Elsevier

Life Sciences

Volume 252, 1 July 2020, 117664
Life Sciences

MicroRNA-374a-5p inhibits neuroinflammation in neonatal hypoxic-ischemic encephalopathy via regulating NLRP3 inflammasome targeted Smad6

https://doi.org/10.1016/j.lfs.2020.117664Get rights and content

Abstract

Aims

Neonatal hypoxic-ischemic encephalopathy (HIE) is still an important cause of neurological dysfunction. At present, there is no reliable biochemical index in clinical examination. Increasing evidence demonstrates that microRNAs (miRNAs) are involved in the process of HIE, and miR-374a-5p is down-regulated in HIE infants. In this study, the aim is to investigate the role and mechanism of miR-374a-5p in HIE.

Main methods

Sprague-Dawley (SD) rats were used to establish model of neonatal HIE, pathologic changes and inflammatory response of brain tissues were measured. Subsequently, primary microglia were induced by LPS (1 μg/ml) in vitro, the miR-374a-5p mimic, Ad-Smad6 adenovirus vector and NLRP3 siRNA oligo were applied for microglial transfection. Furthermore, the target relationship between miR-374a-5p and Smad6 was analyzed, while microglia activity and inflammatory factor (IL-1β, TNF-α and IL-6) levels were detected.

Key findings

Herein, we found that over-expression of miR-374a-5p significantly attenuated brain injury and strongly inhibited the release of pro-inflammatory cytokines in neonatal rat HIE model. In vitro, miR-374a-5p inhibited LPS-induced microglial pro-inflammatory cytokines production by regulating NLRP3 inflammasome. In addition, Smad6 was identified as a direct target of miR-374a-5p, and miR-374a-5p had a negative regulatory effect on Smad6 expression. By targeting Smad6, miR-374a-5p inhibited the activation of NLRP3 inflammatory signals in microglia and the subsequent release of pro-inflammatory factors.

Significance

Our study recognized that miR-374a-5p as a novel regulator of microglial activation in neonatal HIE highlighted potential therapeutic target for the treatment of neonatal hypoxic-ischemic brain injury.

Introduction

Neonatal hypoxic-ischemic encephalopathy (HIE) is attributed to hypoxia in the brain, causing approximately one million deaths each year, and is one of the leading causes of neonatal morbidity and mortality worldwide [1,2]. Although the continued progress of the treatment in the neonates, the molecular mechanisms of brain damage in neonates with hypoxia-ischemia is still unclear. Brain damage intensifies in the first few hours of birth and may be accompanied by long-term neurological disorders [3]. Consequently, targeted therapeutic interventions should be taken immediately after delivery for infants at risk of HIE. Hypothermia treatment (HT) can reduce oxygen, metabolic demand and inflammation, and inhibit apoptosis and cell death in clinical setting, thus improving the nervous system [4]. However, the range of treatment is very narrow, and nearly half of the infants who received hypothermia still died or had obvious neurological dysfunction [5]. At present, the criteria used to predict neonatal HIE severity are unreliable, so early biochemical markers will greatly contribute to clinical decisions.

Neuroinflammation is a chronic pathological process involving all classes of brain injury and disorder [6]. More and more reports confirm that the neuroinflammation induced by cerebral hypoxia-ischemia is the main factor of perinatal brain injury [7,8]. Cytokines released by neuroinflammation are considered to be the mediators of neonatal brain injury [9]. Dammann et al. expounded from the animal model of perioperative brain injury in infected environment [10]. Typically, the levels of inflammatory cytokines such as interleukin-1β (IL)-1β, IL-6 and IL-8 in cerebrospinal fluid of infants after HIE were increased [11]. Neuroinflammation is characterized by the activation of glial cells, especially microglia. Microglia are innate immune cells of the central nervous system that monitor and deal with invasive pathogens and environmental deterioration, and show resting phenotype under normal circumstances. In the case of stimuli such as brain injury, it may lead to adjacent neurons injury, eliciting more microglia reactivation [12]. In view of the implication of microglia in the neonatal HIE. Consequently, comprehending the regulation mechanism of microglia activation is conducive to the prevention of neonatal HIE.

MicroRNAs (miRNAs) are a highly conserved small RNA molecule (16–25 nucleotides in length) that recognize the 3′ untranslated region (3′UTR) of downstream target genes and induce mRNA degradation and/or inhibit translation. According to our understanding, the research on miRNA in HIE is very limited. Looney et al. published the first description about the miRNA analysis of the neonates, and reported that the miR-374a-5p is down-regulated in the umbilical blood of HIE infants [13]. Since miRNA regulates the expression of target mRNA by inhibition or degradation, it is a reliable bio-marker in molecular biological diagnosis [14] and may provide novel insights for HIE and other diseases. The aim of this study was to elucidate the biological function of miR-374a-5p and further explore its role in neonatal HIE.

Section snippets

Animals treatment

Pregnant Sprague-Dawley (SD) rats were obtained from Lab Animal Center of Sichuan University (Chengdu, China). After delivery, their pups were kept in a room of 20 ± 2 °C for 12 hour light/dark cycle, and were free to access clean food and water. Postnatal day 7 (P7) pups were used to construct HIE model, P0-1 pups were used to isolate primary microglia.

