The University of Southampton
University of Southampton Institutional Repository

Widespread translational control of fibrosis in the human heart by RNA-binding proteins

Widespread translational control of fibrosis in the human heart by RNA-binding proteins
Widespread translational control of fibrosis in the human heart by RNA-binding proteins
Background:
Fibrosis is a common pathology in many cardiac disorders and is driven by the activation of resident fibroblasts. The global posttranscriptional mechanisms underlying fibroblast-to-myofibroblast conversion in the heart have not been explored.

Methods:
Genome-wide changes of RNA transcription and translation during human cardiac fibroblast activation were monitored with RNA sequencing and ribosome profiling. We then used RNA-binding protein-based analyses to identify translational regulators of fibrogenic genes. The integration with cardiac ribosome occupancy levels of 30 dilated cardiomyopathy patients demonstrates that these posttranscriptional mechanisms are also active in the diseased fibrotic human heart.

Results:
We generated nucleotide-resolution translatome data during the transforming growth factor β1-driven cellular transition of human cardiac fibroblasts to myofibroblasts. This identified dynamic changes of RNA transcription and translation at several time points during the fibrotic response, revealing transient and early-responder genes. Remarkably, about one-third of all changes in gene expression in activated fibroblasts are subject to translational regulation, and dynamic variation in ribosome occupancy affects protein abundance independent of RNA levels. Targets of RNA-binding proteins were strongly enriched in posttranscriptionally regulated genes, suggesting genes such as MBNL2 can act as translational activators or repressors. Ribosome occupancy in the hearts of patients with dilated cardiomyopathy suggested the same posttranscriptional regulatory network was underlying cardiac fibrosis. Key network hubs include RNA-binding proteins such as Pumilio RNA binding family member 2 (PUM2) and Quaking (QKI) that work in concert to regulate the translation of target transcripts in human diseased hearts. Furthermore, silencing of both PUM2 and QKI inhibits the transition of fibroblasts toward profibrotic myofibroblasts in response to transforming growth factor β1.

