1
|
Sato Y, Habara M, Hanaki S, Masaki T, Tomiyasu H, Miki Y, Sakurai M, Morimoto M, Kobayashi D, Miyamoto T, Shimada M. Calcineurin-mediated dephosphorylation stabilizes E2F1 protein by suppressing binding of the FBXW7 ubiquitin ligase subunit. Proc Natl Acad Sci U S A 2024; 121:e2414618121. [PMID: 39361641 PMCID: PMC11474076 DOI: 10.1073/pnas.2414618121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 08/26/2024] [Indexed: 10/05/2024] Open
Abstract
The transcription factor E2F1 serves as a regulator of the cell cycle and promotes cell proliferation. It is highly expressed in cancer tissues and contributes to their malignant transformation. Degradation by the ubiquitin-proteasome system may help to prevent such overexpression of E2F1 and thereby to suppress carcinogenesis. A detailed understanding of the mechanisms underlying E2F1 degradation may therefore inform the development of new cancer treatments. We here identified SCFFBXW7 as a ubiquitin ligase for E2F1 by comprehensive analysis. We found that phosphorylation of E2F1 at serine-403 promotes its binding to FBXW7 (F-box/WD repeat-containing protein 7) followed by its ubiquitination and degradation. Furthermore, calcineurin, a Ca2+/calmodulin-dependent serine-threonine phosphatase, was shown to stabilize E2F1 by mediating its dephosphorylation at serine-403 and thereby preventing FBXW7 binding. Treatment of cells with Ca2+ channel blockers resulted in downregulation of both E2F1 protein and the expression of E2F1 target genes, whereas treatment with the Ca2+ ionophore ionomycin induced upregulation of E2F1. Finally, the calcineurin inhibitor FK506 attenuated xenograft tumor growth in mice in association with downregulation of E2F1 in the tumor tissue. Impairment of the balance between the opposing actions of FBXW7 and calcineurin in the regulation of E2F1 abundance may therefore play an important role in carcinogenesis.
Collapse
Affiliation(s)
- Yuki Sato
- Department of Veterinary Biochemistry, Yamaguchi University, Yamaguchi753-8511, Japan
| | - Makoto Habara
- Department of Veterinary Biochemistry, Yamaguchi University, Yamaguchi753-8511, Japan
| | - Shunsuke Hanaki
- Department of Veterinary Biochemistry, Yamaguchi University, Yamaguchi753-8511, Japan
| | - Takahiro Masaki
- Department of Veterinary Biochemistry, Yamaguchi University, Yamaguchi753-8511, Japan
| | - Haruki Tomiyasu
- Department of Veterinary Biochemistry, Yamaguchi University, Yamaguchi753-8511, Japan
| | - Yosei Miki
- Department of Veterinary Biochemistry, Yamaguchi University, Yamaguchi753-8511, Japan
| | - Masashi Sakurai
- Department of Veterinary Pathology, Yamaguchi University, Yamaguchi753-8511, Japan
| | - Masahiro Morimoto
- Department of Veterinary Pathology, Yamaguchi University, Yamaguchi753-8511, Japan
| | - Daigo Kobayashi
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Yamaguchi University, Ube, Yamaguchi 755-8505, Japan
| | - Tatsuo Miyamoto
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Yamaguchi University, Ube, Yamaguchi 755-8505, Japan
- Division of Advanced Genome Editing Therapy Research, Research Institute for Cell Design Medical Science, Yamaguchi University, Ube, Yamaguchi 755-8505, Japan
| | - Midori Shimada
- Department of Veterinary Biochemistry, Yamaguchi University, Yamaguchi753-8511, Japan
- Department of Molecular Biology, Graduate School of Medicine, Nagoya University, Nagoya, Aichi 466-8550, Japan
| |
Collapse
|
2
|
Sato Y, Habara M, Hanaki S, Sharif J, Tomiyasu H, Miki Y, Shimada M. Calcineurin/NFATc1 pathway represses cellular cytotoxicity by modulating histone H3 expression. Sci Rep 2024; 14:14732. [PMID: 38926604 PMCID: PMC11208570 DOI: 10.1038/s41598-024-65769-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 06/24/2024] [Indexed: 06/28/2024] Open
Abstract
Excess amounts of histones in the cell induce mitotic chromosome loss and genomic instability, and are therefore detrimental to cell survival. In yeast, excess histones are degraded by the proteasome mediated via the DNA damage response factor Rad53. Histone expression, therefore, is tightly regulated at the protein level. Our understanding of the transcriptional regulation of histone genes is far from complete. In this study, we found that calcineurin inhibitor treatment increased histone protein levels, and that the transcription factor NFATc1 (nuclear factor of activated T cells 1) repressed histone transcription and acts downstream of the calcineurin. We further revealed that NFATc1 binds to the promoter regions of many histone genes and that histone transcription is downregulated in a manner dependent on intracellular calcium levels. Indeed, overexpression of histone H3 markedly inhibited cell proliferation. Taken together, these findings suggest that NFATc1 prevents the detrimental effects of histone H3 accumulation by inhibiting expression of histone at the transcriptional level.
Collapse
Affiliation(s)
- Yuki Sato
- Department of Veterinary Biochemistry, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8511, Japan
| | - Makoto Habara
- Department of Veterinary Biochemistry, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8511, Japan
| | - Shunsuke Hanaki
- Department of Veterinary Biochemistry, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8511, Japan
| | - Jafar Sharif
- Developmental Genetics Group, Center for Integrative Medical Sciences (IMS), RIKEN, 1-7-22 Suehiro, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
| | - Haruki Tomiyasu
- Department of Veterinary Biochemistry, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8511, Japan
| | - Yosei Miki
- Department of Veterinary Biochemistry, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8511, Japan
| | - Midori Shimada
- Department of Veterinary Biochemistry, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8511, Japan.
