Next Article in Journal
Transient Expression in HEK-293 Cells in Suspension Culture as a Rapid and Powerful Tool: SARS-CoV-2 N and Chimeric SARS-CoV-2N-CD154 Proteins as a Case Study
Next Article in Special Issue
Chronic Thromboembolic Pulmonary Hypertension: A Review of the Multifaceted Pathobiology
Previous Article in Journal
A Prospective Study on the Roles of the Lymphocyte-to-Monocyte Ratio (LMR), Neutrophil-to-Lymphocyte Ratio (NLR), and Platelet-to-Lymphocyte Ratio (PLR) in Patients with Locally Advanced Rectal Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Review of Serotonin in the Developing Lung and Neonatal Pulmonary Hypertension

by
Jamie L. Archambault
and
Cassidy A. Delaney
*
Section of Neonatology, Department of Pediatrics, University of Colorado, Aurora, CO 80045, USA
*
Author to whom correspondence should be addressed.
Biomedicines 2023, 11(11), 3049; https://doi.org/10.3390/biomedicines11113049
Submission received: 11 October 2023 / Revised: 3 November 2023 / Accepted: 9 November 2023 / Published: 14 November 2023

Abstract

:
Serotonin (5-HT) is a bioamine that has been implicated in the pathogenesis of pulmonary hypertension (PH). The lung serves as an important site of 5-HT synthesis, uptake, and metabolism with signaling primarily regulated by tryptophan hydroxylase (TPH), the 5-HT transporter (SERT), and numerous unique 5-HT receptors. The 5-HT hypothesis of PH was first proposed in the 1960s and, since that time, preclinical and clinical studies have worked to elucidate the role of 5-HT in adult PH. Over the past several decades, accumulating evidence from both clinical and preclinical studies has suggested that the 5-HT signaling pathway may play an important role in neonatal cardiopulmonary transition and the development of PH in newborns. The expression of TPH, SERT, and the 5-HT receptors is developmentally regulated, with alterations resulting in pulmonary vasoconstriction and pulmonary vascular remodeling. However, much remains unknown about the role of 5-HT in the developing and newborn lung. The purpose of this review is to discuss the implications of 5-HT on fetal and neonatal pulmonary circulation and summarize the existing preclinical and clinical literature on 5-HT in neonatal PH.

1. Introduction

Serotonin (5-HT) is a bioamine most appreciated for its role as a neurotransmitter and its effect on mood and behavior. However, the majority of 5-HT is synthesized in the periphery where 5-HT modulates numerous other biologic processes including respiratory, cardiovascular, and gastrointestinal functions [1,2,3]. Over the past several decades, 5-HT has emerged as a potent pulmonary vasoconstrictor and angiogenic agent that plays an important role in the development of pulmonary hypertension (PH).
5-HT is synthesized from L-tryptophan through the activity of tryptophan hydroxylase (TPH), the rate-limiting enzyme in 5-HT synthesis. TPH has two distinct isoforms. TPH1 is primarily present in the enterochromaffin cells of the small intestine and is responsible for the production of peripheral 5-HT, whereas TPH2 is present in the central and enteric nervous systems [4,5]. TPH converts L-tryptophan to 5-hydroxytroptophan (5-HTP). 5-HTP is then converted by amino acid decarboxylase (AAAD) to 5-HT (Figure 1). On first pass, 30–80% of 5-HT synthesized in the intestine is metabolized by the liver and 90% of the remainder is metabolized by the lungs. The remaining 10% is primarily stored in platelets which take up 5-HT via the 5-HT transporter (SERT) (Figure 2). Consequently, only a small amount of 5-HT freely circulates and plasma levels are extremely low [6].
The diverse biologic functions of 5-HT are mediated by fifteen unique receptors divided into seven families (5-HT 1–7) based on molecular structure, signal transduction properties, and pharmacological properties [7,8]. The 5-HT 1, 2, and 4–7 receptors couple with G-proteins, while the 5-HT 3 receptors are 5-HT-gated ion channels. The altered expression and activity of both SERT and 5-HT receptors have been implicated in the pathophysiology of a wide spectrum of conditions in the fetus, newborns, and adults. This review aims to describe the role of pulmonary 5-HT signaling, discuss its implications on the adult lung, and summarize the existing preclinical and clinical literature investigating the role of 5-HT in the developing lung and neonatal PH.

2. Serotonin in Adult Pulmonary Circulation

2.1. Serotonin Signaling in the Adult Lung

The lung is an important site of 5-HT synthesis, uptake, and metabolism (Figure 3). Pulmonary artery endothelial cells (PA-EC) that line the intima of the pulmonary vasculature, along with pulmonary neuroendocrine cells (PNEC) that line the airway epithelium, express TPH1 and contribute to local 5-HT synthesis. Preclinical studies have demonstrated that PA-EC synthesis of TPH1 is upregulated under hypoxic conditions, leading to increased local production of 5-HT [9,10]. The lung also plays an important role in the clearance of 5-HT via SERT-mediated uptake by PA-ECs, primarily at the site of pulmonary arterioles and capillaries [11,12,13,14]. The PA-EC uptake of 5-HT is a saturable, energy-dependent process that serves as the rate-limiting step in pulmonary clearance of circulating 5-HT [12]. Once taken up by PA-ECs, 5-HT is converted into its active metabolite 5-hydroxyindolacetic acid (5-HIAA) by the intracellular mitochondrial enzyme monoamine oxidase (MAO) [15,16].
SERT and the 5-HT G-protein-coupled receptors 1B, 2A, and 2B expressed by pulmonary artery smooth muscle cells (PA-SMC) serve to mediate 5-HT-induced vasoconstriction and pulmonary vascular remodeling [17,18,19,20,21]. 5-HT intracellular signaling is complex and binding to these receptors stimulates the induction of reactive oxygen species, activation of Rho-kinase, and mitogen activation of the protein kinase pathway [22,23]. Phosphorylated kinases travel to the nucleus to increase the transcription of growth factors, promote cellular proliferation, and contribute to vasoconstriction [24]. In non-human mammals the 5-HT 2A receptor mediates pulmonary vasoconstriction, while the 5-HT 1B receptor mediates both pulmonary vasoconstriction and PA-SMC proliferation [18,20,25]. When aberrantly regulated, these processes significantly contribute to the pathophysiology underlying PH

2.2. Serotonin in Adult Pulmonary Hypertension

The 5-HT hypothesis of PH was first proposed in the 1960s following the observation that the use of appetite suppressants such as aminorex, fenfluramine, and chlorphentermine led to the development of PH [26,27,28,29,30,31,32]. These drugs are known to increase extracellular levels of 5-HT in both the brain and the lung by exchanging exogenous drug molecules for endogenous 5-HT, thus providing substrate for 5-HT uptake/transport and contributing to the development of PH. This association was further supported by the observation that patients with altered platelet 5-HT storage develop PH [33,34,35,36,37].
The World Health Organization (WHO) categorizes PH into five distinct categories: (1) pulmonary arterial hypertension (PAH); (2) PH due to left heart disease; (3) PH due to chronic lung disease and/or hypoxia; (4) chronic thromboembolic PH; (5) PH due to unknown causes [38]. PH is characterized by vasoconstriction, pulmonary vascular remodeling, and thrombosis leading to increased pulmonary vascular resistance and right-sided heart failure [39,40,41]. Morbidity and mortality are significant with outcomes closely tied to the status of the right ventricle [42,43,44,45]. While most of the literature discussing the role of 5-HT in the pathogenesis of PH refers to the WHO Group 1 classification, or PAH, it is reasonable to conclude that 5-HT may play a role in the pathogenesis of all groups.
The genetic expression of TPH1 is increased in the lungs and PA-ECs of patients with idiopathic PAH (IPAH) [10]. The pharmacologic and/or genetic depletion of 5-HT synthesis prevents the development of PH in adult mice. When exposed to hypoxia, 5-HT deficient mice (TPH1 knockout mice) demonstrate decreased PA-SMC proliferation, decreased right ventricular hypertrophy, and decreased right ventricular systolic pressures compared to wild-type mice [46,47]. Despite these associations, studies attempting to quantify circulating 5-HT levels in patients with idiopathic PH have yielded conflicting results, perhaps related to the heterogeneity of the disease and difficulty measuring free 5-HT [6].
SERT and 5-HT receptor expression are increased in PH. SERT is highly expressed by lung PA-SMCs and expression is increased in PA-SMCs from patients with IPAH [48]. Pulmonary SERT overexpression is associated with more severe disease in experimental PH, and mice deficient in SERT display attenuated hypoxia-induced pulmonary vascular remodeling and right ventricular hypertrophy compared to wild-type mice [46,49,50,51]. Pulmonary artery expression of the 5-HT 1B receptor is increased in both patients with PAH and mice with experimental PH [24,52]. Further, 5-HT 1B knockout mice display attenuated pulmonary artery vasoconstriction under hypoxic conditions and pharmacologic inhibition of the 1B and 2A receptors prevents the development of experimental PH [21,53,54].
As a result of these findings in the adult population, there is currently a phase 2 multicenter clinical trial (ELEVATE 2) investigating the use of a rodatristat ethyl, a novel TPH1 inhibitor, for the treatment of PH in adults [55]. Blocking the biosynthesis of peripheral 5-HT from tryptophan is hypothesized to decrease circulating 5-HT concentration, thus reducing available SERT substrate, limiting binding to 5-HT receptors, and ultimately ameliorating downstream vasoconstriction and smooth muscle cell proliferation of the pulmonary arteries. This drug has demonstrated efficacy in preclinical models of PH and shown a dose-dependent reduction of the 5-HT metabolite, 5-HIAA, in the plasma and urine of healthy human subjects, though outcomes in humans with PH have yet to be determined [56]. It is clear based on this work that 5-HT plays a well-defined role in the development of adult PH, though relatively little is known about the pathogenesis of PH in the developing lung.

