Elsevier

Neuroscience

Volume 443, 1 September 2020, Pages 188-205
Neuroscience

Research Article
Third Ventricular Injection of CCL2 in Rat Embryo Stimulates CCL2/CCR2 Neuroimmune System in Neuroepithelial Radial Glia Progenitor Cells: Relation to Sexually Dimorphic, Stimulatory Effects on Peptide Neurons in Lateral Hypothalamus

https://doi.org/10.1016/j.neuroscience.2020.01.020Get rights and content

Highlights

  • Embryonic intracerebroventricular (ICV) injection of CCL2 has effects like maternal CCL2.

  • ICV CCL2 stimulates CCL2 in hypothalamic neuroepithelial radial glia progenitor cells.

  • ICV CCL2 increases CCL2 and MCH neurons in lateral hypothalamus that colocalize CCL2.

  • These effects are sexually dimorphic with stronger effects in females than males.

  • Embryonic CCL2/CCR2 system may be involved in mediating effects of maternal ethanol.

Abstract

Clinical and animal studies show maternal alcohol consumption during pregnancy causes in offspring persistent alterations in neuroimmune and neurochemical systems known to increase alcohol drinking and related behaviors. Studies in lateral hypothalamus (LH) demonstrate in adolescent offspring that maternal oral administration of ethanol stimulates the neuropeptide, melanin-concentrating hormone (MCH), together with the inflammatory chemokine C–C motif ligand 2 (CCL2) and its receptor CCR2 which are increased in most MCH neurons. These effects, consistently stronger in females than males, are detected in embryos, not only in LH but hypothalamic neuroepithelium (NEP) along the third ventricle where neurons are born and CCL2 is stimulated within radial glia progenitor cells and their laterally projecting processes that facilitate MCH neuronal migration toward LH. With ethanol’s effects similarly produced by maternal peripheral CCL2 administration and blocked by CCR2 antagonist, we tested here using in utero intracerebroventricular (ICV) injections whether CCL2 acts locally within the embryonic NEP. After ICV injection of CCL2 (0.1 µg/µl) on embryonic day 14 (E14) when neurogenesis peaks, we observed in embryos just before birth (E19) a significant increase in endogenous CCL2 within radial glia cells and their processes in NEP. These auto-regulatory effects, evident only in female embryos, were accompanied by increased density of CCL2 and MCH neurons in LH, more strongly in females than males. These results support involvement of embryonic CCL2/CCR2 neuroimmune system in radial glia progenitor cells in mediating sexually dimorphic effects of maternal challenges such as ethanol on LH MCH neurons that colocalize CCL2 and CCR2.

Introduction

Recent studies have demonstrated that the neuroimmune system in the brain, functioning beyond the signaling for immune responses, produces profound changes in the development of neurons that result in long-term behavioral consequences (Cui et al., 2014, Crews et al., 2015, Chang et al., 2015, Chang et al., 2018). The wide-ranging and long-lasting functionalities of neuroimmune signaling are evident in the progression of ethanol’s effects on neuronal development in the embryo and subsequent changes in the offspring’s behavior associated with increased alcohol consumption. While chronic high doses or binge drinking episodes of ethanol are found to affect inflammatory pathways involving such molecules as Toll-like receptors which cause considerable neurodegeneration that exacerbates existing alcohol use disorder (Lippai et al., 2013, Crews and Vetreno, 2014, Flores-Bastias and Karahanian, 2018), there is evidence that lower levels and shorter periods of ethanol exposure can have very different effects. Along with elevated neuroimmune activity, these include an increase in cell proliferation, neurogenesis and migration (Camarillo and Miranda, 2008, Mooney and Miller, 2010, Skorput and Yeh, 2015) and in the expression and density of neurons in the lateral hypothalamus (LH) that express melanin-concentrating hormone (MCH) (Chang et al., 2015, Chang et al., 2018), a neuropeptide that promotes alcohol drinking and other behaviors associated with alcohol use disorder (Duncan et al., 2005, Morganstern et al., 2010, Karlsson et al., 2016). These studies of MCH in the LH demonstrate in the rat that maternal intraoral administration of ethanol, at low-to-moderate doses from embryonic day 10 (E10) to E15 during the period of peak hypothalamic neurogenesis (Ifft, 1972), increases in adolescent offspring the density of neurons that express the inflammatory chemokine C–C motif ligand 2 (CCL2) and its main receptor CCR2, a neuroimmune system also positively linked to alcohol intake (Blednov et al., 2005, June et al., 2015, Valenta and Gonzales, 2016). It also stimulates the co-expression of CCL2 and CCR2 in up to 90% of the MCH neurons (Chang et al., 2015, Chang et al., 2018)