- Department of Molecular Biology, Graduate School of Medicine, Nagoya University, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| |
Collapse
|
3
|
Fonódi M, Nagy L, Boratkó A. Role of Protein Phosphatases in Tumor Angiogenesis: Assessing PP1, PP2A, PP2B and PTPs Activity. Int J Mol Sci 2024; 25:6868. [PMID: 38999976 PMCID: PMC11241275 DOI: 10.3390/ijms25136868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 07/14/2024] Open
Abstract
Tumor angiogenesis, the formation of new blood vessels to support tumor growth and metastasis, is a complex process regulated by a multitude of signaling pathways. Dysregulation of signaling pathways involving protein kinases has been extensively studied, but the role of protein phosphatases in angiogenesis within the tumor microenvironment remains less explored. However, among angiogenic pathways, protein phosphatases play critical roles in modulating signaling cascades. This review provides a comprehensive overview of the involvement of protein phosphatases in tumor angiogenesis, highlighting their diverse functions and mechanisms of action. Protein phosphatases are key regulators of cellular signaling pathways by catalyzing the dephosphorylation of proteins, thereby modulating their activity and function. This review aims to assess the activity of the protein tyrosine phosphatases and serine/threonine phosphatases. These phosphatases exert their effects on angiogenic signaling pathways through various mechanisms, including direct dephosphorylation of angiogenic receptors and downstream signaling molecules. Moreover, protein phosphatases also crosstalk with other signaling pathways involved in angiogenesis, further emphasizing their significance in regulating tumor vascularization, including endothelial cell survival, sprouting, and vessel maturation. In conclusion, this review underscores the pivotal role of protein phosphatases in tumor angiogenesis and accentuate their potential as therapeutic targets for anti-angiogenic therapy in cancer.
Collapse
Affiliation(s)
| | | | - Anita Boratkó
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary; (M.F.); (L.N.)
| |
Collapse
|
4
|
Wang H, Shi Y, Zhou X, Zhang L, Yang A, Zhou D, Ma T. HNRNPA2B1 stabilizes NFATC3 levels to potentiate its combined actions with FOSL1 to mediate vasculogenic mimicry in GBM cells. Cell Biol Toxicol 2024; 40:44. [PMID: 38862832 PMCID: PMC11166796 DOI: 10.1007/s10565-024-09890-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 06/03/2024] [Indexed: 06/13/2024]
Abstract
BACKGROUND Vasculogenic mimicry (VM) is an enigmatic physiological feature that influences blood supply within glioblastoma (GBM) tumors for their sustained growth. Previous studies identify NFATC3, FOSL1 and HNRNPA2B1 as significant mediators of VEGFR2, a key player in vasculogenesis, and their molecular relationships may be crucial for VM in GBM. AIMS The aim of this study was to understand how NFATC3, FOSL1 and HNRNPA2B1 collectively influence VM in GBM. METHODS We have investigated the underlying gene regulatory mechanisms for VM in GBM cell lines U251 and U373 in vitro and in vivo. In vitro cell-based assays were performed to explore the role of NFATC3, FOSL1 and HNRNPA2B1 in GBM cell proliferation, VM and migration, in the context of RNA interference (RNAi)-mediated knockdown alongside corresponding controls. Western blotting and qRT-PCR assays were used to examine VEGFR2 expression levels. CO-IP was employed to detect protein-protein interactions, ChIP was used to detect DNA-protein complexes, and RIP was used to detect RNA-protein complexes. Histochemical staining was used to detect VM tube formation in vivo. RESULTS Focusing on NFATC3, FOSL1 and HNRNPA2B1, we found each was significantly upregulated in GBM and positively correlated with VM-like cellular behaviors in U251 and U373 cell lines. Knockdown of NFATC3, FOSL1 or HNRNPA2B1 each resulted in decreased levels of VEGFR2, a key growth factor gene that drives VM, as well as the inhibition of proliferation, cell migration and extracorporeal VM activity. Chromatin immunoprecipitation (ChIP) studies and luciferase reporter gene assays revealed that NFATC3 binds to the promoter region of VEGFR2 to enhance VEGFR2 gene expression. Notably, FOSL1 interacts with NFATC3 as a co-factor to potentiate the DNA-binding capacity of NFATC3, resulting in enhanced VM-like cellular behaviors. Also, level of NFATC3 protein in cells was enhanced through HNRNPA2B1 binding of NFATC3 mRNA. Furthermore, RNAi-mediated silencing of NFATC3, FOSL1 and HNRNPA2B1 in GBM cells reduced their capacity for tumor formation and VM-like behaviors in vivo. CONCLUSION Taken together, our findings identify NFATC3 as an important mediator of GBM tumor growth through its molecular and epistatic interactions with HNRNPA2B1 and FOSL1 to influence VEGFR2 expression and VM-like cellular behaviors.
