1
|
Guo C, Tang H, Ren M, Zhang Y. BHLHE40-mediated RGS16 upregulation: a driver propelling gastric cancer progression via ferroptosis suppression. Hereditas 2025; 162:87. [PMID: 40413527 PMCID: PMC12102885 DOI: 10.1186/s41065-025-00447-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Accepted: 05/05/2025] [Indexed: 05/27/2025] Open
Abstract
BACKGROUND Gastric cancer (GC), a malignant neoplasm that arises from the epithelium of the gastric mucosa, endangers patients' lives and health severely. Regulator of G-protein signaling 16 (RGS16) has been found to be correlated with the malignant progression of various cancers, and BHLHE40 is highly expressed in GC. However, it remains unclear whether there is a regulatory mechanism between the them. METHODS The bioinformatics tools were applied to assess the differentially expressed genes in GC. Next, the expression levels of mRNA and protein were evaluated by qRT-PCR and Western blot. Cellular behaviors were assessed using CCK-8, EdU, Transwell, and flow cytometry assays. Meanwhile, the ferroptosis-related indicators were measured. Subsequently, the xenograft models were set up to estimate the role of RGS16 in vivo. Besides, the interaction between BHLHE40 and RGS16 was determined using ChIP assay and dual-luciferase reporter assay. RESULTS RGS16 exhibited an upregulated pattern in GC. In addition, silencing RGS16 impeded the proliferation, migration and invasion of GC cells while reinforcing apoptosis and ferroptosis. Moreover, RGS16 boosted the growth of tumors in vivo. Furthermore, BHLHE40 could bind to RGS16 and positively regulate its expression. Overexpression of RGS16 reversed the effects of silencing BHLHE40 on GC cells. CONCLUSION BHLHE40 curbed ferroptosis and oxidative stress of GC cells by modulating the expression of RGS16, thereby facilitating the malignant progression of GC.
Collapse
Affiliation(s)
- Caiyun Guo
- Department of Gastroenterology, Xi'an International Medical Center Hospital, Xi'an, 710117, Shaanxi, China
| | - Hua Tang
- Department of Gastroenterology, Xi'an International Medical Center Hospital, Xi'an, 710117, Shaanxi, China.
| | - Maifang Ren
- Department of Gastroenterology, Xi'an International Medical Center Hospital, Xi'an, 710117, Shaanxi, China
| | - Yongli Zhang
- Department of Gastroenterology, Tongchuan People's Hospital, Tongchuan, 727100, Shaanxi, China.
| |
Collapse
|
2
|
Tang Y, Fahira A, Lin S, Shao Y, Huang Z. Shared and specific competing endogenous RNAs network mining in four digestive system tumors. Comput Struct Biotechnol J 2024; 23:4271-4287. [PMID: 39669749 PMCID: PMC11635987 DOI: 10.1016/j.csbj.2024.11.005] [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: 08/02/2024] [Revised: 10/17/2024] [Accepted: 11/02/2024] [Indexed: 12/14/2024] Open
Abstract
Background Digestive system malignancies, including esophageal carcinoma (ESCA), stomach adenocarcinoma (STAD), liver hepatocellular carcinoma (LIHC), and colon adenocarcinoma (COAD), pose significant global health challenges. Identifying shared and distinct regulatory mechanisms across these cancers can lead to improved therapies. This study aims to construct and compare competing endogenous RNA (ceRNA) networks across ESCA, STAD, LIHC, and COAD to identify RNA biomarkers that could serve as precision therapeutic targets to enhance clinical outcomes and advance personalized cancer care. Methods Clinical and transcriptomic data from The Cancer Genome Atlas (TCGA) were analyzed to predict differentially expressed RNAs using the edgeR package. The ceRNA networks were constructed using the miRcode and ENCORI databases. Functional enrichment analysis and prognostic RNA screening were performed with ConsensusPathDB and univariate Cox regression analysis. Results we identified 6, 88, 55, and 41 RNA biomarkers in ESCA, STAD, LIHC, and COAD, respectively. Network analysis revealed shared and specific elements, with shared nodes enriched in cell cycle and mitotic processes. Several biomarkers, including HMGB3 and RGS16 (ESCA), COL4A1 and COL6A3 (STAD), CDCA5 and CDCA8 (LIHC), and LIMK1 and OSBPL3 (COAD), were consistent with prior studies, while novel biomarkers, such as C3P1 (ESCA), P2RY6 (STAD), and N4BP2L1 and PPP1R3B (LIHC), were discovered. Based on RNA correlation analysis, 1, 23, and 2 potential ceRNA regulatory axes were identified in STAD (PVT1/miR-490-3p/HMGA2), LIHC (DLX6-AS1/miR-139-5p/TOP2A, etc.), and COAD (STRCP1 & LINC00488/miR-142-3p/GAB1), respectively. Conclusions This study advances the understanding of ceRNA networks in digestive cancers, highlighting RNA biomarkers with potential as therapeutic targets for personalized treatment strategies.
