Skip to main content
Sage Choice logoLink to Sage Choice
. 2024 Aug 7;31(3):234–261. doi: 10.1177/10738584241268754

Navigating Central Oxytocin Transport: Known Realms and Uncharted Territories

Deniz Parmaksiz 1,2, Yongsoo Kim 1,2,
PMCID: PMC12103645  PMID: 39113465

Abstract

Complex mechanisms govern the transport and action of oxytocin (Oxt), a neuropeptide and hormone that mediates diverse physiologic processes. While Oxt exerts site-specific and rapid effects in the brain via axonal and somatodendritic release, volume transmission via CSF and the neurovascular interface can act as an additional mechanism to distribute Oxt signals across distant brain regions on a slower timescale. This review focuses on modes of Oxt transport and action in the CNS, with particular emphasis on the roles of perivascular spaces, the blood-brain barrier (BBB), and circumventricular organs in coordinating the triadic interaction among circulating blood, CSF, and parenchyma. Perivascular spaces, critical conduits for CSF flow, play a pivotal role in Oxt diffusion and distribution within the CNS and reciprocally undergo Oxt-mediated structural and functional reconstruction. While the BBB modulates the movement of Oxt between systemic and cerebral circulation in a majority of brain regions, circumventricular organs without a functional BBB can allow for diffusion, monitoring, and feedback regulation of bloodborne peripheral signals such as Oxt. Recognition of these additional transport mechanisms provides enhanced insight into the systemic propagation and regulation of Oxt activity.

Keywords: oxytocin, transport mechanism, volume transmission, axonal release, somatodendritic release, cerebrospinal fluid, neurovasculature, perivascular space, blood-brain barrier, circumventricular organs, neurohypophyseal system, hypothalamus, neuroendocrinology

Introduction

Oxytocin (Oxt), a highly conserved neuropeptide and hormone, is widely recognized for its pivotal roles in mammalian reproduction and prosocial behaviors (Box 1). Within the CNS, Oxt is predominantly synthesized in the hypothalamus, notably the supraoptic nucleus (SON) and paraventricular (PVH) regions, alongside several accessory nuclei (Grinevich and others 2016; Jurek and Neumann 2018; Son and others 2022). Hypothalamic Oxt is supplemented by Oxt locally produced in various peripheral sites across the reproductive, cardiovascular, gastrointestinal, immune, and musculoskeletal systems of both sexes (Assinder 2022; Figure 1).

Box 1.

Oxytocin.

In 1906, Sir Henry Dale noted that extracts from the human posterior pituitary caused uterine contractions, leading to the discovery of oxytocin (Oxt), a highly conserved nonapeptide hormone derived from the larger precursor protein neurophysin (Brownstein and others 1980; Dale 1906). Often referred to as the “love hormone” or “bonding hormone” due to its prosocial effects, Oxt’s influence extends across a spectrum of behavioral, cognitive, and physiologic processes, encompassing parturition, lactation, pair bonding, parental behaviors, sociability, anxiety regulation, appetite control, and body fluid homeostasis (Gimpl and Fahrenholz 2001; Jurek and Neumann 2018; Quintana and Guastella 2020; Son and others 2022). These effects are mediated by the Oxt receptor, a G protein–coupled receptor ubiquitously expressed throughout the brain and body (Jurek and Neumann 2018). Disruptions in Oxt signaling have been linked to several conditions, ranging from autism spectrum disorders to preeclampsia, prompting research into the therapeutic potential of exogenous Oxt treatment (Ghazy and others 2023; D. A. Martins and others 2020; Martins and others 2022).

Figure 1.

Figure 1.

Oxytocin and oxytocin receptor expression in the body. For information on expression in each peripheral site, refer to Supplementary Table S1. Oxt = oxytocin; Oxtr = oxytocin receptor. Created with BioRender.com.

In the CNS, Oxt acts as a neuromodulator through two complementary mechanisms. Somatodendritic Oxt release mediates localized effects in the hypothalamus and neighboring brain areas, fine-tuning the sensitivity of the Oxt system, while axonal transmission extends Oxt signaling to remote brain regions (Grinevich and others 2016; Figure 2). Furthermore, synaptically and dendritically released Oxt can diffuse into the CSF, constituting a third mode of Oxt transport known as volume transmission (Box 2), through which Oxt functions as a hormone, eliciting widespread effects on a slower timescale (Veening and others 2010). Notably, as CSF traverses perivascular spaces, Oxt, known for its cardiovascular effects (Gutkowska and Jankowski 2012), intimately and reciprocally interacts with cerebral circulation (Alonso and others 2005; Miyata and Hatton 2002). Despite extensive research on Oxt transport in CSF, the neurovascular interface’s role in volume transmission and overall central Oxt function has been largely neglected. Additionally, significant efforts are needed to understand the interplay between central and peripheral Oxt systems and the blood-brain barrier’s (BBB) permeability to Oxt, particularly regarding exogenous Oxt treatments for neuropsychiatric disorders (D. A. Martins and others 2020; Valstad and others 2017; Yamamoto and others 2019). However, studies examining potential transport mechanisms often overlook circumventricular organs (CVOs), specialized structures devoid of a functional BBB, which are anatomically connected with oxytocinergic nuclei and share significant functional overlap with the Oxt system (Jeong and others 2021; Sisó and others 2010). This review explores the mechanisms governing Oxt transport and activity in the CNS, with a focus on the crucial roles of the neurovascular unit, emphasizing perivascular spaces, the BBB, and CVOs.

Figure 2.

Figure 2.

Established modes of central oxytocin transport and action. (a) Somatodendritic release. Oxytocin released from dendrites of magnocellular and parvocellular neurons of hypothalamic nuclei engages in autocrine and paracrine signaling by activating oxytocin receptors on the neuron of origin and its neighbors. The remaining oxytocin diffuses from the parenchyma into the CSF. (b) En passant release. Oxytocin can be released extrasynaptically from undilated axons and axonal varicosities. Several axon collaterals of oxytocinergic neurons are strategically positioned to facilitate en passant release directly into the third ventricle or subarachnoid space, thereby contributing to volume transmission of oxytocin in CSF. (c) Synaptic release. Axons of parvocellular oxytocin neurons make synaptic contacts in various brain regions, including the isocortex, cortical subplate, hippocampal formation, pallidum, striatum, midbrain, brainstem, and spinal cord. In some species, cortical and subcortical projections of magnocellular neurons have also been reported. Upon release from synaptic terminals, oxytocin can bind to pre- and postsynaptic oxytocin receptors. Oxytocin is thought to mediate NMDAR-dependent plasticity via co-release with glutamate at specific synapses within the frontal cortex. Spillover from the synaptic cleft can also contribute to volume transmission via CSF. 3V = third ventricle; CCV = clear core vesicle; Hyp = hypothalamus; LDCV = large dense core vesicle; NMDAR = N-methyl-D-aspartate receptor; Oxt = oxytocin; OXTR = oxytocin receptor; PVH = paraventricular nucleus of the hypothalamus. Created with BioRender.com.

Box 2.

Volume Transmission.

Volume transmission refers to a mode of communication within the brain that involves the diffusion of signaling molecules through the extracellular fluid and CSF rather than traditional point-to-point synaptic transmission (Agnati and others 1986; Agnati and others 2010). Volume transmission encompasses various release mechanisms beyond direct delivery into the CSF, with contributions from somatodendritic release, en passant (or extrasynaptic) release from axonal collaterals, and spillover from synaptic transmission. Diffusion of neurotransmitters and peptides is mediated by flow along spaces surrounding blood vessels and myelinated fiber bundles, as influenced by many factors, including concentration and pressure gradients (Iliff and others 2012; Iliff and others 2013). Key diffusion parameters include 1) volume fraction of extracellular fluid and CSF as compared with total brain volume; 2) clearance rate of neurotransmitters; and 3) tortuosity, which describes increases in diffusional path length due to extracellular matrix components and glial processes (Agnati and others 1986; Agnati and others 2010). Unlike synaptic transmission, which is rapid, precise, and localized, volume transmission allows for more widespread and flexible modulation of neural circuits across an extended timescale (Zoli and others 1999). Because of its brainwide effects, volume transmission is thought to play a crucial role in modulating mood and arousal.

Axonal and Somatodendritic Transmission

Axons of Oxt magnocellular neurons (MCNs) residing in the PVH extend ventrally and intersect with supraoptic projections, establishing the hypothalamoposthypophyseal tract (HPH; Carey 1959; Currie and others 1960). These fibers terminate as Herring bodies in the posterior pituitary, where “burst firing” or synchronized high-frequency Oxt release into plasma elicits well-established physiologic responses, such as milk ejection and uterine contraction (Summerlee 1981; Wakerley and Lincoln 1973). Oxt, implicated in various bodily functions in both sexes, is continuously secreted into the bloodstream to sustain basal concentrations of 1 to 3 pg/mL by Oxt MCNs, firing at a rate of ~1 to 5 spikes per second (Brown 2016).

The traditional view held that MCN axons exclusively target the posterior pituitary and release Oxt somatodendritically (Figure 2a), while parvocellular neurons (PCNs) send out central projections to regulate Oxt’s behavioral effects (Swanson and Sawchenko 1980, 1983). This view has been challenged by the detection of MCN axon collaterals in the rat brain that extend to cortical areas (e.g., piriform and auditory cortices) and subcortical regions such as the central amygdala, caudate putamen, and nucleus accumbens (Knobloch and others 2012; Zhang and others 2021). However, recent investigations in mice report that PVH MCNs projecting to the pituitary do not extend axonal arbors to extrahypothalamic regions (Li and others 2024), possibly highlighting interspecies differences in morphologic patterns and connectivity of Oxt neurons. Conversely, projections of PCNs have been reported in the isocortex, cortical subplate, hippocampal formation, pallidum, striatum, midbrain, brainstem, and spinal cord (Althammer and Grinevich 2017; Hosoya and others 1991; Li and others 2024; Pyner and Coote 2000; Shafton and others 1998; Swanson and Sawchenko 1980, 1983; Xiao and others 2017), where Oxt action can regulate nociception (Eliava and others 2016; Iwasaki and others 2023), satiety (Blevins and others 2004; Uchoa and others 2009), and cardiovascular and autonomic processes (Mack and others 2002; Roy and others 2018; Xu and others 2012). PCNs are thought to not only receive and integrate sensory information but also modulate MCN activity somatodendritically (Eliava and others 2016), and they have been implicated in social behaviors (Loth and others 2014; Tang and others 2020; Wei and others 2021) and dysfunction thereof in animal models of autism spectrum disorder (Lewis and others 2020; Tsurutani and others 2023). Furthermore, continued research into morphoelectric characteristics of Oxt neuron populations and application of advanced methods, such as single-cell and nucleus RNA sequencing, have begun to shed light to the structural, functional, and genetic diversity of Oxt neurons, identifying several types of Oxt neurons beyond the canonical dichotomous categorization (Althammer and Grinevich 2017; Chen and others 2022; Xu and others 2020).

It remains uncertain whether MCNs make synaptic contacts outside the posterior pituitary (Li and others 2024), despite reports of such contacts in the central amygdala (Knobloch and others 2012; Morris and Pow 1991), which may serve a modulatory function via co-release of glutamate with Oxt (Figure 2c), as supported by expression of the glutamate transporter VGLUT2 (Hrabovszky and Liposits 2008). The remainder of Oxt from MCN axons is released extrasynaptically, acting on glutamatergic and GABAergic synapses that show abundant Oxt receptor (Oxtr) expression in pre- and postsynaptic terminals (Mitre and others 2016). This constitutes en passant release (Figure 2b), or exocytosis from varicosities and undilated axons, which may not only mediate social fear inhibition in the lateral septum but also contribute to volume transmission of Oxt via CSF (Chini and others 2017; Li and others 2024; Menon and others 2018), as elaborated in the next section.

Besides their known role in receiving and propagating synaptic inputs, dendrites are capable of synthesizing and releasing neurotransmitters (Gao 1998), which then exert autocrine and paracrine effects by acting on pre- and postsynaptic terminals (Ludwig and Leng 2006). This mechanism, known as somatodendritic release, accounts for a majority of Oxt output within the CNS (Figure 2a) and has been extensively studied and reviewed in the context of Oxt signaling (Brown and others 2020; Ludwig and Leng 2006; Pow and Morris 1989).

Unlike neurotransmitters, neuropeptides such as Oxt are stored in large dense-core vesicles (LDCVs) that do not require vesicular transport proteins (Li and Kim 2008) and can be released from all neuronal compartments, including dendrites (Morris and Pow 1991). Somatodendritic release of Oxt LDCVs is regulated by multiple mechanisms. As in synaptic release, action potential–mediated calcium influx through voltage-gated channels prompts exocytosis of LDCVs (Ludwig and Leng 2006). The activation of NMDARs (N-methyl-D-aspartate receptors), which contribute to calcium influx, is required for somatodendritic transmission at basal firing rates and enhances activity-driven release (Pitra and others 2019), presumably by increasing LDCV trafficking velocities (Kirchner and others 2023). Additionally, Oxt MCNs express Oxtrs (Freund-Mercier and others 1994), which, upon activation (e.g., in lactating females), prompt further Oxt release by de-sequestering intracellular calcium stores via dual actions of CD38-dependent second messengers cADPR and NAADP (Lopatina and others 2010; Salmina and others 2010) and the IP3 pathway (Dayanithi and others 2000). This priming process involves the transport of LDCVs to the readily releasable pool, as well as direct release from the reserve pool (Sedej and others 2009; Tobin and others 2004), accompanied by heightened density of N-type calcium channels, which are functionally implicated in dendritic exocytosis (Tobin and others 2011). Cytoskeletal network restructuring, particularly fluctuations in F-actin polymerization (Tobin and Ludwig 2007) and microtubule density (Hicks and others 2020), further modulate somatodendritic release under various physiologic conditions, such as salt loading.

Oxt action on its cell of origin and neighboring cells can have diverse effects. Under basal conditions, Oxtr binding suppresses excitatory postsynaptic currents and promotes endocannabinoid release, leading to retrograde inhibition of afferent terminals and reduced excitatory tone through negative feedback (Hirasawa and others 2001; Hirasawa and others 2004). However, under osmotic stimulation, endocannabinoid action can switch from inhibition to excitation via TRPV-1 channel activation (Sharif Naeini and others 2006). Likewise, GABAergic transmission becomes excitatory during pregnancy, as accompanied by augmented glutamatergic inputs onto MCNs (Lee and others 2015). These electrochemical adjustments and priming mechanisms can create a positive feedback loop for somatodendritic Oxt release under physiologic conditions such as pregnancy, lactation, and osmotic stimulation, which also trigger enhanced Oxt release into plasma from synaptic terminals in the posterior pituitary (Althammer and others 2021; Landgraf and others 1988). Yet, the two modes of release are not always synchronized (Neumann and others 2008; Valstad and others 2017). For example, axonal secretion into the posterior pituitary lags behind the somatodendritic response to suckling in lactating rats (Moos and others 1989), whereas upsurge in plasma Oxt levels precedes dendritic release during osmotic stimulation (Ludwig and others 1994). Furthermore, electrical decoupling between dendrites and other cellular compartments through regulatory mechanisms (i.e., NMDAR gating of dendritic release probability; Pitra and others 2019) and modulation of microgeometrical dendritic factors such as stem diameter, varicosities, and glial sheath wrapping size (Korogod and others 2023) allows liberation of dendritic intracellular calcium stores independent of action potentials. Additionally, α-melanocyte–stimulating hormone, originating from proopiomelanocortin neurons of the arcuate nucleus, promotes somatodendritic Oxt release under basal conditions by elevating intracellular calcium levels while lowering the probability of action potential generation and thus peripheral secretion (Sabatier and Leng 2006; Sabatier and others 2003).

The fine-tuned process of somatodendritic Oxt release in turn regulates the activity of other neurons, especially PCNs. Corticotropin-releasing hormone–secreting PCNs in the PVN are subject to attenuation of afferent glutamatergic inputs upon Oxt binding, potentially suppressing the hypothalamic-pituitary-adrenal axis and mitigating the stress response (Dabrowska and others 2011). Somatodendritic Oxt can also reach brain areas neighboring the PVN and SON that express Oxtrs such as the ventromedial hypothalamus and the medial nucleus of the amygdala, where it may mediate effects on satiety and social recognition, respectively (Klockars and others 2017; Manning and others 2012; Patisaul and others 2003; Takayanagi and others 2017).

Although the influence of somatodendritic Oxt in such regions adjacent to SON and PVN is well established, its ability to reach distant brain areas is still debated, as detailed in the next section.

Volume Transmission via CSF

Volume transmission of neuropeptides such as Oxt in CSF has long been recognized as a counterpart to axonal projections, which may exert lasting changes to neural function and behavior by operating across a broader spatial and temporal scale (Box 2; Agnati and others 1986; Agnati and others 2010). Oxt has been detected in the CSF of many species, including humans, primates, and rodents (Amico and others 1983; Artman and others 1982; Devarajan and Rusak 2004), with microdialysis studies confirming its effective diffusion into CSF and brain regions beyond the PVH and SON (Ebner and others 2000; Engelmann and others 2006). Yet, some argue that basal CSF Oxt levels fall significantly below the threshold needed to activate Oxtrs across the brain, with constraints from peptidases and tissue-specific diffusion barriers confining its effects locally (Chini and others 2017). In vivo responses are typically observed within the range of 1 to 100nM Oxt, consistent with Oxtr ligand-binding assays (Elands and others 1988), but significantly higher than picomolar CSF Oxt concentrations (Monte and others 2014; Striepens and others 2013). In vivo electrophysiology experiments typically report effects at 1 to 5μM concentrations due to limited peptide penetration, which may yield conflicting results, as distinct Oxtr signaling cascades can be activated at different Oxt concentrations (Busnelli and others 2012; Chini and others 2017). However, CSF measurements might be underestimated, based on the low recovery rate (~10%) of microdialysis probes (Landgraf and Ludwig 1991). Recent technological advancements, such as the development of fluorescent sensor MTRIAOT (Ino and others 2022) and the genetically encoded sensor GRABOT (Qian and others 2023), have enabled the measurement of extracellular Oxt and the detection of Oxt release with high spatiotemporal resolution. These sensors offer significant potential for elucidating CSF Oxt concentrations and determining whether CSF Oxt reaches brain areas such as the somatosensory or primary motor cortices, which lack oxytocinergic projections. Yet, their current sensitivity is in the nanomolar range, whereas basal CSF Oxt concentrations are believed to be in the picomolar range. Consequently, while these sensors are likely to perform well in brain regions with consistently high Oxt concentrations, such as the PVH and SON, and show promise in detecting activity-dependent increases in Oxt release, enhancing their sensitivity is crucial to resolving the CSF Oxt debate under basal conditions. Another alternative was pioneered by Nakamura and colleagues (2022): click chemistry, whereby Oxt tagged with a small alkyne molecule is applied to acute brain slices, fixed, and conjugated with azide-tagged fluorophores. These authors identified high-affinity binding sites in the hippocampus, which respond to low concentrations of Oxt presumed to reach the target receptors through volume transmission, as supported by fast turnover of Oxt binding and lack of uptake into cells. As noted by the authors, click chemistry could also be used in vivo to explore Oxt delivery via CSF and shed light on spatiotemporal dynamics of extracellular Oxt in different sites of action.

