Abstract
The developing brain, particularly in premature infants, is highly susceptible to environmental and pharmacological influences. Premature neonates often require prolonged stays in the NICU, where midazolam (MDZ), a benzodiazepine, is commonly used as a sedative, despite concerns raised by the FDA in 2016 regarding its potential neurological complications in infants. Understanding the long-term effects of MDZ on these vulnerable patients is hindered by ethical considerations and limited research. This review emphasizes the vulnerability of premature infants to sedation and anesthesia and outlines how early exposure to MDZ can impact brain development at both molecular and behavioral levels, drawing from clinical and preclinical data. Additionally, we highlighted existing knowledge gaps and suggested avenues for further research to better comprehend the enduring consequences of MDZ exposure on neurodevelopment in this population.
Introduction
In the Neonatal Intensive Care Unit (NICU), where medication choices are constantly evolving, one major concern for caregivers is the early exposure of neonates to anesthesia and sedatives. This is particularly crucial given the vulnerability of neonates during the critical period of brain development.
Midazolam (MDZ), the most used sedative for neonates approved by the FDA, is widely administered in the NICU [1, 2]. Initially considered a safe sedative, recent research since 2003 has raised concerns about its neural complications.
This review aims to present current evidence on the effects of MDZ, alone or in combination with other anesthetics, on the neurodevelopment of animals and humans. We will begin by briefly outlining the mechanism, pharmacodynamics, and pharmacokinetics of MDZ. Subsequently, we will delve into the effects of MDZ on children, encompassing both preclinical and clinical evidence of altered behavior. Following this, we will explore the impact of MDZ on neural biology systems, including neuronal, glial, dendritic, and synaptic functions. Finally, we will discuss existing limitations and propose areas for future research.
The vulnerabilities of premature brain
Preterm infant is defined as an infant born prior to 37 weeks’ gestation. Such infants are categorized based on their gestational age as extremely preterm (<28 weeks), very preterm (28–31 weeks), moderately preterm (32–33 weeks), and late preterm (34–36 weeks) [3]. In 2022, approximately 10 % of infants were born preterm, with a rise by 4 % in the 2020–2021 period [4]. During pregnancy, the final months and weeks play a crucial role in the development of vital organs such as the brain, lungs, and liver. Babies born prematurely, especially before 32 weeks, exhibit higher rates of mortality and disability [5]. In 2020, preterm birth and low birth weight accounted for about 16 % of infant deaths (deaths before 1 year). Surviving neonates may have breathing problems, feeding difficulties, cerebral palsy, developmental delay, vision problems, and hearing problems [4].
The Neonatal Intensive Care Unit (NICU) is a pivotal environment for managing and providing essential care to preterm infants, with the duration of stay varying based on factors such as birth weight, gestational age, and maternal complications. Notably, infants weighing less than 1,000 g may spend an average of 79 days [6] in the NICU, whereas full-term infants typically have a mean stay of 10 days [7]. Prolonged NICU stays are influenced by factors like growth retardation, infections, feeding issues, and respiratory complications [7–10].
The last trimester period of development is crucial for normal brain development. During this period, the human brain undergoes significant growth, doubling in size by 6 months and tripling by 12 months after birth [11–13]. Tremendous changes can occur even before birth. Neuroimaging studies reveal substantial differences in brain structure between 28 and 38 weeks of gestation, emphasizing the intricate nature of early brain development [11–13]. Specifically, at the 28th week of gestation, the brain is small and simple, with less folding than the brain of full-term infants (at the 38th week of gestation). The speed and complexity of the neurobiological processes governing early brain development place them at higher risk of developmental adversity.
Neurobiological processes, including neurogenesis, neuron migration, neuroapoptosis, synapse proliferation, pruning, and myelination, shape the developing brain [14–16]. The process of neurogenesis begins in the early stages of gestation following conception, with neuron migration occurring between the 12th and 20th weeks of gestation, facilitated by the development in the germinal matrix and subventricular zone [15]. Subsequently, cortical neurons navigate along a scaffold of glial cells to reach their final destinations. Following the migration, neuroapoptosis, characterized by programmed cell death, becomes notably active from 24 weeks of gestation until approximately 4 weeks after birth [14]. An additional crucial neurodevelopmental phase involves the proliferation of neuronal synapses, beginning around the 20th week of gestation and progressing rapidly. Synapse numbers peak at approximately 1–2 years of age, with figures 50 % higher than those observed in adults [14, 17, 18]. Following this peak, certain synapses undergo pruning, eliminating “unneeded” neurons, resulting in the loss of synaptic connections. The fourth major neurodevelopmental process, myelination, initiates towards the end of the second trimester and continues more gradually throughout childhood.
Notably, neonates in the NICU also have a high incidence of neonatal brain injuries. Some common brain injuries that preterm infants often encounter are intraventricular hemorrhage (IVH) or periventricular leukomalacia (PVL) [19]. IVH, a leading cause of brain damage in preterm newborns, occurs when the germinal matrix and subventricular zone sustain injury [19]. This vulnerability arises from the immaturity of blood vessels in the highly vascular germinal matrix, coupled with poor tissue vascular support, making this region especially prone to IVH [19, 20]. PVL is another important brain lesion that affects preterm newborns, which further can cause cerebral palsy and cognitive, behavioral, and attention deficits [19]. This type of injury specifically affects the cerebral white matter, resulting in decreased volumes in areas like the cortex, thalamus, and basal ganglia [19]. The lesion typically begins as a focal point in deep periventricular white matter zones due to neuronal necrosis, reactive gliosis, and microglial activation [19].
Given the complexities of these developmental phases, exposure to chemicals, including anesthetics and sedatives, may disrupt the normal morphogenesis of brain structures, leading to significant long-term damage. The heightened activity during synaptogenesis, synaptic pruning, and myelination – referred to as the brain’s growth spurt – in the second and third trimesters makes the fetal brain highly sensitive to anesthetic and sedative agents [21, 22]. Premature infants, often born during or before this growth spurt, are particularly vulnerable to the neurotoxic impacts of these drugs, potentially resulting in enduring cognitive impairment [21–24]. The underdeveloped state of neonatal organ systems, including the central nervous system (CNS), renders premature infants more prone to the depressant effects of these drugs, potentially inducing cerebral hypoperfusion and metabolic disruptions [23]. While the specific molecular mechanisms underlying this vulnerability are not yet fully understood [24], factors such as the immaturity of neurotransmitter systems and the delicate balance of neurochemical processes during synaptogenesis contribute to the heightened sensitivity of the premature brain to anesthetic neurotoxicity. The debated clinical significance arises as some children exposed to multiple and prolonged anesthesia report deficits larger than those with single exposures [25–28]. Further investigation is essential to elucidate the connection between exposure and brain cell loss and to formulate protective strategies.
Overview of midazolam
Among all benzodiazepines (BDZ), MDZ is the preferred choice for inducing sedation in neonates, primarily due to its rapid onset of action and shorter half-life. MDZ is the first-identified water-soluble BDZ, featuring an imidazole ring fused to a benzene ring integrated with a seven-membered diazepine ring. MDZ structure is unique in a way that the environmental pH-dependent ring-opening phenomenon characterizes its property. MDZ is highly water-soluble in a mild acidic environment (pH<4.0). Once the pH increases to physiological pH, the MDZ’s imidazole ring closes, which makes it much more lipid soluble. This unique structural characteristic is a primary contributor to making MDZ accessible to the blood-brain barrier and executing the rapid onset action of the mechanism [29–31]. A summary of MDZ structure, pharmacokinetics, and pharmacodynamics is presented in Figure 1.

Overview of midazolam metabolism and mechanism of action. Midazolam (MDZ) can be administered orally, intranasally, or intravenously. Once MDZ is in the bloodstream, it can be bound to albumin and transported to either the brain, where it induces its mechanism of action, or to the liver to be converted to the metabolites formed by CYP3A enzymes and later excreted by the kidneys. In the brain, MDZ bound allosterically to the GABAA receptor, which promotes the gamma-aminobutyric acid (GABA) binding to the receptors, which increases the influx of chloride ions (Cl−) into the postsynaptic cell. The Cl− influx increases the negativity of the cell potential, subsequently leading to hyperpolarization. The figure is created by Biorender.com.
While the pharmacodynamics of MDZ in preterm neonates have not been thoroughly investigated, it is important to note that there is currently no established correlation between MDZ concentration and clinical effects in term neonates or children [32, 33]. Nevertheless, in the NICU, MDZ is often used in combination with opioids (fentanyl, morphine, and diamorphine) for ventilated neonates [2, 34]. Notably, endotracheal intubation and mechanical ventilation are major components of routine intensive care for very low birthweight and critically ill full-term infants [2, 34]. MDZ can provide anxiolytics, sedation, muscle relaxants, anterograde amnesia, and anticonvulsant effects by binding on γ2 subunit of the gamma-aminobutyric acid alpha (GABAA) receptors. Once bound, MDZ modulates the activation of the GABAA receptors, which increases the frequency of opening of chloride channels that subsequently leads to the influx of Cl− into the cell membrane, which results in membrane hyperpolarization and neuronal inhibition [29–31].
Approximately 97 % of MDZ is bound to plasma protein, primarily albumin, in adults and pediatric patients. Metabolized in the liver through cytochrome P450-3A family (CYP3A) enzymes, particularly CYP3A4 and CYP3A5, MDZ undergoes hydroxylation as the main metabolic pathway. This results in the formation of the active metabolite alpha-1 hydroxy midazolam (about 60–70 % of biotransformation products), 4-hydroxy-midazolam (5 %), or dihydroxy derivative [35, 36]. Following hydroxylation, glucuronidation occurs, a process in which glucuronic acid is attached to the metabolite, enhancing its water solubility and facilitating elimination via urine.
Neonates can perceive pain when undergoing invasive procedures such as surgeries, mechanical ventilation, and intubation, which can then induce agitation in newborns [37]. In the clinical setting, the optimal dose of MDZ is varied due to clinical situations and factors such as the patient’s age, coexisting medical conditions, and concomitant medications. Pacifici wrote a review on the clinical pharmacology of MDZ in neonates and children, which also reviewed the dosages of MDZ in clinical settings [2]. The neonates and infant patients’ ages can vary between less than 28 weeks of gestation to 8 years of life [2]. Depending on health conditions, age, and procedure requirements, MDZ can be administered as bolus injections or followed by continuous infusion.
