1
|
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
|
2
|
Zhu YT, Wu SY, Yang S, Ying J, Tian L, Xu HL, Zhang HP, Yao H, Zhang WY, Jin QQ, Yang YT, Jiang XY, Zhang N, Yao S, Zhou SG, Chen G. Identification and validation of a novel anoikis-related signature for predicting prognosis and immune landscape in ovarian serous cystadenocarcinoma. Heliyon 2023; 9:e18708. [PMID: 37554782 PMCID: PMC10404752 DOI: 10.1016/j.heliyon.2023.e18708] [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: 03/08/2023] [Revised: 07/23/2023] [Accepted: 07/25/2023] [Indexed: 08/10/2023] Open
Abstract
BACKGROUND Ovarian serous cystadenocarcinoma (OSC) is the most prevalent histological subtype of ovarian cancer (OV) and presents a serious threat to women's health. Anoikis is an essential component of metastasis, and tumor cells can get beyond it to become viable. The impact of anoikis on OSC, however, has only been the topic of a few studies. METHODS The mRNA sequencing and clinical information of OSC came from The Cancer Genome Atlas Target Genotype-Tissue Expression (TCGA TARGET GTEx) dataset. Anoikis-related genes (ARGs) were collected by Harmonizome and GeneCards websites. Centered on these ARGs, we used unsupervised consensus clustering to explore potential tumor typing and filtered hub ARGs to create a model of predictive signature for OSC patients. Furthermore, we presented clinical specialists with a novel nomogram based on ARGs, revealing the underlying clinical relevance of this signature. Finally, we explored the immune microenvironment among various risk groups. RESULTS We identified 24 ARGs associated with the prognosis of OSC and classified OSC patients into three subtypes, and the subtype with the best prognosis was more enriched in immune-related pathways. Seven ARGs (ARHGEF7, NOTCH4, CASP2, SKP2, PAK4, LCK, CCDC80) were chosen to establish a risk model and a nomogram that can provide practical clinical decision support. Risk scores were found to be an independent and significant prognostic factor in OSC patients. The CIBERSORTx result revealed an inflammatory microenvironment is different for risk groups, and the proportion of immune infiltrates of Macrophages M1 is negatively correlated with risk score (rs = -0.21, P < 0.05). Ultimately, quantitative reverse transcription polymerase chain reaction (RT-PCR) was utilized to validate the expression of the seven pivotal ARGs. CONCLUSION In this study, based on seven ARGs, a risk model and nomogram established can be used for risk stratification and prediction of survival outcomes in patients with OSC, providing a reliable reference for individualized therapy of OSC patients.
Collapse
Affiliation(s)
- Yu-Ting Zhu
- Department of Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
- Department of Gynecology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - Shuang-Yue Wu
- Department of Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
- Department of Gynecology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - Song Yang
- Department of Pain Treatment, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Jie Ying
- Department of Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
- Department of Gynecology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - Lu Tian
- Department of Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
- Department of Gynecology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - Hong-Liang Xu
- Department of Pathology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - He-Ping Zhang
- Department of Pathology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - Hui Yao
- Department of Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
- Department of Gynecology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - Wei-Yu Zhang
- Department of Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
- Department of Gynecology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - Qin-Qin Jin
- Department of Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
- Department of Gynecology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - Yin-Ting Yang
- Department of Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
- Department of Gynecology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - Xi-Ya Jiang
- Department of Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
- Department of Gynecology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - Nan Zhang
- Department of Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
- Department of Gynecology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - Shun Yao
- Department of Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
- Department of Gynecology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - Shu-Guang Zhou
