1
|
Zhang H, Zhang L, Wu Z. Interaction of STIL with FOXM1 regulates SF3A3 transcription in the hepatocellular carcinoma development. Cell Div 2025; 20:1. [PMID: 39825314 PMCID: PMC11740530 DOI: 10.1186/s13008-025-00142-4] [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: 07/18/2024] [Accepted: 01/09/2025] [Indexed: 01/20/2025] Open
Abstract
BACKGROUND Dysregulation of SF3A3 has been related to the development of many cancers. Here, we investigated the functional role of SF3A3 in hepatocellular carcinoma (HCC). METHODS SF3A3 expression in HCC tissues and cell lines was examined using RT-qPCR. Changes in malignant behavior of HCC cells after downregulation of SF3A3 were assessed by EdU, colony formation, flow cytometry, wound healing, and Transwell invasion assays. Multiple datasets were combined to identify the upstream modifiers of SF3A3. The binding relationship between STIL and FOXM1 was explored by co-IP assay, and the effect of STIL and FOXM1 on the binding of FOXM1 at the SF3A3 promoter was detected by ChIP-qPCR assay. A xenograft tumor model was established to explore the changes of tumors in vivo, and the expression of Ki67, GPC3, and p53 in tumor tissues was detected by immunohistochemistry. RESULTS SF3A3 and STIL were overexpressed in HCC tissues and cells, and downregulation of SF3A3 or STIL inhibited the malignant behavior of HCC cells by promoting the expression of p53. An interaction between STIL and FOXM1 regulated the SF3A3 expression in HCC cells. Knockdown of FOXM1 further enhanced the anti-tumor effects of STIL loss on HCC cells in vitro and in vivo, whereas SF3A3 overexpression overturned the impact of STIL loss on HCC cells in vitro and in vivo. CONCLUSIONS Our findings indicate that STIL/FOXM1 expedites HCC development by activating SF3A3, which highlights the importance of SF3A3 as a promising prognostic marker and therapeutic target for HCC.
Collapse
Affiliation(s)
- Haijun Zhang
- Second Department of General Surgery, the First Hospital of Qiqihar, No. 700, Pukui avenue, Long sha District, Qiqihar, Heilongjiang, 161000, P. R. China.
| | - Lin Zhang
- Department of Pharmacy, the Second Affiliated Hospital of Qiqihar Medical College, Qiqihar, Heilongjiang, 161006, P. R. China
| | - Ziqi Wu
- Second Department of General Surgery, the First Hospital of Qiqihar, No. 700, Pukui avenue, Long sha District, Qiqihar, Heilongjiang, 161000, P. R. China
| |
Collapse
|
2
|
Matsuki T, Tabata H, Ueda M, Ito H, Nagata KI, Tsuneura Y, Eda S, Kasai K, Nakayama A. The MCPH7 Gene Product STIL Is Essential for Dendritic Spine Formation. Cells 2025; 14:62. [PMID: 39851490 PMCID: PMC11764357 DOI: 10.3390/cells14020062] [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: 11/23/2024] [Revised: 12/14/2024] [Accepted: 12/23/2024] [Indexed: 01/26/2025] Open
Abstract
Dendritic spine formation/maintenance is highly dependent on actin cytoskeletal dynamics, which is regulated by small GTPases Rac1 and Cdc42 through their downstream p21-activated kinase/LIM-kinase-I/cofilin pathway. ARHGEF7, also known as ß-PIX, is a guanine nucleotide exchange factor for Rac1 and Cdc42, thereby activating Rac1/Cdc42 and the downstream pathway, leading to the upregulation of spine formation/maintenance. We found that STIL, one of the primary microcephaly gene products, is associated with ARHGEF7 in dendritic spines and that knockdown of Stil resulted in a significant reduction in dendritic spines in neurons both in vitro and in vivo. Rescue experiments indicated that the STIL requirement for spine formation/maintenance depended on its coiled coil domain that mediates the association with ARHGEF7. The overexpression of Rac1/Cdc42 compensated for the spine reduction caused by STIL knockdown. FRET experiments showed that Rac activation is impaired in STIL knockdown neurons. Chemical long-term potentiation, which triggers Rac activation, promoted STIL accumulation in the spine and its association with ARHGEF7. The dynamics of these proteins further supported their coordinated involvement in spine formation/maintenance. Based on these findings, we concluded that the centrosomal protein STIL is a novel regulatory factor essential for spine formation/maintenance by activating Rac and its downstream pathway, possibly through the association with ARHGEF7.
Collapse
Affiliation(s)
- Tohru Matsuki
- Department of Cellular Pathology, Institute for Developmental Research, Aichi Developmental Disability Center, Kasugai 480-0392, Aichi, Japan (S.E.)
| | - Hidenori Tabata
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Developmental Disability Center, Kasugai 480-0392, Aichi, Japan; (H.T.); (K.-i.N.)
| | - Masashi Ueda
- Department of Cellular Pathology, Institute for Developmental Research, Aichi Developmental Disability Center, Kasugai 480-0392, Aichi, Japan (S.E.)
| | - Hideaki Ito
- Department of Pathology, Aichi Medical University School of Medicine, Nagakute 480-1195, Aichi, Japan (K.K.)
| | - Koh-ichi Nagata
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Developmental Disability Center, Kasugai 480-0392, Aichi, Japan; (H.T.); (K.-i.N.)
- Department of Neurochemistry, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Aichi, Japan
| | - Yumi Tsuneura
- Department of Cellular Pathology, Institute for Developmental Research, Aichi Developmental Disability Center, Kasugai 480-0392, Aichi, Japan (S.E.)
| | - Shima Eda
- Department of Cellular Pathology, Institute for Developmental Research, Aichi Developmental Disability Center, Kasugai 480-0392, Aichi, Japan (S.E.)
| | - Kenji Kasai
- Department of Pathology, Aichi Medical University School of Medicine, Nagakute 480-1195, Aichi, Japan (K.K.)
| | - Atsuo Nakayama
- Department of Cellular Pathology, Institute for Developmental Research, Aichi Developmental Disability Center, Kasugai 480-0392, Aichi, Japan (S.E.)
- Department of Neurochemistry, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Aichi, Japan
| |
Collapse
|
3
|
Zhang Z, Wang Z, Zhang H, Gong Y, Sun H, Zhang W. Regulatory factor X-5/SCL/TAL1 interruption site axis promotes aerobic glycolysis and hepatocellular carcinoma cell stemness. Kaohsiung J Med Sci 2025; 41:e12922. [PMID: 39718123 PMCID: PMC11724169 DOI: 10.1002/kjm2.12922] [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: 08/01/2024] [Revised: 11/18/2024] [Accepted: 12/02/2024] [Indexed: 12/25/2024] Open
Abstract
The incidence and development of various tumors, such as hepatocellular carcinoma (HCC), are linked to tumor stem cells. Although research has revealed how important SCL/TAL1 interruption site (STIL) is in many human tumors, the impact of STIL on HCC stem cells is poorly understood. This study aimed to examine the regulatory mechanisms and the function of STIL in the stemness of HCC tumor cells. Bioinformatics analysis was applied to determine the STIL and regulatory factor X-5 (RFX5) expression in HCC tissues. Immunohistochemistry (IHC) was used to detect the expression of STIL and RFX5 in HCC tissues. Quantitative real-time polymerase chain reaction was utilized to measure the STIL and RFX5 expression levels in HCC cells. The viability of the cells was assessed by the Cell Counting Kit-8 assay. The sphere formation assay was used to evaluate the sphere-forming capacity. The expression levels of the stem cell markers SOX2, Oct-4, CD133, CD44, the glycolysis-related proteins LDHA, HK2, AKT, p-AKT, and β-catenin were assessed by Western blot. Lactate production, oxygen consumption rate, and extracellular acidification rate were measured to assess the glycolytic capacity of HCC cells. Chromatin immunoprecipitation and dual-luciferase experiments were performed to validate the connection between RFX5 and STIL. Bioinformatics analysis determined that STIL exhibited high expression in HCC tissues and was enriched in the glycolysis pathway. In addition, the expression of glycolysis marker genes was positively correlated with STIL expression. Cell experiments verified that the activation of the glycolysis pathway by overexpression of STIL promoted stemness in HCC. Molecular experiments also revealed the binding relationship between STIL and RFX5. IHC detected high expression of STIL and RFX5 in HCC tissues. Cell functional experiments revealed that RFX5 could influence the HCC cells stemness by activating the STIL transcription via the glycolysis pathway. This study identified a novel role for the RFX5/STIL axis in HCC progression, which may offer treatment targets for HCC.