3. Serotonin in Fetal Pulmonary Circulation

3.1. Serotonin Signaling during Pregnancy and Fetal Development

During pregnancy, 5-HT levels remain constant in the mother but increase in both the placenta and fetus [57]. Consequently, the demand for tryptophan, the essential amino acid that serves as the precursor for 5-HT synthesis, increases to match the needs for fetal growth and development. The fetus is initially dependent on maternal amino acid intake and the placental transport of tryptophan [58]. The placenta serves as an important site of 5-HT synthesis up until mid-gestation, with a subsequent decline throughout the second and third trimesters as the fetus begins independently synthesizing 5-HT [57,59,60]. Human studies have revealed platelet levels of 5-HT are relatively low at birth, but quickly increase and reach concentrations close to adult level by 1 month of life [61].
Disruption in 5-HT signaling during pregnancy has a significant negative impact on fetal growth and development [62,63,64,65,66,67,68,69]. Unsurprisingly, given its known function as a neurotransmitter, 5-HT plays a critical role in fetal brain development and impacts cell migration, proliferation, and maturation [63,70,71,72]. Increased placental 5-HT synthesis may affect fetal forebrain development, leading to long-term behavioral abnormalities such as anxiety [64]. Maternal stress increases tryptophan synthesis and fetal brain concentrations of 5-HT in rats, leading to increased anxiety in the offspring [73]. Meanwhile, impaired placental degradation of tryptophan in humans is associated with an increased risk of schizophrenia, bipolar disorder, and autism [74,75]. While these neuropsychologic consequences have been long appreciated, more recent literature has begun to examine the impact of 5-HT signaling on other biologic processes, including the respiratory and cardiovascular systems.

3.2. Serotonin Signaling in the Fetal Lung

In utero, PNECs, which are known to express TPH1, emerge early in gestation during the embryonic stage of human lung development [76]. PNECs are considered to be the most important oxygen and stimuli-sensing epithelial cells in the respiratory system [77]. Despite this important role, they make up just 0.01% of human lung cells [78,79]. 5-HT-containing PNECs, as compared to others (e.g., bombesin), are more prominent during the early stages of development and are present as early as 8 weeks gestation. They are mostly found in larger airways as the epithelium centrifugally differentiates. Preclinical studies in rats have found that 5-HT PNECs increase in number throughout gestation as the fetus nears term and TPH1 expression gradually increases, suggesting 5-HT may play an important role in the neonatal cardiopulmonary transition [60].
SERT expression in the rat lung, amongst other organs (brain, intestine, liver), significantly increases throughout gestation, allowing for improved regulation of 5-HT clearance [60,80]. In the human lung, pulmonary SERT expression begins at 30 weeks gestation, increases to term, and remains postnatally elevated [81]. Studies quantifying 5-HT receptor expression throughout gestation are limited, but have revealed that the pulmonary expression of 5-HT receptors is developmentally regulated, similar to that of SERT. We reported the 5-HT 2A receptor is expressed in the fetal ovine lung and that, compared to the adult ovine lung, the expression of the 5-HT 1B, 2A, and 2B receptors is markedly decreased [82]. Hodge et al. found there is a trend towards increased 5-HT 4 receptor expression in the human fetal lung from 19 days to 19 weeks of gestation [83]. Nikolic et al. analyzed the expression of the 5-HT receptors 5HT 1A, 2A, and 3A across different stages of human lung development from 12 to 40 weeks of gestation [84]. They too found receptor expression was developmentally regulated, with the strongest expression in the epithelium of the proximal airways. 5-HT 1A was also strongly expressed in the smooth muscle cells of the proximal airway, and 5-HT 1A and 2A were expressed by both vascular smooth muscle cells and endothelial cells.
These developmental differences in expression likely have physiologic consequences. While the infusion of 5-HT induces pulmonary vasoconstriction in rabbits of all developmental stages, from fetal through adult life, only 5-HT 2A receptors mediate vasoconstriction in fetal pulmonary vessels, while 5-HT 1B and 1D receptors mediate vasoconstriction in adult vessels [85]. This finding is supported by Goyal et al., who demonstrated the 5-HT 2A receptor is the predominant 5-HT receptor involved in 5-HT-induced vasoconstriction in isolated ovine fetal pulmonary arteries exposed to long-term maternal hypoxia [86]. 5-HT clearly plays a defined role in pulmonary vasoconstriction, but the nuanced mechanisms vary across developmental stages.

4. Serotonin in Neonatal Pulmonary Circulation

4.1. Neonatal Cardiopulmonary Transition

The definition and classification of PH are the same for children as they are for adults. While there are indeed differences in the etiology, presentation, and outcomes of pediatric PH compared to adult PH, the histopathology and mechanisms for targeted therapies remain similar [87]. However, it is unclear if this same framework should also be applied to infants. In utero, PVR is significantly greater than systemic vascular resistance, allowing for the preferential shunting of oxygenated placental blood to the developing fetus. At birth, ventilation of the newborn lung causes a dramatic decrease in PVR and pulmonary blood flow increases by 8–10 fold to accommodate the postnatal dependence on alveolar gas exchange [88,89]. Pulmonary artery pressures continue to decrease for 2–3 months after birth, a process that takes longer in infants born at higher altitudes. Failure to complete this physiologic transition after birth results in a persistent pulmonary hypertension of the newborn (PPHN), a disease process characterized by sustained elevation in PVR and intracardiac shunting, leading to severe hypoxemia. While the exact incidence and prevalence of PH in the pediatric population are unknown, current registries have begun to examine the etiology associated with pediatric PH. In addition to PPHN, the majority of the remaining cases of PH are caused by idiopathic PAH, PH due to congenital heart disease, or PH due to chronic lung disease of prematurity [90,91,92].

4.2. Selective Serotonin Reuptake Inhibitors and Persistent Pulmonary Hypertension of the Newborn

In the early 2000s, several retrospective case-control studies and meta-analyses investigated the association between the maternal use of selective serotonin reuptake inhibitors (SSRI) and the incidence of PPHN. Based on the concerning findings reported by these epidemiologic studies, the United States Food and Drug Administration issued an advisory statement warning against the potential consequences of SSRI use during pregnancy.
SSRIs are a class of medications that block the neuronal reuptake and reabsorption of 5-HT. Approximately 8–10% of pregnant women are prescribed selective SSRIs to treat anxiety and/or depression [93,94,95]. SSRIs bind to SERT expressed by placental syncytiotrophoblasts or cross the placenta and bind to SERT expressed by fetal platelets and organ tissues, resulting in increased local concentrations of 5-HT [96,97,98]. The lung, in addition to the brain, serves as a primary uptake site for SSRIs and may function as a reservoir for antidepressants with a high affinity to SERT [99]. SSRIs are also known to inhibit nitric oxide synthase (NOS) and may impact the concentration of nitric oxide, a potent vasodilator that plays a critical role in the regulation of vascular tone in utero and during postnatal transition [100,101]. Maternal use of SSRIs may compromise uterine/placental vascular perfusion and impact fetal growth. In humans, maternal SSRI use is associated with changes in fetal heart rate and cerebral blood flow, consistent with reductions in placental perfusion and resultant fetal hypoxia [102].
The early epidemiological studies found a six-fold increased risk of PPHN in neonates when exposed to maternal SSRI in pregnancy, specifically during the second and third trimesters [62,103,104,105,106,107,108,109]. However, further examination of this association revealed conflicting data, with an absolute risk difference of 0.6–2 per 1000 live births [95,110,111,112,113,114]. Clinical practice has shown the potential effects of SSRI exposure on respiratory distress are short lived, with no reported deaths. Thus, current obstetric and psychiatric/psychologic practice advises against cessation of SSRI use before or during pregnancy, acknowledging the significant maternal and infant morbidity associated with untreated perinatal depression and anxiety [115].
Despite conflicting evidence regarding the clinical significance of SSRI exposure and PPHN, there is a clear impact of maternal SSRI use on fetomaternal circulation [116,117,118]. Large animal models have shown that acute infusion of the SSRI fluoxetine into pregnant sheep leads to a transient decrease in uterine artery blood flow, fetal PaO2, and oxygen saturation [118]. Similarly, the offspring of rats treated with fluoxetine in late gestation displayed lower postnatal arterial oxygen saturation and PH characterized by increased pulmonary artery medial wall thickness and increased right ventricular mass [119].
Our group utilized the chronically prepared fetal sheep model to study the in vivo hemodynamic effects of SSRIs in the developing lung. We found that brief infusions of the SSRIs sertraline and fluoxetine directly to the fetal ovine lung caused potent and sustained elevation in pulmonary vascular resistance, which could be reversed by the 5-HT 2A receptor antagonist ketanserin [82,120]. Acetylcholine-induced vasodilation persisted after sertraline treatment, suggesting sertraline does not directly impair NOS activity. A similar study in mice revealed sertraline and fluoxetine contribute to in utero constriction of the ductus arteriosus, a well-known mechanism for elevated prenatal pulmonary pressures and PPHN [121]. Based on these mechanisms, it is plausible to conclude the accumulation of lung 5-HT from SSRI exposure could contribute to increased vasoconstriction, smooth muscle cell proliferation, and elevated pulmonary vascular resistance in exposed infants leading to the clinical syndrome known as PPHN.

