scispace - formally typeset
Open AccessJournal ArticleDOI

Interaction of reactive astrocytes with type I collagen induces astrocytic scar formation through the integrin–N-cadherin pathway after spinal cord injury

TLDR
In a mouse model of spinal cord injury, pharmacological blockade of reactive astrocyte–type I collagen interaction preventedAstrocytic scar formation, thereby leading to improved axonal regrowth and better functional outcomes, and reveal environmental cues regulating astROcytic fate decisions, thereby providing a potential therapeutic target for CNS injury.
Abstract
Central nervous system (CNS) injury transforms naive astrocytes into reactive astrocytes, which eventually become scar-forming astrocytes that can impair axonal regeneration and functional recovery. This sequential phenotypic change, known as reactive astrogliosis, has long been considered unidirectional and irreversible. However, we report here that reactive astrocytes isolated from injured spinal cord reverted in retrograde to naive astrocytes when transplanted into a naive spinal cord, whereas they formed astrocytic scars when transplanted into injured spinal cord, indicating the environment-dependent plasticity of reactive astrogliosis. We also found that type I collagen was highly expressed in the spinal cord during the scar-forming phase and induced astrocytic scar formation via the integrin-N-cadherin pathway. In a mouse model of spinal cord injury, pharmacological blockade of reactive astrocyte-type I collagen interaction prevented astrocytic scar formation, thereby leading to improved axonal regrowth and better functional outcomes. Our findings reveal environmental cues regulating astrocytic fate decisions, thereby providing a potential therapeutic target for CNS injury.

read more

Content maybe subject to copyright    Report

九州大学学術情報リポジト
Kyushu University Institutional Repository
Interaction of reactive astrocytes with type I
collagen induces astrocytic scar formation
through the integrin-N-cadherin pathway after
spinal cord injury
原, 正
http://hdl.handle.net/2324/1931776
出版情報:九州大学, 2017, 博士(医学), 課程博
バージョン:
権利関係:やむを得ない事由により本文ファイル非公開 (2)

1
1
Interaction of reactive astrocytes with collagen type I induces astrocytic scar formation 2
through the integrin/N-cadherin pathway after spinal cord injury 3
4
5
6
7
8
Authors: 9
Masamitsu Hara
1,2
, Kazu Kobayakawa
2
, Yasuyuki Ohkawa
3
, Hiromi Kumamaru
2
, Kazuya 10
Yokota
2
, Takeyuki Saito
1,2
, Ken Kijima
1,2
, Shingo Yoshizaki
1,2
, Katsumi Harimaya
2
, Yasuharu 11
Nakashima
2
and Seiji Okada
1,2
12
13
Affiliations: 14
1
Department of Advanced Medical Initiatives, Graduate School of Medical Sciences, Kyushu 15
University, Fukuoka, Japan 16
2
Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 17
Fukuoka, Japan 18
3
Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 19
Japan 20
Correspondence should be addressed to S.O. (seokada@ortho.med.kyushu-u.ac.jp) 21

2
Abstract 1
Central nervous system (CNS) injury activates naïve astrocytes into reactive astrocytes, which 2
eventually transform into scar-forming astrocytes that can impair axonal regeneration and 3
functional recovery. This sequential phenotypic change, known as reactive astrogliosis, has 4
long been considered unidirectional and irreversible. However, we report here that reactive 5
astrocytes isolated from injured spinal cords retrogradely reverted to naïve astrocytes when 6
transplanted into a naïve spinal cord, whereas they formed astrocytic scars when transplanted 7
into an injured spinal cord, indicating the environment-dependent reversibility of reactive 8
astrogliosis. We also found that collagen type I was highly expressed during the scar-forming 9
phase and induced astrocytic scar formation via the integrin/N-cadherin pathway. 10
Pharmacological blockade of reactive astrocyte-collagen type I interaction prevented astrocytic 11
scar formation, thereby leading to improved axonal regrowth and better functional outcomes 12
in a mouse model of spinal cord injury. Our findings reveal environmental cues regulating 13
astrocytic fate decisions, thereby providing a potential therapeutic target for CNS injury. 14