The adverse effects of MDZ or BDZs used on neonates are debatable since some studies did not note any adverse outcomes, while others did [2, 32, 38], [39], [40], [41], [42]. Life-threatening adverse effects of MDZ administration are related to respiratory depression, hypotension, cardiac arrest, decreased blood flow, and increased brain injuries such as IVH and PVL [43, 44]. Other major adverse effects associated with long-term MDZ administration are tolerance development, withdrawal symptoms, and delayed awakening [2, 32, 38], [39], [40], [41], [42]. In addition, The Neonatal Outcome and Prolonged Analgesia in Neonate (NOPAIN) trial has shown that MDZ is associated with poor short-term neurologic outcomes with about 33 % of MDZ-exposed neonates affected by phase III of IVH, PVL, and death, which is much higher when comparing to placebo or morphine cohort [43].
Numerous animal studies compellingly demonstrate that general anesthetics and sedative medications elicit diverse morpho-functional changes throughout brain development [45]. For MDZ exposure or related MDZ-anesthesia models, the rodent model is often the first choice for the preclinical model. Though lacking nonhuman primates (NHP) model data in early MDZ exposure model, rodent models of MDZ found similar apoptotic injury patterns as NHP exposed to other anesthetics agents such as ketamine and can offer several advantages given the cost and ethical concerns of studies using NHP. The MDZ preclinical models are also varied but can be categorized into 2 groups: anesthesia (single exposure in only 1 day) and sedation (multiple exposures or multiple days) models. For the preclinical model, rodents are often used. In the anesthesia model, MDZ is often co-administered with anesthetic agents such as isoflurane and nitrous oxide (N2O) for 6 h (Tables 1 and 2). Meanwhile, in the sedation model, MDZ is often treated alone and can be repeated from 3 to 18 days. Tables 1 and 2 present the age of the rodent animals, often starting on postnatal day 3 or day 7 (rats) and P10–P18 (mice). Most of the available rodents often start the treatment on P7, which is considered a peak in synaptogenesis in rodents. Very few in vitro models often use cortical primary neurons or embryonic hippocampi with dose ranges of 50–150 nM [46, 47] up to 30 mM [48].
Behavioral alterations post-midazolam exposure in young animals.
Species (postnatal day, P) | Treatment | Study significances | Ref. | |
---|---|---|---|---|
Components | Dosage and duration | |||
Sprague Dawley rats (P7) | Isoflurane + MDZ + N2O | 0.75 % + 9 mg/kg + 75 % for 6 h | Impaired spatial reference memories capabilities at adolescence and adulthood | [22] |
Sprague Dawley rats (P7) | Isoflurane + MDZ + N2O | 0.75 % + 9 mg/kg + 70 % for 6 h | Lasting influences on socio-emotional status in young adult rats (P63–87): – ↑ risk-taking tendencies and alterations in anxiety-related behaviors – ↑ novelty-seeking tendencies and less guarded-behavior |
[49] |
Sprague-Dawley rat (P7) | MDZ ± pre-treated minocycline (30 min before MDZ injection) | 9 mg/kg ± 40 mg/kg | Adult rats showed low spatial learning in MDZ group. Pre-treated with minocycline improved spatial learning and memory in adulthood. |
[50] |
Sprague-Dawley rat (P3, 10, 21, and 40) | MDZ | 0.1 mg/kg, 1.0 mg/kg, and 10 mg/kg | Neonates: ↓ reflex thresholds and ↑ reflex magnitudes Older rat pups and adults: ↑ reflex thresholds and ↓ reflex magnitude |
[51] |
C57BL/6J mice (P10 and P20) | MDZ | 50 mg/kg | P20 mice: acute ↓ locomotor activity at 3 and 24 h post-exposure but not after 72 h no long-term effects on learning and memory behaviors in adult (P105) | [52] |
C57BL/6 mice (P18–22) | MDZ | 10 mg/kg and 20 mg/kg, maintained for 12 h per day, repeated for 5 consecutive days | Impaired spatial and learning memory in adult (P60–63): – No differences in exploration time between original and novel Y-maze arm – ↓ freezing time percentage of contextual and cued fear conditioning |
[46] |
Sprague-Dawley rats (P7) | MDZ | 10, 25, or 40 mg/kg at 15 min prior to H/R | ↓ learning and memory impairment in H/R-induced model | [53] |
C57BL/6 mice (P7–9) | MDZ | 10 mg/kg, repeated single dose, 3 days | 12 w mice: – ↓ preference for an object placed at the NLR – ↓ contextual fear memory in FC Recovered cognitive ability post-voluntary exercise |
[47] |
-
Key: ↑, increased/elevated; ↓, decrease/reduced; FC, fear conditioning; H/R, hypoxia/reoxygenation; NLR, novel location recognition; P, postnatal day.
Midazolam’s effects on developing neurobiology in the in vivo and in vitro models.
Species (postnatal day, P) | Treatment | Brain regions | Study significances | Ref. | |
---|---|---|---|---|---|
Components | Dosage and duration | ||||
Sprague Dawley rats (P7) | MDZ | 3, 6, or 9 mg/kg | Whole brain | No increased in neuroapoptosis ↓ LTP in hippocampus slices at P29–35 |
[22] |
Isoflurane + MDZ | 0.75 % + 9 mg/kg for 6 h | LD, AV thalamic nuclei, parietal cortex (layer II) | ↑ neuroapoptosis | ||
Isoflurane + MDZ + N2O | 0.75 % + 9 mg/kg + 75 % for 6 h | Thalamus, parietal cortex, widespread effects in other regions | ↑ neuroapoptosis ↓ LTP in hippocampus slices at P29–35 |
||
Sprague Dawley rats (P1, 3, 7, 10 and 14) | MDZ | 3, 6, or 9 mg/kg | No increased apoptosis | [54] | |
Isoflurane + MDZ + N2O | 0.75 % + 9 mg/kg + 75 %; for 2, 4, or 6 h | Posterior cingulate/ retrosplenial cortex, the parietal and occipital cortex, subiculum, anterior thalamus, amygdala | ↑ neuroapoptosis (age-dependent) No disturbance in cortical cerebral blood flow, hypoxia, and hypoventilation Activated intrinsic apoptotic pathway: – alterations of bclxL – ↑ cytochrome C release and caspase-3, −8 and −9 activation |
||
Sprague Dawley rats (P7 and 17) | Isoflurane + MDZ + N2O | 0.75 % + 9 mg/kg + 75 % for 6 h | CC and anterior thalamus | ↑ neuroapoptosis but alleviated when treated with melatonin | [55] |
Sprague Dawley rats (P7) | Isoflurane + MDZ + N2O | 0.75 % + 9 mg/kg + 75 %; for 2, 4, or 6 h | CC and anterior thalamus | Disturbance in BDNF-activated apoptotic cascade (thalamus: ↓BDNF; CC: ↑BDNF) via Trk-dependent (thalamus) and Trk-independent p75NTR-dependent (CC) pathways ↓neuroapotosis via β-estradiol application |
[56] |
Sprague Dawley rats (P7) | Isoflurane + MDZ + N2O | 0.75 % + 9 mg/kg + 75 % for 6 h | CC and anterior thalamus | Altered synaptic proteins level after anesthesia administration Induced permanent neuronal loss after 3–23 days post-anesthesia |
[57] |
Sprague Dawley rats (P7) | Isoflurane + MDZ + N2O | 0.75 % + 9 mg/kg + 75 % for 6 h | Subiculum | Impaired neuropil structure, ↓ MSBs at 2 weeks post-exposure ↓ number of synapses per μm No preferential loss of different types of synaptic contacts or excitatory vs. inhibitory synapses |
[58] |
Sprague Dawley rats (P7) | Isoflurane + MDZ + N2O | 0.75 % + 9 mg/kg + 70 % for 6 h | Hippocampus | ↓ H3-acetylation in BDNF and c-Fos genes, manifested by CBP fragmentation ↓ BDNF and ↓c-Fos proteins: impaired neuronal development and synaptic neurotransmission |
[59] |
Sprague Dawley rats (P7) | Isoflurane + MDZ + N2O | 0.75 % + 9 mg/kg + 70 % for 6 h | Subiculum | Impairment of mitochondrial morphogenesis (↑ mitochondria area), structural integrity (↑ complex IV activity, ↓ regional distribution in presynaptic) ↑ autophagic activity and ↓ mitochondria density Disturbances in inhibitory synaptic neurotransmission |
[60] |
Sprague Dawley rats (P7) | Isoflurane + MDZ + N2O | 0.75 % + 9 mg/kg + 70 % for 6 h | Subiculum | 30 % rise ROS and a 2-fold decline in superoxide dismutase. Disrupts mitochondrial dynamics by promoting excessive fission via dynamin-related protein 1 sequestration and mitofusin-2 downregulation. |
[61] |
C57BL6 mice (P7) | MDZ ± ketamine | 9 mg/kg ± 40 mg/kg | CC, caudate putamen | ↑ neuroapoptosis, but more robust when combined with ketamine | [62] |
YFP-expressed mice (P15, 20, and 30) | MDZ | 25 mg/kg, repeated for 3 times per 90 min intervals | Somatosensory cortex, hippocampus | ↑ dendritic spine protrusion density and ↓ protrusions head width and lasts for several days | [48] |
P6-P7 hippocampus slices | MDZ | 30 μM, 5-hour-long bath | Hippocampus CA1 pyramidal neurons | ↑ dendritic spine protrusion and ↑ formation density rapidly after treatment, and lasted up to 48 h ↑ spinogenesis |
|
GFP-M transgenic mice (P8–12) | MDZ | 50 mg/kg, repeated single dose, 5 days | Hippocampus pyramidal neurons | ↓ spine density and ↑ spine length of hippocampal CA1 pyramidal neurons at P13 but not P30 | [63] |
E19 Wistar rats | MDZ | 100 and 300 nM on 15 DIV | Hippocampus | ↑ neuronal apoptosis ↑ cytosolic Ca2+-concentration Suppresses neuronal Ca2+-oscillations amplitude and frequency (dose dependent) ↓ synapses number after 24 h |
[64] |
Wistar rats (P5 and P15) | MDZ | Initially with 25 mg/kg then 12.5 mg/kg to achieve total of 6 h sedation; repeated up to 7 injection in a single day | mPFC | ↑ neuroapoptosis without impacting total neurons and neuronal density at both early (P5) and later (P15) stages of brain development Altered GABAergic neuronal phenotype, with stage-dependent effects on PV and CR neurons |
[65] |
Sprague-Dawley rat (P7) | MDZ ± pre-treated minocycline (30 min before MDZ injection) | 9 mg/kg ± 40 mg/kg | SVZ, SGZ of hippocampus, LV, DG | ↓ cell proliferation SVZ and SGZ after 7 days ↑ cell proliferation when pre-treated with minocycline |
[50] |
C57BL/6 mice (P18–22) | MDZ | 10 mg/kg and 20 mg/kg, maintained for 12 h per day, repeated for 5 consecutive days | Hippocampus, DG | ↓ long-term neurons proliferations ↓ presynaptic and ↑ post-synaptic density |
[46] |
C57BL/6 mice (P18–22) | MDZ ± morphine | MDZ: 5–9 mg/kg, MDZ + morphine: 2.5–4 mg/kg morphine and 5–9 mg/kg MDZ; escalating dose, twice daily for 5 days (from P18 to 22) | Whole brain | MDZ only: ↑ S100B expression and 17–18.5 kDa MBP splicing isoform expressions MDZ + morphine: − 20 % ↑ presynaptic marker synaptophysin − ↓ % weight gained − ↑ drebrin levels |
[66] |
C57BL/6 mice (P7–9) | MDZ | 10 mg/kg, repeated single dose, 3 days | Hippocampus, DG | ↓ NSC proliferation and neurogenesis in the DG at both P10 and P38 Inhibits NSC activation and neurogenesis in the adult DG but recover post voluntary exercise |
[47] |