Conclusions:
We reveal widespread translational effects of transforming growth factor β1 and define novel posttranscriptional regulatory networks that control the fibroblast-to-myofibroblast transition. These networks are active in human heart disease, and silencing of hub genes limits fibroblast activation. Our findings show the central importance of translational control in fibrosis and highlight novel pathogenic mechanisms in heart failure.
0009-7322
937-951
Chothani, Sonia
24850611-01f3-46ae-af99-8c2693e6ca8f
Schäfer, Sebastian
c19c756b-f15d-4c9d-b415-f8dcfb05b2d9
Adami, Eleonora
c68076ed-059b-4f12-bb41-22f55de4cbae
Viswanathan, Sivakumar
3a7c01b3-ebb9-4329-96a6-f1b111a8a765
Widjaja, Anissa A.
11120825-c28e-4fe6-9831-8602ff1651d0
Langley, Sarah R.
897e76ce-ff32-43dc-abe6-6b347bad7fd1
Tan, Jessie
bb37d387-6f91-4a8c-b3a3-ff7c547e0075
Wang, Mao
fcf5d364-110d-4677-ac0f-84e6f1c7ff5c
Quaife, Nicholas M.
e4fbf8a0-15dc-4a31-a152-798b64465c92
Jian Pua, Chee
7a579c21-4a9d-4225-9d52-b827edeeb5e5
D’agostino, Giuseppe
dae9770f-b48c-48f5-b898-5483fef65cb9
Guna Shekeran, Shamini
cc73b94b-b230-4b81-8e1e-65b5d2890ab1
George, Benjamin L.
663ae7f3-5ecc-4ff3-9303-4759adbdecf8
Lim, Stella
15aa5f9e-2371-4248-84e7-40f03c5a9b89
Yiqun Cao, Elaine
f1c98fe8-3ab7-402f-97b9-1e8d23d6ff5c
Van Heesch, Sebastiaan
c1c1a412-c3f8-4947-b6e5-304abd5e60b9
Witte, Franziska
e31036f4-f7cc-4ca2-93e7-6b7bd3cb17f9
Felkin, Leanne E.
5324f950-1234-4c33-85d2-6d7be8555ee3
Christodoulou, Eleni G.
999f9e0f-ea83-4778-9544-2b46a49a1f7d
Dong, Jinrui
8bd11d54-eb9e-4e77-9635-91fb3a46f97c
Blachut, Susanne
2e40cff8-8067-4632-a680-21573d015d01
Patone, Giannino
90e32170-a40c-45f0-a4f4-a3eb9d6e82c0
Barton, Paul J.r.
240f673f-f2f1-466b-9f95-6410b41230f4
Hubner, Norbert
e01e1d75-f475-4d26-a3de-ba4150133c34
Cook, Stuart A.
2c9730f9-0c9b-4d81-a32b-2fa1180b8153
Rackham, Owen J.l.
8122eb1f-6e9f-4da5-90e1-ce108ccbbcbf
Chothani, Sonia
24850611-01f3-46ae-af99-8c2693e6ca8f
Schäfer, Sebastian
c19c756b-f15d-4c9d-b415-f8dcfb05b2d9
Adami, Eleonora
c68076ed-059b-4f12-bb41-22f55de4cbae
Viswanathan, Sivakumar
3a7c01b3-ebb9-4329-96a6-f1b111a8a765
Widjaja, Anissa A.
11120825-c28e-4fe6-9831-8602ff1651d0
Langley, Sarah R.
897e76ce-ff32-43dc-abe6-6b347bad7fd1
Tan, Jessie
bb37d387-6f91-4a8c-b3a3-ff7c547e0075
Wang, Mao
fcf5d364-110d-4677-ac0f-84e6f1c7ff5c
Quaife, Nicholas M.
e4fbf8a0-15dc-4a31-a152-798b64465c92
Jian Pua, Chee
7a579c21-4a9d-4225-9d52-b827edeeb5e5
D’agostino, Giuseppe
dae9770f-b48c-48f5-b898-5483fef65cb9
Guna Shekeran, Shamini
cc73b94b-b230-4b81-8e1e-65b5d2890ab1
George, Benjamin L.
663ae7f3-5ecc-4ff3-9303-4759adbdecf8
Lim, Stella
15aa5f9e-2371-4248-84e7-40f03c5a9b89
Yiqun Cao, Elaine
f1c98fe8-3ab7-402f-97b9-1e8d23d6ff5c
Van Heesch, Sebastiaan
c1c1a412-c3f8-4947-b6e5-304abd5e60b9
Witte, Franziska
e31036f4-f7cc-4ca2-93e7-6b7bd3cb17f9
Felkin, Leanne E.
5324f950-1234-4c33-85d2-6d7be8555ee3
Christodoulou, Eleni G.
999f9e0f-ea83-4778-9544-2b46a49a1f7d
Dong, Jinrui
8bd11d54-eb9e-4e77-9635-91fb3a46f97c
Blachut, Susanne
2e40cff8-8067-4632-a680-21573d015d01
Patone, Giannino
90e32170-a40c-45f0-a4f4-a3eb9d6e82c0
Barton, Paul J.r.
240f673f-f2f1-466b-9f95-6410b41230f4
Hubner, Norbert
e01e1d75-f475-4d26-a3de-ba4150133c34
Cook, Stuart A.
2c9730f9-0c9b-4d81-a32b-2fa1180b8153
Rackham, Owen J.l.
8122eb1f-6e9f-4da5-90e1-ce108ccbbcbf

Chothani, Sonia, Schäfer, Sebastian, Adami, Eleonora, Viswanathan, Sivakumar, Widjaja, Anissa A., Langley, Sarah R., Tan, Jessie, Wang, Mao, Quaife, Nicholas M., Jian Pua, Chee, D’agostino, Giuseppe, Guna Shekeran, Shamini, George, Benjamin L., Lim, Stella, Yiqun Cao, Elaine, Van Heesch, Sebastiaan, Witte, Franziska, Felkin, Leanne E., Christodoulou, Eleni G., Dong, Jinrui, Blachut, Susanne, Patone, Giannino, Barton, Paul J.r., Hubner, Norbert, Cook, Stuart A. and Rackham, Owen J.l. (2019) Widespread translational control of fibrosis in the human heart by RNA-binding proteins. Circulation, 140 (11), 937-951. (doi:10.1161/CIRCULATIONAHA.119.039596).

