotype, awaiting nutrient availability [91]. For example, dormant brain microbes were documented in Alzheimer’s disease (AD), while a virome (viral microbiome) was recently identified in SCZ brains (Broadman area 46) [92,93,94]. The dormant microbes can be reactivated when iron becomes available, causing neuropathology. Moreover, iron regulatory proteins, including hepcidin, are AhR ligands, emphasizing a previously unknown relationship between AhR and nutritional immunity [92].

AhR is located at the biological barriers and comes readily into contact with endogenous and exogenous nutrients and toxins that bind this receptor with various affinities, eliciting different degrees of activation. Excessive AhR activation likely disrupts TJs, opening paracellular pathways utilized by microbes or their components to migrate outside the GI tract. In this regard, SCZ with negative symptoms was associated with antibodies against translocated Hafnei alvei, Pseudomonas aeruginosa, Pseudomonas putida, and Klebsiella pneumonia, which are proof of concept of microbial migration in this disorder [93].

Viruses also induce cellular senescence by manipulating AhR via viral antigens. For example, the SARS-CoV-2 virus usurps OXT via the OXT-activating motif found in calcium calmodulin kinase II (CaMKII), inducing senescence [94]. Moreover, the S antigen of the SARS-CoV-2 virus contains soluble N-ethylmaleimide-sensitive fusion protein attachment protein receptors (SNARE) repeats that can interfere with the vesicular release of OXT, further lowering the systemic level of this hormone [95].

Molecular imitation of placental proteins by viral arginine repeats, HAP2, tubulin, tau, and human endogenous retroviruses (HERVs) facilitates viral infection and the development of antibodies against host proteins, likely accounting for the reduced birth rates documented worldwide after the pandemic. Viral repeats (motifs) are known AhR activators, an action that manipulates AhR into adopting a pro-viral role. Activated AhR lowers interferon-gamma, a potent antiviral defense [96]. Therefore, AhR inhibitors exert not only anti-translocation but also antiviral properties.

6. Possible Interventions

The microbial translocation hypothesis opens new therapeutic strategies beyond the synapse and dopamine. These strategies include senotherapeutics, barrier enhancers, and AhR inhibitors.

6.1 Senotherapeutics

SCZ has been associated with premature molecular aging, which is reflected in decreased longevity compared to the general population. Premature molecular aging likely results in dysfunctional TJs and subsequent microbial translocation in the host’s tissues and organs.

Senotherapeutic drugs include senolytic agents (which promote the elimination of senescent cells) and senomorphic compounds (which delete senescent markers).

Senotherapeutics are underutilized in SCZ, but we believe they could be beneficial. For example, eliminating neurotoxic microglia could avert neurodegeneration and SCZ by sparing the healthy neuronal and glial cells (Table 2).

Table 2 Common natural and synthetic senolytic and senomorphic drugs that have been demonstrated to benefit both patients with schizophrenia and viral infections.

In the past, it was believed that cellular senescence was irreversible. However, newer studies have found that inhibiting 3-phosphoinositide-dependent protein kinase-1 (PDK1) can reverse this phenotype [104]. For this reason, PDK1 inhibitors will likely play a significant role in future antipsychotic treatments.

6.2 Barrier Enhancers

Premature cellular senescence in SCZ affects most cell types, including those comprising the biological barriers. The senescent phenotype disrupts the TJs, enabling the translocation of gut bacteria into the host circulation. The GI tract barrier is comprised of epithelial and endothelial cells. IECs can produce OXT in the presence of the gastric hormone secretin. Earlier studies have associated secretin with symptomatic improvement of refractory SCZ, suggesting an indirect action via OXT [88]. Along this line, a recent study found that gut microbes influence OXT levels, implicating commensal flora, including Limosilactobacillus reuterii, in the biosynthesis of this hormone [105,106]. For this reason, probiotics with L. reuterii should be supplemented for SCZ patients. In addition, previous studies found that Bifidobacterium breve A-1 ameliorated depression, anxiety, and poor cognition, suggesting that it could benefit SCZ in combination with L. reuterii. Furthermore, as both microbes likely upregulate IL-22, “the guardian of the gut barrier,” they likely avert microbial translocation [107].

During the 1980s HIV epidemic, an IL-22 deficit was linked to increased intestinal permeability and HIV-induced translocation of gut microbes into host tissues. This occurred as the virus targeted innate lymphoid cell type 3 (ILC-3), the producers of IL-22.

Depletion of IL-22 led to microbial migration outside the gut lumen. For this reason, we have proposed human recombinant IL-22 as a novel SCZ treatment [108]. Other interventions for enhancing the gut barrier include serine proteases, such as nafamostat mesylate and campostat mesylate, which are known for decreasing paracellular spaces, thus lowering translocation [109]. Indigo and indirubin, Chinese herbal medicines, are enhancers of the gut barrier and have beneficial effects in IBD and probably SCZ.

6.3 OSU 03012 and Other PDK1 Inhibitors

Novel studies have demonstrated that aging cells can be “rejuvenated” via phosphatidylinositol-dependent kinase 1 (PDK1) inhibitors, including the BBB-crossing agent, OSU-03012 [110] (Figure 4). Aside from PDK1 inhibitors, inhibiting the downstream protein kinase B (Akt)/glycogen synthase kinase 3β (GSK3β) pathway may accomplish the same effect [111]. Indeed, many drugs, including some antipsychotics and lithium, inhibit GSK3β, suggesting that natural compounds exerting this effect may be beneficial against affective and psychotic disorders. For example, kaempferol, a polyphenol, and berberine, along with membrane lipid replacement (MLR), may exhibit antipsychotic properties [112,113].