Collapse
Affiliation(s)
- Yulai Tang
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523710, China
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
- Dongguan Key Laboratory of Sepsis Translational Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523710
| | - Aamir Fahira
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Siying Lin
- Dongguan Key Laboratory of Sepsis Translational Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523710
| | - Yiming Shao
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523710, China
| | - Zunnan Huang
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523710, China
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| |
Collapse
|
3
|
Shen H, Yuan J, Tong D, Chen B, Yu E, Chen G, Peng C, Chang W, E J, Cao F. Regulator of G protein signaling 16 restrains apoptosis in colorectal cancer through disrupting TRAF6-TAB2-TAK1-JNK/p38 MAPK signaling. Cell Death Dis 2024; 15:438. [PMID: 38906869 PMCID: PMC11192724 DOI: 10.1038/s41419-024-06803-6] [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: 02/17/2024] [Revised: 05/28/2024] [Accepted: 05/31/2024] [Indexed: 06/23/2024]
Abstract
Colorectal cancer (CRC) remains a major global cause of cancer-related mortality, lacking effective biomarkers and therapeutic targets. Revealing the critical pathogenic factors of CRC and the underlying mechanisms would offer potential therapeutic strategies for clinical application. G protein signaling (RGS) protein family modulators play essential role within regulating downstream signaling of GPCR receptors, with function in cancers unclear. Our study focused on the expression patterns of RGS proteins in CRC, identifying Regulator of G protein signaling 16 (RGS16) as a prospective diagnostic and therapeutic target. Analyzing 899 CRC tissues revealed elevated RGS16 levels, correlating with clinicopathological features and CRC prognosis by immunohistochemistry (IHC) combined with microarray. We confirmed the elevated RGS16 protein level in CRC, and found that patients with RGS16-high tumors exhibited decreased disease-specific survival (DSS) and disease-free survival (DFS) compared to those with low RGS16 expression. Functional assays demonstrated that RGS16 promoted the CRC progression, knockdown of RGS16 led to significantly increased apoptosis rates of CRC in vitro and in vivo. Notably, we also confirmed these phenotypes of RGS16 in organoids originated from resected primary human CRC tissues. Mechanistically, RGS16 restrained JNK/P38-mediated apoptosis in CRC cells through disrupting the recruitment of TAB2/TAK1 to TRAF6. This study provides insights into addressing the challenges posed by CRC, offering avenues for clinical translation.
Collapse
Affiliation(s)
- Hao Shen
- Department of Colorectal Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
- Department of Environmental and Occupational Health, Naval Medical University, Shanghai, China
| | - Jie Yuan
- Department of Colorectal Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
- Department of Health Management, Beidaihe Rest and Recuperation Center of PLA Joint Logistics Support Force, Qinhuangdao, China
| | - Dafeng Tong
- Department of Colorectal Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Bingchen Chen
- Department of Colorectal Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Enda Yu
- Department of Colorectal Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Guanglei Chen
- Department of Health Management, Beidaihe Rest and Recuperation Center of PLA Joint Logistics Support Force, Qinhuangdao, China
| | - Cheng Peng
- Department of Health Management, Beidaihe Rest and Recuperation Center of PLA Joint Logistics Support Force, Qinhuangdao, China
| | - Wenjun Chang
- Department of Environmental and Occupational Health, Naval Medical University, Shanghai, China.
| | - Jifu E
- Department of Colorectal Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China.
| | - Fuao Cao
- Department of Colorectal Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China.
| |
Collapse
|
4
|
Swierczynski M, Kasprzak Z, Makaro A, Salaga M. Regulators of G-Protein Signaling (RGS) in Sporadic and Colitis-Associated Colorectal Cancer. Int J Mol Sci 2024; 25:577. [PMID: 38203748 PMCID: PMC10778579 DOI: 10.3390/ijms25010577] [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: 11/13/2023] [Revised: 12/21/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
Colorectal cancer (CRC) is one of the most common neoplasms worldwide. Among the risk factors of CRC, inflammatory bowel disease (IBD) is one of the most important ones leading to the development of colitis-associated CRC (CAC). G-protein coupled receptors (GPCR) are transmembrane receptors that orchestrate a multitude of signaling cascades in response to external stimuli. Because of their functionality, they are promising targets in research on new strategies for CRC diagnostics and treatment. Recently, regulators of G-proteins (RGS) have been attracting attention in the field of oncology. Typically, they serve as negative regulators of GPCR responses to both physiological stimuli and medications. RGS activity can lead to both beneficial and harmful effects depending on the nature of the stimulus. However, the atypical RGS-AXIN uses its RGS domain to antagonize key signaling pathways in CRC development through the stabilization of the β-catenin destruction complex. Since AXIN does not limit the efficiency of medications, it seems to be an even more promising pharmacological target in CRC treatment. In this review, we discuss the current state of knowledge on RGS significance in sporadic CRC and CAC with particular emphasis on the regulation of GPCR involved in IBD-related inflammation comprising opioid, cannabinoid and serotonin receptors.