Collapse
Affiliation(s)
- Zhi‐Zhong Zhang
- Department of General Surgery Ward OneAnyang Tumor HospitalAnyangHenanChina
| | - Zi‐Ming Wang
- Department of General Surgery Ward OneAnyang Tumor HospitalAnyangHenanChina
| | - Hao‐Wen Zhang
- Department of General Surgery Ward OneAnyang Tumor HospitalAnyangHenanChina
| | - Yan‐Xin Gong
- Department of General Surgery Ward OneAnyang Tumor HospitalAnyangHenanChina
| | - Hao‐Ran Sun
- Pathological CenterAnyang Tumor HospitalAnyangHenanChina
| | - Wei Zhang
- Department of General Surgery Ward OneAnyang Tumor HospitalAnyangHenanChina
| |
Collapse
|
4
|
Ohtsuka S, Kato H, Ishikawa R, Watanabe H, Miyazaki R, Katsuragi SY, Yoshimura K, Yamada H, Sakai Y, Inoue Y, Takanashi Y, Sekihara K, Funai K, Sugimura H, Shinmura K. STIL Overexpression Is Associated with Chromosomal Numerical Abnormalities in Non-Small-Cell Lung Carcinoma Through Centrosome Amplification. Curr Oncol 2024; 31:7936-7949. [PMID: 39727708 PMCID: PMC11674966 DOI: 10.3390/curroncol31120585] [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: 10/31/2024] [Revised: 12/09/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024] Open
Abstract
STIL is a regulatory protein essential for centriole biogenesis, and its dysregulation has been implicated in various diseases, including malignancies. However, its role in non-small-cell lung carcinoma (NSCLC) remains unclear. In this study, we examined STIL expression and its potential association with chromosomal numerical abnormalities (CNAs) in NSCLC using The Cancer Genome Atlas (TCGA) dataset, immunohistochemical analysis, and in vitro experiments with NSCLC cell lines designed to overexpress STIL. TCGA data revealed upregulated STIL mRNA expression in lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC), the two major subtypes of NSCLC. Immunohistochemical analysis of cases from our hospital (LUAD, n = 268; LUSC, n = 98) revealed STIL protein overexpression. To elucidate the functional role of STIL, an inducible STIL-overexpressing H1299 NSCLC cell line was generated. Overexpression of STIL in these cells promoted centrosome amplification, leading to chromosomal instability. Finally, analysis of arm-level chromosomal copy number alterations from the TCGA dataset revealed that elevated STIL mRNA expression was associated with CNAs in both LUAD and LUSC. These findings suggest that STIL overexpression is associated with CNAs in NSCLC, likely through centrosome amplification, which is linked to chromosomal instability and might represent a potential therapeutic target for NSCLC treatment.
Collapse
Affiliation(s)
- Shunsuke Ohtsuka
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; (S.O.); (H.K.); (R.I.); (H.W.); (R.M.); (S.-y.K.); (K.Y.); (H.Y.); (Y.S.); (H.S.)
| | - Hisami Kato
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; (S.O.); (H.K.); (R.I.); (H.W.); (R.M.); (S.-y.K.); (K.Y.); (H.Y.); (Y.S.); (H.S.)
| | - Rei Ishikawa
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; (S.O.); (H.K.); (R.I.); (H.W.); (R.M.); (S.-y.K.); (K.Y.); (H.Y.); (Y.S.); (H.S.)
| | - Hirofumi Watanabe
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; (S.O.); (H.K.); (R.I.); (H.W.); (R.M.); (S.-y.K.); (K.Y.); (H.Y.); (Y.S.); (H.S.)
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan;
| | - Ryosuke Miyazaki
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; (S.O.); (H.K.); (R.I.); (H.W.); (R.M.); (S.-y.K.); (K.Y.); (H.Y.); (Y.S.); (H.S.)
| | - Shin-ya Katsuragi
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; (S.O.); (H.K.); (R.I.); (H.W.); (R.M.); (S.-y.K.); (K.Y.); (H.Y.); (Y.S.); (H.S.)
| | - Katsuhiro Yoshimura
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; (S.O.); (H.K.); (R.I.); (H.W.); (R.M.); (S.-y.K.); (K.Y.); (H.Y.); (Y.S.); (H.S.)
| | - Hidetaka Yamada
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; (S.O.); (H.K.); (R.I.); (H.W.); (R.M.); (S.-y.K.); (K.Y.); (H.Y.); (Y.S.); (H.S.)
| | - Yasuhiro Sakai
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; (S.O.); (H.K.); (R.I.); (H.W.); (R.M.); (S.-y.K.); (K.Y.); (H.Y.); (Y.S.); (H.S.)
| | - Yusuke Inoue
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan;
| | - Yusuke Takanashi
- Department of Surgery 1, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; (Y.T.); (K.S.); (K.F.)
| | - Keigo Sekihara
- Department of Surgery 1, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; (Y.T.); (K.S.); (K.F.)
| | - Kazuhito Funai
- Department of Surgery 1, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; (Y.T.); (K.S.); (K.F.)
| | - Haruhiko Sugimura
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; (S.O.); (H.K.); (R.I.); (H.W.); (R.M.); (S.-y.K.); (K.Y.); (H.Y.); (Y.S.); (H.S.)
- Kyoundo Hospital, Sasaki Foundation, Tokyo 101-0062, Japan
| | - Kazuya Shinmura
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; (S.O.); (H.K.); (R.I.); (H.W.); (R.M.); (S.-y.K.); (K.Y.); (H.Y.); (Y.S.); (H.S.)
| |
Collapse
|
5
|
Wang M, Pan B, Hu Y, Gao J, Hou L, Yu Z, Li M, Zhao Z. STIL facilitates the development and malignant progression of triple-negative breast cancer through activation of Fanconi anemia pathway via interacting with KLF16. Transl Oncol 2024; 46:102010. [PMID: 38823260 PMCID: PMC11177054 DOI: 10.1016/j.tranon.2024.102010] [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: 07/30/2023] [Revised: 05/08/2024] [Accepted: 05/25/2024] [Indexed: 06/03/2024] Open
Abstract
BACKGROUND STIL is an important cell cycle-regulating protein specifically recruited to the mitotic centrosome to promote the replication of centrioles in dividing cells. However, the potential role of STIL in the regulation of the biological functions of triple-negative breast cancer remains still unclear. METHODS We screened for differentially expressed STIL in the Cancer Genome Atlas database. The expression of STIL protein in 10 pairs of breast cancer tissues and adjacent normal tissues was further assessed by western blotting. Functionally, the knockdown and overexpression of STIL have been used to explore the effects of STIL on breast cancer cell proliferation, migration, and invasion. Mechanistically, RNA-seq, dual-luciferase reporter assay, chromatin immunoprecipitation assay, mass spectrometry, immunoprecipitation assay, and DNA pull-down assay were performed. RESULTS Breast cancer tissues and cells have higher STIL expression than normal tissues and cells. STIL knockdown impairs breast cancer cell growth, migration, and invasion, whereas STIL overexpression accelerates these processes. STIL promotes breast cancer progression by regulating FANCD2 expression, and exploration of its molecular mechanism demonstrated that STIL interacts with KLF16 to regulate the expression of FANCD2. CONCLUSIONS Collectively, our findings identified STIL as a critical promoter of breast cancer progression that interacts with KLF16 to regulate Fanconi anemia pathway protein FANCD2. In summary, STIL is a potential novel biomarker and therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Meiling Wang
- Department of Breast Surgery & Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, 116023 China
| | - Bo Pan
- Department of Breast Surgery & Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, 116023 China
| | - Ye Hu
- Department of Breast Surgery & Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, 116023 China
| | - Jiyue Gao
- Department of Breast Surgery & Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, 116023 China
| | - Lu Hou
- Department of Breast Surgery & Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, 116023 China
| | - Zhenlong Yu
- College of Pharmacy, Dalian Medical University, Dalian, China.