Sprague-Dawley rats (P3–P21) | MDZ | 1 mg/kg to 10 mg/kg; repeated single dose, escalate from P3 to P21 | Cortex | Altered synaptic proteome GO analysis associated with actin binding, cytochrome c-oxidase, TCA cycle, etc. Validated ↑ ADD1 protein expression in MDZ synaptosome |
[67] |
Sprague-Dawley rats (P3–P21) | MDZ | 1 mg/kg to 10 mg/kg; repeated single dose, escalate from P3 to P21 | Cortex | Altered BDEV proteome GO analysis associated with sodium channel activity, adenylate cyclase, corticosteroid receptor signaling pathway, etc. Validated ↓ YWHAH post-MDZ exposure as BDEV marker |
[68] |
-
Key: ↑, increased/elevated; ↓, decrease/reduced; ADD1, alpha adducin protein; AV, anterodorsal; BDEV, brain-derived extracellular vesicle; BDNF, brain-derived neurotrophic factors; CBP, CREB-binding protein; CC, cerebral cortex; CR, calretinin; DG, dentate gyrus; DIV, days in vitro; EAAT2, Excitatory amino acid transporter 2; GO, gene ontology; H/R, hypoxia/reoxygenation; LD, lateral dorsal; LTP, long-term potentiation; MBP, myelin basic protein; mPFC, medial prefrontal cortex; MSB, multiple synaptic boutons; NSC, neural stem cell; P, postnatal day; PV, Parvalbumin; ROS, reactive oxygen species; SGZ, subgranular zone; SVZ, subventricular zone; S100B, calcium binding S 100 protein; YWHAH, 14-3-3 eta protein.
Midazolam exposure during childhood: effects on neurobehavioral outcomes
Young animals and children are particularly vulnerable to the effects of anesthetics and sedatives, a topic extensively explored in the literature [69]. While challenges persist in translating findings from animal studies to humans, concerns about alterations in brain systems and subsequent changes in behavioral states are significant, especially when indicators are not obvious. In this part, we will comprehensively evaluate animal and human data, delving into the nuanced landscape of behavioral changes resulting from exposure to MDZ.
Anesthetics and sedatives during early development have been linked to behavioral changes, as observed in rodent models [69]. In 2009, Wilder et al. conducted a pioneering large-scale retrospective study on anesthetic exposure in individuals under 4 years of age, examining the incidence of later behavioral, learning, or developmental issues during school age [25]. The findings revealed that subjects with prolonged or repeated exposure to anesthesia exhibited an approximately two-fold increase in the identification rate of such problems compared to those without exposure. Since then, various retrospective studies have been published, yielding mixed results concerning the association between anesthetic exposure and subsequent neurobehavioral and developmental problems [70, 71]. Yet, the precise age at which the human brain undergoes its most vulnerable phase to the neurotoxic impacts of anesthetic and sedative drugs remains unclear and contentious. This uncertainty arises as there is a limited endorsement for postponing essential procedures in young children to minimize neurodevelopmental risks. In addition, not only direct exposure to anesthesia but also exposure to anesthesia via maternal also affects behavior in fetal and newborn rats. For instance, multiple works have shown that in utero or prenatal exposure to opioids such as morphine or oxycodone could induce heightened social anxiety and declined cognitive scores [72, 73].
Many available clinical studies consistently indicate an intensified risk of learning and behavioral disabilities in individuals with early exposure to general anesthesia and sedative agents [69, 71, 74–77]. While ample evidence explores the impact of opioids, such as fentanyl and morphine, on learning abilities and cognitive functions, limited research focuses on the cognitive and behavioral outcomes of children exposed to MDZ despite it being a commonly chosen sedative for neonatal and childhood care.
Duerden et al. conducted studies on very preterm neonates, assessing the long-term effects of MDZ exposure. Magnetic resonance and diffusion tensor imaging scans revealed decreased hippocampal volumes and lower cognitive scores at 18 months and 8 years of corrected age [78, 79]. McGraw reported increased postoperative behavior changes in children given preoperative oral MDZ, indicating potential long-term effects beyond the immediate postoperative period [80]. Research by Puia-Dumitrescu et al. on extremely preterm infants exposed to opioids and/or benzodiazepines for more than 7 days highlighted lower cognitive, motor, and language scores at 2 years corrected age. This study emphasizes the potential cumulative impact of prolonged exposure to these sedatives on developmental trajectories, warranting further research to understand specific mechanisms and long-term consequences [81]. Furthermore, the implications of MDZ in pediatric cardiac surgery are noteworthy, with evidence indicating that the total MDZ dose may adversely affect neurodevelopmental outcomes in infants [82]. Additionally, the combined use of MDZ and phenobarbital, both GABAergic drugs, has been linked to increased neuronal injury in neonatal seizures [83]. These findings are particularly concerning given the potential long-term impact on neurodevelopment, especially in vulnerable populations such as preterm infants.
While clinical data on neonatal exposure to MDZ is limited, preclinical investigations suggest potentially harmful neurodevelopmental outcomes. The ‘midazolam effect,’ observed in animal studies, reveals a dose-dependent impact on reflex thresholds and behavioral changes. Seminal research by Jevtovic-Tedorovic et al. demonstrated that postnatal day 7 rats, continuously exposed for 6 h to a general anesthesia ‘triple cocktail’ including isoflurane, nitrous oxide (N2O), and MDZ, exhibited lasting cognitive, behavioral, and memory deficiencies [22]. Another study found that MDZ administration in neonatal rats produced a dose-dependent reduction in mechanical reflex thresholds and a concurrent increase in both mechanical and thermal reflex magnitudes [51]. Conversely, in older rat pups and adults, MDZ produced the opposite outcome, elevating reflex thresholds and diminishing reflex magnitude [51]. Beyond traditional measures of intelligence and cognitive outcomes, researchers are expanding their focus to include behavioral testing and examining changes in anxiety and social interactions. For instance, one study by Diana et al. demonstrated that early exposure to a triple anesthetic cocktail similar to Jevtovic-Tedorovic’s study led to altered behavior in the open field and elevated plus maze tests, suggesting increased risk-taking and anxiety [49]. Additionally, a study by Xu et al. found that repeated, long-duration exposure to MDZ can result in deficits in performance on the Ymaze and fear conditioning testing in young adult mice [46]. A summary of the effects of early MDZ exposure on behavior is provided in Table 1.
Both clinical and preclinical studies presented herein reveal that exposure to MDZ, particularly in vulnerable populations such as preterm infants and young children, can lead to lasting cognitive and behavioral deficits. Despite mixed findings and ongoing debates about the critical periods of vulnerability, the evidence indicates the necessity for cautious use of MDZ in specific patient groups. Further investigation is warranted to elucidate the mechanisms underlying these effects and develop strategies to mitigate potential long-term harm.
Effects of MDZ on neurobiology
Effects on neurons
Neurons are fundamental functional units of the nervous system responsible for controlling signal transmission and informational processing throughout the body. During the critical period of rapid synaptogenesis, the developing brain is particularly vulnerable to anesthetic agents [22, 54, 57, 69, 84, 85]. In rodents, this susceptibility peaks around postnatal day 7, while in humans, rapid synaptogenesis extends from mid-fetal gestation to several years after birth [14].
Preclinical studies have demonstrated that MDZ is associated with disruption in synaptogenesis and alterations in neuronal structure and function. Specifically, rodents exposed to MDZ followed by isoflurane and N2O have been associated with increased neuroapoptosis [22, 54–57], activation of intrinsic apoptotic pathway [56], long-term hippocampal potentiation suppression [54], decreased synapse density [58], and impaired synaptic mitochondria structure and dynamic [60, 61]. A single exposure to only MDZ is debatable since some studies observed no neuroapoptosis [22, 62] while others presented increased neuronal cell death [62, 64] and were even robust when co-treated with ketamine [62]. Moreover, in vitro and in vivo studies also showed that exposure to MDZ can reduce neuronal proliferation [46, 50]. Repeated and prolonged exposure to MDZ during the neonatal period also led to enduring alterations in neuronal structure and function and can persist long-term. Studies employing prolonged MDZ sedation revealed significant dose-dependent changes in synaptic morphology (i.e., alterations in dendritic spine protrusion and density) and number of connections [48, 63, 64], reduced hippocampal neurogenesis, cell proliferation, and impaired learning and memory in rodents [46, 47, 50]. In addition, repeated exposure to MDZ at both early and later stages of development can also alter medial prefrontal cortex (mPFC) GABAergic neuronal phenotypes with a direct impact on parvalbumin and calretinin interneurons [65]. Giri and colleagues also showed that a single MDZ exposure could inhibit cell proliferation in the neurogenic regions of neonatal rats, suggesting potential impairment of neurogenesis during neonatal MDZ exposure. Interestingly, in the same study, the adverse effects on neurogenesis were reversed when neonate rats were treated with minocycline, a long-acting tetracycline agent with neuroprotective and anti-inflammatory properties, before MDZ injection [50]. Similarly, Doi et al. found that repetitive neonatal MDZ exposure negatively affects neurogenesis and cognitive ability, yet these effects were mitigated by voluntary exercise in adulthood [47]. Whether combining MDZ with other known neuroprotective agents or implementing exercise at a later developmental stage could mitigate the harmful effects of neonatal MDZ exposure remains to be further investigated.
