Elsevier

Neurobiology of Disease

Volume 35, Issue 2, August 2009, Pages 234-240
Neurobiology of Disease

The effect of mutant SOD1 dismutase activity on non-cell autonomous degeneration in familial amyotrophic lateral sclerosis

https://doi.org/10.1016/j.nbd.2009.05.002Get rights and content

Abstract

Mutant superoxide dismutase type 1 (MTSOD1), the most common known cause of familial amyotrophic lateral sclerosis (FALS), is believed to cause FALS as a result of a toxicity of the protein. MTSOD1s with full dismutase enzymatic activity (e.g., G37R) and without any enzymatic activity (e.g., G85R) cause FALS, demonstrating that the ability of MTSOD1 to cause FALS is not dependent on the dismutase activity; however, it remains unclear whether MTSOD1 dismutase activity can influence disease phenotype. In the present study, we selectively knocked down G85R expression in particular cell types of G85R mice. Results following knockdown of G85R in motor neurons (MNs)/interneurons of G85R mice were similar to results from a published study involving knockdown of G37R in G37R mice; however, G85R knockdown in microglia/macrophages induced a prolonged early and late disease phase while G37R knockdown in the same cells only affected late phase. These results show that: (i) MN as well as non-MN expression of G85R, like G37R, has a significant effect on disease in transgenic mice — indicating the role of non-cell autonomous degeneration in both dismutase-active and inactive MTSOD1s. (ii) The effect of MTSOD1 expression in microglia/macrophages varies with different mutants, and may be influenced by the MTSOD1's dismutase activity.

Introduction

Amyotrophic lateral sclerosis (ALS) is a relentless motor system degeneration that results in death usually 3–5 years after the onset. Approximately 10% of cases are inherited, and 15% of these familial ALS (FALS) cases are caused by mutations of Cu/Zn superoxide dismutase type 1 (SOD1). Scientists have vigorously investigated mutant (MT) SOD1-induced FALS under the assumption that this disease involves pathways of motor neuron (MN) death that are similar to those operative in sporadic ALS.

MTSOD1 is believed to cause cell death by means of a toxic gain of function rather than a loss of function. The mechanism by which MTSOD1 exerts its toxicity, however, remains unclear. MTSOD1s with full dismutase enzymatic activity (e.g., G37R) and without any enzymatic activity (e.g., G85R) cause FALS (Borchelt et al., 1994), demonstrating that the ability of the mutant protein to cause FALS is not dependent on its dismutase activity. Despite evidence indicating that a gain of function of MTSOD1 leads to FALS, the presence or absence of dismutase activity of the MT enzyme may still affect the pathogenesis or phenotype of the disease.

One important issue related to ALS is whether MN death is non-cell autonomous, i.e., whether MN death is influenced by the production of pathogenic factors produced in other cell types besides MNs. Compelling data related to this topic and especially relevant to the present study came from the use of CreLoxP technology to decrease the expression of MTSOD1 in one neural cell type with a constant MTSOD1 expression in the remainder of cell types (Boillee et al., 2006a, Yamanaka et al., 2008). In these experiments, mice were generated that carried a Cre recombinase transgene (that can excise a “floxed” gene that is flanked by LoxP sites) under the control of a cell type-specific transcriptional control element along with a floxed G37R MTSOD1 (G37Rflox) transgene. These mice were crossed with a mouse carrying Cre expressed in MNs (under the Islet-1 or VAChT promoter) or microglia/macrophages (under the CD11b promoter) to determine the effect of disease following a knockdown in G37R expression in these cell types. A decrease in G37R expression in the G37Rflox mice in MNs and other neurons delayed the onset and early phase of disease, while a decrease in expression of G37R in microglia and activated macrophages had little effect on the onset or early phase of disease, but significantly increased disease duration because of slowing of the late phase of disease.