Click to view original image

Figure 4 In the absence of OXT, senescent IECs enable microbial translocation outside the GI tract. Many viruses, including SARS-CoV-2, exploit OXT and promote VIS (virus-induced senescence). VIS, like SCZ, has been associated with GMC reduction and subsequent cognitive sequelae.

PDK1 inhibitors, including alpha-lipoic acid N acetylcysteine (NAC), have been found to lower lipid peroxidation and are currently being evaluated as SCZ treatments [114,115,116,117] (NCT03788759).

The aging brain retains iron, which can enhance the oxidation of the membrane lipid bilayer. This pathology could be ameliorated or reversed by membrane lipid replacement (MLR) [118,119,120]. Interestingly, aside from signaling with its receptors, DA also binds directly to membrane lipids, triggering peroxidation via 6-hydroxydopamine (6-OHDA), an established neurotoxin [121,122].

We propose combining MLR with OSU 03012, a synthetic PDK1 inhibitor, a combination discussed in other articles that will not be repeated here [123] (Figure 5). Initially described by Professor Garth Nicolson, who studied Gulf War Illness since early 1990s, MLR was expanded to fatiguing disorders and SCZ, a condition marked by extensive oxidation of membrane lipids.

Click to view original image

Figure 5 When the growth factor binds to its receptor, PDK-1 is recruited to the cell membrane and activated by PI3K through PIP1 and PIP2. MLR, particularly the glycerophospholipid phosphatidylserine (PS), inhibits PDK1 by maintaining its inactive conformation. Consequently, Akt phosphorylation is hindered, allowing GSK-3β to be released from inhibition. Some second-generation antipsychotic drugs upregulate GSK-3β by increasing its phosphorylation. OSU-03012 works synergistically with MLR to inhibit PDK1.

Interestingly, OXT inhibits the PI3K/AKT pathway, including PDK1, thus acting synergistically with PDK1 inhibitors. This suggests that combining this hormone with MLR could bring more benefit in SCZ than each component alone [124].

7. Conclusions

Synthesized in the CNS and the gut, OXT is far from being only the “hormone of love” and prosocial behaviors. In the GI tract, OXT maintains the homeostasis of the gut barrier, preventing microbial translocation that likely improves the SCZ outcome.

Interventions to upregulate OXT via AhR and PDK-1 inhibitors represent new treatment strategies applicable to SCZ and IBD. A particular intervention, combining OXT, MLR, OSU-03012, and human recombinant IL-22, may be beneficial by maintaining gut barrier homeostasis and eliminating already translocated microbes.

Author Contributions

Adonis Sfera: conceptualization and writing. Sabine Hazan: data curation. Jacob Anton: review, editing. Ioana Ciuperca: editing, supervision. Carolina Klein: editing, writing, and supervision. Karina G. Thomas: reviewing data.

Competing Interests

The authors have declared that no competing interests exist.

