Cargando…

KPNA2 promotes angiogenesis by regulating STAT3 phosphorylation

PURPOSE: Angiogenesis is involved in many pathological and physiological processes and is mainly driven by hypoxia. Karyopherin subunit alpha 2 (KPNA2), a member of the nuclear transport protein family, was recently shown to be induced by hypoxia in various types of tumours, so we aimed to investiga...

Descripción completa

Detalles Bibliográficos
Autores principales: Jia, Yujie, Wang, Qi, Liang, Minglu, Huang, Kai
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9798605/
https://www.ncbi.nlm.nih.gov/pubmed/36578083
http://dx.doi.org/10.1186/s12967-022-03841-6
_version_ 1784860939537874944
author Jia, Yujie
Wang, Qi
Liang, Minglu
Huang, Kai
author_facet Jia, Yujie
Wang, Qi
Liang, Minglu
Huang, Kai
author_sort Jia, Yujie
collection PubMed
description PURPOSE: Angiogenesis is involved in many pathological and physiological processes and is mainly driven by hypoxia. Karyopherin subunit alpha 2 (KPNA2), a member of the nuclear transport protein family, was recently shown to be induced by hypoxia in various types of tumours, so we aimed to investigate the role and mechanism of KPNA2 in angiogenesis under hypoxia. MATERIALS AND METHODS: After overexpression or knockdown of KPNA2 in human umbilical vein endothelial cells (HUVEC) by adenovirus vector infection, the tube formation, proliferation and migration of HUVEC under hypoxia were detected by tubule formation assay, 5-ethynyl-2′-deoxyuridine (EdU) staining and Transwell assay, respectively. After overexpression or knockdown of KPNA2 in a murine hindlimb ischemia model by local injection of purified adenovirus vector into the gastrocnemius muscle, blood flow changes were examined with a laser Doppler system. Changes in KPNA2-binding proteins under hypoxia were detected by immunoprecipitation-mass spectrometry (IP-MS) and co-immunoprecipitation (Co-IP). The effect of KPNA2 on signal transducer and activator of transcription 3 (STAT3) was detected by Western blotting and quantitative RT‒PCR. RESULTS: KPNA2 was upregulated in the HUVEC hypoxia model and murine hindlimb ischemia model. Overexpression of KPNA2 increased the proliferation, migration and tube formation of HUVEC under hypoxia, while knockdown of KPNA2 reduced the proliferation, migration and tube formation of HUVEC. Overexpression of KPNA2 promoted the restoration of blood flow in the murine hindlimb ischemia model, while knockout of KPNA2 inhibited the restoration of blood flow in the murine hindlimb ischemia model. Mechanistically, hypoxia promoted the binding of STAT3 to KPNA2. Overexpression of KPNA2 promoted STAT3 phosphorylation and then upregulated vascular endothelial growth factor (VEGF) and angiopoietin 2(ANGPT2), whereas knockdown of KPNA2 inhibited STAT3 phosphorylation and then downregulated VEGF and ANGPT2. CONCLUSION: Our study demonstrates that hypoxia promotes the binding of STAT3 to KPNA2 and KPNA2 promotes angiogenesis under hypoxia by promoting the binding of STAT3 and JAK1 and regulating STAT3 phosphorylation. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12967-022-03841-6.
format Online
Article
Text
id pubmed-9798605
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-97986052022-12-30 KPNA2 promotes angiogenesis by regulating STAT3 phosphorylation Jia, Yujie Wang, Qi Liang, Minglu Huang, Kai J Transl Med Research PURPOSE: Angiogenesis is involved in many pathological and physiological processes and is mainly driven by hypoxia. Karyopherin subunit alpha 2 (KPNA2), a member of the nuclear transport protein family, was recently shown to be induced by hypoxia in various types of tumours, so we aimed to investigate the role and mechanism of KPNA2 in angiogenesis under hypoxia. MATERIALS AND METHODS: After overexpression or knockdown of KPNA2 in human umbilical vein endothelial cells (HUVEC) by adenovirus vector infection, the tube formation, proliferation and migration of HUVEC under hypoxia were detected by tubule formation assay, 5-ethynyl-2′-deoxyuridine (EdU) staining and Transwell assay, respectively. After overexpression or knockdown of KPNA2 in a murine hindlimb ischemia model by local injection of purified adenovirus vector into the gastrocnemius muscle, blood flow changes were examined with a laser Doppler system. Changes in KPNA2-binding proteins under hypoxia were detected by immunoprecipitation-mass spectrometry (IP-MS) and co-immunoprecipitation (Co-IP). The effect of KPNA2 on signal transducer and activator of transcription 3 (STAT3) was detected by Western blotting and quantitative RT‒PCR. RESULTS: KPNA2 was upregulated in the HUVEC hypoxia model and murine hindlimb ischemia model. Overexpression of KPNA2 increased the proliferation, migration and tube formation of HUVEC under hypoxia, while knockdown of KPNA2 reduced the proliferation, migration and tube formation of HUVEC. Overexpression of KPNA2 promoted the restoration of blood flow in the murine hindlimb ischemia model, while knockout of KPNA2 inhibited the restoration of blood flow in the murine hindlimb ischemia model. Mechanistically, hypoxia promoted the binding of STAT3 to KPNA2. Overexpression of KPNA2 promoted STAT3 phosphorylation and then upregulated vascular endothelial growth factor (VEGF) and angiopoietin 2(ANGPT2), whereas knockdown of KPNA2 inhibited STAT3 phosphorylation and then downregulated VEGF and ANGPT2. CONCLUSION: Our study demonstrates that hypoxia promotes the binding of STAT3 to KPNA2 and KPNA2 promotes angiogenesis under hypoxia by promoting the binding of STAT3 and JAK1 and regulating STAT3 phosphorylation. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12967-022-03841-6. BioMed Central 2022-12-28 /pmc/articles/PMC9798605/ /pubmed/36578083 http://dx.doi.org/10.1186/s12967-022-03841-6 Text en © The Author(s) 2022 https://creativecommons.org/licenses/by/4.0/Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) . The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/ (https://creativecommons.org/publicdomain/zero/1.0/) ) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Research
Jia, Yujie
Wang, Qi
Liang, Minglu
Huang, Kai
KPNA2 promotes angiogenesis by regulating STAT3 phosphorylation
title KPNA2 promotes angiogenesis by regulating STAT3 phosphorylation
title_full KPNA2 promotes angiogenesis by regulating STAT3 phosphorylation
title_fullStr KPNA2 promotes angiogenesis by regulating STAT3 phosphorylation
title_full_unstemmed KPNA2 promotes angiogenesis by regulating STAT3 phosphorylation
title_short KPNA2 promotes angiogenesis by regulating STAT3 phosphorylation
title_sort kpna2 promotes angiogenesis by regulating stat3 phosphorylation
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9798605/
https://www.ncbi.nlm.nih.gov/pubmed/36578083
http://dx.doi.org/10.1186/s12967-022-03841-6
work_keys_str_mv AT jiayujie kpna2promotesangiogenesisbyregulatingstat3phosphorylation
AT wangqi kpna2promotesangiogenesisbyregulatingstat3phosphorylation
AT liangminglu kpna2promotesangiogenesisbyregulatingstat3phosphorylation
AT huangkai kpna2promotesangiogenesisbyregulatingstat3phosphorylation