GRP75-driven, cell-cycle-dependent macropinocytosis of Tat/pDNA-Ca2+ nanoparticles underlies distinct gene therapy effect in ovarian cancer

Abstract Practice of tumor-targeted suicide gene therapy is hampered by unsafe and low efficient delivery of plasmid DNA (pDNA). Using HIV-Tat-derived peptide (Tat) to non-covalently form Tat/pDNA complexes advances the delivery performance. However, this innovative approach is still limited by intr...

Full description

Bibliographic Details
Main Authors: Linjia Su, Zhe Sun, Fangzheng Qi, Huishan Su, Luomeng Qian, Jing Li, Liang Zuo, Jinhai Huang, Zhilin Yu, Jinping Li, Zhinan Chen, Sihe Zhang
Format: Article
Language:English
Published: BMC 2022-07-01
Series:Journal of Nanobiotechnology
Subjects:
Online Access:https://doi.org/10.1186/s12951-022-01530-6
_version_ 1811277735230701568
author Linjia Su
Zhe Sun
Fangzheng Qi
Huishan Su
Luomeng Qian
Jing Li
Liang Zuo
Jinhai Huang
Zhilin Yu
Jinping Li
Zhinan Chen
Sihe Zhang
author_facet Linjia Su
Zhe Sun
Fangzheng Qi
Huishan Su
Luomeng Qian
Jing Li
Liang Zuo
Jinhai Huang
Zhilin Yu
Jinping Li
Zhinan Chen
Sihe Zhang
author_sort Linjia Su
collection DOAJ
description Abstract Practice of tumor-targeted suicide gene therapy is hampered by unsafe and low efficient delivery of plasmid DNA (pDNA). Using HIV-Tat-derived peptide (Tat) to non-covalently form Tat/pDNA complexes advances the delivery performance. However, this innovative approach is still limited by intracellular delivery efficiency and cell-cycle status. In this study, Tat/pDNA complexes were further condensed into smaller, nontoxic nanoparticles by Ca2+ addition. Formulated Tat/pDNA-Ca2+ nanoparticles mainly use macropinocytosis for intercellular delivery, and their macropinocytic uptake was persisted in mitosis (M-) phase and highly activated in DNA synthesis (S-) phase of cell-cycle. Over-expression or phosphorylation of a mitochondrial chaperone, 75-kDa glucose-regulated protein (GRP75), promoted monopolar spindle kinase 1 (MPS1)-controlled centrosome duplication and cell-cycle progress, but also driven cell-cycle-dependent macropinocytosis of Tat/pDNA-Ca2+ nanoparticles. Further in vivo molecular imaging based on DF (Fluc-eGFP)-TF (RFP-Rluc-HSV-ttk) system showed that Tat/pDNA-Ca2+ nanoparticles exhibited highly suicide gene therapy efficiency in mouse model xenografted with human ovarian cancer. Furthermore, arresting cell-cycle at S-phase markedly enhanced delivery performance of Tat/pDNA-Ca2+ nanoparticles, whereas targeting GRP75 reduced their macropinocytic delivery. More importantly, in vivo targeting GRP75 combined with cell-cycle or macropinocytosis inhibitors exhibited distinct suicide gene therapy efficiency. In summary, our data highlight that mitochondrial chaperone GRP75 moonlights as a biphasic driver underlying cell-cycle-dependent macropinocytosis of Tat/pDNA-Ca2+ nanoparticles in ovarian cancer.
first_indexed 2024-04-13T00:22:04Z
format Article
id doaj.art-47c34d7741b94ee0b201803d80bfcabd
institution Directory Open Access Journal
issn 1477-3155
language English
last_indexed 2024-04-13T00:22:04Z
publishDate 2022-07-01
publisher BMC
record_format Article
series Journal of Nanobiotechnology
spelling doaj.art-47c34d7741b94ee0b201803d80bfcabd2022-12-22T03:10:44ZengBMCJournal of Nanobiotechnology1477-31552022-07-0120112010.1186/s12951-022-01530-6GRP75-driven, cell-cycle-dependent macropinocytosis of Tat/pDNA-Ca2+ nanoparticles underlies distinct gene therapy effect in ovarian cancerLinjia Su0Zhe Sun1Fangzheng Qi2Huishan Su3Luomeng Qian4Jing Li5Liang Zuo6Jinhai Huang7Zhilin Yu8Jinping Li9Zhinan Chen10Sihe Zhang11Department of Cell Biology, School of Medicine, Nankai University, Nankai DistrictSchool of Life Sciences, Tianjin UniversityDepartment of Cell Biology, School of Medicine, Nankai University, Nankai DistrictDepartment of Cell Biology, School of Medicine, Nankai University, Nankai DistrictDepartment of Cell Biology, School of Medicine, Nankai University, Nankai DistrictDepartment of Cell Biology, School of Medicine, Nankai University, Nankai DistrictDepartment of Cell Biology, School of Medicine, Nankai University, Nankai DistrictSchool of Life Sciences, Tianjin UniversityState Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai UniversityDepartment of Medical Biochemistry and Microbiology, Uppsala UniversityNational Translational Science Center for Molecular Medicine, Department of Cell Biology, State Key Laboratory of Cancer Biology, Fourth Military Medical UniversityDepartment of Cell Biology, School of Medicine, Nankai University, Nankai DistrictAbstract Practice of tumor-targeted suicide gene therapy is hampered by unsafe and low efficient delivery of plasmid DNA (pDNA). Using HIV-Tat-derived peptide (Tat) to non-covalently form Tat/pDNA complexes advances