Elsevier

Neuroscience Research

Volume 133, August 2018, Pages 48-57
Neuroscience Research

Dissociation of blood-brain barrier disruption and disease manifestation in an aquaporin-4-deficient mouse model of amyotrophic lateral sclerosis

https://doi.org/10.1016/j.neures.2017.11.001Get rights and content

Highlights

  • AQP4 overexpression is detected in ALS model mice and patients with ALS.

  • No aberrant AQP4 localization is observed in other chronic or acute gliosis models.

  • AQP4 deficiency improves BBB disruption in ALS mice.

  • AQP4 deficiency accelerates disease onset and shortens lifespan in ALS model mice.

Abstract

Aquaporin-4 (AQP4) is abundantly expressed in the central nervous system and is involved in the water balance in the cellular environment. Previous studies have reported that AQP4 expression is upregulated in rat models of amyotrophic lateral sclerosis (ALS), a fatal disease that affects motor neurons in the brain and spinal cord. In this study, we report that astrocytic AQP4 overexpression is evident during the course of disease in the spinal cord of an ALS mouse model, as well as in tissue from patients with ALS. AQP4 overexpression appears to be specifically associated with ALS because it was not induced by other experimental manipulations that produced acute or chronic gliosis. In order to examine the contribution of AQP4 to ALS disease development, we crossed AQP4 knockout mice with a mouse model of ALS. Significant improvement in blood-brain barrier (BBB) permeability was observed in the AQP4-deficient ALS mouse model. However, the time to disease onset and total lifespan were reduced in the AQP4-deficient ALS mouse model. The contradictory results suggest that changes in AQP4 may be context-dependent and further studies are required to understand the precise contribution of brain water balance in ALS.

Introduction

Astrocytes constitute one of the major building blocks of the blood-brain barrier (BBB), which tightly regulates the brain microenvironment to maintain proper neuronal functioning. Brain microvessels are wrapped by the basal lamina and astrocyte endfeet, which are known to serve as physical barriers and mutual functional bridges between neurons and endothelial cells/pericytes; this anatomical/physiological multi-cellular complex is termed the “neurovascular unit” (Abbott et al., 2006, Zhao et al., 2015, Zlokovic, 2005). Astrocyte proliferation (astrogliosis) and BBB breakdown have been implicated in the pathogenesis of many neurological or mental disorders (Abbott et al., 2006, Najjar et al., 2017, Zlokovic, 2005).

Amyotrophic lateral sclerosis (ALS) is a progressive degenerative disease affecting the upper and lower motor neurons in the brain and spinal cord (van Es et al., 2017). Many studies have reported that astrogliosis is one of the main pathological hallmarks observed in the spinal cords of patients with ALS, as well as in animal models of the disease (Schiffer et al., 1996, Hall et al., 1998). Recent studies have shown that reactive microglial cells and astrocytes actively contribute to the death of motor neurons in ALS mouse models and in patients with ALS (Turner et al., 2004, Philips and Robberecht, 2011). Astrocytes were shown to contribute to microglial activation, leading to further damage to motor neurons and accelerated disease progression via a non-cell autonomous mechanism (Yamanaka et al., 2008, Ilieva et al., 2009). Moreover, BBB disruption has been observed at an early, presymptomatic stage in mouse models of ALS (Garbuzova-Davis et al., 2007, Zhong et al., 2008) and in patients with ALS (Garbuzova-Davis et al., 2012), suggesting that vascular disease can occur simultaneously in ALS.

Aquaporin-4 (AQP4), the most abundantly expressed AQP isoform in the brain, is specifically expressed in astrocytes. AQP4 shows highly polarized localization (Nielsen et al., 1997) and serves as a marker for astrocyte endfeet, which suggests a possible role in maintaining BBB properties (Nicchia et al., 2004, Tomás-Camardiel et al., 2005, Wolburg et al., 2009). In a rat model of ALS in which mutant human superoxide dismutase 1 (SOD1) is expressed, the overexpression and mislocalization of AQP4 was observed in the affected motor nuclei in the brainstem and spinal cord (Nicaise et al., 2009, Bataveljić et al., 2012). However, it is not known whether the observed AQP4 abnormalities are a cause or consequence of ALS. Recent studies showed that AQP4 upregulation induced BBB disruption and brain edema, while inhibition of AQP4 attenuated BBB disruption in mouse models of ischemia (Tang et al., 2014, Wang et al., 2015).

In the present study, we investigated the expression of AQP4 in a mouse model of ALS, as well as in tissue from patients with ALS. In both cases, AQP4 was upregulated, which is consistent with previous studies in a rat model of ALS (Garbuzova-Davis et al., 2007, Zhong et al., 2008). We then utilized AQP4 knockout mice to create an AQP4-deficient ALS mouse model, and examined the role of AQP4 in BBB permeability and ALS disease progression.