Record type: Article

Abstract

Background:
Fibrosis is a common pathology in many cardiac disorders and is driven by the activation of resident fibroblasts. The global posttranscriptional mechanisms underlying fibroblast-to-myofibroblast conversion in the heart have not been explored.

Methods:
Genome-wide changes of RNA transcription and translation during human cardiac fibroblast activation were monitored with RNA sequencing and ribosome profiling. We then used RNA-binding protein-based analyses to identify translational regulators of fibrogenic genes. The integration with cardiac ribosome occupancy levels of 30 dilated cardiomyopathy patients demonstrates that these posttranscriptional mechanisms are also active in the diseased fibrotic human heart.

Results:
We generated nucleotide-resolution translatome data during the transforming growth factor β1-driven cellular transition of human cardiac fibroblasts to myofibroblasts. This identified dynamic changes of RNA transcription and translation at several time points during the fibrotic response, revealing transient and early-responder genes. Remarkably, about one-third of all changes in gene expression in activated fibroblasts are subject to translational regulation, and dynamic variation in ribosome occupancy affects protein abundance independent of RNA levels. Targets of RNA-binding proteins were strongly enriched in posttranscriptionally regulated genes, suggesting genes such as MBNL2 can act as translational activators or repressors. Ribosome occupancy in the hearts of patients with dilated cardiomyopathy suggested the same posttranscriptional regulatory network was underlying cardiac fibrosis. Key network hubs include RNA-binding proteins such as Pumilio RNA binding family member 2 (PUM2) and Quaking (QKI) that work in concert to regulate the translation of target transcripts in human diseased hearts. Furthermore, silencing of both PUM2 and QKI inhibits the transition of fibroblasts toward profibrotic myofibroblasts in response to transforming growth factor β1.

Conclusions:
We reveal widespread translational effects of transforming growth factor β1 and define novel posttranscriptional regulatory networks that control the fibroblast-to-myofibroblast transition. These networks are active in human heart disease, and silencing of hub genes limits fibroblast activation. Our findings show the central importance of translational control in fibrosis and highlight novel pathogenic mechanisms in heart failure.

This record has no associated files available for download.

More information

Accepted/In Press date: 31 July 2019
Published date: 10 September 2019

Identifiers

Local EPrints ID: 444313
URI: http://eprints.soton.ac.uk/id/eprint/444313
ISSN: 0009-7322
PURE UUID: ffb02ab7-fc23-4511-9db5-aab6d9c1408e
ORCID for Owen J.l. Rackham: ORCID iD orcid.org/0000-0002-4390-0872

Catalogue record

Date deposited: 12 Oct 2020 16:31
Last modified: 17 Mar 2024 04:03

Export record

Altmetrics

Contributors

Author: Sonia Chothani
Author: Sebastian Schäfer
Author: Eleonora Adami
Author: Sivakumar Viswanathan
Author: Anissa A. Widjaja
Author: Sarah R. Langley
Author: Jessie Tan
Author: Mao Wang
Author: Nicholas M. Quaife
Author: Chee Jian Pua
Author: Giuseppe D’agostino
Author: Shamini Guna Shekeran
Author: Benjamin L. George
Author: Stella Lim
Author: Elaine Yiqun Cao
Author: Sebastiaan Van Heesch
Author: Franziska Witte
Author: Leanne E. Felkin
Author: Eleni G. Christodoulou
Author: Jinrui Dong
Author: Susanne Blachut
Author: Giannino Patone
Author: Paul J.r. Barton
Author: Norbert Hubner
Author: Stuart A. Cook

Download statistics

Downloads from ePrints over the past year. Other digital versions may also be available to download e.g. from the publisher's website.

View more statistics

Atom RSS 1.0 RSS 2.0

Contact ePrints Soton: eprints@soton.ac.uk

ePrints Soton supports OAI 2.0 with a base URL of http://eprints.soton.ac.uk/cgi/oai2

This repository has been built using EPrints software, developed at the University of Southampton, but available to everyone to use.

We use cookies to ensure that we give you the best experience on our website. If you continue without changing your settings, we will assume that you are happy to receive cookies on the University of Southampton website.

×