the delivery performance. However, this innovative approach is still limited by intracellular delivery efficiency and cell-cycle status. In this study, Tat/pDNA complexes were further condensed into smaller, nontoxic nanoparticles by Ca2+ addition. Formulated Tat/pDNA-Ca2+ nanoparticles mainly use macropinocytosis for intercellular delivery, and their macropinocytic uptake was persisted in mitosis (M-) phase and highly activated in DNA synthesis (S-) phase of cell-cycle. Over-expression or phosphorylation of a mitochondrial chaperone, 75-kDa glucose-regulated protein (GRP75), promoted monopolar spindle kinase 1 (MPS1)-controlled centrosome duplication and cell-cycle progress, but also driven cell-cycle-dependent macropinocytosis of Tat/pDNA-Ca2+ nanoparticles. Further in vivo molecular imaging based on DF (Fluc-eGFP)-TF (RFP-Rluc-HSV-ttk) system showed that Tat/pDNA-Ca2+ nanoparticles exhibited highly suicide gene therapy efficiency in mouse model xenografted with human ovarian cancer. Furthermore, arresting cell-cycle at S-phase markedly enhanced delivery performance of Tat/pDNA-Ca2+ nanoparticles, whereas targeting GRP75 reduced their macropinocytic delivery. More importantly, in vivo targeting GRP75 combined with cell-cycle or macropinocytosis inhibitors exhibited distinct suicide gene therapy efficiency. In summary, our data highlight that mitochondrial chaperone GRP75 moonlights as a biphasic driver underlying cell-cycle-dependent macropinocytosis of Tat/pDNA-Ca2+ nanoparticles in ovarian cancer.https://doi.org/10.1186/s12951-022-01530-6MacropinocytosisCell-cycleGlucose-regulated proteinTat/pDNA-Ca2+ nanoparticleSuicide gene therapyOvarian cancer
spellingShingle Linjia Su
Zhe Sun
Fangzheng Qi
Huishan Su
Luomeng Qian
Jing Li
Liang Zuo
Jinhai Huang
Zhilin Yu
Jinping Li
Zhinan Chen
Sihe Zhang
GRP75-driven, cell-cycle-dependent macropinocytosis of Tat/pDNA-Ca2+ nanoparticles underlies distinct gene therapy effect in ovarian cancer
Journal of Nanobiotechnology
Macropinocytosis
Cell-cycle
Glucose-regulated protein
Tat/pDNA-Ca2+ nanoparticle
Suicide gene therapy
Ovarian cancer
title GRP75-driven, cell-cycle-dependent macropinocytosis of Tat/pDNA-Ca2+ nanoparticles underlies distinct gene therapy effect in ovarian cancer
title_full GRP75-driven, cell-cycle-dependent macropinocytosis of Tat/pDNA-Ca2+ nanoparticles underlies distinct gene therapy effect in ovarian cancer
title_fullStr GRP75-driven, cell-cycle-dependent macropinocytosis of Tat/pDNA-Ca2+ nanoparticles underlies distinct gene therapy effect in ovarian cancer
title_full_unstemmed GRP75-driven, cell-cycle-dependent macropinocytosis of Tat/pDNA-Ca2+ nanoparticles underlies distinct gene therapy effect in ovarian cancer
title_short GRP75-driven, cell-cycle-dependent macropinocytosis of Tat/pDNA-Ca2+ nanoparticles underlies distinct gene therapy effect in ovarian cancer
title_sort grp75 driven cell cycle dependent macropinocytosis of tat pdna ca2 nanoparticles underlies distinct gene therapy effect in ovarian cancer
topic Macropinocytosis
Cell-cycle
Glucose-regulated protein
Tat/pDNA-Ca2+ nanoparticle
Suicide gene therapy
Ovarian cancer
url https://doi.org/10.1186/s12951-022-01530-6
work_keys_str_mv AT linjiasu grp75drivencellcycledependentmacropinocytosisoftatpdnaca2nanoparticlesunderliesdistinctgenetherapyeffectinovariancancer
AT zhesun grp75drivencellcycledependentmacropinocytosisoftatpdnaca2nanoparticlesunderliesdistinctgenetherapyeffectinovariancancer
AT fangzhengqi grp75drivencellcycledependentmacropinocytosisoftatpdnaca2nanoparticlesunderliesdistinctgenetherapyeffectinovariancancer
AT huishansu grp75drivencellcycledependentmacropinocytosisoftatpdnaca2nanoparticlesunderliesdistinctgenetherapyeffectinovariancancer
AT luomengqian grp75drivencellcycledependentmacropinocytosisoftatpdnaca2nanoparticlesunderliesdistinctgenetherapyeffectinovariancancer
AT jingli grp75drivencellcycledependentmacropinocytosisoftatpdnaca2nanoparticlesunderliesdistinctgenetherapyeffectinovariancancer
AT liangzuo grp75drivencellcycledependentmacropinocytosisoftatpdnaca2nanoparticlesunderliesdistinctgenetherapyeffectinovariancancer
AT jinhaihuang grp75drivencellcycledependentmacropinocytosisoftatpdnaca2nanoparticlesunderliesdistinctgenetherapyeffectinovariancancer
AT zhilinyu grp75drivencellcycledependentmacropinocytosisoftatpdnaca2nanoparticlesunderliesdistinctgenetherapyeffectinovariancancer
AT jinpingli grp75drivencellcycledependentmacropinocytosisoftatpdnaca2nanoparticlesunderliesdistinctgenetherapyeffectinovariancancer
AT zhinanchen grp75drivencellcycledependentmacropinocytosisoftatpdnaca2nanoparticlesunderliesdistinctgenetherapyeffectinovariancancer
AT sihezhang grp75drivencellcycledependentmacropinocytosisoftatpdnaca2nanoparticlesunderliesdistinctgenetherapyeffectinovariancancer