Cargando…

CAMSAP2-mediated noncentrosomal microtubule acetylation drives hepatocellular carcinoma metastasis

Rationale: Emerging evidence suggests that noncentrosomal microtubules play an essential role in intracellular transport, cell polarity and cell motility. Whether these noncentrosomal microtubules exist or function in cancer cells remains unclear. Methods: The expression and prognostic values of CAM...

Descripción completa

Detalles Bibliográficos
Autores principales: Li, Dongxiao, Ding, Xiangming, Xie, Meng, Huang, Zheng, Han, Ping, Tian, Dean, Xia, Limin
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Ivyspring International Publisher 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7069094/
https://www.ncbi.nlm.nih.gov/pubmed/32206120
http://dx.doi.org/10.7150/thno.42596
_version_ 1783505710926004224
author Li, Dongxiao
Ding, Xiangming
Xie, Meng
Huang, Zheng
Han, Ping
Tian, Dean
Xia, Limin
author_facet Li, Dongxiao
Ding, Xiangming
Xie, Meng
Huang, Zheng
Han, Ping
Tian, Dean
Xia, Limin
author_sort Li, Dongxiao
collection PubMed
description Rationale: Emerging evidence suggests that noncentrosomal microtubules play an essential role in intracellular transport, cell polarity and cell motility. Whether these noncentrosomal microtubules exist or function in cancer cells remains unclear. Methods: The expression and prognostic values of CAMSAP2 and its functional targets were analyzed by immunohistochemistry in two independent HCC cohorts. Immunofluorescence and co-immunoprecipitation were used for detection of CAMSAP2-decorated noncentrosomal microtubule. Chromatin immunoprecipitation and luciferase report assays were used to determine the c-Jun binding sites in HDAC6 promoter region. In vitro migration and invasion assays and in vivo orthotopic metastatic models were utilized to investigate invasion and metastasis. Results: We reported a microtubule minus‑end‑targeting protein, CAMSAP2, is significantly upregulated in hepatocellular carcinoma (HCC) and correlated with poor prognosis. CAMSAP2 was specifically deposited on microtubule minus ends to serve as a “seed” for noncentrosomal microtubule outgrowth in HCC cells. Upon depletion of CAMSAP2, the noncentrosomal microtubule array was transformed into a completely radial centrosomal pattern, thereby impairing HCC cell migration and invasion. We further demonstrated that CAMSAP2 cooperates with EB1 to regulate microtubule dynamics and invasive cell migration via Trio/Rac1 signaling. Strikingly, both immunofluorescence staining and western blotting showed that CAMSAP2 depletion strongly reduced the abundance of acetylated microtubules in HCC cells. Our results revealed that HDAC6, a promising target for cancer therapy, was inversely downregulated in HCC and uniquely endowed with tumor-suppressive activity by regulation CAMSAP2-mediated microtubule acetylation. Mechanistically, CAMSAP2 activates c-Jun to induce transrepression of HDAC6 through Trio-dependent Rac1/JNK pathway. Furthermore, NSC23766, a Rac1-specific inhibitor significantly inhibited CAMSAP2-mediated HCC invasion and metastasis. Conclusions: CAMSAP2 is functionally, mechanistically, and clinically oncogenic in HCC. Targeting CAMSAP2-mediated noncentrosomal microtubule acetylation may provide new therapeutic strategies for HCC metastasis.
format Online
Article
Text
id pubmed-7069094
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher Ivyspring International Publisher
record_format MEDLINE/PubMed