- Department of Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
- Department of Gynecology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - Guo Chen
- Department of Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
- Department of Gynecology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| |
Collapse
|
3
|
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
|
4
|
Chen B, Mao T, Qin X, Zhang W, Watanabe N, Li J. Role of estrogen receptor signaling pathway-related genes in diffuse large B-cell lymphoma and identification of key targets via integrated bioinformatics analysis and experimental validation. Front Oncol 2022; 12:1029998. [PMID: 36531013 PMCID: PMC9749266 DOI: 10.3389/fonc.2022.1029998] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 11/07/2022] [Indexed: 11/17/2023] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is a highly heterogeneous malignancy. Epidemiologically, the incidence of DLBCL is higher in men, and the female sex is a favorable prognostic factor, which can be explained by estrogen. This study aimed to explore the potential targets of the estrogen receptor (ER) signaling pathway and provide a meaningful way to treat DLBCL patients. Datasets were obtained from the Gene Expression Omnibus (GEO) to identify differentially expressed genes (DEGs). Representative gene sets estrogen receptor pathways, and growth regulatory pathways were identified based on Gene Set Enrichment Analysis (GSEA) analysis. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) were used for function and pathway analysis. STRING and Cytoscape were used to construct the interaction network, and the MCODE plug-in performed the module analysis. GEPIA, TCGA, and LOGpc databases were used for expression and predictive analysis. The Human Protein Atlas (HPA) database was used to analyze the protein expression levels, cBioPortal was used to explore genetic alterations, and ROC analysis and prognostic assessment were used to predict the diagnostic value of genes. Finally, BJAB cells were treated with ER inhibitor fulvestrant and specific shRNA, and the expression of hub genes was verified by RT-qPCR. We identified 81 overlapping DEGs and CDC6, CDC20, KIF20A, STIL, and TOP2A as novel biomarkers affecting the prognosis of DLBCL. In addition, the STAT and KRAS pathways are considered potential growth regulatory pathways. These results hold promise for new avenues for the treatment of DLBCL patients.
Collapse
Affiliation(s)
- Bo Chen
- Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Tianjiao Mao
- Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiuni Qin
- Guangzhou Concord Cancer Center, Guangzhou, Guangdong, China
| | - Wenqi Zhang
- School of Basic Medicine, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Nobumoto Watanabe
- Chemical Biology Research Group, RIKEN Center for Sustainable Resource Science, Wako, Saitama, Japan
- Bio-Active Compounds Discovery Unit, RIKEN Center for Sustainable Resource Science, Wako, Saitama, Japan
| | - Jiang Li
- Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
5
|
STIL Promotes Tumorigenesis of Bladder Cancer by Activating PI3K/AKT/mTOR Signaling Pathway and Targeting C-Myc. Cancers (Basel) 2022; 14:cancers14235777. [PMID: 36497260 PMCID: PMC9739707 DOI: 10.3390/cancers14235777] [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: 11/02/2022] [Revised: 11/19/2022] [Accepted: 11/20/2022] [Indexed: 11/25/2022] Open
Abstract
SCL/TAL1 interrupting locus (STIL) regulates centriole replication and causes chromosome instability, which is closely related to malignant tumors. The purpose of our study was to investigate the role of STIL in bladder cancer (BC) tumorigenesis for the first time. The public database indicated that STIL is highly expressed and correlated with the cell cycle in BC. Immunohistochemistry staining showed that STIL expression is significantly elevated in BC tissues compared with paracancer tissues. CRISPR-Cas9 gene editing technology was used to induce BC cells to express STIL-specific sgRNA, revealing a significantly delayed growth rate in STIL knockout BC cells. Moreover, cell cycle arrest in the G0/G1 phase was triggered by decreasing STIL, which led to delayed BC cell growth in vitro and in vivo. Mechanically, STIL knockout inhibited the PI3K/AKT/mTOR pathway and down-regulated the expression of c-myc. Furthermore, SC79 (AKT activating agent) partially reversed the inhibitory effects of STIL knockout on the proliferation and migration of BC cells. In conclusion, STIL enhanced the PI3K/AKT/mTOR pathway, resulting in increased expression of c-myc, ultimately promoting BC occurrence and progression. These results indicate that STIL might be a potential target for BC patients.
