Next Article in Journal
Therapeutic Potential of Ginsenosides on Bone Metabolism: A Review of Osteoporosis, Periodontal Disease and Osteoarthritis
Previous Article in Journal
Triple-Negative Breast Cancer Subclassified by Immunohistochemistry: Correlation with Clinical and Pathological Outcomes in Patients Receiving Neoadjuvant Chemotherapy
Previous Article in Special Issue
Circulating microRNA Profiles Identify a Patient Subgroup with High Inflammation and Severe Symptoms in Schizophrenia Experiencing Acute Psychosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Fetal Brain-Derived Exosomal miRNAs from Maternal Blood: Potential Diagnostic Biomarkers for Fetal Alcohol Spectrum Disorders (FASDs)

1
Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
2
Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, USA
3
Medical College of Wisconsin-Prevea Health, Green Bay, WI 54304, USA
4
Department of Obstetrics & Gynecology, University of Texas, Houston, TX 77030, USA
5
Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
6
Department of Neurology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
*
Authors to whom correspondence should be addressed.
These authors contributed equally as co-first authors.
Int. J. Mol. Sci. 2024, 25(11), 5826; https://doi.org/10.3390/ijms25115826
Submission received: 29 March 2024 / Revised: 20 May 2024 / Accepted: 20 May 2024 / Published: 27 May 2024

Abstract

:
Fetal alcohol spectrum disorders (FASDs) are leading causes of neurodevelopmental disability but cannot be diagnosed early in utero. Because several microRNAs (miRNAs) are implicated in other neurological and neurodevelopmental disorders, the effects of EtOH exposure on the expression of these miRNAs and their target genes and pathways were assessed. In women who drank alcohol (EtOH) during pregnancy and non-drinking controls, matched individually for fetal sex and gestational age, the levels of miRNAs in fetal brain-derived exosomes (FB-Es) isolated from the mothers’ serum correlated well with the contents of the corresponding fetal brain tissues obtained after voluntary pregnancy termination. In six EtOH-exposed cases and six matched controls, the levels of fetal brain and maternal serum miRNAs were quantified on the array by qRT-PCR. In FB-Es from 10 EtOH-exposed cases and 10 controls, selected miRNAs were quantified by ddPCR. Protein levels were quantified by ELISA. There were significant EtOH-associated reductions in the expression of several miRNAs, including miR-9 and its downstream neuronal targets BDNF, REST, Synapsin, and Sonic hedgehog. In 20 paired cases, reductions in FB-E miR-9 levels correlated strongly with reductions in fetal eye diameter, a prominent feature of FASDs. Thus, FB-E miR-9 levels might serve as a biomarker to predict FASDs in at-risk fetuses.

1. Introduction

Prenatal exposure to ethanol (EtOH) causes variable abnormalities known as fetal alcohol spectrum disorders (FASDs), of which the most severe and multisystemic is fetal alcohol syndrome (FAS), which is the leading cause of the only known preventable neurodevelopmental impairment. The estimated global prevalence of FASDs among the general population is 7.7 cases/1000 individuals. In fact, FASDs’ prevalence is apparently highest in the WHO European Region (19.8 per 1000) and lowest in the WHO Eastern Mediterranean Region (0.1 per 1000) [1]. Unfortunately, many women drink alcohol (EtOH) before they know they are pregnant, and this poses a potential diagnostic problem because FASDs cannot be diagnosed early in utero [1,2] when it might be easiest to intervene therapeutically. In addition, almost half of the estimated 80,000 children born with FASDs each year in the US go undiagnosed. Children with FASDs have facial abnormalities, small eyes and head size, and prominent cognitive and behavioral deficits [3]. The exact mechanisms for these abnormalities are not known, and the mechanisms by which EtOH disrupts fetal brain development are complex and incompletely understood, as are the genetic factors that modify individual vulnerability. The present study is part of a series aimed at identifying potential biomarkers for FASDs, with the goal of testing their predictive value in identifying which at-risk fetuses will present the clinical features of FASDs in post-natal life [4,5].
Since FASDs cannot be diagnosed early in utero by conventional imaging, there is a need to find early biomarkers that can predict which at-risk fetuses will go on to have FASDs postnatally and provide clues to early interventions that might prevent or ameliorate FASDs. Among the potential biomarkers, microRNAs (miRNAs) have attracted attention [6,7,8,9,10,11]. miRNAs act by binding to or destabilizing mRNAs and repressing protein translation and play important roles in brain development [12]. They also have been implicated in neurological diseases, including those caused by prenatal exposure to EtOH. Because individual miRNAs are involved in regulating expressions of hundreds of genes on average, they represent attractive subjects of investigation to elucidate the pathogenesis of complex disorders [13], such as FASDs. A summary of published data on some of the molecular targets of specific miRNAs and their associated neurodevelopmental functions or disorders are shown in Supplemental Data, Table S1 [14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143].
Changes in miRNA levels have been described in animal models of FASDs, as well as in human fetuses exposed to EtOH in vivo [144,145,146,147]. Thus, FASDs might be predicted by altered fetal brain levels of specific miRNAs, including microRNA-9 (miR-9) and its target molecules, e.g., brain-derived neurotrophic factor (BDNF), restrictive element-1 silencing transcription factor (REST), and Sonic hedgehog protein (Shh). Our laboratory has developed methods to measure the cargo of fetal brain-specific exosomes (FB-Es) that can be harvested non-invasively from the mother’s blood [148,149]. Correlation between FB-E miRNA changes and changes in an anatomical hallmark of FASDs, e.g., eye diameter [150,151], would suggest that specific miRNAs in FB-Es might be useful in the detection of FASDs early in utero and suggest possible therapeutic approaches aimed at preventing or ameliorating FASDs.

2. Results

2.1. Prenatal EtOH Exposure Was Associated with Reduced miRNA Levels in Fetal Brain but Increased miRNA Levels in Maternal Serum during Development

To understand the dynamics of miRNA changes, we quantified the expression levels of 84 miRNAs that are important for neurogenesis and neuronal development, including miR-9 and miR-132, in human fetal brain samples and matching maternal serum samples obtained from a previously organized human tissue biobank, created to study the effects of prenatal EtOH exposure on fetal brain development. miR-9 and miR-132 share molecular targets, and in rodent and ovine fetuses, had been shown to be downregulated on exposure to EtOH [147]. Each EtOH-exposed case was paired with a fetal sex-, GA-, maternal age-, and race-matched control from GA 9–23. Biometric and clinical characteristics of the subjects are shown in Table 1.
Table 1. Fetal brain and maternal blood used in miRNA studies. EtOH cases (n = 40) vs. Controls (n = 40). Ethanol-exposed fetal brain samples and blood samples from mothers who consumed EtOH during pregnancy were matched with non-EtOH-exposed controls by sex, GA, and race (white: Caucasian; black: African-American, some of them brown-skinned). In our population, other racial minorities were relatively few. Therefore, for this limited study, other non-Caucasian groups were not included but will be incorporated in future larger-scale studies, along with maternal age. PCR for the SRY gene was performed to determine fetal sex.
The 84 assayed miRNAs were represented on a Neurologic Disorders-Associated miRNA Array (Qiagen), which included markers previously reported to be associated with either normal neurodevelopment or neurological/neuropsychiatric disorders. Table 2 lists the miRNAs included in the array, together with the associated conditions that are pertinent to the neurodevelopment and neuropathology of FASDs. Major miRNAs implicated in CNS development and neurogenesis are presented in Table 3 [152,153,154,155,156,157,158,159,160,161,162,163,164,165,166,167,168,169,170,171,172,173,174,175,176,177,178,179,180,181,182,183].
Codes
  • NPCs: neural progenitor cells
  • ELAVL3/4: embryonic lethal abnormal vision-like 3/4;
  • EZH2: enhancer of zeste 2 polycomb repressive complex 2 subunit;
  • MSI1: Musashi1
  • PTBP1/2: polypyrimidine tract binding protein 1/2
  • REST: RE1 Silencing Transcription Factor
  • SCP1: CTD small phosphatase 1 (also known as CTDSP1)
  • ZFP36: ZFP36 ring finger protein;
  • ZFP36L1: ZFP36 ring finger protein like 1
  • Sox9: SRY-Box Transcription Factor 9
  • NOVA1: NOVA Alternative Splicing Regulator 1, Neuro-Oncological Ventral Antigen 1 N NOVA Alternative Splic1
  • Rbfox1: RNA Binding Fox-1 Homolog 1
  • Phf6: X-linked syndromic intellectual disability gene
  • PCM1: pericentriolar material 1
  • AMPARs: AMPA-type glutamate receptors
  • CREB: cAMP response element binding protein
  • MeCP2: methyl CpG-binding protein 2
  • Cdh5: vascular endothelial cadherin (VE-cadherin)
  • FMRP: fragile X mental retardation protein
Figure 1A illustrates the developmental changes in overall miRNA expression in the maternal serum and fetal brain, presented as heat maps of the 84 miRNAs in the microarray, relative values at 11.3 weeks gestational age (GA), which is defined as baseline. Changes in the developmental pattern associated with exposure to EtOH are illustrated in Figure 1B. The position of each miRNA in the 96-well plate is specified in Figure 1C. Each sample was used for one 96-well pre-plated miRNA array. Thus, a total of 24 plates were used for 24 samples in the array: six control maternal sera, six EtOH-exposed maternal sera, six control fetal brains, and six EtOH-exposed fetal brains.
Compared to the 11.3 weeks baseline, miRNA expression in the unexposed control maternal serum was reduced (green) at 12.2 weeks GA and even lower at 18.3 weeks (Figure 1A, top). This pattern was less consistent in the fetal brain (mostly neocortex, Figure 1A, bottom). When pregnant women who consumed EtOH were compared to their individually matched, unexposed controls, the EtOH-exposed group showed greatly reduced (green) serum miRNA levels at 11.3 weeks GA, but then the levels increased (red) dramatically by 18.3 weeks (Figure 1B, top). The relative changes in miRNA levels in the fetal brains were less consistent, but generally, the levels were slightly lower in the EtOH-exposed fetal brains than in their individually matched controls.

2.2. EtOH Exposure Was Associated with Changes in Expression of Array miRNAs

EtOH exposure was associated with dysregulation of miRNA signaling in both the fetal brain and maternal serum. From a combined heatmap expression of several miRNAs appeared to be most dramatically changed. Nine of these miRNAs and their downstream targets are listed in Table 4 and were assayed by qRT-PCR in six EtOH-exposed and six unexposed matched control maternal sera and the corresponding fetal brain homogenates. All but one miRNA (mIR-509) showed significant changes in expression.
In the control cases, a general upregulation of miRNA levels was seen in the secondtrimester maternal serum, compared to the first trimester (Figure 2A). Prenatal exposure to EtOH was associated with a reduction of this upregulation from 3.8-fold to 2.0-fold (Figure 2A). By contrast, in EtOH-exposed fetal brains, the normal 1.9-fold downregulation of miRNA levels seen in controls was converted to a 2.0-fold increase late in the second trimester (Figure 2B). These responses differed by GA. In the first-trimester samples, screened miRNAs were upregulated 2.5–5.5-fold in the fetal brains and downregulated 3.5–6.5-fold in the maternal serum, compared to their matched unexposed controls. In the second-trimester samples, EtOH exposure was associated with smaller effects in the opposite direction–downregulation in the fetal brain specimens and slight upregulation in the maternal serum. Thus, there was a positive correlation between the paired maternal and fetal specimens for several miRNAs at early GAs but a smaller or even negative correlation at late GAs.

2.3. EtOH Exposure Was Associated with Reduced miR-9 and miR-132 in FB-Es

EtOH exposure was associated with reduced miR-9 and miR-132 in FB-Es. Fluctuations of miR-9 and miR-132 levels were analyzed in FB-Es, which carry fetal brain cell-specific nucleic acids, proteins, lipids, and metabolites. FB-Es are produced in the endosomal compartment of the fetal brain cells and released into the maternal blood. Their contents reflect the metabolic and functional state of their cells of origin. FB-Es isolated from the maternal serum were assayed by qRT-PCR for two EtOH-responsive miRNAs, miR-9 and miR-132, which are enriched in neural cells and have been implicated in the regulation of neurogenesis, axon extension, dendritic growth, synaptic structure and function, vascular integrity, and microglial homeostasis [164,172,173,174,175,176,177,178,179,180,181,182,183]. A larger number of cases was analyzed by limiting the control-matching to GA and fetal sex only. Twenty EtOH-exposed cases were compared with their individually matched unexposed controls, as well as with non-pregnant controls. In these same cases, both miR-9 (Figure 3A) and miR-132 (Figure 3B) were downregulated in FB-Es from pregnant mothers who consumed EtOH.

2.4. miR-9 Expression in FB-Es

Copy numbers of miR-9 were measured in FB-Es from the first 10 available pairs of EtOH-exposed mothers and their GA- and fetal sex-matched controls using digital droplet PCR (ddPCR; Figure 4). In each case, the EtOH-exposed sample had fewer copies than its control. The miR-9 levels in the control FB-Es ranged from 200 to 1200 copies per 1 μL, while 180 to 820 copies were found in the EtOH-exposed FB-Es. With the accumulation of additional cases, it was possible to add subjects for other assays reported below.

2.5. miRNA-9 Targets in a Double Negative Feedback Loop Pathway

In both the developing and adult vertebrate brains, miR-9 is expressed at high levels and is involved in regulating the proliferation of neural progenitors [137]. It also is important in the regulation of axon extension and local branching by targeting BDNF, which affects critical components of the cytoskeleton [184]. To understand whether EtOH exposure affects not only miR-9 but also its targets, we performed ELISA and quantitative Western-blot assays (qWestern) on the FB-E cargo from 20 pregnant women and their individually matched controls. There was a 50% reduction in levels of Synapsin (↓2.1-fold; p < 0.01; Figure 5A). Similarly, downregulation was observed in the miR-9 downstream double-negative feedback targets REST (↓1.8-fold; p < 0.05; Figure 5B), Shh (↓2.2-fold; p < 0.05; Figure 5C), and BDNF (↓1.4-fold; p < 0.05; Figure 5D), suggesting that EtOH exposure might be associated with synaptic injury and that this could be detected non-invasively using FB-Es. The postulated double-negative feedback loop between miR-9 and its molecular target REST and a triple-negative net positive feedback loop through BDNF were previously shown by others [184,185,186,187,188,189] and are diagrammed in Figure 5E. The net effect of these feedback loops is that miR-9 controls its own expression via the regulation of its targets.

2.6. Reductions in FB-E miR-9 Levels in Fetuses Exposed to EtOH Correlate with Reductions in Eye Diameter in Fetuses Exposed to EtOH in a Larger Population

Previously, we measured eye diameters in 10 histological sections of human fetuses that had or had not been exposed to EtOH. Levels of miR-9 were measured by ddPCR in FB-Es isolated from the maternal blood drawn at the time of voluntary pregnancy termination. A linear correlation was observed between the reduction in eye size (arithmetic difference between the EtOH-exposed fetus and its paired control) and the reduction in exosomal miR-9 levels. To determine whether FB-E miR-9 levels might be generally useful as a molecular marker to identify those at-risk fetuses that are destined to be born with FASDs, we performed ddPCR assays for FB-E miR-9 in a larger number of cases (N = 40 EtOH-exposed cases and their individually GA-matched unexposed controls), achieved by eliminating maternal age, maternal race, and fetal sex as control variables. The correlations persisted in this larger population (Figure 6). The first-trimester pregnancies ranged from 9 to 14 weeks GA, and the second-trimester pregnancies ranged from 14.1 to 23 weeks GA. The differences in FB-E miR-9 levels between the EtOH and control groups were highly significant (p = 0.000000464825; Figure 6A). *** p < 0.001. Each assay was performed in triplicate, and the values indicated by the dots are the averages of the three determinations. The levels of miR-9 in the control FB-Es rose dramatically early in the second trimester, whereas the miR-9 levels of the EtOH-exposed group did not. Each EtOH-exposed case is compared with its individual control in Figure 6B. Except for the two youngest fetuses, miR-9 levels were lower in each EtOH-exposed case than in its control. Correlations between the reduction in eye size and the reduction in exosomal miR-9 levels are presented as a scatter plot in (Figure 6C). As in Figure 6, the correlation appeared to be almost constant throughout the observed gestational periods when the data were presented as % change (Figure 6D).

3. Discussion

This preliminary study suggests that miRNAs are important cellular targets of fetal alcohol exposure that can be detected early in gestation, even in the first trimester. In particular, miR-9 is identified as a possible molecular biomarker for FASDs that can be quantified in FB-Es isolated non-invasively from maternal blood. These two findings may allow us to conduct large-scale population-based studies to determine whether FB-E miR-9 levels can be used to predict the emergence of FASDs postnatally. Compared with the controls, subjects with EtOH exposure in the first and/or second trimester showed changes in miRNA levels in both the fetal brain and maternal serum. In the first trimester samples, screened miRNAs were downregulated in the maternal serum but upregulated in the fetal brain. In the early second trimester samples, there was a positive correlation between the paired maternal and fetal specimens for several miRNAs, but in the late second trimester, the correlation was much less, or even negative. Because the changes in the fetal brain did not mirror the changes in maternal blood, the effect of EtOH exposure on the fetal brain cannot be attributed to the passive transmission of maternal blood miRNAs to the fetus. The early emergence of changes in molecular markers is important because they might suggest potential therapeutic approaches to limiting the negative effects of exposure to EtOH soon after a woman learns she is pregnant and realizes that her fetus is at risk.