Section snippets

Animals

Transgenic mice carrying the human mutant SOD1G93A gene (B6.Cg-Tg(SOD1*G93A)1Gur/J) were purchased from Jackson Laboratory (Bar Harbor, ME, USA) and maintained on a C57BL/6 background. Mice carrying the human SOD1G85R gene (line 148) or the wild-type human SOD1 gene (B6.Cg-Tg(SOD1)76Dpr) were kind gifts from Dr. Don Cleveland (University of California, San Diego). LoxSOD1G37R mice were previously described (Yamanaka et al., 2008). The AQP4 knockout mice (Ikeshima-Kataoka et al., 2013, Acc. No.

Overexpression of AQP4 in a mouse model of ALS and in tissue from patients with ALS

Immunohistochemical analysis revealed that AQP4 was specifically observed in astrocyte endfeet surrounding blood vessels in the lumbar spinal cord of wild-type (WT) control mice. AQP4 was shown to be mislocalized in age-matched SOD1G93A mice during the progression of ALS symptoms (Fig. 1). Numerous studies have reported the presence of activated astrocytes in ALS animal models and in patients with ALS (Bataveljić et al., 2012, Hall et al., 1998, Nicaise et al., 2009). In the present study,

Discussion

The overexpression of astrocytic AQP4 in the SOD1G93A rat model of ALS has been reported in previous studies (Bataveljić et al., 2012, Nicaise et al., 2009). In this study, we also showed that astrocytic AQP4 is overexpressed in mouse models of ALS, as well as in tissue from patients with ALS. In the spinal cord, abnormal AQP4 localization was more conspicuous in the ventral horn than in the dorsal horn. Moreover, we showed that AQP4 overexpression is uniquely associated with ALS disease state,

Author contributions

S.W-M., Y.M., T.O., and H.M. designed the experiments. S.W-M. and H.M. performed the experiments. K.Y., Y.A., and M.Y. contributed critical analytic tools and input. S.O. provided clinical and pathological data and tissue samples of human cases. S.W-M. and H.M. wrote the manuscript, and all authors discussed the data and commented on the manuscript.

Acknowledgments

We thank Airi Iwasaki, Yumiko Date, Akihiro Kondo, Yuki Sudo for help with the experiments. This work was supported by Grants-in-Aid for Scientific Research on Innovative Areas (Grant Number 23111006 to KY and HM) from the Ministry of Education, Culture, Sports, Science and Technology of Japan; JSPS KAKENHI Grant Number 16K15129 (to HM) and 26293208 (to KY).

References (44)

  • A.S. Verkman et al.

    Three distinct roles of aquaporin-4 in brain function revealed by knockout mice

    Biochim. Biophys. Acta

    (2006)
  • M.L. Vizuete et al.

    Differential upregulation of aquaporin-4 mRNA expression in reactive astrocytes after brain injury: potential role in brain edema

    Neurobiol. Dis.

    (1999)
  • M.A. van Es et al.

    Amyotrophic lateral sclerosis

    Lancet

    (2017)
  • Z. Zhao et al.

    Establishment and dysfunction of the blood-Brain barrier

    Cell

    (2015)
  • B.V. Zlokovic

    Neurovascular mechanisms of Alzheimer's neurodegeneration

    Trends Neurosci.

    (2005)
  • N.J. Abbott et al.

    Astrocyte–endothelial interactions at the blood–brain barrier

    Nat. Rev. Neurosci.

    (2006)
  • D. Bataveljić et al.

    Changes in the astrocytic aquaporin-4 and inwardly rectifying potassium channel expression in the brain of the amyotrophic lateral sclerosis SOD1(G93A) rat model

    Glia

    (2012)
  • O. Bloch et al.

    Aquaporin-4 gene deletion in mice increases focal edema associated with staphylococcal brain abscess

    J. Neurochem.

    (2005)
  • O. Bloch et al.

    Accelerated progression of kaolin-induced hydrocephalus in aquaporin-4-deficient mice

    J. Cereb. Blood Flow Metab.

    (2006)
  • V.A. Eidsvaag et al.

    Human and mouse cortical astrocytes differ in aquaporin-4 polarization toward microvessels

    Glia

    (2017)
  • E.D. Hall et al.

    Relationship of microglial and astrocytic activation to disease onset and progression in a transgenic model of familial ALS

    Glia

    (1998)
  • H. Ilieva et al.

    Non-cell autonomous toxicity in neurodegenerative disorders: ALS and beyond

    J. Cell Biol.

    (2009)
  • Cited by (22)

    • Stagnation of glymphatic interstitial fluid flow and delay in waste clearance in the SOD1-G93A mouse model of ALS

      2021, Neuroscience Research
      Citation Excerpt :

      In AQP4-deficient mice, ISF flow is weakened due to a decrease in the inflow of CSF from the perivascular space, and excretion of waste products from the parenchyma is delayed compared to that in WT mice (Fig. 4B). On the other hand, in SOD1G93A mice in which perivascular AQP4 localization is preserved, due to the overexpression and mislocalization of AQP4, the distribution of AQP4 is not limited to the endfeet of astrocytes, but is also localized in reactive astrocytes present in neuropil (Watanabe-Matsumoto et al., 2018). We consider that this abnormal AQP4 distribution causes abnormalities of the glymphatic system in SOD1G93A mice.

    View all citing articles on Scopus
    View full text