The TGFβ/Notch axis facilitates Müller cell-to-epithelial transition to ultimately form a chronic glial scar.

Conedera, Federica Maria; Quintela Pousa, Ana Maria; Mercader Huber, Nadia Isabel; Tschopp, Markus; Enzmann, Volker (2021). The TGFβ/Notch axis facilitates Müller cell-to-epithelial transition to ultimately form a chronic glial scar. Molecular neurodegeneration, 16(1), p. 69. BioMed Central 10.1186/s13024-021-00482-z

[img]
Preview
Text
s13024-021-00482-z.pdf - Published Version
Available under License Creative Commons: Attribution (CC-BY).

Download (8MB) | Preview

BACKGROUND

Contrasting with zebrafish, retinal regeneration from Müller cells (MCs) is largely limited in mammals, where they undergo reactive gliosis that consist of a hypertrophic response and ultimately results in vision loss. Transforming growth factor β (TGFβ) is essential for wound healing, including both scar formation and regeneration. However, targeting TGFβ may affect other physiological mechanisms, owing its pleiotropic nature. The regulation of various cellular activities by TGFβ relies on its interaction with other pathways including Notch. Here, we explore the interplay of TGFβ with Notch and how this regulates MC response to injury in zebrafish and mice. Furthermore, we aimed to characterize potential similarities between murine and human MCs during chronic reactive gliosis.

METHODS

Focal damage to photoreceptors was induced with a 532 nm diode laser in TgBAC (gfap:gfap-GFP) zebrafish (ZF) and B6-Tg (Rlbp1-GFP) mice. Transcriptomics, immunofluorescence, and flow cytometry were employed for a comparative analysis of MC response to laser-induced injury between ZF and mouse. The laser-induced injury was paired with pharmacological treatments to inhibit either Notch (DAPT) or TGFβ (Pirfenidone) or TGFβ/Notch interplay (SIS3). To determine if the murine laser-induced injury model translates to the human system, we compared the ensuing MC response to human donors with early retinal degeneration.

RESULTS

Investigations into injury-induced changes in murine MCs revealed TGFβ/Notch interplay during reactive gliosis. We found that TGFβ1/2 and Notch1/2 interact via Smad3 to reprogram murine MCs towards an epithelial lineage and ultimately to form a glial scar. Similar to what we observed in mice, we confirmed the epithelial phenotype of human Müller cells during gliotic response.

CONCLUSION

The study indicates a pivotal role for TGFβ/Notch interplay in tuning MC stemness during injury response and provides novel insights into the remodeling mechanism during retinal degenerative diseases.

Item Type:

Journal Article (Original Article)

Division/Institute:

04 Faculty of Medicine > Department of Head Organs and Neurology (DKNS) > Clinic of Ophthalmology
04 Faculty of Medicine > Pre-clinic Human Medicine > Institute of Anatomy
04 Faculty of Medicine > Pre-clinic Human Medicine > BioMedical Research (DBMR) > Forschungsbereich Augenklinik > Forschungsgruppe Augenheilkunde

Graduate School:

Graduate School for Cellular and Biomedical Sciences (GCB)

UniBE Contributor:

Conedera, Federica Maria, Quintela Pousa, Ana Maria, Mercader Huber, Nadia Isabel, Tschopp, Markus, Enzmann, Volker

Subjects:

600 Technology > 610 Medicine & health
500 Science > 570 Life sciences; biology

ISSN:

1750-1326

Publisher:

BioMed Central

Language:

English

Submitter:

Volker Enzmann

Date Deposited:

10 Jan 2022 11:47

Last Modified:

05 Dec 2022 15:56

Publisher DOI:

10.1186/s13024-021-00482-z

PubMed ID:

34593012

Uncontrolled Keywords:

Laser injury Müller cells Notch pathway Retinal degeneration Retinal regeneration Smad3 TGFβ signaling Vertebrates

BORIS DOI:

10.48350/162195

URI:

https://boris.unibe.ch/id/eprint/162195

Actions (login required)

Edit item Edit item
Provide Feedback