Collapse
Affiliation(s)
| | | | | | - Maciej Salaga
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (M.S.); (Z.K.); (A.M.)
| |
Collapse
|
5
|
Yang C, Zhang X, Yang X, Lian F, Sun Z, Huang Y, Shen W. Function and regulation of RGS family members in solid tumours: a comprehensive review. Cell Commun Signal 2023; 21:316. [PMID: 37924113 PMCID: PMC10623796 DOI: 10.1186/s12964-023-01334-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 09/25/2023] [Indexed: 11/06/2023] Open
Abstract
G protein-coupled receptors (GPCRs) play a key role in regulating the homeostasis of the internal environment and are closely associated with tumour progression as major mediators of cellular signalling. As a diverse and multifunctional group of proteins, the G protein signalling regulator (RGS) family was proven to be involved in the cellular transduction of GPCRs. Growing evidence has revealed dysregulation of RGS proteins as a common phenomenon and highlighted the key roles of these proteins in human cancers. Furthermore, their differential expression may be a potential biomarker for tumour diagnosis, treatment and prognosis. Most importantly, there are few systematic reviews on the functional/mechanistic characteristics and clinical application of RGS family members at present. In this review, we focus on the G-protein signalling regulator (RGS) family, which includes more than 20 family members. We analysed the classification, basic structure, and major functions of the RGS family members. Moreover, we summarize the expression changes of each RGS family member in various human cancers and their important roles in regulating cancer cell proliferation, stem cell maintenance, tumorigenesis and cancer metastasis. On this basis, we outline the molecular signalling pathways in which some RGS family members are involved in tumour progression. Finally, their potential application in the precise diagnosis, prognosis and treatment of different types of cancers and the main possible problems for clinical application at present are discussed. Our review provides a comprehensive understanding of the role and potential mechanisms of RGS in regulating tumour progression. Video Abstract.
Collapse
Affiliation(s)
- Chenglong Yang
- Key Laboratory of Precision Oncology in Universities of Shandong, Institute of Precision Medicine, Jining Medical University, Jining, 272067, China
| | - Xiaoyuan Zhang
- Key Laboratory of Precision Oncology in Universities of Shandong, Institute of Precision Medicine, Jining Medical University, Jining, 272067, China
| | - Xiaowen Yang
- Key Laboratory of Precision Oncology in Universities of Shandong, Institute of Precision Medicine, Jining Medical University, Jining, 272067, China
| | - Fuming Lian
- Key Laboratory of Precision Oncology in Universities of Shandong, Institute of Precision Medicine, Jining Medical University, Jining, 272067, China
| | - Zongrun Sun
- Key Laboratory of Precision Oncology in Universities of Shandong, Institute of Precision Medicine, Jining Medical University, Jining, 272067, China
| | - Yongming Huang
- Department of General Surgery, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272067, China.
| | - Wenzhi Shen
- Key Laboratory of Precision Oncology in Universities of Shandong, Institute of Precision Medicine, Jining Medical University, Jining, 272067, China.
| |
Collapse
|
6
|
Zhang Y, Zhu Q, Cao X, Ni B. RGS16 regulates Hippo-YAP activity to promote esophageal cancer cell proliferation and migration. Biochem Biophys Res Commun 2023; 675:122-129. [PMID: 37473526 DOI: 10.1016/j.bbrc.2023.04.033] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 04/07/2023] [Accepted: 04/14/2023] [Indexed: 07/22/2023]
Abstract
Esophageal Squamous Cell Carcinoma (ESCC) is a common malignant tumor of digestive tract, accounting for 90% of all pathological types of esophageal cancer. Despite the rapid development of multi-disciplinary treatment such as surgery, chemotherapy, radiotherapy and chemoradiotherapy, the prognosis of patients with ESCC is still poor. Regulators of G-protein signaling (RGSs) are involved in the processes of various cancers. The expression levels of its family member RGS16 are abnormally elevated in a variety of tumors, but its role in ESCC is still unclear. We found that RGS16 expression is aberrantly increased in ESCC tissues and correlated with poor prognosis of ESCC patients from The Cancer Genome Atlas (TCGA) database and our collected ESCC tissues. Moreover, knockdown of RGS16 in two ESCC cells could indeed inhibit their proliferation and migration. We further explored the molecular mechanism of RGS16 in ESCC, and the correlation analysis from TCGA database showed that the mRNA levels of RGS16 was positively correlated with that of CTGF and CYR61, two target genes of Hippo-YAP signaling. Consistently, RGS16- knockdown significantly inhibited the expression of CTGF and CYR61 in ESCC cells. We found that the phosphorylation levels of LATS1 and YAP were significantly increased and YAP translocated into the cytoplasm after depletion of RGS16 in ESCC cells. Also, RGS16-knockdown promoted the interaction between LATS1 and upstream kinase MST1. In addition, reintroduction of a constitutive active YAP5A mutant significantly rescued CTGF expression and cell proliferation in RGS16-knockdown cells. Together, our work revealed that RGS16 promoted YAP activity through disrupting the interaction between LATS1 and MST1, thus promoting the proliferation and migration of ESCC cells.
Collapse
Affiliation(s)
- Yanzhou Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Qing Zhu
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Xiufeng Cao
- Department of Cardiothoracic Surgery, Nanjing Yimin Hospital, Nanjing, 211103, Jiangsu, China
| | - Bin Ni
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
7
|
Li L, Xu Q, Tang C. RGS proteins and their roles in cancer: friend or foe? Cancer Cell Int 2023; 23:81. [PMID: 37118788 PMCID: PMC10148553 DOI: 10.1186/s12935-023-02932-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 04/21/2023] [Indexed: 04/30/2023] Open
Abstract
As negative modulators of G-protein-coupled receptors (GPCRs) signaling, regulators of G protein signaling (RGS) proteins facilitate various downstream cellular signalings through regulating kinds of heterotrimeric G proteins by stimulating the guanosine triphosphatase (GTPase) activity of G-protein α (Gα) subunits. The expression of RGS proteins is dynamically and precisely mediated by several different mechanisms including epigenetic regulation, transcriptional regulation -and post-translational regulation. Emerging evidence has shown that RGS proteins act as important mediators in controlling essential cellular processes including cell proliferation, survival -and death via regulating downstream cellular signaling activities, indicating that RGS proteins are fundamentally involved in sustaining normal physiological functions and dysregulation of RGS proteins (such as aberrant expression of RGS proteins) is closely associated with pathologies of many diseases such as cancer. In this review, we summarize the molecular mechanisms governing the expression of RGS proteins, and further discuss the relationship of RGS proteins and cancer.