Collapse
|
6
|
Keum S, Yang SJ, Park E, Kang T, Choi JH, Jeong J, Hwang YE, Kim JW, Park D, Rhee S. Beta-Pix-dynamin 2 complex promotes colorectal cancer progression by facilitating membrane dynamics. Cell Oncol (Dordr) 2021; 44:1287-1305. [PMID: 34582006 PMCID: PMC8648671 DOI: 10.1007/s13402-021-00637-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 09/14/2021] [Indexed: 12/24/2022] Open
Abstract
PURPOSE Spatiotemporal regulation of cell membrane dynamics is a major process that promotes cancer cell invasion by acting as a driving force for cell migration. Beta-Pix (βPix), a guanine nucleotide exchange factor for Rac1, has been reported to be involved in actin-mediated cellular processes, such as cell migration, by interacting with various proteins. As yet, however, the molecular mechanisms underlying βPix-mediated cancer cell invasion remain unclear. METHODS The clinical significance of βPix was analyzed in patients with colorectal cancer (CRC) using public clinical databases. Pull-down and immunoprecipitation assays were employed to identify novel binding partners for βPix. Additionally, various cell biological assays including immunocytochemistry and time-lapse video microscopy were performed to assess the effects of βPix on CRC progression. A βPix-SH3 antibody delivery system was used to determine the effects of the βPix-Dyn2 complex in CRC cells. RESULTS We found that the Src homology 3 (SH3) domain of βPix interacts with the proline-rich domain of Dynamin 2 (Dyn2), a large GTPase. The βPix-Dyn2 interaction promoted lamellipodia formation, along with plasma membrane localization of membrane-type 1 matrix metalloproteinase (MT1-MMP). Furthermore, we found that Src kinase-mediated phosphorylation of the tyrosine residue at position 442 of βPix enhanced βPix-Dyn2 complex formation. Disruption of the βPix-Dyn2 complex by βPix-SH3 antibodies targeting intracellular βPix inhibited CRC cell invasion. CONCLUSIONS Our data indicate that spatiotemporal regulation of the Src-βPix-Dyn2 axis is crucial for CRC cell invasion by promoting membrane dynamics and MT1-MMP recruitment into the leading edge. The development of inhibitors that disrupt the βPix-Dyn2 complex may be a useful therapeutic strategy for CRC.
Collapse
Affiliation(s)
- Seula Keum
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Soo Jung Yang
- Translational Research Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, 98101, USA
| | - Esther Park
- School of Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - TaeIn Kang
- School of Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jee-Hye Choi
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Jangho Jeong
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Ye Eun Hwang
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Jung-Woong Kim
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Dongeun Park
- School of Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sangmyung Rhee
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea.
| |
Collapse
|
7
|
García-Jiménez I, Cervantes-Villagrana RD, Del-Río-Robles JE, Castillo-Kauil A, Beltrán-Navarro YM, García-Román J, Reyes-Cruz G, Vázquez-Prado J. Gβγ mediates activation of Rho guanine nucleotide exchange factor ARHGEF17 that promotes metastatic lung cancer progression. J Biol Chem 2021; 298:101440. [PMID: 34808208 PMCID: PMC8703085 DOI: 10.1016/j.jbc.2021.101440] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 11/10/2021] [Accepted: 11/18/2021] [Indexed: 12/12/2022] Open
Abstract
Metastatic lung cancer is a major cause of death worldwide. Dissemination of cancer cells can be facilitated by various agonists within the tumor microenvironment, including by lysophosphatidic acid (LPA). We postulate that Rho guanine nucleotide exchange factors (RhoGEFs), which integrate signaling cues driving cell migration, are critical effectors in metastatic cancer. Specifically, we addressed the hypothetical role of ARHGEF17, a RhoGEF, as a potential effector of Gβγ in metastatic lung cancer cells responding to LPA. Here, we show that ARHGEF17, originally identified as a tumor endothelial marker, is involved in tumor growth and metastatic dissemination of lung cancer cells in an immunocompetent murine model. Gene expression–based analysis of lung cancer datasets showed that increased levels of ARHGEF17 correlated with reduced survival of patients with advanced-stage tumors. Cellular assays also revealed that this RhoGEF participates in the invasive and migratory responses elicited by Gi protein–coupled LPA receptors via the Gβγ subunit complex. We demonstrate that this signaling heterodimer promoted ARHGEF17 recruitment to the cell periphery and actin fibers. Moreover, Gβγ allosterically activates ARHGEF17 by the removal of inhibitory intramolecular restrictions. Taken together, our results indicate that ARHGEF17 may be a valid potential target in the treatment of metastatic lung cancer.
