Human lncRNA <i>SUGCT-AS1</i> Regulates the Proinflammatory Response of Macrophage

Macrophages are the major primary immune cells that mediate the inflammatory response. In this process, long non-coding RNAs (lncRNAs) play an important, yet largely unknown role. Therefore, utilizing several publicly available RNA sequencing datasets, we predicted and selected lncRNAs that are diff...

Full description

Bibliographic Details
Main Authors: Yeong-Hwan Lim, Gwangho Yoon, Yeongseo Ryu, Dahee Jeong, Juhyun Song, Yong Sook Kim, Youngkeun Ahn, Hyun Kook, Young-Kook Kim
Format: Article
Language:English
Published: MDPI AG 2023-08-01
Series:International Journal of Molecular Sciences
Subjects:
Online Access:https://www.mdpi.com/1422-0067/24/17/13315
_version_ 1797582415363833856
author Yeong-Hwan Lim
Gwangho Yoon
Yeongseo Ryu
Dahee Jeong
Juhyun Song
Yong Sook Kim
Youngkeun Ahn
Hyun Kook
Young-Kook Kim
author_facet Yeong-Hwan Lim
Gwangho Yoon
Yeongseo Ryu
Dahee Jeong
Juhyun Song
Yong Sook Kim
Youngkeun Ahn
Hyun Kook
Young-Kook Kim
author_sort Yeong-Hwan Lim
collection DOAJ
description Macrophages are the major primary immune cells that mediate the inflammatory response. In this process, long non-coding RNAs (lncRNAs) play an important, yet largely unknown role. Therefore, utilizing several publicly available RNA sequencing datasets, we predicted and selected lncRNAs that are differentially expressed in M1 or M2 macrophages and involved in the inflammatory response. We identified <i>SUGCT-AS1</i>, which is a human macrophage-specific lncRNA whose expression is increased upon M1 macrophage stimulation. Conditioned media of <i>SUGCT-AS1</i>-depleted M1 macrophages induced an inflammatory phenotype of vascular smooth muscle cells, which included increased expression of inflammatory genes (<i>IL1B</i> and <i>IL6</i>), decreased contractile marker proteins (ACTA2 and SM22α), and increased cell migration. Depletion of <i>SUGCT-AS1</i> promoted the expression and secretion of proinflammatory cytokines, such as <i>TNF</i>, <i>IL1B</i>, and <i>IL6</i>, in M1 macrophages, and transcriptomic analysis showed that <i>SUGCT-AS1</i> has functions related to inflammatory responses and cytokines. Furthermore, we found that <i>SUGCT-AS1</i> directly binds to hnRNPU and regulates its nuclear–cytoplasmic translocation. This translocation of hnRNPU altered the proportion of the <i>MALT1</i> isoforms by regulating the alternative splicing of <i>MALT1</i>, a mediator of NF-κB signaling. Overall, our findings suggest that lncRNAs can be used for future studies on macrophage regulation. Moreover, they establish the <i>SUGCT-AS1</i>/hnRNPU/<i>MALT1</i> axis, which is a novel inflammatory regulatory mechanism in macrophages.
first_indexed 2024-03-10T23:21:53Z
format Article
id doaj.art-cbe709ab29624df9ac782836659a6e5f
institution Directory Open Access Journal
issn 1661-6596
1422-0067
language English
last_indexed 2024-03-10T23:21:53Z
publishDate 2023-08-01
publisher MDPI AG
record_format Article
series International Journal of Molecular Sciences
spelling doaj.art-cbe709ab29624df9ac782836659a6e5f2023-11-19T08:15:29ZengMDPI AGInternational Journal of Molecular Sciences1661-65961422-00672023-08-0124171331510.3390/ijms241713315Human lncRNA <i>SUGCT-AS1</i> Regulates the Proinflammatory Response of MacrophageYeong-Hwan Lim0Gwangho Yoon1Yeongseo Ryu2Dahee Jeong3Juhyun Song4Yong Sook Kim5Youngkeun Ahn6Hyun Kook7Young-Kook Kim8Basic Research Laboratory for Vascular Remodeling, Chonnam National University Medical School, Hwasun 58128, Republic of KoreaDivision of Brain Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju 28159, Republic of KoreaDepartment of Biochemistry, Chonnam National University Medical School, Hwasun 58128, Republic of KoreaDepartment of Biochemistry, Chonnam National University Medical School, Hwasun 58128, Republic of KoreaBioMedical Sciences Graduate Program (BMSGP), Chonnam National University, Hwasun 58128, Republic of KoreaBasic Research Laboratory for Vascular Remodeling, Chonnam National University