As alluded to in the hippocampus example, variability in receptor expression, efficacy, and binding sites across brain regions and physiologic states can influence the sensitivity of different areas to identical Oxt concentrations (Jurek and Neumann 2018; Meddle and others 2007). Oxt binding induces diverse conformational changes in Oxtrs, activating various downstream effectors (Reversi and others 2005). While Gi and β-arrestins require higher concentrations for activation, the Gq pathway can be turned on by lower levels of Oxt, particularly by homodimeric Oxtrs (~1000-fold lower), as compared with monovalent forms (Busnelli and others 2012; Busnelli and others 2016). Gq signaling is the primary driver of Oxt action, inducing calcium influx and downstream proliferative effects (Wang and Hatton 2007), although elevated Oxt levels or enhanced Gi expression can modify this response (Busnelli and others 2012). For instance, atosiban, once viewed as an Oxtr antagonist, is now recognized as a biased agonist that selectively engages the Gi pathway upon Oxtr binding to increase cytokine synthesis and inhibit cell growth (Kim and others 2016; Reversi and others 2005). Furthermore, owing to structural homology, high Oxt concentrations can activate arginine vasopressin (Avp) receptors, especially vasopressin receptor 1A (V1aR), and induce a response similar to Gi signaling (Chini and others 2017; Manning and others 2012; Neumann and Landgraf 2012). Hence, under basal conditions, low CSF Oxt levels selectively recruit the Oxtr-Gq pathway, while activity-dependent increases in Oxt concentrations may engage Oxtr-Gi and V1aR in select brain areas to modulate region-specific responses.

The efficacy of volume transmission is expected to be further influenced by various regulatory mechanisms, such as steroid hormone action (Gimpl and Fahrenholz 2001), that govern local receptor sensitivity and recycling dynamics (Conti and others 2009). Such an influence of steroids is evident in the ventromedial hypothalamus and preoptic area, where receptor sensitivity fluctuates throughout the estrous cycle, affecting the responsiveness of these regions to basal Oxt levels (Bale and others 1995; Caldwell and others 1994; Jirikowski and others 2018). On top of these considerations, significant variations (up to fivefold) in diurnal CSF Oxt levels complicate the definition of “basal” levels (Artman and others 1982; Devarajan and Rusak 2004). Even if basal concentrations are insufficient to elicit a brainwide response, CSF could propagate activity-dependent increases in Oxt signaling, turning local signals into global responses during states of heightened Oxt release, as evidenced by elevated CSF Oxt levels during osmotic stimulation and nursing (Kendrick and others 1987; Morris and others 1984).

Volume transmission of Oxt is supported by the consistently reported mismatch between Oxt projections and Oxtr expression (Herkenham 1987; Manjila and others 2022; Son and others 2022). For instance, the olfactory bulb, crucial for social bonding and parental behavior in rodents (Meddle and others 2000), exhibits abundant Oxtr expression despite lacking direct projections from hypothalamic Oxt neurons (Son and others 2022), lending support to the functional significance of volume transmission of Oxt via CSF.

In accordance, the SON and PVH, which house most Oxt neurons, are strategically located near the ventral subarachnoid space and third ventricle (3V), respectively, facilitating interaction with CSF (Veening and others 2010). While Oxt PCNs project to ventricles and other ventricular system–related areas, dendrites of MCNs in the PVH extend centromedially, penetrating the ventricular lining either subependymally or running along its surface to reach the supraependymal plexus on the 3V floor (Buijs 1978; Cloft and Mitchell 1994; Ju and others 1992; Krisch 1986; Li and others 2024). Some dendritic processes and axon collaterals reach across to the dorsal 3V (Rhodes and others 1981), contributing to bilateral PVH synchronization in lactating females (Belin and Moos 1986). SON MCNs also project dendritic processes toward the ventral surface and into the subarachnoid space, instrumental to contralateral SON activation during unilateral Oxt neuron stimulation in lactating animals (Neumann and others 1995). Dendritic processes of periventricular nucleus Oxt neurons penetrate the ventricular lumen as well (Jirikowski 2019; Vigh-Teichmann and others 1970). Furthermore, axon terminals from SON and PVH can contact CSF through gaps in the ependymal lining of the infundibular recess, a CSF-filled space connecting the 3V to the pituitary (Jirikowski 2019; Wittkowski 1969).

While the anatomic features detailed here facilitate the direct release of Oxt into CSF, these examples alone are insufficient to supply adequate Oxt for diffusion into distant brain regions. Somatodendritic release, accounting for 95% of central Oxt output (Rossoni and others 2008), is considered the primary source of CSF Oxt (Veening and others 2010; Figure 2a), although there are notable constraints in the time frame and effectiveness of this route. Diffusion of dendritically released Oxt into CSF and brain regions is hindered by factors such as tortuosity (i.e., diffusional path length) of the extracellular space and local variations in aminopeptidase expression. On one hand, only about 10% to 15% of extracellular fluid drains into the ventricular system (Weller and others 2009), and Oxt has a short half-life (<1 minute) in parenchyma (Stark and others 1989). On the other, continuous LDCV exocytosis sustains peptide availability for diffusion (Xia and others 2009), and the fraction reaching CSF can circulate longer due to a protracted half-life (~25 minutes; Mens and others 1983). Furthermore, additional factors, such as glial retraction, can substantially broaden the range of diffusion and increase Oxt release probability (Theodosis 2002). Oxt availability in parenchyma is also significantly influenced by levels of placental leucine aminopeptidase (P-LAP), which degrades Oxt (Matsumoto and others 2001). Despite being present in all Oxtr-expressing brain regions, P-LAP concentrations do not correlate with Oxtr density (Grinevich and others 2016). Although P-LAP decreases Oxt availability at excess, moderate levels can coordinate lactation-induced burst firing and enhance tissue sensitivity by countering Oxtr desensitization (Beyer and others 2010; Tobin and others 2014).

Others in the field postulate that axonal, specifically en passant, release (Figure 2b and 2c) contributes significantly to volume transmission (Chini and others 2017), challenging the predominant view of somatodendritic release in the SON and PVH as the main source of CSF Oxt. While en passant release may explain some Oxt actions previously attributed to somatodendritic release (Li and others 2024), such as satiety signaling through transsynaptic co-release of glutamate in the ventromedial hypothalamus (Griffin and others 2010), sparse collaterals of Oxt axons are unlikely to maintain basal CSF Oxt levels alone. It is likely that the two modalities work together such that somatodendritic transmission establishes a baseline and fine-tunes excitation, while axonal release provides precise control over timing and location, efficiently targeting Oxtrs in distant brain regions.

Transmission via Perivascular Spaces, Circulation, and Circumventricular Organs

The peripheral role of Oxt as a hormone is well established, as is the close interaction between the ventricular system and cerebral vessels. However, the role of circulation in central Oxt transport, whether it be direct through blood flow or indirect via CSF movement through perivascular spaces, is often overlooked.

While CSF production was historically thought to be limited to the choroid plexus and ependymal cells of the ventricular system, with negligible contributions from parenchyma (McComb 1983), recent evidence suggests that the walls of CNS capillaries are responsible for a bulk of interstitial fluid and CSF formation and reabsorption (Hladky and Barrand 2016; Oresković and Klarica 2010). In a similar vein, although CSF absorption into circulation is traditionally thought to occur across arachnoid villi (Davson and others 1973) and cervical lymphatics (McComb 1983), studies with radioiodinated albumin have shown notable drainage through cerebral perivascular spaces and olfactory lobe subarachnoid spaces (Bradbury and others 1981), highlighting plasma and CSF as interconnected modes of molecule transport within the CNS.

Perivascular Spaces

Perivascular space, also known as Virchow-Robin space (Weed 1923), describes the area between astrocyte end feet that make up glia limitans and either the pial sheath (in arteries) or endothelial basement membrane (in capillaries and veins; Ineichen and others 2022; Figure 3). There has been considerable confusion about the terminology, structure, and function of perivascular spaces, with challenges in their distinction from other spaces made up of layers of basement membranes enclosed by smooth muscle cells around arteries, as well as questions regarding the plausibility of circulation driven by steady pressure in either space (Faghih and Sharp 2018). Here, we refer to perivascular space to describe extensions of the subarachnoid spaces surrounding cerebrovasculature, which follow the course of penetrating arteries down to the level of capillaries (Hadaczek and others 2006), making up the so-called glymphatic system (Jessen and others 2015), implicated in the flow of CSF from the subarachnoid space into parenchyma (Rennels and others 1985), as well as clearance of substances such as amyloid β from interstitial fluid into CSF (Hawkes and others 2014; Weller and others 2009). This conceptual framework suggests that CSF permeates into perivascular spaces around penetrating arteries, moves into parenchyma via aquaporin 4 channels (Iliff and others 2012; Figure 3a), and flows out through perivascular spaces surrounding deep veins to be drained into the cervical lymphatic system (Bacyinski and others 2017; Jessen and others 2015). The precise mechanisms underlying perivascular CSF flow remain a subject of ongoing debate: arterial pulsations were initially thought to be a main driver (Hadaczek and others 2006; Iliff and others 2013); however, arteriolar wall movements were found to propel oscillatory flow but not directional pumping (Kedarasetti, Drew, and others 2020), with suggestions that directional CSF flow may instead be explained by pressure differences, respiration, peristaltic motion in perivascular networks (Gjerde and others 2023), or “valves” formed by astrocyte end feet (Gan and others 2023), while functional hyperemia following neural activity may serve to promote CSF transport and metabolite clearance (Kedarasetti, Turner, and others 2020; Kim and others 2023).

Figure 3.

Figure 3.

Perivascular spaces and stimulus-induced neurovascular remodeling. (a) Schematic representation of the perivascular space. CSF flows within perivascular spaces that surround cerebrovasculature before drainage into the cervical lymphatic system. At the level of arteries, the inner border of perivascular spaces is demarcated by the pial sheath, which disappears and yields to the endothelial basement membrane at the capillary and venous levels. The outer boundary is formed by astrocyte end feet, which constitute the glia limitans of the blood-brain barrier. Aquaporin 4 channels mediate CSF movement between perivascular spaces and parenchyma, with direction and velocity of flow determined by various factors, including arterial pulsations, pressure gradients, respiration, and neural activity. (b) Oxytocin neurons and the perivascular space at baseline activity levels. (c) Stimulus-induced neurovascular remodeling and angiogenesis. Reversible changes to the neurovascular unit have been reported in the hypothalamus and posterior pituitary after activity-dependent increases (e.g., lactation, salt loading) in oxytocin signaling—namely, retraction of astrocyte end feet and concurrent extension of pericyte processes. Subsequent to this surface expansion, dendrites and axons of oxytocin neurons come in close contact with the perivascular space and endothelial basement membrane, facilitating diffusion of oxytocin into the CSF and potentially enabling penetration into vessels. The reversible increases in capillary density and neurovascular remodeling are orchestrated by the upregulated expression of angiogenic factors: VEGFA in astrocytes; VEGFR2, PDGFB, and PDGFRB in endothelial cells; PDGFB and VEGFR3 in oxytocin neurons; and PDGFRB in pericytes. AQP4 = aquaporin 4; PDGFB = platelet-derived growth factor β; PDGFRB = platelet-derived growth factor receptor; VEGFA = vascular endothelial growth factor α; VEGFR2 = vascular endothelial growth factor receptor 2; VEGFR3 = vascular endothelial growth factor receptor 3. Created with BioRender.com.

Perivascular clusters of Oxt neurons that extend projections into the vascular endothelium (e.g., circular nucleus; Møller and others 2018; Rovsing and others 2013) can be found in between the PVH and SON as well as in other accessory areas, mainly in preoptic and tuberal regions in rodents (Palkovits and others 1974; Sofroniew and Glasmann 1981), primates (Ichimiya and others 1988), and humans (Krolewski and others 2010). While some of their projections are thought to join axons from the canonical oxytocinergic nuclei and terminate at the posterior pituitary, others may penetrate perivascular spaces, releasing Oxt directly into cerebrovasculature (Jirikowski 2019; Møller and others 2018) or CSF (Morris and others 1998; Figure 3b and 3c). This is exemplified by the highly efficient delivery of CSF messages to ventromedial medullary neurons involved in synchronization of bilateral Oxt-secreting hypothalamic nuclei, thought to be facilitated by the vascular pump (Moos and others 2004). Additionally, increased numbers of Oxt neurons surrounding vasculature have been noted in various hypothalamic nuclei of parturient rats (Jirikowski and others 1988), hinting at a role for perivascular Oxt release in child delivery. Studies utilizing electron microscopical immunocytochemistry and morphometry (Blanco and others 1991) showed that in late pregnancy and after 2 days of lactation, numerous Oxt neurons can be found in direct contact with the outer basement membrane of vessels, at a much lower distance than that seen in ovariectomized controls and even after 9 days of lactation. These transitory changes may be driven by steroid-induced glial retraction (Hatton and others 1984) and may serve to enhance activity-dependent somatodendritic Oxt release (Kamei 1986), which could then diffuse into CSF and potentially supplement plasma Oxt levels (Wagner and others 1974). Through simultaneous release into circulation and CSF, perivascular Oxt neurons could serve to synchronize systemic and central Oxt actions under special physiologic circumstances, such as parturition and lactation.

Additionally, perivascular Oxt release may serve to modulate cerebral perfusion pressure and cerebrovascular tone through endothelial cell (EC) Oxtr signaling (Schini and others 1990; Thibonnier and others 1999), inducing endothelial nitric oxide (NO) synthase production (Cattaneo and others 2009), and calcium-dependent vasodilation and trophic responses (Thibonnier and others 1999). This could have implications for cerebral circulation of not only blood but also CSF: functional hyperemia—that is, elevated local blood flow triggered by neuronal (specifically sensorimotor) activity—is thought to increase CSF circulation and metabolite exchange (Kedarasetti, Turner, and others 2020; Kim and others 2023), and several types of sensory stimulation are known to trigger Oxt release (e.g., suckling, scent of offspring, physical touch; Uvnäs Moberg and others 2022). Oxt-positive feedback on its own release may hence be reinforced by CSF-perivascular mechanisms, whereby somatosensory activity leads to Oxt release and the ensuing functional hyperemia and Oxt-induced vasodilation promote further Oxt neuron activity and neuropeptide diffusion in CSF.

Evidence backing this idea is present in studies of hyperosmotic stimulation, a known trigger of Oxt signaling (Balment and others 1980; Balment and others 1986; Ueno and others 2020), that show activity-dependent reversible remodeling and angiogenesis in hypothalamic and posterior pituitary microvessels (Alonso and others 2005; Miyata and Hatton 2002; Figure 3b and 3c). Similar morphologic adaptations have been reported in both regions in response to physiologic stimuli that elicit increased Oxt release, such as lactation, parturition, hemorrhage, restraint stress, as well as central administration of Oxt itself (Blanco and others 1991; Hatton 1988; Miyata and others 1994; Theodosis and others 1986; Tweedle and Hatton 1987). In the posterior pituitary, neurovascular reconstruction is driven by shape conversion of pericytes and astrocytes, increasing vascular surface area and permeability and enhancing direct contact of Oxt axonal terminals with the outer basement membrane (Furube and others 2014; Nishikawa and others 2017). These changes are accompanied by increased Oxt release and heightened expression of angiogenic factors in Oxt axonal terminals, astrocytes, ECs, and pericytes (Furube and others 2014; Nishikawa and others 2017; Figure 3c). Such remodeling in the pituitary presumably serves a secretory function by increasing the surface area for Oxt diffusion into peripheral circulation (and perhaps into CSF) to meet the high demand for Oxt, whereas morphologic plasticity in the hypothalamus is thought to have several functional consequences. For example, retraction of astrocytic processes can promote contact between Oxt neurons (facilitating paracrine somatodendritic release and diffusion), increase neuronal excitability via reductions in glutamate clearance and membrane potential, and concurrently allow synaptic remodeling and formation of new synapses onto Oxt neurons (Baudon and others 2022; Theodosis 2002; Theodosis and others 2006). Furthermore, increased blood delivery following angiogenesis could enhance the monitoring of osmotic agents such as sodium (Roy and others 2021), and elevated oxygen (O2) levels could serve to modulate neural activity (Zhang and others 2023).

Interestingly, inverse neurovascular coupling was discovered to induce positive feedback excitation of Avp neurons such that hyperosmotic stimulation (i.e., salt loading) progressively increased Avp release while evoking vasoconstriction that reduced local blood flow (Roy and others 2021). These seemingly contradictory neurovascular responses are compatible with the complementary actions of the two neuropeptides. While Oxt and Avp converge in their response to hyperosmolality (Balment and others 1986), they have opposite effects in plasma volume regulation: Oxt promotes volume contraction by modulating autonomic nervous system activity (Higa and others 2002) and inducing atrial natriuretic peptide release (Favaretto and others 1997), whereas Avp increases fluid retention and water intake (Kimura and others 1976; Szczepanska-Sadowska and others 1982). As neurosecretory cells synthesizing Oxt and Avp reside in the same hypothalamic nuclei, their local milieu must be under very stringent control to ensure the proper response to changes in osmolality and volume from both cell groups.

In support of this notion, it was previously shown that Avp and NO may be involved in the balance between vasoconstriction and vasodilation in response to osmotic stimulation (Du and others 2015). Blockade of V1aRs showing inverse neurovascular coupling unmasked underlying salt-induced vasodilation (Roy and others 2021) that could presumably be driven by Oxt neurons, which, upon osmotic stimulation, upregulate expression of neuronal NO synthase (Kadowaki and others 1994; Srisawat and others 2004), a modulator of Oxt and Avp MCN activity in various contexts, such as LTP maintenance and acute stress response (Matsuura and others 2023; Orlando and others 2007). In turn, basal Oxt and Avp release is tonically suppressed by NO-mediated potentiation of GABAergic inputs (Stern and Ludwig 2000). Conversely, under circumstances that elicit activity-dependent release, the inhibitory effect of NO on Avp is alleviated, while its suppression of Oxt secretion is acutely intensified (Antunes-Rodrigues and others 2004; Kadekaro and Summy-Long 2000). However, elevated O2 levels, which promote NO degradation (Zhang and others 2023), potentially boost Oxt MCN activity by alleviating NO-mediated negative feedback. Besides NO availability, O2 concentration directly regulates potassium channel expression and the rate of neuromodulator synthesis (Zhang and others 2023). Specifically, potassium channels TASK-1 and TASK-3 (Turner and Buckler 2013) as well as Kv3 channels (Kaczmarek and Zhang 2017) can be inhibited by declining O2 levels, leading to membrane depolarization and increased neuronal excitability. The presence of these potassium channels has been confirmed in hypothalamic MCNs (Han and others 2003; Kaczmarek and Zhang 2017), with stronger expression of some in Avp neurons (Shevchenko and others 2004). Therefore, increases in O2 concentrations may enhance Oxt activity, while declining O2 levels may promote Avp signaling, providing a basis for the antagonistic neurovascular coupling responses elicited by the two neuronal populations. Such distinct regulatory mechanisms likely fine-tune the hypothalamic microenvironment to establish a balance between Oxt and Avp and thus achieve stimulus-dependent neural discrimination.

