2013
DOI: 10.1038/nature12681
|View full text |Cite
|
Sign up to set email alerts
|

Divergent angiocrine signals from vascular niche balance liver regeneration and fibrosis

Abstract: Summary Chemical or traumatic damage to the liver is frequently associated with aberrant healing(fibrosis) that overrides liver regeneration1–5. The mechanism by which hepatic niche cells differentially modulate regeneration and fibrosis during liver repair remains to be defined6–8. Hepatic vascular niche predominantly represented by liver sinusoidal endothelial cells (LSECs), deploys paracrine trophogens, known as angiocrine factors, to stimulate regeneration9–15. Nevertheless, it remains unknown how pro-rege… Show more

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
2
1

Citation Types

18
504
1
5

Year Published

2014
2014
2018
2018

Publication Types

Select...
5
3

Relationship

0
8

Authors

Journals

citations
Cited by 499 publications
(528 citation statements)
references
References 33 publications
18
504
1
5
Order By: Relevance
“…Because vascular density was not changed either upon endothelial-specific overexpression or deletion of KLF2, we hypothesized that KLF2 does not affect angiogenesis or endothelial cell survival, but regulates liver regeneration by controlling the endothelial secretome. Endothelial-mediated paracrine regulation of liver regeneration was shown to be critically controlled by the SDF-1 receptors CXCR4, which mediates the release of profibrotic cytokines, and CXCR7, which was shown to provide a proregenerative niche (4). Interestingly, we demonstrate that in vitro overexpression of KLF2 in ECs reciprocally regulates CXCR4 and CXCR7 expression on mRNA as well as protein level (Fig.…”
Section: Discussionmentioning
confidence: 66%
See 2 more Smart Citations
“…Because vascular density was not changed either upon endothelial-specific overexpression or deletion of KLF2, we hypothesized that KLF2 does not affect angiogenesis or endothelial cell survival, but regulates liver regeneration by controlling the endothelial secretome. Endothelial-mediated paracrine regulation of liver regeneration was shown to be critically controlled by the SDF-1 receptors CXCR4, which mediates the release of profibrotic cytokines, and CXCR7, which was shown to provide a proregenerative niche (4). Interestingly, we demonstrate that in vitro overexpression of KLF2 in ECs reciprocally regulates CXCR4 and CXCR7 expression on mRNA as well as protein level (Fig.…”
Section: Discussionmentioning
confidence: 66%
“…1 C and D). Hepatocyte proliferation was significantly elevated in CCL 4 -treated EC-KLF2-deficient mice compared with WT mice (Fig. 1E).…”
Section: Significancementioning
confidence: 89%
See 1 more Smart Citation
“…8 Furthermore, differential expression of CXCL12 and its receptors promotes profibrotic changes within the hepatic vascular niche during injury. 26 Finally, in prostate and colon cancers, CXCR4 expression by tumor cells signifies their metastatic potential, and hepatic metastases of these tumors demonstrate a predilection for CXCL12-expressing niches within the liver. 27e29 Despite numerous reports demonstrating immunohistochemical evidence of CXCL12 expression by cholangiocytes, we show that human and murine cholangiocytes do not appear to secrete or express CXCL12.…”
Section: Discussionmentioning
confidence: 99%
“…The different structural and metabolic requirements of organs and tissues prompt the formation of specialized blood vessels during development 1 , which deliver molecules that instruct tissues on their fates 2,3 . This process requires crosstalk between organs and the vasculature: organs generate local growth factors that attract blood vessels toward metabolically active regions, and the vessels release molecules (known as angiocrine signals) that affect cell differentiation in the developing organs.…”
mentioning
confidence: 99%