3.1. MicroRNAs as Indicators of Alcohol-Associated Fetal Pathology

The effects of prenatal EtOH exposure on fetal development are complex and often severe. Abnormalities include prenatal and postnatal growth retardation, CNS injury, and facial abnormalities [3,190,191,192]. These effects are difficult to detect early in pregnancy with the current imaging technology. Thus, it is important to develop novel non-invasive tools to predict FASDs in utero. The present study focused on a molecular biomarker, miR-9, which is highly expressed in the fetal brain, can be detected early in fetal development, and is important in regulating axonal elongation and branching via its reciprocal interactions with BDNF and consequent effects on the cytoskeleton. miR-9 also is involved in regulating the proliferation of neural progenitors.
By use of qRT-PCR arrays and even more sensitive ddPCR, we have demonstrated the following: (i) the developmental regulation of miRNAs in the human fetal brain; (ii) the effects of in utero EtOH exposure on miRNA levels, both in the developing brain and in FB-Es; (iii) exposure to EtOH reduces miR-9 levels in FB-Es; and (iv) exposure to EtOH is associated with reduced levels of miR-9 target proteins. It has been suggested that the toxic effects of EtOH on miRNA expression occur primarily during the second trimester, when neurogenesis is active [191]. Interestingly, the numbers of EtOH-sensitive miRNAs increased during the neural differentiation stage, from 4% in fetal neural stem cells (NSCs) to 11% in differentiated neurons, due to the increasing complexity of miRNA contents during neuronal maturation [193,194]. Also, the EtOH-sensitive miRNAs miR-335 and miR-21, which play important roles in regulating NSC fate [194], are no longer EtOH-sensitive by the end of the second trimester. Other EtOH-sensitive miRNAs, such as miR-10a/10b, negative regulators of the Hox gene family, can dysregulate neuronal migration when upregulated during a critical window for neuronal migration, while miR-21 and miR-335 regulate NSC behavior at earlier times [193,195]. Several miRNAs, including miR-9, are EtOH-sensitive during multiple developmental periods (from the embryonic and fetal stages to adulthood [195,196,197,198]). EtOH reduced miR-9 expression early in mouse and fish development [196,198,199] and increased miR-9 expression at later stages of fetal development and in adult animals [195,197]. This developmental switch is probably due to the effects on miR-9 targets, but the exact mechanisms are not completely understood. In the present study, we looked not only at the expression of miR-9 but also its targets (BDNF, Shh, Synapsin, and REST), which are affected by EtOH via its action on miR-9 throughout development. It is likely that downstream effects on the translation of those proteins would change during neuronal maturation and into adulthood. By assessing miR-9 in FB-Es, we avoided confusion caused by the mixing of fetal and maternal miR-9 and its targets.

3.2. FB-Es as Non-Invasive Tools to Assess Potential Molecular Biomarkers

In previous studies, we showed that fetal brain-derived exosomes (FB-Es) can be isolated non-invasively from the maternal blood and contain neuron- [148] and oligodendrocyte-specific [149,150,151] molecules that might be useful as biomarkers for the diagnosis of pathological conditions. Some of these markers were associated with anatomical abnormalities characteristic of FASDs, including reduced eye size. In the present study, we determined how exposure to EtOH during pregnancy affects miRNA levels in both the fetal brain and maternal serum. Based on the results of previous microarray analysis (not published), we focused attention on whether FASDs can be predicted by altered levels of miR-9 in fetal brain-derived exosomes (FB-Es) isolated non-invasively from maternal serum. We tested whether the results from fetal brains and FB-Es were congruent and whether any observed abnormalities are associated with the effects of EtOH on eye diameter. This anatomical hallmark was used because eyes appear early in embryogenesis and because EtOH-associated reductions of eye diameter have been detected even in the first trimester.
The association between early exposure to EtOH and changes in FB-E miRNA levels was detected long before facial features of FASDs could be seen by conventional ultrasound imaging or even MRI. Of particular importance, the EtOH-associated reduction in miR-9 levels, not only in the fetal brains but also in FB-Es, correlated with the reduction in fetal eye diameters, a common morphological feature of FASDs [3]. A similar correlation had been found previously between FB-E myelin basic protein (MBP) levels and reduced eye diameter [150].

3.3. The Association between Exposure to EtOH and Reduced Fetal miRNA Expression Probably Is Due to a Direct Effect of EtOH on Fetal Brain

EtOH exposure was associated with the upregulation of target miRNAs in maternal serum during the second trimester but with downregulation during this time in the fetal brain. Therefore, the effects of EtOH on fetal brain miRNA expression are unlikely to result passively from changes in maternal blood concentrations. Exposure to EtOH was associated with changes in the expression of eight neurodevelopment- and neurological disorder-related miRNAs in maternal serum and fetal brain homogenates. A general downregulation seen in the first trimester disappeared later in development, suggesting that if miRNAs were to be used as biomarkers for FASDs, they would have to be assayed during the first trimester of pregnancy, when imaging methods are least able to resolve the anatomical abnormalities associated with FASDs.

3.4. Limitations

The present study should be viewed as preliminary, in part because the number of EtOH-exposed cases and controls was too small to permit control for all the potential variables that might affect fetal sensitivity to EtOH, such as EtOH dose, maternal obesity, use of tobacco and other drugs of abuse, medications, and socioeconomic status. An ethically unavoidable limitation is that the study was not prospectively randomized, i.e., although we tried to pair each EtOH-exposed fetus with an unexposed control, women were not randomly assigned prospectively to either use EtOH or not use EtOH. Because the biobank from which the samples were obtained was not collected solely to study FASDs, it did not include tissue specimens for biochemical testing to verify alcohol use. However, any under-reporting of EtOH use would have the effect of reducing rather than exaggerating the reported effects of EtOH exposure. Moreover, in studying some other drugs of abuse in our patient population, there was a very high correlation between self-report and drug test results. Finally, since the pregnancies all were terminated, we cannot say for certain which fetuses would have gone on to develop FASDs postnatally. We now are conducting a larger prospective study of pregnancies brought to term (including tissue ethylene glycol testing to verify EtOH use), so that we can determine which at-risk children develop FASDs, and possibly identify molecular determinants of the subtypes and severities of the FASD. Detailed studies on other miRNAs, e.g., miR-124, will be included in future publications.

4. Materials and Methods

4.1. Clinical Samples

Pregnant women who used EtOH were compared with those who did not use alcohol during pregnancy and did not use any drugs or medications (Table 1). Cases were selected based on the availability of fetal brain and eye tissues, matching maternal blood samples, and, at a minimum, on data for fetal sex and GA. In some cases, the controls also were matched with regard to maternal race. Each EtOH-exposed fetus was paired with a sex- and GA-matched control. Consenting women were enrolled between 9- and 23-week GA under Temple University Institutional Review Board (IRB)-approved protocol. Data for both sexes were combined in all assays.
Subject Recruitment. Pregnant women with or without EtOH use during pregnancy were grouped in two GA windows within the first or second trimester (9–23 weeks). GA was confirmed by an ultrasound performed prior to recruitment. Samples from 40 women with or without EtOH use were collected.
Assessment of EtOH Exposure in Pregnancy. Maternal EtOH exposure was determined using a face-to-face questionnaire. Exposure status was based on self-reported EtOH use (modified timeline follow-back). Pregnant women were screened for EtOH use, and then maternal blood, fetal brain (mostly cortex), and eye tissue were collected.
The amount of EtOH was calculated as the total number of drinks consumed in a week multiplied by the number of weeks of exposure. A detailed questionnaire was used based on the NICHD PASS study [200]. Each drink was estimated as the equivalent of one shot (1.5 oz of brandy or 5 oz of wine [148].
Tissue collection. Fetal brain and eye tissues and maternal blood from subjects undergoing elective termination of pregnancy were collected according to an IRB-approved protocol by a trained study coordinator and were transferred to the laboratory within 60 min.

4.2. RNA Preparation

Total RNA was isolated from the fetal brain and FB-Es using the RNeasy Kit (Qiagen, Valencia, CA, USA) with on-column DNA digestion. Fetal sex determination was performed using SuperScript One-Step RT-PCR with Platinum Taq (Life Technologies, Carlsbad, CA, USA), BioRad C1000 Touch Thermal Cycler, and sex-determining SRY primers. The qRT-PCR reaction was performed using the One-Step FAST SYBR Green PCR Master Mix (Qiagen, Valencia, CA, USA). For relative quantification, the expression level of genes was normalized to the housekeeping gene β-actin.

4.3. miRNA Preparation and Real-Time qRT-PCR

Human fetal brain and maternal plasma total miRNA/RNA was isolated with the miRNeasy kit (Qiagen, Valencia, CA, USA), using QIAzol Lysis Reagent with on-column DNA digestion. The qRT-PCR reaction was performed with 1 μg total RNA/miRNA using the One-Step FAST RT-PCR SYBR Green PCR Master Mix (Qiagen). The StepOnePlus Real-Time PCR system thermocycler was used (Applied Biosystems, Waltham, MA, USA). PCR conditions were as follows: activation 95 °C 5 min, PCR 45 cycles: 95 °C 10 s, 60 °C 20 s, 72 °C 30 s, melting curve (95–65 °C), cool to 40 °C 30 s. For the relative quantification, the expression levels of genes were normalized to the housekeeping gene β-actin.
cDNA was obtained using the miRNAs and miScript PCR Systems, containing 5× miScript HighSpec Buffer, 10× miScript Nucleics Mix, and miRNA Reverse Transcriptase mix (Qiagen, Valencia, CA, USA). miRNA Neurological Development and Disease Pathway array (Qiagen) for 90 miRNAs (Table 2, Table 3 and Table S1) was assayed by real-time PCR with the Applied Biosciences Cycler using the fetal brain RNAs, primers, and Cyber Green mix (Qiagen Universal PCR Master Mix). Of the 90 miRNAs on the array, 6 were controls that enabled data analysis using the ΔΔCT method of relative quantification, assessment of reverse transcription performance, and assessment of PCR performance. Real-time PCR experiments were performed on an Applied Biosystems instrument with the following thermal-cycling procedure: 95 °C for 10 min, followed by 40 cycles of 95 °C for 15 s, and 56 °C for 1 min, as specified in the RNA assay protocol provided by Applied Biosciences.

4.4. Droplet Digital PCR (ddPCR)

For the absolute quantitation of mRNA copies, ddPCR was performed using the QX200 Droplet Digital PCR (ddPCR) System (Bio-Rad Laboratories, Inc., Hercules, CA, USA) with QuantaSoft Analysis Pro Software v 1.4 (AP) (Bio-Rad, Hercules, CA, USA). Fifty nanograms of human fetal total RNA was used with the 1st Strand cDNA Synthesis Kit (Qiagen, Valencia, CA, USA). The cDNA (300 dilution) aliquots were added to the BioRad master mix to conduct ddPCR (EvaGreen ddPCR Supermix, BioRad, Hercules, CA, USA). The prepared ddPCR master mix for each sample (20-μL aliquots) was used for droplet formation. PCR conditions were as follows: activation at 95 °C 5 min, PCR 45 cycles at 95 °C 10 s, 60 °C 20 s, 72 °C 30 s, melting curve (95–65 °C), cool to 40 °C 30 s. The absolute quantity of DNA per sample (copies/µL) was calculated using QuantaSoft Analysis Pro Software v 1.4 (AP) (Bio-Rad, Hercules, CA, USA) to analyze ddPCR data for technical errors (Poisson errors). With 20,000 droplets, the above ddPCR protocol yields a linear dynamic range of detection between 1 and 100,000 target mRNA copies/µL. The ddPCR data were exported to Microsoft Excel (Microsoft 365) for further statistical analysis.

4.5. Primers (IDT Inc., Coralville, IA, USA)

β-actin: S: 5′-CTACAATGAGCTGCG TGTGGC-3′,
AS: 5′-CAGGTCCAGACGCAGGATGGC-3′,
BDNF: S: 5′-CAGGGGCATAGACAAAAG-3′, AS: 5′-CTTCCCCTTTTAATGGTC-3′,
AS: 5′-GAGGGACTGAGCTGGACAACCCAC-3′
SRY: Forward 5′-CAT GAA CGC ATT CAT CGT GTG GTC-3′; reverse 5′-CTG CGG GAA GCA AAC TGC AAT TCT T-3′.
Hsa-mir-26b MI0000084
5′CCGGGACCCAGUUCAAGUAAUUCAGGAUAGGUUGUGUGCUGUCCAGCCUGUUCUCCAUUACUUGGCUCGGGGACCGG-3′
Hsa-mir-132 MI0000449
5′CCGCCCCCGCGUCUCCAGGGCAACCGUGGCUUUCGAUUGUUACUGUGGGAACUGGAGGUAACAGUCUACAGCCAUGGUCGCCCCGCAGCACGCCCACGCGC-3′
Hsa-mir-124-1 MI0000443
5′AGGCCUCUCUCUCCGUGUUCACAGCGGACCUUGAUUUAAAUGUCCAUACAAUUAAGGCACGCGGUGAAUGCCAAGAAUGGGGCUG-3′
Hsa-mir-125b-1 MI0000446
5′UGCGCUCCUCUCAGUCCCUGAGACCCUAACUUGUGAUGUUUACCGUUUAAAUCCACGGGUUAGGCUCUUGGGAGCUGCGAGUCGUGCU-3′
Hsa-mir-138-1 MI0000476
5′CCCUGGCAUGGUGUGGUGGGGCAGCUGGUGUUGUGAAUCAGGCCGUUGCCAAUCAGAGAACGGCUACUUCACAACACCAGGGCCACACCACACUACAGG-3′
Hsa-mir-128-1 MI0000447
5′UGAGCUGUUGGAUUCGGGGCCGUAGCACUGUCUGAGAGGUUUACAUUUCUCACAGUGAACCGGUCUCUUUUUCAGCUGCUUC-3′
Hsa-mir-509-1 MI0003196
5′CAUGCUGUGUGUGGUACCCUACUGCAGACAGUGGCAAUCAUGUAUAAUUAAAAAUGAUUGGUACGUCUGUGGGUAGAGUACUGCAUGACACAUG-3′
Hsa-mir-9-1 MI0000466
5′CGGGGUUGGUUGUUAUCUUUGGUUAUCUAGCUGUAUGAGUGGUGUGGAGUCUUCAUAAAGCUAGAUAACCGAAAGUAAAAAUAACCCCA-3′
Hsa-mir-134 MI0000474
5′CAGGGUGUGUGACUGGUUGACCAGAGGGGCAUGCACUGUGUUCACCCUGUGGGCCACCUAGUCACCAACCCUC-3′
Hsa-mir-485 MI0002469
5′ACUUGGAGAGAGGCUGGCCGUGAUGAAUUCGsAUUCAUCAAAGCGAGUCAUACACGGCUCUCCUCUCUUUUAGU-3′
SNORD61-11
5′GCTATGATGAATTTGATTGCATTGATCGTCTGACATGATAATGTATTTTTGTCCTCTAAGAAGTTCTGAGCTT-3′

4.6. ELISA Quantification of Exosomal Proteins

Proteins and CD81 (American Research Products-Cusabio) were quantified by human-specific ELISAs according to the suppliers’ instructions. ELISA data were statistically evaluated using Excel (Microsoft 365, software version 2404) and statistical analysis tools: CurveExpert for ELISA statistics (CUSABIO) or APP 96-well Plate Assay Data Analysis Software 5.0.apk (Cloud-Clone, Katy, TX, USA), available online.

4.7. Quantitative Western Blots

Thirty micrograms of proteins in Laemmli sample buffer were heated at 95 °C for 10 min and separated by 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE), then transferred to supported nitrocellulose membranes. Primary antibodies: anti-REST rabbit polyclonal (AB10514P, Millipore, Burlington, MA, USA), anti-Synapsin, and anti-α-tubulin clone B512 (Sigma-Aldrich, St. Louis, MO, USA). Secondary IRDye® antibodies were used to detect band intensity (normalized to tubulin) using an Odyssey® CLx Imaging System (iS Image Studio™ Software version 3.1) and LiCor dyes.