Collapse
Affiliation(s)
- Hanting Wang
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China
| | - Yiwen Shi
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China
| | - Xinxin Zhou
- Liaoning University of Traditional Chinese Medicine, Shenyang, 110034, China
| | - Lu Zhang
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China
| | - Aodan Yang
- The First Clinical College of China Medical University, Shenyang, 110002, China
| | - Dabo Zhou
- School and Hospital of Stomatology, China Medical University, Shenyang, 110002, China.
| | - Teng Ma
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China.
| |
Collapse
|
5
|
Tomiyasu H, Habara M, Hanaki S, Sato Y, Miki Y, Shimada M. FOXO1 promotes cancer cell growth through MDM2-mediated p53 degradation. J Biol Chem 2024; 300:107209. [PMID: 38519029 PMCID: PMC11021968 DOI: 10.1016/j.jbc.2024.107209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 03/07/2024] [Accepted: 03/13/2024] [Indexed: 03/24/2024] Open
Abstract
FOXO1 is a transcription factor and potential tumor suppressor that is negatively regulated downstream of PI3K-PKB/AKT signaling. Paradoxically, FOXO also promotes tumor growth, but the detailed mechanisms behind this role of FOXO are not fully understood. In this study, we revealed a molecular cascade by which the Thr24 residue of FOXO1 is phosphorylated by AKT and is dephosphorylated by calcineurin, which is a Ca2+-dependent protein phosphatase. Curiously, single nucleotide somatic mutations of FOXO1 in cancer occur frequently at and near Thr24. Using a calcineurin inhibitor and shRNA directed against calcineurin, we revealed that calcineurin-mediated dephosphorylation of Thr24 regulates FOXO1 protein stability. We also found that FOXO1 binds to the promoter region of MDM2 and activates transcription, which in turn promotes MDM2-mediated ubiquitination and degradation of p53. FOXO3a and FOXO4 are shown to control p53 activity; however, the significance of FOXO1 in p53 regulation remains largely unknown. Supporting this notion, FOXO1 depletion increased p53 and p21 protein levels in association with the inhibition of cell proliferation. Taken together, these results indicate that FOXO1 is stabilized by calcineurin-mediated dephosphorylation and that FOXO1 supports cancer cell proliferation by promoting MDM2 transcription and subsequent p53 degradation.
Collapse
Affiliation(s)
- Haruki Tomiyasu
- Department of Veterinary Biochemistry, Yamaguchi University, Yamaguchi, Yamaguchi, Japan
| | - Makoto Habara
- Department of Veterinary Biochemistry, Yamaguchi University, Yamaguchi, Yamaguchi, Japan
| | - Shunsuke Hanaki
- Department of Veterinary Biochemistry, Yamaguchi University, Yamaguchi, Yamaguchi, Japan
| | - Yuki Sato
- Department of Veterinary Biochemistry, Yamaguchi University, Yamaguchi, Yamaguchi, Japan
| | - Yosei Miki
- Department of Veterinary Biochemistry, Yamaguchi University, Yamaguchi, Yamaguchi, Japan
| | - Midori Shimada
- Department of Veterinary Biochemistry, Yamaguchi University, Yamaguchi, Yamaguchi, Japan; Department of Molecular Biology, Nagoya University, Graduate School of Medicine, Showa-ku, Nagoya, Japan.
| |
Collapse
|
6
|
Hanaki S, Habara M, Sato Y, Tomiyasu H, Miki Y, Shibutani S, Shimada M. Dephosphorylation of NFAT by Calcineurin inhibits Skp2-mediated degradation. J Biochem 2024; 175:235-244. [PMID: 38030387 DOI: 10.1093/jb/mvad103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 11/26/2023] [Indexed: 12/01/2023] Open
Abstract
The transcription factor NFAT plays key roles in multiple biological activities, such as immune responses, tissue development and malignant transformation. NFAT is dephosphorylated by calcineurin, which is activated by intracellular calcium levels, and translocated into the nucleus, resulting in transcriptional activation. Calcineurin dephosphorylates various target proteins and regulates their functions. However, the regulation of NFAT degradation is largely unknown, and it is unclear whether calcineurin contributes to the stability of NFAT. We investigated the effect of calcineurin inhibition on NFAT protein stability and found that the dephosphorylation of NFAT by calcineurin promotes the NFAT stabilization, whereas calcineurin mutant that is defective in phosphatase activity was unable to stabilize NFAT. Increased intracellular calcium ion concentration, which is essential for calcineurin activation, also induced NFAT stability. In addition, we identified S-phase kinase associated protein 2 (Skp2), an F-box protein of the SCF ubiquitin ligase complex, as a factor mediating degradation of NFAT when calcineurin was depleted. In summary, these findings revealed that the dephosphorylation of NFAT by calcineurin protects NFAT from degradation by Skp2 and promotes its protein stability.
Collapse
Affiliation(s)
- Shunsuke Hanaki
- Department of Veterinary Biochemistry, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, Yamaguchi, 753-8511, Japan
| | - Makoto Habara
- Department of Veterinary Biochemistry, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, Yamaguchi, 753-8511, Japan
| | - Yuki Sato
- Department of Veterinary Biochemistry, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, Yamaguchi, 753-8511, Japan
| | - Haruki Tomiyasu
- Department of Veterinary Biochemistry, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, Yamaguchi, 753-8511, Japan
| | - Yosei Miki
- Department of Veterinary Biochemistry, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, Yamaguchi, 753-8511, Japan
| | - Shusaku Shibutani
- Laboratory of Veterinary Hygiene, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, Yamaguchi, 753-8511, Japan
| | - Midori Shimada
- Department of Veterinary Biochemistry, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, Yamaguchi, 753-8511, Japan
- Department of Molecular Biology, Nagoya University, Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| |
Collapse
|
7
|
Masaki T, Habara M, Hanaki S, Sato Y, Tomiyasu H, Miki Y, Shimada M. Calcineurin-mediated dephosphorylation enhances the stability and transactivation of c-Myc. Sci Rep 2023; 13:13116. [PMID: 37573463 PMCID: PMC10423207 DOI: 10.1038/s41598-023-40412-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 08/09/2023] [Indexed: 08/14/2023] Open
Abstract
c-Myc, a transcription factor, induces cell proliferation and is often aberrantly or highly expressed in cancers. However, molecular mechanisms underlying this aberrantly high expression remain unclear. Here, we found that intracellular Ca2+ concentration regulates c-Myc oncoprotein stability. We identified that calcineurin, a Ca2+-dependent protein phosphatase, is a positive regulator of c-Myc expression. Calcineurin depletion suppresses c-Myc targeted gene expression and c-Myc degradation. Calcineurin directly dephosphorylates Thr58 and Ser62 in c-Myc, which inhibit binding to the ubiquitin ligase Fbxw7. Mutations within the autoinhibitory domain of calcineurin, most frequently observed in cancer, may increase phosphatase activity, increasing c-Myc transcriptional activity in turn. Notably, calcineurin inhibition with FK506 decreased c-Myc expression with enhanced Thr58 and Ser62 phosphorylation in a mouse xenograft model. Thus, calcineurin can stabilize c-Myc, promoting tumor progression. Therefore, we propose that Ca2+ signaling dysfunction affects cancer-cell proliferation via increased c-Myc stability and that calcineurin inhibition could be a new therapeutic target of c-Myc-overexpressing cancers.