spelling pubmed-70690942020-03-23 CAMSAP2-mediated noncentrosomal microtubule acetylation drives hepatocellular carcinoma metastasis Li, Dongxiao Ding, Xiangming Xie, Meng Huang, Zheng Han, Ping Tian, Dean Xia, Limin Theranostics Research Paper Rationale: Emerging evidence suggests that noncentrosomal microtubules play an essential role in intracellular transport, cell polarity and cell motility. Whether these noncentrosomal microtubules exist or function in cancer cells remains unclear. Methods: The expression and prognostic values of CAMSAP2 and its functional targets were analyzed by immunohistochemistry in two independent HCC cohorts. Immunofluorescence and co-immunoprecipitation were used for detection of CAMSAP2-decorated noncentrosomal microtubule. Chromatin immunoprecipitation and luciferase report assays were used to determine the c-Jun binding sites in HDAC6 promoter region. In vitro migration and invasion assays and in vivo orthotopic metastatic models were utilized to investigate invasion and metastasis. Results: We reported a microtubule minus‑end‑targeting protein, CAMSAP2, is significantly upregulated in hepatocellular carcinoma (HCC) and correlated with poor prognosis. CAMSAP2 was specifically deposited on microtubule minus ends to serve as a “seed” for noncentrosomal microtubule outgrowth in HCC cells. Upon depletion of CAMSAP2, the noncentrosomal microtubule array was transformed into a completely radial centrosomal pattern, thereby impairing HCC cell migration and invasion. We further demonstrated that CAMSAP2 cooperates with EB1 to regulate microtubule dynamics and invasive cell migration via Trio/Rac1 signaling. Strikingly, both immunofluorescence staining and western blotting showed that CAMSAP2 depletion strongly reduced the abundance of acetylated microtubules in HCC cells. Our results revealed that HDAC6, a promising target for cancer therapy, was inversely downregulated in HCC and uniquely endowed with tumor-suppressive activity by regulation CAMSAP2-mediated microtubule acetylation. Mechanistically, CAMSAP2 activates c-Jun to induce transrepression of HDAC6 through Trio-dependent Rac1/JNK pathway. Furthermore, NSC23766, a Rac1-specific inhibitor significantly inhibited CAMSAP2-mediated HCC invasion and metastasis. Conclusions: CAMSAP2 is functionally, mechanistically, and clinically oncogenic in HCC. Targeting CAMSAP2-mediated noncentrosomal microtubule acetylation may provide new therapeutic strategies for HCC metastasis. Ivyspring International Publisher 2020-02-19 /pmc/articles/PMC7069094/ /pubmed/32206120 http://dx.doi.org/10.7150/thno.42596 Text en © The author(s) This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/). See http://ivyspring.com/terms for full terms and conditions.
spellingShingle Research Paper
Li, Dongxiao
Ding, Xiangming
Xie, Meng
Huang, Zheng
Han, Ping
Tian, Dean
Xia, Limin
CAMSAP2-mediated noncentrosomal microtubule acetylation drives hepatocellular carcinoma metastasis
title CAMSAP2-mediated noncentrosomal microtubule acetylation drives hepatocellular carcinoma metastasis
title_full CAMSAP2-mediated noncentrosomal microtubule acetylation drives hepatocellular carcinoma metastasis
title_fullStr CAMSAP2-mediated noncentrosomal microtubule acetylation drives hepatocellular carcinoma metastasis
title_full_unstemmed CAMSAP2-mediated noncentrosomal microtubule acetylation drives hepatocellular carcinoma metastasis
title_short CAMSAP2-mediated noncentrosomal microtubule acetylation drives hepatocellular carcinoma metastasis
title_sort camsap2-mediated noncentrosomal microtubule acetylation drives hepatocellular carcinoma metastasis
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7069094/
https://www.ncbi.nlm.nih.gov/pubmed/32206120
http://dx.doi.org/10.7150/thno.42596
work_keys_str_mv AT lidongxiao camsap2mediatednoncentrosomalmicrotubuleacetylationdriveshepatocellularcarcinomametastasis
AT dingxiangming camsap2mediatednoncentrosomalmicrotubuleacetylationdriveshepatocellularcarcinomametastasis
AT xiemeng camsap2mediatednoncentrosomalmicrotubuleacetylationdriveshepatocellularcarcinomametastasis
AT huangzheng camsap2mediatednoncentrosomalmicrotubuleacetylationdriveshepatocellularcarcinomametastasis
AT hanping camsap2mediatednoncentrosomalmicrotubuleacetylationdriveshepatocellularcarcinomametastasis
AT tiandean camsap2mediatednoncentrosomalmicrotubuleacetylationdriveshepatocellularcarcinomametastasis
AT xialimin camsap2mediatednoncentrosomalmicrotubuleacetylationdriveshepatocellularcarcinomametastasis