Effects on glial cells
While considerable attention is given to neuronal impacts, it is essential not to disregard the influence of MDZ on glial function. Glial cells (i.e., astrocytes, microglia, and oligodendrocytes) play a pivotal role in the development and maturation of neurons. The emerging field of research underscores the contributions of glial to neural function and behaviors, including learning and memory [86, 87]. However, limited information is available on how early exposure to MDZ affects glial cells. Iqbal O’meara et al. demonstrated that repeated MDZ exposure in young mice significantly modulates the expression of specific myelin basic protein isoforms and S100B calcium-binding protein in the developing brain [66]. Another study by Tanabe et al., while not specifically focused on neonates or young animals, suggested that MDZ is associated with reducing cerebral endothelial ICAM-1 expression and suppressing IL-1β-induced IL-6 release from rat glial cells [88]. Alterations in acetylcholinesterase and butyrylcholinesterase activities further contribute to the intricate relationship between MDZ and glial dynamics.
Despite the uncertainties about MDZ’s influence on developing glial cells, studies on other GABAA agonist sedatives and anesthetics with similar mechanisms of action (e.g., propofol, sevoflurane, desoflurane) indicate potential toxic effects on glial cells, particularly astrocytes, oligodendrocytes, and microglia. In vitro and in vivo studies on isoflurane revealed the toxic effects of isoflurane on impairments of cytoskeletal structure and organization during astrocyte development and maturation [89–92]. Additionally, a 3-h exposure to propofol in rat dentate gyrus nascent cells significantly increased astrocyte expression 24 h post-exposure [92]. Furthermore, 2 h of exposure to 3 % sevoflurane in P6 mice repeatedly at multiple time points was associated with microglia activation in the hippocampus [93].
Current evidence also suggests that the impairment in brain and cognitive function may be due to the negative impacts that anesthesia exposure had on glial cell architecture and modulated neuroimmune responses [94–98]. Anesthetics and sedative agents can exert both anti- and pro-inflammatory effects by modulating microglial activation, suggesting that anesthetics may play dual roles in cognitive impairments. Several studies have highlighted the effects of BDZs on modulating neuroimmune responses in adult in vivo or in vitro models [99–103]. A recent study by Shi et al. suggested that chronic exposure to diazepam (DZP), a common sedative BDZ, induces cognitive deficits in adult mice via the mitochondrial 18 kDa translocator protein (TSPO) by altering microglial morphology and impairing synaptic material phagocytosis. These findings highlight a mechanism by which TSPO ligands modulate synaptic plasticity and consequentially lead to cognitive impairment [100]. TSPO, previously known as the peripheral BDZ receptor, is a transmembrane protein expressed ubiquitously in peripheral tissues and binds to BDZs. Although TSPO has been implicated in multiple biological functions, including steroid biosynthesis, apoptosis, and cell proliferation, its exact mechanism in regulating immune response remains ambiguous [104]. Some studies suggest that TSPO signaling can reduce pro-inflammatory cytokine production in microglia [99, 101, 105]. For instance, Horiguchi and colleagues demonstrated that MDZ exerts anti-inflammatory effects on LPS-stimulated macrophages by suppressing NF-κB/AP-1 and MAPK activation through TSPO mediation. This suppression reduces the activation of pro-inflammatory genes, including those encoding CD80, IL-6, and TNF-α [99]. The authors suggested that such interference with macrophage activation may be linked to the adverse side effects seen in some critically ill patients following MDZ administration [106, 107]. Wilms et al. also suggested that these BZDs may hinder microglial proliferation and mitigate inflammatory responses associated with activated microglia [101].
Molecular correlates
The brain is a complex network of interconnected circuits responsible for specific functions. Neurons communicate by releasing neurotransmitters into the synapse – the gap between neurons. These neurotransmitters bind to receptors on the receiving neuron, influencing its activity. Transporters recycle neurotransmitters, regulating the signal between neurons. MDZ, like other BDZs, directly binds to the GABAA receptor, enhancing GABA binding and limiting excitatory signals. This action can induce dopamine release by disinhibiting interneurons and promoting the activity of dopaminergic neurons [108]. The complex impact of MDZ on neurotransmitters also involves the disruptions in cholinesterase homeostasis, suppression of neuronal Ca2+ oscillations, and inhibition of hippocampal long-term potentiation and learning through BDZ receptors [64, 109, 110].
Additionally, the microenvironment of the synapse plays a crucial role in controlling the synaptic activities. Nguyen et al. demonstrated alterations in the proteome of synaptosomes, isolated subcellular fractions of synaptic terminals, in early neonatal rats exposed to prolonged MDZ. Their findings revealed up- and downregulated differentially expressed proteins (DEPs) involved in various molecular functions and biological processes, such as actin-binding, cytochrome c oxidase, pyridoxal phosphate binding, protein depolymerization, tricarboxylic cycle, and central nervous system neuron development [67].
In addition to exploring neurochemistry and the proteome, current research highlights potential long-term changes in gene transcription and functional deficits in learning and behavior induced by anesthesia. These effects may be mediated through epigenetic modifications. Notably, the literature emphasizes the significance of histone modifications in neural development and brain function [111]. Preclinical studies underscore the involvement of cyclic-AMP-response element binding protein (CREB) signaling pathways in anesthesia-induced neurodegenerative changes [111–113]. CREB-binding protein (CBP), also known as CBP, is a transcription activator and co-activator of CREB. Additionally, CBP functions as a histone acetyltransferase (HAT), regulating histone acetylation through its intrinsic HAT domain. Exposure to a sedative dose of MDZ, followed by a combination of nitrous oxide and isoflurane for 6 h in P7 rat pups, leads to the fragmentation of CBP and a subsequent decrease in its HAT activity at 2 h or 24 h (at P8) post-exposure. This subsequently results in hypoacetylation of H3, causing down-regulation in the transcription and expression of BDNF and c-Fos [59]. The long-term epigenetic changes in rodents exposed to MDZ during their neonatal period have yet to be explored.
The evidence discussed herein suggests that MDZ and other BDZs may affect neurodevelopment through various mechanisms. These effects are not limited to neuronal damage, alterations in synaptic function, modulation of glial cell activity, disruption of immune responses, changes in neurotransmitter dynamics, and epigenetic modifications. Therefore, it is imperative to further investigate the mechanisms by which MDZ influences neurodevelopmental biology and to assess the long-term consequences of both acute and chronic early exposure. Although few studies have explored therapeutic interventions to mitigate the cognitive impairments or adverse effects of neonatal MDZ exposure, this area of research holds significant promise for clinical application. This knowledge is essential for developing safer anesthetic and sedation practices, particularly for vulnerable populations such as preterm infants and young children, to mitigate potential adverse outcomes and ensure proper neurodevelopment. Detailed impacts of MDZ exposure during early development on neurobiology are provided in Table 2, and a summary of risks associated with MDZ exposure is described in Figure 2.

Overview of risks associated with exposure to midazolam during early development. Graphic summary of significant molecular, cellular, and behavioral deficits in neonatal exposure to MDZ. This figure is created by Biorender.com.
Future perspectives
The use of anesthesia and sedatives to prevent pain and manage agitation and stress has been a major advance in medicine and neonatal intensive care [37]. Despite the potential for limiting painful procedures, critically ill infants often undergo unavoidable stress-inducing interventions, such as intubation and catheterization. The utilization of analgesics (opioids and nonsteroidal anti-inflammatory drugs) and sedatives (benzodiazepines and other anesthetic agents) is widespread, but its application varies significantly across different hospital units. While the mechanism of action of many anesthetics, including MDZ, is still not completely understood, the potential for these agents to induce longer-term alterations in brain function, particularly in vulnerable populations such as premature neonates, warrants future investigation. Despite ongoing research on MDZ’s potential longer-term effects, establishing a definitive link between general anesthesia/surgery/sedation and lasting behavioral or cognitive alterations in children remains debatable.
A significant gap in our understanding pertains to the cumulative impact of life stresses on the long-term effects of MDZ. Concerns persist, especially regarding the safety of prolonged MDZ use throughout developmental stages like early childhood, adolescence, and adulthood. Unfortunately, preclinical data is lacking for these specific time points. In addition, clinical studies emphasize that a considerable number of children undergo multiple and prolonged exposures to anesthetics and sedatives, resulting in reported deficits larger than those observed with a single exposure [25, 26, 28, 81]. The current preclinical model predominantly examines a single exposure, with very few models exploring the impact of multiple MDZ exposures, leaving uncertainties about the consequences of multiple exposures on molecular, cellular, and behavioral aspects.
The process of synaptogenesis is profoundly affected by MDZ alone or in combination with other anesthetic agents (Table 2). However, synapses are also regulated by intricate networks of proteins and signaling molecules that modulate the synaptogenesis process. Therefore, identifying biomarkers indicating abnormalities associated with MDZ exposure can offer insights into potential injury and toxicity-induced mechanisms. Biomarkers can aid in examining proposed preclinical mechanisms and facilitate translation between human and animal models. Additionally, since synapses house numerous mitochondria, it is crucial to acknowledge the potential impact on synaptic mitochondrial functions and the expression of mitochondria-associated proteins. Notably, growth factors acting as signaling molecules can directly or indirectly influence various biological processes, including synaptogenesis and synaptic pruning. However, there is limited data on the impact of chronic MDZ exposure during childhood on growth factor profiles.