Collapse
Affiliation(s)
- Takahiro Masaki
- Department of Veterinary Biochemistry, Joint Faculty of Veterinary Science, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8511, Japan
| | - Makoto Habara
- Department of Veterinary Biochemistry, Joint Faculty of Veterinary Science, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8511, Japan
| | - Shunsuke Hanaki
- Department of Veterinary Biochemistry, Joint Faculty of Veterinary Science, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8511, Japan
| | - Yuki Sato
- Department of Veterinary Biochemistry, Joint Faculty of Veterinary Science, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8511, Japan
| | - Haruki Tomiyasu
- Department of Veterinary Biochemistry, Joint Faculty of Veterinary Science, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8511, Japan
| | - Yosei Miki
- Department of Veterinary Biochemistry, Joint Faculty of Veterinary Science, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8511, Japan
| | - Midori Shimada
- Department of Veterinary Biochemistry, Joint Faculty of Veterinary Science, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8511, Japan.
| |
Collapse
|
8
|
METTL3 Promotes Endothelium-Mesenchymal Transition of Pulmonary Artery Endothelial Cells by Regulating TRPC6/Calcineurin/NFAT Signaling Pathways. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2023; 2023:8269356. [PMID: 36865750 PMCID: PMC9974285 DOI: 10.1155/2023/8269356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 01/17/2023] [Accepted: 02/02/2023] [Indexed: 02/25/2023]
Abstract
Background Endothelium-mesenchymal transition (EndMT) is a process of phenotypic and functional transition from activated endothelial cells to mesenchymal cells. Recently, EndMT has been proved to be one of the main pathological mechanisms of pulmonary artery hypertension (PAH). However, the molecular mechanism is not clear. Methods Primary rat pulmonary arterial endothelial cells (rPAECs) were isolated from Sprague-Dawley rats and verified by CD31 immunofluorescence staining. rPAECs were exposed to hypoxic conditions to induce EndMT. RNA and protein levels in cells were detected by RT-qPCR and Western blot. The migration ability was verified by the transwell assay. The RIP experiment was used to test the m6A modification of TRPC6 mRNA and the binding relationship between TRPC6 and METTL3. Calcineurin/NFAT signaling was measured by using commercial kits. Results METTL3 was found to be highly expressed by hypoxia treatment in a time-dependent manner. Knockdown of METTL3 significantly suppressed cell migration, downregulated the levels of interstitial cell-related markers like α-SMA and vimentin, and increased the levels of endothelial cell markers including CD31 and VE-cadherin. Mechanistically, METTL3 increased TRPC6 expression by enhancing the m6A modification of TRPC6 mRNA, thus activating calcineurin/NFAT signaling. Our experiments showed that METTL3 silencing mediated the inhibitory roles in the hypoxia-mediated EndMT process, which were significantly reversed by TRPC6/calcineurin/NFAT signaling activation. Conclusion Our results elucidated that METTL3 knockdown inhibited the hypoxia-mediated EndMT process by inactivating TRPC6/calcineurin/NFAT signaling.
Collapse
|
9
|
Masaki T, Habara M, Shibutani S, Hanaki S, Sato Y, Tomiyasu H, Shimada M. Dephosphorylation of the EGFR protein by calcineurin at serine 1046/1047 enhances its stability. Biochem Biophys Res Commun 2023; 641:84-92. [PMID: 36525928 DOI: 10.1016/j.bbrc.2022.12.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022]
Abstract
The epidermal growth factor receptor (EGFR) is highly expressed or abnormally activated in several types of cancers, such as lung and colorectal cancers. Inhibitors that suppress the tyrosine kinase activity of EGFR have been used in the treatment of lung cancer. However, resistance to these inhibitors has become an issue in cancer treatment, and the development of new therapies that inhibit EGFR is desired. We found that calcineurin, a Ca2+/calmodulin-activated serine/threonine phosphatase, is a novel regulator of EGFR. Inhibition of calcineurin by FK506 treatment or calcineurin depletion promoted EGFR degradation in cancer cells. In addition, we found that calcineurin dephosphorylates EGFR at serine (S)1046/1047, which in turn stabilizes EGFR. Furthermore, in human colon cancer cells transplanted into mice, the inhibition of calcineurin by FK506 decreased EGFR expression. These results indicate that calcineurin stabilizes EGFR by dephosphorylating S1046/1047 and promotes tumor growth. These findings suggest that calcineurin may be a new therapeutic target for cancers with high EGFR expression or activation.
