Elsevier

Acta Biomaterialia

Volume 113, 1 September 2020, Pages 597-613
Acta Biomaterialia

Full length article
Mesenchymal stromal cell-derived extracellular vesicles modulate microglia/macrophage polarization and protect the brain against hypoxia-ischemic injury in neonatal mice by targeting delivery of miR-21a-5p

https://doi.org/10.1016/j.actbio.2020.06.037Get rights and content

Abstract

Mesenchymal stromal cell (MSC)-derived extracellular vesicles (EVs) (MSC-EVs) exhibit protective effects in damaged or diseased tissues. However, the role of EVs secreted by MSC in hypoxia-ischemic (HI) injury in neonatal mice remains unknown. Systemic administration of MSC-EVs attenuated acute brain damage and neuroinflammation, and skewed CD11b+/CD45low microglia and CD11b+/CD45high brain monocyte/macrophage towards a more anti-inflammatory property as determined at 72 h post-HI. In addition, MSC-EVs remarkably improve the injury outcomes pups prior to weaning (P21), while no effect on long-term memory impairment (P42). Importantly, these effects were preceded by incorporation of MSC-EVs into a large number of neurons and microglia within HI group. Abundant levels of miR-21a-5p were present in EVs as determined with next-generation sequencing. Notably, MSC-EVs treatment further increased miR-21a-5p levels at 72 h post HI. Knockdown analyses revealed that miR-21a-5p, and its target-Timp3, were essential for this neuroprotective property of MSC-EVs following HI exposure as demonstrated in both in vitro and in vivo models. These findings suggest that a systemic administration of EVs derived from MSC, have the capacity to incorporated into neurons and microglia where they can then exert neuroprotection against HI-induced injury in neonates through the delivery of miR-21a-5p.

Introduction

Neonatal hypoxic–ischemia encephalopathy (HIE) is a common neurologic condition encountered in newborns and currently no promising therapy exists. One recent approach has involved that of cell therapy with use of mesenchymal stromal cell (MSC). This protocol has shown promise in animal models of perinatal hypoxia-ischemic (HI) brain injury, including decreases in the volume of ipsilateral hemisphere and improved functional outcomes [1,2]. The mechanisms through which MSC mediate these effects are unclear and continue to be investigated. While only a small number of transplanted MSC actually are able to engraft in host tissues, and most are not permanently retained, their therapeutic benefits, in part, appear to involve a promotion of endogenous repair processes such as inducing neurogenesis and angiogenesis and a reduction in glial scarring resulting from the release of paracrine factors [3], [4], [5], [6].

Findings from recent studies suggested that the paracrine effects of MSCadministration are largely due to extracellular vesicles (EVs) secreted by MSC [7,8]. EVs are small membrane vesicle, and classified into exosomes (30–200 nm), microvesicles (200–1000 nm) and apoptosomes (1–10 μm) depending on their size [9]. Exosome-enriched EV fractions mediate intercellular communication via transferring bio-active molecules, including DNA, messengerRNAs (mRNAs), microRNAs (miRNAs), proteins and lipids [10]; and, it has been suggested that miRNAs can be taken up by recipient cells and, in this way, influence the fate of target cells [11,12].

Results from recent studies have indicated that MSC-EVs can exert beneficial effects in ischemic injury. For example, a systemic administrations of EVs derived from MSC have been shown to offer neuroprotective effects against ischemia in adult mice [13,14]; and intravenous administration of human bone marrow-MSC-EVs prevented global HI induced functional impairment and structural injury within the preterm brain [15]. Moreover, MSC-derived EVs have the potential to prevent oxygen–glucose deprivation/reoxygenation (OGD/R)-initiated apoptosis in mouse neuroblastoma cells [16]. However, whether EVs secreted by MSC display any functional activity within neonatal HI injury remain unknown. In this study, we investigated the effects of a short-term MSC-EVs administration on acute brain injury, and on long-term neurological functioning within neonatal HI mice. In addition, we characterized some of the underlying mechanisms involved in these effects.

Section snippets

HI model and treatments

A total of 385 male mice were used in this study. All animal experiments were performed in accordance with the International Guiding Principles for Animal Research provided by the Council for International Organizations of Medical Sciences (CIOMS), and procedures were approved by the Animal Ethical and Welfare Committee of Shandong University (approval No. ECSBMSSDU2018–2–059). Participants who worked with the animal models were trained following Institutional Animal Care and Use Committee

EVs treatment suppressed edema, infarction volume and tissue loss

At passage 3, C57BL/6 J mouse bone marrow MSC was identified by morphology and Flow Cytometric Analysis (FACS). Cells showed a typical spindle-like morphology after reaching about 90% confluence (Fig. S1A). Alizarin Red staining and Oli Red O staining were utilized to identify the osteogenic and adipogenic differentiation of MSC respectively (Fig. S1B). FACS was applied to confirm that MSC were positive for CD29 and CD44 but negative for CD45 or CD11b (Fig. S1C).

