Mechanically sensitive HSF1 is a key regulator of left-right symmetry breaking in zebrafish embryos

Summary: The left-right symmetry breaking of vertebrate embryos requires nodal flow. However, the molecular mechanisms that mediate the asymmetric gene expression regulation under nodal flow remain elusive. Here, we report that heat shock factor 1 (HSF1) is asymmetrically activated in the Kupffer’s...

Full description

Bibliographic Details
Main Authors: Jing Du, Shu-Kai Li, Liu-Yuan Guan, Zheng Guo, Jiang-Fan Yin, Li Gao, Toru Kawanishi, Atsuko Shimada, Qiu-Ping Zhang, Li-Sha Zheng, Yi-Yao Liu, Xi-Qiao Feng, Lin Zhao, Dong-Yan Chen, Hiroyuki Takeda, Yu-Bo Fan
Format: Article
Language:English
Published: Elsevier 2023-10-01
Series:iScience
Subjects:
Online Access:http://www.sciencedirect.com/science/article/pii/S2589004223019417
_version_ 1797647831359553536
author Jing Du
Shu-Kai Li
Liu-Yuan Guan
Zheng Guo
Jiang-Fan Yin
Li Gao
Toru Kawanishi
Atsuko Shimada
Qiu-Ping Zhang
Li-Sha Zheng
Yi-Yao Liu
Xi-Qiao Feng
Lin Zhao
Dong-Yan Chen
Hiroyuki Takeda
Yu-Bo Fan
author_facet Jing Du
Shu-Kai Li
Liu-Yuan Guan
Zheng Guo
Jiang-Fan Yin
Li Gao
Toru Kawanishi
Atsuko Shimada
Qiu-Ping Zhang
Li-Sha Zheng
Yi-Yao Liu
Xi-Qiao Feng
Lin Zhao
Dong-Yan Chen
Hiroyuki Takeda
Yu-Bo Fan
author_sort Jing Du
collection DOAJ
description Summary: The left-right symmetry breaking of vertebrate embryos requires nodal flow. However, the molecular mechanisms that mediate the asymmetric gene expression regulation under nodal flow remain elusive. Here, we report that heat shock factor 1 (HSF1) is asymmetrically activated in the Kupffer’s vesicle of zebrafish embryos in the presence of nodal flow. Deficiency in HSF1 expression caused a significant situs inversus and disrupted gene expression asymmetry of nodal signaling proteins in zebrafish embryos. Further studies demonstrated that HSF1 is a mechanosensitive protein. The mechanical sensation ability of HSF1 is conserved in a variety of mechanical stimuli in different cell types. Moreover, cilia and Ca2+-Akt signaling axis are essential for the activation of HSF1 under mechanical stress in vitro and in vivo. Considering the conserved expression of HSF1 in organisms, these findings unveil a fundamental mechanism of gene expression regulation by mechanical clues during embryonic development and other physiological and pathological transformations.
first_indexed 2024-03-11T15:23:17Z
format Article
id doaj.art-909450b7fbb444c8a3993fb727560073
institution Directory Open Access Journal
issn 2589-0042
language English
last_indexed 2024-03-11T15:23:17Z
publishDate 2023-10-01
publisher Elsevier
record_format Article
series iScience
spelling doaj.art-909450b7fbb444c8a3993fb7275600732023-10-28T05:08:47ZengElsevieriScience2589-00422023-10-012610107864Mechanically sensitive HSF1 is a key regulator of left-right symmetry breaking in zebrafish embryosJing Du0Shu-Kai Li1Liu-Yuan Guan2Zheng Guo3Jiang-Fan Yin4Li Gao5Toru Kawanishi6Atsuko Shimada7Qiu-Ping Zhang8Li-Sha Zheng9Yi-Yao Liu10Xi-Qiao Feng11Lin Zhao12Dong-Yan Chen13Hiroyuki Takeda14Yu-Bo Fan15Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China; Institute of Biomechanics and Medical Engineering, Department of Mechanical Engineering, School of Aerospace, Tsinghua University, Beijing 100084, China; Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo 113-0033, Japan; Corresponding authorKey Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, ChinaInstitute of Biomechanics and Medical Engineering, Department of Mechanical Engineering, School of Aerospace, Tsinghua University, Beijing 100084, ChinaKey Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, ChinaCollege of life science, Hebei Normal University, Shijiazhuang 050024, ChinaCollege of life science, Hebei Normal University, Shijiazhuang 050024, ChinaDepartment of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo 113-0033, JapanDepartment of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo 113-0033, JapanTianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Department of Histology and Embryology, School of Medicine, Nankai University, Tianjin 300071, ChinaKey Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, ChinaDepartment of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, ChinaInstitute of Biomechanics and Medical Engineering, Department of Mechanical Engineering, School of Aerospace, Tsinghua University, Beijing 100084, ChinaTianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Department