In the present study, we carried out investigations of non-cell autonomous degeneration in G85Rflox mice similar to those carried out with G37Rflox mice (Boillee et al., 2006a, Yamanaka et al., 2008). Since G85R has no dismutase activity, while G37R has full dismutase activity, we reasoned that a comparison of our results with those obtained with G37Rflox transgenic mice might clarify the importance of the mutant's enzymatic activity in different neural cell types in FALS in the following way. One could envision that overexpression of a dismutase-active MTSOD1, such as G37R, in a particular cell type could have neuroprotective activity (in addition to its toxicity) since oxidative stress has been implicated as a pathogenic factor in the disease; relevant to the issue of oxidative stress in particular cell types is a recent study that showed that overexpression of another anti-oxidant defense, Nrf2, in astrocytes ameliorated disease in the FALS transgenic mouse (Vargas et al., 2008). If the protective properties of a dismutase-active MTSOD1 in a particular cell type outweigh its toxicity, a knockdown of the MTSOD1 in this cell type would be expected to actually accelerate disease. In fact, results from a recent report showed that knockdown of G37R in Schwann cells ameliorated FALS, suggesting that synthesis of G37R in Schwann cells is neuroprotective (Lobsiger et al., 2009). On the other hand, if the toxicity in a particular cell type of a dismutase-active MTSOD1, such as G37R, outweighed its neuroprotective properties, a knockdown would be expected to extend the onset and/or duration of disease compared to that seen in a mouse with expression of that same active MTSOD1 in all cell types. One might expect that the amount of disease amelioration seen with a knockdown of a dismutase-active mutant in the same cell type would be less than that seen with knockdown of dismutase-inactive MTSOD1 such as G85R — since G85R does not provide anti-oxidant activity.

Section snippets

Mice and breeding

A G85R genomic clone generously provided by Dr. David Borchelt was used to generate a G85Rflox SOD1 transgenic mouse, as described (Wang et al., 2009). This mouse was crossed with Lhx3:Cre and CD11b:Cre mice (a gift from Drs. G. Kassiotis and G. Kollias), both of which have been previously described (Boillee et al., 2006a, Peng et al., 2007, Sharma et al., 1998). The G85Rflox mouse was prepared on a C57BL/6J background. The Lhx3:Cre (Peng et al., 2007, Sharma et al., 1998) and CD11b:Cre mice (

The preparation and breeding of G85Rflox transgenic mice

A G85Rflox transgenic mouse was generated, as previously described (Wang et al., 2009). The G85Rflox transgenic mice developed progressive paralysis culminating in death. These G85Rflox transgenic mice were crossed with either Lhx3:Cre or CD11b:Cre mice to generate G85Rflox/Lhx3:Cre and G85Rflox/CD11b:Cre mice respectively. The Lhx3:Cre mice express Cre in spinal and hindbrain MNs and a population of interneurons in the spinal cord (Peng et al., 2007, Sharma et al., 1998), while CD11b:Cre mice

Discussion

Although MTSOD1-induced FALS makes up only ∼ 2% of the total number of ALS cases, it has provided a model system to explore the pathogenesis of this disease. There is convincing evidence that non-cell autonomous degeneration is important in the pathogenesis of MTSOD1-induced FALS, and that non-MN cell types may contribute to FALS pathogenesis. Although non-MN cell types play this important role, other reports show that transgenic mice with MTSOD1 expression restricted to neurons can develop MN

Acknowledgments

This work was supported by National Institutes of Health [NS049333-01 to RPR], ALS Association Foundation [#1211 to RPR], and Les Turner ALS Foundation [to RPR]. We thank Drs. George Kollias and George Kassiotis of the Alexander Fleming Biomedical Sciences Research Centre for the use of the CD11b:Cre mice.

References (25)

  • DengH.X. et al.

    Conversion to the amyotrophic lateral sclerosis phenotype is associated with intermolecular linked insoluble aggregates of SOD1 in mitochondria

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

    (2006)
  • Di GiorgioF.P. et al.

    Non-cell autonomous effect of glia on motor neurons in an embryonic stem cell-based ALS model

    Nat. Neurosci.

    (2007)
  • Cited by (92)

    • Translatomic analysis of regenerating and degenerating spinal motor neurons in injury and ALS

      2021, iScience
      Citation Excerpt :

      One of the challenges to understanding the etiology of ALS is that there are significant cell non-autonomous components to disease progression (Ilieva et al., 2009). This has been well demonstrated for SOD1 mutations in which the expression of mutant SOD1 in astrocytes and microglia does not affect the onset of disease but significantly contributes to disease progression (Boillee et al., 2006; Wang et al., 2009; Yamanaka et al., 2008). This underscores the need to evaluate motor neuron degeneration in the context of their complex environment.

    View all citing articles on Scopus
    View full text