The possibility that the CCL2/CCR2 system actually mediates these stimulatory effects of ethanol on MCH neurons is supported by the additional findings that they are similarly produced by maternal peripheral administration of CCL2 and are blocked by maternal administration of the CCR2 antagonist INCB3344 during the period of ethanol exposure (Chang et al., 2018, Chang et al., 2020a). A recent study of the developmental origins of this CCL2/CCR2-mediated stimulatory effect on neuronal development has shown that ethanol’s effects on MCH neurons that colocalize CCL2 and CCR2 in LH are evident in the embryo and neonatal offspring and are reversed by maternal administration of a CCL2 antibody that neutralizes endogenous CCL2 and a CCR2 antagonist that blocks CCL2’s main receptor (Chang et al., 2020a). Also in the embryo, maternal ethanol administration stimulates CCL2 cells dense in the hypothalamic neuroepithelium (NEP), a primary source of neurons along the third ventricle (Bedont et al., 2015), and it also increases the colocalization of CCL2 within radial glia progenitor cells and their laterally projecting processes, effects that are mimicked by maternal CCL2 administration and accompanied by an increased number of MCH neurons close to the radial glia cells and positioned along their processes (Chang et al., 2020b). Further tests demonstrate that these effects of maternal ethanol and CCL2 administration on the development of CCL2 and MCH neurons are consistently stronger in females than males, as shown in the embryo as well as adolescent offspring (Chang et al., 2018, Chang et al., 2020a, Chang et al., 2020b) Thus, in response to inflammatory challenges such as ethanol, these studies suggest the involvement of endogenous CCL2-rich radial glia progenitor cells in embryos in promoting, in a sexually dimorphic manner, the genesis and migration of MCH neurons toward their final destination in the LH.

While bringing attention to the CCL2/CCR2 system and radial glia in the NEP as a possible mechanism underlying these strong stimulatory effects on neuronal development, these studies with maternal peripheral manipulations of ethanol or CCL2 in pregnant rats leave open the question as to whether these alterations in this neuroimmune mechanism result from direct effects on the embryonic brain or whether they are indirect consequences of effects in the mother produced by peripheral manipulations. To investigate this question, we employed here in utero intracerebroventricular (ICV) injections to directly manipulate the embryonic brain during the period of peak hypothalamic neurogenesis, while having minimal impact on maternal neuroimmune factors. This ICV technique, allowing delivery of substances directly into the microenvironment of developing neural systems, not only avoids the confounding variables of maternal peripheral injections but also reduces some uncertainty regarding the molecular dynamics of neuroimmune compounds crossing the fetal blood brain barrier. While studies over the years have employed ICV injections to alter brain microenvironments in adult, adolescent, and postnatal rodents (Lewinski et al., 1984, Jung et al., 1994, Fang et al., 2013, Li et al., 2016), this technique has more recently been used in the embryo to administer genetic material with electroporation to induce localized changes in gene expression in both superficial and deeper embryonic brain regions (Walantus et al., 2007, Vomund et al., 2013, Fekete et al., 2017, Rosin and Kurrasch, 2018). These investigations have reported good success in terms of both embryo survival rates and the effectiveness of gene transfer as revealed by measurements after birth and into adolescence. To our knowledge, there are only a few in utero ICV injection studies in rodent models that have administered non-genetic material without electroporation directly into the embryonic brain for purposes of providing pharmacological manipulations. In two such reports, direct injections of neuroimmune signaling molecules, the growth factor TGF-B1 that promotes radial glia differentiation (Stipursky et al., 2014) and maternal autoantibodies related to autism spectrum disorders (Camacho et al., 2014), have been performed, with little complication and significant success in demonstrating subsequent changes in target molecules and target regions.