of Histology and Embryology, School of Medicine, Nankai University, Tianjin 300071, ChinaTianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Department of Histology and Embryology, School of Medicine, Nankai University, Tianjin 300071, China; Corresponding authorDepartment of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo 113-0033, Japan; Corresponding authorKey Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China; Corresponding authorSummary: The left-right symmetry breaking of vertebrate embryos requires nodal flow. However, the molecular mechanisms that mediate the asymmetric gene expression regulation under nodal flow remain elusive. Here, we report that heat shock factor 1 (HSF1) is asymmetrically activated in the Kupffer’s vesicle of zebrafish embryos in the presence of nodal flow. Deficiency in HSF1 expression caused a significant situs inversus and disrupted gene expression asymmetry of nodal signaling proteins in zebrafish embryos. Further studies demonstrated that HSF1 is a mechanosensitive protein. The mechanical sensation ability of HSF1 is conserved in a variety of mechanical stimuli in different cell types. Moreover, cilia and Ca2+-Akt signaling axis are essential for the activation of HSF1 under mechanical stress in vitro and in vivo. Considering the conserved expression of HSF1 in organisms, these findings unveil a fundamental mechanism of gene expression regulation by mechanical clues during embryonic development and other physiological and pathological transformations.http://www.sciencedirect.com/science/article/pii/S2589004223019417Molecular biologyCell biologyDevelopmental biologyEmbryology
spellingShingle Jing Du
Shu-Kai Li
Liu-Yuan Guan
Zheng Guo
Jiang-Fan Yin
Li Gao
Toru Kawanishi
Atsuko Shimada
Qiu-Ping Zhang
Li-Sha Zheng
Yi-Yao Liu
Xi-Qiao Feng
Lin Zhao
Dong-Yan Chen
Hiroyuki Takeda
Yu-Bo Fan
Mechanically sensitive HSF1 is a key regulator of left-right symmetry breaking in zebrafish embryos
iScience
Molecular biology
Cell biology
Developmental biology
Embryology
title Mechanically sensitive HSF1 is a key regulator of left-right symmetry breaking in zebrafish embryos
title_full Mechanically sensitive HSF1 is a key regulator of left-right symmetry breaking in zebrafish embryos
title_fullStr Mechanically sensitive HSF1 is a key regulator of left-right symmetry breaking in zebrafish embryos
title_full_unstemmed Mechanically sensitive HSF1 is a key regulator of left-right symmetry breaking in zebrafish embryos
title_short Mechanically sensitive HSF1 is a key regulator of left-right symmetry breaking in zebrafish embryos
title_sort mechanically sensitive hsf1 is a key regulator of left right symmetry breaking in zebrafish embryos
topic Molecular biology
Cell biology
Developmental biology
Embryology
url http://www.sciencedirect.com/science/article/pii/S2589004223019417
work_keys_str_mv AT jingdu mechanicallysensitivehsf1isakeyregulatorofleftrightsymmetrybreakinginzebrafishembryos
AT shukaili mechanicallysensitivehsf1isakeyregulatorofleftrightsymmetrybreakinginzebrafishembryos
AT liuyuanguan mechanicallysensitivehsf1isakeyregulatorofleftrightsymmetrybreakinginzebrafishembryos
AT zhengguo mechanicallysensitivehsf1isakeyregulatorofleftrightsymmetrybreakinginzebrafishembryos
AT jiangfanyin mechanicallysensitivehsf1isakeyregulatorofleftrightsymmetrybreakinginzebrafishembryos
AT ligao mechanicallysensitivehsf1isakeyregulatorofleftrightsymmetrybreakinginzebrafishembryos
AT torukawanishi mechanicallysensitivehsf1isakeyregulatorofleftrightsymmetrybreakinginzebrafishembryos
AT atsukoshimada mechanicallysensitivehsf1isakeyregulatorofleftrightsymmetrybreakinginzebrafishembryos
AT qiupingzhang mechanicallysensitivehsf1isakeyregulatorofleftrightsymmetrybreakinginzebrafishembryos
AT lishazheng mechanicallysensitivehsf1isakeyregulatorofleftrightsymmetrybreakinginzebrafishembryos
AT yiyaoliu mechanicallysensitivehsf1isakeyregulatorofleftrightsymmetrybreakinginzebrafishembryos
AT xiqiaofeng mechanicallysensitivehsf1isakeyregulatorofleftrightsymmetrybreakinginzebrafishembryos
AT linzhao mechanicallysensitivehsf1isakeyregulatorofleftrightsymmetrybreakinginzebrafishembryos
AT dongyanchen mechanicallysensitivehsf1isakeyregulatorofleftrightsymmetrybreakinginzebrafishembryos
AT hiroyukitakeda mechanicallysensitivehsf1isakeyregulatorofleftrightsymmetrybreakinginzebrafishembryos
AT yubofan mechanicallysensitivehsf1isakeyregulatorofleftrightsymmetrybreakinginzebrafishembryos