| | - Man Li
- Department of Breast Surgery & Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, 116023 China.
| | - Zuowei Zhao
- Department of Breast Surgery & Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, 116023 China.
| |
Collapse
|
6
|
Athwal H, Kochiyanil A, Bhat V, Allan AL, Parsyan A. Centrosomes and associated proteins in pathogenesis and treatment of breast cancer. Front Oncol 2024; 14:1370565. [PMID: 38606093 PMCID: PMC11007099 DOI: 10.3389/fonc.2024.1370565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 03/04/2024] [Indexed: 04/13/2024] Open
Abstract
Breast cancer is the most prevalent malignancy among women worldwide. Despite significant advances in treatment, it remains one of the leading causes of female mortality. The inability to effectively treat advanced and/or treatment-resistant breast cancer demonstrates the need to develop novel treatment strategies and targeted therapies. Centrosomes and their associated proteins have been shown to play key roles in the pathogenesis of breast cancer and thus represent promising targets for drug and biomarker development. Centrosomes are fundamental cellular structures in the mammalian cell that are responsible for error-free execution of cell division. Centrosome amplification and aberrant expression of its associated proteins such as Polo-like kinases (PLKs), Aurora kinases (AURKs) and Cyclin-dependent kinases (CDKs) have been observed in various cancers, including breast cancer. These aberrations in breast cancer are thought to cause improper chromosomal segregation during mitosis, leading to chromosomal instability and uncontrolled cell division, allowing cancer cells to acquire new genetic changes that result in evasion of cell death and the promotion of tumor formation. Various chemical compounds developed against PLKs and AURKs have shown meaningful antitumorigenic effects in breast cancer cells in vitro and in vivo. The mechanism of action of these inhibitors is likely related to exacerbation of numerical genomic instability, such as aneuploidy or polyploidy. Furthermore, growing evidence demonstrates enhanced antitumorigenic effects when inhibitors specific to centrosome-associated proteins are used in combination with either radiation or chemotherapy drugs in breast cancer. This review focuses on the current knowledge regarding the roles of centrosome and centrosome-associated proteins in breast cancer pathogenesis and their utility as novel targets for breast cancer treatment.
Collapse
Affiliation(s)
- Harjot Athwal
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Arpitha Kochiyanil
- Faculty of Science, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Vasudeva Bhat
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- London Regional Cancer Program, London Health Sciences Centre, Lawson Health Research Institute, London, ON, Canada
| | - Alison L. Allan
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- London Regional Cancer Program, London Health Sciences Centre, Lawson Health Research Institute, London, ON, Canada
- Department of Oncology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Armen Parsyan
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- London Regional Cancer Program, London Health Sciences Centre, Lawson Health Research Institute, London, ON, Canada
- Department of Oncology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Division of General Surgery, Department of Surgery, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Department of Surgery, St. Joseph’s Health Care London and London Health Sciences Centre, London, ON, Canada
| |
Collapse
|
7
|
Lou H, Wu Z, Wei G. CDC6 may serve as an indicator of lung adenocarcinoma prognosis and progression based on TCGA and GEO data mining and experimental analyses. Oncol Rep 2024; 51:35. [PMID: 38186304 PMCID: PMC10807357 DOI: 10.3892/or.2024.8694] [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: 08/01/2023] [Accepted: 11/29/2023] [Indexed: 01/09/2024] Open
Abstract
Lung adenocarcinoma (LUAD) is one of the most lethal types of cancer worldwide, and accurately predicting patient prognosis is an important challenge. Gene prediction models, which are known for their simplicity and efficiency, have the potential to be used for prognostic predictions. However, the availability of models with true clinical value is limited. The present study integrated tissue sequencing and the clinical information of patients with LUAD from The Cancer Genome Atlas and Gene Expression Omnibus databases using bioinformatics. This comprehensive approach enabled the identification of 252 differentially expressed genes. Subsequently, univariate and multivariate Cox analyses were performed using these genes, and 14 and 3 genes [including cell division cycle 6 (CDC6), hyaluronan mediated motility receptor and STIL centriolar assembly protein] were selected for the construction of two prognostic models. Notably, the 3‑gene prognostic model exhibited a comparable predictive ability to that of the 14‑gene model. Functionally, pathway enrichment analysis revealed that CDC6 played a role in regulating the cell cycle and promoting tumor staging. To further investigate the relevance of CDC6, in vitro experiments involving the downregulation of CDC6 expression were conducted, which resulted in significant inhibition of tumor cell migration, invasion and proliferation. Moreover, in vivo experiments demonstrated that downregulating CDC6 expression significantly reduced the burden and metastasis of in situ lung tumors in mice. These findings suggested that CDC6 may be a critical gene involved in the development and prognosis of LUAD. In summary, the present study successfully constructed a simple yet accurate prognostic prediction model consisting of 3 genes. Additionally, the functional importance of CDC6 as a key gene in the model was identified. These findings lay a crucial foundation for further exploration of prognostic prediction models and a deeper understanding of the functional mechanisms of CDC6. Notably, these results have potential clinical implications for improving personalized treatment and prognosis evaluation for patients with LUAD.
Collapse
Affiliation(s)
- Hao Lou
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
| | - Zelai Wu
- Department of Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, P.R. China
| | - Guangyou Wei
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
- Department of Pediatrics, Bozhou Municipal People's Hospital, Bozhou, Anhui 236800, P.R. China
- Department of Pediatrics, Bozhou Clinical Medicine of Anhui University of Science and Technology School, Bozhou, Anhui 236800, P.R. China
| |
Collapse
|
8
|
Qu P, Shao Z, Zhang Y, He J, Lu D, Wei W, Hua J, Wang W, Wang J, Ding N. Primary cilium participates in radiation-induced bystander effects through TGF-β1 signaling. J Cell Physiol 2024; 239:e31163. [PMID: 38009273 DOI: 10.1002/jcp.31163] [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: 03/21/2023] [Revised: 11/09/2023] [Accepted: 11/15/2023] [Indexed: 11/28/2023]
Abstract
Many studies have indicated that tumor growth factor-beta (TGF-β) signaling mediates radiation-induced bystander effects (RIBEs). The primary cilium (PC) coordinates several signaling pathways including TGF-β signaling to regulate diverse cellular processes. But whether the PC participates in TGF-β induced RIBEs remains unclear. The cellular levels of TGF-β1 were detected by western blot analysis and the secretion of TGF-β1 was measured by ELISA kit. The ciliogenesis was altered by CytoD treatment, STIL siRNA transfection, IFT88 siRNA transfection, or KIF3a siRNA transfection, separately, and was detected by western blot analysis and immunofluorescence staining. G0 /G1 phase cells were arrested by serum starvation and S phase cells were induced by double thymidine block. The TGF-β1 signaling was interfered by LY2109761, a TGF-β receptor 1 (TβR1) inhibitor, or TGF-β1 neutral antibody. The DNA damages were induced by TGF-β1 or radiated conditional medium (RCM) from irradiated cells and were reflected by p21 expression, 53BP1 foci, and γH2AX foci. Compared with unirradiated control, both A549 and Beas-2B cells expressed and secreted more TGF-β1 after carbon ion beam or X-ray irradiation. RCM collected from irradiated cells or TGF-β1 treatment caused an increase of DNA damage in cocultured unirradiated Beas-2B cells while blockage of TGF-β signaling by TβR1 inhibitor or TGF-β1 neutral antibody alleviates this phenomenon. IFT88 siRNA or KIF3a siRNA impaired PC formation resulted in an aggravated DNA damage in bystander cells, while elevated PC formation by CytoD or STIL siRNA resulted in a decrease of DNA damage. Furthermore, TGF-β1 induced more DNA damages in S phases cells which showed lower PC formation rate and less DNA damages in G0 /G1 phase cells which showed higher PC formation rate. This study demonstrates the particular role of primary cilia during RCM induced DNA damages through TGF-β1 signaling restriction and thereby provides a functional link between primary cilia and RIBEs.