In this manuscript, we use this technique of in utero ICV injection to accomplish two goals. The first is to determine if this technique is a viable method for investigating and directly manipulating mechanisms in the embryonic brain. The second is to test whether the effects in the offspring produced by maternal manipulations of a neuroimmune agent during pregnancy can be reproduced by delivering this agent into the embryonic brain, indicating that they reflect direct actions on the embryo rather than indirect consequences from changes in the mother. Specifically, using the in utero ICV injection technique, we delivered CCL2, a potent signaling molecule, directly into the third ventricle of the embryo at E14, when hypothalamic neurogenesis peaks (Ifft, 1972), and investigated in the embryo just before birth at E19 its effects on neuroimmune function in both the highly proliferative NEP surrounding the third ventricle and the more distant neuronal systems in the LH. In the NEP, we examined the CCL2-containing radial glia progenitor cells in addition to their processes in the medial hypothalamus (mHYP) that facilitate neuronal migration and then tested the CCL2 and MCH neurons in the LH, the site of their final destination. The results obtained with embryonic ICV CCL2 injection reveal very similar effects on the embryo as those produced by maternal peripheral administration of CCL2. In addition to validating this technique which negates potential confounding effects of maternal manipulations that indirectly alter the embryonic and amniotic environment, these results provide strong evidence for the involvement of a localized CCL2 system in the embryo brain in mediating the effects of maternal manipulations. They specifically demonstrate how CCL2 functions in an autoregulatory and sexually dimorphic manner to stimulate the radial glia progenitor cells and processes in which it colocalizes and the development of CCL2 neurons in the LH along with MCH neurons which themselves grow to colocalize CCL2 as well as CCR2.

Section snippets

Experimental procedures

All procedures were conducted in a fully accredited AAALAC facility (22 °C, 12:12-h light–dark cycle with lights off at 8 am), in accordance with protocols approved by The Rockefeller University Animal Care and Use Committee and consistent with the NIH Guide to the Care and Use of Laboratory Animals.

Technique involving ICV injection of CCL2 at E14 and brain analyses at E19

To more fully understand the technique of in utero ICV injection, we used a dye solution (0.1% Fast Green in normal saline) to reveal the nature and extent of its spread and performed histological analyses to verify the site of the injection and assess the extent of damage. As illustrated in the photomicrograph of an E16 embryo sacrificed 2 h after injection (Fig. 2A), the injection through the anterior fissure of the dye solution (1 µl) shows from the surface of the skull how the dye is almost

Discussion

Studies investigating prenatal exposure to teratogenic compounds are generally limited to maternal peripheral manipulations, precluding an analysis of disturbances that reflect actions directly within the fetal brain. Such functional clarity at the fetal level, however, is crucial for a comprehensive understanding of embryonic mechanisms mediating the effects observed subsequently in the offspring, as well as for developing effective therapeutic interventions. The current study demonstrates the

Acknowledgements

This research was supported by National Institute on Alcohol Abuse and Alcoholism of the National Institutes of Health under Award Number R01AA024798. The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH. The authors would like to thank Ms. Gazal Gulati for her assistance with the literature, references and figures. We also extend gratitude to The Rockefeller University’s Bio-Imaging Resource Center and Translational Technology

Declaration of interest

None.

References (76)

  • O. Flores-Bastias et al.

    Neuroinflammation produced by heavy alcohol intake is due to loops of interactions between toll-like 4 and tnf receptors, peroxisome proliferator-activated receptors and the central melanocortin system: a novel hypothesis and new therapeutic avenues

    Neuropharmacology

    (2018)
  • X.B. Gao

    Electrophysiological effects of mch on neurons in the hypothalamus

    Peptides

    (2009)
  • J.S. Jung et al.

    Effects of intraventricular injection of morphine and beta-endorphin on serotonin release from the spinal cord in rats

    Pharmacol Biochem Behav

    (1994)
  • M.M. McCarthy et al.

    Fast, furious and enduring: Sensitive versus critical periods in sexual differentiation of the brain

    Physiol Behav

    (2018)
  • S.M. Mooney et al.

    Prenatal exposure to ethanol affects postnatal neurogenesis in thalamus

    Exp Neurol

    (2010)
  • I. Morganstern et al.