References

  1. Simanjuntak Y, Liang JJ, Lee YL, Lin YL. Japanese encephalitis virus exploits dopamine D2 receptor-phospholipase C to target dopaminergic human neuronal cells. Front Microbiol. 2017; 8: 651. [CrossRef] [Google scholar] [PubMed]
  2. Lu X, Liu Y, Zhang J, Wu X, Li X. Oxytocin increases pregnancy rates after fixed time artificial insemination in Kazak ewes. Reprod Domest Anim. 2021; 56: 942-947. [CrossRef] [Google scholar] [PubMed]
  3. Okumura T, Nozu T, Ishioh M, Igarashi S, Funayama T, Kumei S, et al. Oxytocin acts centrally in the brain to improve leaky gut through the vagus nerve and a cannabinoid signaling in rats. Physiol Behav. 2022; 254: 113914. [CrossRef] [Google scholar] [PubMed]
  4. Liu Y, Wu B, Wang X, Li W, Zhang T, Wu X, et al. Oxytocin effects on self-referential processing: Behavioral and neuroimaging evidence. Soc Cogn Affect Neurosci. 2017; 12: 1845-1858. [CrossRef] [Google scholar] [PubMed]
  5. Wang SC, Zhang F, Zhu H, Yang H, Liu Y, Wang P, et al. Potential of endogenous oxytocin in endocrine treatment and prevention of COVID-19. Front Endocrinol. 2022; 13: 799521. [CrossRef] [Google scholar] [PubMed]
  6. Wang C, Zhang T, He L, Fu JY, Deng HX, Xue XL, et al. Bacterial translocation associates with aggression in schizophrenia inpatients. Front Syst Neurosci. 2021; 15: 704069. [CrossRef] [Google scholar] [PubMed]
  7. Winkler-Dworak M, Zeman K, Sobotka T. Birth rate decline in the later phase of the COVID-19 pandemic: The role of policy interventions, vaccination programmes, and economic uncertainty. Hum Reprod Open. 2024; 2024: hoae052. [CrossRef] [Google scholar] [PubMed]
  8. Rahman A, Russell M, Zheng W, Eckrich D, Ahmed I, N3C Consortium. SARS-CoV-2 infection is associated with an increase in new diagnoses of schizophrenia spectrum and psychotic disorder: A study using the US national COVID cohort collaborative (N3C). PLoS One. 2024; 19: e0295891. [CrossRef] [Google scholar]
  9. Giron LB, Dweep H, Yin X, Wang H, Damra M, Goldman AR, et al. Plasma markers of disrupted gut permeability in severe COVID-19 patients. Front Immunol. 2021; 12: 686240. [CrossRef] [Google scholar] [PubMed]
  10. Mawson AR, Croft AM. Rubella virus infection, the congenital rubella syndrome, and the link to autism. Int J Environ Res Public Health. 2019; 16: 3543. [CrossRef] [Google scholar]
  11. Leboyer M, Tamouza R, Charron D, Faucard R, Perron H. Human endogenous retrovirus type W (HERV-W) in schizophrenia: A new avenue of research at the gene-environment interface. World J Biol Psychiatry. 2013; 14: 80-90. [CrossRef] [Google scholar] [PubMed]
  12. O’Donnell MP, Fox BW, Chao PH, Schroeder FC, Sengupta P. A neurotransmitter produced by gut bacteria modulates host sensory behaviour. Nature. 2020; 583: 415-420. [CrossRef] [Google scholar] [PubMed]
  13. Clarke G, Stilling RM, Kennedy PJ, Stanton C, Cryan JF, Dinan TG. Minireview: Gut microbiota: The neglected endocrine organ. Mol Endocrinol. 2014; 28: 1221-1238. [CrossRef] [Google scholar] [PubMed]
  14. Brown JM, Hazen SL. The gut microbial endocrine organ: Bacterially derived signals driving cardiometabolic diseases. Annu Rev Med. 2015; 66: 343-359. [CrossRef] [Google scholar] [PubMed]
  15. Mu Q, Tavella VJ, Luo XM. Role of Lactobacillus reuteri in human health and diseases. Front Microbiol. 2018; 9: 757. [CrossRef] [Google scholar] [PubMed]
  16. Du T, Lei A, Zhang N, Zhu C. The beneficial role of probiotic Lactobacillus in respiratory diseases. Front Immunol. 2022; 13: 908010. [CrossRef] [Google scholar] [PubMed]
  17. Danhof HA, Lee J, Thapa A, Britton RA, Di Rienzi SC. Microbial stimulation of oxytocin release from the intestinal epithelium via secretin signaling. Gut Microbes. 2023; 15: 2256043. [CrossRef] [Google scholar] [PubMed]
  18. Sheitman BB, Knable MB, Jarskog LF, Chakos M, Boyce LH, Early J, et al. Secretin for refractory schizophrenia. Schizophr Res. 2004; 66: 177-181. [CrossRef] [Google scholar] [PubMed]
  19. Gimpl G, Fahrenholz F. The oxytocin receptor system: Structure, function, and regulation. Physiol Rev. 2001; 81: 629-683. [CrossRef] [Google scholar] [PubMed]
  20. Caria A. Hypothalamus, neuropeptides and socioemotional behavior. Brain Sci. 2023; 13: 1303. [CrossRef] [Google scholar] [PubMed]
  21. Mei L, Osakada T, Lin D. Hypothalamic control of innate social behaviors. Science. 2023; 382: 399-404. [CrossRef] [Google scholar] [PubMed]
  22. Li XH, Matsuura T, Xue M, Chen QY, Liu RH, Lu JS, et al. Oxytocin in the anterior cingulate cortex attenuates neuropathic pain and emotional anxiety by inhibiting presynaptic long-term potentiation. Cell Rep. 2021; 36: 109411. [CrossRef] [Google scholar] [PubMed]