The size of MSC-EVs within the

Discussion

In the present study, we demonstrated that a systemic administration of MSC- derived EVs following HI markedly improved acute injury and neuroinflammation in immature mice. These effects were associated with promoting CD11b+/CD45low microglia and CD11b+/CD45high monocyte/macrophage towards a more anti-inflammatory property following MSC-EVs treatment. Moreover, MSC-EVs remarkably improved behavioral impairments pups prior to weaning (P21), while no effect on long-term memory impairment (P42).

Declaration of Competing Interest

The authors declare no conflict of interest.

Acknowledgments

Research funding support for this work was from the National Natural Science Foundation of China (No. 81873768 and 81671213 to Dr. Zhen Wang), the Key Research and Development Foundation of Shandong Province (No.2017GSF218091 to Dr. Zhen Wang), the National Key Research and Development Program of China (No. 2017YFC0820203 to Dr. Dexiang Liu). We gratefully acknowledge the help of Fang Pan for assistance with the MWM test.

Compliance with ethical standards

The animal experiments were performed based on the International Guiding Principles for Animal Research provided by the Council for International Organizations of Medical Sciences (CIOMS), and procedures were approved by the Animal Ethical and Welfare Committee of Shandong University. Participants who worked with the animal models were trained according to Institutional Animal Care and Use Committee Guidebook (IACUC) rules.

Autors contribution

ZW: made substantial contributions to study design, data interpretation, writing and revising of the manuscript, and final revision of the manuscript; DQX: made substantial contributions to laboratory work, the analysis of data, and edited the manuscript; TTL: performed cell cultures and EVs labeling; XLC, HFK, ZYY, and LLC: performed behavioral testing; XMB: performed EVs analysis; DXL: revised the manuscript.

All authors read and approved the final manuscript.

Reference (51)

  • X. Zhou et al.

    Mesenchymal stem cell derived EVs mediate neuroprotection after spinal cord injury in rats via the microRNA-21-5p/FasL gene axis

    Biomed. Pharmac.

    (2019)
  • Z. Yin et al.

    Neuron-derived exosomes with high miR-21-5p expression promoted polarization of M1 microglia in culture

    Brain Behav. Immun.

    (2020)
  • C.Y. Kuan et al.

    Mechanisms of programmed cell death in the developing brain

    Trends Neurosci.

    (2000)
  • B. Oppliger et al.

    Intranasal delivery of umbilical cord-derived mesenchymal stem cells preserves myelination in perinatal brain damage

    Stem Cells Dev.

    (2016)
  • C.T. van Velthoven et al.

    Mesenchymal stem cells as a treatment for neonatal ischemic brain damage

    Pediatr. Res.

    (2012)
  • G. Qiu et al.

    Mesenchymal stem cell-derived extracellular vesicles affect disease outcomes via transfer of microRNAs

    Stem Cell Res. Ther.

    (2018)
  • J.D. Anderson et al.

    Comprehensive Proteomic Analysis of Mesenchymal Stem Cell Exosomes Reveals Modulation of Angiogenesis via Nuclear Factor-KappaB Signaling

    Stem Cells

    (2016)
  • N. Heldring et al.

    Therapeutic Potential of Multipotent Mesenchymal Stromal Cells and Their Extracellular Vesicles

    Hum. Gene Ther.

    (2015)
  • C. Thery et al.

    Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines

    J. Extracell. Vesicles

    (2018)
  • S.D. Kadam et al.

    Neuropathogical features of a rat model for perinatal hypoxic-ischemic encephalopathy with associated epilepsy

    J. Comp. Neurol.

    (2007)
  • H. Xin et al.

    Exosome-mediated transfer of miR-133b from multipotent mesenchymal stromal cells to neural cells contributes to neurite outgrowth

    Stem Cells

    (2012)
  • H. Xin et al.

    MicroRNA cluster miR-17-92 Cluster in exosomes enhance neuroplasticity and functional recovery after stroke in rats

    Stroke

    (2017)
  • M. Deng et al.

    Mesenchymal Stem Cell-Derived Extracellular Vesicles Ameliorates Hippocampal Synaptic Impairment after Transient Global Ischemia

    Front. Cell. Neurosci.

    (2017)
  • G.W. Hu et al.

    Exosomes secreted by human-induced pluripotent stem cell-derived mesenchymal stem cells attenuate limb ischemia by promoting angiogenesis in mice

    Stem Cell. Res. Ther.

    (2015)
  • D.R. Ophelders et al.

    Mesenchymal stromal cell-derived extracellular vesicles protect the fetal brain after hypoxia-ischemia

    Stem Cells Transl. Med.

    (2016)
  • Cited by (46)

    • Bioengineering exosomes for treatment of organ ischemia-reperfusion injury

      2022, Life Sciences
      Citation Excerpt :

      Among different parent cells, native exosomes derived from stem cells (MSC-Exo) have received more attention [77–79]. For example, the uptake of mesenchymal stromal cell-derived exosomes by neurons and microglia increases expression of miR-21a-5p, which has been linked to a neuroprotective effect against hypoxic brain ischemia injury [80]. Furthermore, exosomes derived from human urine stem cells (USCs) have elevated levels of in miR-216a-5p, which activates Akt phosphorylation and reduces renal I/R injury [81].

    View all citing articles on Scopus
    View full text