Collapse
Affiliation(s)
- Takahiro Masaki
- Department of Veterinary Biochemistry, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, Yamaguchi, 753-8511, Japan
| | - Makoto Habara
- Department of Veterinary Biochemistry, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, Yamaguchi, 753-8511, Japan
| | - Shusaku Shibutani
- Department of Veterinary Hygiene, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, Yamaguchi, 753-8511, Japan
| | - Shunsuke Hanaki
- Department of Veterinary Biochemistry, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, Yamaguchi, 753-8511, Japan
| | - Yuki Sato
- Department of Veterinary Biochemistry, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, Yamaguchi, 753-8511, Japan
| | - Haruki Tomiyasu
- Department of Veterinary Biochemistry, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, Yamaguchi, 753-8511, Japan
| | - Midori Shimada
- Department of Veterinary Biochemistry, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, Yamaguchi, 753-8511, Japan.
| |
Collapse
|
10
|
FBXL16 Promotes Endometrial Progesterone Resistance via PP2AB55α/Cyclin D1 Axis in Ishikawa. J Immunol Res 2022; 2022:7372202. [PMID: 36106050 PMCID: PMC9467819 DOI: 10.1155/2022/7372202] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/25/2022] [Accepted: 08/06/2022] [Indexed: 11/18/2022] Open
Abstract
Background F-box proteins are essential components of the E3 ubiquitin ligases which are involved in the regulation of almost all life activities such as cell cycle, proliferation, and apoptosis, which have become an important research and drug target. However, there are few studies on F-box and leucine-rich repeat protein 16 (FBXL16) in endometrial carcinoma. Methods Clinical samples were collected for determining the correlation between FBXL16 and endometrial carcinoma. Cells were screened and established with Ishikawa cells which proved the fundamental role of FBXL16 in regulating cell proliferation and cell cycle. The MPA-resistant endometrial carcinoma cell line Ishikawa/MPA was established. FBXL16, PP2AB55α, and cyclin D1 were analyzed separately in MPA sensitive and resistant Ishikawa cells in vitro and in vivo. Results The high expression of FBXL16 was positively correlated with MPA resistance and poor prognosis of endometrial cancer. MPA tolerance of endometrial cancer cells was inhibited by knockdown of FBXL16 in DNA content assessment, CCK-8, and colony formation. It was confirmed that FBXL16 inhibited the activity of substrate PP2AB55α by binding to PP2A, reduced the phosphorylation level at Thr308 site of AKT1, inhibited the expression of GSK-3β, and thus led to a significant decrease in the phosphorylation level of cyclin D1, which prevented the ubiquitination recognition and degradation of cyclin D1. Conclusion In our experiments, FBXL16 binds PP2A to promote the dephosphorylation of Thr286 site of cyclin D1 via AKT1/GSK3β/cyclin D1 pathway, which is required for resisting the ubiquitination degradation and enhances the MPA resistance of Ishikawa.
Collapse
|
11
|
Chen L, Song M, Yao C. Calcineurin in development and disease. Genes Dis 2022; 9:915-927. [PMID: 35685477 PMCID: PMC9170610 DOI: 10.1016/j.gendis.2021.03.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/27/2021] [Accepted: 03/05/2021] [Indexed: 12/26/2022] Open
Abstract
Calcineurin (CaN) is a unique calcium (Ca2+) and calmodulin (CaM)-dependent serine/threonine phosphatase that becomes activated in the presence of increased intracellular Ca2+ level. CaN then functions to dephosphorylate target substrates including various transcription factors, receptors, and channels. Once activated, the CaN signaling pathway participates in the development of multiple organs as well as the onset and progression of various diseases via regulation of different cellular processes. Here, we review current literature regarding the structural and functional properties of CaN, highlighting its crucial role in the development and pathogenesis of immune system disorders, neurodegenerative diseases, kidney disease, cardiomyopathy and cancer.
Collapse
Affiliation(s)
- Lei Chen
- Department of Blood Transfusion, First Affiliated Hospital, The Third Military Medical University (Army Medical University), Chongqing 400038, PR China
| | - Min Song
- Department of Blood Transfusion, First Affiliated Hospital, The Third Military Medical University (Army Medical University), Chongqing 400038, PR China
| | - Chunyan Yao
- Department of Blood Transfusion, First Affiliated Hospital, The Third Military Medical University (Army Medical University), Chongqing 400038, PR China
| |
Collapse
|
12
|
Lao M, Zhang X, Yang H, Bai X, Liang T. RCAN1-mediated calcineurin inhibition as a target for cancer therapy. Mol Med 2022; 28:69. [PMID: 35717152 PMCID: PMC9206313 DOI: 10.1186/s10020-022-00492-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 05/26/2022] [Indexed: 11/10/2022] Open
Abstract
Cancer is the leading cause of mortality worldwide. Regulator of calcineurin 1 (RCAN1), as a patent endogenous inhibitor of calcineurin, plays crucial roles in the pathogenesis of cancers. Except for hypopharyngeal and laryngopharynx cancer, high expression of RCAN1 inhibits tumor progression. Molecular antitumor functions of RCAN1 are largely dependent on calcineurin. In this review, we highlight current research on RCAN1 characteristics, and the interaction between RCAN1 and calcineurin. Moreover, the dysregulation of RCAN1 in various cancers is reviewed, and the potential of targeting RCAN1 as a new therapeutic approach is discussed.
Collapse
Affiliation(s)
- Mengyi Lao
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310009, Zhejiang, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China.,Zhejiang Provincial Innovation Center for the Study of Pancreatic Diseases, Hangzhou, 310009, Zhejiang, China.,Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310009, Zhejiang, China
| | - Xiaozhen Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310009, Zhejiang, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China.,Zhejiang Provincial Innovation Center for the Study of Pancreatic Diseases, Hangzhou, 310009, Zhejiang, China.,Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310009, Zhejiang, China
| | - Hanshen Yang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310009, Zhejiang, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China.,Zhejiang Provincial Innovation Center for the Study of Pancreatic Diseases, Hangzhou, 310009, Zhejiang, China.,Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310009, Zhejiang, China
| | - Xueli Bai
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310009, Zhejiang, China. .,Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China. .,Zhejiang Provincial Innovation Center for the Study of Pancreatic Diseases, Hangzhou, 310009, Zhejiang, China. .,Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310009, Zhejiang, China.