  23. Allman JM, Tetreault NA, Hakeem AY, Manaye KF, Semendeferi K, Erwin JM, et al. The von economo neurons in the frontoinsular and anterior cingulate cortex. Ann N Y Acad Sci. 2011; 1225: 59-71. [CrossRef] [Google scholar] [PubMed]
  24. Sridharan D, Levitin DJ, Menon V. A critical role for the right fronto-insular cortex in switching between central-executive and default-mode networks. Proc Natl Acad Sci USA. 2008; 105: 12569-12574. [CrossRef] [Google scholar] [PubMed]
  25. Ishioh M, Nozu T, Okumura T. Brain neuropeptides, neuroinflammation, and irritable bowel syndrome. Digestion. 2024; 105: 34-39. [CrossRef] [Google scholar] [PubMed]
  26. Chen F, Ohashi N, Li W, Eckman C, Nguyen JH. Disruptions of occludin and claudin‐5 in brain endothelial cells in vitro and in brains of mice with acute liver failure. Hepatology. 2009; 50: 1914-1923. [CrossRef] [Google scholar] [PubMed]
  27. Seoane R, Vidal S, Bouzaher YH, El Motiam A, Rivas C. The interaction of viruses with the cellular senescence response. Biology. 2020; 9: 455. [CrossRef] [Google scholar] [PubMed]
  28. Papanastasiou E, Gaughran F, Smith S. Schizophrenia as segmental progeria. J R Soc Med. 2011; 104: 475-484. [CrossRef] [Google scholar] [PubMed]
  29. Stevenson JR, McMahon EK, Boner W, Haussmann MF. Oxytocin administration prevents cellular aging caused by social isolation. Psychoneuroendocrinology. 2019; 103: 52-60. [CrossRef] [Google scholar] [PubMed]
  30. Greene C, Hanley N, Campbell M. Claudin-5: gatekeeper of neurological function. Fluids Barriers CNS. 2019; 16: 3. [CrossRef] [Google scholar] [PubMed]
  31. Momenabadi S, Vafaei AA, Zahedi Khorasani M, Vakili A. Pre-ischemic oxytocin treatment alleviated neuronal injury via suppressing NF-κB, MMP-9, and apoptosis regulator proteins in a mice model of stroke. Cell J. 2022; 24: 337-345. [Google scholar]
  32. Yan L, Sun X, Wang Z, Song M, Zhang Z. Regulation of social behaviors by p-Stat3 via oxytocin and its receptor in the nucleus accumbens of male Brandt's voles (Lasiopodomys brandtii). Horm Behav. 2020; 119: 104638. [CrossRef] [Google scholar] [PubMed]
  33. Sfera A, Imran H, Sfera DO, Anton JJ, Kozlakidis Z, Hazan S. Novel insights into psychosis and antipsychotic interventions: From managing symptoms to improving outcomes. Int J Mol Sci. 2024; 25: 5904. [CrossRef] [Google scholar] [PubMed]
  34. Zhang Y, Catts VS, Sheedy D, McCrossin T, Kril JJ, Shannon Weickert C. Cortical grey matter volume reduction in people with schizophrenia is associated with neuro-inflammation. Transl Psychiatry. 2016; 6: e982. [CrossRef] [Google scholar] [PubMed]
  35. Feng Y, Shen J, He J, Lu M. Schizophrenia and cell senescence candidate genes screening, machine learning, diagnostic models, and drug prediction. Front Psychiatry. 2023; 14: 1105987. [CrossRef] [Google scholar] [PubMed]
  36. Caspi A, Shireby G, Mill J, Moffitt TE, Sugden K, Hannon E. Accelerated pace of aging in schizophrenia: Five case-control studies. Biol Psychiatry. 2024; 95: 1038-1047. [CrossRef] [Google scholar] [PubMed]
  37. Nguyen TT, Eyler LT, Jeste DV. Systemic biomarkers of accelerated aging in schizophrenia: A critical review and future directions. Schizophr Bull. 2018; 44: 398-408. [CrossRef] [Google scholar] [PubMed]
  38. Denholm M, Rintoul RC, Muñoz-Espín D. SARS-CoV-2-induced senescence as a potential therapeutic target. Eur Respir J. 2022; 60: 2201101. [CrossRef] [Google scholar] [PubMed]
  39. Chen P, Wang Y, Zhou B. Insights into targeting cellular senescence with senolytic therapy: The journey from preclinical trials to clinical practice. Mech Ageing Dev. 2024; 218: 111918. [CrossRef] [Google scholar] [PubMed]
  40. Evangelou K, Veroutis D, Paschalaki K, Foukas PG, Lagopati N, Dimitriou M, et al. Pulmonary infection by SARS-CoV-2 induces senescence accompanied by an inflammatory phenotype in severe COVID-19: Possible implications for viral mutagenesis. Eur Respir J. 2022; 60: 2102951. [CrossRef] [Google scholar] [PubMed]
  41. Cho SY, Kim AY, Kim J, Choi DH, Son ED, Shin DW. Oxytocin alleviates cellular senescence through oxytocin receptor‐mediated extracellular signal‐regulated kinase/Nrf2 signalling. Br J Dermatol. 2019; 181: 1216-1225. [CrossRef] [Google scholar] [PubMed]
  42. Benameur T, Panaro MA, Porro C. The antiaging role of oxytocin. Neural Regen Res. 2021; 16: 2413-2414. [CrossRef] [Google scholar] [PubMed]
  43. Dickerson F, Vaidya D, Liu Y, Yolken R. Levels of matrix metalloproteinase 9 are elevated in persons with schizophrenia or bipolar disorder: The role of modifiable factors. Biol Psychiatry Glob Open Sci. 2023; 3: 766-772. [CrossRef] [Google scholar] [PubMed]