4.3. Serotonin in Neonatal Pulmonary Hypertension

Several decades after the original 5-HT hypothesis was proposed in the literature, a pediatric clinical study quantified 5-HT PNECs in infants who died due to respiratory distress syndrome (RDS) and bronchopulmonary dysplasia (BPD) [122]. Interestingly, the number of 5-HT immunoreactive PNECs was decreased in preterm infants who died from RDS, but significantly increased in those who died from BPD. These results were most pronounced at 2 months of age, when infants with BPD had a 34-fold increase in 5-HT immunoreactive PNECs. The significance of the 5-HT pathway in the process of pulmonary vascular remodeling at birth is further supported by evidence that SERT expression is decreased in infants with alveolar capillary dysplasia with misalignment of pulmonary veins (ACD/MPV) [81]. ACD/MPV is a uniformly lethal condition characterized by severe PPHN. The pulmonary microvasculature of infants with ACD/MPV is markedly depleted of SERT expression, suggesting alterations in 5-HT signaling may contribute to abnormal vascular development and PH. 5-HT metabolism is also increased in pediatric patients with PH due to congenital heart disease [123]. In this population, there is no appreciable difference in the concentration of plasma total and free 5-HT. However, these patients have increased urinary excretion of 5-HIAA, which may be due to increased 5-HT metabolism.
Numerous preclinical studies have demonstrated an important association between 5-HT and PH in both small and large animal models of neonatal PH. 5-HT exacerbates pulmonary artery constriction in a hyperoxic rat model of BPD-induced PH, and pulmonary TPH1 is increased in both murine and ovine models of PH [120,124,125]. In an experimental model of congenital diaphragmatic hernia, a developmental lung disease associated with PPHN and high rates of neonatal morbidity and mortality, the expression of SERT and the 5-HT 2A receptor are upregulated in the pulmonary vasculature, suggesting 5-HT may mediate pulmonary vascular remodeling seen in this disease [126].
Our group studied the hemodynamic effects of 5-HT and 5-HT receptor antagonists in the late-gestation ovine fetus for over a decade [82]. We found that a brief infusion of 5-HT into the pulmonary vasculature increases PVR in a dose-dependent manner. Pharmacologic inhibition of the 5-HT 2A receptor with direct pulmonary infusions of the 5-HT 2A receptor antagonist ketanserin results in pulmonary vasodilation, suggesting endogenous 5-HT contributes to the maintenance of normal pulmonary vascular tone in the ovine fetus via activation of the 5-HT 2A receptor. Ketanserin also prevents exogenous 5-HT-induced pulmonary vasoconstriction. However, pharmacologic inhibition of the 5-HT 1B and 2B receptors does not have this effect. We concluded from this work that the pulmonary vasoconstrictor effects of 5-HT are mediated by the 5-HT 2A receptor and, in part, through rho kinase activation.
We further demonstrated the hemodynamic effects of 5-HT infusion and 5-HT 2A receptor antagonism in a PHHN model of the ovine fetus [120]. As demonstrated in other animal models, prolonged constriction of the ductus arteriosus in fetal sheep leads to elevated pulmonary pressures. This elevation is augmented by 5-HT infusion and ameliorated by 5-HT 2A receptor antagonism. From this we conclude that 5-HT contributes to high PVR in the fetus and speculate that prolonged exposure may impact fetal pulmonary circulation and postnatal transition.
Because both TPH1 and 5-HT 2A receptor expression are increased in experimental PH and the pharmacologic blockade of the 5-HT 2A receptor is protective against the development of experimental PH, we hypothesized that genetic depletion of TPH1 is also protective against PH. Interestingly, this was not the case [127]. Contrary to our hypothesis, circulating and pulmonary 5-HT is decreased in hypoxic wild-type mice compared to the controls. TPH1 knock out mice have a similar degree of hypoxia-induced alveolar simplification and pulmonary vascular remodeling as wild-type mice. Interestingly, the hypoxia-induced increase in right ventricular systolic pressures is appropriately attenuated in TPH1 knock out mice compared to wild-type mice, suggesting local pulmonary 5-HT contributes to pulmonary vascular tone. While the genetic and pharmacologic inhibitions of TPH1 have shown protection in adult models, the mixed findings in neonatal suggest inhibition may have a variable impact in the developing lung.

5. Conclusions

PH is a devastating disease that affects adults, children, and infants. Its pathophysiology is driven by a variety of mechanisms that ultimately contribute to disease severity. 5-HT is a potent pulmonary vasoconstrictor and smooth muscle mitogen that has been implicated in the pathogenesis of this disease. 5-HT is both centrally and peripherally synthesized from L-tryptophan through the activity of TPH. Its biologic functions are primarily mediated by SERT and 5-HT receptors. Targeting 5-HT signaling with novel therapeutic agents via these pathways has shown great promise for adults with PH. However, pulmonary SERT and 5-HT receptor expression is developmentally regulated. The limited body of literature describing the role of 5-HT in the developing lung and neonatal PH highlights that this mechanism is quite complicated, and may differ from what is known about 5-HT in adult pulmonary circulation.
PNECs expressing TPH1 are present early in the embryonic phase of development, whereas SERT expression begins later in gestation but gradually rises as the fetus nears term. The 5-HT 2A receptor is the predominant receptor that regulates 5-HT-induced vasoconstriction in the perinatal circulation. Both clinical and preclinical studies suggest that 5-HT signaling contributes to the fetal to neonatal cardiopulmonary transition and that elevated levels of 5-HT may contribute to the development of neonatal PH. Preclinical studies targeting the 5-HT signaling pathway have demonstrated a variable protective effect against 5-HT-mediated pulmonary vasoconstriction, highlighting that we have much to learn about the impact of 5-HT on both lung development and the initiation and progression of PH in newborns. We are hopeful that this field will continue to expand towards improving outcomes for neonates with PH.