Collapse
Affiliation(s)
- Pei Qu
- Key Laboratory of Space Radiobiology of Gansu Province & Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Zhiang Shao
- Key Laboratory of Space Radiobiology of Gansu Province & Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Yanan Zhang
- Key Laboratory of Space Radiobiology of Gansu Province & Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Jinpeng He
- Key Laboratory of Space Radiobiology of Gansu Province & Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Dong Lu
- Key Laboratory of Space Radiobiology of Gansu Province & Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Wenjun Wei
- Key Laboratory of Space Radiobiology of Gansu Province & Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Junrui Hua
- Key Laboratory of Space Radiobiology of Gansu Province & Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Wei Wang
- Department of Urological Surgery, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Jufang Wang
- Key Laboratory of Space Radiobiology of Gansu Province & Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Nan Ding
- Key Laboratory of Space Radiobiology of Gansu Province & Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
9
|
Shamir M, Martin FJO, Woolfson DN, Friedler A. Molecular Mechanism of STIL Coiled-Coil Domain Oligomerization. Int J Mol Sci 2023; 24:14616. [PMID: 37834064 PMCID: PMC10572602 DOI: 10.3390/ijms241914616] [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: 08/20/2023] [Revised: 09/16/2023] [Accepted: 09/21/2023] [Indexed: 10/15/2023] Open
Abstract
Coiled-coil domains (CCDs) play key roles in regulating both healthy cellular processes and the pathogenesis of various diseases by controlling protein self-association and protein-protein interactions. Here, we probe the mechanism of oligomerization of a peptide representing the CCD of the STIL protein, a tetrameric multi-domain protein that is over-expressed in several cancers and associated with metastatic spread. STIL tetramerization is mediated both by an intrinsically disordered domain (STIL400-700) and a structured CCD (STIL CCD718-749). Disrupting STIL oligomerization via the CCD inhibits its activity in vivo. We describe a comprehensive biophysical and structural characterization of the concentration-dependent oligomerization of STIL CCD peptide. We combine analytical ultracentrifugation, fluorescence and circular dichroism spectroscopy to probe the STIL CCD peptide assembly in solution and determine dissociation constants of both the dimerization, (KD = 8 ± 2 µM) and tetramerization (KD = 68 ± 2 µM) of the WT STIL CCD peptide. The higher-order oligomers result in increased thermal stability and cooperativity of association. We suggest that this complex oligomerization mechanism regulates the activated levels of STIL in the cell and during centriole duplication. In addition, we present X-ray crystal structures for the CCD containing destabilising (L736E) and stabilising (Q729L) mutations, which reveal dimeric and tetrameric antiparallel coiled-coil structures, respectively. Overall, this study offers a basis for understanding the structural molecular biology of the STIL protein, and how it might be targeted to discover anti-cancer reagents.
Collapse
Affiliation(s)
- Mai Shamir
- Institute of Chemistry, The Hebrew University of Jerusalem, Safra Campus Givat Ram, Jerusalem 91904, Israel;
| | - Freddie J. O. Martin
- School of Chemistry, University of Bristol, Cantock’s Close, Bristol BS8 1TS, UK;
| | - Derek N. Woolfson
- School of Chemistry, University of Bristol, Cantock’s Close, Bristol BS8 1TS, UK;
- School of Biochemistry, University of Bristol, Biomedical Sciences Building, University Walk, Bristol BS8 1TD, UK
- Bristol BioDesign Institute, University of Bristol, Cantock’s Close, Bristol BS8 1TS, UK
| | - Assaf Friedler
- Institute of Chemistry, The Hebrew University of Jerusalem, Safra Campus Givat Ram, Jerusalem 91904, Israel;
| |
Collapse
|
10
|
Bergwell M, Smith A, Smith E, Dierlam C, Duran R, Haastrup E, Napier-Jameson R, Seidel R, Potter W, Norris A, Iyer J. A primary microcephaly-associated sas-6 mutation perturbs centrosome duplication, dendrite morphogenesis, and ciliogenesis in Caenorhabditis elegans. Genetics 2023; 224:iyad105. [PMID: 37279547 PMCID: PMC10411591 DOI: 10.1093/genetics/iyad105] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 06/08/2023] Open
Abstract
The human SASS6(I62T) missense mutation has been linked with the incidence of primary microcephaly in a Pakistani family, although the mechanisms by which this mutation causes disease remain unclear. The SASS6(I62T) mutation corresponds to SAS-6(L69T) in Caenorhabditis elegans. Given that SAS-6 is highly conserved, we modeled this mutation in C. elegans and examined the sas-6(L69T) effect on centrosome duplication, ciliogenesis, and dendrite morphogenesis. Our studies revealed that all the above processes are perturbed by the sas-6(L69T) mutation. Specifically, C. elegans carrying the sas-6(L69T) mutation exhibit an increased failure of centrosome duplication in a sensitized genetic background. Further, worms carrying this mutation also display shortened phasmid cilia, an abnormal phasmid cilia morphology, shorter phasmid dendrites, and chemotaxis defects. Our data show that the centrosome duplication defects caused by this mutation are only uncovered in a sensitized genetic background, indicating that these defects are mild. However, the ciliogenesis and dendritic defects caused by this mutation are evident in an otherwise wild-type background, indicating that they are stronger defects. Thus, our studies shed light on the novel mechanisms by which the sas-6(L69T) mutation could contribute to the incidence of primary microcephaly in humans.
Collapse
Affiliation(s)
- Mary Bergwell
- Oklahoma Medical Research Foundation, Cell Cycle & Cancer Biology Research Program, Oklahoma City, OK 73104, USA
| | - Amy Smith
- Pfizer Inc., Pharmaceutical R&D – Drug Product Design & Development, Chesterfield, MO 63017, USA
| | - Ellie Smith
- University of Tulsa, Department of Chemistry and Biochemistry, Tulsa, OK 74104, USA
| | - Carter Dierlam
- University of Tulsa, Department of Chemistry and Biochemistry, Tulsa, OK 74104, USA
| | - Ramon Duran
- University of Tulsa, Department of Chemistry and Biochemistry, Tulsa, OK 74104, USA
| | - Erin Haastrup
- University of Tulsa, Department of Chemistry and Biochemistry, Tulsa, OK 74104, USA
| | | | - Rory Seidel
- University of Tulsa, Department of Chemistry and Biochemistry, Tulsa, OK 74104, USA
| | - William Potter
- University of Tulsa, Department of Chemistry and Biochemistry, Tulsa, OK 74104, USA
| | - Adam Norris
- Southern Methodist University, Department of Biological Sciences, Dallas, TX 75275, USA
| | - Jyoti Iyer
- University of Tulsa, Department of Chemistry and Biochemistry, Tulsa, OK 74104, USA
| |
Collapse
|
11
|
Li J, Qi Y, Li B, Liu Y, Yang K, Zhang Z, Zhu J, Du E. STIL/AURKA axis promotes cell proliferation by influencing primary cilia formation in bladder cancer. J Transl Med 2023; 21:281. [PMID: 37101292 PMCID: PMC10131372 DOI: 10.1186/s12967-023-04118-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 04/09/2023] [Indexed: 04/28/2023] Open
Abstract
BACKGROUND The primary cilia (PC) is a microtubule-based and nonmotile organelle which protrudes from the surface of almost all mammalian cells. At present, PC has been found to be a deficiency or loss in multiple cancers. Restoring PC could be a novel targeting therapy strategy. Our research showed that PC was reduced in human bladder cancer (BLCA) cells, and PC deficiency promotes cell proliferation. However, the concrete mechanisms remain unknown. SCL/TAL1 interrupting locus (STIL), a PC-related protein, was screened in our previous study and could influence the cell cycle by regulating PC in tumor cells. In this study, we aimed to elucidate the function of STIL for PC to explore the underlying mechanism of PC in BLCA. METHODS Public database analysis, western blot, and enzyme-linked immunosorbent assay (ELISA) were used to screen genes and explore gene expression alteration. Immunofluorescence and western blot were utilized to investigate PC. Wound healing assay, clone formation assay, and CCK-8 assay were used to explore cell migration, growth, and proliferation. The co-immunoprecipitation and western blot were employed to reveal the interaction of STIL and AURKA. RESULTS We found that high STIL expression is correlated with poor outcomes of BLCA patients. Further analysis revealed that STIL overexpression could inhibit PC formation, activate SHH signaling pathways, and promote cell proliferation. In contrast, STIL-knockdown could promote PC formation, inactivate SHH signaling, and inhibit cell proliferation. Furthermore, we found that the regulatory functions of STIL for PC depend on AURKA. STIL could influence proteasome activity and maintain AURKA stabilization. AURKA-knockdown could reverse PC deficiency caused by STIL overexpression for PC in BLCA cells. We observed that co-knockdown in STIL and AURKA significantly enhanced PC assembly. CONCLUSION In summary, our result provides a potential therapy target for BLCA based on the restoration of PC.