Collapse
Affiliation(s)
- Lin Li
- National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, No. 3333, Binsheng Rd., Hangzhou, 310052, People's Republic of China
- Department of Urology, Third Affiliated Hospital of the Second Military Medical University, Shanghai, 201805, China
| | - Qiang Xu
- National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, No. 3333, Binsheng Rd., Hangzhou, 310052, People's Republic of China
| | - Chao Tang
- National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, No. 3333, Binsheng Rd., Hangzhou, 310052, People's Republic of China.
| |
Collapse
|
8
|
High-Throughput Sequencing Reveals That Rotundine Inhibits Colorectal Cancer by Regulating Prognosis-Related Genes. J Pers Med 2023; 13:jpm13030550. [PMID: 36983731 PMCID: PMC10052610 DOI: 10.3390/jpm13030550] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/13/2023] [Accepted: 03/14/2023] [Indexed: 03/22/2023] Open
Abstract
Background: Rotundine is an herbal medicine with anti-cancer effects. However, little is known about the anti-cancer effect of rotundine on colorectal cancer. Therefore, our study aimed to investigate the specific molecular mechanism of rotundine inhibition of colorectal cancer. Methods: MTT and cell scratch assay were performed to investigate the effects of rotundine on the viability, migration, and invasion ability of SW480 cells. Changes in cell apoptosis were analyzed by flow cytometry. DEGs were detected by high-throughput sequencing after the action of rotundine on SW480 cells, and the DEGs were subjected to function enrichment analysis. Bioinformatics analyses were performed to screen out prognosis-related DEGs of COAD. Followed by enrichment analysis of prognosis-related DEGs. Furthermore, prognostic models were constructed, including ROC analysis, risk curve analysis, PCA and t-SNE, Nomo analysis, and Kaplan–Meier prognostic analysis. Results: In this study, we showed that rotundine concentrations of 50 μM, 100 μM, 150 μM, and 200 μM inhibited the proliferation, migration, and invasion of SW480 cells in a time- and concentration-dependent manner. Rotundine does not induce SW480 cell apoptosis. Compared to the control group, high-throughput results showed that there were 385 DEGs in the SW480 group. And DEGs were associated with the Hippo signaling pathway. In addition, 16 of the DEGs were significantly associated with poorer prognosis in COAD, with MEF2B, CCDC187, PSD2, RGS16, PLXDC1, HELB, ASIC3, PLCH2, IGF2BP3, CLHC1, DNHD1, SACS, H1-4, ANKRD36, and ZNF117 being highly expressed in COAD and ARV1 being lowly expressed. Prognosis-related DEGs were mainly enriched in cancer-related pathways and biological functions, such as inositol phosphate metabolism, enterobactin transmembrane transporter activity, and enterobactin transport. Prognostic modeling also showed that these 16 DEGs could be used as predictors of overall survival prognosis in COAD patients. Conclusions: Rotundine inhibits the development and progression of colorectal cancer by regulating the expression of these prognosis-related genes. Our findings could further provide new directions for the treatment of colorectal cancer.
Collapse
|
9
|
Microdissecting the Hypoxia Landscape in Colon Cancer Reveals Three Distinct Subtypes and Their Potential Mechanism to Facilitate the Development of Cancer. JOURNAL OF ONCOLOGY 2023; 2023:9346621. [PMID: 36925652 PMCID: PMC10014161 DOI: 10.1155/2023/9346621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/09/2022] [Accepted: 11/25/2022] [Indexed: 03/09/2023]
Abstract
Background Hypoxia contributes to tumor progression and confers drug resistance. We attempted to microdissect the hypoxia landscape in colon cancer (CC) and explore its correlation with immunotherapy response. Materials and Methods The hypoxia landscape in CC patients was microdissected through unsupervised clustering. The "xCell" algorithms were applied to decipher the tumor immune infiltration characteristics. A hypoxia-related index signature was developed via the LASSO (least absolute shrinkage and selection operator) Cox regression in The Cancer Genome Atlas (TCGA)-colon adenocarcinoma (COAD) cohort and validated in an independent dataset from the Gene Expression Omnibus (GEO) database. The tumor immune dysfunction and exclusion (TIDE) algorithm was utilized to evaluate the correlation between the hypoxia-related index (HRI) signature and immunotherapy response. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) and western blotting were performed to verify the mRNA expression levels of five key genes. The Cell Counting Kit-8 (CCK-8) assay and flow cytometry were performed to examine the cell viability and cell apoptosis. Results Patients were classified into hypoxia-high, hypoxia-median, and hypoxia-low clusters in TCGA-COAD and verified in the GSE 17538 dataset. Compared with the hypoxia-low cluster, the hypoxia-high cluster consistently presented an unfavorable prognosis, higher immune scores, and stromal scores and elevated infiltration levels of several critical immune and stromal cells. Otherwise, we also found 600 hypoxia-related differentially expressed genes (HRDEGs) between the hypoxia-high cluster and the hypoxia-low cluster. Based on the 600 HRDEGs, we constructed the HRI signature which consists of 11 genes and shows a good prognostic value in both TCGA-COAD and GSE 17538 (AUC of 6-year survival prediction >0.75). Patients with low HRI scores were consistently predicted to be more responsive to immunotherapy. Of the 11 HRI signature genes, RGS16, SNAI1, CDR2L, FRMD5, and FSTL3 were differently expressed between tumors and adjacent tissues. Low expression of SNAI1, CDR2L, FRMD5, and FSTL3 could induce cell viability and promote tumor cell apoptosis. Conclusion In our study, we discovered three hypoxia clusters which correlate with the clinical outcome and the tumor immune microenvironment in CC. Based on the hypoxia cluster and HRDEGs, we constructed a reliable HRI signature that could accurately predict the prognosis and immunotherapeutic responsiveness in CC patients and discovered four key genes that could affect tumor cell viability and apoptosis.