Author Contributions

J.L.A. and C.A.D. made substantial contributions to the conception and design of this review paper. J.L.A. and C.A.D. drafted the article or revised it critically for important intellectual content. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Rapport, M.M.; Green, A.; Page, I.H. Serum Vasoconstriction (Serotonin): Isolation and Characterization. J. Biol. Chem. 1948, 176, 1243–1251. [Google Scholar] [CrossRef] [PubMed]
  2. Kroeze, W.K.; Kristiansen, K.; Roth, B.L. Molecular biology of serotonin receptors structure and function at the molecular level. Curr. Top. Med. Chem. 2002, 2, 507–528. [Google Scholar] [CrossRef] [PubMed]
  3. Berger, M.; Gray, J.A.; Roth, B.L. The expanded biology of serotonin. Annu. Rev. Med. 2009, 60, 355–366. [Google Scholar] [CrossRef] [PubMed]
  4. Walther, D.J.; Bader, M. A unique central tryptophan hydroxylase isoform. Biochem. Pharmacol. 2003, 66, 1673–1680. [Google Scholar] [CrossRef]
  5. Walther, D.J.; Peter, J.U.; Bashammakh, S.; Hörtnagl, H.; Voits, M.; Fink, H.; Bader, M. Synthesis of Serotonin by a Second Tryptophan Hydroxylase Isoform. Science 2003, 299, 5603. [Google Scholar] [CrossRef]
  6. Brenner, B.; Harney, J.T.; Ahmed, B.A.; Jeffus, B.C.; Unal, R.; Mehta, J.L.; Kilic, F. Plasma serotonin levels and the platelet serotonin transporter. J. Neurochem. 2007, 102, 206–215. [Google Scholar] [CrossRef]
  7. Raymond, J.R.; Mukhin, Y.V.; Gelasco, A.; Turner, J.; Collinsworth, G.; Gettys, T.W.; Grewal, J.S.; Garnovskaya, M.N. Multiplicity of mechanisms of serotonin receptor signal transduction. Pharmacol. Ther. 2001, 92, 179–212. [Google Scholar] [CrossRef]
  8. Hannon, J.; Hoyer, D. Molecular biology of 5-HT receptors. Behav. Brain Res. 2008, 195, 198–213. [Google Scholar] [CrossRef]
  9. Izikki, M.; Hanoun, N.; Marcos, E.; Savale, L.; Barlier-Mur, A.M.; Saurini, F.; Eddahibi, S.; Hamon, M.; Adnot, S. Tryptophan hydroxylase 1 knockout and tryptophan hydroxylase 2 polymorphism: Effects on hypoxic pulmonary hypertension in mice. Am. J. Physiol. Lung Cell Mol. Physiol. 2007, 293, L1045–L1052. [Google Scholar] [CrossRef]
  10. Eddahibi, S.; Guignabert, C.; Barlier-Mur, A.-M.; Dewachter, L.; Fadel, E.; Dartevelle, P.; Humbert, M.; Simonneau, G.; Hanoun, N.; Saurini, F.; et al. Cross talk between endothelial and smooth muscle cells in pulmonary hypertension: Critical role for serotonin-induced smooth muscle hyperplasia. Circulation 2006, 113, 1857–1864. [Google Scholar] [CrossRef]
  11. Thomas, D.P.; Vane, J.R. 5-hydroxytryptamine in the circulation of the dog. Nature 1967, 216, 335–338. [Google Scholar] [CrossRef] [PubMed]
  12. Strum, J.M.; Junod, A.F. Radioautographic demonstration of 5-hydroxytryptamine-3H uptake by pulmonary endothelial cells. J. Cell Biol. 1972, 54, 456–467. [Google Scholar] [CrossRef] [PubMed]
  13. Alabaster, V.A.; Bakhle, Y.S. Removal of 5-hydroxytryptamine by rat isolated lung. Br. J. Pharmacol. 1970, 38, 440P–441P. [Google Scholar]
  14. Gillis, C.N.; Roth, J.A. The fate of biogenic monoamines in perfused rabbit lung. Br. J. Pharmacol. 1977, 59, 585–590. [Google Scholar] [CrossRef]
  15. Pickett, R.D.; Anderson, M.W.; Orton, T.C.; Eling, T.E. The pharmacodynamics of 5-hydroxytryptamine uptake and metabolism by the isolated perfused rabbit lung. J. Pharmacol. Exp. Ther. 1975, 194, 545–553. [Google Scholar]
  16. Gao, Y.; Chen, T.; Raj, J.U. Endothelial and Smooth Muscle Cell Interactions in the Pathobiology of Pulmonary Hypertension. Am. J. Respir. Cell Mol. Biol. 2016, 54, 451–460. [Google Scholar] [CrossRef] [PubMed]
  17. Ullmer, C.; Schmuck, K.; Kalkman, H.O.; Lübbert, H. Expression of serotonin receptor mRNAs in blood vessels. FEBS Lett. 1995, 370, 215–221. [Google Scholar] [CrossRef]
  18. MacLean, M.R.; Sweeney, G.; Baird, M.; McCulloch, K.M.; Houslay, M.; Morecroft, I. 5-Hydroxytryptamine receptors mediating vasoconstriction in pulmonary arteries from control and pulmonary hypertensive rats. Br. J. Pharmacol. 1996, 119, 917–930. [Google Scholar] [CrossRef]
  19. Cortijo, J.; Martí-Cabrera, M.; Bernabeu, E.; Domènech, T.; Bou, J.; Fernández, A.G.; Beleta, J.; Palacios, J.M.; Morcillo, E.J. Characterization of 5-HT receptors on human pulmonary artery and vein: Functional and binding studies. Br. J. Pharmacol. 1997, 122, 1455–1463. [Google Scholar] [CrossRef]
  20. Morecroft, I.; Heeley, R.P.; Prentice, H.M.; Kirk, A.; MacLean, M.R. 5-hydroxytryptamine receptors mediating contraction in human small muscular pulmonary arteries: Importance of the 5-HT1B receptor. Br. J. Pharmacol. 1999, 128, 730–734. [Google Scholar] [CrossRef]
  21. Keegan, A.; Morecroft, I.; Smillie, D.; Hicks, M.N.; MacLean, M.R. Contribution of the 5-HT(1B) receptor to hypoxia-induced pulmonary hypertension: Converging evidence using 5-HT(1B)-receptor knockout mice and the 5-HT(1B/1D)-receptor antagonist GR127935. Circ. Res. 2001, 89, 1231–1239. [Google Scholar] [CrossRef] [PubMed]
  22. MacLean, M.R.; Herve, P.; Eddahibi, S.; Adnot, S. 5-hydroxytryptamine and the pulmonary circulation: Receptors, transporters and relevance to pulmonary arterial hypertension. Br. J. Pharmacol. 2000, 131, 161–168. [Google Scholar] [CrossRef] [PubMed]
  23. Watts, S.W.; Yang, P.; Banes, A.K.; Baez, M. Activation of Erk mitogen-activated protein kinase proteins by vascular serotonin receptors. J. Cardiovasc. Pharmacol. 2001, 38, 539–551. [Google Scholar] [CrossRef] [PubMed]
  24. Launay, J.-M.; Hervé, P.; Peoc’h, K.; Tournois, C.; Callebert, J.; Nebigil, C.; Etienne, N.; Drouet, L.; Humbert, M.; Simonneau, G.; et al. Function of the serotonin 5-hydroxytryptamine 2B receptor in pulmonary hypertension. Nat. Med. 2002, 8, 1129–1135. [Google Scholar] [CrossRef]
  25. Lawrie, A.; Spiekerkoetter, E.; Martinez, E.C.; Ambartsumian, N.; Sheward, W.J.; MacLean, M.R.; Harmar, A.J.; Schmidt, A.-M.; Lukanidin, E.; Rabinovitch, M.; et al. Interdependent serotonin transporter and receptor pathways regulate S100A4/Mts1, a gene associated with pulmonary vascular disease. Circ. Res. 2005, 97, 227–235. [Google Scholar] [CrossRef]
  26. Fahlén, M.; Bergman, H.; Helder, G.; Rydén, L.; Wallentin, I.; Zettergren, L. Phenformin and pulmonary hypertension. Br. Heart J. 1973, 35, 824–828. [Google Scholar] [CrossRef]
  27. Gurtner, H.P. Aminorex and pulmonary hypertension. A review. Cor Vasa. 1985, 27, 160–171. [Google Scholar]
  28. Pouwels, H.M.; Smeets, J.L.; Cheriex, E.C.; Wouters, E.F. Pulmonary hypertension and fenfluramine. Eur. Respir. J. 1990, 3, 606–607. [Google Scholar] [CrossRef]
  29. Brenot, F.; Herve, P.; Petitpretz, P.; Parent, F.; Duroux, P.; Simonneau, G. Primary pulmonary hypertension and fenfluramine use. Br. Heart J. 1993, 70, 537–541. [Google Scholar] [CrossRef]
  30. Brenot, F. Primary pulmonary hypertension. Case series from France. Chest 1994, 105 (Suppl. S2), 33S–36S. [Google Scholar] [CrossRef]
  31. Abenhaim, L.; Moride, Y.; Brenot, F.; Rich, S.; Benichou, J.; Kurz, X.; Higenbottam, T.; Oakley, C.; Wouters, E.; Aubier, M.; et al. Appetite-suppressant drugs and the risk of primary pulmonary hypertension. International Primary Pulmonary Hypertension Study Group. N. Engl. J. Med. 1996, 335, 609–616. [Google Scholar] [CrossRef]