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310009, Zhejiang, China. .,Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China. .,Zhejiang Provincial Innovation Center for the Study of Pancreatic Diseases, Hangzhou, 310009, Zhejiang, China. .,Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310009, Zhejiang, China. .,Cancer Center, Zhejiang University, Hangzhou, 310058, Zhejiang, China.
| |
Collapse
|
13
|
Stagni F, Bartesaghi R. The Challenging Pathway of Treatment for Neurogenesis Impairment in Down Syndrome: Achievements and Perspectives. Front Cell Neurosci 2022; 16:903729. [PMID: 35634470 PMCID: PMC9130961 DOI: 10.3389/fncel.2022.903729] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 04/19/2022] [Indexed: 12/17/2022] Open
Abstract
Down syndrome (DS), also known as trisomy 21, is a genetic disorder caused by triplication of Chromosome 21. Gene triplication may compromise different body functions but invariably impairs intellectual abilities starting from infancy. Moreover, after the fourth decade of life people with DS are likely to develop Alzheimer’s disease. Neurogenesis impairment during fetal life stages and dendritic pathology emerging in early infancy are thought to be key determinants of alterations in brain functioning in DS. Although the progressive improvement in medical care has led to a notable increase in life expectancy for people with DS, there are currently no treatments for intellectual disability. Increasing evidence in mouse models of DS reveals that pharmacological interventions in the embryonic and neonatal periods may greatly benefit brain development and cognitive performance. The most striking results have been obtained with pharmacotherapies during embryonic life stages, indicating that it is possible to pharmacologically rescue the severe neurodevelopmental defects linked to the trisomic condition. These findings provide hope that similar benefits may be possible for people with DS. This review summarizes current knowledge regarding (i) the scope and timeline of neurogenesis (and dendritic) alterations in DS, in order to delineate suitable windows for treatment; (ii) the role of triplicated genes that are most likely to be the key determinants of these alterations, in order to highlight possible therapeutic targets; and (iii) prenatal and neonatal treatments that have proved to be effective in mouse models, in order to rationalize the choice of treatment for human application. Based on this body of evidence we will discuss prospects and challenges for fetal therapy in individuals with DS as a potential means of drastically counteracting the deleterious effects of gene triplication.
Collapse
Affiliation(s)
- Fiorenza Stagni
- Department for Life Quality Studies, University of Bologna, Rimini, Italy
| | - Renata Bartesaghi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
- *Correspondence: Renata Bartesaghi,
| |
Collapse
|
14
|
FKBP52 and FKBP51 differentially regulate the stability of estrogen receptor in breast cancer. Proc Natl Acad Sci U S A 2022; 119:e2110256119. [PMID: 35394865 PMCID: PMC9169630 DOI: 10.1073/pnas.2110256119] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
SignificanceEstrogen receptor α (ERα) is a transcription factor that induces cell proliferation and exhibits increased expression in a large subset of breast cancers. We comprehensively searched for indicators of poor prognosis in ERα-positive breast cancer through the multiple databases, including interactome, transcriptome, and survival analysis, and identified FKBP52. We found that two immunophilins, FKBP52 and FKBP51, have opposing effects on ERα stability and propose that therapeutic targeting of FKBP52 could be useful for the prevention and treatment of ERα-positive breast cancers, including endocrine therapy-resistant breast cancers.
Collapse
|
15
|
Decoding the Phosphatase Code: Regulation of Cell Proliferation by Calcineurin. Int J Mol Sci 2022; 23:ijms23031122. [PMID: 35163061 PMCID: PMC8835043 DOI: 10.3390/ijms23031122] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/18/2022] [Accepted: 01/18/2022] [Indexed: 02/06/2023] Open
Abstract
Calcineurin, a calcium-dependent serine/threonine phosphatase, integrates the alterations in intracellular calcium levels into downstream signaling pathways by regulating the phosphorylation states of several targets. Intracellular Ca2+ is essential for normal cellular physiology and cell cycle progression at certain critical stages of the cell cycle. Recently, it was reported that calcineurin is activated in a variety of cancers. Given that abnormalities in calcineurin signaling can lead to malignant growth and cancer, the calcineurin signaling pathway could be a potential target for cancer treatment. For example, NFAT, a typical substrate of calcineurin, activates the genes that promote cell proliferation. Furthermore, cyclin D1 and estrogen receptors are dephosphorylated and stabilized by calcineurin, leading to cell proliferation. In this review, we focus on the cell proliferative functions and regulatory mechanisms of calcineurin and summarize the various substrates of calcineurin. We also describe recent advances regarding dysregulation of the calcineurin activity in cancer cells. We hope that this review will provide new insights into the potential role of calcineurin in cancer development.
Collapse
|
16
|
Calcineurin regulates the stability and activity of estrogen receptor α. Proc Natl Acad Sci U S A 2021; 118:2114258118. [PMID: 34711683 DOI: 10.1073/pnas.2114258118] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 08/27/2021] [Indexed: 12/14/2022] Open
Abstract
Estrogen receptor α (ER-α) mediates estrogen-dependent cancer progression and is expressed in most breast cancer cells. However, the molecular mechanisms underlying the regulation of the cellular abundance and activity of ER-α remain unclear. We here show that the protein phosphatase calcineurin regulates both ER-α stability and activity in human breast cancer cells. Calcineurin depletion or inhibition down-regulated the abundance of ER-α by promoting its polyubiquitination and degradation. Calcineurin inhibition also promoted the binding of ER-α to the E3 ubiquitin ligase E6AP, and calcineurin mediated the dephosphorylation of ER-α at Ser294 in vitro. Moreover, the ER-α (S294A) mutant was more stable and activated the expression of ER-α target genes to a greater extent compared with the wild-type protein, whereas the extents of its interaction with E6AP and polyubiquitination were attenuated. These results suggest that the phosphorylation of ER-α at Ser294 promotes its binding to E6AP and consequent degradation. Calcineurin was also found to be required for the phosphorylation of ER-α at Ser118 by mechanistic target of rapamycin complex 1 and the consequent activation of ER-α in response to β-estradiol treatment. Our study thus indicates that calcineurin controls both the stability and activity of ER-α by regulating its phosphorylation at Ser294 and Ser118 Finally, the expression of the calcineurin A-α gene (PPP3CA) was associated with poor prognosis in ER-α-positive breast cancer patients treated with tamoxifen or other endocrine therapeutic agents. Calcineurin is thus a promising target for the development of therapies for ER-α-positive breast cancer.