Collapse
Affiliation(s)
- Jingxian Li
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Yuanjiong Qi
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Bo Li
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Yan Liu
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Kuo Yang
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Zhihong Zhang
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China.
| | - Jianqiang Zhu
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China.
| | - E Du
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China.
| |
Collapse
|
12
|
Xu L, Zhang S, Feng J, Tan D, Sun H, Guo H. ncRNAs-mediated overexpression of STIL predict unfavorable prognosis and correlated with the efficacy of immunotherapy of hepatocellular carcinoma. Cancer Cell Int 2023; 23:44. [PMID: 36899391 PMCID: PMC10007768 DOI: 10.1186/s12935-023-02869-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 02/13/2023] [Indexed: 03/12/2023] Open
Abstract
BACKGROUND STIL centriolar assembly protein (STIL) is a cytoplasmic protein implicated in cellular growth and proliferation as well as chromosomal stability, which abnormal condition affected tumor immunity and tumor progression. However, the role of STIL in the biological mechanism of hepatocellular carcinoma (HCC) remains unclear. METHODS Comprehensive bioinformatic approaches, in vitro functional assays, and validation were conducted to elucidate the oncogenic value of STIL in HCC. RESULTS In the present study, we found that STIL may serve as an independent prognostic indicator and a potential oncogene in HCC. Gene set enrichment analysis (GSEA), and Gene set variation analysis (GSVA) showed that upregulated expression of STIL was positively associated with pathways enriched in the cell cycle and DNA damage response. Subsequently, we identified several non-coding RNAs (ncRNAs) accounting for the upregulation of STIL expression using a combination of in silico bioinformatics approaches (including expression analysis, correlation analysis, and survival analysis). Finally, CCNT2-AS1/SNHG1-has-miR-204-5p-STIL axis was screened out as the most potential upstream ncRNA-related pathway of STIL in HCC. Moreover, STIL expression is highly associated with the infiltration of immune cells, the expression of immune checkpoints, as well as the survival benefit of immunotherapy/chemotherapy. CONCLUSIONS Our study discloses that ncRNAs-mediated overexpression of STIL independently predicted poor prognosis and correlated with the efficacy of PD-1-targeted immunotherapy in HCC.
Collapse
Affiliation(s)
- Longwen Xu
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China
| | - Shirong Zhang
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China
| | - Jinteng Feng
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China
| | - Deli Tan
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China
| | - Hong Sun
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China
| | - Hui Guo
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China.
| |
Collapse
|
13
|
Li J, Yang Z, Qi Y, Liu X, Liu Y, Gao X, Li S, Zhu J, Zhang C, Du E, Zhang Z. STIL Acts as an Oncogenetic Driver in a Primary Cilia-Dependent Manner in Human Cancer. Front Cell Dev Biol 2022; 10:804419. [PMID: 35155425 PMCID: PMC8826476 DOI: 10.3389/fcell.2022.804419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 01/07/2022] [Indexed: 11/13/2022] Open
Abstract
SCL/TAL1 Interrupting locus (STIL) is a ciliary-related gene involved in regulating the cell cycle and duplication of centrioles in dividing cells. STIL has been found disordered in multiple cancers and driven carcinogenesis. However, the molecular mechanisms and biological functions of STIL in cancers remain ambiguous. Here, we systematically analyzed the genetic alterations, molecular mechanisms, and clinical relevance of STIL across >10,000 samples representing 33 cancer types in The Cancer Genome Atlas (TCGA) dataset. We found that STIL expression is up-regulated in most cancer types compared with their adjacent normal tissues. The expression dysregulation of STIL was affected by copy number variation, mutation, and DNA methylation. High STIL expression was associated with worse outcomes and promoted the progression of cancers. Gene Ontology (GO) enrichment analysis and Gene Set Variation Analysis (GSVA) further revealed that STIL is involved in cell cycle progression, Mitotic spindle, G2M checkpoint, and E2F targets pathways across cancer types. STIL expression was negatively correlated with multiple genes taking part in ciliogenesis and was positively correlated with several genes which participated with centrosomal duplication or cilia degradation. Moreover, STIL silencing could promote primary cilia formation and inhibit cell cycle protein expression in prostate and kidney cancer cell lines. The phenotype and protein expression alteration due to STIL silencing could be reversed by IFT88 silencing in cancer cells. These results revealed that STIL could regulate the cell cycle through primary cilia in tumor cells. In summary, our results revealed the importance of STIL in cancers. Targeting STIL might be a novel therapeutic approach for cancers.
Collapse
Affiliation(s)
- Jingxian Li
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Zikun Yang
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Yuanjiong Qi
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Xun Liu
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Yang Liu
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Xinyu Gao
- Department of Graduate School, Tianjin Medical University, Tianjin, China
| | - Shuai Li
- Department of Graduate School, Tianjin Medical University, Tianjin, China
| | - Jianqiang Zhu
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Changwen Zhang
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - E Du
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
- *Correspondence: E Du, ; Zhihong Zhang,
| | - Zhihong Zhang
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
- *Correspondence: E Du, ; Zhihong Zhang,
| |
Collapse
|
14
|
Abstract
In this review, Phan et al. discuss the different models that have been proposed to explain how centrosome dysfunction impairs cortical development, and review the evidence supporting a unified model in which centrosome defects reduce cell proliferation in the developing cortex by prolonging mitosis and activating a mitotic surveillance pathway. Last, they also extend their discussion to centrosome-independent microcephaly mutations, such as those involved in DNA replication and repair Primary microcephaly is a brain growth disorder characterized by a severe reduction of brain size and thinning of the cerebral cortex. Many primary microcephaly mutations occur in genes that encode centrosome proteins, highlighting an important role for centrosomes in cortical development. Centrosomes are microtubule organizing centers that participate in several processes, including controlling polarity, catalyzing spindle assembly in mitosis, and building primary cilia. Understanding which of these processes are altered and how these disruptions contribute to microcephaly pathogenesis is a central unresolved question. In this review, we revisit the different models that have been proposed to explain how centrosome dysfunction impairs cortical development. We review the evidence supporting a unified model in which centrosome defects reduce cell proliferation in the developing cortex by prolonging mitosis and activating a mitotic surveillance pathway. Finally, we also extend our discussion to centrosome-independent microcephaly mutations, such as those involved in DNA replication and repair.