  32. Rothman, R.B.; Ayestas, M.A.; Dersch, C.M.; Baumann, M.H. Aminorex, fenfluramine, and chlorphentermine are serotonin transporter substrates. Implications for primary pulmonary hypertension. Circulation 1999, 100, 869–875. [Google Scholar] [CrossRef]
  33. Marasini, B.; Biondi, M.L.; Bianchi, E.; Dell’Orto, P.; Agostoni, A. Ketanserin treatment and serotonin in patients with primary and secondary Raynaud’s phenomenon. Eur. J. Clin. Pharmacol. 1988, 35, 419–421. [Google Scholar] [CrossRef]
  34. Klimiuk, P.S.; Grennan, A.; Weinkove, C.; Jayson, M.I. Platelet serotonin in systemic sclerosis. Ann. Rheum. Dis. 1989, 48, 586–589. [Google Scholar] [CrossRef] [PubMed]
  35. Herve, P.; Drouet, L.; Dosquet, C.; Launay, J.-M.; Rain, B.; Simonneau, G.; Caen, J.; Duroux, P. Primary pulmonary hypertension in a patient with a familial platelet storage pool disease: Role of serotonin. Am. J. Med. 1990, 89, 117–120. [Google Scholar] [CrossRef] [PubMed]
  36. Hervé, P.; Launay, J.-M.; Scrobohaci, M.-L.; Brenot, F.; Simonneau, G.; Petitpretz, P.; Poubeau, P.; Cerrina, J.; Duroux, P.; Drouet, L. Increased plasma serotonin in primary pulmonary hypertension. Am. J. Med. 1995, 99, 249–254. [Google Scholar] [CrossRef] [PubMed]
  37. Kéreveur, A.; Callebert, J.; Humbert, M.; Hervé, P.; Simonneau, G.; Launay, J.-M.; Drouet, L. High plasma serotonin levels in primary pulmonary hypertension. Effect of long-term epoprostenol (prostacyclin) therapy. Arterioscler. Thromb. Vasc. Biol. 2000, 20, 2233–2239. [Google Scholar] [CrossRef] [PubMed]
  38. Writing Committee Members; McLaughlin, V.V.; Archer, S.L.; Badesch, D.B.; Barst, R.J.; Farber, H.W.; Lindner, J.R.; Mathier, M.A.; McGoon, M.D.; Park, M.H.; et al. ACCF/AHA 2009 expert consensus document on pulmonary hypertension: A report of the American College of Cardiology Foundation Task Force on Expert Consensus Documents and the American Heart Association: Developed in collaboration with the American College of Chest Physicians, American Thoracic Society, Inc., and the Pulmonary Hypertension Association. Circulation 2009, 119, 2250–2294. [Google Scholar] [CrossRef]
  39. Tuder, R.M.; Archer, S.L.; Dorfmüller, P.; Erzurum, S.C.; Guignabert, C.; Michelakis, E.; Rabinovitch, M.; Schermuly, R.; Stenmark, K.R.; Morrell, N.W. Relevant issues in the pathology and pathobiology of pulmonary hypertension. J. Am. Coll. Cardiol. 2013, 62 (Suppl. S25), D4–D12. [Google Scholar] [CrossRef]
  40. Ivy, D.D.; Abman, S.H.; Barst, R.J.; Berger, R.M.; Bonnet, D.; Fleming, T.R.; Haworth, S.G.; Raj, J.U.; Rosenzweig, E.B.; Neick, I.S.; et al. Pediatric pulmonary hypertension. J. Am. Coll. Cardiol. 2013, 62 (Suppl. S25), D117–D126. [Google Scholar] [CrossRef]
  41. Ruoss, J.L.; Rios, D.R.; Levy, P.T. Updates on Management for Acute and Chronic Phenotypes of Neonatal Pulmonary Hypertension. Clin. Perinatol. 2020, 47, 593–615. [Google Scholar] [CrossRef] [PubMed]
  42. D’Alonzo, G.E.; Barst, R.J.; Ayres, S.M.; Bergofsky, E.H.; Brundage, B.H.; Detre, K.M.; Fishman, A.P.; Goldring, R.M.; Groves, B.M.; Kernis, J.T.; et al. Survival in patients with primary pulmonary hypertension. Results from a national prospective registry. Ann. Intern. Med. 1991, 115, 343–349. [Google Scholar] [CrossRef] [PubMed]
  43. Frank, D.B.; Crystal, M.A.; Morales, D.L.S.; Gerald, K.; Hanna, B.D.; Mallory, G.B.; Rossano, J.W. Trends in pediatric pulmonary hypertension-related hospitalizations in the United States from 2000–2009. Pulm Circ. 2015, 5, 339–348. [Google Scholar] [CrossRef] [PubMed]
  44. Maxwell, B.G.; Nies, M.K.; Ajuba-Iwuji, C.C.; Coulson, J.D.; Romer, L.H. Trends in Hospitalization for Pediatric Pulmonary Hypertension. Pediatrics 2015, 136, 241–250. [Google Scholar] [CrossRef] [PubMed]
  45. Haworth, S.G.; Hislop, A.A. Treatment and survival in children with pulmonary arterial hypertension: The UK Pulmonary Hypertension Service for Children 2001–2006. Heart 2009, 95, 312–317. [Google Scholar] [CrossRef]
  46. Eddahibi, S.; Hanoun, N.; Lanfumey, L.; Lesch, K.; Raffestin, B.; Hamon, M.; Adnot, S. Attenuated hypoxic pulmonary hypertension in mice lacking the 5-hydroxytryptamine transporter gene. J. Clin. Investig. 2000, 105, 1555–1562. [Google Scholar] [CrossRef]
  47. Morecroft, I.; Dempsie, Y.; Bader, M.; Walther, D.J.; Kotnik, K.; Loughlin, L.; Nilsen, M.; MacLean, M.R. Effect of tryptophan hydroxylase 1 deficiency on the development of hypoxia-induced pulmonary hypertension. Hypertension 2007, 49, 232–236. [Google Scholar] [CrossRef]
  48. Marcos, E.; Fadel, E.; Sanchez, O.; Humbert, M.; Dartevelle, P.; Simonneau, G.; Hamon, M.; Adnot, S.; Eddahibi, S. Serotonin-induced smooth muscle hyperplasia in various forms of human pulmonary hypertension. Circ. Res. 2004, 94, 1263–1270. [Google Scholar] [CrossRef]
  49. Eddahibi, S.; Humbert, M.; Fadel, E.; Raffestin, B.; Darmon, M.; Capron, F.; Simonneau, G.; Dartevelle, P.; Hamon, M.; Adnot, S. Serotonin transporter overexpression is responsible for pulmonary artery smooth muscle hyperplasia in primary pulmonary hypertension. J. Clin. Investig. 2001, 108, 1141–1150. [Google Scholar] [CrossRef]
  50. Guignabert, C.; Izikki, M.; Tu, L.I.; Li, Z.; Zadigue, P.; Barlier-Mur, A.-M.; Hanoun, N.; Rodman, D.; Hamon, M.; Adnot, S.; et al. Transgenic mice overexpressing the 5-hydroxytryptamine transporter gene in smooth muscle develop pulmonary hypertension. Circ. Res. 2006, 98, 1323–1330. [Google Scholar] [CrossRef]
  51. MacLean, M.R.; Deuchar, G.A.; Hicks, M.N.; Morecroft, I.; Shen, S.; Sheward, J.; Colston, J.; Loughlin, L.; Nilsen, M.; Dempsie, Y.; et al. Overexpression of the 5-hydroxytryptamine transporter gene: Effect on pulmonary hemodynamics and hypoxia-induced pulmonary hypertension. Circulation 2004, 109, 2150–2155. [Google Scholar] [CrossRef] [PubMed]
  52. Rondelet, B.; Van Beneden, R.; Kerbaul, F.; Motte, S.; Fesler, P.; McEntee, K.; Brimioulle, S.; Ketelslegers, J.-M.; Naeije, R. Expression of the serotonin 1b receptor in experimental pulmonary hypertension. Eur. Respir. J. 2003, 22, 408–412. [Google Scholar] [CrossRef] [PubMed]
  53. Hironaka, E.; Hongo, M.; Sakai, A.; Mawatari, E.; Terasawa, F.; Okumura, N.; Yamazaki, A.; Ushiyama, Y.; Yazaki, Y.; Kinoshita, O. Serotonin receptor antagonist inhibits monocrotaline-induced pulmonary hypertension and prolongs survival in rats. Cardiovasc. Res. 2003, 60, 692–699. [Google Scholar] [CrossRef] [PubMed]
  54. Hood, K.Y.; Mair, K.M.; Harvey, A.P.; Montezano, A.C.; Touyz, R.M.; MacLean, M.R. Serotonin Signaling Through the 5-HT1B Receptor and NADPH Oxidase 1 in Pulmonary Arterial Hypertension. Arter. Thromb. Vasc. Biol. 2017, 37, 1361–1370. [Google Scholar] [CrossRef]
  55. Lazarus, H.M.; Denning, J.; Wring, S.; Palacios, M.; Hoffman, S.; Crizer, K.; Kamau-Kelley, W.; Symonds, W.; Feldman, J. A trial design to maximize knowledge of the effects of rodatristat ethyl in the treatment of pulmonary arterial hypertension (ELEVATE 2). Pulm. Circ. 2022, 12, e12088. [Google Scholar] [CrossRef]
  56. Lazarus, H.; Denning, J.; Kamau-Kelley, W.; Wring, S.; Palacios, M.; Humbert, M. ELEVATE 2: A multicenter study of rodatristat ethyl in patients with WHO Group 1 pulmonary arterial hypertension (PAH). Eur. Respir. J. 2021, 58 (Suppl. S65). [Google Scholar] [CrossRef]
  57. Robson, J.M.; Senior, J.B. The 5-Hydroxytryptamine content of the placenta and foetus during pregnancy in mice. Br. J. Pharmacol. Chemother. 1964, 22, 380–391. [Google Scholar] [CrossRef]