Blood Circulation and the BBB

Despite Oxt regulation of vasculature throughout the body (Gutkowska and Jankowski 2012; Gutkowska and others 2000), most research on Oxt and cerebrovasculature has focused on Oxt’s ability to permeate the BBB (Bowen 2019), as motivated by many studies and trials investigating the use of exogenous (specifically intranasal) Oxt for treatment of disorders such as autism spectrum disorder (Martins and others 2022). It was believed the BBB prevented the diffusion of Oxt in meaningful quantities from peripheral circulation into the brain and vice versa (Mens and others 1983), prompting mutually exclusive regulation of the two Oxt systems. Indeed, peripheral and central release of Oxt can occur independently (Landgraf and Neumann 2004); for example, dendritic release is thought to lag behind increases in plasma Oxt levels during osmotic stimulation (Neumann and others 2008). However, the cardiovascular system, namely the heart and large vessels, is able to synthesize Oxt in relatively large quantities (Gutkowska and others 1997; Jankowski and others 1998; Thibonnier and others 1999; Figure 1) and may contribute to the early rise in plasma levels prior to release in the pituitary, although this remains to be studied. Additionally, numerous physiologic cues, such as parturition, lactation, sexual activity, and stressors, can trigger Oxt to be released centrally and peripherally (Althammer and others 2021; Neumann and Landgraf 2012). Elicited behavioral effects, such as maternal care, anxiolysis, and social recognition, complement peripheral Oxt actions (i.e., labor and milk let-down; Engelmann and others 2004; Landgraf and others 1988; Neumann 2007). Nonetheless, co-release into plasma and CSF by collaterals of MCNs projecting to the pituitary can partially explain this phenomenon (Zhang and others 2021), and many Oxt-mediated behavioral effects (Jurek and Neumann 2018; Marsh and others 2021; Rigney and others 2022) are not accompanied by peripheral secretion (Althammer and others 2021).

Recent studies and meta-analyses point to a positive, albeit moderate and highly heterogeneous, correlation between Oxt concentrations in the CNS and periphery (Valstad and others 2017). While basal levels show no association, a positive correlation was observed after intranasal Oxt (IN-Oxt) treatment or introduction of experimental stressors, such as maternal separation (Kojima and others 2012) and forced swim test (Williams and others 2012), and was taken as supporting the hypothesis that basal Oxt levels in the blood and CNS are uncoordinated (Valstad and others 2017). The emergence of a correlation after experimental stressors could be attributed to hypothalamic-pituitary-adrenal axis activation, prompting interaction between corticotropin-releasing hormone PCNs and Oxt MCNs in the PVH (Ferguson and others 2008; Williams and others 2012) as well as autonomic feedback on the Oxt system via peripheral corticosteroid action (Quintana, Alvares, and others 2015). However, there are many caveats to such premature conclusions, as many issues have been reported with regard to the accurate measurement of peripheral plasma Oxt concentrations—specifically, discrepancies between radioimmunoassay and enzyme immunoassay methods that also depend on sample extraction (Szeto and others 2011; for extensive reviews and recommendations for best practice, see Leng and Sabatier 2016; MacLean and others 2019; D. Martins and others 2020; McCullough and others 2013; Tabak and others 2023). Furthermore, basal Oxt dynamics show oscillations that lead to tonic release (Ino and others 2022), and the half-life of Oxt is much shorter in blood (~3-8 minutes; Morin and others 2008) than in CSF (~20-30 minutes; Jones and Robinson 2008; Mens and others 1983). Therefore, even if peripheral and central release occurs simultaneously, random sampling of CSF and plasma could show discrepant concentrations due to differences in the timeline of Oxt degradation in the two media (also highlighted by Tabak and others 2023). In this line of reasoning, correlations detected after IN-Oxt administration or experimental stressors could be explained by stimulus-evoked release that is not constrained by baseline release dynamics. Moreover, sampling of CSF and plasma after experimental procedures would not be random but rather likely fall in the time window preceding degradation of plasma Oxt, leading to the detection of a correlation. Future research using sensors for real-time detection of Oxt release dynamics (Ino and others 2022; Qian and others 2023) and validated methods, such as combined two-dimensional liquid chromatography and mass spectrometry, to measure peripheral Oxt concentrations (Zhang and others 2011) could settle this debate on the correlation between peripheral and central Oxt (MacLean and others 2019; Tabak and others 2023). Moreover, establishing whether peripherally synthesized Oxt contributes to plasma Oxt levels and, if so, quantifying the extent of this contribution will aid in distinguishing increases attributable to release from the posterior pituitary (Assinder 2022; Figure 1).

Discrepant release dynamics and concentrations do not necessarily preclude an interaction between peripheral and central Oxt, especially in the setting of exogenous Oxt administration. Mode of delivery appears to be a key determinant, as indicated by findings of studies comparing IN-Oxt with intravenous (IV) Oxt administration, suggesting that while these methods of delivery lead to comparable peripheral Oxt concentrations, only IN-Oxt is able to elicit social cognitive effects (Quintana, Westlye, and others 2015) and neural effects (Quintana and others 2016), which implies IN-Oxt specific penetration of brain tissue. However, others reported that IV Oxt and IN-Oxt can activate Fos expression at the PVH, the area postrema, and the dorsal motor nucleus of the vagus (Maejima and others 2015). Importantly, the area postrema, located outside the BBB, is not accessible to CSF-Oxt, indicating a potential role for CVOs (Jeong and others 2021) for the detection of peripheral Oxt levels, as detailed later in this section. Similarly, increased Oxt levels in systemic circulation were found to explain Oxt-induced decreases in blood flow in the left amygdala and anterior cingulate cortex following IN-Oxt and IV Oxt (D. A. Martins and others 2020). Yet, this same study found additional changes in cerebral perfusion induced by IN-Oxt only, which could not be accounted for by changes in plasma Oxt, corroborating results from another group reporting that Oxt in amygdalar extracellular fluid was increased more by IN-Oxt delivery as compared with intraperitoneal (Smith and others 2019). In contrast, a recent clever experiment by Yao and colleagues (2023) demonstrated that the use of a vasoconstrictor pretreatment restricting IN-Oxt entry into systemic circulation not only impeded the increases in peripheral Oxt concentrations but also abolished a majority of resting-state neural responses to IN-Oxt, as measured by delta-beta cross-frequency coupling. These findings collectively suggest that peripheral routes of Oxt delivery are sufficient to elicit most central effects, while IN-Oxt may target additional brain regions, presumably via direct CSF penetration (Figure 4).

Figure 4.

Figure 4.

Oxytocin and proposed pathways across the blood-brain and blood-cerebrospinal fluid barriers. (a) Potential mechanisms of oxytocin absorption into the CNS following intranasal administration can involve direct delivery through axonal and paracellular transport, as well as indirect routes such as signal transduction following V1aR binding and RAGE-mediated uptake into nasal vessels. (b) In RAGE-mediated transport across the blood-brain barrier, oxytocin can either bind to membrane-bound RAGE on the basal side of vascular endothelial cells or attach to soluble RAGE, which then dimerizes with the membrane-bound form. The oxytocin-RAGE complex undergoes endocytosis into the cell and subsequent exocytosis to the apical side, facilitating diffusion into the neural parenchyma. (c) Oxytocin transport from plasma to CSF follows a similar process. At the blood-CSF barrier, oxytocin diffuses freely out of fenestrated vessels and is then shuttled from the basal to apical side of choroid plexus ependymal cells via RAGE. mRAGE = membrane-bound RAGE; RAGE = receptor for advanced glycation end products; SAS = subarachnoid space; sRAGE = soluble RAGE; V1aR = vasopressin receptor 1A. Created with BioRender.com.

Strikingly, even with the delivery of very high concentrations of IN-Oxt, only negligible increases have been detected in CSF despite significant elevations in plasma levels (Born and others 2002; Chang and others 2012; Monte and others 2014). Based on these findings, two potential pathways of IN-Oxt entry into the brain were described, the first being axonal transport following uptake into olfactory and trigeminal neurons (Chen and others 1998; Thorne and others 1995, pathway 1) and the second consisting of passage into subarachnoid space (Born and others 2002; Illum 2000; Figure 4a). The first pathway could explain reports of low CSF levels following IN-Oxt delivery: Oxt is rapidly degraded in neural parenchyma, and only a fraction of centrally released Oxt reaches CSF (Born and others 2002; Thorne and others 1995); thus, it is possible that IN-Oxt could reach some brain regions via axonal transport without entering CSF. The second pathway suggests that IN-Oxt diffuses passively through perineural spaces of the nasal epithelium, bypassing the BBB entirely (Ermisch and others 1985; Fehm and others 2000; Illum 2000; Vyas and others 2005; Figure 4a, pathway 2). The finding that diverse patterns of Oxt-induced changes are seen in cerebral blood flow upon application of the same dose of IN-Oxt via standard nasal spray and nebulizer endorses this idea, as such differences can be related to small spatial variations in the deposition of IN-Oxt in paranasal cavities that make up entry points to the brain (D. A. Martins and others 2020).

However, the potential mechanisms of IN-Oxt are not limited to these two pathways. As mentioned, experimental use of extra physiologically high Oxt concentrations can lead to atypical effects via Oxt action at V1aRs (Chini and others 2017; Manning and others 2012; Neumann and Landgraf 2012). These receptors are expressed in the olfactory epithelium (Levasseur and others 2004), where Oxt binding could trigger signaling cascades back to the brain through synaptic pathways (Figure 4a, pathway 3, dashed line). Another explanation is that small but functionally significant amounts of Oxt can cross the BBB (Neumann and Landgraf 2012), exerting effects exclusively in areas with high Oxtr expression (e.g., amygdala; D. A. Martins and others 2020). Support for this hypothesis can be found in studies showing increased synthetic Oxt in CSF of primates after IV infusion of labeled synthetic Oxt despite no effects on endogenous release (Lee and others 2018). Similarly, intraperitoneal delivery of synthetic Oxt in wild type and Oxt knockout (KO) mice led to increases in extracellular fluid Oxt in the amygdala and dorsal hippocampus (Smith and others 2019). This hypothesis has gained further traction in the last few years with the discovery that the receptor for advanced glycation end products (RAGE) can act as an Oxt-specific transporter (Higashida and others 2017; Yamamoto and others 2019; Figure 4). RAGE was initially demonstrated to shuttle Oxt across the intestinal lumen, leading to absorption of Oxt from breastmilk into infant blood via intestinal permeability (Higashida and others 2017). This finding prompted studies of binding and transport characteristics of RAGE at the BBB, leading to the discovery that RAGE is expressed by vascular ECs and choroid plexus epithelium in the brain (Figure 4b and 4c) and mediates Oxt transport from peripheral circulation into the brain about 10 times more efficiently than the opposite direction (Figure 4a, pathway 4; Gerasimenko and others 2021; Leerach and others 2021; Munesue and others 2021; Shimizu and others 2020). Furthermore, this directional transport into the brain was obstructed in RAGE KO and knockdown mice, as demonstrated by little or no increase in CSF Oxt after IN-Oxt application (Yamamoto and others 2019). Notably, RAGE KO mice showed hyperactivity and deficits in maternal bonding (Yamamoto and others 2019) that were exacerbated by additional stress (Gerasimenko and others 2021), recapitulating behaviors seen in Oxt and Oxtr KO models as well as mice with deficiencies in Oxt secretion (i.e., CD38 KO; Ferguson and others 2000; Jin and others 2007; Takayanagi and others 2005). Importantly, these behavioral effects were not caused by a disruption of central Oxt signaling, as hypothalamic Oxt release was not affected by RAGE deficiency (Yamamoto and others 2019). Moreover, defects in Oxt transport and abnormalities in maternal behavior were rescued in RAGE KO mice upon transgenic EC expression of RAGE (Yamamoto and others 2019).

These findings suggest not only that peripheral Oxt can be transported into the brain but also that the delivered amounts are functionally meaningful, as the obstruction of such transport can abolish the central effects of exogenous Oxt (Yao and others 2023) and cause behavioral consequences without disruption of endogenous central Oxt signaling (Gerasimenko and others 2021; Yamamoto and others 2019). Munesue and colleagues (2021) proposed that transport of Oxt from peripheral circulation into the brain could be an alternative to volume transmission and en passant release in supplying areas without direct projections from Oxt neurons. The plausibility of this assertion is controversial, as the amount of Oxt that would need to be transported from the periphery into neural parenchyma to elicit behavioral and cognitive responses may be much higher than the transport capacity of RAGE alone, especially considering that Oxt cannot compete with the interaction of RAGE with other ligands (Leerach and others 2021). Moreover, despite Higashida and colleagues’ (2017) assertion that RAGE is expressed only by vascular ECs and choroid plexus epithelium in the brain, others have detected RAGE in a subset of neurons in the cerebral cortex (Brett and others 1993), with further studies showing RAGE involvement in embryonic and adult neuronal differentiation, neurite outgrowth, and nerve regeneration (Rong and others 2004; Wang and others 2008), as well as pathogenesis of neurologic disorders (Juranek and others 2022). It is therefore not yet possible to establish a causal relationship between the RAGE KO behavioral phenotype and impaired Oxt transport, even in EC-specific RAGE KO models, as RAGE is also implicated in vascular development and homeostasis (Adamopoulos and others 2016; Liu and others 2018) and disruption of RAGE signaling in cerebrovasculature would presumably affect neural function. That being said, RAGE is not the only candidate for an endogenous Oxt transporter: recent investigations by Værøy and colleagues (2023) indicate that a significant amount of plasma Oxt is naturally and reversibly bound to immunoglobulin G (IgG), a molecule capable of crossing the BBB (Chang and others 2018), and that the Oxt/IgG immune complexes are functionally active at the Oxtr, as demonstrated by calcium mobilization and receptor internalization assays (Værøy and others 2023). Furthermore, the same study showed that violent inmates have higher levels of Oxt-reactive IgG that are less efficient at carrying Oxt, as demonstrated by affinity kinetic parameters, which were correlated with psychological measures of aggressiveness. Overall, recent studies investigating potential transport mechanisms provide compelling support for the notion that peripheral Oxt reaches the brain and is essential for the complete spectrum of Oxt-induced behavioral and neural effects, although the exact amount of Oxt transported from blood to brain, as well as the details of such candidate mechanisms, remains to be fully elucidated. As with volume transmission, methods such as click chemistry or biosensors, which are minimally invasive and have superior resolution versus alternatives such as radiolabeling, fluorescence-based tagging, and microdialysis, could be utilized to determine whether peripherally administered Oxt reaches the brain (Ino and others 2022; Nakamura and others 2022; Qian and others 2023). Moreover, there are further avenues of interaction between neural tissue and peripheral circulation that are unrestricted by the BBB, which could supplement the proposed RAGE-mediated Oxt transport mechanism in various ways.

Circumventricular Organs

CVOs are brain structures found in association with the third and fourth ventricles (Hofer 1958), distinguished by their fenestrated or otherwise permeable vascular network and proximity to the ventricular system (Petrov and others 1994; Wislocki and King 1936), making them a triadic interface among peripheral blood, CSF, and brain parenchyma (Sisó and others 2010). The proximity of all CVOs to either ventricular recesses or junctions and the orientation (and sometimes contact) of CVO-associated vessels and capillary loops toward ventricular cavities suggest a functional component to this physical relationship that can facilitate molecular interactions (Duvernoy and Risold 2007). There are 8 CVOs, although this number is contested (Duvernoy and Risold 2007; Oldfield and Mckinley 2004). Three of these—namely, the subfornical organ (SFO), the organum vasculosum of the lamina terminalis (OVLT), and the area postrema are sensory—while the neurohypophysis (or posterior pituitary), the median eminence, the pineal gland, and the subcommisural organ are considered secretory (Sisó and others 2010). The choroid plexus, which does not house any neural tissue, is sometimes referred to as a CVO, while the CVO identity of the subcommissural organ is questionable due to its lack of fenestrated endothelium (Petrov and others 1994). Sensory CVOs sample molecules from blood and interstitial fluid, relaying this information via projections to central hubs controlling autonomic functions, body fluid balance, and immune surveillance (Sisó and others 2010), implying significant overlap of CVO function with Oxt activity. Of note, all three sensory CVOs are in very close proximity to and/or have functional connectivity with hypothalamic nuclei that house Oxt neurons (Jeong and others 2021; Figure 5a).

Figure 5.

Figure 5.

Circumventricular organs and the oxytocin system. (a) Hypothalamic oxytocinergic nuclei exhibit extensive reciprocal connections with select sensory CVOs, such as the SFO, OVLT, and AP, as well as certain secretory CVOs, such as the ME and PP. (b) The HPH tract and the hypophyseal portal system. The HPH tract containing oxytocinergic fibers courses through the median eminence and infundibulum before terminating as Herring bodies at the posterior pituitary capillary network. The primary plexus of the pituitary portal system, situated within the infundibular stalk, receives hypothalamic releasing hormones (e.g., GnRH, CRH) from the TH tract and delivers them to the anterior pituitary via the secondary plexus. As depicted in the callout, fenestrated vessels allow interaction of the two tracts at the level of the median eminence, primary plexus, and short portal vessels connecting the anterior and posterior pituitary. (c) Gross and magnified structure of the OVLT in relation to oxytocin signaling. Through its primary capillary networks, the OVLT makes contact with the subarachnoid space and third ventricle, where fenestrated vessels and a leaky blood-brain barrier allow monitoring of CSF and plasma osmolality via sodium channels, osmosensors, and other receptors present on neurons and glia. Additionally, some cells in the OVLT express oxytocin receptors and may even synthesize oxytocin. The OVLT and SFO communicate information regarding body fluid homeostasis to oxytocinergic nuclei, eliciting volume contraction and natriuresis via oxytocin signaling. 3V = third ventricle; AP = area postrema; Arc = arcuate nucleus; CP = choroid plexus; CRH = corticotropin-releasing hormone; CVO = circumventricular organ; GnRH = gonadotropin-releasing hormone; HPH = hypothalamoposthypophyseal; ME = median eminence; OC = optic chiasm; OR = optic recess; OVLT = organum vasculosum of lamina terminalis; Oxt = oxytocin; PG = pineal gland; PoA = preoptic area; PP = posterior pituitary; PVH = paraventricular nucleus of hypothalamus; SCO = subcommisural organ; SFO = subfornical organ; SON = supraoptic nucleus; TH = tuberohypophyseal. Created with BioRender.com.