Collapse
|
15
|
Pradhan T, Kumar V, Surya H E, Krishna R, John S, Jissa VT, Anjana S, Chandramohan K, Nair SA. STIL Endows Oncogenic and Stem-Like Attributes to Colorectal Cancer Plausibly by Shh and Wnt Signaling. Front Oncol 2021; 11:581671. [PMID: 34485108 PMCID: PMC8416176 DOI: 10.3389/fonc.2021.581671] [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: 07/09/2020] [Accepted: 07/21/2021] [Indexed: 11/28/2022] Open
Abstract
The discovery of a potent gene regulating tumorigenesis and drug resistance is of high clinical importance. STIL is an oncogene; however, its molecular associations and role in colorectal oncogenesis are unknown. In this study, we have explored the role of STIL gene in tumorigenesis and studied its molecular targets in colorectal cancer (CRC). STIL silencing reduced proliferation and tumor growth in CRC. Further, STIL was found to regulate stemness markers CD133 and CD44 and drug resistant markers thymidylate synthase, ABCB1, and ABCG2 both in in-vitro and in-vivo CRC models. In addition, high expression of STIL mRNA was found to be associated with reduced disease-free survival in CRC cases. Interestingly, we observed that STIL-mediated regulation of stemness and drug resistant genes is not exclusively governed by Sonic hedgehog (Shh) signaling. Remarkably, we found STIL regulate β-catenin levels through p-AKT, independent of Shh pathway. This partially answers Shh independent regulatory mechanism of cancer stem cell (CSC) markers by STIL. Our study suggests an instrumental role of STIL in molecular manifestation of CRC and progression.
Collapse
Affiliation(s)
- Tapas Pradhan
- Cancer Research Program 4, Rajiv Gandhi Centre for Biotechnology, Trivandrum, India
| | - Vikas Kumar
- Cardiovascular Diseases & Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Trivandrum, India
| | - Evangeline Surya H
- Cancer Research Program 4, Rajiv Gandhi Centre for Biotechnology, Trivandrum, India
| | - R Krishna
- Cancer Research Program 4, Rajiv Gandhi Centre for Biotechnology, Trivandrum, India
| | - Samu John
- Cancer Research Program 4, Rajiv Gandhi Centre for Biotechnology, Trivandrum, India
| | - V T Jissa
- Achutha Menon Centre for Health Science Studies (AMCHSS), Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, India
| | - S Anjana
- Cancer Research Program 4, Rajiv Gandhi Centre for Biotechnology, Trivandrum, India
| | - K Chandramohan
- Department of Surgical Oncology, Regional Cancer Centre, Trivandrum, India
| | - S Asha Nair
- Cancer Research Program 4, Rajiv Gandhi Centre for Biotechnology, Trivandrum, India
| |
Collapse
|
16
|
Integration of multiomics data with graph convolutional networks to identify new cancer genes and their associated molecular mechanisms. NAT MACH INTELL 2021. [DOI: 10.1038/s42256-021-00325-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
17
|
Zhou X, Zhi Y, Yu J, Xu D. The Yin and Yang of Autosomal Recessive Primary Microcephaly Genes: Insights from Neurogenesis and Carcinogenesis. Int J Mol Sci 2020; 21:ijms21051691. [PMID: 32121580 PMCID: PMC7084222 DOI: 10.3390/ijms21051691] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/23/2020] [Accepted: 02/26/2020] [Indexed: 12/26/2022] Open
Abstract
The stem cells of neurogenesis and carcinogenesis share many properties, including proliferative rate, an extensive replicative potential, the potential to generate different cell types of a given tissue, and an ability to independently migrate to a damaged area. This is also evidenced by the common molecular principles regulating key processes associated with cell division and apoptosis. Autosomal recessive primary microcephaly (MCPH) is a neurogenic mitotic disorder that is characterized by decreased brain size and mental retardation. Until now, a total of 25 genes have been identified that are known to be associated with MCPH. The inactivation (yin) of most MCPH genes leads to neurogenesis defects, while the upregulation (yang) of some MCPH genes is associated with different kinds of carcinogenesis. Here, we try to summarize the roles of MCPH genes in these two diseases and explore the underlying mechanisms, which will help us to explore new, attractive approaches to targeting tumor cells that are resistant to the current therapies.
Collapse
Affiliation(s)
- Xiaokun Zhou
- College of Biological Science and Engineering, Institute of Life Sciences, Fuzhou University, Fuzhou 350108, China; (X.Z.); (Y.Z.); (J.Y.)
| | - Yiqiang Zhi
- College of Biological Science and Engineering, Institute of Life Sciences, Fuzhou University, Fuzhou 350108, China; (X.Z.); (Y.Z.); (J.Y.)
| | - Jurui Yu
- College of Biological Science and Engineering, Institute of Life Sciences, Fuzhou University, Fuzhou 350108, China; (X.Z.); (Y.Z.); (J.Y.)
| | - Dan Xu
- College of Biological Science and Engineering, Institute of Life Sciences, Fuzhou University, Fuzhou 350108, China; (X.Z.); (Y.Z.); (J.Y.)
- Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou 350005, China
- Correspondence: ; Tel.: +86-17085937559
| |
Collapse
|
18
|
|
19
|
Vandervore LV, Schot R, Kasteleijn E, Oegema R, Stouffs K, Gheldof A, Grochowska MM, van der Sterre MLT, van Unen LMA, Wilke M, Elfferich P, van der Spek PJ, Heijsman D, Grandone A, Demmers JAA, Dekkers DHW, Slotman JA, Kremers GJ, Schaaf GJ, Masius RG, van Essen AJ, Rump P, van Haeringen A, Peeters E, Altunoglu U, Kalayci T, Poot RA, Dobyns WB, Bahi-Buisson N, Verheijen FW, Jansen AC, Mancini GMS. Heterogeneous clinical phenotypes and cerebral malformations reflected by rotatin cellular dynamics. Brain 2019; 142:867-884. [PMID: 30879067 PMCID: PMC6439326 DOI: 10.1093/brain/awz045] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 11/26/2018] [Accepted: 01/07/2019] [Indexed: 12/16/2022] Open
Abstract
Recessive mutations in RTTN, encoding the protein rotatin, were originally identified as cause of polymicrogyria, a cortical malformation. With time, a wide variety of other brain malformations has been ascribed to RTTN mutations, including primary microcephaly. Rotatin is a centrosomal protein possibly involved in centriolar elongation and ciliogenesis. However, the function of rotatin in brain development is largely unknown and the molecular disease mechanism underlying cortical malformations has not yet been elucidated. We performed both clinical and cell biological studies, aimed at clarifying rotatin function and pathogenesis. Review of the 23 published and five unpublished clinical cases and genomic mutations, including the effect of novel deep intronic pathogenic mutations on RTTN transcripts, allowed us to extrapolate the core phenotype, consisting of intellectual disability, short stature, microcephaly, lissencephaly, periventricular heterotopia, polymicrogyria and other malformations. We show that the severity of the phenotype is related to residual function of the protein, not only the level of mRNA expression. Skin fibroblasts from eight affected individuals were studied by high resolution immunomicroscopy and flow cytometry, in parallel with in vitro expression of RTTN in HEK293T cells. We demonstrate that rotatin regulates different phases of the cell cycle and is mislocalized in affected individuals. Mutant cells showed consistent and severe mitotic failure with centrosome amplification and multipolar spindle formation, leading to aneuploidy and apoptosis, which could relate to depletion of neuronal progenitors often observed in microcephaly. We confirmed the role of rotatin in functional and structural maintenance of primary cilia and determined that the protein localized not only to the basal body, but also to the axoneme, proving the functional interconnectivity between ciliogenesis and cell cycle progression. Proteomics analysis of both native and exogenous rotatin uncovered that rotatin interacts with the neuronal (non-muscle) myosin heavy chain subunits, motors of nucleokinesis during neuronal migration, and in human induced pluripotent stem cell-derived bipolar mature neurons rotatin localizes at the centrosome in the leading edge. This illustrates the role of rotatin in neuronal migration. These different functions of rotatin explain why RTTN mutations can lead to heterogeneous cerebral malformations, both related to proliferation and migration defects.