  58. Badawy, A.A.B. Tryptophan metabolism, disposition and utilization in pregnancy. Biosci. Rep. 2015, 35, e00261. [Google Scholar] [CrossRef]
  59. Sano, M.; Ferchaud-Roucher, V.; Kaeffer, B.; Poupeau, G.; Castellano, B.; Darmaun, D. Maternal and fetal tryptophan metabolism in gestating rats: Effects of intrauterine growth restriction. Amino Acids. 2016, 48, 281–290. [Google Scholar] [CrossRef]
  60. Abad, C.; Karahoda, R.; Kastner, P.; Portillo, R.; Horackova, H.; Kucera, R.; Nachtigal, P.; Staud, F. Profiling of Tryptophan Metabolic Pathways in the Rat Fetoplacental Unit During Gestation. Int. J. Mol. Sci. 2020, 21, 7578. [Google Scholar] [CrossRef]
  61. Anderson, G.M.; Czarkowski, K.; Ravski, N.; Epperson, C.N. Platelet serotonin in newborns and infants: Ontogeny, heritability, and effect of in utero exposure to selective serotonin reuptake inhibitors. Pediatr. Res. 2004, 56, 418–422. [Google Scholar] [CrossRef] [PubMed]
  62. Oberlander, T.F.; Warburton, W.; Misri, S.; Aghajanian, J.; Hertzman, C. Neonatal outcomes after prenatal exposure to selective serotonin reuptake inhibitor antidepressants and maternal depression using population-based linked health data. Arch. Gen. Psychiatry 2006, 63, 898–906. [Google Scholar] [CrossRef] [PubMed]
  63. Bonnin, A.; Torii, M.; Wang, L.; Rakic, P.; Levitt, P. Serotonin modulates the response of embryonic thalamocortical axons to netrin-1. Nat. Neurosci. 2007, 10, 588–597. [Google Scholar] [CrossRef] [PubMed]
  64. Oberlander, T.F.; Gingrich, J.A.; Ansorge, M.S. Sustained neurobehavioral effects of exposure to SSRI antidepressants during development: Molecular to clinical evidence. Clin. Pharmacol. Ther. 2009, 86, 672–677. [Google Scholar] [CrossRef]
  65. Davidson, S.; Prokonov, D.; Taler, M.; Maayan, R.; Harell, D.; Gil-Ad, I.; Weizman, A. Effect of Exposure to Selective Serotonin Reuptake Inhibitors In Utero on Fetal Growth: Potential Role for the IGF-I and HPA Axes. Pediatr. Res. 2009, 65, 236–241. [Google Scholar] [CrossRef]
  66. Narboux-Nême, N.; Angenard, G.; Mosienko, V.; Klempin, F.; Pitychoutis, P.M.; Deneris, E.; Bader, M.; Giros, B.; Alenina, N.; Gaspar, P. Postnatal growth defects in mice with constitutive depletion of central serotonin. ACS Chem. Neurosci. 2013, 4, 171–181. [Google Scholar] [CrossRef]
  67. Ranzil, S.; Ellery, S.; Walker, D.W.; Vaillancourt, C.; Alfaidy, N.; Bonnin, A.; Borg, A.; Wallace, E.M.; Ebeling, P.R.; Erwich, J.J.; et al. Disrupted placental serotonin synthetic pathway and increased placental serotonin: Potential implications in the pathogenesis of human fetal growth restriction. Placenta 2019, 84, 74–83. [Google Scholar] [CrossRef]
  68. Rosenfeld, C.S. Placental serotonin signaling, pregnancy outcomes, and regulation of fetal brain development†. Biol. Reprod. 2020, 102, 532–538. [Google Scholar] [CrossRef]
  69. Domingues, R.R.; Beard, A.D.; Connelly, M.K.; Wiltbank, M.C.; Hernandez, L.L. Fluoxetine-induced perinatal morbidity in a sheep model. Front. Med. 2022, 9, 955560. [Google Scholar] [CrossRef]
  70. Lauder, J.M.; Krebs, H. Serotonin as a differentiation signal in early neurogenesis. Dev. Neurosci. 1978, 1, 15–30. [Google Scholar] [CrossRef]
  71. Rakic, P.; Lidow, M.S. Distribution and density of monoamine receptors in the primate visual cortex devoid of retinal input from early embryonic stages. J. Neurosci. 1995, 15 Pt 2, 2561–2574. [Google Scholar] [CrossRef]
  72. Vitalis, T.; Cases, O.; Passemard, S.; Callebert, J.; Parnavelas, J.G. Embryonic depletion of serotonin affects cortical development. Eur. J. Neurosci. 2007, 26, 331–344. [Google Scholar] [CrossRef] [PubMed]
  73. Peters, D.A. Maternal stress increases fetal brain and neonatal cerebral cortex 5-hydroxytryptamine synthesis in rats: A possible mechanism by which stress influences brain development. Pharmacol. Biochem. Behav. 1990, 35, 943–947. [Google Scholar] [CrossRef] [PubMed]
  74. Miller, C.L.; Murakami, P.; Ruczinski, I.; Ross, R.G.; Sinkus, M.; Sullivan, B.; Leonard, S. Two complex genotypes relevant to the kynurenine pathway and melanotropin function show association with schizophrenia and bipolar disorder. Schizophr. Res. 2009, 113, 259–267. [Google Scholar] [CrossRef]
  75. Tsuang, D.W.; Bird, T.D. Genetic factors in neurodegenerative diseases. Am. J. Med. Genet. B Neuropsychiatr. Genet. 2017, 174, 3–4. [Google Scholar] [CrossRef]
  76. Cutz, E.; Gillan, J.E.; Bryan, A.C. Neuroendocrine cells in the developing human lung: Morphologic and functional considerations. Pediatr. Pulmonol. 1985, 1 (Suppl. S3), S21–S29. [Google Scholar] [PubMed]
  77. Garg, A.; Sui, P.; Verheyden, J.M.; Young, L.R.; Sun, X. Consider the lung as a sensory organ: A tip from pulmonary neuroendocrine cells. Curr. Top. Dev. Biol. 2019, 132, 67–89. [Google Scholar] [CrossRef]
  78. Boers, J.E.; den Brok, J.L.; Koudstaal, J.; Arends, J.W.; Thunnissen, F.B. Number and proliferation of neuroendocrine cells in normal human airway epithelium. Am. J. Respir. Crit. Care Med. 1996, 154 Pt 1, 758–763. [Google Scholar] [CrossRef]
  79. Travaglini, K.J.; Nabhan, A.N.; Penland, L.; Sinha, R.; Gillich, A.; Sit, R.V.; Chang, S.; Conley, S.D.; Mori, Y.; Seita, J.; et al. A molecular cell atlas of the human lung from single-cell RNA sequencing. Nature 2020, 587, 619–625. [Google Scholar] [CrossRef]
  80. Karahoda, R.; Horackova, H.; Kastner, P.; Matthios, A.; Cerveny, L.; Kucera, R.; Kacerovsky, M.; Tebbens, J.D.; Bonnin, A.; Abad, C.; et al. Serotonin homeostasis in the materno-foetal interface at term: Role of transporters (SERT/SLC6A4 and OCT3/SLC22A3) and monoamine oxidase A (MAO-A) in uptake and degradation of serotonin by human and rat term placenta. Acta Physiol. 2020, 229, e13478. [Google Scholar] [CrossRef]
  81. Castro, E.C.C.; Sen, P.; Parks, W.T.; Langston, C.; Galambos, C. The Role of Serotonin Transporter in Human Lung Development and in Neonatal Lung Disorders. Can. Respir. J. 2017, 2017, 9064046. [Google Scholar] [CrossRef]
  82. Delaney, C.; Gien, J.; Grover, T.R.; Roe, G.; Abman, S.H. Pulmonary vascular effects of serotonin and selective serotonin reuptake inhibitors in the late-gestation ovine fetus. Am. J. Physiol. Lung Cell Mol. Physiol. 2011, 301, L937–L944. [Google Scholar] [CrossRef]
  83. Hodge, E.; Nelson, C.P.; Miller, S.; Billington, C.K.; Stewart, C.E.; Swan, C.; Malarstig, A.; Henry, A.P.; Gowland, C.; Melén, E.; et al. HTR4 gene structure and altered expression in the developing lung. Respir. Res. 2013, 14, 77. [Google Scholar] [CrossRef]
  84. Nikolić, J.; Vukojević, K.; Šoljić, V.; Mišković, J.; Vlaho, M.O.; Saraga-Babić, M.; Filipović, N. Expression Patterns of Serotonin Receptors 5-HT1A, 5-HT2A, and 5-HT3A during Human Fetal Lung Development. Int. J. Mol. Sci. 2023, 24, 2965. [Google Scholar] [CrossRef]
  85. Morecroft, I.; MacLean, M.R. 5-hydroxytryptamine receptors mediating vasoconstriction and vasodilation in perinatal and adult rabbit small pulmonary arteries. Br. J. Pharmacol. 1998, 125, 69–78. [Google Scholar] [CrossRef] [PubMed]
  86. Goyal, R.; Papamatheakis, D.G.; Loftin, M.; Vrancken, K.; Dawson, A.S.; Osman, N.J.; Blood, A.B.; Pearce, W.J.; Longo, L.D.; Wilson, S.M. Long-term maternal hypoxia: The role of extracellular Ca2+ entry during serotonin-mediated contractility in fetal ovine pulmonary arteries. Reprod. Sci. 2011, 18, 948–962. [Google Scholar] [CrossRef] [PubMed]