Collapse
|
8
|
Cheng K, Larabee SM, Tolaymat M, Hanscom M, Shang AC, Schledwitz A, Hu S, Drachenberg CB, Zhan M, Chahdi A, Raufman JP. Targeted intestinal deletion of Rho guanine nucleotide exchange factor 7, βPIX, impairs enterocyte proliferation, villus maturation, and mucosal defenses in mice. Am J Physiol Gastrointest Liver Physiol 2021; 320:G627-G643. [PMID: 33566751 PMCID: PMC8238171 DOI: 10.1152/ajpgi.00415.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/01/2021] [Accepted: 02/10/2021] [Indexed: 01/31/2023]
Abstract
Rho guanine nucleotide exchange factors (RhoGEFs) regulate Rho GTPase activity and cytoskeletal and cell adhesion dynamics. βPix, a CDC42/RAC family RhoGEF encoded by ARHGEF7, is reported to modulate human colon cancer cell proliferation and postwounding restitution of rat intestinal epithelial monolayers. We hypothesized that βPix plays a role in maintaining intestinal epithelial homeostasis. To test this hypothesis, we examined βPix distribution in the human and murine intestine and created mice with intestinal epithelial-selective βPix deletion [βPixflox/flox/Tg(villin-Cre); Arhgef7 CKO mice]. Using Arhgef7 conditional knockout (CKO) and control mice, we investigated the consequences of βPix deficiency in vivo on intestinal epithelial and enteroid development, dextran sodium sulfate-induced mucosal injury, and gut permeability. In normal human and murine intestines, we observed diffuse cytoplasmic and moderate nuclear βPix immunostaining in enterocytes. Arhgef7 CKO mice were viable and fertile, with normal gross intestinal architecture but reduced small intestinal villus height, villus-to-crypt ratio, and goblet cells; small intestinal crypt cells had reduced Ki67 staining, compatible with impaired cell proliferation. Enteroids derived from control mouse small intestine were viable for more than 20 passages, but those from Arhgef7 CKO mice did not survive beyond 24 h despite addition of Wnt proteins or conditioned media from normal enteroids. Adding a Rho kinase (ROCK) inhibitor partially rescued CKO enteroid development. Compared with littermate control mice, dextran sodium sulfate-treated βPix-deficient mice lost more weight and had greater impairment of intestinal barrier function, and more severe colonic mucosal injury. These findings reveal βPix expression is important for enterocyte development, intestinal homeostasis, and resistance to toxic injury.NEW & NOTEWORTHY To explore the role of βPix, a guanine nucleotide exchange factor encoded by ARHGEF7, in intestinal development and physiology, we created mice with intestinal epithelial cell Arhgef7/βPix deficiency. We found βPix essential for normal small intestinal epithelial cell proliferation, villus development, and mucosal resistance to injury. Moreover, Rho kinase signaling mediated developmental arrest observed in enteroids derived from βPix-deficient small intestinal crypts. Our studies provide insights into the role Arhgef7/βPix plays in intestinal epithelial homeostasis.
Collapse
Affiliation(s)
- Kunrong Cheng
- Veterans Affairs Maryland Healthcare System, Baltimore, Maryland
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Shannon M Larabee
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland
| | - Mazen Tolaymat
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Marie Hanscom
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Aaron C Shang
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Alyssa Schledwitz
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Shien Hu
- Veterans Affairs Maryland Healthcare System, Baltimore, Maryland
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Cinthia B Drachenberg
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Min Zhan
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, Maryland
| | - Ahmed Chahdi
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Jean-Pierre Raufman
- Veterans Affairs Maryland Healthcare System, Baltimore, Maryland
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, Maryland
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
9
|
Li XY, Shi LX, Yao XM, Jing M, Li QQ, Wang YL, Li QS. Functional vinorelbine plus schisandrin B liposomes destroying tumor metastasis in treatment of gastric cancer. Drug Dev Ind Pharm 2021; 47:100-112. [PMID: 33295825 DOI: 10.1080/03639045.2020.1862169] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Gastric cancer is one of the leading causes of cancer-related death worldwide with a poor prognosis. Gastric cancer is usually treated with surgery and chemotherapy, accompanied by a high rate of metastasis and recurrence. In this paper, R8 (RRRRRRRR) modified vinorelbine plus schisandrin B liposomes had been successfully constructed for treating gastric cancer. In the liposomes, R8 was used to enhance the intracellular uptake, schisandrin B was incorporated into liposomes for inhibiting tumor cells metastasis, and vinorelbine was encapsulated into liposomes as antitumor drugs. Studies were performed on BGC-823 cells in vitro and were verified in the BGC-823 cell xenografts nude mice in vivo. Results in vitro demonstrated that the targeting liposomes could induce BGC-823 cells apoptosis, inhibit the metastasis of tumor cells, and increase targeting effects to tumor cells. Meanwhile, action mechanism studies showed that the targeting liposomes could down-regulate VEGF, VE-Cad, HIF-1a, PI3K, MMP-2, and FAK to inhibit tumor metastasis. In vivo results exhibited that the targeting liposomes displayed an obvious antitumor efficacy by accumulating selectively in tumor site and induce tumor cell apoptosis. Hence, R8 modified vinorelbine plus schisandrin B liposomes might provide a safe and efficient therapy strategy for gastric cancer.