Despite recent advancements in understanding the potential impact of MDZ on synapses, there remains a need for a more comprehensive exploration of the long-term effects of MDZ exposure on synaptic functionality. Some studies indicate that MDZ may decrease the long-term potentiation of synapses. Additionally, synaptic activities, such as neurotransmitter release and uptake, vesicle docking and recycling, and dendritic spine alterations, require substantial ATP, primarily regulated by mitochondria at pre- and post-synaptic sites. The scarcity of information on the short and long-term effects of MDZ on mitochondria and its influence on synaptic activities emphasizes the importance of understanding these aspects. Such knowledge can contribute to elucidating the mechanisms of neuroapoptosis and cellular dysfunction, thereby aiding the development of appropriate interventions and therapeutic strategies to address adverse effects associated with MDZ exposure in neonates and infants.
Conclusions
Midazolam, a widely used benzodiazepine sedative in the NICU, serves to alleviate agitation and induce sedation in neonates. Despite its effectiveness, its administration often accompanies invasive procedures such as surgeries and mechanical ventilation, contributing to various adverse outcomes encompassing respiratory, cardiovascular, and neurological domains. Recent studies have scrutinized the impact of single MDZ exposures on long-term outcomes, revealing adverse effects on cognitive function and behavioral deficits. This scrutiny extends to the molecular and cellular levels, where MDZ is found to induce apoptosis and alter cell morphology and functions. Although progress has been made in understanding baseline deficits, notable knowledge gaps persist. Particularly, there is a paucity of information on the consequences of prolonged/chronic MDZ exposures and their potential long-term effects on neurological and developmental outcomes. Addressing these gaps is crucial for advancing our understanding of MDZ’s comprehensive impact in clinical settings.
Funding source: Startup funds from the Department of Anesthesiology, UNMC
Funding source: Lieberman Endowment
Funding source: Pilot funding from the Child Health Research Institute, UNMC, Omaha, NE
-
Research ethics: The local Institutional Review Board deemed the study exempt from review.
-
Informed consent: Not applicable.
-
Author contributions: NMN: investigation, writing – original draft; writing – review and editing. GP: funding acquisition; project administration; supervision; writing – review and editing.
-
Competing interests: The authors declare no conflict of interest. The funders had no role in the design of the study, in the collection, analyses, or interpretation of data, in the writing of the manuscript, or in the decision to publish the results.
-
Research funding: This work was supported by Lieberman Endowment, startup funds from the Department of Anesthesiology, and pilot funding from the Child Health Research Institute, UNMC to G.P. The funding entity played no role in the study design or its conduction.
-
Data availability: All data accessed is included in this review article.
References
1. Stark, A, Smith, PB, Hornik, CP, Zimmerman, KO, Hornik, CD, Pradeep, S, et al.. Medication use in the neonatal intensive care unit and changes from 2010 to 2018. J Pediatr 2022;240:66–71.e4. https://doi.org/10.1016/j.jpeds.2021.08.075.Search in Google Scholar PubMed PubMed Central
2. Pacifici, GM. Clinical pharmacology of midazolam in neonates and children: effect of disease – a review. Int J Pediatr 2014;2014:309342. https://doi.org/10.1155/2014/309342.Search in Google Scholar PubMed PubMed Central
3. Song, IG. Neurodevelopmental outcomes of preterm infants. Clin Exp Pediatr 2023;66:281–7. https://doi.org/10.3345/cep.2022.00822.Search in Google Scholar PubMed PubMed Central
4. CDC. Preterm birth: division of reproductive health; 2023. Available from: https://www.cdc.gov/reproductivehealth/maternalinfanthealth/pretermbirth.htm.Search in Google Scholar
5. Barfield, WD. Public health implications of very preterm birth. Clin Perinatol 2018;45:565–77. https://doi.org/10.1016/j.clp.2018.05.007.Search in Google Scholar PubMed PubMed Central
6. Lee, HC, Bennett, MV, Schulman, J, Gould, JB, Profit, J. Estimating length of stay by patient type in the neonatal intensive care unit. Am J Perinatol 2016;33:751–7. https://doi.org/10.1055/s-0036-1572433.Search in Google Scholar PubMed
7. Yang, X, Meng, T. Admission of full-term infants to the neonatal intensive care unit: a 9.5-year review in a tertiary teaching hospital. J Matern Fetal Neonatal Med 2020;33:3003–9. https://doi.org/10.1080/14767058.2019.1566901.Search in Google Scholar PubMed
8. Bakri, MH, Ismail, EA, Ali, MS, Elsedfy, GO, Sayed, TA, Ibrahim, A. Behavioral and emotional effects of repeated general anesthesia in young children. Saudi J Anaesth 2015;9:161–6. https://doi.org/10.4103/1658-354x.152843.Search in Google Scholar PubMed PubMed Central
9. Durrmeyer, X, Vutskits, L, Anand, KJS, Rimensberger, PC. Use of analgesic and sedative drugs in the NICU: integrating clinical trials and laboratory data. Pediatr Res 2010;67:117–27. https://doi.org/10.1203/pdr.0b013e3181c8eef3.Search in Google Scholar PubMed
10. Ng, E, Taddio, A, Ohlsson, A. Intravenous midazolam infusion for sedation of infants in the neonatal intensive care unit. Cochrane Database Syst Rev 2012:Cd002052. https://doi.org/10.1002/14651858.CD002052.Search in Google Scholar PubMed
11. Wolke, D. The SAGE handbook of developmental disorders. London: SAGE Publications Ltd; 2011. https://sk.sagepub.com/reference/hdbk_developmentaldisorders [Accessed 2 Feb 2024].Search in Google Scholar
12. Gholipour, A, Rollins, CK, Velasco-Annis, C, Ouaalam, A, Akhondi-Asl, A, Afacan, O, et al.. A normative spatiotemporal MRI atlas of the fetal brain for automatic segmentation and analysis of early brain growth. Sci Rep 2017;7:476. https://doi.org/10.1038/s41598-017-00525-w.Search in Google Scholar PubMed PubMed Central
13. Belfort, MB, Inder, TE. Human milk and preterm infant brain development: a narrative review. Clin Therapeut 2022;44:612–21. https://doi.org/10.1016/j.clinthera.2022.02.011.Search in Google Scholar PubMed PubMed Central
14. Semple, BD, Blomgren, K, Gimlin, K, Ferriero, DM, Noble-Haeusslein, LJ. Brain development in rodents and humans: identifying benchmarks of maturation and vulnerability to injury across species. Prog Neurobiol 2013;106–107:1–16. https://doi.org/10.1016/j.pneurobio.2013.04.001.Search in Google Scholar PubMed PubMed Central
15. Tymofiyeva, O, Hess, CP, Xu, D, Barkovich, AJ. Structural MRI connectome in development: challenges of the changing brain. Br J Radiol 2014;87:20140086. https://doi.org/10.1259/bjr.20140086.Search in Google Scholar PubMed PubMed Central
16. Lenroot, RK, Giedd, JN. Brain development in children and adolescents: insights from anatomical magnetic resonance imaging. Neurosci Biobehav Rev 2006;30:718–29. https://doi.org/10.1016/j.neubiorev.2006.06.001.Search in Google Scholar PubMed
17. Herschkowitz, N, Kagan, J, Zilles, K. Neurobiological bases of behavioral development in the first year. Neuropediatrics 1997;28:296–306. https://doi.org/10.1055/s-2007-973720.Search in Google Scholar PubMed
18. Huttenlocher, PR. Synaptic density in human frontal cortex - developmental changes and effects of aging. Brain Res 1979;163:195–205. https://doi.org/10.1016/0006-8993(79)90349-4.Search in Google Scholar PubMed
19. Volpe, JJ. Brain injury in premature infants: a complex amalgam of destructive and developmental disturbances. Lancet Neurol 2009;8:110–24. https://doi.org/10.1016/s1474-4422(08)70294-1.Search in Google Scholar PubMed PubMed Central
20. Whitelaw, A. Intraventricular haemorrhage and posthaemorrhagic hydrocephalus: pathogenesis, prevention and future interventions. Semin Neonatol 2001;6:135–46. https://doi.org/10.1053/siny.2001.0047.Search in Google Scholar PubMed
21. Culley, DJ, Xie, Z, Crosby, G. General anesthetic-induced neurotoxicity: an emerging problem for the young and old? Curr Opin Anaesthesiol 2007;20:408–13. https://doi.org/10.1097/aco.0b013e3282efd18b.Search in Google Scholar
22. Jevtovic-Todorovic, V, Hartman, RE, Izumi, Y, Benshoff, ND, Dikranian, K, Zorumski, CF, et al.. Early exposure to common anesthetic agents causes widespread neurodegeneration in the developing rat brain and persistent learning deficits. J Neurosci 2003;23:876–82. https://doi.org/10.1523/jneurosci.23-03-00876.2003.Search in Google Scholar PubMed PubMed Central
23. Weiss, M, Bissonnette, B, Engelhardt, T, Soriano, S. Anesthetists rather than anesthetics are the threat to baby brains. Paediatr Anaesth 2013;23:881–2. https://doi.org/10.1111/pan.12200.Search in Google Scholar PubMed