4.8. Isolation of Fetal Brain-Derived Exosomes (FB-Es) from Maternal Serum and ELISA Quantification of Exosomal Proteins

Human FB-Es were isolated as described previously [148]. In brief, two hundred and fifty microliters of serum was incubated with exosome precipitation solution (EXOQ; System Biosciences, Inc., Mountainview, CA, USA). The resultant suspensions were centrifuged at 1500× g for 30 min at 4 °C, and pellets were resuspended in 400 mL of distilled water with protease and phosphatase inhibitor cocktail for immunochemical enrichment of exosomes. To isolate exosomes from fetal neural sources, total exosome suspensions were incubated for 90 min at 20 °C with 50 μL of 3% bovine serum albumin (BSA; Thermo Scientific, Inc., Waltham, MA, USA) containing 2 μg of mouse monoclonal IgG1 antihuman contactin-2/TAG1 antibody (clone 372913, R&D Systems, Inc., Minneapolis, MN, USA) that had been biotinylated (EZLink sulfo-NHS-biotin System, Thermo Scientific, Inc., USA). Then, 10 μL of Streptavidin-Plus UltraLink resin (PierceThermo Scientific Inc., Waltham, MA, USA) in 40 μL of 3% BSA was added, and the incubation continued for 60 min at 20 °C. After centrifugation at 300× g for 10 min at 4 °C and removal of supernatants, pellets were resuspended in 75 μL of 0.05 mol/L glycine-HCl (pH 3.0), incubated at 4 °C for 10 min, and recentrifuged at 4000× g for 10 min at 4 °C. Each supernatant was mixed in a new 1.5 mL Eppendorf tube with 5 mL of 1 mol/L Tris-HCl (pH 8.0) and 20 μL of 3% BSA, followed by the addition of 0.4 mL of mammalian protein extraction reagent (M-PER; Thermo Scientific Inc., Waltham, MA, USA) containing protease and phosphatase inhibitors prior to storage at −80 °C. For the exosome counts, immunoprecipitated pellets were resuspended in 0.25 mL of 0.05 mol/L glycine-HCl (pH 3.0) at 4 °C with a pH of 7.0 and 1 mol/L Tris-HCl (pH 8.6). The exosome suspensions were diluted 1:200 to permit counting in the range of 1–5 × 108/mL with an NS500 nanoparticle tracking system (NanoSight, Amesbury, UK).

4.9. Statistical Analysis

For the statistical analysis, we used SPSS Statistics from IBM Corp., released in 2017 for Windows, Version 25.0 (Armonk, NY, USA). All data are represented as the mean ± SD for all performed repetitions. Means were analyzed by a one-way ANOVA, with Bonferroni correction. Statistical significance was defined as p < 0.05. Sample numbers are indicated in the figure legends. Data from ddPCR, which measures absolute quantities of DNA per sample (copies/µL), were processed using QuantaSoft Analysis Pro Software (Bio-Rad) to analyze for technical errors (Poisson errors) and then exported to EXCEL for further statistical analysis.

4.10. Ethics: Human Subjects

Consenting mothers were enrolled between 9–23 weeks gestation under a protocol approved by Temple University Institutional Review Board (IRB). This protocol involved no invasive procedures other than routine care. Maternal EtOH exposure was determined with a face-to-face questionnaire that also included questions regarding many types of drugs/medications used. The questionnaire was adapted from that designed to identify and quantify maternal EtOH exposure in the NIH/NIAAA Prenatal Alcohol and SIDS and Stillbirth (PASS) study [199].
All procedures for collection and processing of human brain tissues and blood were performed according to NIH Guidelines by a trained Study Coordinator. All investigators completed Citi Program-Human Subject training, Blood-Borne Pathogens Training, and Biohazard Waste Safety Training annually.
Written informed consent has been obtained from the parents of the patient(s) for studies, and deidentified samples were used for this publication. Informed Consent forms were maintained by the Study Coordinator. The de-identified log sheets contain an assigned accession number, and the age, sex, ethnicity, and race of the patient. Except for an assigned accession number, no identification was kept on the blood samples.
a. Eligibility Criteria: The blood and tissue samples were obtained according to NIH Guidelines through a trained Study Coordinator. Samples were collected regardless of sex, ethnic background, and race.
b. Treatment Plan: Each patient was asked to sign a separate consent form for research on blood and tissue samples. Blood obtained was processed for collection of serum and plasma. No invasive procedures were performed on the mother, other than those used in her routine medical care. Fetal tissues were processed for RNA or protein isolation.
c. Risk and Benefits: There are very small risks of loss of privacy as with any research study where protected health information is viewed. The samples were depersonalized before they were sent to the lab for analysis. There were no additional risks of blood sampling as this was only performed in patients with clinically indicated venous access. There was little anticipated risk from obtaining approximately 2–3 cc of blood, but a well-trained Study Coordinator collected all samples.
There was no direct benefit to the research subjects from participation, but there is significant potential benefit for the future FASDs subjects and the general population. This research represents a reasonable opportunity to further the understanding, prevention, or alleviation of a serious problem affecting the health or welfare of FASDs patients.
d. Informed Consent: Consent forms were maintained by the Study Coordinator and were not sent to the investigator with the samples. The de-identified log sheets and IRB protocol were sent by the Study Coordinator to Principal Investigator with each blood and tissue sample. This sheet contains an assigned accession number, the age, sex, ethnicity, and race of the patient. Except for an assigned accession number, no identification was kept on the blood and tissue samples.

5. Conclusions

Many women drink EtOH before they know they are pregnant and discontinue drinking once their pregnancy is diagnosed. Not all fetuses that are exposed to EtOH develop FASDs, but EtOH-associated reductions in fetal eye diameter occur even in the first trimester. Unfortunately, this and other somatic features of FASDs cannot be detected early in gestation noninvasively by conventional imaging. Thus, it is important to find early molecular biomarkers to predict which at-risk fetuses will go on to develop FASDs. The present study proposes a novel class of biomarkers based on miRNAs that are developmentally regulated and can be detected as early as 9 weeks of gestation. In the present study, EtOH exposure was associated with reduced expression of miR-9 and of its target proteins, BDNF, REST, Shh, and Synapsin, in FB-Es. This may allow biomarkers for FASDs to be assayed non-invasively from a small sample of maternal blood. The biomarkers so identified also may provide hints to therapeutic approaches that could prevent or ameliorate FASDs.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms25115826/s1.

Author Contributions

N.D.: designing the experiments, developing exosome studies, managing the project, supervising all experimental processes, biostatistical search, writing the first draft, participation in reviewing and editing the manuscript, and supporting the project financially. M.H.: organizing tables, working with literature, participation in the data analysis, reviewing and editing the manuscript. D.M.: qRT-PCR and miRNA arrays, data collection and analysis. A.B.: qRT-PCR analysis and RNA isolation. A.D.: methodology, conceptualization, data analysis, writing, reviewing, editing, and visualization. N.M.: supervising students, participation in exosome isolation, ELISA assays, and editing the manuscript. G.T.: editing the manuscript, participation in qRT-PCR, and ELISA assays. L.G.: editing the manuscript, developing exosome studies, and supporting the project financially at its inception. S.A.: editing and reviewing the manuscript. M.E.S.: designing the study and interpreting the data, providing overall scientific expertise, writing the first draft of the manuscript, reviewing and editing the final manuscript, and supporting the project financially. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by USA Pennsylvania State Health Department grant Project 10: 420491-04400-02 to Nune Darbinian; NIH grant R01HD069238 and Gates Foundation grant OPP1119489 to Laura Goetzl; NIH grants R01NS97846, R01NS097846-02S1, R01NS092876 and the National Institute On Alcohol Abuse And Alcoholism (NIAAA) of NIH grant R01AA031319 to Michael Selzer.

Institutional Review Board Statement

IRB protocol Number #21476 from 2013 to 28 February 2023, Early Gestation Alcohol Exposure: Mechanisms of Human Developmental Injury was approved by Temple University IRB Committee. IRB protocols, all questionnaire amendments, and the informed consent documents have been approved by the Temple University IRB Committee. The study was conducted in accordance with the Declaration of Helsinki and approved by the Institutional Review Board of Temple University for studies involving humans. Informed consent was obtained from all subjects involved in the study.

Informed Consent Statement

Procedures involving the collection and processing of tissues and blood were performed according to NIH Guidelines by a trained Study Coordinator. All investigators were trained annually to complete Citi Program-Human Subject training, all required safety training including, Biohazard Waste Safety Training and Blood–Borne Pathogens Training. Samples were de-identified and written informed consent was obtained from the patients for studies by the Study Coordinator. The de-identified log sheets did contain an assigned accession number, fetal sex, age, and race of the patient. Except for an assigned accession number, no other identification was kept on the blood samples. Although this study was performed with de-identified subjects and without follow-up examinations, the study coordinator did meet the mother once and explained the risks of alcohol consumption during pregnancy for the health of both the fetus and mother. It is hoped that the women will consider all the risks during any subsequent pregnancies. The women were not encouraged to terminate their pregnancies, were not paid, and were not provided with any gifts for answering questionnaires.

Data Availability Statement

This study collected demographic, behavioral, and laboratory data from normal, healthy women and from women who drank alcohol during pregnancy. Our research team supports all these activities and has developed a data-sharing plan. We also recognize that additional benefits from data sharing may arise in the future that are not apparent at this time, and we are prepared to work specifically with NIH in addressing all requests for raw data. At the present time, we have not deposited any of these raw data in an existing databank, but will make the data available to other investigators on request, in a manner consistent with NIH guidelines. Consistent with NIH policy, shared data will be rendered “free of identifiers that would permit linkages to individual research participants and variables that could lead to deductive disclosure of the identity of individual subjects” Intellectual property and data generated under this project will be administered in accordance with both University and NIH policies, including the NIH Data Sharing Policy and Implementation Guidance of 5 March 2003, and 0925-0001 and 0925-0002 (Rev 07/2022 through 01/31/2026). With this caveat observed, data will be made available to the NIH/NICHD/NIAAA. Sufficient identifiers will be provided to the NIH so that research participants can be assigned a Global Unique Identifier (GUID), which is a universal subject ID that protects personally identifiable information (PII). Using the GUID, NDAR can bring together multiple types of data collected from a single participant, regardless of where and when those data were collected. Biological samples (blood, serum, exosomes, and RNAs) and data that are shared will be completely free of identifiers that would permit linkages to individual research participants. We will make biological samples, deidentified data, and associated documentation available to users only under a data-sharing agreement that provides for (1) a commitment to using the data only for research purposes, (2) a commitment to securing the data using appropriate computer technology; and (3) a commitment to destroying or returning remaining samples after analyses are completed. Intellectual property and data generated under this project will be administered in accordance with both University and NIH policies, including the NIH Data Sharing Policy and Implementation Guidance of 5 March 2003. As the FAIR data bank receives approval from the NIH, the data will be made available to that group as well. The NIH will be implementing a new specific policy regarding data sharing https://grants.nih.gov/grants/guide/notice-files/NOT-OD-21-014.html, as of 25 January 2023. We will adopt that policy also. Data will be also available at https://www.mdpi.com/ethics accessed on 1 January 2025).

Acknowledgments

We thank study coordinator Tamara Tatevosian-Geller, MPH for her assistance in the collecting of clinical samples. We also thank Bohdan Metenko for the help with miRNA array studies. This work was supported by the USA Pennsylvania State Health Department grant Project 10: 420491-04400-02 to Nune Darbinian; the National Institute of Neurological Disorders and Stroke (NINDS) of the National Institutes of Health (NIH) grants R01NS97846, R01NS097846-02S1, and R01NS092876 to Michael Selzer; the National Institute of Child Health and Human Development (NICHD) of NIH grant R01HD069238 to Laura Goetzl; the National Institute On Alcohol Abuse And Alcoholism (NIAAA) of NIH grant R01AA031319 to Michael Selzer; and the Gates Foundation grant OPP1119489 to Laura Goetzl. The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

BDNFbrain-derived neurotrophic factor
ddPCRdigital droplet polymerase chain reaction
ELISAenzyme-linked immunosorbent assay
EtOHethanol, alcohol
FASfetal alcohol syndrome
FASDfetal alcohol spectrum disorders
FB-Esfetal brain-derived exosomes
GAgestation age
miR-9microRNA-9
qRT-PCRquantitative reverse transcription polymerase chain reaction
RESTrestrictive element-1 silencing transcription factor
ShhSonic hedgehog protein
SNORDsmall nucleolar RNA D
SRYsex-determining region of the Y chromosome