  87. Barst, R.J.; Ertel, S.I.; Beghetti, M.; Ivy, D.D. Pulmonary arterial hypertension: A comparison between children and adults. Eur. Respir. J. 2011, 37, 665–677. [Google Scholar] [CrossRef] [PubMed]
  88. Dawes, G.S.; Mott, J.C.; Widdicombe, J.G.; Wyatt, D.G. Changes in the lungs of the new-born lamb. J. Physiol. 1953, 121, 141–162. [Google Scholar] [CrossRef]
  89. Rudolph, A.M. Distribution and regulation of blood flow in the fetal and neonatal lamb. Circ. Res. 1985, 57, 811–821. [Google Scholar] [CrossRef]
  90. Fraisse, A.; Jais, X.; Schleich, J.-M.; di Filippo, S.; Maragnès, P.; Beghetti, M.; Gressin, V.; Voisin, M.; Dauphin, C.; Clerson, P.; et al. Characteristics and prospective 2-year follow-up of children with pulmonary arterial hypertension in France. Arch. Cardiovasc. Dis. 2010, 103, 66–74. [Google Scholar] [CrossRef] [PubMed]
  91. Barst, R.J.; McGoon, M.D.; Elliott, C.G.; Foreman, A.J.; Miller, D.P.; Ivy, D.D. Survival in childhood pulmonary arterial hypertension: Insights from the registry to evaluate early and long-term pulmonary arterial hypertension disease management. Circulation 2012, 125, 113–122. [Google Scholar] [CrossRef] [PubMed]
  92. Berger, R.M.; Beghetti, M.; Humpl, T.; Raskob, G.E.; Ivy, D.D.; Jing, Z.-C.; Bonnet, D.; Schulze-Neick, I.; Barst, R.J. Clinical features of paediatric pulmonary hypertension: A registry study. Lancet 2012, 379, 537–546. [Google Scholar] [CrossRef] [PubMed]
  93. Mitchell, A.A.; Gilboa, S.M.; Werler, M.M.; Kelley, K.E.; Louik, C.; Hernández-Díaz, S.; National Birth Defects Prevention Study. Medication use during pregnancy, with particular focus on prescription drugs: 1976–2008. Am. J. Obstet. Gynecol. 2011, 205, 51.e1–51.e8. [Google Scholar] [CrossRef] [PubMed]
  94. Huybrechts, K.F.; Palmsten, K.; Mogun, H.; Kowal, M.; Avorn, J.; Setoguchi-Iwata, S.; Hernández-Díaz, S. National trends in antidepressant medication treatment among publicly insured pregnant women. Gen. Hosp. Psychiatry 2013, 35, 265–271. [Google Scholar] [CrossRef]
  95. Huybrechts, K.F.; Bateman, B.T.; Palmsten, K.; Desai, R.J.; Patorno, E.; Gopalakrishnan, C.; Levin, R.; Mogun, H.; Hernandez-Diaz, S. Antidepressant use late in pregnancy and risk of persistent pulmonary hypertension of the newborn. JAMA 2015, 313, 2142–2151. [Google Scholar] [CrossRef]
  96. Epperson, C.N.; Jatlow, P.I.; Czarkowski, K.; Anderson, G.M. Maternal fluoxetine treatment in the postpartum period: Effects on platelet serotonin and plasma drug levels in breastfeeding mother-infant pairs. Pediatrics 2003, 112, e425. [Google Scholar] [CrossRef]
  97. Ewing, G.; Tatarchuk, Y.; Appleby, D.; Schwartz, N.; Kim, D. Placental transfer of antidepressant medications: Implications for postnatal adaptation syndrome. Clin. Pharmacokinet. 2015, 54, 359–370. [Google Scholar] [CrossRef]
  98. Horackova, H.; Karahoda, R.; Cerveny, L.; Vachalova, V.; Ebner, R.; Abad, C.; Staud, F. Effect of Selected Antidepressants on Placental Homeostasis of Serotonin: Maternal and Fetal Perspectives. Pharmaceutics 2021, 13, 1306. [Google Scholar] [CrossRef]
  99. Suhara, T.; Sudo, Y.; Yoshida, K.; Okubo, Y.; Fukuda, H.; Obata, T.; Yoshikawa, K.; Suzuki, K.; Sasaki, Y. Lung as reservoir for antidepressants in pharmacokinetic drug interactions. Lancet 1998, 351, 332–335. [Google Scholar] [CrossRef]
  100. Finkel, M.S.; Laghrissi-Thode, F.; Pollock, B.G.; Rong, J. Paroxetine is a novel nitric oxide synthase inhibitor. Psychopharmacol. Bull. 1996, 32, 653–658. [Google Scholar]
  101. Ikenouchi, A.; Okamoto, N.; Konno, Y.; Fujii, R.; Fujino, Y.; Yoshimura, R. Influence of antidepressants on plasma levels of nitric oxide metabolites in patients with major depressive disorder. BJPsych Open 2021, 8, e14. [Google Scholar] [CrossRef]
  102. Rurak, D.; Lim, K.; Sanders, A.; Brain, U.; Riggs, W.; Oberlander, T.F. Third trimester fetal heart rate and Doppler middle cerebral artery blood flow velocity characteristics during prenatal selective serotonin reuptake inhibitor exposure. Pediatr. Res. 2011, 70, 96–101. [Google Scholar] [CrossRef] [PubMed]
  103. Chambers, C.D.; Hernandez-Diaz, S.; Van Marter, L.J.; Werler, M.M.; Louik, C.; Jones, K.L.; Mitchell, A.A. Selective serotonin-reuptake inhibitors and risk of persistent pulmonary hypertension of the newborn. N. Engl. J. Med. 2006, 354, 579–587. [Google Scholar] [CrossRef] [PubMed]
  104. Reis, M.; Källén, B. Delivery outcome after maternal use of antidepressant drugs in pregnancy: An update using Swedish data. Psychol. Med. 2010, 40, 1723–1733. [Google Scholar] [CrossRef]
  105. Galbally, M.; Gentile, S.; Lewis, A.J. Further findings linking SSRIs during pregnancy and persistent pulmonary hypertension of the newborn: Clinical implications. CNS Drugs 2012, 26, 813–822. [Google Scholar] [CrossRef]
  106. Grigoriadis, S.; VonderPorten, E.H.; Mamisashvili, L.; Tomlinson, G.; Dennis, C.-L.; Koren, G.; Steiner, M.; Mousmanis, P.; Cheung, A.; Ross, L.E. Prenatal exposure to antidepressants and persistent pulmonary hypertension of the newborn: Systematic review and meta-analysis. BMJ 2014, 348, f6932. [Google Scholar] [CrossRef]
  107. Chambers, C.D.; Johnson, K.A.; Dick, L.M.; Felix, R.J.; Jones, K.L. Birth outcomes in pregnant women taking fluoxetine. N. Engl. J. Med. 1996, 335, 1010–1015. [Google Scholar] [CrossRef] [PubMed]
  108. Källén, B.; Olausson, P.O. Maternal use of selective serotonin re-uptake inhibitors and persistent pulmonary hypertension of the newborn. Pharmacoepidemiol. Drug Saf. 2008, 17, 801–806. [Google Scholar] [CrossRef]
  109. Kieler, H.; Artama, M.; Engeland, A.; Ericsson, Ö.; Furu, K.; Gissler, M.; Nielsen, R.B.; Nørgaard, M.; Stephansson, O.; Valdimarsdottir, U.; et al. Selective serotonin reuptake inhibitors during pregnancy and risk of persistent pulmonary hypertension in the newborn: Population based cohort study from the five Nordic countries. BMJ 2012, 344, d8012. [Google Scholar] [CrossRef]
  110. Andrade, S.E.; McPhillips, H.; Loren, D.; Raebel, M.A.; Lane, K.; Livingston, J.; Boudreau, D.M.; Smith, D.H.; Davis, R.L.; Willy, M.E.; et al. Antidepressant medication use and risk of persistent pulmonary hypertension of the newborn. Pharmacoepidemiol. Drug Saf. 2009, 18, 246–252. [Google Scholar] [CrossRef]
  111. Wichman, C.L.; Moore, K.M.; Lang, T.R.; St Sauver, J.L.; Heise, R.H., Jr.; Watson, W.J. Congenital heart disease associated with selective serotonin reuptake inhibitor use during pregnancy. Mayo Clin. Proc. 2009, 84, 23–27. [Google Scholar] [CrossRef] [PubMed]
  112. Wilson, K.L.; Zelig, C.M.; Harvey, J.P.; Cunningham, B.S.; Dolinsky, B.M.; Napolitano, P.G. Persistent pulmonary hypertension of the newborn is associated with mode of delivery and not with maternal use of selective serotonin reuptake inhibitors. Am. J. Perinatol. 2011, 28, 19–24. [Google Scholar] [CrossRef] [PubMed]
  113. Ng, Q.X.; Venkatanarayanan, N.; Ho, C.Y.X.; Sim, W.S.; Lim, D.Y.; Yeo, W.S. Selective Serotonin Reuptake Inhibitors and Persistent Pulmonary Hypertension of the Newborn: An Updated Meta-Analysis. J. Womens Health 2019, 28, 331–338. [Google Scholar] [CrossRef]