OVLT, which is the sensory CVO closest to the SON and PVH, is positioned at the anteroventral border of the optic chiasm and serves as the vascular component of the lamina terminalis, constituting the anterior boundary of the 3V (Sisó and others 2010). Of the two primary capillary networks of OVLT, the superficial intrapial plexus associates with the subarachnoid space, while the deep plexus is located subependymally and connects to the 3V (Figure 5c; Shaver and others 1992). Ependymocytes and tanycytes, distinct types of specialized glial cells that line the ventricles of the brain (Duvernoy and Risold 2007), make up the ependymal lining at points of contact between the OVLT and ventricular recesses (Sisó and others 2010), earning CVOs the nickname “hypendymal organs” owing to these specialized glial networks, a lack of BBB, and high aquaporin 4 expression (Figure 5c; Goren and others 2006). While ependymocytes are linked by tight junctions (Duvernoy and Risold 2007), tanycytes extend processes that contact the CSF and the OVLT (Mullier and others 2010), presumably allowing the OVLT to track CSF composition. Functionally, the dorsal cap of the OVLT serves as a primary hub for efferent projections to various brain regions, including the SON, PVH, cerebral cortex, limbic structures, midbrain areas, and medullary raphe nucleus (Camacho and Phillips 1981; McKinley and others 1992). Highly relevant to Oxt and Avp actions in regulating blood volume and osmolality via natriuresis (Antunes-Rodrigues and others 2004), sodium-sensitive OVLT neurons monosynaptically communicate with the PVN (Shi and others 2008), and polysynaptic efferent pathways connecting the OVLT, PVH, and kidneys have been confirmed by experiments using viral tracers (McKinley and others 2003). Similarly, cells within the SFO interact with MCNs of the PVN directly as well as indirectly through projections to other hypothalamic nuclei, including the bed nucleus of the stria terminalis, arcuate nucleus, OVLT, and median preoptic nucleus (Abbott and others 2016; Kolaj and Renaud 2010; Matsuda and others 2017; Yeo and others 2019), with reciprocal connections between SFO and median preoptic nucleus potentially creating a feedback loop (Kawano 2017). Electrical stimulation of SFO is sufficient to induce Oxt and Avp release (Ferguson and Kasting 1987). In this vein, water deprivation, angiotensin I and II signaling, as well as hypertonic saline infusion and hyponatremia, which are all triggers for Oxt and Avp release, have been reported to activate OVLT and SFO osmoreceptors (Anderson and others 2000; Negoro and others 1988; Richard and Bourque 1995). Furthermore, OVLT and SFO neurons express high levels of angiotensin II type 1 receptors and angiotensin-converting enzyme (Giles and others 1999). Oxt-immunoreactive fibers have been observed in the OVLT, which were mostly traced back to astrocytes and basement membranes of the primary capillary network (Sisó and others 2010). Similarly, the superficial capillary plexus of the SFO shows Oxt staining, with immunohistochemical methods and mRNA measurements indicating that this signal might be coming from microglia (Blackmore and others 2018; Maejima and others 2022).

Such findings provide not only support for the notion that CVOs are functionally upstream of PVN and SON in establishing a balance of neuroendocrine-mediated metabolic pathways (Jeong and others 2021) but also raise questions regarding the purpose of Oxt expression in CVO glial cells of some species. Despite its classification as a sensory CVO, OVLT has been proposed to serve as a “double organ,” which acts as a sensor of molecules circulating in blood and CSF, as well as a secretory structure receiving dense afferent inputs from the hypothalamus (Oldfield and Mckinley 2004). This raises the unlikely but exciting probability that glial cells may not only sense but also secrete Oxt in CVOs (Maejima and others 2022). The purpose of such Oxt activity is up to debate; however, as with neurovascular reconstruction, it is possible that it may serve to modulate neuronal excitability. Oxt activity is known to increase BBB integrity by preventing excessive leakiness of the barrier (Momenabadi and others 2020; Stary and others 2019), and Oxt neurons themselves are able to sense the osmolarity of their environment, which increases their excitability (Mecawi and others 2022). Moreover, glial cells in the SFO were shown to regulate neuronal activity by sensing sodium levels in body fluids (Shimizu and others 2007). Oxt detection and secretion in CVOs could thus serve as a feedback mechanism aimed at regulating dispersion of peripheral signals in the CNS by 1) modulating afferent inputs from CVOs into hypothalamic nuclei and 2) monitoring the amount of osmolytes diffusing into CSF from circulation. Such regulatory mechanisms would serve to prevent hyperexcitability of Oxt neurons while ensuring optimal Oxt secretion in response to osmotic stimulation. The discovery of Oxtr expression in the OVLT, SFO, area postrema, and median eminence (Yoshida and others 2009) as well as the MPOA surrounding the OVLT (Sharma and others 2019) supports the notion that local Oxt signaling may serve to fine-tune afferent outputs from CVOs, potentially detecting and responding to peripheral Oxt that can freely diffuse into CVOs that lack a BBB.

Projections of Oxt MCNs are found within two of the secretory CVOs—namely, the structurally and functionally related neurohypophysis and the median eminence (Carey 1959; Currie and others 1960), implicating a role for these structures in Oxt release. The neurohypophysis (or posterior pituitary), originating from the floor of the 3V, consists of a distal part, the neural lobe, and a proximal part: the median eminence, separated by the hypophyseal recess (Duvernoy and Risold 2007). It is vascularized by an extensive capillary network receiving Oxt and Avp through the HPH tract (Figure 5b, yellow projections). The median eminence comprises an internal layer containing the HPH tract and an external layer receiving tuberohypophyseal fibers carrying releasing hormones from the hypothalamus to the adenohypophysis (or anterior pituitary; Yin and Gore 2010; Figure 5b, green projections), whereby the hypophyseal portal system facilitates one-way traffic of hormones, with a primary plexus receiving neurohormones and portal vessels reaching a secondary plexus vascularizing the adenohypophysis (Amar and Weiss 2003). The secondary plexus comprises long capillary loops that cross the external and internal layers of the median eminence. The fenestrated endothelium of these median eminence capillaries, coupled with large perivascular spaces (Wittkowski 1969), could allow interaction of the HPH tract with the tuberohypophyseal tract within these long capillaries (Jirikowski 2019), as well as within short portal vessels that link the lobes of the pituitary (Baertschi and others 1980; Bergland and Page 1978). Oxt and Avp are secretagogues of ACTH, which is released by the anterior pituitary upon corticotropin-releasing hormone signaling from the hypothalamus (Engelmann and others 2004), although how these neuropeptides reach the corticotrophs of the anterior pituitary is undetermined. It is likely that en passant release of Oxt and Avp in the median eminence (Buma and Nieuwenhuys 1987) could trigger ACTH secretion (Engelmann and others 2004; Figure 5b). Oxt and prolactin, another hormone of the anterior pituitary, are both involved in lactation and regulate the release of each other, with Oxt stimulating prolactin release (Egli and others 2006) and coordinating its pulsatile secretion via negative feedback (Bertram and others 2010), leading to the suggestion that Oxt might be the evasive prolactin-releasing hormone (Villegas-Gabutti and others 2018). If this is true, Oxt could enter the tuberohypophyseal system at the level of the median eminence and promote prolactin release in the anterior pituitary. Other than facilitating communication between the two hypothalamohypophyseal tracts, the long capillary loops of the secondary plexus reach the hypophyseal recess, suggesting a potential interaction of portal blood of the two tracts and CSF (Duvernoy and Risold 2007). The fenestrated capillaries of the median eminence contain tanycytes, which line the floor of the 3V and form a permeable layer, thus controlling barrier properties and allowing the exchange of molecules between CSF and parenchyma at the level of the arcuate nucleus (Mullier and others 2010). Axonal release at the level of the median eminence and perhaps posterior pituitary could therefore contribute to CSF Oxt levels and synchronize peripheral and central Oxt actions.

Conclusion

Diverse yet interrelated mechanisms intricately regulate the transport and action of Oxt within the CNS. The coordinated interplay between axonal and somatodendritic Oxt release orchestrates rapid and localized physiologic, behavioral, and cognitive responses, complemented by volume transmission, which propagates Oxt signaling across the entire brain over an extended time frame. Here, we have highlighted the function of perivascular spaces as conduits for Oxt exchange between the CSF and neural parenchyma, exemplifying dynamic structural and functional adaptations induced by stimulus-driven increases in Oxt activity. While much emphasis has been placed on Oxt’s neuromodulatory role, its significant cardiovascular functions within the CNS have been comparatively overlooked. Exploring reciprocal interactions between the oxytocinergic and vascular systems in the brain holds promise for elucidating neurodevelopmental pathophysiology and age-related disorders, where concurrent alterations in both systems have been observed (El Assar and others 2012; Ghazy and others 2023; Ouellette and others 2020). Further investigations into these interactions hold promise for deepening our understanding of CNS function and introducing novel avenues for Oxt-based therapeutic interventions.

Supplemental Material

sj-docx-1-nro-10.1177_10738584241268754 – Supplemental material for Navigating Central Oxytocin Transport: Known Realms and Uncharted Territories

Supplemental material, sj-docx-1-nro-10.1177_10738584241268754 for Navigating Central Oxytocin Transport: Known Realms and Uncharted Territories by Deniz Parmaksiz and Yongsoo Kim in The Neuroscientist

Acknowledgments

Figures were generated using BioRender.com.

Footnotes

The authors declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Funding: The authors disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This work was supported by National Institutes of Health grants R01MH116176 and R01NS108407 to Y.K.

Supplemental Material: Supplemental material for this article is available online.