References

  1. Popova, S.; Charness, M.E.; Burd, L.; Crawford, A.; Hoyme, H.E.; Mukherjee RA, S.; Riley, E.P.; Elliott, E.J. Fetal alcohol spectrum disorders. Nat. Rev. Dis. Primers 2023, 9, 11. [Google Scholar] [CrossRef] [PubMed]
  2. May, P.A.; Baete, A.; Russo, J.; Elliott, A.J.; Blankenship, J.; Kalberg, W.O.; Buckley, D.; Brooks, M.; Hasken, J.; Abdul-Rahman, O.; et al. Prevalence and characteristics of fetal alcohol spectrum disorders. Pediatrics 2014, 134, 855–866. [Google Scholar] [CrossRef] [PubMed]
  3. Hoyme, H.E.; Kalberg, W.O.; Elliott, A.J.; Blankenship, J.; Buckley, D.; Marais, A.S.; Manning, M.A.; Robinson, L.K.; Adam, M.P.; Abdul-Rahman, O.; et al. Updated Clinical Guidelines for Diagnosing Fetal Alcohol Spectrum Disorders. Pediatrics 2016, 138, e20154256. [Google Scholar] [CrossRef] [PubMed]
  4. Balaraman, S.; Schafer, J.J.; Tseng, A.M.; Wertelecki, W.; Yevtushok, L.; Zymak-Zakutnya, N.; Chambers, C.D.; Miranda, R.C. Plasma miRNA Profiles in Pregnant Women Predict Infant Out- 946 comes following Prenatal Alcohol Exposure. PLoS ONE 2016, 11, e0165081. [Google Scholar] [CrossRef] [PubMed]
  5. Mahnke, A.H.; Salem, N.A.; Tseng, A.M.; Chung, D.D.; Miranda, R.C. Nonprotein-coding 952 RNAs in Fetal Alcohol Spectrum Disorders. Prog. Mol. Biol. Transl. Sci. 2018, 157, 299–342. [Google Scholar] [CrossRef] [PubMed]
  6. Zhao, J.; He, Z.; Wang, J. MicroRNA-124: A Key Player in Microglia-Mediated Inflammation in Neurological Diseases. Front. Cell. Neurosci. 2021, 15, 771898. [Google Scholar] [CrossRef] [PubMed]
  7. Zhang, S.F.; Gao, J.; Liu, C.M. The Role of Non-Coding RNAs in Neurodevelopmental Disorders. Front. Genet. 2019, 10, 1033. [Google Scholar] [CrossRef] [PubMed]
  8. Lungu, G.; Stoica, G.; Ambrus, A. MicroRNA profiling and the role of microRNA-132 in neurodegeneration using a rat model. Neurosci. Lett. 2013, 553, 153–158. [Google Scholar] [CrossRef]
  9. Jimenez-Mateos, E.M.; Engel, T.; Merino-Serrais, P.; Fernaud-Espinosa, I.; Rodriguez-Alvarez, N.; Reynolds, J.; Reschke, C.R.; Conroy, R.M.; McKiernan, R.C.; deFelipe, J.; et al. Antagomirs targeting microRNA-134 increase hippocampal pyramidal neuron spine volume in vivo and protect against pilocarpine-induced status epilepticus. Brain Struct. Funct. 2015, 220, 2387–2399. [Google Scholar] [CrossRef]
  10. Ma, Z.; Li, C.Y.; Wang, L.J.; Xia, Y.; Feng, C.A.; Peng, Y.F.; Han, Y.B.; Fan, Y.; Ba, Y.C. MicroRNA-138 Regulates Spinal Cord Development by Activating the Shh in Fetal Rats. Pediatr. Neurosurg. 2022, 57, 407–421. [Google Scholar] [CrossRef]
  11. Dong, L.; Wang, M.; Gao, X.; Zheng, X.; Zhang, Y.; Sun, L.; Zhao, N.; Ding, C.; Ma, Z.; Wang, Y. miR-9-5p promotes myogenic differentiation via the Dlx3/Myf5 axis. PeerJ 2022, 10, e13360. [Google Scholar] [CrossRef] [PubMed]
  12. Bartel, D.P. MicroRNAs: Target recognition and regulatory functions. Cell 2009, 136, 215–233. [Google Scholar] [CrossRef] [PubMed]
  13. Ghibaudi, M.; Boido, M.; Vercelli, A. Functional integration of complex miRNA networks in central and peripheral lesion and axonal regeneration. Prog. Neurobiol. 2017, 158, 69–93. [Google Scholar] [CrossRef]
  14. Shi, Y.; Zhang, B.; Zhu, J.; Huang, W.; Han, B.; Wang, Q.; Qi, C.; Wang, M.; Liu, F. miR-106b-5p Inhibits IRF1/IFN-β Signaling to Promote M2 Macrophage Polarization of Glioblastoma. OncoTargets Ther. 2020, 13, 7479–7492. [Google Scholar] [CrossRef]
  15. Shi, Z.; Piccus, Z.; Zhang, X.; Yang, H.; Jarrell, H.; Ding, Y.; Teng, Z.; Tchernichovski, O.; Li, X. miR-9 regulates basal ganglia-dependent developmental vocal learning and adult vocal performance in songbirds. Elife 2018, 7, e29087. [Google Scholar] [CrossRef] [PubMed]
  16. Bludau, A.; Schwartz, U.; Zeitler, D.M.; Royer, M.; Meister, G.; Neumann, I.D.; Menon, R. Functional involvement of septal miR-132 in extinction and oxytocin-mediated reversal of social fear. Mol. Psychiatry 2023. [Google Scholar] [CrossRef]
  17. Xiao, X.; Ai, R.; Tian, Y.; Mi, N.; Cheng, L.; Qian, N.; Zhu, X. Study on the Mechanism of Action of MicroRNA-140-5p in the Treatment of Autism by Regulating the Nuclear Factor Kappa B Signaling Pathway. Indian J. Pharm. Sci. 2021, 83, 133–139. [Google Scholar] [CrossRef]
  18. Wu, X.; Li, W.; Zheng, Y. Recent Progress on Relevant microRNAs in Autism Spectrum Disorders. Int. J. Mol. Sci. 2020, 21, 5904. [Google Scholar] [CrossRef]
  19. Jing, X.L.; Xing, A.Y.; Bai, H.; Wu, L. miRNA-148b-3p Influences Glucose Metabolism of Offspring with Maternal Cholestasis by Regulating GLUT1 Expression in Placental Trophoblast Cells. Sichuan Da Xue Xue Bao Yi Xue Ban J. Sichuan Univ. (Med. Sci. Ed.) 2019, 50, 328–333. [Google Scholar]
  20. Ni, Y.; Yang, Y.; Ran, J.; Zhang, L.; Yao, M.; Liu, Z.; Zhang, L. miR-15a-5p inhibits metastasis and lipid metabolism by suppressing histone acetylation in lung cancer. Free. Radic. Biol. Med. 2020, 161, 150–162. [Google Scholar] [CrossRef]
  21. Hosokawa, R.; Yoshino, Y.; Funahashi, Y.; Horiuchi, F.; Iga, J.I.; Ueno, S.I. MiR-15b-5p Expression in the Peripheral Blood: A Potential Diagnostic Biomarker of Autism Spectrum Disorder. Brain Sci. 2022, 13, 27. [Google Scholar] [CrossRef]
  22. Tonacci, A.; Bagnato, G.; Pandolfo, G.; Billeci, L.; Sansone, F.; Conte, R.; Gangemi, S. MicroRNA Cross-Involvement in Autism Spectrum Disorders and Atopic Dermatitis: A Literature Review. J. Clin. Med. 2019, 8, 88. [Google Scholar] [CrossRef] [PubMed]
  23. Shen, L.; Lin, Y.; Sun, Z.; Yuan, X.; Chen, L.; Shen, B. Knowledge-Guided Bioinformatics Model for Identifying Autism Spectrum Disorder Diagnostic MicroRNA Biomarkers. Sci. Rep. 2016, 6, 39663. [Google Scholar] [CrossRef] [PubMed]
  24. Cava, C.; Manna, I.; Gambardella, A.; Bertoli, G.; Castiglioni, I. Potential Role of miRNAs as Theranostic Biomarkers of Epilepsy. Mol. Ther. Nucleic Acids 2018, 13, 275–290. [Google Scholar] [CrossRef]
  25. Wu, H.; Pula, T.; Tews, D.; Amri, E.Z.; Debatin, K.M.; Wabitsch, M.; Fischer-Posovszky, P.; Roos, J. microRNA-27a-3p but Not -5p Is a Crucial Mediator of Human Adipogenesis. Cells 2021, 10, 3205. [Google Scholar] [CrossRef] [PubMed]
  26. Vasu, M.; Anitha, A.; Thanseem, I.; Suzuki, K.; Yamada, K.; Takahashi, T.; Wakuda, T.; Iwata, K.; Tsujii, M.; Sugiyama, T.; et al. Serum microRNA profiles in children with autism. Mol. Autism 2014, 5, 40. [Google Scholar] [CrossRef]
  27. Wang, Z.; Lu, T.; Li, X.; Jiang, M.; Jia, M.; Liu, J.; Zhang, D.; Li, J.; Wang, L. Altered Expression of Brain-specific Autism-Associated miRNAs in the Han Chinese Population. Front. Genet. 2022, 13, 865881. [Google Scholar] [CrossRef]
  28. Wang, Y.; Zhang, K.; Yuan, X.; Xu, N.; Zhao, S.; Hou, L.; Yang, L.; Zhang, N. miR-431-5p regulates cell proliferation and apoptosis in fibroblast-like synoviocytes in rheumatoid arthritis by targeting XIAP. Arthritis Res. Ther. 2020, 22, 231. [Google Scholar] [CrossRef]
  29. Honorato-Mauer, J.; Xavier, G.; Ota, V.K.; Chehimi, S.N.; Mafra, F.; Cuóco, C.; Ito, L.T.; Ormond, R.; Asprino, P.F.; Oliveira, A.; et al. Alterations in microRNA of extracellular vesicles associated with major depression, attention-deficit/hyperactivity and anxiety disorders in adolescents. Transl. Psychiatry 2023, 13, 47. [Google Scholar] [CrossRef]
  30. Fujitani, M.; Zhang, S.; Fujiki, R.; Fujihara, Y.; Yamashita, T. A chromosome 16p13.11 microduplication causes hyperactivity through dysregulation of miR-484/protocadherin-19 signaling. Mol. Psychiatry 2017, 22, 364–374. [Google Scholar] [CrossRef]
  31. Li, J.; Xu, X.; Liu, J.; Zhang, S.; Tan, X.; Li, Z.; Zhang, J.; Wang, Z. Decoding microRNAs in autism spectrum disorder. Mol. Ther. Nucleic Acids 2022, 30, 535–546. [Google Scholar] [CrossRef]
  32. Nuzziello, N.; Craig, F.; Simone, M.; Consiglio, A.; Licciulli, F.; Margari, L.; Grillo, G.; Liuni, S.; Liguori, M. Integrated Analysis of microRNA and mRNA Expression Profiles: An Attempt to Disentangle the Complex Interaction Network in Attention Deficit Hyperactivity Disorder. Brain Sci. 2019, 9, 288. [Google Scholar] [CrossRef]
  33. Kim, J.Y.; Kim, W.; Lee, K.H. The role of microRNAs in the molecular link between circadian rhythm and autism spectrum disorder. Anim. Cells Syst. 2023, 27, 38–52. [Google Scholar] [CrossRef]
  34. Zhou, M.; Hou, Y.; Wu, J.; Li, G.; Cao, P.; Chen, W.; Hu, L.; Gan, D. miR-93-5p promotes insulin resistance to regulate type 2 diabetes progression in HepG2 cells by targeting HGF. Mol. Med. Rep. 2021, 23, 329. [Google Scholar] [CrossRef] [PubMed]
  35. Mansur, F.A.; Raman, N.F.A.; Rahman, H.A.; Mohd Manzor, N.F. Mechanism of Autism Spectrum Disorder and The Involvement of microRNA. Malays. J. Sci. Health Technol. 2021, 6. [Google Scholar] [CrossRef]
  36. Popa, N.; Boyer, F.; Jaouen, F.; Belzeaux, R.; Gascon, E. Social Isolation and Enrichment Induce Unique miRNA Signatures in the Prefrontal Cortex and Behavioral Changes in Mice. iScience 2020, 23, 101790. [Google Scholar] [CrossRef]
  37. Wang, C.; Liu, S.; Li, J.; Cheng, Y.; Wang, Z.; Feng, T.; Lu, G.; Wang, S.; Song, J.; Xia, P.; et al. Biological Functions of Let-7e-5p in Promoting the Differentiation of MC3T3-E1 Cells. Front. Cell Dev. Biol. 2021, 9, 671170. [Google Scholar] [CrossRef] [PubMed]
  38. He, B.; Zhang, K.; Han, X.; Su, C.; Zhao, J.; Wang, G.; Wang, G.; Zhang, L.; Hu, W. Extracellular Vesicle-Derived miR-105-5p Promotes Malignant Phenotypes of Esophageal Squamous Cell Carcinoma by Targeting SPARCL1 via FAK/AKT Signaling Pathway. Front. Genet. 2022, 13, 819699. [Google Scholar] [CrossRef]
  39. Xiang, W.; He, J.; Huang, C.; Chen, L.; Tao, D.; Wu, X.; Wang, M.; Luo, G.; Xiao, X.; Zeng, F.; et al. miR-106b-5p targets tumor suppressor gene SETD2 to inactive its function in clear cell renal cell carcinoma. Oncotarget 2015, 6, 4066–4079. [Google Scholar] [CrossRef]
  40. Beveridge, N.J.; Gardiner, E.; Carroll, A.P.; Tooney, P.A.; Cairns, M.J. Schizophrenia is associated with an increase in cortical microRNA biogenesis. Mol. Psychiatry 2010, 15, 1176–1189. [Google Scholar] [CrossRef]
  41. Pan, J.; Qu, M.; Li, Y.; Wang, L.; Zhang, L.; Wang, Y.; Tang, Y.; Tian, H.L.; Zhang, Z.; Yang, G.Y. MicroRNA-126-3p/-5p Overexpression Attenuates Blood-Brain Barrier Disruption in a Mouse Model of Middle Cerebral Artery Occlusion. Stroke 2020, 51, 619–627. [Google Scholar] [CrossRef] [PubMed]
  42. Budi, H.S.; Younus, L.A.; Lafta, M.H.; Parveen, S.; Mohammad, H.J.; Al-Qaim, Z.H.; Jawad, M.A.; Parra, R.M.R.; Mustafa, Y.F.; Alhachami, F.R.; et al. The role of miR-128 in cancer development, prevention, drug resistance, and immunotherapy. Front. Oncol. 2023, 12, 1067974. [Google Scholar] [CrossRef] [PubMed]
  43. Liu, L.; Wang, P.; Wang, Y.S.; Zhang, Y.N.; Li, C.; Yang, Z.Y.; Liu, Z.H.; Zhan, T.Z.; Xu, J.; Xia, C.M. MiR-130a-3p Alleviates Liver Fibrosis by Suppressing HSCs Activation and Skewing Macrophage to Ly6Clo Phenotype. Front. Immunol. 2021, 12, 696069. [Google Scholar] [CrossRef] [PubMed]
  44. Daswani, R.; Gilardi, C.; Soutschek, M.; Nanda, P.; Weiss, K.; Bicker, S.; Fiore, R.; Dieterich, C.; Germain, P.L.; Winterer, J.; et al. MicroRNA-138 controls hippocampal interneuron function and short-term memory in mice. Elife 2022, 11, e74056. [Google Scholar] [CrossRef] [PubMed]
  45. Wang, Y.; Wang, D.; Xie, G.; Yin, Y.; Zhao, E.; Tao, K.; Li, R. MicroRNA-152 regulates immune response via targeting B7-H1 in gastric carcinoma. Oncotarget 2017, 8, 28125–28134. [Google Scholar] [CrossRef] [PubMed]
  46. Weissman, R.; Diamond, E.L.; Haroche, J.; Durham, B.H.; Cohen, F.; Buthorn, J.; Amoura, Z.; Emile, J.F.; Mazor, R.D.; Shomron, N.; et al. MicroRNA-15a-5p acts as a tumor suppressor in histiocytosis by mediating CXCL10-ERK-LIN28a-let-7 axis. Leukemia 2022, 36, 1139–1149. [Google Scholar] [CrossRef] [PubMed]
  47. Eyileten, C.; Sharif, L.; Wicik, Z.; Jakubik, D.; Jarosz-Popek, J.; Soplinska, A.; Postula, M.; Czlonkowska, A.; Kaplon-Cieslicka, A.; Mirowska-Guzel, D. The Relation of the Brain-Derived Neurotrophic Factor with MicroRNAs in Neurodegenerative Diseases and Ischemic Stroke. Mol. Neurobiol. 2021, 58, 329–347. [Google Scholar] [CrossRef] [PubMed]
  48. Saba, R.; Störchel, P.H.; Aksoy-Aksel, A.; Kepura, F.; Lippi, G.; Plant, T.D.; Schratt, G.M. Dopamine-regulated microRNA MiR-181a controls GluA2 surface expression in hippocampal neurons. Mol. Cell. Biol. 2012, 32, 619–632. [Google Scholar] [CrossRef] [PubMed]
  49. Ding, H.; Yao, J.; Xie, H.; Wang, C.; Chen, J.; Wei, K.; Ji, Y.; Liu, L. MicroRNA-195-5p Downregulation Inhibits Endothelial Mesenchymal Transition and Myocardial Fibrosis in Diabetic Cardiomyopathy by Targeting Smad7 and Inhibiting Transforming Growth Factor Beta 1-Smads-Snail Pathway. Front. Physiol. 2021, 12, 709123. [Google Scholar] [CrossRef]
  50. Arzhanov, I.; Sintakova, K.; Romanyuk, N. The Role of miR-20 in Health and Disease of the Central Nervous System. Cells 2022, 11, 1525. [Google Scholar] [CrossRef]
  51. Guan, C.; Luan, L.; Li, J.; Yang, L. MiR-212-3p improves rat functional recovery and inhibits neurocyte apoptosis in spinal cord injury models via PTEN downregulation-mediated activation of AKT/mTOR pathway. Brain Res. 2021, 1768, 147576. [Google Scholar] [CrossRef]
  52. Oladejo, A.O.; Li, Y.; Imam, B.H.; Ma, X.; Shen, W.; Wu, X.; Jiang, W.; Yang, J.; Lv, Y.; Ding, X.; et al. MicroRNA miR-24-3p Mediates the Negative Regulation of Lipopolysaccharide-Induced Endometrial Inflammatory Response by Targeting TNF Receptor-Associated Factor 6 (TRAF6). J. Inflamm. Res. 2022, 15, 807–825. [Google Scholar] [CrossRef] [PubMed]
  53. Xiao, Y.; Zheng, S.; Duan, N.; Li, X.; Wen, J. MicroRNA-26b-5p alleviates cerebral ischemia-reperfusion injury in rats via inhibiting the N-myc/PTEN axis by downregulating KLF10 expression. Hum. Exp. Toxicol. 2021, 40, 1250–1262. [Google Scholar] [CrossRef] [PubMed]
  54. Harati, R.; Hammad, S.; Tlili, A.; Mahfood, M.; Mabondzo, A.; Hamoudi, R. miR-27a-3p regulates expression of intercellular junctions at the brain endothelium and controls the endothelial barrier permeability. PLoS ONE 2022, 17, e0262152. [Google Scholar] [CrossRef]
  55. Camkurt, M.A. Blood microRNA dysregulation in schizophrenia. Psychiatry Clin. Psychopharmacol. 2015, 25, S112. [Google Scholar]
  56. Wang, J.; Zhu, M.; Ye, L.; Chen, C.; She, J.; Song, Y. MiR-29b-3p promotes particulate matter-induced inflammatory responses by regulating the C1QTNF6/AMPK pathway. Aging 2020, 12, 1141–1158. [Google Scholar] [CrossRef] [PubMed]
  57. Hu, Z.; Cai, M.; Zhang, Y.; Tao, L.; Guo, R. miR-29c-3p inhibits autophagy and cisplatin resistance in ovarian cancer by regulating FOXP1/ATG14 pathway. Cell Cycle 2020, 19, 193–206. [Google Scholar] [CrossRef]
  58. Yuan, J.N.; Hong, Y.; Ma, Z.L.; Pang, R.P.; Lei, Q.Q.; Lv, X.F.; Zhou, J.G.; Huang, H.; Zhang, T.T. MiR-302a Limits Vascular Inflammation by Suppressing Nuclear Factor-κ B Pathway in Endothelial Cells. Front. Cell Dev. Biol. 2021, 9, 682574. [Google Scholar] [CrossRef]
  59. Yasukawa, K.; Kinoshita, D.; Yaku, K.; Nakagawa, T.; Koshiba, T. The microRNAs miR-302b and miR-372 regulate mitochondrial metabolism via the SLC25A12 transporter, which controls MAVS-mediated antiviral innate immunity. J. Biol. Chem. 2020, 295, 444–457. [Google Scholar] [CrossRef]
  60. Li, J.; Salvador, A.M.; Li, G.; Valkov, N.; Ziegler, O.; Yeri, A.; Xiao, C.Y.; Meechoovet, B.; Alsop, E.; Rodosthenous, R.S.; et al. Mir-30d Regulates Cardiac Remodeling by Intracellular and Paracrine Signaling. Circ. Res. 2021, 128, e1–e23. [Google Scholar] [CrossRef]
  61. Zhang, R.; Shi, H.; Ren, F.; Feng, W.; Cao, Y.; Li, G.; Liu, Z.; Ji, P.; Zhang, M. MicroRNA-338-3p suppresses ovarian cancer cells growth and metastasis: Implication of Wnt/catenin beta and MEK/ERK signaling pathways. J. Exp. Clin. Cancer Res. 2019, 38, 494. [Google Scholar] [CrossRef]
  62. Fletcher, C.E.; Deng, L.; Orafidiya, F.; Yuan, W.; Lorentzen, M.P.G.S.; Cyran, O.W.; Varela-Carver, A.; Constantin, T.A.; Leach, D.A.; Dobbs, F.M.; et al. A non-coding RNA balancing act: miR-346-induced DNA damage is limited by the long non-coding RNA NORAD in prostate cancer. Mol. Cancer 2022, 21, 82. [Google Scholar] [CrossRef] [PubMed]
  63. Zhao, C.; Zhou, Y.; Ran, Q.; Yao, Y.; Zhang, H.; Ju, J.; Yang, T.; Zhang, W.; Yu, X.; He, S. MicroRNA-381-3p Functions as a Dual Suppressor of Apoptosis and Necroptosis and Promotes Proliferation of Renal Cancer Cells. Front. Cell Dev. Biol. 2020, 8, 290. [Google Scholar] [CrossRef]
  64. Wan, L.; Zhu, L.; Xu, J.; Lu, B.; Yang, Y.; Liu, F.; Wang, Z. MicroRNA-409-3p functions as a tumor suppressor in human lung adenocarcinoma by targeting c-Met. Cell. Physiol. Biochem. 2014, 34, 1273–1290. [Google Scholar] [CrossRef] [PubMed]
  65. Deng, X.; Zuo, M.; Pei, Z.; Xie, Y.; Yang, Z.; Zhang, Z.; Jiang, M.; Kuang, D. MicroRNA-455-5p Contributes to Cholangiocarcinoma Growth and Mediates Galangin’s Anti-Tumor Effects. J. Cancer 2021, 12, 4710–4721. [Google Scholar] [CrossRef] [PubMed]
  66. Zhao, Z.; Shuang, T.; Gao, Y.; Lu, F.; Zhang, J.; He, W.; Qu, L.; Chen, B.; Hao, Q. Targeted delivery of exosomal miR-484 reprograms tumor vasculature for chemotherapy sensitization. Cancer Lett. 2022, 530, 45–58. [Google Scholar] [CrossRef] [PubMed]
  67. Gao, F.; Wu, H.; Wang, R.; Guo, Y.; Zhang, Z.; Wang, T.; Zhang, G.; Liu, C.; Liu, J. MicroRNA-485-5p suppresses the proliferation, migration and invasion of small cell lung cancer cells by targeting flotillin-2. Bioengineered 2019, 10, 1–12. [Google Scholar] [CrossRef]
  68. Ma, M.; He, M.; Jiang, Q.; Yan, Y.; Guan, S.; Zhang, J.; Yu, Z.; Chen, Q.; Sun, M.; Yao, W.; et al. MiR-487a Promotes TGF-β1-induced EMT, the Migration and Invasion of Breast Cancer Cells by Directly Targeting MAGI2. Int. J. Biol. Sci. 2016, 12, 397–408. [Google Scholar] [CrossRef] [PubMed]