Medical School, Hwasun 58128, Republic of KoreaBasic Research Laboratory for Vascular Remodeling, Chonnam National University Medical School, Hwasun 58128, Republic of KoreaBasic Research Laboratory for Vascular Remodeling, Chonnam National University Medical School, Hwasun 58128, Republic of KoreaBasic Research Laboratory for Vascular Remodeling, Chonnam National University Medical School, Hwasun 58128, Republic of KoreaMacrophages are the major primary immune cells that mediate the inflammatory response. In this process, long non-coding RNAs (lncRNAs) play an important, yet largely unknown role. Therefore, utilizing several publicly available RNA sequencing datasets, we predicted and selected lncRNAs that are differentially expressed in M1 or M2 macrophages and involved in the inflammatory response. We identified <i>SUGCT-AS1</i>, which is a human macrophage-specific lncRNA whose expression is increased upon M1 macrophage stimulation. Conditioned media of <i>SUGCT-AS1</i>-depleted M1 macrophages induced an inflammatory phenotype of vascular smooth muscle cells, which included increased expression of inflammatory genes (<i>IL1B</i> and <i>IL6</i>), decreased contractile marker proteins (ACTA2 and SM22α), and increased cell migration. Depletion of <i>SUGCT-AS1</i> promoted the expression and secretion of proinflammatory cytokines, such as <i>TNF</i>, <i>IL1B</i>, and <i>IL6</i>, in M1 macrophages, and transcriptomic analysis showed that <i>SUGCT-AS1</i> has functions related to inflammatory responses and cytokines. Furthermore, we found that <i>SUGCT-AS1</i> directly binds to hnRNPU and regulates its nuclear–cytoplasmic translocation. This translocation of hnRNPU altered the proportion of the <i>MALT1</i> isoforms by regulating the alternative splicing of <i>MALT1</i>, a mediator of NF-κB signaling. Overall, our findings suggest that lncRNAs can be used for future studies on macrophage regulation. Moreover, they establish the <i>SUGCT-AS1</i>/hnRNPU/<i>MALT1</i> axis, which is a novel inflammatory regulatory mechanism in macrophages.https://www.mdpi.com/1422-0067/24/17/13315long non-coding RNAs<i>SUGCT-AS1</i>hnRNPU<i>MALT1</i>macrophagevascular smooth muscle cells
spellingShingle Yeong-Hwan Lim
Gwangho Yoon
Yeongseo Ryu
Dahee Jeong
Juhyun Song
Yong Sook Kim
Youngkeun Ahn
Hyun Kook
Young-Kook Kim
Human lncRNA <i>SUGCT-AS1</i> Regulates the Proinflammatory Response of Macrophage
International Journal of Molecular Sciences
long non-coding RNAs
<i>SUGCT-AS1</i>
hnRNPU
<i>MALT1</i>
macrophage
vascular smooth muscle cells
title Human lncRNA <i>SUGCT-AS1</i> Regulates the Proinflammatory Response of Macrophage
title_full Human lncRNA <i>SUGCT-AS1</i> Regulates the Proinflammatory Response of Macrophage
title_fullStr Human lncRNA <i>SUGCT-AS1</i> Regulates the Proinflammatory Response of Macrophage
title_full_unstemmed Human lncRNA <i>SUGCT-AS1</i> Regulates the Proinflammatory Response of Macrophage
title_short Human lncRNA <i>SUGCT-AS1</i> Regulates the Proinflammatory Response of Macrophage
title_sort human lncrna i sugct as1 i regulates the proinflammatory response of macrophage
topic long non-coding RNAs
<i>SUGCT-AS1</i>
hnRNPU
<i>MALT1</i>
macrophage
vascular smooth muscle cells
url https://www.mdpi.com/1422-0067/24/17/13315
work_keys_str_mv AT yeonghwanlim humanlncrnaisugctas1iregulatestheproinflammatoryresponseofmacrophage
AT gwanghoyoon humanlncrnaisugctas1iregulatestheproinflammatoryresponseofmacrophage
AT yeongseoryu humanlncrnaisugctas1iregulatestheproinflammatoryresponseofmacrophage
AT daheejeong humanlncrnaisugctas1iregulatestheproinflammatoryresponseofmacrophage
AT juhyunsong humanlncrnaisugctas1iregulatestheproinflammatoryresponseofmacrophage
AT yongsookkim humanlncrnaisugctas1iregulatestheproinflammatoryresponseofmacrophage
AT youngkeunahn humanlncrnaisugctas1iregulatestheproinflammatoryresponseofmacrophage
AT hyunkook humanlncrnaisugctas1iregulatestheproinflammatoryresponseofmacrophage
AT youngkookkim humanlncrnaisugctas1iregulatestheproinflammatoryresponseofmacrophage