Collapse
|
10
|
RGS16 regulated by let-7c-5p promotes glioma progression by activating PI3K-AKT pathway. Front Med 2022; 17:143-155. [PMID: 36414916 DOI: 10.1007/s11684-022-0929-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 04/21/2022] [Indexed: 11/24/2022]
Abstract
Gliomas are the most common central nervous system tumours; they are highly aggressive and have a poor prognosis. RGS16 belongs to the regulator of G-protein signalling (RGS) protein family, which plays an important role in promoting various cancers, such as breast cancer, pancreatic cancer, and colorectal cancer. Moreover, previous studies confirmed that let-7c-5p, a well-known microRNA, can act as a tumour suppressor to regulate the progression of various tumours by inhibiting the expression of its target genes. However, whether RGS16 can promote the progression of glioma and whether it is regulated by miR let-7c-5p are still unknown. Here, we confirmed that RGS16 is upregulated in glioma tissues and that high expression of RGS16 is associated with poor survival. Ectopic deletion of RGS16 significantly suppressed glioma cell proliferation and migration both in vitro and in vivo. Moreover, RGS16 was validated as a direct target gene of miR let-7c-5p. The overexpression of miR let-7c-5p obviously downregulated the expression of RGS16, and knocking down miR let-7c-5p had the opposite effect. Thus, we suggest that the suppression of RGS16 by miR let-7c-5p can promote glioma progression and may serve as a potential prognostic biomarker and therapeutic target in glioma.
Collapse
|
11
|
Abstract
BACKGROUND Ulcerative colitis is a chronic and progressive inflammatory disorder. The regulator of the G-protein signaling (RGS) is involved in the pathogenesis of several immune system disorders. RGS16, a member of the RGS protein superfamily, has been shown to play critical roles in several immune system-related diseases. However, the roles of RGS16 in ulcerative colitis remain to be elucidated. METHODS We analyzed the expression of RGS16 in peripheral blood mononuclear cells (PBMCs) and inflamed mucosa of ulcerative colitis patients using quantitative reverse transcription-PCR, western blotting and immunohistochemistry. We performed Spearman's correlation to analyze the correlation between RGS16 expression and the ulcerative colitis endoscopic index of severity (UCEIS), Mayo index, erythrocyte sedimentation rate (ESR) and serum tumor necrosis factor alpha (TNF-a) and IL-17A levels. Further, PBMCs were stimulated with inflammatory cytokines in vitro . RESULTS RGS16 expression significantly increased in the colonic mucosa and PBMCs from patients with ulcerative colitis and significantly correlated with the Mayo index, UCEIS, ESR and serum TNF-α and IL-17A levels. TNF-α upregulated RGS16 expression in PBMCs in a dose- and time-dependent manner via the nuclear factor kappa beta (NF-kB) signaling pathway. Moreover, anti-TNF treatment with infliximab significantly decreased RGS16 expression in PBMCs and intestinal mucosa of patients with ulcerative colitis. CONCLUSION Our study revealed a novel mechanism by which RGS16 expression in ulcerative colitis is positively correlated with disease activity. Thus, RGS16 might serve as a potential therapeutic marker for the treatment of ulcerative colitis.