24. Istaphanous, GK, Loepke, AW. General anesthetics and the developing brain. Curr Opin Anaesthesiol 2009;22:368–73. https://doi.org/10.1097/aco.0b013e3283294c9e.Search in Google Scholar PubMed
25. Wilder, RT, Flick, RP, Sprung, J, Katusic, SK, Barbaresi, WJ, Mickelson, C, et al.. Early exposure to anesthesia and learning disabilities in a population-based birth cohort. Anesthesiology 2009;110:796–804. https://doi.org/10.1097/01.anes.0000344728.34332.5d.Search in Google Scholar PubMed PubMed Central
26. Sprung, J, Flick, RP, Katusic, SK, Colligan, RC, Barbaresi, WJ, Bojanić, K, et al.. Attention-deficit/hyperactivity disorder after early exposure to procedures requiring general anesthesia. Mayo Clin Proc 2012;87:120–9. https://doi.org/10.1016/j.mayocp.2011.11.008.Search in Google Scholar PubMed PubMed Central
27. Hu, D, Flick, RP, Zaccariello, MJ, Colligan, RC, Katusic, SK, Schroeder, DR, et al.. Association between exposure of young children to procedures requiring general anesthesia and learning and behavioral outcomes in a population-based birth cohort. Anesthesiology 2017;127:227–40. https://doi.org/10.1097/aln.0000000000001735.Search in Google Scholar
28. Warner, DO, Zaccariello, MJ, Katusic, SK, Schroeder, DR, Hanson, AC, Schulte, PJ, et al.. Neuropsychological and behavioral outcomes after exposure of young children to procedures requiring general anesthesia: the mayo anesthesia safety in kids (MASK) study. Anesthesiology 2018;129:89–105. https://doi.org/10.1097/aln.0000000000002232.Search in Google Scholar
29. Dundee, JW, Halliday, NJ, Harper, KW, Brogden, RN. Midazolam. A review of its pharmacological properties and therapeutic use. Drugs 1984;28:519–43. https://doi.org/10.2165/00003495-198428060-00002.Search in Google Scholar PubMed
30. Kanto, JH. Midazolam: the first water-soluble benzodiazepine. Pharmacology, pharmacokinetics and efficacy in insomnia and anesthesia. Pharmacotherapy 1985;5:138–55. https://doi.org/10.1002/j.1875-9114.1985.tb03411.x.Search in Google Scholar PubMed
31. Wang, J, Sun, P, Liang, P. Neuropsychopharmacological effects of midazolam on the human brain. Brain Inf 2020;7:15. https://doi.org/10.1186/s40708-020-00116-y.Search in Google Scholar PubMed PubMed Central
32. Jacqz-Aigrain, E, Wood, C, Robieux, I. Pharmacokinetics of midazolam in critically ill neonates. Eur J Clin Pharmacol 1990;39:191–2. https://doi.org/10.1007/bf00280059.Search in Google Scholar
33. de Wildt, SN, de Hoog, M, Vinks, AA, Joosten, KF, van Dijk, M, van den Anker, JN. Pharmacodynamics of midazolam in pediatric intensive care patients. Ther Drug Monit 2005;27:98–102. https://doi.org/10.1097/00007691-200502000-00018.Search in Google Scholar PubMed
34. Aranda, JV, Carlo, W, Hummel, P, Thomas, R, Lehr, VT, Anand, KJ. Analgesia and sedation during mechanical ventilation in neonates. Clin Therapeut 2005;27:877–99. https://doi.org/10.1016/j.clinthera.2005.06.019.Search in Google Scholar PubMed
35. Swart, EL, Slort, PR, Plötz, FB. Growing up with midazolam in the neonatal and pediatric intensive care. Curr Drug Metabol 2012;13:760–6. https://doi.org/10.2174/138920012800840347.Search in Google Scholar PubMed
36. PubChem Compound Summary for CID 4192. Midazolam. National Center for Biotechnology Information; 2024. Available from: https://pubchem.ncbi.nlm.nih.gov/compound/Midazolam.Search in Google Scholar
37. McPherson, C, Miller, SP, El-Dib, M, Massaro, AN, Inder, TE. The influence of pain, agitation, and their management on the immature brain. Pediatr Res 2020;88:168–75. https://doi.org/10.1038/s41390-019-0744-6.Search in Google Scholar PubMed PubMed Central
38. Hughes, J, Gill, A, Leach, HJ, Nunn, AJ, Billingham, I, Ratcliffe, J, et al.. A prospective study of the adverse effects of midazolam on withdrawal in critically ill children. Acta Paediatr 1994;83:1194–9. https://doi.org/10.1111/j.1651-2227.1994.tb18280.x.Search in Google Scholar PubMed
39. Jacqz-Aigrain, E, Daoud, P, Burtin, P, Maherzi, S, Beaufils, F. Pharmacokinetics of midazolam during continuous infusion in critically ill neonates. Eur J Clin Pharmacol 1992;42:329–32. https://doi.org/10.1007/bf00266357.Search in Google Scholar
40. Treluyer, JM, Zohar, S, Rey, E, Hubert, P, Iserin, F, Jugie, M, et al.. Minimum effective dose of midazolam for sedation of mechanically ventilated neonates. J Clin Pharm Therapeut 2005;30:479–85. https://doi.org/10.1111/j.1365-2710.2005.00678.x.Search in Google Scholar PubMed
41. Jacqz-Aigrain, E, Daoud, P, Burtin, P, Desplanques, L, Beaufils, F. Placebo-controlled trial of midazolam sedation in mechanically ventilated newborn babies. Lancet 1994;344:646–50. https://doi.org/10.1016/s0140-6736(94)92085-0.Search in Google Scholar PubMed
42. Arya, V, Ramji, S. Midazolam sedation in mechanically ventilated newborns: a double blind randomized placebo controlled trial. Indian Pediatr 2001;38:967–74.Search in Google Scholar
43. Anand, KJ, Barton, BA, McIntosh, N, Lagercrantz, H, Pelausa, E, Young, TE, et al.. Analgesia and sedation in preterm neonates who require ventilatory support: results from the NOPAIN trial. Neonatal outcome and prolonged analgesia in neonates. Arch Pediatr Adolesc Med 1999;153:331–8. https://doi.org/10.1001/archpedi.153.4.331.Search in Google Scholar PubMed
44. Harte, GJ, Gray, PH, Lee, TC, Steer, PA, Charles, BG. Haemodynamic responses and population pharmacokinetics of midazolam following administration to ventilated, preterm neonates. J Paediatr Child Health 1997;33:335–8. https://doi.org/10.1111/j.1440-1754.1997.tb01611.x.Search in Google Scholar PubMed
45. Vutskits, L, Xie, Z. Lasting impact of general anaesthesia on the brain: mechanisms and relevance. Nat Rev Neurosci 2016;17:705–17. https://doi.org/10.1038/nrn.2016.128.Search in Google Scholar PubMed
46. Xu, J, Mathena, RP, Singh, S, Kim, J, Long, JJ, Li, Q, et al.. Early developmental exposure to repetitive long duration of midazolam sedation causes behavioral and synaptic alterations in a rodent model of neurodevelopment. J Neurosurg Anesthesiol 2019;31:151–62. https://doi.org/10.1097/ana.0000000000000541.Search in Google Scholar PubMed PubMed Central
47. Doi, H, Matsuda, T, Sakai, A, Matsubara, S, Hoka, S, Yamaura, K, et al.. Early-life midazolam exposure persistently changes chromatin accessibility to impair adult hippocampal neurogenesis and cognition. Proc Natl Acad Sci USA 2021;118. https://doi.org/10.1073/pnas.2107596118.Search in Google Scholar PubMed PubMed Central
48. De Roo, M, Klauser, P, Briner, A, Nikonenko, I, Mendez, P, Dayer, A, et al.. Anesthetics rapidly promote synaptogenesis during a critical period of brain development. PLoS One 2009;4:e7043. https://doi.org/10.1371/journal.pone.0007043.Search in Google Scholar PubMed PubMed Central
49. Diana, P, Joksimovic, SM, Faisant, A, Jevtovic-Todorovic, V. Early exposure to general anesthesia impairs social and emotional development in rats. Mol Neurobiol 2020;57:41–50. https://doi.org/10.1007/s12035-019-01755-x.Search in Google Scholar PubMed PubMed Central
50. Giri, PK, Lu, Y, Lei, S, Li, W, Zheng, J, Lu, H, et al.. Pretreatment with minocycline improves neurogenesis and behavior performance after midazolam exposure in neonatal rats. Neuroreport 2018;29:153–9. https://doi.org/10.1097/wnr.0000000000000937.Search in Google Scholar
51. Koch, SC, Fitzgerald, M, Hathway, GJ. Midazolam potentiates nociceptive behavior, sensitizes cutaneous reflexes, and is devoid of sedative action in neonatal rats. Anesthesiology 2008;108:122–9. https://doi.org/10.1097/01.anes.0000296079.45446.15.Search in Google Scholar PubMed
52. Xu, H, Liu, ZQ, Liu, Y, Zhang, WS, Xu, B, Xiong, YC, et al.. Administration of midazolam in infancy does not affect learning and memory of adult mice. Clin Exp Pharmacol Physiol 2009;36:1144–8. https://doi.org/10.1111/j.1440-1681.2009.05208.x.Search in Google Scholar PubMed
53. Tang, Z, Yang, F, Dong, Y, Ma, C, Sun, S, Shan, Y, et al.. Midazolam contributes to neuroprotection against hypoxia/reoxygenation-induced brain injury in neonatal rats via regulation of EAAT2. Brain Res Bull 2020;161:136–46. https://doi.org/10.1016/j.brainresbull.2020.04.016.Search in Google Scholar PubMed
54. Yon, JH, Daniel-Johnson, J, Carter, LB, Jevtovic-Todorovic, V. Anesthesia induces neuronal cell death in the developing rat brain via the intrinsic and extrinsic apoptotic pathways. Neuroscience 2005;135:815–27. https://doi.org/10.1016/j.neuroscience.2005.03.064.Search in Google Scholar PubMed
55. Yon, JH, Carter, LB, Reiter, RJ, Jevtovic-Todorovic, V. Melatonin reduces the severity of anesthesia-induced apoptotic neurodegeneration in the developing rat brain. Neurobiol Dis 2006;21:522–30. https://doi.org/10.1016/j.nbd.2005.08.011.Search in Google Scholar PubMed
56. Lu, LX, Yon, JH, Carter, LB, Jevtovic-Todorovic, V. General anesthesia activates BDNF-dependent neuroapoptosis in the developing rat brain. Apoptosis 2006;11:1603–15. https://doi.org/10.1007/s10495-006-8762-3.Search in Google Scholar PubMed