    Increased orexin and melanin-concentrating hormone expression in the perifornical lateral hypothalamus of rats prone to overconsuming a fat-rich diet

    Pharmacol Biochem Behav

    (2010)
  • E. Pellegrini et al.

    Steroid modulation of neurogenesis: Focus on radial glial cells in zebrafish

    J Steroid Biochem Mol Biol

    (2016)
  • M.R. Peltier et al.

    Sex differences in stress-related alcohol use

    Neurobiol Stress

    (2019)
  • Z. Ren et al.

    Ethanol-induced damage to the developing spinal cord: The involvement of ccr2 signaling

    Biochim Biophys Acta

    (2017)
  • M. Takeda et al.

    Chemokine ligand 2/chemokine receptor 2 signaling in the trigeminal ganglia contributes to inflammatory hyperalgesia in rats

    Neurosci Res

    (2018)
  • K. Zhang et al.

    Role of mcp-1 and ccr2 in alcohol neurotoxicity

    Pharmacol Res

    (2019)
  • P. Abe et al.

    Intermediate progenitors facilitate intracortical progression of thalamocortical axons and interneurons through cxcl12 chemokine signaling

    J Neurosci

    (2015)
  • N. Adachi et al.

    Corticotropin-releasing hormone-binding protein is up-regulated by brain-derived neurotrophic factor and is secreted in an activity-dependent manner in rat cerebral cortical neurons

    J Neurochem

    (2018)
  • F. Akhtar et al.

    Acute maternal oxidant exposure causes susceptibility of the fetal brain to inflammation and oxidative stress

    J Neuroinflammation

    (2017)
  • J. Altman et al.

    Atlas of prenatal rat brain development

    (1995)
  • J. Bakker et al.

    Alpha-fetoprotein protects the developing female mouse brain from masculinization and defeminization by estrogens

    Nat Neurosci

    (2006)
  • J.R. Barson et al.

    Neurochemical heterogeneity of rats predicted by different measures to be high ethanol consumers

    Alcohol Clin Exp Res

    (2013)
  • J.L. Bedont et al.

    Patterning, specification, and differentiation in the developing hypothalamus

    Wiley Interdiscip Rev Dev Biol

    (2015)
  • F. Brischoux et al.

    Ontogenetic development of the diencephalic mch neurons: a hypothalamic ‘mch area’ hypothesis

    Eur J Neurosci

    (2001)
  • M.E. Brocca et al.

    Non-reproductive functions of aromatase in the central nervous system under physiological and pathological conditions

    Cell Mol Neurobiol

    (2019)
  • C. Camarillo et al.

    Ethanol exposure during neurogenesis induces persistent effects on neural maturation: evidence from an ex vivo model of fetal cerebral cortical neuroepithelial progenitor maturation

    Gene Expr

    (2008)
  • J. Cazareth et al.

    Molecular and cellular neuroinflammatory status of mouse brain after systemic lipopolysaccharide challenge: Importance of ccr2/ccl2 signaling

    J Neuroinflammation

    (2014)
  • C. Cerri et al.

    The chemokine ccl2 mediates the seizure-enhancing effects of systemic inflammation

    J Neurosci

    (2016)
  • M. Chakrabarti et al.

    Molecular mechanisms of estrogen for neuroprotection in spinal cord injury and traumatic brain injury

    Rev Neurosci

    (2016)
  • G.Q. Chang et al.

    Prenatal exposure to nicotine stimulates neurogenesis of orexigenic peptide-expressing neurons in hypothalamus and amygdala

    J Neurosci

    (2013)
  • G.Q. Chang et al.

    Radial glia progenitor cells and ccl2/ccr2 system in embryonic neuroepithelium: Involvement in sexually dimorphic, stimulatory effects of maternal ethanol administration on development of hypothalamic melanin-concentrating hormone neurons in offspring

    J Neuroinflammation

    (2020)
  • G.Q. Chang et al.

    Hypothalamic ccl2/ccr2 chemokine system: Role in sexually dimorphic effects of maternal ethanol exposure on melanin-concentrating hormone and behavior in adolescent offspring

    J Neurosci

    (2018)
  • F.T. Crews et al.

    Neuroimmune function and the consequences of alcohol exposure

    Alcohol Res

    (2015)
  • Cited by (0)

    View full text