References

  1. Abbott SBG, Machado NLS, Geerling JC, Saper CB. 2016. Reciprocal control of drinking behavior by median preoptic neurons in mice. J Neurosci 36(31):8228–37. 10.1523/JNEUROSCI.1244-16.2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Adamopoulos C, Piperi C, Gargalionis AN, Dalagiorgou G, Spilioti E, Korkolopoulou P, and others. 2016. Advanced glycation end products upregulate lysyl oxidase and endothelin-1 in human aortic endothelial cells via parallel activation of ERK1/2-NF-κB and JNK-AP-1 signaling pathways. Cell Mol Life Sci 73(8):1685–98. 10.1007/s00018-015-2091-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Agnati LF, Fuxe K, Zoli M, Ozini I, Toffano G, Ferraguti F. 1986. A correlation analysis of the regional distribution of central enkephalin and β-endorphin immunoreactive terminals and of opiate receptors in adult and old male rats. Evidence for the existence of two main types of communication in the central nervous system: the volume transmission and the wiring transmission. Acta Physiol Scand 128(2):201–7. 10.1111/j.1748-1716.1986.tb07967.x [DOI] [PubMed] [Google Scholar]
  4. Agnati LF, Guidolin D, Guescini M, Genedani S, Fuxe K. 2010. Understanding wiring and volume transmission. Brain Res Rev 64(1):137–59. 10.1016/j.brainresrev.2010.03.003 [DOI] [PubMed] [Google Scholar]
  5. Alonso G, Galibert E, Duvoid-Guillou A, Vincent A. 2005. Hyperosmotic stimulus induces reversible angiogenesis within the hypothalamic magnocellular nuclei of the adult rat: a potential role for neuronal vascular endothelial growth factor. BMC Neurosci 6:20. 10.1186/1471-2202-6-20 [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Althammer F, Eliava M, Grinevich V. 2021. Central and peripheral release of oxytocin: relevance of neuroendocrine and neurotransmitter actions for physiology and behavior. Handb Clin Neurol 180:25–44. 10.1016/B978-0-12-820107-7.00003-3 [DOI] [PubMed] [Google Scholar]
  7. Althammer F, Grinevich V. 2017. Diversity of oxytocin neurons: beyond magno- and parvocellular cell types? J Neuroendocrinol. 10.1111/jne.12549 [DOI] [PubMed]
  8. Amar AP, Weiss MH. 2003. Pituitary anatomy and physiology. Neurosurg Clin N Am 14(1):11–23. 10.1016/s1042-3680(02)00017-7 [DOI] [PubMed] [Google Scholar]
  9. Amico JA, Tenicela R, Johnston J, Robinson AG. 1983. A time-dependent peak of oxytocin exists in cerebrospinal fluid but not in plasma of humans. J Clin Endocrinol Metab 57(5):947–51. 10.1210/jcem-57-5-947 [DOI] [PubMed] [Google Scholar]
  10. Anderson JW, Washburn DL, Ferguson AV. 2000. Intrinsic osmosensitivity of subfornical organ neurons. Neuroscience 100(3):539–47. 10.1016/s0306-4522(00)00313-4 [DOI] [PubMed] [Google Scholar]
  11. Antunes-Rodrigues J, De Castro M, Elias LLK, Valença MM, McCann SM. 2004. Neuroendocrine control of body fluid metabolism. Physiol Rev 84(1):169–208. 10.1152/physrev.00017.2003 [DOI] [PubMed] [Google Scholar]
  12. Artman HG, Reppert SM, Perlow MJ, Swaminathan S, Oddie TH, Fisher DA. 1982. Characterization of the daily oxytocin rhythm in primate cerebrospinal fluid. J Neurosci 2(5):598–603. 10.1523/JNEUROSCI.02-05-00598.1982 [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Assinder SJ. 2022. The importance of experimental investigation of the peripheral oxytocin system. Methods Mol Biol 2384:1–27. 10.1007/978-1-0716-1759-5_1 [DOI] [PubMed] [Google Scholar]
  14. Bacyinski A, Xu M, Wang W, Hu J. 2017. The paravascular pathway for brain waste clearance: current understanding, significance and controversy. Front Neuroanat 11. https://www.frontiersin.org/articles/10.3389/fnana.2017.00101 [DOI] [PMC free article] [PubMed]
  15. Baertschi AJ, Vallet P, Baumann JB, Girard J. 1980. Neural lobe of pituitary modulates corticotropin release in the rat. Endocrinology 106(3):878–82. 10.1210/endo-106-3-878 [DOI] [PubMed] [Google Scholar]
  16. Bale TL, Dorsa DM, Johnston CA. 1995. Oxytocin receptor mRNA expression in the ventromedial hypothalamus during the estrous cycle. J Neurosci 15(7):5058–64. 10.1523/JNEUROSCI.15-07-05058.1995 [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Balment RJ, Brimble MJ, Forsling ML. 1980. Release of oxytocin induced by salt loading and its influence on renal excretion in the male rat. J Physiol 308:439–49. 10.1113/jphysiol.1980.sp013481 [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Balment RJ, Brimble MJ, Forsling ML, Kelly LP, Musabayane CT. 1986. A synergistic effect of oxytocin and vasopressin on sodium excretion in the neurohypophysectomized rat. J Physiol 381:453–64. 10.1113/jphysiol.1986.sp016338 [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Baudon A, Clauss Creusot E, Althammer F, Schaaf CP, Charlet A. 2022. Emerging role of astrocytes in oxytocin-mediated control of neural circuits and brain functions. Prog Neurobiol 217:102328. 10.1016/j.pneurobio.2022.102328 [DOI] [PubMed] [Google Scholar]
  20. Belin V, Moos F. 1986. Paired recordings from supraoptic and paraventricular oxytocin cells in suckled rats: recruitment and synchronization. J Physiol 377(1):369–90. 10.1113/jphysiol.1986.sp016192 [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Bergland RM, Page RB. 1978. Can the pituitary secrete directly to the brain? (Affirmative anatomical evidence). Endocrinology 102(5):1325–38. 10.1210/endo-102-5-1325 [DOI] [PubMed] [Google Scholar]
  22. Bertram R, Helena C, Gonzalez-Iglesias AE, Tabak J, Freeman ME. 2010. A tale of two rhythms: the emerging roles of oxytocin in rhythmic prolactin release. J Neuroendocrinol 22(7):778–84. 10.1111/j.1365-2826.2010.02012.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Beyer CE, Dwyer JM, Platt BJ, Neal S, Luo B, Ling H-P, and others. 2010. Angiotensin IV elevates oxytocin levels in the rat amygdala and produces anxiolytic-like activity through subsequent oxytocin receptor activation. Psychopharmacology 209(4):303–11. 10.1007/s00213-010-1791-1 [DOI] [PubMed] [Google Scholar]
  24. Blackmore KA, Jeong JK, Young CN. 2018. A novel oxytocin expressing microglia population in the brain subfornical organ. FASEB J 32(S1):598.9. 10.1096/fasebj.2018.32.1_supplement.598.9 [DOI] [Google Scholar]
  25. Blanco E, Pilgrim C, Vazquez R, Jirikowski GF. 1991. Plasticity of the interface between oxytocin neurons and the vasculature in late pregnant rats: an ultrastructural morphometric study. Acta Histochemica 91(2):165–72. 10.1016/S0065-1281(11)80272-2 [DOI] [PubMed] [Google Scholar]
  26. Blevins JE, Schwartz MW, Baskin DG. 2004. Evidence that paraventricular nucleus oxytocin neurons link hypothalamic leptin action to caudal brain stem nuclei controlling meal size. Am J Physiol Regul Integr Comp Physiol 287(1):R87–96. 10.1152/ajpregu.00604.2003 [DOI] [PubMed] [Google Scholar]
  27. Born J, Lange T, Kern W, McGregor GP, Bickel U, Fehm HL. 2002. Sniffing neuropeptides: a transnasal approach to the human brain. Nat Neurosci 5(6):514–6. 10.1038/nn0602-849 [DOI] [PubMed] [Google Scholar]
  28. Bowen MT. 2019. Does peripherally administered oxytocin enter the brain? Compelling new evidence in a long-running debate. Pharmacol Res 146:104325. 10.1016/j.phrs.2019.104325 [DOI] [PubMed] [Google Scholar]
  29. Bradbury MW, Cserr HF, Westrop RJ. 1981. Drainage of cerebral interstitial fluid into deep cervical lymph of the rabbit. Am J Physiol 240(4):F329–36. 10.1152/ajprenal.1981.240.4.F329 [DOI] [PubMed] [Google Scholar]
  30. Brett J, Schmidt AM, Yan SD, Zou YS, Weidman E, Pinsky D, and others. 1993. Survey of the distribution of a newly characterized receptor for advanced glycation end products in tissues. Am J Pathol 143(6):1699–712. [PMC free article] [PubMed] [Google Scholar]
  31. Brown CH. 2016. Magnocellular neurons and posterior pituitary function. Compr Physiol 6(4):1701–41. 10.1002/cphy.c150053 [DOI] [PubMed] [Google Scholar]
  32. Brown CH, Ludwig M, Tasker JG, Stern JE. 2020. Somato-dendritic vasopressin and oxytocin secretion in endocrine and autonomic regulation. J Neuroendocrinol 32(6):e12856. 10.1111/jne.12856 [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Brownstein MJ, Russell JT, Gainer H. 1980. Synthesis, transport, and release of posterior pituitary hormones. Science 207(4429):373–78. 10.1126/science.6153132 [DOI] [PubMed] [Google Scholar]
  34. Buijs RM. 1978. Intra- and extrahypothalamic vasopressin and oxytocin pathways in the rat. Cell Tissue Res 192(3):423–35. 10.1007/BF00212323 [DOI] [PubMed] [Google Scholar]
  35. Buma P, Nieuwenhuys R. 1987. Ultrastructural demonstration of oxytocin and vasopressin release sites in the neural lobe and median eminence of the rat by tannic acid and immunogold methods. Neurosci Lett 74(2):151–7. 10.1016/0304-3940(87)90141-8 [DOI] [PubMed] [Google Scholar]
  36. Busnelli M, Kleinau G, Muttenthaler M, Stoev S, Manning M, Bibic L, and others. 2016. Design and characterization of superpotent bivalent ligands targeting oxytocin receptor dimers via a channel-like structure. J Med Chem 59(15):7152–66. 10.1021/acs.jmedchem.6b00564 [DOI] [PubMed] [Google Scholar]
  37. Busnelli M, Saulière A, Manning M, Bouvier M, Galés C, Chini B. 2012. Functional selective oxytocin-derived agonists discriminate between individual g protein family subtypes. J Biol Chem 287(6):3617–29. 10.1074/jbc.M111.277178 [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Caldwell JD, Johns JM, Faggin BM, Senger MA, Pedersen CA. 1994. Infusion of an oxytocin antagonist into the medial preoptic area prior to progesterone inhibits sexual receptivity and increases rejection in female rats. Horm Behav 28(3):288–302. 10.1006/hbeh.1994.1024 [DOI] [PubMed] [Google Scholar]
  39. Camacho A, Phillips MI. 1981. Horseradish peroxidase study in rat of the neural connections of the organum vasculosum of the lamina terminalis. Neurosci Lett 25(3):201–4. 10.1016/0304-3940(81)90391-8 [DOI] [PubMed] [Google Scholar]
  40. Carey HM. 1959. Posterior pituitary oxytocin. J Obstet Gynaecol Br Emp 66:869–70. [PubMed] [Google Scholar]
  41. Cattaneo MG, Lucci G, Vicentini LM. 2009. Oxytocin stimulates in vitro angiogenesis via a Pyk-2/Src-dependent mechanism. Exp Cell Res 315(18):3210–9. 10.1016/j.yexcr.2009.06.022 [DOI] [PubMed] [Google Scholar]
  42. Chang HY, Morrow K, Bonacquisti E, Zhang W, Shah DK. 2018. Antibody pharmacokinetics in rat brain determined using microdialysis. mAbs 10(6):843–53. 10.1080/19420862.2018.1473910 [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Chang SWC, Barter JW, Ebitz RB, Watson KK, Platt ML. 2012. Inhaled oxytocin amplifies both vicarious reinforcement and self reinforcement in rhesus macaques (Macaca mulatta). Proc Natl Acad Sci U S A 109(3):959–64. 10.1073/pnas.1114621109 [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Chen S, Xu H, Dong S, Xiao L. 2022. Morpho-electric properties and diversity of oxytocin neurons in paraventricular nucleus of hypothalamus in female and male mice. J Neurosci 42(14):2885–904. 10.1523/JNEUROSCI.2494-21.2022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Chen XQ, Fawcett JR, Rahman YE, Ala TA, Frey WH, II. 1998. Delivery of nerve growth factor to the brain via the olfactory pathway. J Alzheimers Dis 1(1):35–44. 10.3233/jad-1998-1102 [DOI] [PubMed] [Google Scholar]
  46. Chini B, Verhage M, Grinevich V. 2017. The action radius of oxytocin release in the mammalian CNS: from single vesicles to behavior. Trends Pharmacol Sci 38(11):982–91. 10.1016/j.tips.2017.08.005 [DOI] [PubMed] [Google Scholar]
  47. Cloft HJ, Mitchell JA. 1994. Immunocytochemical detection of oxytocin in the supraependymal neuronal complex of the golden hamster. Brain Res 639(2):233–9. 10.1016/0006-8993(94)91735-3 [DOI] [PubMed] [Google Scholar]
  48. Conti F, Sertic S, Reversi A, Chini B. 2009. Intracellular trafficking of the human oxytocin receptor: evidence of receptor recycling via a Rab4/Rab5 “short cycle.” Am J Physiol Endocrinol Metab 296(3):E532–42. 10.1152/ajpendo.90590.2008 [DOI] [PubMed] [Google Scholar]
  49. Currie AR, Adamsons H, Van Dyke HB. 1960. Vasopressin and oxytocin in the posterior lobe of the pituitary of man. J Clin Endocrinol Metab 20:947–51. 10.1210/jcem-20-7-947 [DOI] [PubMed] [Google Scholar]
  50. Dabrowska J, Hazra R, Ahern TH, Guo J-D, McDonald AJ, Mascagni F, and others. 2011. Neuroanatomical evidence for reciprocal regulation of the corticotrophin-releasing factor and oxytocin systems in the hypothalamus and the bed nucleus of the stria terminalis of the rat: implications for balancing stress and affect. Psychoneuroendocrinology 36(9):1312–26. 10.1016/j.psyneuen.2011.03.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Dale HH. 1906. On some physiological actions of ergot. J Physiol 34(3):163–206. 10.1113/jphysiol.1906.sp001148 [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Davson H, Domer FR, Hollingsworth JR. 1973. The mechanism of drainage of the cerebrospinal fluid. Brain 96(2):329–36. 10.1093/brain/96.2.329 [DOI] [PubMed] [Google Scholar]
  53. Dayanithi G, Sabatier N, Widmer H. 2000. Intracellular calcium signalling in magnocellular neurones of the rat supraoptic nucleus: understanding the autoregulatory mechanisms. Exp Physiol 85 Spec No.:75S–84S. 10.1111/j.1469-445x.2000.tb00010.x [DOI] [PubMed]
  54. Devarajan K, Rusak B. 2004. Oxytocin levels in the plasma and cerebrospinal fluid of male rats: effects of circadian phase, light and stress. Neurosci Lett 367(2):144–7. 10.1016/j.neulet.2004.05.112 [DOI] [PubMed] [Google Scholar]
  55. Du W, Stern JE, Filosa JA. 2015. Neuronal-derived nitric oxide and somatodendritically released vasopressin regulate neurovascular coupling in the rat hypothalamic supraoptic nucleus. J Neurosci 35(13):5330–41. 10.1523/JNEUROSCI.3674-14.2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Duvernoy HM, Risold P-Y. 2007. The circumventricular organs: an atlas of comparative anatomy and vascularization. Brain Res Rev 56(1):119–47. 10.1016/j.brainresrev.2007.06.002 [DOI] [PubMed] [Google Scholar]
  57. Ebner K, Wotjak CT, Landgraf R, Engelmann M. 2000. A single social defeat experience selectively stimulates the release of oxytocin, but not vasopressin, within the septal brain area of male rats. Brain Res 872(1–2):87–92. 10.1016/s0006-8993(00)02464-1 [DOI] [PubMed] [Google Scholar]
  58. Egli M, Bertram R, Toporikova N, Sellix MT, Blanco W, Freeman ME. 2006. Prolactin secretory rhythm of mated rats induced by a single injection of oxytocin. Am J Physiol Endocrinol Metab 290(3):E566–72. 10.1152/ajpendo.00427.2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. El Assar De La Fuente M, Angulo Frutos J, Vallejo Fernán S, Peiró Vallejo C, Sánchez-Ferrer CF, Rodríguez-Mañas L. 2012. Mechanisms involved in the aging-induced vascular dysfunction. Front Physiol 3. 10.3389/fphys.2012.00132 [DOI] [PMC free article] [PubMed]
  60. Elands J, Barberis C, Jard S. 1988. [3H]-[Thr4,Gly7]OT: a highly selective ligand for central and peripheral OT receptors. Am J Physiol 254(1 pt 1):E31–8. 10.1152/ajpendo.1988.254.1.E31 [DOI] [PubMed] [Google Scholar]
  61. Eliava M, Melchior M, Knobloch-Bollmann HS, Wahis J, da Silva Gouveia M, Tang Y, and others. 2016. A new population of parvocellular oxytocin neurons controlling magnocellular neuron activity and inflammatory pain processing. Neuron 89(6):1291–304. 10.1016/j.neuron.2016.01.041 [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Engelmann M, Ebner K, Landgraf R, Wotjak CT. 2006. Effects of Morris water maze testing on the neuroendocrine stress response and intrahypothalamic release of vasopressin and oxytocin in the rat. Horm Behav 50(3):496–501. 10.1016/j.yhbeh.2006.04.009 [DOI] [PubMed] [Google Scholar]
  63. Engelmann M, Landgraf R, Wotjak CT. 2004. The hypothalamic-neurohypophysial system regulates the hypothalamic-pituitary-adrenal axis under stress: an old concept revisited. Front Neuroendocrinol 25(3):132–49. 10.1016/j.yfrne.2004.09.001 [DOI] [PubMed] [Google Scholar]
  64. Ermisch A, Rühle HJ, Landgraf R, Hess J. 1985. Blood-brain barrier and peptides. J Cereb Blood Flow Metab 5(3):350–7. 10.1038/jcbfm.1985.49 [DOI] [PubMed] [Google Scholar]
  65. Faghih MM, Sharp MK. 2018. Is bulk flow plausible in perivascular, paravascular and paravenous channels? Fluids Barriers CNS 15:17. 10.1186/s12987-018-0103-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Favaretto AL, Ballejo GO, Albuquerque-Araújo WI, Gutkowska J, Antunes-Rodrigues J, McCann SM. 1997. Oxytocin releases atrial natriuretic peptide from rat atria in vitro that exerts negative inotropic and chronotropic action. Peptides 18(9):1377–81. 10.1016/s0196-9781(97)00209-x [DOI] [PubMed] [Google Scholar]
  67. Fehm HL, Perras B, Smolnik R, Kern W, Born J. 2000. Manipulating neuropeptidergic pathways in humans: a novel approach to neuropharmacology? Eur J Pharmacol 405(1–3):43–54. 10.1016/s0014-2999(00)00540-9 [DOI] [PubMed] [Google Scholar]
  68. Ferguson AV, Kasting NW. 1987. Activation of subfornical organ efferents stimulates oxytocin secretion in the rat. Regul Pept 18(2):93–100. 10.1016/0167-0115(87)90039-5 [DOI] [PubMed] [Google Scholar]
  69. Ferguson AV, Latchford KJ, Samson WK. 2008. The paraventricular nucleus of the hypothalamus—a potential target for integrative treatment of autonomic dysfunction. Expert Opin Ther Targets 12(6):717–27. 10.1517/14728222.12.6.717 [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Ferguson JN, Young LJ, Hearn EF, Matzuk MM, Insel TR, Winslow JT. 2000. Social amnesia in mice lacking the oxytocin gene. Nat Genet 25(3):284–8. 10.1038/77040 [DOI] [PubMed] [Google Scholar]
  71. Freund-Mercier MJ, Stoeckel ME, Klein MJ. 1994. Oxytocin receptors on oxytocin neurones: histoautoradiographic detection in the lactating rat. J Physiol 480(Pt 1):155–61. 10.1113/jphysiol.1994.sp020349 [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Furube E, Mannari T, Morita S, Nishikawa K, Yoshida A, Itoh M, and others. 2014. VEGF-dependent and PDGF-dependent dynamic neurovascular reconstruction in the neurohypophysis of adult mice. J Endocrinol 222(1):161–79. 10.1530/JOE-14-0075 [DOI] [PubMed] [Google Scholar]
  73. Gan Y, Holstein-Rønsbo S, Nedergaard M, Boster KAS, Thomas JH, Kelley DH. 2023. Perivascular pumping of cerebrospinal fluid in the brain with a valve mechanism. J R Soc Interface 20(206):20230288. 10.1098/rsif.2023.0288 [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Gao FB. 1998. Messenger RNAs in dendrites: localization, stability, and implications for neuronal function. Bioessays 20(1):70–8. . [DOI] [PubMed] [Google Scholar]
  75. Gerasimenko M, Lopatina O, Munesue S, Harashima A, Yokoyama S, Yamamoto Y, and others. 2021. Receptor for advanced glycation end-products (RAGE) plays a critical role in retrieval behavior of mother mice at early postpartum. Physiol Behav 235:113395. 10.1016/j.physbeh.2021.113395 [DOI] [PubMed] [Google Scholar]
  76. Ghazy AA, Soliman OA, Elbahnasi AI, Alawy AY, Mansour AM, Gowayed MA. 2023. Role of oxytocin in different neuropsychiatric, neurodegenerative, and neurodevelopmental disorders. Rev Physiol Biochem Pharmacol 186:95–134. 10.1007/112_2022_72 [DOI] [PubMed] [Google Scholar]