57. Nikizad, H, Yon, JH, Carter, LB, Jevtovic-Todorovic, V. Early exposure to general anesthesia causes significant neuronal deletion in the developing rat brain. Ann N Y Acad Sci 2007;1122:69–82. https://doi.org/10.1196/annals.1403.005.Search in Google Scholar PubMed
58. Lunardi, N, Ori, C, Erisir, A, Jevtovic-Todorovic, V. General anesthesia causes long-lasting disturbances in the ultrastructural properties of developing synapses in young rats. Neurotox Res 2010;17:179–88. https://doi.org/10.1007/s12640-009-9088-z.Search in Google Scholar PubMed PubMed Central
59. Dalla, ML, Osuru, HP, Oklopcic, A, Milanovic, D, Joksimovic, SM, Caputo, V, et al.. General anesthesia causes epigenetic histone modulation of c-fos and brain-derived neurotrophic factor, target genes important for neuronal development in the immature rat Hippocampus. Anesthesiology 2016;124:1311–27. https://doi.org/10.1097/aln.0000000000001111.Search in Google Scholar PubMed PubMed Central
60. Sanchez, V, Feinstein, SD, Lunardi, N, Joksovic, PM, Boscolo, A, Todorovic, SM, et al.. General anesthesia causes long-term impairment of mitochondrial morphogenesis and synaptic transmission in developing rat brain. Anesthesiology 2011;115:992–1002. https://doi.org/10.1097/aln.0b013e3182303a63.Search in Google Scholar PubMed PubMed Central
61. Boscolo, A, Milanovic, D, Starr, JA, Sanchez, V, Oklopcic, A, Moy, L, et al.. Early exposure to general anesthesia disturbs mitochondrial fission and fusion in the developing rat brain. Anesthesiology 2013;118:1086–97. https://doi.org/10.1097/aln.0b013e318289bc9b.Search in Google Scholar PubMed PubMed Central
62. Young, C, Jevtovic-Todorovic, V, Qin, YQ, Tenkova, T, Wang, H, Labruyere, J, et al.. Potential of ketamine and midazolam, individually or in combination, to induce apoptotic neurodegeneration in the infant mouse brain. Br J Pharmacol 2005;146:189–97. https://doi.org/10.1038/sj.bjp.0706301.Search in Google Scholar PubMed PubMed Central
63. Tan, H, Ren, RR, Xiong, ZQ, Wang, YW. Effects of ketamine and midazolam on morphology of dendritic spines in hippocampal CA1 region of neonatal mice. Chin Med J 2009;122:455–9. https://doi.org/10.3760/cma.j.issn.0366-6999.2009.04.0018.Search in Google Scholar
64. Sinner, B, Friedrich, O, Zausig, Y, Bein, T, Graf, BM. Toxic effects of midazolam on differentiating neurons in vitro as a consequence of suppressed neuronal Ca2+-oscillations. Toxicology 2011;290:96–101. https://doi.org/10.1016/j.tox.2011.08.022.Search in Google Scholar PubMed
65. Osterop, SF, Virtanen, MA, Loepke, JR, Joseph, B, Loepke, AW, Vutskits, L. Developmental stage-dependent impact of midazolam on calbindin, calretinin and parvalbumin expression in the immature rat medial prefrontal cortex during the brain growth spurt. Int J Dev Neurosci 2015;45:19–28. https://doi.org/10.1016/j.ijdevneu.2015.04.002.Search in Google Scholar PubMed
66. Iqbal, O, Meara, AM, Miller Ferguson, N, Zven, SE, Karam, OL, Meyer, LC, et al.. Potential neurodevelopmental effects of pediatric intensive care sedation and analgesia: repetitive benzodiazepine and opioid exposure alters expression of glial and synaptic proteins in juvenile rats. Crit Care Explor 2020;2:e0105. https://doi.org/10.1097/cce.0000000000000105.Search in Google Scholar PubMed PubMed Central
67. Nguyen, NM, Vellichirammal, NN, Guda, C, Pendyala, G. Decoding the synaptic proteome with long-term exposure to midazolam during early development. Int J Mol Sci 2022;23. https://doi.org/10.3390/ijms23084137.Search in Google Scholar PubMed PubMed Central
68. Nguyen, NM, Meyer, D, Meyer, L, Chand, S, Jagadesan, S, Miravite, M, et al.. Identification of YWHAH as a novel brain-derived extracellular vesicle marker post long-term midazolam exposure during early development. Cells 2023;12. https://doi.org/10.3390/cells12060966.Search in Google Scholar PubMed PubMed Central
69. Colon, E, Bittner, EA, Kussman, B, McCann, ME, Soriano, S, Borsook, D. Anesthesia, brain changes, and behavior: insights from neural systems biology. Prog Neurobiol 2017;153:121–60. https://doi.org/10.1016/j.pneurobio.2017.01.005.Search in Google Scholar PubMed
70. DiMaggio, C, Sun, LS, Ing, C, Li, G. Pediatric anesthesia and neurodevelopmental impairments: a Bayesian meta-analysis. J Neurosurg Anesthesiol 2012;24:376–81. https://doi.org/10.1097/ana.0b013e31826a038d.Search in Google Scholar
71. DiMaggio, C, Sun, LS, Kakavouli, A, Byrne, MW, Li, G. A retrospective cohort study of the association of anesthesia and hernia repair surgery with behavioral and developmental disorders in young children. J Neurosurg Anesthesiol 2009;21:286–91. https://doi.org/10.1097/ana.0b013e3181a71f11.Search in Google Scholar PubMed PubMed Central
72. Flores, A, Nguyen, NM, Pendyala, G. Developmental outcomes with perinatal exposure (DOPE) to prescription opioids. NeuroImmune Pharm Therapeut 2023;2:339–51. https://doi.org/10.1515/nipt-2023-0017.Search in Google Scholar PubMed PubMed Central
73. Odegaard, KE, Schaal, VL, Clark, AR, Koul, S, Gowen, A, Sankarasubramani, J, et al.. Characterization of the intergenerational impact of in utero and postnatal oxycodone exposure. Transl Psychiatry 2020;10:329. https://doi.org/10.1038/s41398-020-01012-z.Search in Google Scholar PubMed PubMed Central
74. Davidson, A, Flick, RP. Neurodevelopmental implications of the use of sedation and analgesia in neonates. Clin Perinatol 2013;40:559–73. https://doi.org/10.1016/j.clp.2013.05.009.Search in Google Scholar PubMed
75. Flick, RP, Katusic, SK, Colligan, RC, Wilder, RT, Voigt, RG, Olson, MD, et al.. Cognitive and behavioral outcomes after early exposure to anesthesia and surgery. Pediatrics 2011;128:e1053–61. https://doi.org/10.1542/peds.2011-0351.Search in Google Scholar PubMed PubMed Central
76. Ing, C, DiMaggio, C, Whitehouse, A, Hegarty, MK, Brady, J, von Ungern-Sternberg, BS, et al.. Long-term differences in language and cognitive function after childhood exposure to anesthesia. Pediatrics 2012;130:e476–85. https://doi.org/10.1542/peds.2011-3822.Search in Google Scholar PubMed
77. Ing, CH, DiMaggio, CJ, Malacova, E, Whitehouse, AJ, Hegarty, MK, Feng, T, et al.. Comparative analysis of outcome measures used in examining neurodevelopmental effects of early childhood anesthesia exposure. Anesthesiology 2014;120:1319–32. https://doi.org/10.1097/aln.0000000000000248.Search in Google Scholar
78. Duerden, EG, Guo, T, Dodbiba, L, Chakravarty, MM, Chau, V, Poskitt, KJ, et al.. Midazolam dose correlates with abnormal hippocampal growth and neurodevelopmental outcome in preterm infants. Ann Neurol 2016;79:548–59. https://doi.org/10.1002/ana.24601.Search in Google Scholar PubMed
79. Duerden, EG, Guo, T, Chau, C, Chau, V, Synnes, A, Grunau, RE, et al.. Association of neonatal midazolam exposure with hippocampal growth and working memory performance in children born preterm. Neurology 2023;101:e1863–72. https://doi.org/10.1212/wnl.0000000000207817.Search in Google Scholar PubMed PubMed Central
80. McGraw, T, Kendrick, A. Oral midazolam premedication and postoperative behaviour in children. Paediatr Anaesth 1998;8:117–21. https://doi.org/10.1046/j.1460-9592.1998.00724.x.Search in Google Scholar PubMed
81. Puia-Dumitrescu, M, Comstock, BA, Li, S, Heagerty, PJ, Perez, KM, Law, JB, et al.. Assessment of 2-year neurodevelopmental outcomes in extremely preterm infants receiving opioids and benzodiazepines. JAMA Netw Open 2021;4:e2115998. https://doi.org/10.1001/jamanetworkopen.2021.15998.Search in Google Scholar PubMed PubMed Central
82. Wise-Faberowski, L, Quinonez, ZA, Hammer, GB. Anesthesia and the developing brain: relevance to the pediatric cardiac surgery. Brain Sci 2014;4:295–310. https://doi.org/10.3390/brainsci4020295.Search in Google Scholar PubMed PubMed Central
83. Torolira, D, Suchomelova, L, Wasterlain, CG, Niquet, J. Phenobarbital and midazolam increase neonatal seizure-associated neuronal injury. Ann Neurol 2017;82:115–20. https://doi.org/10.1002/ana.24967.Search in Google Scholar PubMed PubMed Central
84. Brambrink, AM, Evers, AS, Avidan, MS, Farber, NB, Smith, DJ, Martin, LD, et al.. Ketamine-induced neuroapoptosis in the fetal and neonatal rhesus macaque brain. Anesthesiology 2012;116:372–84. https://doi.org/10.1097/aln.0b013e318242b2cd.Search in Google Scholar
85. Lei, S, Davis, N, Lee, M, Ing, C. Engaging stakeholders in research related to anesthesia and neurodevelopment in children. J Neurosurg Anesthesiol 2014;26:387–90. https://doi.org/10.1097/ana.0000000000000122.Search in Google Scholar PubMed
86. Fields, RD, Araque, A, Johansen-Berg, H, Lim, SS, Lynch, G, Nave, KA, et al.. Glial biology in learning and cognition. Neuroscientist 2014;20:426–31. https://doi.org/10.1177/1073858413504465.Search in Google Scholar PubMed PubMed Central