Collapse
Affiliation(s)
- Xiu-Ying Li
- School of Pharmacy, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Luan-Xia Shi
- School of Pharmacy, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Xue-Min Yao
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Ming Jing
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Qin-Qing Li
- School of Pharmacy, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Ying-Li Wang
- School of Pharmacy, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Qing-Shan Li
- School of Pharmacy, Shanxi University of Chinese Medicine, Jinzhong, China
| |
Collapse
|
10
|
Connor K, Murray DW, Jarzabek MA, Tran NL, White K, Dicker P, Sweeney KJ, O’Halloran PJ, MacCarthy B, Shiels LP, Lodi F, Lambrechts D, Sarkaria JN, Schiffelers RM, Symons M, Byrne AT. Targeting the RhoGEF βPIX/COOL-1 in Glioblastoma: Proof of Concept Studies. Cancers (Basel) 2020; 12:cancers12123531. [PMID: 33256106 PMCID: PMC7761123 DOI: 10.3390/cancers12123531] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/15/2020] [Accepted: 11/19/2020] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Glioblastoma (GBM) is an incurable disease with a 14-month average life-expectancy following diagnosis, and clinical management has not improved in four decades. GBM mortality is due to rapid tumour growth and invasion into surrounding normal brain. Invasive cells make complete surgical removal of the tumour impossible, and result in disease relapse. Thus, it is imperative that any new treatment strategy takes these invading cells into consideration. Bevacizumab (Bev), which prevents the formation of new blood vessels, is an FDA approved therapy, but it has failed to increase overall survival in GBM and has even been shown to increase tumour invasion in some cases. Complementary anti-invasive therapies are therefore urgently required to enhance bevacizumab efficacy. We have identified βPIX/COOL-1, a RhoGEF protein which plays an important role in GBM cell invasion and angiogenesis and could be a useful target in this setting. Abstract Glioblastoma (GBM), a highly invasive and vascular malignancy is shown to rapidly develop resistance and evolve to a more invasive phenotype following bevacizumab (Bev) therapy. Rho Guanine Nucleotide Exchange Factor proteins (RhoGEFs) are mediators of key components in Bev resistance pathways, GBM and Bev-induced invasion. To identify GEFs with enhanced mRNA expression in the leading edge of GBM tumours, a cohort of GEFs was assessed using a clinical dataset. The GEF βPix/COOL-1 was identified, and the functional effect of gene depletion assessed using 3D-boyden chamber, proliferation, and colony formation assays in GBM cells. Anti-angiogenic effects were assessed in endothelial cells using tube formation and wound healing assays. In vivo effects of βPix/COOL-1-siRNA delivered via RGD-Nanoparticle in combination with Bev was studied in an invasive model of GBM. We found that siRNA-mediated knockdown of βPix/COOL-1 in vitro decreased cell invasion, proliferation and increased apoptosis in GBM cell lines. Moreover βPix/COOL-1 mediated endothelial cell migration in vitro. Mice treated with βPix/COOL-1 siRNA-loaded RGD-Nanoparticle and Bev demonstrated a trend towards improved median survival compared with Bev monotherapy. Our hypothesis generating study suggests that the RhoGEF βPix/COOL-1 may represent a target of vulnerability in GBM, in particular to improve Bev efficacy.