  44. Ding L, Guo H, Zhang C, Jin H, Guo X, Li T. Elevated matrix metalloproteinase‑9 expression is associated with COVID‑19 severity: A meta‑analysis. Exp Ther Med. 2023; 26: 545. [CrossRef] [Google scholar] [PubMed]
  45. Momenabadi S, Vafaei AA, Khorasani MZ, Vakili A. Pre-ischemic oxytocin treatment alleviated neuronal injury via suppressing NF-κB, MMP-9, and apoptosis regulator proteins in a mice model of stroke. Cell J. 2022; 24: 337-345. [Google scholar]
  46. Chiu PS, Lai SC. Matrix metalloproteinase-9 leads to claudin-5 degradation via the NF-κB pathway in BALB/c mice with eosinophilic meningoencephalitis caused by Angiostrongylus cantonensis. PLoS One. 2013; 8: e53370. [CrossRef] [Google scholar] [PubMed]
  47. Krishnamoorthy S, Sylaja PN, Sreedharan SE, Singh G, Damayanthi D, Gopala S, et al. Biomarkers predict hemorrhagic transformation and stroke severity after acute ischemic stroke. J Stroke Cerebrovasc Dis. 2023; 32: 106875. [CrossRef] [Google scholar] [PubMed]
  48. Vaswani CM, Varkouhi AK, Gupta S, Ektesabi AM, Tsoporis JN, Yousef S, et al. Preventing occludin tight-junction disruption via inhibition of microRNA-193b-5p attenuates viral load and influenza-induced lung injury. Mol Therapy. 2023; 31: 2681-2701. [CrossRef] [Google scholar] [PubMed]
  49. Chen W, Man X, Zhang Y, Yao G, Chen J. Medial prefrontal cortex oxytocin mitigates epilepsy and cognitive impairments induced by traumatic brain injury through reducing neuroinflammation in mice. Sci Rep. 2023; 13: 5214. [CrossRef] [Google scholar] [PubMed]
  50. Greene C, Kealy J, Humphries MM, Gong Y, Hou J, Hudson N, et al. Dose-dependent expression of claudin-5 is a modifying factor in schizophrenia. Mol Psychiatry. 2018; 23: 2156-2166. [CrossRef] [Google scholar] [PubMed]
  51. Ju S, Shin Y, Han S, Kwon J, Choi TG, Kang I, et al. The gut-brain axis in schizophrenia: The implications of the gut microbiome and SCFA production. Nutrients. 2023; 15: 4391. [CrossRef] [Google scholar] [PubMed]
  52. Cornelissen FM, Markert G, Deutsch G, Antonara M, Faaij N, Bartelink I, et al. Explaining blood-brain barrier permeability of small molecules by integrated analysis of different transport mechanisms. J Med Chem. 2023; 66: 7253-7267. [CrossRef] [Google scholar] [PubMed]
  53. Spindler KR, Hsu TH. Viral disruption of the blood-brain barrier. Trends Microbiol. 2012; 20: 282-290. [CrossRef] [Google scholar] [PubMed]
  54. Tsounis EP, Triantos C, Konstantakis C, Marangos M, Assimakopoulos SF. Intestinal barrier dysfunction as a key driver of severe COVID-19. World J Virol. 2023; 12: 68-90. [CrossRef] [Google scholar] [PubMed]
  55. Erdman SE. Oxytocin and the microbiome. Curr Opin Endocr Metab Res. 2021; 19: 8-14. [CrossRef] [Google scholar]
  56. Tang Y, Shi Y, Gao Y, Xu X, Han T, Li J, et al. Oxytocin system alleviates intestinal inflammation by regulating macrophages polarization in experimental colitis. Clin Sci. 2019; 133: 1977-1992. [CrossRef] [Google scholar] [PubMed]
  57. Front Matter. Scientific American Mind. 2008; 19: 2. Available from: http://www.jstor.org/stable/24939820.
  58. Cardinale V, Capurso G, Ianiro G, Gasbarrini A, Arcidiacono PG, Alvaro D. Intestinal permeability changes with bacterial translocation as key events modulating systemic host immune response to SARS-CoV-2: A working hypothesis. Dig Liver Dis. 2020; 52: 1383-1389. [CrossRef] [Google scholar] [PubMed]
  59. Gunin AG. Oxytocin is produced by gastrointestinal epithelial cells. Eur J Obstet Gynecol Reprod Biol. 2024; 302: 381. [CrossRef] [Google scholar] [PubMed]
  60. Gur RE, Turetsky BI, Bilker WB, Gur RC. Reduced gray matter volume in schizophrenia. Arch Gen Psychiatry. 1999; 56: 905-911. [CrossRef] [Google scholar] [PubMed]
  61. Duggan MR, Peng Z, Sipilä PN, Lindbohm JV, Chen J, Lu Y, et al. Proteomics identifies potential immunological drivers of postinfection brain atrophy and cognitive decline. Nat Aging. 2024; 4: 1263-1278. [CrossRef] [Google scholar] [PubMed]
  62. Haaf R, Brandi ML, Albantakis L, Lahnakoski JM, Henco L, Schilbach L. Peripheral oxytocin levels are linked to hypothalamic gray matter volume in autistic adults: A cross-sectional secondary data analysis. Sci Rep. 2024; 14: 1380. [CrossRef] [Google scholar] [PubMed]
  63. Lewis DA, Lieberman JA. Catching up on schizophrenia: Natural history and neurobiology. Neuron. 2000; 28: 325-334. [CrossRef] [Google scholar] [PubMed]