  69. Gao, W.; Zheng, W.; Sun, Y.; Xu, T. microRNA-489 negatively modulates RIG-I signaling pathway via targeting TRAF6 in miiuy croaker after poly(I:C) stimulation. Fish Shellfish. Immunol. 2021, 113, 61–68. [Google Scholar] [CrossRef]
  70. Ravegnini, G.; De Leo, A.; Coada, C.; Gorini, F.; de Biase, D.; Ceccarelli, C.; Dondi, G.; Tesei, M.; De Crescenzo, E.; Santini, D.; et al. Identification of miR-499a-5p as a Potential Novel Biomarker for Risk Stratification in Endometrial Cancer. Front. Oncol. 2021, 11, 757678. [Google Scholar] [CrossRef]
  71. Chen, S.; Zhou, H.; Zhang, B.; Hu, Q. Exosomal miR-512-3p derived from mesenchymal stem cells inhibits oxidized low-density lipoprotein-induced vascular endothelial cells dysfunction via regulating Keap1. J. Biochem. Mol. Toxicol. 2021, 35, 1–11. [Google Scholar] [CrossRef]
  72. Liu, M.; Wang, Y.; Lu, H.; Wang, H.; Shi, X.; Shao, X.; Li, Y.X.; Zhao, Y.; Wang, Y.L. miR-518b Enhances Human Trophoblast Cell Proliferation Through Targeting Rap1b and Activating Ras-MAPK Signal. Front. Endocrinol. 2018, 9, 100. [Google Scholar] [CrossRef] [PubMed]
  73. Augenlicht, A.; Saiselet, M.; Decaussin-Petrucci, M.; Andry, G.; Dumont, J.E.; Maenhaut, C. MiR-7-5p inhibits thyroid cell proliferation by targeting the EGFR/MAPK and IRS2/PI3K signaling pathways. Oncotarget 2021, 12, 1587–1599. [Google Scholar] [CrossRef] [PubMed]
  74. Zhen, J.; Zhang, H.; Dong, H.; Tong, X. miR-9-3p inhibits glioma cell proliferation and apoptosis by directly targeting FOXG1. Oncol. Lett. 2020, 20, 2007–2015. [Google Scholar] [CrossRef]
  75. Huang, J.; Zhou, Q.; Chen, C.; Chen, L.; Xu, X.; Wang, Y.; An, Z.; Lin, C.; Han, H. MicroRNA miR-92a-3p regulates breast cancer cell proliferation and metastasis via regulating B-cell translocation gene 2 (BTG2). Bioengineered 2021, 12, 2033–2044. [Google Scholar]
  76. Bhattacharyya, P.; Biswas, A.; Biswas, S.C. Brain-enriched miR-128: Reduced in exosomes from Parkinson’s patient plasma, improves synaptic integrity, and prevents 6-OHDA mediated neuronal apoptosis. Front. Cell. Neurosci. 2023, 16, 1037903. [Google Scholar] [CrossRef]
  77. Koh, H.S.; Lee, S.; Lee, H.J.; Min, J.W.; Iwatsubo, T.; Teunissen, C.E.; Cho, H.J.; Ryu, J.H. Targeting MicroRNA-485-3p Blocks Alzheimer’s Disease Progression. Int. J. Mol. Sci. 2021, 22, 13136. [Google Scholar] [CrossRef]
  78. Yoon, S.; Han, E.; Choi, Y.C.; Kee, H.; Jeong, Y.; Yoon, J.; Baek, K. Inhibition of cell proliferation and migration by miR-509-3p that targets CDK2, Rac1, and PIK3C2A. Mol. Cells 2014, 37, 314–321. [Google Scholar] [CrossRef]
  79. Regis, S.; Dondero, A.; Spaggiari, G.M.; Serra, M.; Caliendo, F.; Bottino, C.; Castriconi, R. miR-24-3p down-regulates the expression of the apoptotic factors FasL and BIM in human natural killer cells. Cell. Signal. 2022, 98, 110415. [Google Scholar] [CrossRef]
  80. Li, L.; Zhang, X.; Lin, Y.; Ren, X.; Xie, T.; Lin, J.; Wu, S.; Ye, Q. Let-7b-5p inhibits breast cancer cell growth and metastasis via repression of hexokinase 2-mediated aerobic glycolysis. Cell Death Discov. 2023, 9, 114. [Google Scholar] [CrossRef]
  81. Liu, J.H.; Li, C.; Cao, L.; Zhang, C.H.; Zhang, Z.H. Cucurbitacin B regulates lung cancer cell proliferation and apoptosis via inhibiting the IL-6/STAT3 pathway through the lncRNA XIST/miR-let-7c axis. Pharm. Biol. 2022, 60, 154–162. [Google Scholar] [CrossRef] [PubMed]
  82. Chen, Y.N.; Ren, C.C.; Yang, L.; Nai, M.M.; Xu, Y.M.; Zhang, F.; Liu, Y. MicroRNA let 7d 5p rescues ovarian cancer cell apoptosis and restores chemosensitivity by regulating the p53 signaling pathway via HMGA1. Int. J. Oncol. 2019, 54, 1771–1784. [Google Scholar] [CrossRef] [PubMed]
  83. Chen, W.; Lin, G.; Yao, Y.; Chen, J.; Shui, H.; Yang, Q.; Wang, X.; Weng, X.; Sun, L.; Chen, F.; et al. MicroRNA hsa-let-7e-5p as a potential prognosis marker for rectal carcinoma with liver metastases. Oncol. Lett. 2018, 15, 6913–6924. [Google Scholar] [CrossRef]
  84. Zhang, K.; Yang, R.; Chen, J.; Qi, E.; Zhou, S.; Wang, Y.; Fu, Q.; Chen, R.; Fang, X. Let-7i-5p Regulation of Cell Morphology and Migration Through Distinct Signaling Pathways in Normal and Pathogenic Urethral Fibroblasts. Front. Bioeng. Biotechnol. 2020, 8, 428. [Google Scholar] [CrossRef] [PubMed]
  85. Xue, P.; Huang, S.; Han, X.; Zhang, C.; Yang, L.; Xiao, W.; Fu, J.; Li, H.; Zhou, Y. Exosomal miR-101-3p and miR-423-5p inhibit medulloblastoma tumorigenesis through targeting FOXP4 and EZH2. Cell Death Differ. 2022, 29, 82–95. [Google Scholar] [CrossRef] [PubMed]
  86. Sagar, S.K. miR-106b as an emerging therapeutic target in cancer. Genes Dis. 2021, 9, 889–899. [Google Scholar] [CrossRef]
  87. Ahonen, M.A.; Haridas, P.A.N.; Mysore, R.; Wabitsch, M.; Fischer-Posovszky, P.; Olkkonen, V.M. miR-107 inhibits CDK6 expression, differentiation, and lipid storage in human adipocytes. Mol. Cell. Endocrinol. 2019, 479, 110–116. [Google Scholar] [CrossRef] [PubMed]
  88. Qu, C.; Liu, X.; Guo, Y.; Fo, Y.; Chen, X.; Zhou, J.; Yang, B. MiR-128-3p inhibits vascular smooth muscle cell proliferation and migration by repressing FOXO4/MMP9 signaling pathway. Mol. Med. 2020, 26, 116. [Google Scholar] [CrossRef]
  89. Khalili, N.; Nouri-Vaskeh, M.; Hasanpour Segherlou, Z.; Baghbanzadeh, A.; Halimi, M.; Rezaee, H.; Baradaran, B. Diagnos-tic, prognostic, and therapeutic significance of miR-139-5p in cancers. Life Sci. 2020, 256, 117865. [Google Scholar] [CrossRef]
  90. Wang, Z.; Liu, F.; Wei, M.; Qiu, Y.; Ma, C.; Shen, L.; Huang, Y. Chronic constriction injury-induced microRNA-146a-5p alle-viates neuropathic pain through suppression of IRAK1/TRAF6 signaling pathway. J. Neuroinflamm. 2018, 15, 179. [Google Scholar] [CrossRef]
  91. Maurel, O.M.; Torrisi, S.A.; Barbagallo, C.; Purrello, M.; Salomone, S.; Drago, F.; Ragusa, M.; Leggio, G.M. Dysregulation of miR-15a-5p, miR-497a-5p and miR-511-5p Is Associated with Modulation of BDNF and FKBP5 in Brain Areas of PTSD-Related Susceptible and Resilient Mice. Int. J. Mol. Sci. 2021, 22, 5157. [Google Scholar] [CrossRef] [PubMed]
  92. Liu, F.; Di Wang, X. miR-150-5p represses TP53 tumor suppressor gene to promote proliferation of colon adenocarcinoma. Sci. Rep. 2019, 9, 6740. [Google Scholar] [CrossRef] [PubMed]
  93. Li, B.; Xia, Y.; Lv, J.; Wang, W.; Xuan, Z.; Chen, C.; Jiang, T.; Fang, L.; Wang, L.; Li, Z.; et al. miR-151a-3p-rich small extracel-lular vesicles derived from gastric cancer accelerate liver metastasis via initiating a hepatic stemness-enhancing niche. Oncogene 2021, 40, 6180–6194. [Google Scholar] [CrossRef] [PubMed]
  94. Su, Y.; Yuan, J.; Zhang, F.; Lei, Q.; Zhang, T.; Li, K.; Guo, J.; Hong, Y.; Bu, G.; Lv, X.; et al. MicroRNA-181a-5p and microRNA-181a-3p cooperatively restrict vascular inflammation and atherosclerosis. Cell Death Dis. 2019, 10, 365. [Google Scholar] [CrossRef] [PubMed]
  95. Yu, Y.; Cai, W.; Xu, Y.; Zuo, W. Down-regulation of miR-19b-3p enhances IGF-1 expression to induce osteoblast differentia-tion and improve osteoporosis. Cell. Mol. Biol. 2022, 68, 160–168. [Google Scholar] [CrossRef] [PubMed]
  96. Han, J.; Hu, J.; Sun, F.; Bian, H.; Tang, B.; Fang, X. MicroRNA-20a-5p suppresses tumor angiogenesis of non-small cell lung cancer through RRM2-mediated PI3K/Akt signaling pathway. Mol. Cell. Biochem. 2021, 476, 689–698. [Google Scholar] [CrossRef] [PubMed]
  97. Gorur, A.; Bayraktar, R.; Ivan, C.; Mokhlis, H.A.; Bayraktar, E.; Kahraman, N.; Karakas, D.; Karamil, S.; Kabil, N.N.; Kan-likilicer, P.; et al. ncRNA therapy with miRNA-22-3p suppresses the growth of triple-negative breast cancer. Mol. Ther. Nucleic Acids 2021, 23, 930–943. [Google Scholar] [CrossRef]
  98. Zhang, H.; Liang, J.; Chen, N. The Potential Role of miRNA-Regulated Autophagy in Alzheimer’s Disease. Int. J. Mol. Sci. 2022, 23, 7789. [Google Scholar] [CrossRef]
  99. Zhou, A.; Pan, H.; Sun, D.; Xu, H.; Zhang, C.; Chen, X.; Li, L.; Wang, T. miR-26b-5p Inhibits the Proliferation, Migration and Invasion of Human Papillary Thyroid Cancer in a β-Catenin-Dependent Manner. OncoTargets Ther. 2020, 13, 1593–1603. [Google Scholar] [CrossRef]
  100. Duwe, L.; Munoz-Garrido, P.; Lewinska, M.; Lafuente-Barquero, J.; Satriano, L.; Høgdall, D.; Taranta, A.; Nielsen, B.S.; Ghazal, A.; Matter, M.S.; et al. MicroRNA-27a-3p targets FoxO signalling to induce tumour-like phenotypes in bile duct cells. J. Hepatol. 2023, 78, 364–375. [Google Scholar] [CrossRef]
  101. Lv, Y.; Yang, H.; Ma, X.; Wu, G. Strand-specific miR-28-3p and miR-28-5p have differential effects on nasopharyngeal cancer cells proliferation, apoptosis, migration and invasion. Cancer Cell Int. 2019, 19, 187. [Google Scholar] [CrossRef] [PubMed]
  102. Sun, H.; Zhong, D.; Wang, C.; Sun, Y.; Zhao, J.; Li, G. MiR-298 Exacerbates Ischemia/Reperfusion Injury Following Ischemic Stroke by Targeting Act1. Cell. Physiol. Biochem. 2018, 48, 528–539. [Google Scholar] [CrossRef] [PubMed]
  103. Zhang, K.; Han, X.; Hu, W.; Su, C.; He, B. miR-29a-3p inhibits the malignant characteristics of non-small cell lung cancer cells by reducing the activity of the Wnt/β-catenin signaling pathway. Oncol. Lett. 2022, 24, 379. [Google Scholar] [CrossRef] [PubMed]
  104. Ma, X.; Yun, H.J.; Elkin, K.; Guo, Y.; Ding, Y.; Li, G. MicroRNA-29b Suppresses Inflammation and Protects Blood-Brain Barrier Integrity in Ischemic Stroke. Mediat. Inflamm. 2022, 2022, 1755416. [Google Scholar] [CrossRef] [PubMed]
  105. Taghehchian, N.; Lotfi, M.; Zangouei, A.S.; Akhlaghipour, I.; Moghbeli, M. MicroRNAs as the critical regulators of Forkhead box protein family during gynecological and breast tumor progression and metastasis. Eur. J. Med. Res. 2023, 28, 330. [Google Scholar] [CrossRef]
  106. Chuang, Y.-T.; Tang, J.-Y.; Shiau, J.-P.; Yen, C.-Y.; Chang, F.-R.; Yang, K.-H.; Hou, M.-F.; Farooqi, A.A.; Chang, H.-W. Modu-lating Effects of Cancer-Derived Exosomal miRNAs and Exosomal Processing by Natural Products. Cancers 2023, 15, 318. [Google Scholar] [CrossRef]
  107. Du, H.; Yin, Z.; Zhao, Y.; Li, H.; Dai, B.; Fan, J.; He, M.; Nie, X.; Wang, C.Y.; Wang, D.W.; et al. miR-320a induces pancreatic β cells dysfunction in diabetes by inhibiting MafF. Mol. Ther. Nucleic Acids 2021, 26, 444–457. [Google Scholar] [CrossRef] [PubMed]
  108. Chen, K.C.; Hsi, E.; Hu, C.Y.; Chou, W.W.; Liang, C.L.; Juo, S.H. MicroRNA-328 may influence myopia development by me-diating the PAX6 gene. Investig. Ophthalmol. Vis. Sci. 2012, 53, 2732–2739. [Google Scholar] [CrossRef] [PubMed]
  109. Wang, Q.; Cai, J.; Cai, X.H.; Chen, L. miR-346 regulates osteogenic differentiation of human bone marrow-derived mesen-chymal stem cells by targeting the Wnt/β-catenin pathway. PLoS ONE 2013, 8, e72266. [Google Scholar]
  110. Zhang, L.; Liao, Y.; Tang, L. MicroRNA-34 family: A potential tumor suppressor and therapeutic candidate in cancer. J. Exp. Clin. Cancer Res. 2019, 38, 53. [Google Scholar] [CrossRef]
  111. Li, H.; Xue, Y.; Ma, J.; Shao, L.; Wang, D.; Zheng, J.; Liu, X.; Yang, C.; He, Q.; Ruan, X.; et al. SNHG1 promotes malignant bio-logical behaviors of glioma cells via microRNA-154-5p/miR-376b-3p- FOXP2- KDM5B participating positive feedback loop. J. Exp. Clin. Cancer Res. 2019, 38, 59. [Google Scholar] [CrossRef] [PubMed]
  112. Jiang, H.; Su, Z.; Hu, W.; Yuan, X.; Yu, T.; Yang, J.; Xiao, X.; Zheng, S.; Lin, B. miR-433 Inhibits Glioblastoma Progression by Suppressing the PI3K/Akt Signaling Pathway Through Direct Targeting of ERBB4. OMICS J. Integr. Biol. 2023, 27, 215–226. [Google Scholar] [CrossRef] [PubMed]
  113. Gao, J.; Dai, C.; Yu, X.; Yin, X.B.; Zhou, F. microRNA-485-5p inhibits the progression of hepatocellular carcinoma through blocking the WBP2/Wnt signaling pathway. Cell. Signal. 2020, 66, 109466. [Google Scholar] [CrossRef] [PubMed]
  114. Yang, Y.; Li, H.; He, Z.; Xie, D.; Ni, J.; Lin, X. MicroRNA-488-3p inhibits proliferation and induces apoptosis by targeting ZBTB2 in esophageal squamous cell carcinoma. J. Cell. Biochem. 2019, 120, 18702–18713. [Google Scholar] [CrossRef] [PubMed]
  115. Yang, L.; Xu, X.; Chen, Z.; Zhang, Y.; Chen, H.; Wang, X. miR-511-3p promotes hepatic sinusoidal obstruction syndrome by activating hedgehog pathway via targeting Ptch1. Am. J. Physiol.-Gastrointest. Liver Physiol. 2021, 321, G344–G354. [Google Scholar] [CrossRef] [PubMed]
  116. Li, X.; He, L.; Yue, Q.; Lu, J.; Kang, N.; Xu, X.; Wang, H.; Zhang, H. MiR-9-5p promotes MSC migration by activating β-catenin signaling pathway. Am. J. Physiol.-Cell Physiol. 2017, 313, C80–C93. [Google Scholar] [CrossRef] [PubMed]
  117. Yang, B.; Feng, X.; Liu, H.; Tong, R.; Wu, J.; Li, C.; Yu, H.; Chen, Y.; Cheng, Q.; Chen, J.; et al. High-metastatic cancer cells derived exosomal miR92a-3p promotes epithelial-mesenchymal transition and metastasis of low-metastatic cancer cells by regulating PTEN/Akt pathway in hepatocellular carcinoma. Oncogene 2020, 39, 6529–6543. [Google Scholar] [CrossRef] [PubMed]
  118. Yang, M.; Xiao, R.; Wang, X.; Xiong, Y.; Duan, Z.; Li, D.; Kan, Q. MiR-93-5p regulates tumorigenesis and tumor immunity by targeting PD-L1/CCND1 in breast cancer. Ann. Transl. Med. 2022, 10, 203. [Google Scholar] [CrossRef] [PubMed]
  119. Du, Y.; Shi, X.; Li, J.; Jia, Y. MicroRNA-98-5p inhibits human mesangial cell proliferation and TNF-α and IL-6 secretion by targeting BTB and CNC homology 1. Exp. Ther. Med. 2021, 22, 1436. [Google Scholar] [CrossRef]
  120. Pajares, M.J.; Alemany-Cosme, E.; Goñi, S.; Bandres, E.; Palanca-Ballester, C.; Sandoval, J. Epigenetic Regulation of mi-croRNAs in Cancer: Shortening the Distance from Bench to Bedside. Int. J. Mol. Sci. 2021, 22, 7350. [Google Scholar] [CrossRef]
  121. Geng, L.; Zhang, T.; Liu, W.; Chen, Y. Inhibition of miR-128 Abates Aβ-Mediated Cytotoxicity by Targeting PPAR-γ via NF-κB Inactivation in Primary Mouse Cortical Neurons and Neuro2a Cells. Yonsei Med. J. 2018, 59, 1096–1106. [Google Scholar] [CrossRef] [PubMed]
  122. Huang, P.; Wei, S.; Luo, M.; Tang, Z.; Lin, Q.; Wang, X.; Luo, M.; He, Y.; Wang, C.; Wei, D.; et al. MiR-139-5p has an antide-pressant-like effect by targeting phosphodiesterase 4D to activate the cAMP/PKA/CREB signaling pathway. Ann. Transl. Med. 2021, 9, 1594. [Google Scholar] [CrossRef]
  123. Liao, Z.; Zheng, R.; Shao, G. Mechanisms and application strategies of miRNA-146a regulating inflammation and fibrosis at molecular and cellular levels (Review). Int. J. Mol. Med. 2023, 51, 7. [Google Scholar] [CrossRef]
  124. Li, W.; Wu, L.; Sun, Q.; Yang, Q.; Xue, J.; Shi, M.; Tang, H.; Zhang, J.; Liu, Q. MicroRNA-191 blocking the translocation of GLUT4 is involved in arsenite-induced hepatic insulin resistance through inhibiting the IRS1/AKT pathway. Ecotoxicol. Environ. Saf. 2021, 215, 112130. [Google Scholar] [CrossRef]
  125. He, J.; Han, Z.; An, Z.; Li, Y.; Xie, X.; Zhou, J.; He, S.; Lv, Y.; He, M.; Qu, H.; et al. The miR-203a Regulatory Network Affects the Proliferation of Chronic Myeloid Leukemia K562 Cells. Front. Cell Dev. Biol. 2021, 9, 616711. [Google Scholar] [CrossRef]
  126. Zhu, X.A.; Gao, L.F.; Zhang, Z.G.; Xiang, D.K. Down-regulation of miR-320 exerts protective effects on myocardial I-R injury via facilitating Nrf2 expression. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 1730–1741. [Google Scholar] [PubMed]
  127. Cui, H.; Song, R.; Wu, J.; Wang, W.; Chen, X.; Yin, J. MicroRNA-337 regulates the PI3K/AKT and Wnt/β-catenin signaling pathways to inhibit hepatocellular carcinoma progression by targeting high-mobility group AT-hook 2. Am. J. Cancer Res. 2018, 8, 405–421. [Google Scholar]
  128. Tian, W.; Yang, X.; Yang, H.; Lv, M.; Sun, X.; Zhou, B. Correction: Exosomal miR-338-3p suppresses non-small-cell lung can-cer cells metastasis by inhibiting CHL1 through the MAPK signaling pathway. Cell Death Dis. 2022, 13, 473. [Google Scholar] [CrossRef] [PubMed]
  129. Gao, J.R.; Shi, M.M.; Jiang, H.; Zhu, X.L.; Wei, L.B.; Qin, X.J. MicroRNA-339-5p inhibits lipopolysaccharide-induced rat mesangial cells by regulating the Syk/Ras/c-Fos pathway. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2022, 395, 1075–1085. [Google Scholar] [CrossRef]
  130. Zhang, S.; Liu, L.; Lv, Z.; Li, Q.; Gong, W.; Wu, H. MicroRNA-342-3p Inhibits the Proliferation, Migration, and Invasion of Osteosarcoma Cells by Targeting Astrocyte-Elevated Gene-1 (AEG-1). Oncol. Res. 2017, 25, 1505–1515. [Google Scholar] [CrossRef]
  131. Li, Z.; Zhao, H.; Chu, S.; Liu, X.; Qu, X.; Li, J.; Liu, D.; Li, H. miR-124-3p promotes BMSC osteogenesis via suppressing the GSK-3β/β-catenin signaling pathway in diabetic osteoporosis rats. Vitr. Cell. Dev. Biol.-Anim. 2020, 56, 723–734. [Google Scholar] [CrossRef]
  132. Fang, H.; Li, H.F.; Pan, Q.; Jin, H.L.; Yang, M.; Wang, R.R.; Wang, Q.Y.; Zhang, J.P. MiR-132-3p Modulates MEKK3-Dependent NF-κB and p38/JNK Signaling Pathways to Alleviate Spinal Cord Ischemia-Reperfusion Injury by Hindering M1 Polariza-tion of Macrophages. Front. Cell Dev. Biol. 2021, 9, 570451. [Google Scholar] [CrossRef] [PubMed]
  133. Si, Y.J.; Ren, Q.H.; Bi, L. miR-135b-5p regulates human mesenchymal stem cell osteogenic differentiation by facilitating the Hippo signaling pathway. Int. J. Clin. Exp. Pathol. 2017, 10, 7767–7775. [Google Scholar] [PubMed]
  134. Deng, X.; Chu, X.; Wang, P.; Ma, X.; Wei, C.; Sun, C.; Yang, J.; Li, Y. MicroRNA-29a-3p Reduces TNFα-Induced Endothelial Dysfunction by Targeting Tumor Necrosis Factor Receptor 1. Mol. Ther. Nucleic Acids 2019, 18, 903–915. [Google Scholar] [CrossRef]
  135. Lian, H.; Zhong, X.S.; Xiao, Y.; Sun, Z.; Shen, Y.; Zhao, K.; Ma, X.; Li, Y.; Niu, Q.; Liu, M.; et al. Exosomal miR-29b of Gut Origin in Patients With Ulcerative Colitis Suppresses Heart Brain-Derived Neurotrophic Factor. Front. Mol. Biosci. 2022, 9, 759689. [Google Scholar] [CrossRef]