Collapse
Affiliation(s)
- Laura V Vandervore
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands.,Neurogenetics Research Group, Research Cluster Reproduction, Genetics and Regenerative Medicine, Vrije Universiteit Brussel, Brussels, Belgium.,Center for Medical Genetics, UZ Brussel, Brussels, Belgium
| | - Rachel Schot
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Esmee Kasteleijn
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Renske Oegema
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands.,Department of Pathology, Clinical Bio-informatics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Katrien Stouffs
- Neurogenetics Research Group, Research Cluster Reproduction, Genetics and Regenerative Medicine, Vrije Universiteit Brussel, Brussels, Belgium.,Center for Medical Genetics, UZ Brussel, Brussels, Belgium
| | - Alexander Gheldof
- Neurogenetics Research Group, Research Cluster Reproduction, Genetics and Regenerative Medicine, Vrije Universiteit Brussel, Brussels, Belgium.,Center for Medical Genetics, UZ Brussel, Brussels, Belgium
| | - Martyna M Grochowska
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Marianne L T van der Sterre
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Leontine M A van Unen
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Martina Wilke
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Peter Elfferich
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Peter J van der Spek
- Dipartimento della Donna, del Bambino, di Chirurgia Generale e Specialistica, Seconda Università degli studi della Campania "L. Vanvitelli", Naples, Italy
| | - Daphne Heijsman
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands.,Dipartimento della Donna, del Bambino, di Chirurgia Generale e Specialistica, Seconda Università degli studi della Campania "L. Vanvitelli", Naples, Italy
| | - Anna Grandone
- Department of Molecular Genetics, Proteomics Center, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Jeroen A A Demmers
- Department of Pathology, Optical Imaging Center, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Dick H W Dekkers
- Department of Pathology, Optical Imaging Center, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Johan A Slotman
- Center for Lysosomal and Metabolic Diseases, Erasmus Medical Center (Erasmus MC), 3015 CN Rotterdam, The Netherlands
| | - Gert-Jan Kremers
- Center for Lysosomal and Metabolic Diseases, Erasmus Medical Center (Erasmus MC), 3015 CN Rotterdam, The Netherlands
| | - Gerben J Schaaf
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands.,Department of Genetics, University of Groningen, University Medical Center Groningen, RB, Groningen, The Netherlands
| | - Roy G Masius
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Anton J van Essen
- Department of Clinical Genetics, LUMC, Leiden University Medical Center, Postzone K-5-R, Postbus 9600, RC Leiden, The Netherlands
| | - Patrick Rump
- Department of Clinical Genetics, LUMC, Leiden University Medical Center, Postzone K-5-R, Postbus 9600, RC Leiden, The Netherlands
| | - Arie van Haeringen
- Department of Pediatric Neurology, Juliana Hospital, Els Borst-Eilersplein 275, 2545 AA Den Haag, The Netherlands
| | - Els Peeters
- Department of Medical genetics, Istanbul Medical Faculty, Istanbul University, Topkapı Mahallesi, Turgut Özal Millet Cd, 34093 Fatih/İstanbul, Turkey
| | - Umut Altunoglu
- Department of Cell biology, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Tugba Kalayci
- Department of Cell biology, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Raymond A Poot
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - William B Dobyns
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA.,Imagine Institute, INSERM UMR-1163, Laboratory Genetics and Embryology of Congenital Malformations, Paris Descartes University, Institut des Maladies Génétiques 24, Boulevard de Montparnasse, Paris, France
| | - Nadia Bahi-Buisson
- Pediatric Neurology Unit, Department of Pediatrics, UZ Brussel, Brussels, Belgium
| | - Frans W Verheijen
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Anna C Jansen
- Neurogenetics Research Group, Research Cluster Reproduction, Genetics and Regenerative Medicine, Vrije Universiteit Brussel, Brussels, Belgium.,Center for Medical Genetics, UZ Brussel, Brussels, Belgium
| | - Grazia M S Mancini
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| |
Collapse
|
20
|
Ito H, Tsunoda T, Riku M, Inaguma S, Inoko A, Murakami H, Ikeda H, Matsuda M, Kasai K. Indispensable role of STIL in the regulation of cancer cell motility through the lamellipodial accumulation of ARHGEF7-PAK1 complex. Oncogene 2019; 39:1931-1943. [PMID: 31754215 DOI: 10.1038/s41388-019-1115-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 11/06/2019] [Accepted: 11/11/2019] [Indexed: 01/08/2023]
Abstract
Cell motility is a tightly regulated phenomenon that supports the accurate formation of organ structure during development and homeostasis, including wound healing and inflammation. Meanwhile, cancer cells exhibit dysregulated motility, which causes spreading and invasion. The Dbl family RhoGEF ARHGEF7/β-PIX and its binding partner p21-activated kinase PAK1 are overexpressed in a variety of cancers and have been shown to be responsible for cancer cell migration. A key step in motility is the intracellular transport of ARHGEF7-PAK1 complex to the migrating front of cells, where lamellipodia protrusion and cytoskeletal remodeling efficiently occur. However, the molecular mechanisms of the intracellular transport of this complex are not fully understood. Here we revealed that SCL/TAL1-interrupting locus (STIL) is indispensable for the efficient migration of cancer cells. STIL forms a ternary complex with ARHGEF7 and PAK1 and accumulates with those proteins at the lamellipodia protrusion of motile cells. Knockdown of STIL impedes the accumulation of ARHGEF7-PAK1 complex within membrane ruffles and attenuates the phosphorylation of PAK1 substrates and cortical actin remodeling at the migrating front. Intriguingly, ARHGEF7 knockdown also diminishes STIL and PAK1 accumulation in membrane ruffles. Either STIL or ARHGEF7 knockdown impedes cell migration and Rac1 activity at the migrating front of cells. These results indicate that STIL is involved in the ARHGEF7-mediated positive-feedback activation of cytoskeletal remodeling through accumulating the ARHGEF7-PAK1 complex in lamellipodia. We conclude that its involvement is crucial for the polarized formation of Rac1-mediated leading edge, which supports the efficient migration of cancer cells.
Collapse
Affiliation(s)
- Hideaki Ito
- Department of Pathology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Takumi Tsunoda
- Department of Pathology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Miho Riku
- Department of Pathology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Shingo Inaguma
- Department of Pathology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Akihito Inoko
- Department of Pathology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Hideki Murakami
- Department of Pathology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Hiroshi Ikeda
- Department of Pathology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Michiyuki Matsuda
- Department of Pathology and Biology of Diseases, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kenji Kasai
- Department of Pathology, Aichi Medical University School of Medicine, Nagakute, Japan.
| |
Collapse
|
21
|
Cuenca A, Insinna C, Zhao H, John P, Weiss MA, Lu Q, Walia V, Specht S, Manivannan S, Stauffer J, Peden AA, Westlake CJ. The C7orf43/TRAPPC14 component links the TRAPPII complex to Rabin8 for preciliary vesicle tethering at the mother centriole during ciliogenesis. J Biol Chem 2019; 294:15418-15434. [PMID: 31467083 DOI: 10.1074/jbc.ra119.008615] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 08/25/2019] [Indexed: 01/08/2023] Open
Abstract
The primary cilium is a cellular sensor that detects light, chemicals, and movement and is important for morphogen and growth factor signaling. The small GTPase Rab11-Rab8 cascade is required for ciliogenesis. Rab11 traffics the guanine nucleotide exchange factor (GEF) Rabin8 to the centrosome to activate Rab8, needed for ciliary growth. Rabin8 also requires the transport particle protein complex (TRAPPC) proteins for centrosome recruitment during ciliogenesis. Here, using an MS-based approach for identifying Rabin8-interacting proteins, we identified C7orf43 (also known as microtubule-associated protein 11 (MAP11)) as being required for ciliation both in human cells and zebrafish embryos. We find that C7orf43 directly binds to Rabin8 and that C7orf43 knockdown diminishes Rabin8 preciliary centrosome accumulation. Interestingly, we found that C7orf43 co-sediments with TRAPPII complex subunits and directly interacts with TRAPPC proteins. Our findings establish that C7orf43 is a TRAPPII-specific complex component, referred to here as TRAPPC14. Additionally, we show that TRAPPC14 is dispensable for TRAPPII complex integrity but mediates Rabin8 association with the TRAPPII complex. Finally, we demonstrate that TRAPPC14 interacts with the distal appendage proteins Fas-binding factor 1 (FBF1) and centrosomal protein 83 (CEP83), which we show here are required for GFP-Rabin8 centrosomal accumulation, supporting a role for the TRAPPII complex in tethering preciliary vesicles to the mother centriole during ciliogenesis. In summary, our findings have revealed an uncharacterized TRAPPII-specific component, C7orf43/TRAPPC14, that regulates preciliary trafficking of Rabin8 and ciliogenesis and support previous findings that the TRAPPII complex functions as a membrane tether.