Neonatal HIE animals model

A HIE model in the P7 pups, using Levine modification in the adult rat, has been widely recognized and applied [15]. Briefly, P7 pups were

Up-regulation of miR-374a-5p attenuated pro-inflammatory factors release and brain injury in HIE

As shown in Fig. 1A, miR-374a-5p was down-regulated in the peripheral blood of HIE model, and the mRNA level of miR-374a-5p was critically increased after transfecting miR-374a-5p Agomir. Next, we detected the expression levels of pro-inflammatory cytokines (IL-1β, TNF-α and IL-6) in tissues and serum, respectively, the results revealed that miR-374a-5p Agomir decreased the expression of IL-1β, TNF-α and IL-6 at 24 h after transfection, compared with HIE group (Fig. 1B). Parallelly, the results

Discussion

Hypoxia-ischemic brain injury is the main cause of acute death and chronic nervous system diseases in infants. A string of neurotoxic events within hours after hypoxia-ischemia in brain, such as oxidative stress, inflammatory response and so on, resulting in extensive neuronal deaths and brain dysfunction. At present, the treatment of neonatal HIE is very limited, and it is urgent to find new treatment strategies. Here, we established a model of HIE in P7 rat pups to explore the role and

Conclusions

In a word, we found that the expression of miR-374a-5p was significantly down-regulated in neonatal HIE and it effectively inhibited microglia-mediated neuroinflammation. In vitro studies further demonstrated that miR-374a-5p overexpression attenuated the secretion of LPS-induced proinflammatory cytokines by modulating the NLRP3 inflammasome. Importantly, this study also revealed that Smad6 was the direct target gene of miR-374a-5p, and miR-374a-5p targeted Smad6 to regulate the inflammatory

Declaration of competing interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgments

The authors thank the lab provided by West China Medical Center of Sichuan University.

Funding

This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

References (42)

  • C. Robertson et al.

    Term infants with hypoxic-ischemic encephalopathy: outcome at 3.5 years

    Dev. Med. Child Neurol.

    (1985)
  • T. Wood et al.

    Treatment temperature and insult severity influence the neuroprotective effects of therapeutic hypothermia

    Sci. Rep.

    (2016)
  • B.S. Mietto et al.

    Neurotrauma and inflammation: CNS and PNS responses

    Mediat. Inflamm.

    (2015)
  • H. Hagberg et al.

    The role of inflammation in perinatal brain injury

    Nat. Rev. Neurol.

    (2015)
  • W. Deng

    Neurobiology of injury to the developing brain

    Nat. Rev. Neurol.

    (2010)
  • D.D. Jenkins et al.

    Serum cytokines in a clinical trial of hypothermia for neonatal hypoxic-ischemic encephalopathy

    J. Cereb. Blood Flow Metab.

    (2012)
  • A.I. Bartha et al.

    Neonatal encephalopathy: association of cytokines with MR spectroscopy and outcome

    Pediatric. Res.

    (2004)
  • C. Porta et al.

    Tolerance and M2 (alternative) macrophage polarization are related processes orchestrated by p50 nuclear factor κB

    Proc. Natl. Acad. Sci. U. S. A.

    (2009)
  • A.M. Looney et al.

    Downregulation of umbilical cord blood levels of miR-374a in neonatal hypoxic ischemic encephalopathy

    J. Pediatr.

    (2015)
  • X. Chen et al.

    Characterization of microRNAs in serum: a novel class of biomarkers for diagnosis of cancer and other diseases

    Cell Res.

    (2008)
  • R.C. Vannucci et al.

    Perinatal hypoxic-ischemic brain damage: evolution of an animal model

    Dev. Neurosci.

    (2005)
  • Cited by (25)

    • Inflammasomes as biomarkers and therapeutic targets in traumatic brain injury and related-neurodegenerative diseases: A comprehensive overview

      2023, Neuroscience and Biobehavioral Reviews
      Citation Excerpt :

      To the best of our knowledge, there are no studies that assessed miRNA to target the inflammasome in TBI, there are other studies that have demonstrated the effectiveness of certain miRNAs targeting inflammasomal components in related diseases. For example, (Chen et al., 2020) found that miR-374a-5p is down-regulated in neonatal hypoxic-ischemic encephalopathy, and overexpressing it regulated the NLRP3 inflammasome and Smad6 leading to a decrease in pro-inflammatory cytokines production (IL-1β, IL-6, and TNFα). Similarly, miR-9a-5p overexpression decreased protein levels of NLRP1, ASC, pro and cleaved-caspase-1, IL-1β and IL-18 in rats in rats subjected to middle cerebral artery occlusion and SY-5Y cells exposed to oxygen-glucose deprivation (Cao et al., 2020).

    • IKZF3 modulates cerebral ischemia/reperfusion injury by inhibiting neuroinflammation

      2023, International Immunopharmacology
      Citation Excerpt :

      Cerebral ischemic injury may be caused by a variety of mechanisms, including the inflammatory response and apoapsis and others [35], and the inflammatory response was deemed to be the leading cause [36]. Moreover, secondary brain injury is exacerbated by reperfusion [37], which leads to tissue damage, cellular structure destruction and impaired nerve function [38–40], causing neuronal injury [41]. Therefore, early suppression of the inflammatory response might be an effective treatment strategy for ischemia.

    • Legumain knockout improved cognitive impairment via reducing neuroinflammation in right unilateral common carotid artery occlusion mice

      2021, Life Sciences
      Citation Excerpt :

      However, the molecular mechanism underlying legumain activation in cerebral ischemic pathologies remains unclear. The inflammatory response is actively involved in mediating multiple human diseases, including ischemic reperfusion injury, Gaucher's disease, and neurodegenerative diseases [20–22]. Ischemia initiates brain inflammation, including the activation of microglia and acute inflammation, which exacerbates ischemic lesions.

    View all citing articles on Scopus
    View full text