Gliogenesis from the subventricular zone modulates the extracellular matrix at the glial scar after brain ischemia

  1. Achucarro Basque Center for Neuroscience, Leioa, Spain
  2. Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Spain
  3. CIBERNED, Madrid, Spain
  4. Donostia International Physics Center (DIPC), San Sebastian, Spain
  5. Aix Marseille Univ, CNRS, IBDM, Campus de Luminy, Marseille, France
  6. Univ. Bordeaux, CNRS, IMN, UMR 5293, F-33000 Bordeaux, France
  7. Ikerbasque, Basque Foundation for Science, Bilbao, Spain
  8. Basque Biomodel Platform for Human Research (BBioH), Leioa, Spain
  9. Instituto Biofisika (CSIC, UPV/EHU), Fundación Biofísica Bizkaia, Leioa, Spain

Editors

  • Reviewing Editor
    Annalisa Scimemi
    University at Albany, State University of New York, Albany, United States of America
  • Senior Editor
    Sofia Araújo
    University of Barcelona, Barcelona, Spain

Reviewer #1 (Public Review):

Summary:

The authors show that SVZ-derived astrocytes respond to a middle carotid artery occlusion (MCAO) hypoxia lesion by secreting and modulating hyaluronan at the edge of the lesion (penumbra) and that hyaluronan is a chemoattractant to SVZ astrocytes. They use lineage tracing of SVZ cells to determine their origin. They also find that SVZ-derived astrocytes express Thbs-4 but astrocytes at the MCAO-induced scar do not. Also, they demonstrate that decreased HA in the SVZ is correlated with gliogenesis. While much of the paper is descriptive/correlative they do overexpress Hyaluronan synthase 2 via viral vectors and show this is sufficient to recruit astrocytes to the injury. Interestingly, astrocytes preferred to migrate to the MCAO than to the region of overexpressed HAS2.

Strengths:

The field has largely ignored the gliogenic response of the SVZ, especially with regard to astrocytic function. These cells and especially newborn cells may provide support for regeneration. Emigrated cells from the SVZ have been shown to be neuroprotective via creating pro-survival environments, but their expression and deposition of beneficial extracellular matrix molecules are poorly understood. Therefore, this study is timely and important. The paper is very well written and the flow of results is logical.

Weaknesses:

The main problem is that they do not show that Hyaluronan is necessary for SVZ astrogenesis and or migration to MCAO lesions. Such loss of function studies have been carried out by studies they cite (e.g. Girard et al., 2014 and Benner et al., 2013). Similar approaches seem to be necessary in this work.

Major points:

(1) How good of a marker for newborn astrocytes is Thbs4? Did you co-label with B cell markers like EGFr? Is the Thbs4 gene expressed in B cells? Do scRNAseq papers show it is expressed in B cells? Are they B1 or B2 cells?

(2) It is curious that there was no increase in Type C cells after MCAO - do the authors propose a direct NSC-astrocyte differentiation?

(3) The paper would be strengthened with orthogonal views of z projections to show co-localization.

(4) It is not clear why the dorsal SVZ is analysed and focused on in Figure 4. This region emanates from the developmental pallium (cerebral cortex anlagen). It generates some excitatory neurons early postnatally and is thought to have differential signalling such as Wnt (Raineteau group).

(5) Several of the images show the lesion and penumbra as being quite close to the SVZ. Did any of the lesions contact the SVZ? If so, I would strongly recommend excluding them from the analysis as such contact is known to hyperactivate the SVZ.

(6) The authors switch to a rat in vitro analysis towards the end of the study. This needs to be better justified. How similar are the molecules involved between mouse and rat?

(7) Similar comment for overexpression of naked mole rat HA.

Reviewer #2 (Public Review):

Summary:

In their manuscript, Ardaya et al have addressed the impact of ischemia-induced gliogenesis from the adult SVZ and their effect on the remodeling of the extracellular matrix (ECM) in the glial scar. They use Thbs4, a marker previously identified to be expressed in astrocytes of the SVZ, to understand its role in ischemia-induced gliogenesis. First, the authors show that Thbs4 is expressed in the SVZ and that its expression levels increase upon ischemia. Next, they claim that ischemia induces the generation of newborn astrocyte from SVZ neural stem cells (NSCs), which migrate toward the ischemic regions to accumulate at the glial scar. Thbs4-expressing astrocytes are recruited to the lesion by Hyaluronan where they modulate ECM homeostasis.