  77. Giles ME, Fernley RT, Nakamura Y, Moeller I, Aldred GP, Ferraro T, and others. 1999. Characterization of a specific antibody to the rat angiotensin II AT1 receptor. J Histochem Cytochem 47(4):507–16. 10.1177/002215549904700409 [DOI] [PubMed] [Google Scholar]
  78. Gimpl G, Fahrenholz F. 2001. The oxytocin receptor system: structure, function, and regulation. Physiol Rev 81(2):629–83. 10.1152/physrev.2001.81.2.629 [DOI] [PubMed] [Google Scholar]
  79. Gjerde IG, Rognes ME, Sánchez AL. 2023. The directional flow generated by peristalsis in perivascular networks: theoretical and numerical reduced-order descriptions. J Appl Phys 134(17):174701. 10.1063/5.0160334 [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Goren O, Adorján I, Kálmán M. 2006. Heterogeneous occurrence of aquaporin-4 in the ependyma and in the circumventricular organs in rat and chicken. Anat Embryol (Berl) 211(2):155–72. 10.1007/s00429-005-0067-8 [DOI] [PubMed] [Google Scholar]
  81. Griffin GD, Ferri-Kolwicz SL, Reyes BAS, Van Bockstaele EJ, Flanagan-Cato LM. 2010. Ovarian hormone-induced reorganization of oxytocin-labeled dendrites and synapses lateral to the hypothalamic ventromedial nucleus in female rats. J Comp Neurol 518(22):4531–45. 10.1002/cne.22470 [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Grinevich V, Knobloch-Bollmann HS, Eliava M, Busnelli M, Chini B. 2016. Assembling the puzzle: pathways of oxytocin signaling in the brain. Biol Psychiatry 79(3):155–64. 10.1016/j.biopsych.2015.04.013 [DOI] [PubMed] [Google Scholar]
  83. Gutkowska J, Jankowski M. 2012. Oxytocin revisited: its role in cardiovascular regulation. J Neuroendocrinol 24(4):599–608. 10.1111/j.1365-2826.2011.02235.x [DOI] [PubMed] [Google Scholar]
  84. Gutkowska J, Jankowski M, Lambert C, Mukaddam-Daher S, Zingg HH, McCann SM. 1997. Oxytocin releases atrial natriuretic peptide by combining with oxytocin receptors in the heart. Proc Natl Acad Sci U S A 94(21):11704–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Gutkowska J, Jankowski M, Mukaddam-Daher S, McCann SM. 2000. Oxytocin is a cardiovascular hormone. Braz J Med Biol Res 33(6):625–33. 10.1590/s0100-879x2000000600003 [DOI] [PubMed] [Google Scholar]
  86. Hadaczek P, Yamashita Y, Mirek H, Tamas L, Bohn MC, Noble C, and others. 2006. The “perivascular pump” driven by arterial pulsation is a powerful mechanism for the distribution of therapeutic molecules within the brain. Mol Ther 14(1):69–78. 10.1016/j.ymthe.2006.02.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Han J, Gnatenco C, Sladek CD, Kim D. 2003. Background and tandem-pore potassium channels in magnocellular neurosecretory cells of the rat supraoptic nucleus. J Physiol 546(Pt 3):625–39. 10.1113/jphysiol.2002.032094 [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Hatton GI. 1988. Pituicytes, glia and control of terminal secretion. The Journal of experimental biology, 139, 67–79. 10.1242/jeb.139.1.67 [DOI] [PubMed] [Google Scholar]
  89. Hatton GI, Perlmutter LS, Salm AK, Tweedle CD. 1984. Dynamic neuronal-glial interactions in hypothalamus and pituitary: implications for control of hormone synthesis and release. Peptides 5(Suppl 1):121–38. 10.1016/0196-9781(84)90271-7 [DOI] [PubMed] [Google Scholar]
  90. Hawkes CA, Jayakody N, Johnston DA, Bechmann I, Carare RO. 2014. Failure of perivascular drainage of β-amyloid in cerebral amyloid angiopathy. Brain Pathol 24(4):396–403. 10.1111/bpa.12159 [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Herkenham M. 1987. Mismatches between neurotransmitter and receptor localizations in brain: observations and implications. Neuroscience 23(1):1–38. 10.1016/0306-4522(87)90268-5 [DOI] [PubMed] [Google Scholar]
  92. Hicks A-I, Barad Z, Sobrero A, Lean G, Jacob-Tomas S, Yang J, and others. 2020. Effects of salt loading on the organisation of microtubules in rat magnocellular vasopressin neurones. J Neuroendocrinol 32(2):e12817. 10.1111/jne.12817 [DOI] [PubMed] [Google Scholar]
  93. Higa KT, Mori E, Viana FF, Morris M, Michelini LC. 2002. Baroreflex control of heart rate by oxytocin in the solitary-vagal complex. Am J Physiol Regul Integr Comp Physiol 282:R537–45. [DOI] [PubMed] [Google Scholar]
  94. Higashida H, Furuhara K, Yamauchi A-M, Deguchi K, Harashima A, Munesue S, and others. 2017. Intestinal transepithelial permeability of oxytocin into the blood is dependent on the receptor for advanced glycation end products in mice. Scientific Reports 7(1):7883. 10.1038/s41598-017-07949-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Hirasawa M, Kombian SB, Pittman QJ. 2001. Oxytocin retrogradely inhibits evoked, but not miniature, EPSCs in the rat supraoptic nucleus: role of N- and P/Q-type calcium channels. J Physiol 532(Pt 3):595–607. 10.1111/j.1469-7793.2001.0595e.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Hirasawa M, Schwab Y, Natah S, Hillard CJ, Mackie K, Sharkey KA, and others. 2004. Dendritically released transmitters cooperate via autocrine and retrograde actions to inhibit afferent excitation in rat brain. J Physiol 559(Pt 2):611–24. 10.1113/jphysiol.2004.066159 [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Hladky SB, Barrand MA. 2016. Fluid and ion transfer across the blood-brain and blood-cerebrospinal fluid barriers; a comparative account of mechanisms and roles. Fluids Barriers CNS 13(1):19. 10.1186/s12987-016-0040-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Hofer H. 1958. Zur Morphologie der circumventriculären Organe des Zwischenhirns der Säugetiere. Verhandlungen Der Deutschen Zoologischen Gesellschaft 55:202–55. [Google Scholar]
  99. Hosoya Y, Sugiura Y, Okado N, Loewy AD, Kohno K. 1991. Descending input from the hypothalamic paraventricular nucleus to sympathetic preganglionic neurons in the rat. Exp Brain Res 85(1):10–20. 10.1007/BF00229982 [DOI] [PubMed] [Google Scholar]
  100. Hrabovszky E, Liposits Z. 2008. Novel aspects of glutamatergic signalling in the neuroendocrine system. J Neuroendocrinol 20(6):743–51. 10.1111/j.1365-2826.2008.01719.x [DOI] [PubMed] [Google Scholar]
  101. Ichimiya Y, Emson PC, Shaw FD. 1988. Localization of vasopressin mRNA-containing neurones in the hypothalamus of the monkey. Brain Res 464(1):81–5. 10.1016/0169-328x(88)90022-8 [DOI] [PubMed] [Google Scholar]
  102. Iliff JJ, Wang M, Liao Y, Plogg BA, Peng W, Gundersen GA, and others. 2012. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid β. Sci Transl Med 4(147):147ra111. 10.1126/scitranslmed.3003748 [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Iliff JJ, Wang M, Zeppenfeld DM, Venkataraman A, Plog BA, Liao Y, and others. 2013. Cerebral arterial pulsation drives paravascular CSF–interstitial fluid exchange in the murine brain. J Neurosci 33(46):18190–9. 10.1523/JNEUROSCI.1592-13.2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Illum L. 2000. Transport of drugs from the nasal cavity to the central nervous system. Eur J Pharm Sci 11(1):1–18. 10.1016/s0928-0987(00)00087-7 [DOI] [PubMed] [Google Scholar]
  105. Ineichen BV, Okar SV, Proulx ST, Engelhardt B, Lassmann H, Reich DS. 2022. Perivascular spaces and their role in neuroinflammation. Neuron 110(21):3566–81. 10.1016/j.neuron.2022.10.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Ino D, Tanaka Y, Hibino H, Nishiyama M. 2022. A fluorescent sensor for real-time measurement of extracellular oxytocin dynamics in the brain. Nature Methods 19(10):1286–94. 10.1038/s41592-022-01597-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Iwasaki M, Lefevre A, Althammer F, Clauss Creusot E, Łąpieś O, Petitjean H, and others. 2023. An analgesic pathway from parvocellular oxytocin neurons to the periaqueductal gray in rats. Nat Commun 14(1):1066. 10.1038/s41467-023-36641-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Jankowski M, Hajjar F, Kawas SA, Mukaddam-Daher S, Hoffman G, McCann SM, and others. 1998. Rat heart: a site of oxytocin production and action. Proc Natl Acad Sci U S A 95(24):14558–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Jeong JK, Dow SA, Young CN. 2021. Sensory circumventricular organs, neuroendocrine control, and metabolic regulation. Metabolites 11(8):494. 10.3390/metabo11080494 [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Jessen NA, Munk ASF, Lundgaard I, Nedergaard M. 2015. The glymphatic system: a beginner’s guide. Neurochem Res 40(12):2583–99. 10.1007/s11064-015-1581-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Jin D, Liu H-X, Hirai H, Torashima T, Nagai T, Lopatina O, and others. 2007. CD38 is critical for social behaviour by regulating oxytocin secretion. Nature 446(7131):41–5. 10.1038/nature05526 [DOI] [PubMed] [Google Scholar]
  112. Jirikowski GF. 2019. Diversity of central oxytocinergic projections. Cell Tissue Res 375(1):41–8. 10.1007/s00441-018-2960-5 [DOI] [PubMed] [Google Scholar]
  113. Jirikowski GF, Caldwell JD, Pedersen CA, Stumpf WE. 1988. Estradiol influences oxytocin-immunoreactive brain systems. Neuroscience 25(1):237–48. 10.1016/0306-4522(88)90022-x [DOI] [PubMed] [Google Scholar]
  114. Jirikowski GF, Ochs SD, Caldwell JD. 2018. Oxytocin and steroid actions. Curr Top Behav Neurosci 35:77–95. 10.1007/7854_2017_9 [DOI] [PubMed] [Google Scholar]
  115. Jones PM, Robinson IC. 2008. Differential clearance of neurophysin and neurohypophysial peptides from the cerebrospinal fluid in conscious guinea pigs. Neuroendocrinology 34(4):297–302. 10.1159/000123316 [DOI] [PubMed] [Google Scholar]
  116. Ju G, Ma D, Duan XQ. 1992. Third ventricular subependymal oxytocin-like immunoreactive neuronal plexus in the rat. Brain Res Bull 28(6):887–96. 10.1016/0361-9230(92)90209-G [DOI] [PubMed] [Google Scholar]
  117. Juranek J, Mukherjee K, Kordas B, Załęcki M, Korytko A, Zglejc-Waszak K, and others. 2022. Role of RAGE in the pathogenesis of neurological disorders. Neurosci Bull 38(10):1248–62. 10.1007/s12264-022-00878-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Jurek B, Neumann ID. 2018. The oxytocin receptor: from intracellular signaling to behavior. Physiol Rev 98(3):1805–908. 10.1152/physrev.00031.2017 [DOI] [PubMed] [Google Scholar]
  119. Kaczmarek LK, Zhang Y. 2017. Kv3 channels: enablers of rapid firing, neurotransmitter release, and neuronal endurance. Physiol Rev 97(4):1431–68. 10.1152/physrev.00002.2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Kadekaro M, Summy-Long JY. 2000. Centrally produced nitric oxide and the regulation of body fluid and blood pressure homeostases. Clin Exp Pharmacol Physiol 27(5–6):450–9. 10.1046/j.1440-1681.2000.03264.x [DOI] [PubMed] [Google Scholar]
  121. Kadowaki K, Kishimoto J, Leng G, Emson PC. 1994. Up-regulation of nitric oxide synthase (NOS) gene expression together with NOS activity in the rat hypothalamo-hypophysial system after chronic salt loading: evidence of a neuromodulatory role of nitric oxide in arginine vasopressin and oxytocin secretion. Endocrinology 134(3):1011–7. 10.1210/endo.134.3.7509733 [DOI] [PubMed] [Google Scholar]
  122. Kamei I. 1986. The role of oxytocin cells around the blood vessels in the rat hypothalamus: immunohistochemical study. Acta Physiol Scand Suppl 552:25–8. [PubMed] [Google Scholar]
  123. Kawano H. 2017. Synaptic contact between median preoptic neurons and subfornical organ neurons projecting to the paraventricular hypothalamic nucleus. Exp Brain Res 235(4):1053–62. 10.1007/s00221-016-4862-7 [DOI] [PubMed] [Google Scholar]
  124. Kedarasetti RT, Drew PJ, Costanzo F. 2020. Arterial pulsations drive oscillatory flow of CSF but not directional pumping. Scientific Reports 10(1):10102. 10.1038/s41598-020-66887-w [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Kedarasetti RT, Turner KL, Echagarruga C, Gluckman BJ, Drew PJ, Costanzo F. 2020. Functional hyperemia drives fluid exchange in the paravascular space. Fluids Barriers CNS 17(1):52. 10.1186/s12987-020-00214-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Kendrick KM, Keverne EB, Baldwin BA. 1987. Intracerebroventricular oxytocin stimulates maternal behaviour in the sheep. Neuroendocrinology 46(1):56–61. 10.1159/000124796 [DOI] [PubMed] [Google Scholar]
  127. Kim E, Van Reet J, Yoo S-S. 2023. Cerebrospinal fluid solute transport associated with sensorimotor brain activity in rodents. Scientific Reports 13(1):17002. 10.1038/s41598-023-43920-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Kim SH, MacIntyre DA, Hanyaloglu AC, Blanks AM, Thornton S, Bennett PR, and others. 2016. The oxytocin receptor antagonist, Atosiban, activates pro-inflammatory pathways in human amnion via Gαi signalling. Mol Cell Endocrinol 420:11–23. 10.1016/j.mce.2015.11.012 [DOI] [PubMed] [Google Scholar]
  129. Kimura T, Minai K, Matsui K, Mouri T, Sato T, Yoshinaga K, and others. 1976. Effect of various states of hydration on plasma ADH and renin in man. J Clin Endocrinol Metab 42(1):79–87. 10.1210/jcem-42-1-79 [DOI] [PubMed] [Google Scholar]
  130. Kirchner MK, Althammer F, Donaldson KJ, Cox DN, Stern JE. 2023. Changes in neuropeptide large dense core vesicle trafficking dynamics contribute to adaptive responses to a systemic homeostatic challenge. iScience 26(11):108243. 10.1016/j.isci.2023.108243 [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Klockars OA, Waas JR, Klockars A, Levine AS, Olszewski PK. 2017. Neural basis of ventromedial hypothalamic oxytocin-driven decrease in appetite. Neuroscience 366:54–61. 10.1016/j.neuroscience.2017.10.008 [DOI] [PubMed] [Google Scholar]
  132. Knobloch HS, Charlet A, Hoffmann LC, Eliava M, Khrulev S, Cetin AH, and others. 2012. Evoked axonal oxytocin release in the central amygdala attenuates fear response. Neuron 73(3):553–66. 10.1016/j.neuron.2011.11.030 [DOI] [PubMed] [Google Scholar]
  133. Kojima S, Stewart RA, Demas GE, Alberts JR. 2012. Maternal contact differentially modulates central and peripheral oxytocin in rat pups during a brief regime of mother-pup interaction that induces a filial huddling preference. J Neuroendocrinol 24(5):831–40. 10.1111/j.1365-2826.2012.02280.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Kolaj M, Renaud LP. 2010. Metabotropic glutamate receptors in median preoptic neurons modulate neuronal excitability and glutamatergic and GABAergic inputs from the subfornical organ. J Neurophysiol 103(2):1104–13. 10.1152/jn.00808.2009 [DOI] [PubMed] [Google Scholar]
  135. Korogod SM, Stern JE, Cymbalyuk GS. 2023. Microgeometrical dendritic factors predict electrical decoupling between somatic and dendritic compartments in magnocellular neurosecretory neurons. Front Cell Neurosci 17:1125029. 10.3389/fncel.2023.1125029 [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Krisch B. 1986. Ultrastructure of regulatory neuroendocrine neurons and functionally related structures. In: Ganten D, Pfaff D, editors. Morphology of hypothalamus and its connections. New York: Springer. p. 251–90. 10.1007/978-3-642-71461-0_8 [DOI] [Google Scholar]
  137. Krolewski DM, Medina A, Kerman IA, Bernard R, Burke S, Thompson RC, and others. 2010. Expression patterns of corticotropin-releasing factor, arginine vasopressin, histidine decarboxylase, melanin-concentrating hormone, and orexin genes in the human hypothalamus. J Comp Neurol 518(22):4591–611. 10.1002/cne.22480 [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Landgraf R, Ludwig M. 1991. Vasopressin release within the supraoptic and paraventricular nuclei of the rat brain: osmotic stimulation via microdialysis. Brain Res 558(2):191–6. 10.1016/0006-8993(91)90768-q [DOI] [PubMed] [Google Scholar]
  139. Landgraf R, Neumann ID. 2004. Vasopressin and oxytocin release within the brain: a dynamic concept of multiple and variable modes of neuropeptide communication. Front Neuroendocrinol 25(3):150–76. 10.1016/j.yfrne.2004.05.001 [DOI] [PubMed] [Google Scholar]
  140. Landgraf R, Neumann I, Schwarzberg H. 1988. Central and peripheral release of vasopressin and oxytocin in the conscious rat after osmotic stimulation. Brain Res 457(2):219–25. 10.1016/0006-8993(88)90689-0 [DOI] [PubMed] [Google Scholar]
  141. Lee MR, Scheidweiler KB, Diao XX, Akhlaghi F, Cummins A, Huestis MA, and others. 2018. Oxytocin by intranasal and intravenous routes reaches the cerebrospinal fluid in rhesus macaques: determination using a novel oxytocin assay. Mol Psychiatry 23(1):115–22. 10.1038/mp.2017.27 [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Lee SW, Kim Y-B, Kim JS, Kim WB, Kim YS, Han HC, and others. 2015. GABAergic inhibition is weakened or converted into excitation in the oxytocin and vasopressin neurons of the lactating rat. Mol Brain 8:34. 10.1186/s13041-015-0123-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Leerach N, Harashima A, Munesue S, Kimura K, Oshima Y, Goto H, and others. 2021. Glycation reaction and the role of the receptor for advanced glycation end-products in immunity and social behavior. Glycoconj J 38(3):303–10. 10.1007/s10719-020-09956-6 [DOI] [PubMed] [Google Scholar]
  144. Leng G, Sabatier N. 2016. Measuring oxytocin and vasopressin: bioassays, immunoassays and random numbers. J Neuroendocrinol 28(10):10.1111/jne.12413. 10.1111/jne.12413 [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Levasseur G, Baly C, Grébert D, Durieux D, Salesse R, Caillol M. 2004. Anatomical and functional evidence for a role of arginine-vasopressin (AVP) in rat olfactory epithelium cells. Eur J Neurosci 20(3):658–70. 10.1111/j.1460-9568.2004.03516.x [DOI] [PubMed] [Google Scholar]
  146. Lewis EM, Stein-O’Brien GL, Patino AV, Nardou R, Grossman CD, Brown M, and others. 2020. Parallel social information processing circuits are differentially impacted in autism. Neuron 108(4):659–75.e6. 10.1016/j.neuron.2020.10.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Li C, Kim K. 2008. Neuropeptides. In: WormBook: the online review of C. elegans biology. 10.1895/wormbook.1.142.1 [DOI] [PMC free article] [PubMed]
  148. Li H, Jiang T, An S, Xu M, Gou L, Ren B, and others. 2024. Single-neuron projectomes of mouse paraventricular hypothalamic nucleus oxytocin neurons reveal mutually exclusive projection patterns. Neuron 112(7):1081–99.e7. 10.1016/j.neuron.2023.12.022 [DOI] [PubMed] [Google Scholar]
  149. Liu Z, Huang S, Hu P, Zhou H. 2018. The role of autophagy in advanced glycation end product-induced proliferation and migration in rat vascular smooth muscle cells. Iran J Basic Med Sci 21(6):634–8. 10.22038/IJBMS.2018.20266.5305 [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Lopatina O, Liu HX, Amina S, Hashii M, Higashida H. 2010. Oxytocin-induced elevation of ADP-ribosyl cyclase activity, cyclic ADP-ribose or Ca(2+) concentrations is involved in autoregulation of oxytocin secretion in the hypothalamus and posterior pituitary in male mice. Neuropharmacology 58(1):50–5. 10.1016/j.neuropharm.2009.06.012 [DOI] [PubMed] [Google Scholar]
  151. Loth E, Poline JB, Thyreau B, Jia T, Tao C, Lourdusamy A, and others. 2014. Oxytocin receptor genotype modulates ventral striatal activity to social cues and response to stressful life events. Biol Psychiatry 76(5):367–76. 10.1016/j.biopsych.2013.07.043 [DOI] [PubMed] [Google Scholar]
  152. Ludwig M, Callahan MF, Neumann I, Landgraf R, Morris M. 1994. Systemic osmotic stimulation increases vasopressin and oxytocin release within the supraoptic nucleus. J Neuroendocrinol 6(4):369–73. 10.1111/j.1365-2826.1994.tb00595.x [DOI] [PubMed] [Google Scholar]