  136. Liu, J.; Zuo, X.; Han, J.; Dai, Q.; Xu, H.; Liu, Y.; Cui, S. MiR-9-5p inhibits mitochondrial damage and oxidative stress in AD cell models by targeting GSK-3β. Biosci. Biotechnol. Biochem. 2020, 84, 2273–2280. [Google Scholar] [CrossRef] [PubMed]
  137. Sim, S.E.; Lim, C.S.; Kim, J.I.; Seo, D.; Chun, H.; Yu, N.K.; Lee, J.; Kang, S.J.; Ko, H.G.; Choi, J.H.; et al. The Brain-Enriched MicroRNA miR-9-3p Regulates Synaptic Plasticity and Memory. J. Neurosci. 2016, 36, 8641–8652. [Google Scholar] [CrossRef]
  138. Lu, X.C.; Zheng, J.Y.; Tang, L.J.; Huang, B.S.; Li, K.; Tao, Y.; Yu, W.; Zhu, R.L.; Li, S.; Li, L.X. MiR-133b Promotes neurite out-growth by targeting RhoA expression. Cell. Physiol. Biochem. 2015, 35, 246–258. [Google Scholar] [CrossRef]
  139. Xu, C.; Bai, Q.; Wang, C.; Meng, Q.; Gu, Y.; Wang, Q.; Xu, W.; Han, Y.; Qin, Y.; Jia, S.; et al. miR-433 Inhibits Neuronal Growth and Promotes Autophagy in Mouse Hippocampal HT-22 Cell Line. Front. Pharmacol. 2022, 11, 536913. [Google Scholar] [CrossRef]
  140. Fernández-de Frutos, M.; Galán-Chilet, I.; Goedeke, L.; Kim, B.; Pardo-Marqués, V.; Pérez-García, A.; Herrero, J.I.; Fernán-dez-Hernando, C.; Kim, J.; Ramírez, C.M. MicroRNA 7 Impairs Insulin Signaling and Regulates Aβ Levels through Post-transcriptional Regulation of the Insulin Receptor Substrate 2, Insulin Receptor, Insulin-Degrading Enzyme, and Liver X Re-ceptor Pathway. Mol. Cell. Biol. 2019, 39, e00170-19. [Google Scholar] [CrossRef]
  141. Liu, N.; Yang, C.; Gao, A.; Sun, M.; Lv, D. MiR-101: An Important Regulator of Gene Expression and Tumor Ecosystem. Cancers 2022, 14, 5861. [Google Scholar] [CrossRef]
  142. Al-hasso IK, Q.; Al-Derzi, A.R.; Abbas AA, H.; Gorial, F.I.; Alnuimi, A.S. Role of circulating miRNA-130b-3p and TGF-β 1cytokine in patients with systemic lupus erythematosus. Gene Rep. 2022, 26, 101476. [Google Scholar] [CrossRef]
  143. Zhao, Y.; Li, A. miR-19b-3p relieves intervertebral disc degeneration through modulating PTEN/PI3K/Akt/mTOR signaling pathway. Aging 2021, 13, 22459–22473. [Google Scholar] [CrossRef] [PubMed]
  144. Tseng, A.M.; Mahnke, A.H.; Wells, A.B.; Salem, N.A.; Allan, A.M.; Roberts, V.H.; Newman, N.; Walter, N.A.; Kroenke, C.D.; Grant, K.A.; et al. Maternal circulating miRNAs that predict infant FASD outcomes influence placental maturation. Life Sci. Alliance 2019, 2, e201800252. [Google Scholar] [CrossRef] [PubMed]
  145. Miranda, R.C. MicroRNAs and ethanol toxicity. Int. Rev. Neurobiol. 2014, 115, 245–284. [Google Scholar] [PubMed]
  146. Mahnke, A.H.; Sideridis, G.D.; Salem, N.A.; Tseng, A.M.; Carter, R.C.; Dodge, N.C.; Rathod, A.B.; Molteno, C.D.; Meintjes, E.M.; Jacobson, S.W.; et al. Infant circulating MicroRNAs as biomarkers of effect in fetal alcohol spectrum disorders. Sci. Rep. 2021, 11, 1429. [Google Scholar] [CrossRef] [PubMed]
  147. Schirle, N.T.; Sheu-Gruttadauria, J.; MacRae, I.J. Gene regulation. Structural basis for microRNA targeting. Science 2014, 346, 608–613. [Google Scholar] [CrossRef] [PubMed]
  148. Goetzl, L.; Darbinian, N.; Merabova, N. Noninvasive assessment of fetal central nervous system insult: Potential application to Prenatal Diagnosis. Prenat. Diagn. 2019, 39, 609–615. [Google Scholar] [CrossRef]
  149. Darbinian, N.; Darbinyan, A.; Merabova, N.; Bajwa, A.; Tatevosian, G.; Martirosyan, D.; Zhao, H.; Selzer, M.E.; Goetzl, L. Ethanol-mediated alterations in oligodendrocyte differentiation in the developing brain. Neurobiol. Dis. 2021, 148, 105181. [Google Scholar] [CrossRef]
  150. Darbinian, N.; Darbinyan, A.; Sinard, J.; Tatevosian, G.; Merabova, N.; D’Amico, F.; Khader, T.; Bajwa, A.; Martirosyan, D.; Gawlinski, A.K.; et al. Molecular Markers in Maternal Blood Exosomes Allow Early Detection of Fetal Alcohol Spectrum Disorders. Int. J. Mol. Sci. 2022, 24, 135. [Google Scholar] [CrossRef]
  151. Darbinian, N.; Merabova, N.; Tatevosian, G.; Morrison, M.; Darbinyan, A.; Zhao, H.; Goetzl, L.; Selzer, M.E. Biomarkers of Affective Dysregulation Associated with In Utero Exposure to EtOH. Cells 2024, 13, 2. [Google Scholar] [CrossRef]
  152. Visvanathan, J.; Lee, S.; Lee, B.; Lee, J.W.; Lee, S.K. The microRNA miR-124 antagonizes the anti-neural REST/SCP1 pathway during embryonic CNS development. Genes Dev. 2007, 21, 744–749. [Google Scholar] [CrossRef]
  153. Conaco, C.; Otto, S.; Han, J.J.; Mandel, G.; Otto, S.; Han, J.J.; Mandel, G.; Han, J.J.; Mandel, G.; Mandel, G. Reciprocal actions of REST and a microRNA promote neuronal identity. Proc. Natl. Acad. Sci. USA 2006, 103, 2422–2427. [Google Scholar] [CrossRef]
  154. Cheng, L.C.; Pastrana, E.; Tavazoie, M.; Doetsch, F. miR-124 regulates adult neurogenesis in the subventricular zone stem cell niche. Nat. Neurosci. 2009, 12, 399–408. [Google Scholar] [CrossRef]
  155. Farrell, B.C.; Power, E.M.; Mc Dermott, K.W. Developmentally regulated expression of Sox9 and microRNAs 124 128 and 23 in neuroepithelial stem cells in the developing spinal cord. Int. J. Dev. Neurosci. 2011, 29, 31–36. [Google Scholar] [CrossRef]
  156. Åkerblom, M.; Sachdeva, R.; Barde, I.; Verp, S.; Gentner, B.; Trono, D.; Jakobsson, J. MicroRNA-124 is a subventricular zone neuronal fate determinant. J. Neurosci. 2012, 32, 8879–8889. [Google Scholar] [CrossRef]
  157. Makeyev, E.V.; Zhang, J.; Carrasco, M.A.; Maniatis, T. The microRNA miR-124 promotes neuronal differentiation by triggering brain-specific alternative pre-mRNA splicing. Mol. Cell 2007, 27, 435–448. [Google Scholar] [CrossRef]
  158. Xue, Y.; Qian, H.; Hu, J.; Zhou, B.; Zhou, Y.; Hu, X.; Karakhanyan, A.; Pang, Z.; Fu, X.D. Sequential regulatory loops as key gatekeepers for neuronal reprogramming in human cells. Nat. Neurosci. 2016, 19, 807–815. [Google Scholar] [CrossRef]
  159. Xue, Y.; Ouyang, K.; Huang, J.; Zhou, Y.; Ouyang, H.; Li, H.; Wang, G.; Wu, Q.; Wei, C.; Bi, Y.; et al. Direct conversion of fibroblasts to neurons by reprogramming PTB-regulated MicroRNA circuits. Cell 2013, 152, 82–96. [Google Scholar] [CrossRef] [PubMed]
  160. Dai, W.; Li, W.; Hoque, M.; Li, Z.; Tian, B.; Makeyev, E.V. A post-transcriptional mechanism pacing expression of neural genes with precursor cell differentiation status. Nat. Commun. 2015, 6, 7576. [Google Scholar] [CrossRef] [PubMed]
  161. Papadimitriou, E.; Koutsoudaki, P.N.; Thanou, I.; Karagkouni, D.; Karamitros, T.; Chroni-Tzartou, D.; Gaitanou, M.; Gkemisis, C.; Margariti, M.; Xingi, E.; et al. A miR-124-mediated post-transcriptional mechanism controlling the cell fate switch of astrocytes to induced neurons. Stem Cell Rep. 2023, 18, 915–935. [Google Scholar] [CrossRef] [PubMed]
  162. Epple, R.; Krüger, D.; Berulava, T.; Brehm, G.; Ninov, M.; Islam, R.; Köster, S.; Fischer, A. The Coding and Small Non-coding Hippocampal Synaptic RNAome. Mol. Neurobiol. 2021, 58, 2940–2953. [Google Scholar] [CrossRef] [PubMed]
  163. Lin, S.L. microRNAs and Fragile X Syndrome. Adv. Exp. Med. Biol. 2015, 888, 107–121. [Google Scholar] [PubMed]
  164. Edbauer, D.; Neilson, J.R.; Foster, K.A.; Wang, C.-F.; Seeburg, D.P.; Batterton, M.N.; Tada, T.; Dolan, B.M.; Sharp, P.A.; Sheng, M. Regulation of Synaptic Structure and Function by FMRP-Associated MicroRNAs miR-125b and miR-132. Neuron 2010, 65, 373–384. [Google Scholar] [CrossRef] [PubMed]
  165. Vo, D.T.; Qiao, M.; Smith, A.D.; Burns, S.C.; Brenner, A.J.; Penalva, L.O.F. The oncogenic RNA-binding protein Musashi1 is regulated by tumor suppressor miRNAs. RNA Biol. 2011, 8, 817–828. [Google Scholar] [CrossRef] [PubMed]
  166. Santos, M.C.T.; Tegge, A.N.; Correa, B.R.; Mahesula, S.; Kohnke, L.Q.; Qiao, M.; Ferreira, M.A.R.; Kokovay, E.; Penalva, L.O.F. MiR-124, -128, and -137 orchestrate neural differentiation by acting on overlapping gene sets containing a highly connected transcription factor network. Stem Cells 2016, 34, 220–232. [Google Scholar] [CrossRef] [PubMed]
  167. Zhang, W.; Kim, P.J.; Chen, Z.; Lokman, H.; Qiu, L.; Zhang, K.; Rozen, S.G.; Tan, E.K.; Je, H.S.; Zeng, L. MiRNA-128 regulates the proliferation and neurogenesis of neural precursors by targeting PCM1 in the developing cortex. Elife 2016, 5, e11324. [Google Scholar] [CrossRef] [PubMed]
  168. Franzoni, E.; Booker, S.A.; Parthasarathy, S.; Rehfeld, F.; Grosser, S.; Srivatsa, S.; Fuchs, H.; Tarabykin, V.; Vida, I.; Wulczyn, F.G. miR-128 regulates neuronal migration, outgrowth and intrinsic excitability via the intellectual disability gene Phf6. Elife 2015, 4, e04263. [Google Scholar] [CrossRef]
  169. Sun, G.; Ye, P.; Murai, K.; Lang, M.F.; Li, S.; Zhang, H.; Li, W.; Fu, C.; Yin, J.; Wang, A.; et al. miR-137 forms a regulatory loop with nuclear receptor TLX and LSD1 in neural stem cells. Nat. Commun. 2011, 2, 529. [Google Scholar] [CrossRef]
  170. Olde Loohuis, N.F.; Ba, W.; Stoerchel, P.H.; Kos, A.; Jager, A.; Schratt, G.; Martens, G.J.; van Bokhoven, H.; Nadif Kasri, N.; Aschrafi, A. MicroRNA-137 Controls AMPA-Receptor-Mediated Transmission and mGluR-Dependent LTD. Cell Rep. 2015, 11, 1876–1884. [Google Scholar] [CrossRef]
  171. Abdelmohsen, K.; Hutchison, E.R.; Lee, E.K.; Kuwano, Y.; Kim, M.M.; Masuda, K.; Srikantan, S.; Subaran, S.S.; Marasa, B.S.; Mattson, M.P.; et al. miR-375 inhibits differentiation of neurites by lowering HuD levels. Mol. Cell. Biol. 2010, 30, 4197. [Google Scholar] [CrossRef] [PubMed]
  172. Magill, S.T.; Cambronne, X.A.; Luikart, B.W.; Lioy, D.T.; Leighton, B.H.; Westbrook, G.L.; Mandel, G.; Goodman, R.H. microRNA-132 regulates dendritic growth and arborization of newborn neurons in the adult hippocampus. Proc. Natl. Acad. Sci. USA 2010, 107, 20382–20387. [Google Scholar] [CrossRef]
  173. Hancock, M.L.; Preitner, N.; Quan, J.; Flanagan, J.G. MicroRNA-132 is enriched in developing axons, locally regulates Rasa1 mRNA, and promotes axon extension. J. Neurosci. 2014, 34, 66–78. [Google Scholar] [CrossRef] [PubMed]
  174. Marler, K.J.; Suetterlin, P.; Dopplapudi, A.; Rubikaite, A.; Adnan, J.; Maiorano, N.A.; Lowe, A.S.; Thompson, I.D.; Pathania, M.; Bordey, A.; et al. BDNF promotes axon branching of retinal ganglion cells via miRNA-132 and p250GAP. J. Neurosci. 2014, 34, 969–979. [Google Scholar] [CrossRef] [PubMed]
  175. Luikart, B.W.; Bensen, A.L.; Washburn, E.K.; Perederiy, J.V.; Su, K.G.; Li, Y.; Kernie, S.G.; Parada, L.F.; Westbrook, G.L. miR-132 mediates the integration of newborn neurons into the adult dentate gyrus. PLoS ONE 2011, 6, e19077. [Google Scholar] [CrossRef]
  176. Wayman, G.A.; Davare, M.; Ando, H.; Fortin, D.; Varlamova, O.; Cheng, H.Y.; Marks, D.; Obrietan, K.; Soderling, T.R.; Goodman, R.H.; et al. An activity-regulated microRNA controls dendritic plasticity by down-regulating p250GAP. Proc. Natl. Acad. Sci. USA 2008, 105, 9093–9098. [Google Scholar] [CrossRef]
  177. Mellios, N.; Sugihara, H.; Castro, J.; Banerjee, A.; Le, C.; Kumar, A.; Crawford, B.; Strathmann, J.; Tropea, D.; Levine, S.S.; et al. miR-132, an experience-dependent microRNA, is essential for visual cortex plasticity. Nat. Neurosci. 2011, 14, 1240–1242. [Google Scholar] [CrossRef] [PubMed]
  178. Tognini, P.; Putignano, E.; Coatti, A.; Pizzorusso, T. Experience-dependent expression of miR-132 regulates ocular dominance plasticity. Nat. Neurosci. 2011, 14, 1237–1239. [Google Scholar] [CrossRef] [PubMed]
  179. Anand, S.; Majeti, B.K.; Acevedo, L.M.; Murphy, E.A.; Mukthavaram, R.; Scheppke, L.; Huang, M.; Shields, D.J.; Lindquist, J.N.; Lapinski, P.E.; et al. MicroRNA-132-mediated loss of p120RasGAP activates the endothelium to facilitate pathological angiogenesis. Nat. Med. 2010, 16, 909–914. [Google Scholar] [CrossRef]
  180. Klein, M.E.; Lioy, D.T.; Ma, L.; Impey, S.; Mandel, G.; Goodman, R.H. Homeostatic regulation of MeCP2 expression by a CREB-induced microRNA. Nat. Neurosci. 2007, 10, 1513–1514. [Google Scholar] [CrossRef]
  181. Lagos, D.; Pollara, G.; Henderson, S.; Gratrix, F.; Fabani, M.; Milne, R.S.; Gotch, F.; Boshoff, C. miR-132 regulates antiviral innate immunity through suppression of the p300 transcriptional co-activator. Nat. Cell Biol. 2010, 12, 513–519. [Google Scholar] [CrossRef] [PubMed]
  182. Xu, B.; Zhang, Y.; Du, X.F.; Li, J.; Zi, H.X.; Bu, J.W.; Yan, Y.; Han, H.; Du, J.L. Neurons secrete miR-132-containing exosomes to regulate brain vascular integrity. Cell Res. 2017, 27, 882–897. [Google Scholar] [CrossRef]
  183. Walgrave, H.; Penning, A.; Tosoni, G.; Snoeck, S.; Davie, K.; Davis, E.; Wolfs, L.; Sierksma, A.; Mars, M.; Bu, T.; et al. microRNA-132 regulates gene expression programs involved in microglial homeostasis. iScience 2023, 26, 106829. [Google Scholar] [CrossRef] [PubMed]
  184. Dajas-Bailador, F.; Bonev, B.; Garcez, P.; Stanley, P.; Guillemot, F.; Papalopulu, N. microRNA-9 regulates axon extension and branching by targeting Map1b in mouse cortical neurons. Nat. Neurosci. 2012, 15, 697–699. [Google Scholar] [CrossRef] [PubMed]
  185. Johnston, R.J., Jr.; Chang, S.; Etchberger, J.F.; Ortiz, C.O.; Hobert, O. MicroRNAs acting in a double-negative feedback loop to control a neuronal cell fate decision. Proc. Natl. Acad. Sci. USA 2005, 102, 12449–12454. [Google Scholar] [CrossRef]
  186. Cai, S.; Zhou, P.; Liu, Z. Functional characteristics of a double negative feedback loop mediated by microRNAs. Cogn. Neurodyn. 2013, 7, 417–429. [Google Scholar] [CrossRef] [PubMed]
  187. Radhakrishnan, B.; Alwin Prem Anand, A. Role of miRNA-9 in Brain Development. J. Exp. Neurosci. 2016, 10, 101–120. [Google Scholar] [CrossRef]
  188. Yuva-Aydemir, Y.; Simkin, A.; Gascon, E.; Gao, F.B. MicroRNA-9: Functional evolution of a conserved small regulatory RNA. RNA Biol. 2011, 8, 557–564. [Google Scholar] [CrossRef] [PubMed]
  189. Packer, A.N.; Xing, Y.; Harper, S.Q.; Jones, L.; Davidson, B.L. The bifunctional microRNA miR-9/miR-9* regulates REST and CoREST and is downregulated in Huntington’s disease. J. Neurosci. 2008, 28, 14341–14346. [Google Scholar] [CrossRef]
  190. Abdelrahman, A.; Conn, R. Eye abnormalities in fetal alcohol syndrome. Ulst. Med. J. 2009, 78, 164–165. [Google Scholar]
  191. Strömland, K. Visual impairment and ocular abnormalities in children with fetal alcohol syndrome. Addict. Biol. 2004, 9, 153–157. [Google Scholar] [CrossRef] [PubMed]
  192. Rice, D.; Barone, S. Critical periods of vulnerability for the developing nervous system: Evidence from humans and animal models. Environ. Health Perspect. 2000, 108 (Suppl. S3), 511–533. [Google Scholar] [PubMed]
  193. Miranda, R.C. MicroRNAs and Fetal Brain Development: Implications for Ethanol Teratology during the Second Trimester Period of Neurogenesis. Front. Genet. 2012, 3, 77. [Google Scholar] [CrossRef] [PubMed]
  194. Guo, Y.; Chen, Y.; Carreon, S.; Qiang, M. Chronic intermittent ethanol exposure and its removal induce a different miRNA expression pattern in primary cortical neuronal cultures. Alcohol. Clin. Exp. Res. 2012, 36, 1058–1066. [Google Scholar] [CrossRef] [PubMed]
  195. Wang, L.L.; Zhang, Z.; Li, Q.; Yang, R.; Pei, X.; Xu, Y.; Wang, J.; Zhou, S.F.; Li, Y. Ethanol exposure induces differential microRNA and target gene expression and teratogenic effects which can be suppressed by folic acid supplementation. Hum. Reprod. 2009, 24, 562–579. [Google Scholar] [CrossRef] [PubMed]
  196. Sathyan, P.; Golden, H.B.; Miranda, R.C. Competing interactions between micro-RNAs determine neural progenitor survival and proliferation after ethanol exposure: Evidence from an ex vivo model of the fetal cerebral cortical neuroepithelium. J. Neurosci. 2007, 27, 8546–8557. [Google Scholar] [CrossRef] [PubMed]
  197. Pietrzykowski, A.Z.; Friesen, R.M.; Martin, G.E.; Puig, S.I.; Nowak, C.L.; Wynne, P.M.; Siegelmann, H.T.; Treistman, S.N. Posttranscriptional regulation of BK channel splice variant stability by miR-9 underlies neuroadaptation to alcohol. Neuron 2008, 59, 274–287. [Google Scholar] [CrossRef] [PubMed]
  198. Balaraman, S.; Winzer-Serhan, U.H.; Miranda, R.C. Opposing actions of ethanol and nicotine on microRNAs are mediated by nicotinic acetylcholine receptors in fetal cerebral cortical-derived neural progenitor cells. Alcohol. Clin. Exp. Res. 2012, 36, 1669–1677. [Google Scholar] [CrossRef]
  199. Tal, T.L.; Franzosa, J.A.; Tilton, S.C.; Philbrick, K.A.; Iwaniec, U.T.; Turner, R.T.; Waters, K.M.; Tanguay, R.L. MicroRNAs control neurobehavioral development and function in zebrafish. FASEB J. 2012, 26, 1452–1461. [Google Scholar] [CrossRef]
  200. Dukes, K.; Tripp, T.; Petersen, J.; Robinson, F.; Odendaal, H.; Elliott, A.; Willinger, M.; Hereld, D.; Raffo, C.; Kinney, H.C.; et al. A modified Timeline Followback assessment to capture alcohol exposure in pregnant women: Application in the Safe Passage Study. Alcohol 2017, 62, 17–27. [Google Scholar] [CrossRef]
Figure 1. Effects of EtOH exposure on the normal reductions in miRNA expressions in fetal brain and maternal plasma during the course of gestation. Non-EtOH-exposed human fetal brain tissues from the first and second trimesters were compared with unexposed controls with respect to the expression of 84 miRNAs, using Real-Time qRT-PCR. The results in fold-changes are displayed as heat maps. To assess the normal developmental changes, in (A), only unexposed controls were measured and expressed as fold-changes relative to their expression levels at the earliest-studied time point, 11.3 weeks GA, which is not shown because, by definition, the values equal 1. Expression of the selected miRNAs generally decreased (green) at later GAs in maternal blood (top). The pattern was less clear in the fetal brain (bottom), with expression levels at first increasing (red) but then decreasing toward the 11.3 values (black). N = 6 for each group. In (B), each box represents the average change in expression for a specific miRNA in EtOH-exposed cases relative to their unexposed individually matched controls. N = 6 paired cases for each group, including the 11.3-week time point in (A). In the second trimester, EtOH exposure was associated with a significant upregulation (red) of target miRNAs in the maternal blood (top) but a downregulation (green) in the fetal brain (bottom). (C) The position of each studied miRNA and housekeeping control gene in the 96-well array is shown (all controls and SNORDs for normalization were positioned in row H from H1 to H12). hsa: human, Homo sapiens; miR-9-3p vs. miR-9-5p: The 5p strand is present in the forward (5′-3′) sense, while the 3p strand is the reverse sense complementary strand.