Strengths:

The findings of these studies are in principle interesting and the experiments are in principle good.

Weaknesses:

The manuscript suffers from an evident lack of clarity and precision in regard to their findings and their interpretation.

Reviewer #3 (Public Review):

Summary:

The authors aimed to study the activation of gliogenesis and the role of newborn astrocytes in a post-ischemic scenario. Combining immunofluorescence, BrdU-tracing, and genetic cellular labelling, they tracked the migration of newborn astrocytes (expressing Thbs4) and found that Thbs4-positive astrocytes modulate the extracellular matrix at the lesion border by synthesis but also degradation of hyaluronan. Their results point to a relevant function of SVZ newborn astrocytes in the modulation of the glial scar after brain ischemia. This work's major strength is the fact that it is tackling the function of SVZ newborn astrocytes, whose role is undisclosed so far.

Strengths:

The article is innovative, of good quality, and clearly written, with properly described Materials and Methods, data analysis, and presentation. In general, the methods are designed properly to answer the main question of the authors, being a major strength. Interpretation of the data is also in general well done, with results supporting the main conclusions of this article.

Weaknesses:

However, there are some points of this article that still need clarification to further improve this work.

- As a first general comment, is it possible that the increase in Thbs4-positive astrocytes can also happen locally close to the glia scar, through the proliferation of local astrocytes or even from local astrocytes at the SVZ? As it was shown in published articles most of the newborn astrocytes in the adult brain actually derive from proliferating astrocytes, and a smaller percentage is derived from NSCs. How can the authors rule out a contribution of local astrocytes to the increase of Thbs4-positive astrocytes? The authors also observed that only about one-third of the astrocytes in the glial scar derived from the SVZ.

- It is known that the local, GFAP-reactive astrocytes at the scar can form the required ECM. The authors propose a role of Thbs4-positive astrocytes in the modulation, and perhaps maintenance, of the ECM at the scar, thus participating in scar formation likewise. So, this means that the function of newborn astrocytes is only to help the local astrocytes in the scar formation and thus contribute to tissue regeneration. Why do we need specifically the Thbs4-positive astrocytes migrating from the SVZ to help the local astrocytes? Can you discuss this further?

- The authors observed that the number of BrdU- and DCX-positive cells decreased 15 dpi in all OB layers (Fig. S5). They further suggest that ischemia-induced a change in the neuroblasts ectopic migratory pathway, depriving the OB layers of the SVZ newborn neurons. Are the authors suggesting that these BrdU/DCX-positive cells now migrate also to the ischemic scar, or do they die? In fact, they see an increase in caspase-3 positive cells in the SVZ after ischemia, but they do not analyse which type of cells are dying. Alternatively, is there a change in the fate of the cells, and astrogliogenesis is increased at the expense of neurogenesis? The authors should understand which cells are Cleaved-caspase-3 positive at the SVZ and clarify if there is a change in cell fate. Also please clarify what happens to the BrdU/DCX-positive cells that are born at the SVZ but do not migrate properly to the OB layers.

- The authors showed decreased Nestin protein levels at 15 dpi by western blot and immunostaining shows a decrease already at 7div (Figure 2). These results mean that there is at least a transient depletion of NSCs due to the promotion of astrogliogenesis. However, the authors show that at 30dpi there is an increase of slow proliferating NSCs (Figure 3). Does this mean, that there is a reestablishment of the SVZ cytogenic process? How does it happen, more specifically, how NSCs number is promoted at 30dpi? Please explain how are the NSCs modulated throughout time after ischemia induction and its impact on the cytogenic process.

- The authors performed a classification of Thbs4-positive cells in the SVZ according to their morphology. This should be confirmed with markers expressed by each of the cell subtypes.

- In Figure S6, the authors quantified HABP spots inside Thbs4-positive astrocytes. Please show a higher magnification picture to show how this quantification was done.

  1. Howard Hughes Medical Institute
  2. Wellcome Trust
  3. Max-Planck-Gesellschaft
  4. Knut and Alice Wallenberg Foundation