87. Pearson-Leary, J, Osborne, DM, McNay, EC. Role of glia in stress-induced enhancement and impairment of memory. Front Integr Neurosci 2015;9:63. https://doi.org/10.3389/fnint.2015.00063.Search in Google Scholar PubMed PubMed Central
88. Tanabe, K, Kozawa, O, Iida, H. Midazolam suppresses interleukin-1β-induced interleukin-6 release from rat glial cells. J Neuroinflammation 2011;8:68. https://doi.org/10.1186/1742-2094-8-68.Search in Google Scholar PubMed PubMed Central
89. Lunardi, N, Hucklenbruch, C, Latham, JR, Scarpa, J, Jevtovic-Todorovic, V. Isoflurane impairs immature astroglia development in vitro: the role of actin cytoskeleton. J Neuropathol Exp Neurol 2011;70:281–91. https://doi.org/10.1097/nen.0b013e31821284e9.Search in Google Scholar PubMed PubMed Central
90. Culley, DJ, Cotran, EK, Karlsson, E, Palanisamy, A, Boyd, JD, Xie, Z, et al.. Isoflurane affects the cytoskeleton but not survival, proliferation, or synaptogenic properties of rat astrocytes in vitro. Br J Anaesth 2013;110:i19–28. https://doi.org/10.1093/bja/aet169.Search in Google Scholar PubMed PubMed Central
91. Ryu, YK, Khan, S, Smith, SC, Mintz, CD. Isoflurane impairs the capacity of astrocytes to support neuronal development in a mouse dissociated coculture model. J Neurosurg Anesthesiol 2014;26:363–8. https://doi.org/10.1097/ana.0000000000000119.Search in Google Scholar
92. Erasso, DM, Camporesi, EM, Mangar, D, Saporta, S. Effects of isoflurane or propofol on postnatal hippocampal neurogenesis in young and aged rats. Brain Res 2013;1530:1–12. https://doi.org/10.1016/j.brainres.2013.07.035.Search in Google Scholar PubMed
93. Shen, X, Dong, Y, Xu, Z, Wang, H, Miao, C, Soriano, SG, et al.. Selective anesthesia-induced neuroinflammation in developing mouse brain and cognitive impairment. Anesthesiology 2013;118:502–15. https://doi.org/10.1097/aln.0b013e3182834d77.Search in Google Scholar
94. Brambrink, AM, Back, SA, Riddle, A, Gong, X, Moravec, MD, Dissen, GA, et al.. Isoflurane-induced apoptosis of oligodendrocytes in the neonatal primate brain. Ann Neurol 2012;72:525–35. https://doi.org/10.1002/ana.23652.Search in Google Scholar PubMed PubMed Central
95. Creeley, C, Dikranian, K, Dissen, G, Martin, L, Olney, J, Brambrink, A. Propofol-induced apoptosis of neurones and oligodendrocytes in fetal and neonatal rhesus macaque brain. Br J Anaesth 2013;110:i29–38. https://doi.org/10.1093/bja/aet173.Search in Google Scholar
96. Nagele, P, Tallchief, D, Blood, J, Sharma, A, Kharasch, ED. Nitrous oxide anesthesia and plasma homocysteine in adolescents. Anesth Analg 2011;113:843–8. https://doi.org/10.1213/ane.0b013e31822402f5.Search in Google Scholar
97. Smith, GT, Chen, TJ, Shah, NM, Agrest, B, Grotticelli, J. Anesthesia-mediated neuroinflammatory sequelae in post operative cognitive dysfunction: mechanisms and therapeutic implications. Front Anesthesiol 2024;3. https://doi.org/10.3389/fanes.2024.1281034.Search in Google Scholar
98. Dominguini, D, Steckert, AV, Michels, M, Spies, MB, Ritter, C, Barichello, T, et al.. The effects of anaesthetics and sedatives on brain inflammation. Neurosci Biobehav Rev 2021;127:504–13. https://doi.org/10.1016/j.neubiorev.2021.05.009.Search in Google Scholar
99. Horiguchi, Y, Ohta, N, Yamamoto, S, Koide, M, Fujino, Y. Midazolam suppresses the lipopolysaccharide-stimulated immune responses of human macrophages via translocator protein signaling. Int Immunopharm 2019;66:373–82. https://doi.org/10.1016/j.intimp.2018.11.050.Search in Google Scholar
100. Shi, Y, Cui, M, Ochs, K, Brendel, M, Strübing, FL, Briel, N, et al.. Long-term diazepam treatment enhances microglial spine engulfment and impairs cognitive performance via the mitochondrial 18 kDa translocator protein (TSPO). Nat Neurosci 2022;25:317–29. https://doi.org/10.1038/s41593-022-01013-9.Search in Google Scholar
101. Wilms, H, Claasen, J, Röhl, C, Sievers, J, Deuschl, G, Lucius, R. Involvement of benzodiazepine receptors in neuroinflammatory and neurodegenerative diseases: evidence from activated microglial cells in vitro. Neurobiol Dis 2003;14:417–24. https://doi.org/10.1016/j.nbd.2003.07.002.Search in Google Scholar
102. Park, CH, Carboni, E, Wood, PL, Gee, KW. Characterization of peripheral benzodiazepine type sites in a cultured murine BV-2 microglial cell line. Glia 1996;16:65–70. https://doi.org/10.1002/(sici)1098-1136(199601)16:1<65::aid-glia7>3.0.co;2-a.10.1002/(SICI)1098-1136(199601)16:1<65::AID-GLIA7>3.0.CO;2-ASearch in Google Scholar
103. Ramirez, K, Niraula, A, Sheridan, JF. GABAergic modulation with classical benzodiazepines prevent stress-induced neuro-immune dysregulation and behavioral alterations. Brain Behav Immun 2016;51:154–68. https://doi.org/10.1016/j.bbi.2015.08.011.Search in Google Scholar
104. Selvaraj, V, Stocco, DM. The changing landscape in translocator protein (TSPO) function. Trends Endocrinol Metabol 2015;26:341–8. https://doi.org/10.1016/j.tem.2015.02.007.Search in Google Scholar
105. Wang, M, Wang, X, Zhao, L, Ma, W, Rodriguez, IR, Fariss, RN, et al.. Macroglia-microglia interactions via TSPO signaling regulates microglial activation in the mouse retina. J Neurosci 2014;34:3793–806. https://doi.org/10.1523/jneurosci.3153-13.2014.Search in Google Scholar
106. Obiora, E, Hubbard, R, Sanders, RD, Myles, PR. The impact of benzodiazepines on occurrence of pneumonia and mortality from pneumonia: a nested case-control and survival analysis in a population-based cohort. Thorax 2013;68:163–70. https://doi.org/10.1136/thoraxjnl-2012-202374.Search in Google Scholar PubMed
107. Riker, RR, Shehabi, Y, Bokesch, PM, Ceraso, D, Wisemandle, W, Koura, F, et al.. Dexmedetomidine vs midazolam for sedation of critically ill patients: a randomized trial. JAMA 2009;301:489–99. https://doi.org/10.1001/jama.2009.56.Search in Google Scholar PubMed
108. Riegel, AC, Kalivas, PW. Neuroscience: lack of inhibition leads to abuse. Nature 2010;463:743–4. https://doi.org/10.1038/463743a.Search in Google Scholar PubMed PubMed Central
109. Rump, K, Holtkamp, C, Bergmann, L, Nowak, H, Unterberg, M, Orlowski, J, et al.. Midazolam impacts acetyl-And butyrylcholinesterase genes: an epigenetic explanation for postoperative delirium? PLoS One 2022;17:e0271119. https://doi.org/10.1371/journal.pone.0271119.Search in Google Scholar PubMed PubMed Central
110. Tokuda, K, O’Dell, KA, Izumi, Y, Zorumski, CF. Midazolam inhibits hippocampal long-term potentiation and learning through dual central and peripheral benzodiazepine receptor activation and neurosteroidogenesis. J Neurosci 2010;30:16788–95. https://doi.org/10.1523/jneurosci.4101-10.2010.Search in Google Scholar
111. Wu, Z, Zhao, P. Epigenetic alterations in anesthesia-induced neurotoxicity in the developing brain. Front Physiol 2018;9:1024. https://doi.org/10.3389/fphys.2018.01024.Search in Google Scholar PubMed PubMed Central
112. Bi, J, Zhang, H, Lu, J, Lei, W. Nobiletin ameliorates isoflurane-induced cognitive impairment via antioxidant, anti-inflammatory and anti-apoptotic effects in aging rats. Mol Med Rep 2016;14:5408–14. https://doi.org/10.3892/mmr.2016.5919.Search in Google Scholar PubMed
113. Ding, ML, Ma, H, Man, YG, Lv, HY. Protective effects of a green tea polyphenol, epigallocatechin-3-gallate, against sevoflurane-induced neuronal apoptosis involve regulation of CREB/BDNF/TrkB and PI3K/Akt/mTOR signalling pathways in neonatal mice. Can J Physiol Pharmacol 2017;95:1396–405. https://doi.org/10.1139/cjpp-2016-0333.Search in Google Scholar PubMed
© 2024 the author(s), published by De Gruyter, Berlin/Boston
This work is licensed under the Creative Commons Attribution 4.0 International License.
Articles in the same Issue
- Frontmatter
- Brief Report
- Elevated cerebral oxygen extraction in patients with post-COVID conditions
- Review Articles
- Build muscles and protect myelin
- Cannabis use, oral dysbiosis, and neurological disorders
- Sedation with midazolam in the NICU: implications on neurodevelopment
- The challenges to detect, quantify, and characterize viral reservoirs in the current antiretroviral era
- Research Articles
- Biodegradable cannabidiol: a potential nanotherapeutic for neuropathic pain
- Motivational dysregulation with melanocortin 4 receptor haploinsufficiency
Articles in the same Issue
- Frontmatter
- Brief Report
- Elevated cerebral oxygen extraction in patients with post-COVID conditions
- Review Articles
- Build muscles and protect myelin
- Cannabis use, oral dysbiosis, and neurological disorders
- Sedation with midazolam in the NICU: implications on neurodevelopment
- The challenges to detect, quantify, and characterize viral reservoirs in the current antiretroviral era
- Research Articles
- Biodegradable cannabidiol: a potential nanotherapeutic for neuropathic pain
- Motivational dysregulation with melanocortin 4 receptor haploinsufficiency