Figure 1. Effects of EtOH exposure on the normal reductions in miRNA expressions in fetal brain and maternal plasma during the course of gestation. Non-EtOH-exposed human fetal brain tissues from the first and second trimesters were compared with unexposed controls with respect to the expression of 84 miRNAs, using Real-Time qRT-PCR. The results in fold-changes are displayed as heat maps. To assess the normal developmental changes, in (A), only unexposed controls were measured and expressed as fold-changes relative to their expression levels at the earliest-studied time point, 11.3 weeks GA, which is not shown because, by definition, the values equal 1. Expression of the selected miRNAs generally decreased (green) at later GAs in maternal blood (top). The pattern was less clear in the fetal brain (bottom), with expression levels at first increasing (red) but then decreasing toward the 11.3 values (black). N = 6 for each group. In (B), each box represents the average change in expression for a specific miRNA in EtOH-exposed cases relative to their unexposed individually matched controls. N = 6 paired cases for each group, including the 11.3-week time point in (A). In the second trimester, EtOH exposure was associated with a significant upregulation (red) of target miRNAs in the maternal blood (top) but a downregulation (green) in the fetal brain (bottom). (C) The position of each studied miRNA and housekeeping control gene in the 96-well array is shown (all controls and SNORDs for normalization were positioned in row H from H1 to H12). hsa: human, Homo sapiens; miR-9-3p vs. miR-9-5p: The 5p strand is present in the forward (5′-3′) sense, while the 3p strand is the reverse sense complementary strand.
Ijms 25 05826 g001aIjms 25 05826 g001b
Figure 2. Exposure to EtOH is associated with changes in the expression of eight neurological disorder-related miRNAs. Expressions of candidate miRNAs from Table 4 were assayed by qRT-PCR on samples of maternal serum (A) and fetal brain homogenates (B) from 12 pregnancies and their fetal GA- and sex-matched controls. Results were expressed as fold change between each EtOH-exposed case and its control. An overall upregulation in miRNA expression was seen in the second trimester compared to the first in maternal serum but not in the fetal brain. The expression of miR-509 was almost unaltered, so it is not included in this graph.
Figure 2. Exposure to EtOH is associated with changes in the expression of eight neurological disorder-related miRNAs. Expressions of candidate miRNAs from Table 4 were assayed by qRT-PCR on samples of maternal serum (A) and fetal brain homogenates (B) from 12 pregnancies and their fetal GA- and sex-matched controls. Results were expressed as fold change between each EtOH-exposed case and its control. An overall upregulation in miRNA expression was seen in the second trimester compared to the first in maternal serum but not in the fetal brain. The expression of miR-509 was almost unaltered, so it is not included in this graph.
Ijms 25 05826 g002
Figure 3. Exposure to EtOH is associated with reduced expression of miR-9 and miR-132 in FB-Es. (A) Levels of miRNA were measured by qRT-PCR in FB-Es of 20 EtOH-exposed maternal plasmas and compared with 20 individually matched unexposed controls, 10 matched pairs from the first trimester and 10 from the second. Expressions of miR-9 (A) and miR-132 (B) were normalized to SNORD, according to miScript Qiagen recommendations. Each assay was performed in triplicate and averaged. Changes in EtOH-exposed cases were expressed in comparison with their fetal sex- and GA-matched controls, measured relative to SNORD, and then their averages were assigned a value of 1. Error bars indicate the standard deviations for the 20 averages per group. Significance levels are for the comparison between all EtOH-exposed and all unexposed controls, based on ANOVA. * p < 0.05, ** p < 0.01.
Figure 3. Exposure to EtOH is associated with reduced expression of miR-9 and miR-132 in FB-Es. (A) Levels of miRNA were measured by qRT-PCR in FB-Es of 20 EtOH-exposed maternal plasmas and compared with 20 individually matched unexposed controls, 10 matched pairs from the first trimester and 10 from the second. Expressions of miR-9 (A) and miR-132 (B) were normalized to SNORD, according to miScript Qiagen recommendations. Each assay was performed in triplicate and averaged. Changes in EtOH-exposed cases were expressed in comparison with their fetal sex- and GA-matched controls, measured relative to SNORD, and then their averages were assigned a value of 1. Error bars indicate the standard deviations for the 20 averages per group. Significance levels are for the comparison between all EtOH-exposed and all unexposed controls, based on ANOVA. * p < 0.05, ** p < 0.01.
Ijms 25 05826 g003
Figure 4. Prenatal exposure to EtOH is associated with inhibition of miR-9 expression in FB-Es. Absolute quantification of miR-9 in FB-Es isolated from the blood of 10 EtOH-exposed pregnant women and their individually matched controls was determined by measuring RNA copy numbers using ddPCR. Five ng of exosomal RNA (including miRNAs) was used in the reaction. Each assay was performed in triplicate. Control FB-Es contained 200–1200 copies of miR-9 per μL, while in EtOH-exposed FB-Es, 180–820 copies were found. In each case, the EtOH-exposed sample contained fewer copies of miR-9 than its GA-and sex-matched control (*** p = 0.006).
Figure 4. Prenatal exposure to EtOH is associated with inhibition of miR-9 expression in FB-Es. Absolute quantification of miR-9 in FB-Es isolated from the blood of 10 EtOH-exposed pregnant women and their individually matched controls was determined by measuring RNA copy numbers using ddPCR. Five ng of exosomal RNA (including miRNAs) was used in the reaction. Each assay was performed in triplicate. Control FB-Es contained 200–1200 copies of miR-9 per μL, while in EtOH-exposed FB-Es, 180–820 copies were found. In each case, the EtOH-exposed sample contained fewer copies of miR-9 than its GA-and sex-matched control (*** p = 0.006).
Ijms 25 05826 g004
Figure 5. Potential targets of miRNA-9 in double-negative feedback loop pathways observed in FB-Es. The cargo of FB-Es from 20 pregnant women who had consumed EtOH and their individually matched controls were analyzed by ELISA and quantitative Western blot analysis (qWestern; (AC)). Relative downregulation was observed quantitatively by qWestern for Synapsin (A), REST (B), and Shh (C) and by ELISA for BDNF (D). * p < 0.05, ** p < 0.01. (E). Proposed double-negative feedback loop pathways for miR-9 expression. In the brain, miR-9 inhibits transcription of the transcription factor REST, which in turn inhibits the expression of miR-9. REST also inhibits the translation of BDNF, which in turn inhibits the expression of miR-9, as well as upregulating Synapsin and reciprocally upregulating Shh. Activation of the Shh pathway induces an increase in BDNF expression and results in neuroprotection to oxidative stress.
Figure 5. Potential targets of miRNA-9 in double-negative feedback loop pathways observed in FB-Es. The cargo of FB-Es from 20 pregnant women who had consumed EtOH and their individually matched controls were analyzed by ELISA and quantitative Western blot analysis (qWestern; (AC)). Relative downregulation was observed quantitatively by qWestern for Synapsin (A), REST (B), and Shh (C) and by ELISA for BDNF (D). * p < 0.05, ** p < 0.01. (E). Proposed double-negative feedback loop pathways for miR-9 expression. In the brain, miR-9 inhibits transcription of the transcription factor REST, which in turn inhibits the expression of miR-9. REST also inhibits the translation of BDNF, which in turn inhibits the expression of miR-9, as well as upregulating Synapsin and reciprocally upregulating Shh. Activation of the Shh pathway induces an increase in BDNF expression and results in neuroprotection to oxidative stress.
Ijms 25 05826 g005
Figure 6. Reductions in FB-E miR-9 levels correlate with the reductions in eye diameter in a larger sample of fetuses exposed to EtOH. To enhance the potential practicality of FB-E miR-9 levels as a biomarker to predict FASDs, FB-Es were isolated from the blood of a larger group of mothers who consumed alcohol during pregnancy and their unexposed controls, matched only for GA from 9–23 weeks, disregarding other control factors, such as fetal sex. FB-E miR-9 levels were measured by ddPCR. Eye diameters were measured in histological sections of the fetuses. (A). Copy numbers of miR-9 were reduced by almost 37.5% in EtOH-exposed cases. (B). Each individual FB-E miR-9 level from the 40 EtOH-exposed fetuses is graphed next to its GA-matched control and arrayed in order of GA. In all but the two youngest cases, the EtOH-exposed levels were higher than those of their GA-matched controls. Note also that the control levels of miR-9 rose dramatically early in the second trimester, whereas the miR-9 levels of the EtOH-exposed group did not show this increase. The assay was performed in triplicate. The dashed green line separates the first-trimester cases (on the left) from the second-trimester cases (on the right). (C). The correlation between the reduction in eye size (difference between EtOH-exposed fetus and its paired control) and the reduction in exosomal miR-9 levels is presented as a scatter plot. First-trimester (9 to 14 weeks GA) and second-trimester pregnancies (14.1 to 23 weeks GA) are graphed together. *** p << 0.001 (p = 0.000000464825), based on Spearman’s correlation with exact two-tailed critical p values. Data in (D) are presented in %.
Figure 6. Reductions in FB-E miR-9 levels correlate with the reductions in eye diameter in a larger sample of fetuses exposed to EtOH. To enhance the potential practicality of FB-E miR-9 levels as a biomarker to predict FASDs, FB-Es were isolated from the blood of a larger group of mothers who consumed alcohol during pregnancy and their unexposed controls, matched only for GA from 9–23 weeks, disregarding other control factors, such as fetal sex. FB-E miR-9 levels were measured by ddPCR. Eye diameters were measured in histological sections of the fetuses. (A). Copy numbers of miR-9 were reduced by almost 37.5% in EtOH-exposed cases. (B). Each individual FB-E miR-9 level from the 40 EtOH-exposed fetuses is graphed next to its GA-matched control and arrayed in order of GA. In all but the two youngest cases, the EtOH-exposed levels were higher than those of their GA-matched controls. Note also that the control levels of miR-9 rose dramatically early in the second trimester, whereas the miR-9 levels of the EtOH-exposed group did not show this increase. The assay was performed in triplicate. The dashed green line separates the first-trimester cases (on the left) from the second-trimester cases (on the right). (C). The correlation between the reduction in eye size (difference between EtOH-exposed fetus and its paired control) and the reduction in exosomal miR-9 levels is presented as a scatter plot. First-trimester (9 to 14 weeks GA) and second-trimester pregnancies (14.1 to 23 weeks GA) are graphed together. *** p << 0.001 (p = 0.000000464825), based on Spearman’s correlation with exact two-tailed critical p values. Data in (D) are presented in %.
Ijms 25 05826 g006
Table 1. Clinical characteristics of subjects used in the miRNA experiments.
Table 1. Clinical characteristics of subjects used in the miRNA experiments.
EtOH-Consuming Subjects (n = 40) Control Subjects (No EtOH, n = 40)
Maternal Age (years ± SD)26.17 ± 2.1522.34 ± 1.70
Gestational Age (weeks ± SD)15.47 ± 1.3315.16 ± 1.42
Race: White vs. Black (%)50 vs. 5050 vs. 50
Fetal Sex (male vs. female, %)50 vs. 5050 vs. 50
Table 2. miRNAs in the Neurological Development and Disease Array.
Table 2. miRNAs in the Neurological Development and Disease Array.
miRNAs in Neurological Development and Disease
Development: miR-124-3p, miR-125b-5p, miR-132-3p, miR-134, miR-138-5p, miR-9-5p.
Autistic Disorders: miR-106b-5p, miR-128, miR-132-3p, miR-140-5p, miR-146b-5p, miR-148b-3p, miR-15a-5p, miR-15b-5p, miR-181d, miR-193b-3p, miR-212-3p, miR-27a-3p, miR-320a, miR-381-3p, miR-431-5p, miR-432-5p, miR-484, miR-539-5p, miR-652-3p, miR-7-5p, miR-93-5p, miR-95.
Schizophrenia: let-7d-5p, let-7e-5p, miR-105-5p, miR-106b-5p, miR-107, miR-126-5p, miR-128, miR-130a-3p, miR-138-5p, miR-152, miR-15a-5p, miR-15b-5p, miR-181a-5p, miR-195-5p, miR-20a-5p, miR-212-3p, miR-24-3p, miR-26b-5p, miR-27a-3p, miR-29a-3p, miR-29b-3p, miR-29c-3p, miR-302a-5p, miR-302b-5p, miR-30d-5p, miR-338-3p, miR-346, miR-381-3p, miR-409-3p, miR-455-5p, miR-484, miR-485-5p, miR-487a, miR-489, miR-499a-5p, miR-512-3p, miR-518b, miR-7-5p, miR-9-3p, miR-92a-3p.
Anxiety Disorder: miR-128, miR-485-3p, miR-509-3p.
Tourette’s Syndrome: miR-24-3p.
Prion Diseases: let-7b-5p, miR-128, miR-139-5p, miR-146a-5p, miR-191-5p, miR-203a, miR-320a, miR-337-3p, miR-338-3p, miR-339-5p, miR-342-3p.
Huntington’s Disease: miR-124-3p, miR-132-3p, miR-135b-5p, miR-29a-3p, miR-29b-3p, miR-9-5p, miR-9-3p.
Parkinson’s Disease: miR-133b, miR-433, miR-7-5p.
Spinocerebellar Ataxia 1: miR-101-3p, miR-130a-3p, miR-19b-3p.
Table 3. Major miRNAs implicated in CNS development and neurogenesis.
Table 3. Major miRNAs implicated in CNS development and neurogenesis.
miRNATarget Molecule/InteractionEffectReferences
miR-124SCP1Stimulation of neurogenesis, neuronal differentiation
During CNS development, timely down-regulation of SCP1 stimulates neurogenesis, and miR-124 contributes to this process by down-regulating SCP1 expression.
[152]
Nonneuronal cells including neural progenitors: REST/SCP1 transcriptionally represses expression of miR-124 and other neuronal genes.[153]
Neurogenesis: miR-124 expression is derepressed, miR-124 post-transcriptionally suppresses multiple anti-neural factors including SCP1, resulting in further inhibition of the anti-neural pathway by REST/SCP1.[154]
miR-124 mediated repression of Sox9 associated with progression along the SVZ stem cell lineage to neurons (miR-124 is a neuronal fate determinant in the subventricular zone).[155]
Sox9Neuronal differentiation as a result of interplay between miR-124, PTBP1, and SCP1/REST[156]
[157]
De-repression of neuronal specific transcripts, including neurogenic RBPs Nova1, Rbfox1 and the nElavls[158]
Stimulation of neurogenesis
Ptbp1 [159]
Zfp36l1 [160]
Ezh2 (a negative regulator of neurogenesis) [161]
microRNA-124-5p Member of the synaptic microRNAome–regulators of the synaptic mRNA pool[162]
miR-125a
miR-125b
FMRPInteraction with FMRP: regulation of the signal transduction of metabolic glutamate receptors (mGluR1) and N-methyl-D-aspartate receptors (NMDAR) and neuronal development[163]
[164]
miR-128 (critical role in cortical neurogenesis)MSI1Commitment of NSPC to the neuronal lineage[165]
[166]
PCM1Reduced NPC proliferation, stimulation of NPC differentiation into neurons[167]
Phf6Cortical lamination: migration of neurons through the cortex, termination of upper neuron migration[168]
miR-137MSI1Commitment of NSPC to the neuronal lineage[165]
[166]
Neuronal differentiation and increased migration of progenitors into the cortical plate[169]
GluA1 subunit of AMPARsSynaptic efficacy and mGluR-dependent synaptic plasticity[170]
miR-9Zfp36
Ezh2
De-repression of neuronal specific transcripts, including neurogenic RBPs Nova1, Rbfox1 and the nElavls; Stimulation of neurogenesis[160]
miR-375Elavl4miR-375 is downregulated during the late stages of cortical development. The decrease of miR-375 leads to the de-repression of ELAVL4, with subsequent enhancement of neurite outgrowth in developing neurons[171]
miR-132p250GAP;Regulation of dendritic growth and arborization of newborn neurons in the adult hippocampus (CREB-mediated signaling)[172]
A positive regulation of developing axon extension[173]
mRNA for the Ras GTPase activator Rasa1;Synaptic structure and function[174]
FMRP [175]
Visual cortex plasticity[164]
MeCP2
Brain vascular integrity[176]
Cdh5 [177]
[178]
[179]
[180]
[181]
Microglial homeostasis[182]
Table 4. Neural differentiation- and cell proliferation-specific miRNAs and their targets.
Table 4. Neural differentiation- and cell proliferation-specific miRNAs and their targets.
miRNATargetsSome Important Roles
138ARHGEF3, ROCK2, VIM, SIRT, ETC.Precursor expressed in all tissues; mature miRNA only expressed in the brain. DNA Damage repair, and possibly sleep regulation.
26bEPHA2, CDK6, CCNE1, ETC.Neural Differentiation, and gene expression.
125bIGF2, IL6R, E2F2, MAPK14, ETC.Immune response, Osteoblast differentiation, and Neuroblastoma.
509NTRK3, CFTR, ETC.Cell proliferation and migration.
134VEGFA, ABCC1, FOXM1, ETC.Brain-specific, memory formation, and overexpressed in Schizophrenia.
132SIRT, CDKN1A, CCNA2, ETC.Neurogenesis, regulation of Inflammation in the brain and in the body, and Angiogenesis.
9REST, NFKB1, SIRT1, VIM, ETC.Neural Differentiation.
485NTRK3, NFYB, ETC.Synaptic formation regulation, and systemic iron balance.
128RELN, TGFBR1, TP53, ETC.Neuronal Migration, outgrowth, and excitability.
124EFNB1, CDK4, CDK6, VIM, ROCK2, NR3C1, IT6B1, SLC16A1, ETC.Neural Differentiation.
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