  114. Masarwa, R.; Bar-Oz, B.; Gorelik, E.; Reif, S.; Perlman, A.; Matok, I. Prenatal exposure to selective serotonin reuptake inhibitors and serotonin norepinephrine reuptake inhibitors and risk for persistent pulmonary hypertension of the newborn: A systematic review, meta-analysis, and network meta-analysis. Am. J. Obstet. Gynecol. 2019, 220, 57.e1–57.e13. [Google Scholar] [CrossRef]
  115. Austin, M.P. To treat or not to treat: Maternal depression, SSRI use in pregnancy and adverse neonatal effects. Psychol. Med. 2006, 36, 1663–1670. [Google Scholar] [CrossRef]
  116. da-Silva, V.A.; Altenburg, S.P.; Malheiros, L.R.; Thomaz, T.G.; Lindsey, C.J. Postnatal development of rats exposed to fluoxetine or venlafaxine during the third week of pregnancy. Braz. J. Med. Biol. Res. 1999, 32, 93–98. [Google Scholar] [CrossRef]
  117. Coleman, F.H.; Christensen, H.D.; Gonzalez, C.L.; Rayburn, W.F. Behavioral changes in developing mice after prenatal exposure to paroxetine (Paxil). Am. J. Obstet. Gynecol. 1999, 181 Pt 1, 1166–1171. [Google Scholar] [CrossRef]
  118. Morrison, J.L.; Chien, C.; Riggs, K.W.; Gruber, N.; Rurak, D. Effect of maternal fluoxetine administration on uterine blood flow, fetal blood gas status, and growth. Pediatr. Res. 2002, 51, 433–442. [Google Scholar] [CrossRef] [PubMed]
  119. Fornaro, E.; Li, D.; Pan, J.; Belik, J. Prenatal exposure to fluoxetine induces fetal pulmonary hypertension in the rat. Am. J. Respir. Crit. Care Med. 2007, 176, 1035–1040. [Google Scholar] [CrossRef]
  120. Delaney, C.; Gien, J.; Roe, G.; Isenberg, N.; Kailey, J.; Abman, S.H. Serotonin contributes to high pulmonary vascular tone in a sheep model of persistent pulmonary hypertension of the newborn. Am. J. Physiol. Lung Cell Mol. Physiol. 2013, 304, L894–L901. [Google Scholar] [CrossRef]
  121. Hooper, C.W.; Delaney, C.; Streeter, T.; Yarboro, M.T.; Poole, S.; Brown, N.; Slaughter, J.C.; Cotton, R.B.; Reese, J.; Shelton, E.L. Selective serotonin reuptake inhibitor exposure constricts the mouse ductus arteriosus in utero. Am. J. Physiol. Heart Circ. Physiol. 2016, 311, H572–H581. [Google Scholar] [CrossRef]
  122. Johnson, D.E.; Kulik, T.J.; Lock, J.E.; Elde, R.P.; Thompson, T.R. Bombesin-, calcitonin-, and serotonin-immunoreactive pulmonary neuroendocrine cells in acute and chronic neonatal lung disease. Pediatr. Pulmonol. 1985, 1 (Suppl. S3), S13–S20. [Google Scholar] [PubMed]
  123. Breuer, J.; Georgaraki, A.; Sieverding, L.; Baden, W.; Apitz, J. Increased turnover of serotonin in children with pulmonary hypertension secondary to congenital heart disease. Pediatr. Cardiol. 1996, 17, 214–219. [Google Scholar] [CrossRef]
  124. de la Roque, E.D.; Smeralda, G.; Quignard, J.-F.; Freund-Michel, V.; Courtois, A.; Marthan, R.; Muller, B.; Guibert, C.; Dubois, M. Altered vasoreactivity in neonatal rats with pulmonary hypertension associated with bronchopulmonary dysplasia: Implication of both eNOS phosphorylation and calcium signaling. PLoS ONE 2017, 12, e0173044. [Google Scholar] [CrossRef] [PubMed]
  125. Delaney, C.; Sherlock, L.; Fisher, S.; Maltzahn, J.; Wright, C.; Nozik-Grayck, E. Serotonin 2A receptor inhibition protects against the development of pulmonary hypertension and pulmonary vascular remodeling in neonatal mice. Am. J. Physiol. Lung Cell Mol. Physiol. 2018, 314, L871–L881. [Google Scholar] [CrossRef]
  126. Hofmann, A.D.; Friedmacher, F.; Hunziker, M.; Takahashi, H.; Duess, J.W.; Gosemann, J.-H.; Puri, P. Upregulation of serotonin-receptor-2a and serotonin transporter expression in the pulmonary vasculature of nitrofen-induced congenital diaphragmatic hernia. J. Pediatr. Surg. 2014, 49, 871–874, discussion 874–875. [Google Scholar] [CrossRef] [PubMed]
  127. Roberts, D.S.; Sherlock, L.G.; Posey, J.N.; Archambault, J.L.; Nozik, E.S.; Delaney, C.A. Serotonin-deficient neonatal mice are not protected against the development of experimental bronchopulmonary dysplasia or pulmonary hypertension. Physiol. Rep. 2022, 10, e15482. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Serotonin (5-HT) is synthesized from the amino acid tryptophan. L-tryptophan is converted to 5-hydroxytryptophan (5-HTP) by tryptophan hydroxylase (TPH). TPH1 exists in the enterochromaffin cells of the small intestine and TPH2 is primarily found in the central and enteric nervous systems. 5-HTP is converted to 5-HT by amino acid decarboxylase (AAAD).
Figure 1. Serotonin (5-HT) is synthesized from the amino acid tryptophan. L-tryptophan is converted to 5-hydroxytryptophan (5-HTP) by tryptophan hydroxylase (TPH). TPH1 exists in the enterochromaffin cells of the small intestine and TPH2 is primarily found in the central and enteric nervous systems. 5-HTP is converted to 5-HT by amino acid decarboxylase (AAAD).
Biomedicines 11 03049 g001
Figure 2. Serotonin (5-HT) is either metabolized by monoamine oxidase to 5-hydroxyindoleacetic acid (5-HIAA) or circulates in the bloodstream. The majority of 5-HT is metabolized on first pass by the liver, with much of the remainder metabolized by the lungs. In addition, 90% of unmetabolized 5-HT is taken up into platelets via the 5-HT transporter (SERT). Only 10% of circulating 5-HT is free in the plasma.
Figure 2. Serotonin (5-HT) is either metabolized by monoamine oxidase to 5-hydroxyindoleacetic acid (5-HIAA) or circulates in the bloodstream. The majority of 5-HT is metabolized on first pass by the liver, with much of the remainder metabolized by the lungs. In addition, 90% of unmetabolized 5-HT is taken up into platelets via the 5-HT transporter (SERT). Only 10% of circulating 5-HT is free in the plasma.
Biomedicines 11 03049 g002
Figure 3. The lung participates in serotonin (5-HT) synthesis, uptake, and metabolism. 5-HT is synthesized by pulmonary artery endothelial cells (PA-EC) and pulmonary neuroendocrine cells (PNEC). 5-HT is taken up via SERT by both endothelial and smooth muscle cells of the pulmonary arteries. Pulmonary artery smooth muscle cells (PA-SMC) also express G-protein-coupled receptors which, upon binding 5-HT, contribute to vasoconstriction and smooth muscle cell proliferation seen in pulmonary hypertension (PH).
Figure 3. The lung participates in serotonin (5-HT) synthesis, uptake, and metabolism. 5-HT is synthesized by pulmonary artery endothelial cells (PA-EC) and pulmonary neuroendocrine cells (PNEC). 5-HT is taken up via SERT by both endothelial and smooth muscle cells of the pulmonary arteries. Pulmonary artery smooth muscle cells (PA-SMC) also express G-protein-coupled receptors which, upon binding 5-HT, contribute to vasoconstriction and smooth muscle cell proliferation seen in pulmonary hypertension (PH).
Biomedicines 11 03049 g003
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Archambault, J.L.; Delaney, C.A. A Review of Serotonin in the Developing Lung and Neonatal Pulmonary Hypertension. Biomedicines 2023, 11, 3049. https://doi.org/10.3390/biomedicines11113049

AMA Style

Archambault JL, Delaney CA. A Review of Serotonin in the Developing Lung and Neonatal Pulmonary Hypertension. Biomedicines. 2023; 11(11):3049. https://doi.org/10.3390/biomedicines11113049

Chicago/Turabian Style

Archambault, Jamie L., and Cassidy A. Delaney. 2023. "A Review of Serotonin in the Developing Lung and Neonatal Pulmonary Hypertension" Biomedicines 11, no. 11: 3049. https://doi.org/10.3390/biomedicines11113049

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

Article Metrics

Back to TopTop