Collapse
|
12
|
Tian M, Ma Y, Li T, Wu N, Li J, Jia H, Yan M, Wang W, Bian H, Tan X, Qi J. Functions of regulators of G protein signaling 16 in immunity, inflammation, and other diseases. Front Mol Biosci 2022; 9:962321. [PMID: 36120550 PMCID: PMC9478547 DOI: 10.3389/fmolb.2022.962321] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Regulators of G protein signaling (RGS) act as guanosine triphosphatase activating proteins to accelerate guanosine triphosphate hydrolysis of the G protein α subunit, leading to the termination of the G protein-coupled receptor (GPCR) downstream signaling pathway. RGS16, which is expressed in a number of cells and tissues, belongs to one of the small B/R4 subfamilies of RGS proteins and consists of a conserved RGS structural domain with short, disordered amino- and carboxy-terminal extensions and an α-helix that classically binds and de-activates heterotrimeric G proteins. However, with the deepening of research, it has been revealed that RGS16 protein not only regulates the classical GPCR pathway, but also affects immune, inflammatory, tumor and metabolic processes through other signaling pathways including the mitogen-activated protein kinase, phosphoinositide 3-kinase/protein kinase B, Ras homolog family member A and stromal cell-derived factor 1/C-X-C motif chemokine receptor 4 pathways. Additionally, the RGS16 protein may be involved in the Hepatitis B Virus -induced inflammatory response. Therefore, given the continuous expansion of knowledge regarding its role and mechanism, the structure, characteristics, regulatory mechanisms and known functions of the small RGS proteinRGS16 are reviewed in this paper to prepare for diagnosis, treatment, and prognostic evaluation of different diseases such as inflammation, tumor, and metabolic disorders and to better study its function in other diseases.
Collapse
Affiliation(s)
- Miaomiao Tian
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yan Ma
- Zibo Central Hospital, Zibo, China
| | - Tao Li
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Nijin Wu
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jiaqi Li
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Huimin Jia
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Meizhu Yan
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Wenwen Wang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Hongjun Bian
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xu Tan
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- *Correspondence: Jianni Qi, ; Xu Tan,
| | - Jianni Qi
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong Provincial Engineering and Technological Research Center for Liver Diseases Prevention and Control, Jinan, China
- *Correspondence: Jianni Qi, ; Xu Tan,
| |
Collapse
|
13
|
Hu J, He Y, Liao K, Yang Q, Xu Y, Cao G, Wang X. Identification of inflammatory factor-related genes associated with the prognostic and immune cell infiltration in colorectal cancer patients. Genes Dis 2022. [PMID: 37492736 PMCID: PMC10363590 DOI: 10.1016/j.gendis.2022.07.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
This study aims to identify the inflammatory factor-related genes which help to predict the prognosis of patients with colorectal cancer. GSEA (Gene Set Enrichment Analysis) was used to acquire inflammation-related genes and the corresponding expression information was collected from TCGA database to determine the DEGs (differentially-expressed genes) in CRC patients. We conducted enrichment analysis and PPI (protein-protein interaction) of these DEGs. Besides, key genes that are both differentially-expressed and prognosis-related were screened out, which were used to establish the prognostic model. We obtained 79 DEGs and 19 prognostic genes, 10 prognostic-related differential genes were eventually screened. These genes were used to construct the prognostic model. We also identified that the immune infiltration score of macrophages between different risk groups was significantly different and similar distinction was witnessed in immune function score of APC (antigen-presenting cell) co-stimulation and type I IFN (interferon) response.
Collapse
|
14
|
Huang R, Li G, Zhao Z, Zeng F, Zhang K, Liu Y, Wang K, Hu H. RGS16 promotes glioma progression and serves as a prognostic factor. CNS Neurosci Ther 2020; 26:791-803. [PMID: 32319728 PMCID: PMC7366748 DOI: 10.1111/cns.13382] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/27/2020] [Accepted: 03/31/2020] [Indexed: 12/13/2022] Open
Abstract
Background RGS protein family members have recently became new potentially promising therapeutic targets in many cancers. However, as a key member of RGS family, RGS16 has seldom been studied in glioma. The present study was designed to investigate the prognostic value and biological function of RGS16 based on large‐scale databases and functional assays in vitro. Methods Here, we performed comprehensive analysis for the expression characteristic of RGS16 in Chinese Glioma Genome Atlas (CGGA) microarray database with 301 patients and validated in The Cancer Genome Atlas (TCGA) microarray and RNA sequencing database. Student's t‐test, one‐way ANOVA test and long‐rank test were used to assess differences between groups. Kaplan‐Meier survival, univariate and multivariate Cox analysis and ROC curve were used to estimate the survival distributions. Biological implication of abnormal expression of RGS16 in glioma was also explored. Functional analysis of RGS16 was performed in several glioblastoma (GBM) cell lines. R language and SPSS were used for statistical analysis and graphical work. Results We found that the expression of RGS16 was positively related to the grade of glioma. High level of RGS16 commonly gathered in glioma of mesenchymal subtype and wild‐type IDH1. Moreover, higher expression level of RGS16 was found to be significantly correlated with poor prognosis. The univariate and multivariate Cox regression analysis and ROC curve showed that RGS16 was an independent prognostic factor for glioma patients. Gene ontology analysis, gene set enrichment analysis, and gene set variation analysis suggested that the overexpression of RGS16 tightly related to cell proliferation, migration, epithelial‐mesenchymal transition (EMT), immune and inflammatory response of glioma. Knockdown of RGS16 in glioma cell lines also showed that RGS16 promoted the malignant progress of glioma cell lines. Conclusions RGS16 plays an important role in glioma progression and serves as an independent prognostic factor, especially in GBM patients.