  153. Ludwig M, Leng G. 2006. Dendritic peptide release and peptide-dependent behaviours. Nat Rev Neurosci 7(2):126–36. 10.1038/nrn1845 [DOI] [PubMed] [Google Scholar]
  154. Mack SO, Kc P, Wu M, Coleman BR, Tolentino-Silva FP, Haxhiu MA. 2002. Paraventricular oxytocin neurons are involved in neural modulation of breathing. J Appl Physiol (1985) 92(2):826–34. 10.1152/japplphysiol.00839.2001 [DOI] [PubMed] [Google Scholar]
  155. MacLean EL, Wilson SR, Martin WL, Davis JM, Nazarloo HP, Carter CS. 2019. Challenges for measuring oxytocin: the blind men and the elephant? Psychoneuroendocrinology 107:225–31. 10.1016/j.psyneuen.2019.05.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Maejima Y, Rita RS, Santoso P, Aoyama M, Hiraoka Y, Nishimori K, and others. 2015. Nasal oxytocin administration reduces food intake without affecting locomotor activity and glycemia with c-Fos induction in limited brain areas. Neuroendocrinology 101(1):35–44. 10.1159/000371636 [DOI] [PubMed] [Google Scholar]
  157. Maejima Y, Yokota S, Ono T, Yu Z, Yamachi M, Hidema S, and others. 2022. Identification of oxytocin expression in human and murine microglia. Prog Neuropsychopharmacol Biol Psychiatry 119:110600. 10.1016/j.pnpbp.2022.110600 [DOI] [PubMed] [Google Scholar]
  158. Manjila SB, Betty R, Kim Y. 2022. Missing pieces in decoding the brain oxytocin puzzle: functional insights from mouse brain wiring diagrams. Front Neurosci 16:1044736. 10.3389/fnins.2022.1044736 [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Manning M, Misicka A, Olma A, Bankowski K, Stoev S, Chini B, and others. 2012. Oxytocin and vasopressin agonists and antagonists as research tools and potential therapeutics. J Neuroendocrinol 24(4):609–28. 10.1111/j.1365-2826.2012.02303.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Marsh N, Marsh AA, Lee MR, Hurlemann R. 2021. Oxytocin and the neurobiology of prosocial behavior. Neuroscientist 27(6):604–19. 10.1177/1073858420960111 [DOI] [PMC free article] [PubMed] [Google Scholar]
  161. Martins D, Gabay AS, Mehta M, Paloyelis Y. 2020. Salivary and plasmatic oxytocin are not reliable trait markers of the physiology of the oxytocin system in humans. eLife 9:e62456. 10.7554/eLife.62456 [DOI] [PMC free article] [PubMed]
  162. Martins D, Paduraru M, Paloyelis Y. 2022. Heterogeneity in response to repeated intranasal oxytocin in schizophrenia and autism spectrum disorders: a meta-analysis of variance. Br J Pharmacol 179(8):1525–43. 10.1111/bph.15451 [DOI] [PubMed] [Google Scholar]
  163. Martins DA, Mazibuko N, Zelaya F, Vasilakopoulou S, Loveridge J, Oates A, and others. 2020. Effects of route of administration on oxytocin-induced changes in regional cerebral blood flow in humans. Nat Commun 11(1):1160. 10.1038/s41467-020-14845-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. Matsuda T, Hiyama TY, Niimura F, Matsusaka T, Fukamizu A, Kobayashi K, and others. 2017. Distinct neural mechanisms for the control of thirst and salt appetite in the subfornical organ. Nat Neurosci 20(2):230–41. 10.1038/nn.4463 [DOI] [PubMed] [Google Scholar]
  165. Matsumoto H, Nagasaka T, Hattori A, Rogi T, Tsuruoka N, Mizutani S, and others. 2001. Expression of placental leucine aminopeptidase/oxytocinase in neuronal cells and its action on neuronal peptides. Eur J Biochem 268(11):3259–66. 10.1046/j.1432-1327.2001.02221.x [DOI] [PubMed] [Google Scholar]
  166. Matsuura T, Kawasaki M, Suzuki H, Fujitani T, Baba K, Nishimura H, and others. 2023. Nitric oxide synthase contributes to the maintenance of LTP in the oxytocin–mRFP1 neuron of the rat hypothalamus. J Neuroendocrinol 35(10):e13340. 10.1111/jne.13340 [DOI] [PubMed] [Google Scholar]
  167. McComb JG. 1983. Recent research into the nature of cerebrospinal fluid formation and absorption. J Neurosurg 59(3):369–83. 10.3171/jns.1983.59.3.0369 [DOI] [PubMed] [Google Scholar]
  168. McCullough ME, Churchland PS, Mendez AJ. 2013. Problems with measuring peripheral oxytocin: can the data on oxytocin and human behavior be trusted? Neurosci Biobehav Rev 37(8):1485–92. 10.1016/j.Neubiorev.2013.04.018 [DOI] [PubMed] [Google Scholar]
  169. McKinley MJ, Bicknell RJ, Hards D, McAllen RM, Vivas L, Weisinger RS, and others. 1992. Efferent neural pathways of the lamina terminalis subserving osmoregulation. Prog Brain Res 91:395–402. 10.1016/s0079-6123(08)62358-4 [DOI] [PubMed] [Google Scholar]
  170. McKinley MJ, McAllen RM, Davern P, Giles ME, Penschow J, Sunn N, and others. 2003. The sensory circumventricular organs of the mammalian brain. Adv Anat Embryol Cell Biol 172:1–122. 10.1007/978-3-642-55532-9 [DOI] [PubMed] [Google Scholar]
  171. Mecawi AS, Varanda WA, da Silva MP. 2022. Osmoregulation and the hypothalamic supraoptic nucleus: from genes to functions. Front Physiol 13. https://www.frontiersin.org/articles/10.3389/fphys.2022.887779 [DOI] [PMC free article] [PubMed]
  172. Meddle SL, Bishop VR, Gkoumassi E, van Leeuwen FW, Douglas AJ. 2007. Dynamic changes in oxytocin receptor expression and activation at parturition in the rat brain. Endocrinology 148(10):5095–104. 10.1210/en.2007-0615 [DOI] [PubMed] [Google Scholar]
  173. Meddle SL, Leng G, Selvarajah JR, Bicknell RJ, Russell JA. 2000. Direct pathways to the supraoptic nucleus from the brainstem and the main olfactory bulb are activated at parturition in the rat. Neuroscience 101(4):1013–21. 10.1016/s0306-4522(00)00300-6 [DOI] [PubMed] [Google Scholar]
  174. Menon R, Grund T, Zoicas I, Althammer F, Fiedler D, Biermeier V, and others. 2018. Oxytocin signaling in the lateral septum prevents social fear during lactation. Curr Biol 28(7):1066–78.e6. 10.1016/j.cub.2018.02.044 [DOI] [PubMed] [Google Scholar]
  175. Mens WBJ, Witter A, Van Wimersma Greidanus TB. 1983. Penetration of neurohypophyseal hormones from plasma into cerebrospinal fluid (CSF): half-times of disappearance of these neuropeptides from CSF. Brain Res 262(1):143–9. 10.1016/0006-8993(83)90478-X [DOI] [PubMed] [Google Scholar]
  176. Mitre M, Marlin BJ, Schiavo JK, Morina E, Norden SE, Hackett TA, and others. 2016. A distributed network for social cognition enriched for oxytocin receptors. J Neurosci 36(8):2517–35. 10.1523/JNEUROSCI.2409-15.2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  177. Miyata S, Hatton GI. 2002. Activity-related, dynamic neuron-glial interactions in the hypothalamo-neurohypophysial system. Microsc Res Tech 56(2):143–57. 10.1002/jemt.10012 [DOI] [PubMed] [Google Scholar]
  178. Miyata S, Itoh T, Matsushima O, Nakashima T, Kiyohara T. 1994. Not only osmotic stress but also repeated restraint stress causes structural plasticity in the supraoptic nucleus of the rat hypothalamus. Brain Res Bull 33(6):669–75. 10.1016/0361-9230(94)90231-3 [DOI] [PubMed] [Google Scholar]
  179. Møller M, Busch JR, Jacobsen C, Lundemose SB, Lynnerup N, Rath MF, and others. 2018. The accessory magnocellular neurosecretory system of the rostral human hypothalamus. Cell Tissue Res 373(2):487–98. 10.1007/s00441-018-2818-x [DOI] [PubMed] [Google Scholar]
  180. Momenabadi S, Vafaei AA, Bandegi AR, Zahedi-Khorasani M, Mazaheri Z, Vakili A. 2020. Oxytocin reduces brain injury and maintains blood-brain barrier integrity after ischemic stroke in mice. Neuromolecular Med 22(4):557–71. 10.1007/s12017-020-08613-3 [DOI] [PubMed] [Google Scholar]
  181. Monte OD, Noble PL, Turchi J, Cummins A, Averbeck BB. 2014. CSF and blood oxytocin concentration changes following intranasal delivery in macaque. PLoS One 9(8):e103677. 10.1371/journal.pone.0103677 [DOI] [PMC free article] [PubMed] [Google Scholar]
  182. Moos F, Marganiec A, Fontanaud P, Guillou-Duvoid A, Alonso G. 2004. Synchronization of oxytocin neurons in suckled rats: possible role of bilateral innervation of hypothalamic supraoptic nuclei by single medullary neurons. Eur J Neurosci 20(1):66–78. 10.1111/j.0953-816X.2004.03455.x [DOI] [PubMed] [Google Scholar]
  183. Moos F, Poulain DA, Rodriguez F, Guerné Y, Vincent JD, Richard P. 1989. Release of oxytocin within the supraoptic nucleus during the milk ejection reflex in rats. Exp Brain Res 76(3):593–602. 10.1007/BF00248916 [DOI] [PubMed] [Google Scholar]
  184. Morin V, Del Castillo JRE, Authier S, Ybarra N, Otis C, Gauvin D, and others. 2008. Evidence for non-linear pharmacokinetics of oxytocin in anesthetizetized rat. J Pharm Pharm Sci 11(4):12–24. 10.18433/j3pk5x [DOI] [PubMed] [Google Scholar]
  185. Morris JF, Budd TC, Epton MJ, Ma D, Pow DV, Wang H. 1998. Functions of the perikaryon and dendrites in magnocellular vasopressin-secreting neurons: new insights from ultrastructural studies. Prog Brain Res 119:21–30. 10.1016/s0079-6123(08)61559-9 [DOI] [PubMed] [Google Scholar]
  186. Morris JF, Pow DV. 1991. Widespread release of peptides in the central nervous system: quantitation of tannic acid–captured exocytoses. Anat Rec 231(4):437–45. 10.1002/ar.1092310406 [DOI] [PubMed] [Google Scholar]
  187. Morris M, Barnard RR, Jr, Sain LE. 1984. Osmotic mechanisms regulating cerebrospinal fluid vasopressin and oxytocin in the conscious rat. Neuroendocrinology 39(5):377–83. 10.1159/000124008 [DOI] [PubMed] [Google Scholar]
  188. Mullier A, Bouret SG, Prevot V, Dehouck B. 2010. Differential distribution of tight junction proteins suggests a role for tanycytes in blood-hypothalamus barrier regulation in the adult mouse brain. J Comp Neurol 518(7):943–62. 10.1002/cne.22273 [DOI] [PMC free article] [PubMed] [Google Scholar]
  189. Munesue S-I, Liang M, Harashima A, Zhong J, Furuhara K, Boitsova EB, and others. 2021. Transport of oxytocin to the brain after peripheral administration by membrane-bound or soluble forms of receptors for advanced glycation end-products. J Neuroendocrinol 33(3):e12963. 10.1111/jne.12963 [DOI] [PubMed] [Google Scholar]
  190. Nakamura K, Karasawa K, Yasui M, Nuriya M. 2022. Probing the spatiotemporal dynamics of oxytocin in the brain tissue using a simple peptide alkyne-tagging approach. Analytical chemistry 94(35):11990–8. 10.1021/acs.analchem.2c00452 [DOI] [PubMed] [Google Scholar]
  191. Negoro H, Higuchi T, Tadokoro Y, Honda K. 1988. Osmoreceptor mechanism for oxytocin release in the rat. Jpn J Physiol 38(1):19–31. 10.2170/jjphysiol.38.19 [DOI] [PubMed] [Google Scholar]
  192. Neumann ID. 2007. Stimuli and consequences of dendritic release of oxytocin within the brain. Biochem Soc Trans 35(5):1252–7. 10.1042/BST0351252 [DOI] [PubMed] [Google Scholar]
  193. Neumann ID, Landgraf R. 2012. Balance of brain oxytocin and vasopressin: implications for anxiety, depression, and social behaviors. Trends Neurosci 35(11):649–59. 10.1016/j.tins.2012.08.004 [DOI] [PubMed] [Google Scholar]
  194. Neumann I, Landgraf R, Bauce L, Pittman QJ. 1995. Osmotic responsiveness and cross talk involving oxytocin, but not vasopressin or amino acids, between the supraoptic nuclei in virgin and lactating rats. J Neurosci 15(5):3408–17. 10.1523/JNEUROSCI.15-05-03408.1995 [DOI] [PMC free article] [PubMed] [Google Scholar]
  195. Neumann I, Ludwig M, Engelmann M, Pittman QJ, Landgraf R. 2008. Simultaneous microdialysis in blood and brain: oxytocin and vasopressin release in response to central and peripheral osmotic stimulation and suckling in the rat. Neuroendocrinology 58(6):637–45. 10.1159/000126604 [DOI] [PubMed] [Google Scholar]
  196. Nishikawa K, Furube E, Morita S, Horii-Hayashi N, Nishi M, Miyata S. 2017. Structural reconstruction of the perivascular space in the adult mouse neurohypophysis during an osmotic stimulation. J Neuroendocrinol 29(2). 10.1111/jne.12456 [DOI] [PubMed] [Google Scholar]
  197. Oldfield BJ, Mckinley MJ. 2004. Circumventricular organs. In: Paxinos G, editor. The rat nervous system. 3rd ed. Cambridge, MA: Academic Press. p. 389–406. 10.1016/B978-012547638-6/50017-1 [DOI] [Google Scholar]
  198. Oresković D, Klarica M. 2010. The formation of cerebrospinal fluid: nearly a hundred years of interpretations and misinterpretations. Brain Res Rev 64(2):241–62. 10.1016/j.brainresrev.2010.04.006 [DOI] [PubMed] [Google Scholar]
  199. Orlando GF, Langnaese K, Landgraf R, Spina MG, Wolf G, Engelmann M. 2007. Neural nitric oxide gene inactivation affects the release profile of oxytocin into the blood in response to forced swimming. Nitric Oxide 16(1):64–70. 10.1016/j.niox.2006.05.002 [DOI] [PubMed] [Google Scholar]
  200. Ouellette J, Toussay X, Comin CH, Costa LDF, Ho M, Lacalle-Aurioles M, Freitas-Andrade M, and others. 2020. Vascular contributions to 16p11.2 deletion autism syndrome modeled in mice. Nat Neurosci 23(9):1090–101. 10.1038/s41593-020-0663-1 [DOI] [PubMed] [Google Scholar]
  201. Palkovits M, Záborszky L, Ambach G. 1974. Accessory neurosecretory cell groups in the rat hypothalamus. Acta Morphol Acad Sci Hung 22(1):21–33. [PubMed] [Google Scholar]
  202. Patisaul HB, Scordalakes EM, Young LJ, Rissman EF. 2003. Oxytocin, but not oxytocin receptor, is regulated by oestrogen receptor beta in the female mouse hypothalamus. J Neuroendocrinol 15(8):787–93. 10.1046/j.1365-2826.2003.01061.x [DOI] [PubMed] [Google Scholar]
  203. Petrov T, Howarth AG, Krukoff TL, Stevenson BR. 1994. Distribution of the tight junction-associated protein ZO-1 in circumventricular organs of the CNS. Brain Res Mol Brain Res 21(3–4):235–46. 10.1016/0169-328x(94)90254-2 [DOI] [PubMed] [Google Scholar]
  204. Pitra S, Zhang M, Cauley E, Stern JE. 2019. NMDA receptors potentiate activity-dependent dendritic release of neuropeptides from hypothalamic neurons. J Physiol 597(6):1735–56. 10.1113/JP277167 [DOI] [PMC free article] [PubMed] [Google Scholar]
  205. Pow DV, Morris JF. 1989. Dendrites of hypothalamic magnocellular neurons release neurohypophysial peptides by exocytosis. Neuroscience 32(2):435–9. 10.1016/0306-4522(89)90091-2 [DOI] [PubMed] [Google Scholar]
  206. Pyner S, Coote JH. 2000. Identification of branching paraventricular neurons of the hypothalamus that project to the rostroventrolateral medulla and spinal cord. Neuroscience 100(3):549–56. 10.1016/s0306-4522(00)00283-9 [DOI] [PubMed] [Google Scholar]
  207. Qian T, Wang H, Wang P, Geng L, Mei L, Osakada T, and others. 2023. A genetically encoded sensor measures temporal oxytocin release from different neuronal compartments. Nature biotechnology 41(7):944–57. 10.1038/s41587-022-01561-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  208. Quintana DS, Alvares GA, Hickie IB, Guastella AJ. 2015. Do delivery routes of intranasally administered oxytocin account for observed effects on social cognition and behavior? A two-level model. Neurosci Biobehav Rev 49:182–92. 10.1016/j.neubiorev.2014.12.011 [DOI] [PubMed] [Google Scholar]
  209. Quintana DS, Guastella AJ. 2020. An allostatic theory of oxytocin. Trends Cogn Sci 24(7):515–28. 10.1016/j.tics.2020.03.008 [DOI] [PubMed] [Google Scholar]
  210. Quintana DS, Westlye LT, Alnæs D, Rustan ØG, Kaufmann T, Smerud KT, and others. 2016. Low dose intranasal oxytocin delivered with breath powered device dampens amygdala response to emotional stimuli: a peripheral effect-controlled within-subjects randomized dose-response fMRI trial. Psychoneuroendocrinology 69:180–8. 10.1016/j.psyneuen.2016.04.010 [DOI] [PubMed] [Google Scholar]
  211. Quintana DS, Westlye LT, Rustan ØG, Tesli N, Poppy CL, Smevik H, and others. 2015. Low-dose oxytocin delivered intranasally with breath powered device affects social-cognitive behavior: a randomized four-way crossover trial with nasal cavity dimension assessment. Translational Psychiatry 5(7):e602. 10.1038/tp.2015.93 [DOI] [PMC free article] [PubMed] [Google Scholar]
  212. Rennels ML, Gregory TF, Blaumanis OR, Fujimoto K, Grady PA. 1985. Evidence for a “paravascular” fluid circulation in the mammalian central nervous system, provided by the rapid distribution of tracer protein throughout the brain from the subarachnoid space. Brain Res 326(1):47–63. 10.1016/0006-8993(85)91383-6 [DOI] [PubMed] [Google Scholar]
  213. Reversi A, Rimoldi V, Marrocco T, Cassoni P, Bussolati G, Parenti M, and others. 2005. The oxytocin receptor antagonist atosiban inhibits cell growth via a “biased agonist” mechanism. J Biol Chem 280(16):16311–8. 10.1074/jbc.M409945200 [DOI] [PubMed] [Google Scholar]
  214. Rhodes CH, Morriell JI, Pfaff DW. 1981. Immunohistochemical analysis of magnocellular elements in rat hypothalamus: distribution and numbers of cells containing neurophysin, oxytocin, and vasopressin. J Comp Neurol 198(1):45–64. 10.1002/cne.901980106 [DOI] [PubMed] [Google Scholar]
  215. Richard D, Bourque CW. 1995. Synaptic control of rat supraoptic neurones during osmotic stimulation of the organum vasculosum lamina terminalis in vitro. J Physiol 489(Pt 2):567–77. 10.1113/jphysiol.1995.sp021073 [DOI] [PMC free article] [PubMed] [Google Scholar]
  216. Rigney N, de Vries GJ, Petrulis A, Young LJ. 2022. Oxytocin, vasopressin, and social behavior: from neural circuits to clinical opportunities. Endocrinology 163(9):bqac111. 10.1210/endocr/bqac111 [DOI] [PMC free article] [PubMed] [Google Scholar]
  217. Rong LL, Yan S-F, Wendt T, Hans D, Pachydaki S, Bucciarelli LG, and others. 2004. RAGE modulates peripheral nerve regeneration via recruitment of both inflammatory and axonal outgrowth pathways. FASEB J 18(15):1818–25. 10.1096/fj.04-1900com [DOI] [PubMed] [Google Scholar]
  218. Rossoni E, Feng J, Tirozzi B, Brown D, Leng G, Moos F. 2008. Emergent synchronous bursting of oxytocin neuronal network. PLoS Comput Biol 4(7):e1000123. 10.1371/journal.pcbi.1000123 [DOI] [PMC free article] [PubMed] [Google Scholar]
  219. Rovsing L, Rath MF, Møller M. 2013. Hypothalamic neurosecretory and circadian vasopressinergic neuronal systems in the blind cone-rod homeobox knockout mouse (Crx-/-) and the 129sv wild-type mouse. J Comp Neurol 521(17):4061–74. 10.1002/cne.23390 [DOI] [PubMed] [Google Scholar]
  220. Roy RK, Althammer F, Seymour AJ, Du W, Biancardi VC, Hamm JP, and others. 2021. Inverse neurovascular coupling contributes to positive feedback excitation of vasopressin neurons during a systemic homeostatic challenge. Cell Reports 37(5):109925. 10.1016/j.celrep.2021.109925 [DOI] [PMC free article] [PubMed] [Google Scholar]
  221. Roy RK, Augustine RA, Brown CH, Schwenke DO. 2018. Activation of oxytocin neurons in the paraventricular nucleus drives cardiac sympathetic nerve activation following myocardial infarction in rats. Commun Biol 1:160. 10.1038/s42003-018-0169-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  222. Sabatier N, Caquineau C, Dayanithi G, Bull P, Douglas AJ, Guan XMM, and others. 2003. Alpha-melanocyte-stimulating hormone stimulates oxytocin release from the dendrites of hypothalamic neurons while inhibiting oxytocin release from their terminals in the neurohypophysis. J Neurosci 23(32):10351–8. 10.1523/JNEUROSCI.23-32-10351.2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  223. Sabatier N, Leng G. 2006. Presynaptic actions of endocannabinoids mediate alpha-MSH-induced inhibition of oxytocin cells. Am J Physiol Regul Integr Comp Physiol 290(3):R577–84. 10.1152/ajpregu.00667.2005 [DOI] [PubMed] [Google Scholar]