3
Introduction 1
Spinal cord injury (SCI) is a devastating trauma that causes persistent severe motor/sensory 2
dysfunction
1,2
. After SCI, astrocytes, the most abundant resident cells in the central nervous 3
system (CNS), play a crucial role in the SCI pathology through a phenotypic change known as 4
reactive astrogliosis. In this process, naïve astrocytes (NAs) sequentially exhibit opposite 5
phenotypes: first as reactive astrocytes (RAs) and then as scar-forming astrocytes (SAs). In the 6
subacute phase of SCI (4-14 days post-injury (dpi) in the mouse), RAs migrate to the lesion 7
epicenter and seclude inflammatory cells, leading to tissue repair and functional improvement 8
after SCI
2
. However, RAs gradually transform into SAs that form astrocytic scars, the main 9
impediment for CNS axonal regeneration, resulting in a limited functional recovery in the 10
chronic phase of SCI
3,4
(more than 14 dpi in the mouse). Astrocytic scars have been shown to 11
be irreversible and permanently inhibit axonal regrowth in both rodents and humans with SCI
4-
12
6
, although there is a different opinion that attenuating astrocytic scar formation failed to 13
promote axonal regeneration after SCI
7,8
. As such, clarifying the mechanism of astrocytic scar 14
formation and regulating this scar formation may be a potential therapeutic strategy for SCI. 15
Astrocytic scars have been studied for more than half a century
9,10
, and their formation has 16
been suggested to be regulated by complex and combinatorial inter- and intra-cellular signaling 17
mechanisms
3,5,11
. However, despite the large number of studies examining these astrocytic 18
changes
5,11,12
, the mechanism underlying astrocytic scar formation remains unclear. One factor 19
limiting basic research in this area is the lack of clear definitions of NAs, RAs, and SAs. A 20
conventional method for distinguishing between these cells is a histological analysis, but this 21
method is neither objective nor quantitative. 22
In this study, we established a clear distinction between NAs, RAs, and SAs based on marker 23
gene expression and investigated the regulatory mechanism underlying astrocytic scar 24

4
formation after SCI. 1

Citations
More filters
Journal ArticleDOI

The Biology of Regeneration Failure and Success After Spinal Cord Injury.

TL;DR: The perineuronal net and how chondroitin sulfate proteoglycans deposited in both the glial scar and net impede axonal outgrowth at the level of the growth cone are discussed.
Journal ArticleDOI

Moving beyond the glial scar for spinal cord repair.

TL;DR: Recent advances in understanding the composition and phenotypic characteristics of the spinal injury scar are discussed, the oversimplification of defining the scar in binary terms as good or bad, and the development of therapeutic approaches to target scar components to enable improved functional outcome after spinal cord injury are discussed.
Journal ArticleDOI

The diversity and disparity of the glial scar.

TL;DR: It is proposed that in-depth analysis of the functionally heterogeneous populations of reactive glia within the scar is needed to fully understand the glial scar’s dual nature.
Journal ArticleDOI

Neuroinflammation in the central nervous system: Symphony of glial cells.

TL;DR: This review will discuss the joint actions of glial cells in three phases of neuroinflammation, including initiation, progression, and prognosis, the three movements of the symphony, as the role of each type of glia cells in neuro inflammation depends on the nature of inflammatory cues and specific course of diseases.
Journal ArticleDOI

Reducing Pericyte-Derived Scarring Promotes Recovery after Spinal Cord Injury

TL;DR: In conclusion, attenuation of pericyte-derived fibrosis represents a promising therapeutic approach to facilitate recovery following CNS injury.
References
More filters
Journal ArticleDOI

Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2

TL;DR: This work presents DESeq2, a method for differential analysis of count data, using shrinkage estimation for dispersions and fold changes to improve stability and interpretability of estimates, which enables a more quantitative analysis focused on the strength rather than the mere presence of differential expression.
Journal ArticleDOI

Fast gapped-read alignment with Bowtie 2

TL;DR: Bowtie 2 combines the strengths of the full-text minute index with the flexibility and speed of hardware-accelerated dynamic programming algorithms to achieve a combination of high speed, sensitivity and accuracy.
Posted ContentDOI

Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2

TL;DR: This work presents DESeq2, a method for differential analysis of count data, using shrinkage estimation for dispersions and fold changes to improve stability and interpretability of estimates, which enables a more quantitative analysis focused on the strength rather than the mere presence of differential expression.
Journal ArticleDOI

Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks

TL;DR: This protocol begins with raw sequencing reads and produces a transcriptome assembly, lists of differentially expressed and regulated genes and transcripts, and publication-quality visualizations of analysis results, which takes less than 1 d of computer time for typical experiments and ∼1 h of hands-on time.
Journal ArticleDOI

DAVID: Database for Annotation, Visualization, and Integrated Discovery

TL;DR: DAMID is a web-accessible program that integrates functional genomic annotations with intuitive graphical summaries that assists in the interpretation of genome-scale datasets by facilitating the transition from data collection to biological meaning.
Related Papers (5)