  64. Hazlett EA, Buchsbaum MS, Haznedar MM, Newmark R, Goldstein KE, Zelmanova Y, et al. Cortical gray and white matter volume in unmedicated schizotypal and schizophrenia patients. Schizophr Res. 2008; 101: 111-123. [CrossRef] [Google scholar] [PubMed]
  65. Ho BC, Andreasen NC, Ziebell S, Pierson R, Magnotta V. Long-term antipsychotic treatment and brain volumes: A longitudinal study of first-episode schizophrenia. Arch Gen Psychiatry. 2011; 68: 128-137. [CrossRef] [Google scholar] [PubMed]
  66. Momenabadi S, Vafaei AA, Bandegi AR, Zahedi-Khorasani M, Mazaheri Z, Vakili A. Oxytocin reduces brain injury and maintains blood-brain barrier integrity after ischemic stroke in mice. Neuromolecular Med. 2020; 22: 557-571. [CrossRef] [Google scholar] [PubMed]
  67. Saffari M, Momenabadi S, Vafaei AA, Vakili A, Zahedi-Khorasani M. Prophylactic effect of intranasal oxytocin on brain damage and neurological disorders in global cerebral ischemia in mice. Iran J Basic Med Sci. 2021; 24: 79-84. [Google scholar]
  68. Holm M, Taipale H, Tanskanen A, Tiihonen J, Mitterdorfer‐Rutz E. Employment among people with schizophrenia or bipolar disorder: A population‐based study using nationwide registers. Acta Psychiatr Scand. 2021; 143: 61-71. [CrossRef] [Google scholar] [PubMed]
  69. Lévesque IS, Abdel-Baki A. Homeless youth with first-episode psychosis: A 2-year outcome study. Schizophr Res. 2020; 216: 460-469. [CrossRef] [Google scholar] [PubMed]
  70. Davidson L, Schmutte T, Dinzeo T, Andres-Hyman R. Remission and recovery in schizophrenia: Practitioner and patient perspectives. Schizophr Bull. 2008; 34: 5-8. [CrossRef] [Google scholar] [PubMed]
  71. Stone WS, Phillips MR, Yang LH, Kegeles LS, Susser ES, Lieberman JA. Neurodegenerative model of schizophrenia: Growing evidence to support a revisit. Schizophr Res. 2022; 243: 154-162. [CrossRef] [Google scholar] [PubMed]
  72. Ng PY, McNeely TL, Baker DJ. Untangling senescent and damage‐associated microglia in the aging and diseased brain. FEBS J. 2023; 290: 1326-1339. [CrossRef] [Google scholar] [PubMed]
  73. Wang XL, Li L. Microglia regulate neuronal circuits in homeostatic and high-fat diet-induced inflammatory conditions. Front Cell Neurosci. 2021; 15: 722028. [CrossRef] [Google scholar] [PubMed]
  74. Nguyen PT, Dorman LC, Pan S, Vainchtein ID, Han RT, Nakao-Inoue H, et al. Microglial remodeling of the extracellular matrix promotes synapse plasticity. Cell. 2020; 182: 388-403.e15. [CrossRef] [Google scholar] [PubMed]
  75. Vilalta A, Brown GC. Neurophagy, the phagocytosis of live neurons and synapses by glia, contributes to brain development and disease. FEBS J. 2018; 285: 3566-3575. [CrossRef] [Google scholar] [PubMed]
  76. Hansen DV, Hanson JE, Sheng M. Microglia in Alzheimer’s disease. J Cell Biol. 2018; 217: 459-472. [CrossRef] [Google scholar] [PubMed]
  77. Masaldan S, Clatworthy SA, Gamell C, Meggyesy PM, Rigopoulos AT, Haupt S, et al. Iron accumulation in senescent cells is coupled with impaired ferritinophagy and inhibition of ferroptosis. Redox Biol. 2018; 14: 100-115. [CrossRef] [Google scholar] [PubMed]
  78. Stockwell BR, Angeli JP, Bayir H, Bush AI, Conrad M, Dixon SJ, et al. Ferroptosis: A regulated cell death nexus linking metabolism, redox biology, and disease. Cell. 2017; 171: 273-285. [CrossRef] [Google scholar] [PubMed]
  79. Zhang YD, Zhou JS, Lu FM, Wang XP. Reduced gray matter volume in male adolescent violent offenders. PeerJ. 2019; 7: e7349. [CrossRef] [Google scholar] [PubMed]
  80. Greenwood EK, Brown DR. Senescent microglia: The key to the ageing brain? Int J Mol Sci. 2021; 22: 4402. [CrossRef] [Google scholar] [PubMed]
  81. Donley DW, Realing M, Gigley JP, Fox JH. Iron activates microglia and directly stimulates indoleamine-2,3-dioxygenase activity in the N171-82Q mouse model of Huntington’s disease. PLoS One. 2021; 16: e0250606. [CrossRef] [Google scholar] [PubMed]
  82. Hagihara H, Shoji H, Otabi H, Toyoda A, Katoh K, Namihira M, et al. Protein lactylation induced by neural excitation. Cell Rep. 2021; 37: 109820. [CrossRef] [Google scholar] [PubMed]
  83. Zhang D, Tang Z, Huang H, Zhou G, Cui C, Weng Y, et al. Metabolic regulation of gene expression by histone lactylation. Nature. 2019; 574: 575-580. [CrossRef] [Google scholar] [PubMed]
  84. Anderson GJ, Frazer DM. Lactate as a regulator of iron homeostasis. Life Metab. 2023; 2: load033. [CrossRef] [Google scholar] [PubMed]
  85. Galle E, Wong CW, Ghosh A, Desgeorges T, Melrose K, Hinte LC, et al. H3K18 lactylation marks tissue-specific active enhancers. Genome Biol. 2022; 23: 207. [CrossRef] [Google scholar] [PubMed]