Collapse
|
17
|
In vitro study on immune response modifiers as novel medical treatment options for cholesteatoma. Int J Pediatr Otorhinolaryngol 2021; 145:110743. [PMID: 33933986 DOI: 10.1016/j.ijporl.2021.110743] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 04/24/2021] [Indexed: 11/21/2022]
Abstract
OBJECTIVES To investigate cytokine profile of cholesteatoma and to collect information about important intercellular signaling pathways by establishing two different cell culture models, to block important intercellular signaling pathways in cholesteatoma by applying immune system modifier drugs to develop alternative medical therapy options for cholesteatoma. METHODS To observe the pathogenesis of cholesteatoma and to apply the immunomodulatory drugs, cholesteatoma tissue culture models were constituted with HEKa cells and cholesteatoma keratinocytes, which were obtained from 3 patients who underwent operations for cholesteatoma. Medicines including 5-fluorourasil, imiquimod, cyclosporine, and tacrolimus were applied on both cholesteatoma keratinocytes and HEKa cells. After 48 h of incubation, IL-1, IL-6, IL-8, IL-10, TNF-α, and Ki67 levels were measured to determine cell viability rates. RESULTS In the cholesteatoma control group, IL-6 and TNF-α levels were found higher than in the HEKa control group. All repurposed drugs in the study demonstrated anti-inflammatory, anti-proliferative, and cytotoxic effects on cholesteatoma. Imiquimod and tacrolimus in particular are potential treatment prospects for cholesteatoma due to their strong anti-inflammatory and cytotoxic effects. CONCLUSION Medical therapy options for cholesteatoma are still missing and surgery is not the ultimate solution. We have focused on intercellular inflammatory processes, which play significant roles in the pathogenesis of cholesteatoma in our paper. Inflammation and proliferation of cholesteatoma decreased after all repurposed drug applications in our study. Anti-inflammatory and anti-proliferative effects of tacrolimus and imiquimod was more significant than other drugs in the study. For this reason, tacrolimus and imiquimod should be examined in depth with in vivo studies in terms of efficacy and safety for medical treatment of cholesteatoma.
Collapse
|
18
|
Maeda K, Habara M, Kawaguchi M, Matsumoto H, Hanaki S, Masaki T, Sato Y, Matsuyama H, Kunieda K, Nakagawa H, Shimada M. FKBP51 and FKBP52 regulate androgen receptor dimerization and proliferation in prostate cancer cells. Mol Oncol 2021; 16:940-956. [PMID: 34057812 PMCID: PMC8847985 DOI: 10.1002/1878-0261.13030] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 04/23/2021] [Accepted: 05/28/2021] [Indexed: 11/09/2022] Open
Abstract
The growth of prostate cancer is dependent on the androgen receptor (AR), which serves as a ligand-specific transcription factor. Although two immunophilins, FKBP51 and FKBP52, are known to regulate AR activity, the precise mechanism remains unclear. We found that depletion of either FKBP51 or FKBP52 reduced AR dimer formation, chromatin binding, and phosphorylation, suggesting defective AR signaling. Furthermore, the peptidyl-prolyl cis/trans isomerase activity of FKBP51 was found to be required for AR dimer formation and cancer cell growth. Treatment of prostate cancer cells with FK506, which binds to the FK1 domain of FKBPs, or with MJC13, an inhibitor of FKBP52-AR signaling, also inhibited AR dimer formation. Finally, elevated expression of FKBP52 was associated with a higher rate of prostate-specific antigen recurrence in patients with prostate cancer. Collectively, these results suggest that FKBP51 and FKBP52 might be promising targets for prostate cancer treatment through the inhibition of AR dimer formation.