Collapse
Affiliation(s)
- Ruoyu Huang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Chinese Glioma Cooperative Group (CGCG), Beijing, China
| | - Guanzhang Li
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Chinese Glioma Cooperative Group (CGCG), Beijing, China
| | - Zheng Zhao
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Chinese Glioma Cooperative Group (CGCG), Beijing, China
| | - Fan Zeng
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Chinese Glioma Cooperative Group (CGCG), Beijing, China
| | - Kenan Zhang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Chinese Glioma Cooperative Group (CGCG), Beijing, China
| | - Yanwei Liu
- Chinese Glioma Cooperative Group (CGCG), Beijing, China.,Department of Radiotherapy, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Kuanyu Wang
- Chinese Glioma Cooperative Group (CGCG), Beijing, China.,Department of Gamma Knife Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Huimin Hu
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Chinese Glioma Cooperative Group (CGCG), Beijing, China
| |
Collapse
|
15
|
Sun X, Chen Y, Yu H, Machan JT, Alladin A, Ramirez J, Taliano R, Hart J, Chen Q, Terek RM. Anti-miRNA Oligonucleotide Therapy for Chondrosarcoma. Mol Cancer Ther 2019; 18:2021-2029. [PMID: 31341031 PMCID: PMC6825546 DOI: 10.1158/1535-7163.mct-18-1020] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 05/10/2019] [Accepted: 07/19/2019] [Indexed: 02/03/2023]
Abstract
Chondrosarcoma is a highly aggressive primary malignant bone tumor mostly occurring in adults. There are no effective systemic treatments, and patients with this disease have poor survival. miR-181a is an oncomiR that is overexpressed in high-grade chondrosarcoma and promotes tumor progression. Regulator of G-protein signaling 16 (RGS16) is a target of miR-181a. Inhibition of RGS16 expression by miR-181a enhances CXC chemokine receptor 4 signaling, which in turn increases MMP1 and VEGF expression, angiogenesis, and metastasis. Here, we report the results of systemic treatment with anti-miRNA oligonucleotides (AMO) directed against miR-181a utilizing a nanopiece delivery platform (NPs). NPs were combined with a molecular beacon or anti-miR-181a oligonucleotides and are shown to transfect chondrosarcoma cells in vitro and in vivo Intratumoral injection and systemic delivery had similar effects on miR-181a expression in nude mice bearing chondrosarcoma xenografts. Systemic delivery of NPs carrying anti-miR-181a also restored RGS16 expression, decreased expression of VEGF and MMP1, MMP activity, and tumor volume by 32% at day 38, and prolonged survival from 23% to 45%. In conclusion, these data support that systemic delivery of AMO shows promise for chondrosarcoma treatment.
Collapse
Affiliation(s)
- Xiaojuan Sun
- Department of Orthopaedics, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island
| | - Yupeng Chen
- Department of Orthopaedics, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut
| | - Hongchuan Yu
- Department of Orthopaedics, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island
| | - Jason T Machan
- Department of Orthopaedics, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island
- Lifespan Biostatistics Core, Lifespan Hospital System, Providence, Rhode Island
- Department of Surgery, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island
| | - Ashna Alladin
- Department of Orthopaedics, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island
| | - Jose Ramirez
- Department of Orthopaedics, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island
| | - Ross Taliano
- Department of Pathology, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island
| | - Jesse Hart
- Department of Pathology, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island
| | - Qian Chen
- Department of Orthopaedics, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island
| | - Richard M Terek
- Department of Orthopaedics, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island.
- Providence Veterans Administration Medical Center, Providence, Rhode Island
| |
Collapse
|
16
|
Teras M, Viisileht E, Pahtma-Hall M, Rump A, Paalme V, Pata P, Pata I, Langevin C, Rüütel Boudinot S. Porcine circovirus type 2 ORF3 protein induces apoptosis in melanoma cells. BMC Cancer 2018; 18:1237. [PMID: 30526524 PMCID: PMC6288936 DOI: 10.1186/s12885-018-5090-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 11/14/2018] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND The current treatment of malignant melanoma is limited by the lack of effective therapeutic approaches, and alternative treatments are needed. Proliferative diseases such as melanoma and other cancers may be treatable by virally-encoded apoptotic proteins that are targeted to rapidly multiplying cells. Caspase-dependent apoptosis, that is frequently used in chemotherapy, can boost the cell proliferation that caspase-independent cell death does not. METHODS In the current study, the porcine circovirus type 2 (PCV2), proapoptotic protein ORF3 was expressed in mouse and human cancer cell lines, and its apoptotic activity was assessed. RESULTS Quantitative assessment of the apoptotic cells by flow cytometry showed that apoptotic cell death was significantly increased in ORF3-expressing malignant cells, compared to ORF3 non-expressing cells. Our data show that PCV2 ORF3 induces apoptosis in a caspase-3 and -8 independent manner. ORF3 expression seems to cause an increase in abnormal mitosis in B16F10 melanoma cells by interacting with centrosomes and thereby disrupting the formation of the mitotic spindle. In addition, we show that ORF3 of PCV2 also exhibits significant anti-tumor effects in vivo. Although the expression of Regulator of G protein Signaling (RGS)-16 by recipient mice inhibited the development of grafted melanoma in vivo, it was not required for the antitumoral activity of ORF3. CONCLUSION PCV2 ORF3 causes abnormal mitosis in rapidly dividing cells and increases the apoptosis of cancer cells. Apoptin might, therefore, be considered to develop future antitumoral strategies.