Collapse
Affiliation(s)
- Kate Connor
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland; (K.C.); (D.W.M.); (M.A.J.); (K.W.); (K.J.S.); (P.J.O.); (B.M.); (L.P.S.)
| | - David W. Murray
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland; (K.C.); (D.W.M.); (M.A.J.); (K.W.); (K.J.S.); (P.J.O.); (B.M.); (L.P.S.)
| | - Monika A. Jarzabek
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland; (K.C.); (D.W.M.); (M.A.J.); (K.W.); (K.J.S.); (P.J.O.); (B.M.); (L.P.S.)
| | - Nhan L. Tran
- Department of Cancer Biology and Neurological Surgery, Mayo Clinic Arizona, Scottsdale, AZ 85054, USA;
| | - Kieron White
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland; (K.C.); (D.W.M.); (M.A.J.); (K.W.); (K.J.S.); (P.J.O.); (B.M.); (L.P.S.)
| | - Patrick Dicker
- Epidemiology & Public Health, Royal College of Surgeons in Ireland, Dublin 2, Ireland;
| | - Kieron J. Sweeney
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland; (K.C.); (D.W.M.); (M.A.J.); (K.W.); (K.J.S.); (P.J.O.); (B.M.); (L.P.S.)
- National Neurosurgical Department, Beaumont Hospital, Dublin 9, Ireland
| | - Philip J. O’Halloran
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland; (K.C.); (D.W.M.); (M.A.J.); (K.W.); (K.J.S.); (P.J.O.); (B.M.); (L.P.S.)
- National Neurosurgical Department, Beaumont Hospital, Dublin 9, Ireland
| | - Brian MacCarthy
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland; (K.C.); (D.W.M.); (M.A.J.); (K.W.); (K.J.S.); (P.J.O.); (B.M.); (L.P.S.)
| | - Liam P. Shiels
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland; (K.C.); (D.W.M.); (M.A.J.); (K.W.); (K.J.S.); (P.J.O.); (B.M.); (L.P.S.)
| | - Francesca Lodi
- Center for Cancer Biology, Laboratory for Translational Genetics, Vlaams Instituut voor Biotechnologie (VIB), B-3000 Leuven, Belgium; (F.L.); (D.L.)
| | - Diether Lambrechts
- Center for Cancer Biology, Laboratory for Translational Genetics, Vlaams Instituut voor Biotechnologie (VIB), B-3000 Leuven, Belgium; (F.L.); (D.L.)
| | - Jann N. Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN 55905, USA;
| | - Raymond M. Schiffelers
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, 100 3584 Utrecht, The Netherlands;
| | - Marc Symons
- Department of Oncology & Cell Biology, Feinstein Institute for Medical Research at North Shore-LIJ, Manhasset, NY 11030, USA;
| | - Annette T. Byrne
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland; (K.C.); (D.W.M.); (M.A.J.); (K.W.); (K.J.S.); (P.J.O.); (B.M.); (L.P.S.)
- Correspondence: ; Tel.: +353-1-402-8673
| |
Collapse
|
11
|
Yao D, Li C, Rajoka MSR, He Z, Huang J, Wang J, Zhang J. P21-Activated Kinase 1: Emerging biological functions and potential therapeutic targets in Cancer. Am J Cancer Res 2020; 10:9741-9766. [PMID: 32863957 PMCID: PMC7449905 DOI: 10.7150/thno.46913] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 07/23/2020] [Indexed: 02/06/2023] Open
Abstract
The p21-Activated kinase 1 (PAK1), a member of serine-threonine kinases family, was initially identified as an interactor of the Rho GTPases RAC1 and CDC42, which affect a wide range of processes associated with cell motility, survival, metabolism, cell cycle, proliferation, transformation, stress, inflammation, and gene expression. Recently, the PAK1 has emerged as a potential therapeutic target in cancer due to its role in many oncogenic signaling pathways. Many PAK1 inhibitors have been developed as potential preclinical agents for cancer therapy. Here, we provide an overview of essential roles that PAK1 plays in cancer, including its structure and autoactivation mechanism, its crucial function from onset to progression to metastasis, metabolism, immune escape and even drug resistance in cancer; endogenous regulators; and cancer-related pathways. We also summarize the reported PAK1 small-molecule inhibitors based on their structure types and their potential application in cancer. In addition, we provide overviews on current progress and future challenges of PAK1 in cancer, hoping to provide new ideas for the diagnosis and treatment of cancer.
Collapse
|