  224. Salmina AB, Lopatina O, Ekimova MV, Mikhutkina SV, Higashida H. 2010. CD38/cyclic ADP-ribose system: a new player for oxytocin secretion and regulation of social behaviour. J Neuroendocrinol 22(5):380–92. 10.1111/j.1365-2826.2010.01970.x [DOI] [PubMed] [Google Scholar]
  225. Schini VB, Katusic ZS, Vanhoutte PM. 1990. Neurohypophyseal peptides and tachykinins stimulate the production of cyclic GMP in cultured porcine aortic endothelial cells. J Pharmacol Exp Ther 255(3):994–1000. [PubMed] [Google Scholar]
  226. Sedej S, Gurung IS, Binz T, Rupnik M. 2009. Phosphatidylinositol-4,5-bisphosphate-dependent facilitation of the ATP-dependent secretory activity in mouse pituitary cells. Ann N Y Acad Sci 1152:165–73. 10.1111/j.1749-6632.2008.04002.x [DOI] [PubMed] [Google Scholar]
  227. Shafton AD, Ryan A, Badoer E. 1998. Neurons in the hypothalamic paraventricular nucleus send collaterals to the spinal cord and to the rostral ventrolateral medulla in the rat. Brain Res 801(1-2):239–43. 10.1016/s0006-8993(98)00587-3 [DOI] [PubMed] [Google Scholar]
  228. Sharif Naeini R, Witty M-F, Séguéla P, Bourque CW. 2006. An N-terminal variant of Trpv1 channel is required for osmosensory transduction. Nat Neurosci 9(1):93–8. 10.1038/nn1614 [DOI] [PubMed] [Google Scholar]
  229. Sharma K, LeBlanc R, Haque M, Nishimori K, Reid MM, Teruyama R. 2019. Sexually dimorphic oxytocin receptor-expressing neurons in the preoptic area of the mouse brain. PLoS One 14(7):e0219784. 10.1371/journal.pone.0219784 [DOI] [PMC free article] [PubMed] [Google Scholar]
  230. Shaver SW, Pang JJ, Wainman DS, Wall KM, Gross PM. 1992. Morphology and function of capillary networks in subregions of the rat tuber cinereum. Cell Tissue Res 267(3):437–48. 10.1007/BF00319366 [DOI] [PubMed] [Google Scholar]
  231. Shevchenko T, Teruyama R, Armstrong WE. 2004. High-threshold, Kv3-like potassium currents in magnocellular neurosecretory neurons and their role in spike repolarization. J Neurophysiol 92(5):3043–55. 10.1152/jn.00431.2004 [DOI] [PubMed] [Google Scholar]
  232. Shi P, Martinez MA, Calderon AS, Chen Q, Cunningham JT, Toney GM. 2008. Intra-carotid hyperosmotic stimulation increases Fos staining in forebrain organum vasculosum laminae terminalis neurones that project to the hypothalamic paraventricular nucleus. J Physiol 586(Pt 21):5231–45. 10.1113/jphysiol.2008.159665 [DOI] [PMC free article] [PubMed] [Google Scholar]
  233. Shimizu Y, Harashima A, Munesue S, Oishi M, Hattori T, Hori O, and others. 2020. Neuroprotective effects of endogenous secretory receptor for advanced glycation end-products in brain ischemia. Aging and Disease 11(3):547–58. 10.14336/AD.2019.0715 [DOI] [PMC free article] [PubMed] [Google Scholar]
  234. Shimizu H, Watanabe E, Hiyama TY, Nagakura A, Fujikawa A, Okado H, and others. 2007. Glial Nax channels control lactate signaling to neurons for brain [Na+] sensing. Neuron 54(1):59–72. 10.1016/j.neuron.2007.03.014 [DOI] [PubMed] [Google Scholar]
  235. Sisó S, Jeffrey M, González L. 2010. Sensory circumventricular organs in health and disease. Acta Neuropathol 120(6):689–705. 10.1007/s00401-010-0743-5 [DOI] [PubMed] [Google Scholar]
  236. Smith AS, Korgan AC, Young WS. 2019. Oxytocin delivered nasally or intraperitoneally reaches the brain and plasma of normal and oxytocin knockout mice. Pharmacol Res 146:104324. 10.1016/j.phrs.2019.104324 [DOI] [PMC free article] [PubMed] [Google Scholar]
  237. Sofroniew MV, Glasmann W. 1981. Golgi-like immunoperoxidase staining of hypothalamic magnocellular neurons that contain vasopressin, oxytocin or neurophysin in the rat. Neuroscience 6(4):619–43. 10.1016/0306-4522(81)90147-0 [DOI] [PubMed] [Google Scholar]
  238. Son S, Manjila SB, Newmaster KT, Wu Y-T, Vanselow DJ, Ciarletta M, and others. 2022. Whole-brain wiring diagram of oxytocin system in adult mice. J Neurosci 42(25):5021–33. 10.1523/JNEUROSCI.0307-22.2022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  239. Srisawat R, Bishop VR, Bull PM, Douglas AJ, Russell JA, Ludwig M, and others. 2004. Regulation of neuronal nitric oxide synthase mRNA expression in the rat magnocellular neurosecretory system. Neurosci Lett 369(3):191–6. 10.1016/j.neulet.2004.07.045 [DOI] [PubMed] [Google Scholar]
  240. Stark H, Burbach JP, Van der Kleij AA, De Wied D. 1989. In vivo conversion of vasopressin after microinjection into limbic brain areas of rats. Peptides 10(4):717–20. 10.1016/0196-9781(89)90102-2 [DOI] [PubMed] [Google Scholar]
  241. Stary CM, Xu L, Voloboueva LA, Alcántara-Hernández M, Arvola OJ, Idoyaga J, and others. 2019. Nursing markedly protects postpartum mice from stroke: associated central and peripheral neuroimmune changes and a role for oxytocin. Front Neurosci 13:609. 10.3389/fnins.2019.00609 [DOI] [PMC free article] [PubMed] [Google Scholar]
  242. Stern JE, Ludwig M. 2000. Nitric oxide modulation of supraoptic oxytocin and vasopressin neurons involves potentiation of gabaergic synaptic inputs. Hypertension 36(suppl 1):700. 10.1161/hyp.36.suppl_1.700-b [DOI] [Google Scholar]
  243. Striepens N, Kendrick KM, Hanking V, Landgraf R, Wüllner U, Maier W, and others. 2013. Elevated cerebrospinal fluid and blood concentrations of oxytocin following its intranasal administration in humans. Sci Rep 3:3440. 10.1038/srep03440 [DOI] [PMC free article] [PubMed] [Google Scholar]
  244. Summerlee AJ. 1981. Extracellular recordings from oxytocin neurones during the expulsive phase of birth in unanaesthetized rats. J Physiol 321:1–9. 10.1113/jphysiol.1981.sp013967 [DOI] [PMC free article] [PubMed] [Google Scholar]
  245. Swanson LW, Sawchenko PE. 1980. Paraventricular nucleus: a site for the integration of neuroendocrine and autonomic mechanisms. Neuroendocrinology 31(6):410–7. 10.1159/000123111 [DOI] [PubMed] [Google Scholar]
  246. Swanson LW, Sawchenko PE. 1983. Hypothalamic integration: organization of the paraventricular and supraoptic nuclei. Annu Rev Neurosci 6:269–324. 10.1146/annurev.ne.06.030183.001413 [DOI] [PubMed] [Google Scholar]
  247. Szczepanska-Sadowska E, Sobocinska J, Sadowski B. 1982. Central dipsogenic effect of vasopressin. Am J Physiol 242(3):R372–9. 10.1152/ajpregu.1982.242.3.R372 [DOI] [PubMed] [Google Scholar]
  248. Szeto A, McCabe PM, Nation DA, Tabak BA, Rossetti MA, McCullough ME, and others. 2011. Evaluation of enzyme immunoassay and radioimmunoassay methods for the measurement of plasma oxytocin. Psychosom Med 73(5):393–400. 10.1097/PSY.0b013e31821df0c2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  249. Tabak BA, Leng G, Szeto A, Parker KJ, Verbalis JG, Ziegler TE, and others. 2023. Advances in human oxytocin measurement: challenges and proposed solutions. Mol Psychiatry 28(1):127–40. 10.1038/s41380-022-01719-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  250. Takayanagi Y, Yoshida M, Bielsky IF, Ross HE, Kawamata M, Onaka T, and others. 2005. Pervasive social deficits, but normal parturition, in oxytocin receptor-deficient mice. Proc Natl Acad Sci U S A 102(44):16096–101. 10.1073/pnas.0505312102 [DOI] [PMC free article] [PubMed] [Google Scholar]
  251. Takayanagi Y, Yoshida M, Takashima A, Takanami K, Yoshida S, Nishimori K, and others. 2017. Activation of supraoptic oxytocin neurons by secretin facilitates social recognition. Biol Psychiatry 81(3):243–51. 10.1016/j.biopsych.2015.11.021 [DOI] [PubMed] [Google Scholar]
  252. Tang Y, Benusiglio D, Lefevre A, Hilfiger L, Althammer F, Bludau A, and others. 2020. Social touch promotes interfemale communication via activation of parvocellular oxytocin neurons. Nat Neurosci 23(9):1125–37. 10.1038/s41593-020-0674-y [DOI] [PubMed] [Google Scholar]
  253. Theodosis DT. 2002. Oxytocin-secreting neurons: a physiological model of morphological neuronal and glial plasticity in the adult hypothalamus. Front Neuroendocrinol 23(1):101–35. 10.1006/frne.2001.0226 [DOI] [PubMed] [Google Scholar]
  254. Theodosis DT, Montagnese C, Rodriguez F, Vincent JD, Poulain DA. 1986. Oxytocin induces morphological plasticity in the adult hypothalamo-neurohypophysial system. Nature 322(6081):738–40. 10.1038/322738a0 [DOI] [PubMed] [Google Scholar]
  255. Theodosis DT, Trailin A, Poulain DA. 2006. Remodeling of astrocytes, a prerequisite for synapse turnover in the adult brain? Insights from the oxytocin system of the hypothalamus. Am J Physiol Regul Integr Comp Physiol 290(5):R1175–82. 10.1152/ajpregu.00755.2005 [DOI] [PubMed] [Google Scholar]
  256. Thibonnier M, Conarty DM, Preston JA, Plesnicher CL, Dweik RA, Erzurum SC. 1999. Human vascular endothelial cells express oxytocin receptors. Endocrinology 140(3):1301–9. 10.1210/endo.140.3.6546 [DOI] [PubMed] [Google Scholar]
  257. Thorne RG, Emory CR, Ala TA, Frey WH, 2nd. 1995. Quantitative analysis of the olfactory pathway for drug delivery to the brain. Brain research 692(1–2):278–82. 10.1016/0006-8993(95)00637-6 [DOI] [PubMed] [Google Scholar]
  258. Tobin VA, Arechaga G, Brunton PJ, Russell JA, Leng G, Ludwig M, and others. 2014. Oxytocinase in the female rat hypothalamus: a novel mechanism controlling oxytocin neurones during lactation. J Neuroendocrinol 26(4):205–16. 10.1111/jne.12141 [DOI] [PubMed] [Google Scholar]
  259. Tobin VA, Douglas AJ, Leng G, Ludwig M. 2011. The involvement of voltage-operated calcium channels in somato-dendritic oxytocin release. PLoS One 6(10):e25366. 10.1371/journal.pone.0025366 [DOI] [PMC free article] [PubMed] [Google Scholar]
  260. Tobin VA, Hurst G, Norrie L, Dal Rio FP, Bull PM, Ludwig M. 2004. Thapsigargin-induced mobilization of dendritic dense-cored vesicles in rat supraoptic neurons. Eur J Neurosci 19(10):2909–12. 10.1111/j.1460-9568.2004.03388.x [DOI] [PubMed] [Google Scholar]
  261. Tobin VA, Ludwig M. 2007. The role of the actin cytoskeleton in oxytocin and vasopressin release from rat supraoptic nucleus neurons. J Physiol 582(Pt 3):1337–48. 10.1113/jphysiol.2007.132639 [DOI] [PMC free article] [PubMed] [Google Scholar]
  262. Tsurutani M, Goto T, Hagihara M, Irie S, Miyamichi M. 2023. Selective vulnerability of parvocellular oxytocin neurons in social dysfunction. bioRxiv. 10.1101/2023.12.02.569733 [DOI] [PMC free article] [PubMed]
  263. Turner PJ, Buckler KJ. 2013. Oxygen and mitochondrial inhibitors modulate both monomeric and heteromeric TASK-1 and TASK-3 channels in mouse carotid body type-1 cells. J Physiol 591(Pt 23):5977–98. 10.1113/jphysiol.2013.262022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  264. Tweedle CD, Hatton GI. 1987. Morphological adaptability at neurosecretory axonal endings on the neurovascular contact zone of the rat neurohypophysis. Neuroscience 20(1):241–6. 10.1016/0306-4522(87)90016-9 [DOI] [PubMed] [Google Scholar]
  265. Uchoa ET, Mendes da Silva LE, de Castro M, Antunes-Rodrigues J, Elias LL. 2009. Hypothalamic oxytocin neurons modulate hypophagic effect induced by adrenalectomy. Horm Behav 56(5):532–8. 10.1016/j.yhbeh.2009.09.007 [DOI] [PubMed] [Google Scholar]
  266. Ueno H, Sanada K, Miyamoto T, Baba K, Tanaka K, Nishimura H, and others. 2020. Oxytocin-monomeric red fluorescent protein 1 synthesis in the hypothalamus under osmotic challenge and acute hypovolemia in a transgenic rat line. Physiol Rep 8(17):e14558. 10.14814/phy2.14558 [DOI] [PMC free article] [PubMed] [Google Scholar]
  267. Uvnäs Moberg K, Julius H, Handlin L, Petersson M. 2022. Editorial: sensory stimulation and oxytocin: their roles in social interaction and health promotion. Front Psychol 13:929741. 10.3389/fpsyg.2022.929741 [DOI] [PMC free article] [PubMed] [Google Scholar]
  268. Værøy H, Lahaye E, Dubessy C, Benard M, Nicol M, Cherifi Y, and others. 2023. Immunoglobulin G is a natural oxytocin carrier which modulates oxytocin receptor signaling: relevance to aggressive behavior in humans. Discov Ment Health 3(1):21. 10.1007/s44192-023-00048-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  269. Valstad M, Alvares GA, Egknud M, Matziorinis AM, Andreassen OA, Westlye LT, and others. 2017. The correlation between central and peripheral oxytocin concentrations: a systematic review and meta-analysis. Neurosci Biobehav Rev 78:117–24. 10.1016/j.neubiorev.2017.04.017 [DOI] [PubMed] [Google Scholar]
  270. Veening JG, de Jong T, Barendregt HP. 2010. Oxytocin-messages via the cerebrospinal fluid: behavioral effects; a review. Physiol Behav 101(2):193–210. 10.1016/j.physbeh.2010.05.004 [DOI] [PubMed] [Google Scholar]
  271. Vigh-Teichmann I, Vigh B, Koritsánszky S. 1970. Liquor contacting neurons in the paraventricular nucleus. Article in German. Z Zellforsch Mikrosk Anat 103(4):483–501. [PubMed] [Google Scholar]
  272. Villegas-Gabutti CM, Pennacchio GE, Vivas L, Jahn GA, Soaje M. 2018. Role of oxytocin in prolactin secretion during late pregnancy. Neuroendocrinology 106(4):324–34. 10.1159/000480669 [DOI] [PubMed] [Google Scholar]
  273. Vyas TK, Shahiwala A, Marathe S, Misra A. 2005. Intranasal drug delivery for brain targeting. Curr Drug Deliv 2(2):165–75. 10.2174/1567201053586047 [DOI] [PubMed] [Google Scholar]
  274. Wagner HJ, Pilgrim C, Brandl J. 1974. Penetration and removal of horseradish peroxidase injected into the cerebrospinal fluid: role of cerebral perivascular spaces, endothelium and microglia. Acta Neuropathol 27(4):299–315. 10.1007/BF00690695 [DOI] [PubMed] [Google Scholar]
  275. Wakerley JB, Lincoln DW. 1973. The milk-ejection reflex of the rat: a 20- to 40-fold acceleration in the firing of paraventricular neurones during oxytocin release. J Endocrinol 57(3):477–93. 10.1677/joe.0.0570477 [DOI] [PubMed] [Google Scholar]
  276. Wang L, Li S, Jungalwala FB. 2008. Receptor for advanced glycation end products (RAGE) mediates neuronal differentiation and neurite outgrowth. J Neurosci Research 86(6):1254–66. 10.1002/jnr.21578 [DOI] [PubMed] [Google Scholar]
  277. Wang Y-F, Hatton GI. 2007. Dominant role of betagamma subunits of G-proteins in oxytocin-evoked burst firing. J Neurosci 27(8):1902–12. 10.1523/JNEUROSCI.5346-06.2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  278. Weed LH. 1923. The absorption of cerebrospinal fluid into the venous system. Am J Anat 31(3):191–221. 10.1002/aja.1000310302 [DOI] [Google Scholar]
  279. Wei F, Zhang L, Ma B, Li W, Deng X, Zheng T, and others. 2021. Oxytocin system driven by experiences modifies social recognition and neuron morphology in female BALB/c mice. Peptides 146:170659. 10.1016/j.peptides.2021.170659 [DOI] [PubMed] [Google Scholar]
  280. Weller RO, Djuanda E, Yow H-Y, Carare RO. 2009. Lymphatic drainage of the brain and the pathophysiology of neurological disease. Acta Neuropathol 117(1):1–14. 10.1007/s00401-008-0457-0 [DOI] [PubMed] [Google Scholar]
  281. Williams SK, Barber JS, Jamieson-Drake AW, Enns JA, Townsend LB, Walker CH, and others. 2012. Chronic cocaine exposure during pregnancy increases postpartum neuroendocrine stress responses. J Neuroendocrinol 24(4):701–11. 10.1111/j.1365-2826.2012.02291.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  282. Wislocki GB, King LS. 1936. The permeability of the hypophysis and hypothalamus to vital dyes, with a study of the hypophyseal vascular supply. Am J Anat 58(2):421–72. 10.1002/aja.1000580206 [DOI] [Google Scholar]
  283. Wittkowski W. 1969. Ependymal secretion and receptors in the wall of the recessus infundibularis in mice and their relationship to the small-cell hypothalamus. Article in German. Z Zellforsch Mikrosk Anat 93(4):530–46. [PubMed] [Google Scholar]
  284. Xia X, Lessmann V, Martin TFJ. 2009. Imaging of evoked dense-core-vesicle exocytosis in hippocampal neurons reveals long latencies and kiss-and-run fusion events. J Cell Sci 122(1):75–82. 10.1242/jcs.034603 [DOI] [PMC free article] [PubMed] [Google Scholar]
  285. Xiao L, Priest MF, Nasenbeny J, Lu T, Kozorovitskiy Y. 2017. Biased oxytocinergic modulation of midbrain dopamine systems. Neuron 95(2):368–84.e5. 10.1016/j.neuron.2017.06.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  286. Xu B, Zheng H, Patel KP. 2012. Enhanced activation of RVLM-projecting PVN neurons in rats with chronic heart failure. Am J Physiol Heart Circ Physiol 302(8):H1700–11. 10.1152/ajpheart.00722.2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  287. Xu S, Yang H, Menon V, Lemire AL, Wang L, Henry FE, and others. 2020. Behavioral state coding by molecularly defined paraventricular hypothalamic cell type ensembles. Science 370(6514):eabb2494. 10.1126/science.abb2494 [DOI] [PMC free article] [PubMed] [Google Scholar]
  288. Yamamoto Y, Liang M, Munesue S, Deguchi K, Harashima A, Furuhara K, and others. 2019. Vascular RAGE transports oxytocin into the brain to elicit its maternal bonding behaviour in mice. Communications Biology 2:76. 10.1038/s42003-019-0325-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  289. Yao S, Chen Y, Zhuang Q, Zhang Y, Lan C, Zhu S, and others. 2023. Sniffing oxytocin: nose to brain or nose to blood? Mol Psychiatry 28(7):3083–91. 10.1038/s41380-023-02075-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  290. Yeo S-H, Kyle V, Blouet C, Jones S, Colledge WH. 2019. Mapping neuronal inputs to Kiss1 neurons in the arcuate nucleus of the mouse. PLoS One 14(3):e0213927. 10.1371/journal.pone.0213927 [DOI] [PMC free article] [PubMed] [Google Scholar]
  291. Yin W, Gore AC. 2010. The hypothalamic median eminence and its role in reproductive aging. Ann N Y Acad Sci 1204:113–22. 10.1111/j.1749-6632.2010.05518.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  292. Yoshida M, Takayanagi Y, Inoue K, Kimura T, Young LJ, Onaka T, and others. 2009. Evidence that oxytocin exerts anxiolytic effects via oxytocin receptor expressed in serotonergic neurons in mice. J Neurosci 29(7):2259. 10.1523/JNEUROSCI.5593-08.2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  293. Zhang B, Qiu L, Xiao W, Ni H, Chen L, Wang F, and others. 2021. Reconstruction of the hypothalamo-neurohypophysial system and functional dissection of magnocellular oxytocin neurons in the brain. Neuron 109(2):331–46.e7. 10.1016/j.neuron.2020.10.032 [DOI] [PubMed] [Google Scholar]
  294. Zhang G, Zhang Y, Fast DM, Lin Z, Steenwyk R. 2011. Ultra sensitive quantitation of endogenous oxytocin in rat and human plasma using a two-dimensional liquid chromatography–tandem mass spectrometry assay. Analytical Biochemistry 416(1):45–52. 10.1016/j.ab.2011.04.041 [DOI] [PubMed] [Google Scholar]
  295. Zhang Q, Haselden WD, Charpak S, Drew PJ. 2023. Could respiration-driven blood oxygen changes modulate neural activity? Pflugers Arch 475(1):37–48. 10.1007/s00424-022-02721-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  296. Zoli M, Jansson A, Syková E, Agnati LF, Fuxe K. 1999. Volume transmission in the CNS and its relevance for neuropsychopharmacology. Trends Pharmacol Sci 20(4):142–50. https://doi-org.ezaccess.libraries.psu.edu/10.1016/s0165-6147(99)01343-7 [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

sj-docx-1-nro-10.1177_10738584241268754 – Supplemental material for Navigating Central Oxytocin Transport: Known Realms and Uncharted Territories

Supplemental material, sj-docx-1-nro-10.1177_10738584241268754 for Navigating Central Oxytocin Transport: Known Realms and Uncharted Territories by Deniz Parmaksiz and Yongsoo Kim in The Neuroscientist


Articles from The Neuroscientist are provided here courtesy of SAGE Publications

RESOURCES