Collapse
Affiliation(s)
- Adrian Cuenca
- Center for Cancer Research, NCI-Frederick, National Institutes of Health, Laboratory of Cellular and Developmental Signaling, Frederick, Maryland 21702
| | - Christine Insinna
- Center for Cancer Research, NCI-Frederick, National Institutes of Health, Laboratory of Cellular and Developmental Signaling, Frederick, Maryland 21702
| | - Huijie Zhao
- Center for Cancer Research, NCI-Frederick, National Institutes of Health, Laboratory of Cellular and Developmental Signaling, Frederick, Maryland 21702
| | - Peter John
- Center for Cancer Research, NCI-Frederick, National Institutes of Health, Laboratory of Cellular and Developmental Signaling, Frederick, Maryland 21702
| | - Matthew A Weiss
- Center for Cancer Research, NCI-Frederick, National Institutes of Health, Laboratory of Cellular and Developmental Signaling, Frederick, Maryland 21702
| | - Quanlong Lu
- Center for Cancer Research, NCI-Frederick, National Institutes of Health, Laboratory of Cellular and Developmental Signaling, Frederick, Maryland 21702
| | - Vijay Walia
- Center for Cancer Research, NCI-Frederick, National Institutes of Health, Laboratory of Cellular and Developmental Signaling, Frederick, Maryland 21702
| | - Suzanne Specht
- Center for Cancer Research, NCI-Frederick, National Institutes of Health, Laboratory of Cellular and Developmental Signaling, Frederick, Maryland 21702
| | - Selvambigai Manivannan
- Department of Biomedical Sciences, University of Sheffield, Sheffield S10 2TN, United Kingdom
| | - Jimmy Stauffer
- Center for Cancer Research, NCI-Frederick, National Institutes of Health, Laboratory of Cellular and Developmental Signaling, Frederick, Maryland 21702
| | - Andrew A Peden
- Department of Biomedical Sciences, University of Sheffield, Sheffield S10 2TN, United Kingdom
| | - Christopher J Westlake
- Center for Cancer Research, NCI-Frederick, National Institutes of Health, Laboratory of Cellular and Developmental Signaling, Frederick, Maryland 21702
| |
Collapse
|
22
|
Kodani A, Moyer T, Chen A, Holland A, Walsh CA, Reiter JF. SFI1 promotes centriole duplication by recruiting USP9X to stabilize the microcephaly protein STIL. J Cell Biol 2019; 218:2185-2197. [PMID: 31197030 PMCID: PMC6605807 DOI: 10.1083/jcb.201803041] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 12/18/2018] [Accepted: 05/10/2019] [Indexed: 01/08/2023] Open
Abstract
In mammals, centrioles participate in brain development, and human mutations affecting centriole duplication cause microcephaly. Here, we identify a role for the mammalian homologue of yeast SFI1, involved in the duplication of the yeast spindle pole body, as a critical regulator of centriole duplication in mammalian cells. Mammalian SFI1 interacts with USP9X, a deubiquitylase associated with human syndromic mental retardation. SFI1 localizes USP9X to the centrosome during S phase to deubiquitylate STIL, a critical regulator of centriole duplication. USP9X-mediated deubiquitylation protects STIL from degradation. Consistent with a role for USP9X in stabilizing STIL, cells from patients with USP9X loss-of-function mutations have reduced STIL levels. Together, these results demonstrate that SFI1 is a centrosomal protein that localizes USP9X to the centrosome to stabilize STIL and promote centriole duplication. We propose that the USP9X protection of STIL to facilitate centriole duplication underlies roles of both proteins in human neurodevelopment.
Collapse
Affiliation(s)
- Andrew Kodani
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
- Division of Genetics and Genomics and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA
| | - Tyler Moyer
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Allen Chen
- Division of Genetics and Genomics and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA
| | - Andrew Holland
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Christopher A Walsh
- Division of Genetics and Genomics and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA
| | - Jeremy F Reiter
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
23
|
Wang J, Zhang Y, Dou Z, Jiang H, Wang Y, Gao X, Xin X. Knockdown of STIL suppresses the progression of gastric cancer by down-regulating the IGF-1/PI3K/AKT pathway. J Cell Mol Med 2019; 23:5566-5575. [PMID: 31187582 PMCID: PMC6653615 DOI: 10.1111/jcmm.14440] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 05/08/2019] [Accepted: 05/16/2019] [Indexed: 02/05/2023] Open
Abstract
SCL/TAL1 interrupting locus (STIL) regulates the mitotic centrosome to promote the centriolar replication and cell cycling, and is associated with malignancies. However, the role and mechanism of STIL in gastric cancer (GC) remain elusive. STIL expression in GC tissue microarray was detected by immunohistochemistry (IHC). GC cells were transduced with control lentivirus or lentivirus for expression STIL‐specific shRNA and the effect of STIL silencing on the malignant behaviors of GC cells was measured in vitro and in vivo. The potential mechanisms underlying the action of STIL were analyzed by transcriptome microarray and bioinformatics. STIL expression was up‐regulated in GC tissues both in our cohort and the data from the cancer genome atlas, and positively associated with T stage and poor overall survival of GC patients. Knockdown of STIL significantly inhibited the proliferation and clonogenicity of human GC cells and attenuated the growth of implanted GC in vivo. Furthermore, STIL silencing induced cell cycle arrest in G2/M phase and apoptosis of GC cells. Transcriptome analysis indicated that STIL silencing modulated many gene expression, particularly for down‐regulating the IGF‐1/PI3K/AKT pathway. In addition, treatment with SC79, an AKT activator, significantly mitigated the effect of STIL‐silencing in GC cells. In conclusion, STIL promotes gastric carcinogenesis and progression by enhancing the IGF‐1/PI3K/AKT signaling, and STIL may be a novel target for intervention of GC.
Collapse
Affiliation(s)
- Ju Wang
- Department of Gastrointestinal Surgery, Inner Mongolia People's Hospital, Hohhot, China
| | - Yong Zhang
- Lung Cancer Centre, State Key Laboratory of Biotherapy and Cancer Centre, West China Hospital, Sichuan University, Chengdu, China
| | - Zhongxia Dou
- Department of Gastrointestinal Surgery, Inner Mongolia People's Hospital, Hohhot, China
| | - Hongwei Jiang
- Department of Gastrointestinal Surgery, Inner Mongolia People's Hospital, Hohhot, China
| | - Yongqiang Wang
- Department of Gastrointestinal Surgery, Inner Mongolia People's Hospital, Hohhot, China
| | - Xiaoping Gao
- Department of Gastrointestinal Surgery, Inner Mongolia People's Hospital, Hohhot, China
| | - Xiangyang Xin
- Department of Ophthalmology, Inner Mongolia Baogang Hospital, Baotou, China
| |
Collapse
|