Collapse
Affiliation(s)
- Keisuke Maeda
- Department of Biochemistry, Joint Faculty of Veterinary Science, Yamaguchi University, Japan
| | - Makoto Habara
- Department of Biochemistry, Joint Faculty of Veterinary Science, Yamaguchi University, Japan
| | | | - Hiroaki Matsumoto
- Department of Urology, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Shunsuke Hanaki
- Department of Biochemistry, Joint Faculty of Veterinary Science, Yamaguchi University, Japan
| | - Takahiro Masaki
- Department of Biochemistry, Joint Faculty of Veterinary Science, Yamaguchi University, Japan
| | - Yuki Sato
- Department of Biochemistry, Joint Faculty of Veterinary Science, Yamaguchi University, Japan
| | - Hideyasu Matsuyama
- Department of Urology, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Kazuki Kunieda
- Graduate School of Pharmaceutical Sciences, Nagoya City University, Japan
| | - Hidehiko Nakagawa
- Graduate School of Pharmaceutical Sciences, Nagoya City University, Japan
| | - Midori Shimada
- Department of Biochemistry, Joint Faculty of Veterinary Science, Yamaguchi University, Japan
| |
Collapse
|
19
|
Tran MT. Overview of Ca2+ signaling in lung cancer progression and metastatic lung cancer with bone metastasis. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2021; 2:249-265. [PMID: 36046435 PMCID: PMC9400727 DOI: 10.37349/etat.2021.00045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 05/06/2021] [Indexed: 12/28/2022] Open
Abstract
Intracellular Ca2+ ions that are thought to be one of the most important second messengers for cellular signaling, have a substantial diversity of roles in regulating a plethora of fundamental cellular physiology such as gene expression, cell division, cell motility and apoptosis. It has been suggestive of the Ca2+ signaling-dependent cellular processes to be tightly regulated by the numerous types of Ca2+ channels, pumps, exchangers and sensing receptors. Consequently, dysregulated Ca2+ homeostasis leads to a series of events connected to elevated malignant phenotypes including uncontrolled proliferation, migration, invasion and metastasis, all of which are frequently observed in advanced stage lung cancer cells. The incidence of bone metastasis in patients with advanced stage lung cancer is estimated in a range of 30% to 40%, bringing about a significant negative impact on both morbidity and survival. This review dissects and summarizes the important roles of Ca2+ signaling transduction in contributing to lung cancer progression, and address the question: if and how Ca2+ signaling might have been engaged in metastatic lung cancer with bone metastasis, thereby potentially providing the multifaceted and promising solutions for therapeutic intervention.
Collapse
Affiliation(s)
- Manh Tien Tran
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
| |
Collapse
|
20
|
Hanaki S, Habara M, Shimada M. UV-induced activation of ATR is mediated by UHRF2. Genes Cells 2021; 26:447-454. [PMID: 33848395 DOI: 10.1111/gtc.12851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/09/2021] [Accepted: 04/09/2021] [Indexed: 11/27/2022]
Abstract
UHRF1 (Ubiquitin-like with PHD and ring finger domains 1) regulates DNA methylation and histone modifications and plays a key role in cell proliferation and the DNA damage response. However, the function of UHRF2, a paralog of UHRF1, in the DNA damage response remains largely unknown. Here, we show that UHRF2 is essential for maintaining cell viability after UV irradiation, as well as for the proliferation of cancer cells. UHRF2 was found to physically interact with ATR in a DNA damage-dependent manner through UHRF2's TTD domain. In addition, phosphorylation of threonine at position 1989, which is required for UV-induced activation of ATR, was impaired in cells depleted of UHRF2, suggesting that UHRF2 is essential in ATR activation. In conclusion, these results suggest a new regulatory mechanism of ATR activation mediated by UHRF2.
Collapse
Affiliation(s)
- Shunsuke Hanaki
- Department of Veterinary Biochemistry, Yamaguchi University, Yamaguchi City, Japan
| | - Makoto Habara
- Department of Veterinary Biochemistry, Yamaguchi University, Yamaguchi City, Japan
| | - Midori Shimada
- Department of Veterinary Biochemistry, Yamaguchi University, Yamaguchi City, Japan
| |
Collapse
|
21
|
Hanaki S, Habara M, Masaki T, Maeda K, Sato Y, Nakanishi M, Shimada M. PP1 regulatory subunit NIPP1 regulates transcription of E2F1 target genes following DNA damage. Cancer Sci 2021; 112:2739-2752. [PMID: 33939241 PMCID: PMC8253265 DOI: 10.1111/cas.14924] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/14/2021] [Accepted: 04/14/2021] [Indexed: 12/21/2022] Open
Abstract
DNA damage induces transcriptional repression of E2F1 target genes and a reduction in histone H3‐Thr11 phosphorylation (H3‐pThr11) at E2F1 target gene promoters. Dephosphorylation of H3‐pThr11 is partly mediated by Chk1 kinase and protein phosphatase 1γ (PP1γ) phosphatase. Here, we isolated NIPP1 as a regulator of PP1γ‐mediated H3‐pThr11 by surveying nearly 200 PP1 interactor proteins. We found that NIPP1 inhibits PP1γ‐mediated dephosphorylation of H3‐pThr11 both in vivo and in vitro. By generating NIPP1‐depleted cells, we showed that NIPP1 is required for cell proliferation and the expression of E2F1 target genes. Upon DNA damage, activated protein kinase A (PKA) phosphorylated the NIPP1‐Ser199 residue, adjacent to the PP1 binding motif (RVxF), and triggered the dissociation of NIPP1 from PP1γ, leading to the activation of PP1γ. Furthermore, the inhibition of PKA activity led to the activation of E2F target genes. Statistical analysis confirmed that the expression of NIPP1 was positively correlated with E2F target genes. Taken together, these findings demonstrate that the PP1 regulatory subunit NIPP1 modulates E2F1 target genes by linking PKA and PP1γ during DNA damage.
Collapse
Affiliation(s)
- Shunsuke Hanaki
- Department of Biochemistry, Joint Faculty of Veterinary Science, Yamaguchi University, Yamaguchi, Japan
| | - Makoto Habara
- Department of Biochemistry, Joint Faculty of Veterinary Science, Yamaguchi University, Yamaguchi, Japan
| | - Takahiro Masaki
- Department of Biochemistry, Joint Faculty of Veterinary Science, Yamaguchi University, Yamaguchi, Japan
| | - Keisuke Maeda
- Department of Biochemistry, Joint Faculty of Veterinary Science, Yamaguchi University, Yamaguchi, Japan
| | - Yuki Sato
- Department of Biochemistry, Joint Faculty of Veterinary Science, Yamaguchi University, Yamaguchi, Japan
| | - Makoto Nakanishi
- Division of Cancer Biology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Midori Shimada
- Department of Biochemistry, Joint Faculty of Veterinary Science, Yamaguchi University, Yamaguchi, Japan
| |
Collapse
|