  86. Modica CM, Zivadinov R, Dwyer MG, Bergsland N, Weeks AR, Benedict RH. Iron and volume in the deep gray matter: Association with cognitive impairment in multiple sclerosis. Am J Neuroradiol. 2015; 36: 57-62. [CrossRef] [Google scholar] [PubMed]
  87. Sfera A, Bullock K, Price A, Inderias L, Osorio C. Ferrosenescence: The iron age of neurodegeneration? Mech Ageing Dev. 2018; 174: 63-75. [CrossRef] [Google scholar] [PubMed]
  88. Lu J, Zheng X, Li F, Yu Y, Chen Z, Liu Z, et al. Tunneling nanotubes promote intercellular mitochondria transfer followed by increased invasiveness in bladder cancer cells. Oncotarget. 2017; 8: 15539-15552. [CrossRef] [Google scholar] [PubMed]
  89. Wang J, Wang Z, Wang Q, Li X, Guo Y. Ubiquitous protein lactylation in health and diseases. Cell Mol Biol Lett. 2024; 29: 23. [CrossRef] [Google scholar] [PubMed]
  90. Hood MI, Skaar EP. Nutritional immunity: Transition metals at the pathogen-host interface. Nat Rev Microbiol. 2012; 10: 525-537. [CrossRef] [Google scholar] [PubMed]
  91. Hennigar SR, McClung JP. Nutritional immunity: Starving pathogens of trace minerals. Am J Lifestyle Med. 2016; 10: 170-173. [CrossRef] [Google scholar] [PubMed]
  92. Ghorbani M. Unveiling the human brain virome in Brodmann area 46: Novel insights into dysbiosis and its association with schizophrenia. Schizophr Bull Open. 2023; 4: sgad029. [CrossRef] [Google scholar] [PubMed]
  93. Maes M, Vojdani A, Geffard M, Moreira EG, Barbosa DS, Michelin AP, et al. Schizophrenia phenomenology comprises a bifactorial general severity and a single-group factor, which are differently associated with neurotoxic immune and immune-regulatory pathways. Biomol Concepts. 2019; 10: 209-225. [CrossRef] [Google scholar] [PubMed]
  94. Żak K, Starek E, Korga-Plewko A, Rasoul-Pelińska K, Abramiuk M, Michalczuk M, et al. Assessment of the impact of SARS-CoV-2 infection on the sexual function of women, levels of oxytocin and prolactin: A prospective cohort study. J Clin Med. 2024; 13: 2230. [CrossRef] [Google scholar] [PubMed]
  95. Hsieh TC, Edwards NC, Bhattacharyya SK, Nitschelm KD, Burnett AL. The epidemic of COVID-19-related erectile dysfunction: A scoping review and health care perspective. Sex Med Rev. 2022; 10: 286-310. [CrossRef] [Google scholar] [PubMed]
  96. Chen J, Liang J, Xu H, Liu W, Liu S, Duan L, et al. Targeting aryl hydrocarbon receptor signaling enhances type I interferon-independent resistance to herpes simplex virus. Microbiol Spectr. 2021; 9: e00473-21. [CrossRef] [Google scholar] [PubMed]
  97. Ren JI, Lu Y, Qian Y, Chen B, Wu TA, Ji G. Recent progress regarding kaempferol for the treatment of various diseases. Exp Ther Med. 2019; 18: 2759-2776. [CrossRef] [Google scholar] [PubMed]
  98. Xia QS, Wu F, Wu WB, Dong H, Huang ZY, Xu L, et al. Berberine reduces hepatic ceramide levels to improve insulin resistance in HFD-fed mice by inhibiting HIF-2α. Biomed Pharmacother. 2022; 150: 112955. [CrossRef] [Google scholar] [PubMed]
  99. Imran M, Ghorat F, Ul-Haq I, Ur-Rehman H, Aslam F, Heydari M, et al. Lycopene as a natural antioxidant used to prevent human health disorders. Antioxidants. 2020; 9: 706. [CrossRef] [Google scholar] [PubMed]
  100. Kim TW. Fisetin, an anti-inflammatory agent, overcomes radioresistance by activating the PERK-ATF4-CHOP axis in liver cancer. Int J Mol Sci. 2023; 24: 9076. [CrossRef] [Google scholar] [PubMed]
  101. Lamming DW. Inhibition of the mechanistic target of rapamycin (mTOR)-rapamycin and beyond. Cold Spring Harb Perspect Med. 2016; 6: a025924. [CrossRef] [Google scholar] [PubMed]
  102. Janić M, Lunder M, Cerkovnik P, Prosenc Zmrzljak U, Novaković S, Šabovič M. Low-dose fluvastatin and valsartan rejuvenate the arterial wall through telomerase activity increase in middle-aged men. Rejuvenation Res. 2016; 19: 115-119. [CrossRef] [Google scholar] [PubMed]
  103. Kuk MU, Kim JW, Lee YS, Cho KA, Park JT, Park SC. Alleviation of senescence via ATM inhibition in accelerated aging models. Mol Cells. 2019; 42: 210-217. [Google scholar]
  104. An S, Cho SY, Kang J, Lee S, Kim HS, Min DJ, et al. Inhibition of 3-phosphoinositide-dependent protein kinase 1 (PDK1) can revert cellular senescence in human dermal fibroblasts. Proc Natl Acad Sci. 2020; 117: 31535-31546. [CrossRef] [Google scholar] [PubMed]
  105. Bolbecker AR, Hetrick WP, Johannesen JK, O'Donnell BF, Steinmetz JE, Shekhar AS. Secretin effects on cerebellar-dependent motor learning in schizophrenia. Am J Psychiatry. 2009; 166: 460-466. [CrossRef] [Google scholar] [PubMed]