Darbinian, N.; Hampe, M.; Martirosyan, D.; Bajwa, A.; Darbinyan, A.; Merabova, N.; Tatevosian, G.; Goetzl, L.; Amini, S.; Selzer, M.E. Fetal Brain-Derived Exosomal miRNAs from Maternal Blood: Potential Diagnostic Biomarkers for Fetal Alcohol Spectrum Disorders (FASDs). Int. J. Mol. Sci. 2024, 25, 5826. https://doi.org/10.3390/ijms25115826

AMA Style

Darbinian N, Hampe M, Martirosyan D, Bajwa A, Darbinyan A, Merabova N, Tatevosian G, Goetzl L, Amini S, Selzer ME. Fetal Brain-Derived Exosomal miRNAs from Maternal Blood: Potential Diagnostic Biomarkers for Fetal Alcohol Spectrum Disorders (FASDs). International Journal of Molecular Sciences. 2024; 25(11):5826. https://doi.org/10.3390/ijms25115826

Chicago/Turabian Style

Darbinian, Nune, Monica Hampe, Diana Martirosyan, Ahsun Bajwa, Armine Darbinyan, Nana Merabova, Gabriel Tatevosian, Laura Goetzl, Shohreh Amini, and Michael E. Selzer. 2024. "Fetal Brain-Derived Exosomal miRNAs from Maternal Blood: Potential Diagnostic Biomarkers for Fetal Alcohol Spectrum Disorders (FASDs)" International Journal of Molecular Sciences 25, no. 11: 5826. https://doi.org/10.3390/ijms25115826

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