  106. Wu Y, Zhang X, Liu X, Zhao Z, Tao S, Xu Q, et al. Galactooligosaccharides and Limosilactobacillus reuteri synergistically alleviate gut inflammation and barrier dysfunction by enriching Bacteroides acidifaciens for pentadecanoic acid biosynthesis. Nat Commun. 2024; 15: 9291. [CrossRef] [Google scholar] [PubMed]
  107. Okubo R, Koga M, Katsumata N, Odamaki T, Matsuyama S, Oka M, et al. Effect of bifidobacterium breve A-1 on anxiety and depressive symptoms in schizophrenia: A proof-of-concept study. J Affect Disord. 2019; 245: 377-385. [CrossRef] [Google scholar] [PubMed]
  108. Sfera A, Thomas KA, Anton J. Cytokines and madness: A unifying hypothesis of schizophrenia involving interleukin-22. Int J Mol Sci. 2024; 25: 12110. [CrossRef] [Google scholar] [PubMed]
  109. Van Spaendonk H, Ceuleers H, Witters L, Patteet E, Joossens J, Augustyns K, et al. Regulation of intestinal permeability: The role of proteases. World J Gastroenterol. 2017; 23: 2106-2123. [CrossRef] [Google scholar] [PubMed]
  110. Nishijima I, Yamagata T, Spencer CM, Weeber EJ, Alekseyenko O, Sweatt JD, et al. Secretin receptor-deficient mice exhibit impaired synaptic plasticity and social behavior. Hum Mol Genet. 2006; 15: 3241-3250. [CrossRef] [Google scholar] [PubMed]
  111. Lawson EA. Understanding oxytocin in human physiology and pathophysiology: A path towards therapeutics. Compr Psychoneuroendocrinol. 2024; 19: 100242. [CrossRef] [Google scholar] [PubMed]
  112. Mitchell JW, Atkins N Jr, Sweedler JV, Gillette MU. Direct cellular peptidomics of hypothalamic neurons. Front Neuroendocrinol. 2011; 32: 377-386. [CrossRef] [Google scholar] [PubMed]
  113. Kim DW, Yao Z, Graybuck LT, Kim TK, Nguyen TN, Smith KA, et al. Multimodal analysis of cell types in a hypothalamic node controlling social behavior. Cell. 2019; 179: 713-728. [CrossRef] [Google scholar] [PubMed]
  114. Yue L, Ren Y, Yue Q, Ding Z, Wang K, Zheng T, et al. α‐lipoic acid targeting PDK1/NRF2 axis contributes to the apoptosis effect of lung cancer cells. Oxid Med Cell Longev. 2021; 2021: 6633419. [CrossRef] [Google scholar] [PubMed]
  115. Rossell SL, Francis PS, Galletly C, Harris A, Siskind D, Berk M, et al. N-acetylcysteine (NAC) in schizophrenia resistant to clozapine: A double blind randomised placebo controlled trial targeting negative symptoms. BMC Psychiatry. 2016; 16: 320. [CrossRef] [Google scholar] [PubMed]
  116. Sanders LL, de Souza Menezes CE, Chaves Filho AJ, de Almeida Viana G, Fechine FV, de Queiroz MG, et al. α-Lipoic acid as adjunctive treatment for schizophrenia: An open-label trial. J Clin Psychopharmacol. 2017; 37: 697-701. [CrossRef] [Google scholar] [PubMed]
  117. Spellmann I, Rujescu D, Musil R, Giegling I, Genius J, Zill P, et al. Pleckstrin homology domain containing 6 protein (PLEKHA6) polymorphisms are associated with psychopathology and response to treatment in schizophrenic patients. Prog Neuropsychopharmacol Biol Psychiatry. 2014; 51: 190-195. [CrossRef] [Google scholar] [PubMed]
  118. Feng L, Wong JC, Mahendran R, Chan ES, Spencer MD. Intranasal oxytocin for autism spectrum disorders (ASD). Cochrane Database Syst Rev. 2017; 2017: CD010928. [CrossRef] [Google scholar]
  119. Nicolson GL, Ash ME. Membrane lipid replacement for chronic illnesses, aging and cancer using oral glycerolphospholipid formulations with fructooligosaccharides to restore phospholipid function in cellular membranes, organelles, cells and tissues. Biochim Biophys Acta Biomembr. 2017; 1859: 1704-1724. [CrossRef] [Google scholar] [PubMed]
  120. Repetto M, Semprine J, Boveris A. Lipid peroxidation: Chemical mechanism, biological implications and analytical. Lipid peroxidation. London, UK: IntechOpen; 2012. [CrossRef] [Google scholar]
  121. Sato T, Shapiro JS, Chang HC, Miller RA, Ardehali H. Aging is associated with increased brain iron through cortex-derived hepcidin expression. Elife. 2022; 11: e73456. [CrossRef] [Google scholar] [PubMed]
  122. Lolicato F, Juhola H, Zak A, Postila PA, Saukko A, Rissanen S, et al. Membrane-dependent binding and entry mechanism of dopamine into its receptor. ACS Chem Neurosci. 2020; 11: 1914-1924. [CrossRef] [Google scholar] [PubMed]
  123. Del Campo CM, Nicolson GL, Sfera A. Neurolipidomics in schizophrenia: A not so well-oiled machine. Neuropharmacology. 2024; 260: 110117. [CrossRef] [Google scholar] [PubMed]
  124. Yang Y, Wang Z, Yao M, Xiong W, Wang J, Fang Y, et al. Oxytocin protects against isoproterenol-induced cardiac hypertrophy by inhibiting PI3K/AKT pathway via a lncRNA GAS5/miR-375-3p/KLF4-dependent mechanism. Front Pharmacol. 2021; 12: 766024. [CrossRef] [Google scholar] [PubMed]
Journal Metrics
2023
CiteScore SJR SNIP
1.00.2320.256
Newsletter
Download PDF Download Citation
0 0

TOP