Collapse
Affiliation(s)
- Marina Teras
- Oncology, North Estonia Medical Centre, 19 J. Sütiste tee st, 13419 Tallinn, Estonia
| | - Edda Viisileht
- Oncology, North Estonia Medical Centre, 19 J. Sütiste tee st, 13419 Tallinn, Estonia
- Department of Chemistry and Biotechnology, Tallinn University of Technology, 15 Akadeemia St, 12618 Tallinn, Estonia
| | - Merlis Pahtma-Hall
- Department of Chemistry and Biotechnology, Tallinn University of Technology, 15 Akadeemia St, 12618 Tallinn, Estonia
| | - Airi Rump
- Department of Chemistry and Biotechnology, Tallinn University of Technology, 15 Akadeemia St, 12618 Tallinn, Estonia
| | - Viiu Paalme
- Department of Chemistry and Biotechnology, Tallinn University of Technology, 15 Akadeemia St, 12618 Tallinn, Estonia
| | - Pille Pata
- Department of Chemistry and Biotechnology, Tallinn University of Technology, 15 Akadeemia St, 12618 Tallinn, Estonia
| | - Illar Pata
- IVEX Lab OU, 15 Akadeemia St, 12618 Tallinn, Estonia
| | - Christelle Langevin
- Virologie et Immunologie Moléculaires, INRA, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Sirje Rüütel Boudinot
- Department of Chemistry and Biotechnology, Tallinn University of Technology, 15 Akadeemia St, 12618 Tallinn, Estonia
| |
Collapse
|
17
|
Squires KE, Montañez-Miranda C, Pandya RR, Torres MP, Hepler JR. Genetic Analysis of Rare Human Variants of Regulators of G Protein Signaling Proteins and Their Role in Human Physiology and Disease. Pharmacol Rev 2018; 70:446-474. [PMID: 29871944 PMCID: PMC5989036 DOI: 10.1124/pr.117.015354] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Regulators of G protein signaling (RGS) proteins modulate the physiologic actions of many neurotransmitters, hormones, and other signaling molecules. Human RGS proteins comprise a family of 20 canonical proteins that bind directly to G protein-coupled receptors/G protein complexes to limit the lifetime of their signaling events, which regulate all aspects of cell and organ physiology. Genetic variations account for diverse human traits and individual predispositions to disease. RGS proteins contribute to many complex polygenic human traits and pathologies such as hypertension, atherosclerosis, schizophrenia, depression, addiction, cancers, and many others. Recent analysis indicates that most human diseases are due to extremely rare genetic variants. In this study, we summarize physiologic roles for RGS proteins and links to human diseases/traits and report rare variants found within each human RGS protein exome sequence derived from global population studies. Each RGS sequence is analyzed using recently described bioinformatics and proteomic tools for measures of missense tolerance ratio paired with combined annotation-dependent depletion scores, and protein post-translational modification (PTM) alignment cluster analysis. We highlight selected variants within the well-studied RGS domain that likely disrupt RGS protein functions and provide comprehensive variant and PTM data for each RGS protein for future study. We propose that rare variants in functionally sensitive regions of RGS proteins confer profound change-of-function phenotypes that may contribute, in newly appreciated ways, to complex human diseases and/or traits. This information provides investigators with a valuable database to explore variation in RGS protein function, and for targeting RGS proteins as future therapeutic targets.
Collapse
Affiliation(s)
- Katherine E Squires
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (K.E.S., C.M.-M., J.R.H.); and School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia (R.R.P., M.P.T.)
| | - Carolina Montañez-Miranda
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (K.E.S., C.M.-M., J.R.H.); and School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia (R.R.P., M.P.T.)
| | - Rushika R Pandya
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (K.E.S., C.M.-M., J.R.H.); and School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia (R.R.P., M.P.T.)
| | - Matthew P Torres
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (K.E.S., C.M.-M., J.R.H.); and School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia (R.R.P., M.P.T.)
| | - John R Hepler
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (K.E.S., C.M.-M., J.R.H.); and School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia (R.R.P., M.P.T.)
| |
Collapse
|
18
|
Polidarová L, Houdek P, Sumová A. Chronic disruptions of circadian sleep regulation induce specific proinflammatory responses in the rat colon. Chronobiol Int 2017; 34:1273-1287. [DOI: 10.1080/07420528.2017.1361436] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Lenka Polidarová
- Department of Neurohumoral Regulations, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Pavel Houdek
- Department of Neurohumoral Regulations, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Alena Sumová
- Department of Neurohumoral Regulations, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
19
|
McCauley HA, Chevrier V, Birnbaum D, Guasch G. De-repression of the RAC activator ELMO1 in cancer stem cells drives progression of TGFβ-deficient squamous cell carcinoma from transition zones. eLife 2017; 6:e22914. [PMID: 28219480 PMCID: PMC5319840 DOI: 10.7554/elife.22914] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 01/27/2017] [Indexed: 01/18/2023] Open
Abstract
Squamous cell carcinomas occurring at transition zones are highly malignant tumors with poor prognosis. The identity of the cell population and the signaling pathways involved in the progression of transition zone squamous cell carcinoma are poorly understood, hence representing limited options for targeted therapies. Here, we identify a highly tumorigenic cancer stem cell population in a mouse model of transitional epithelial carcinoma and uncover a novel mechanism by which loss of TGFβ receptor II (Tgfbr2) mediates invasion and metastasis through de-repression of ELMO1, a RAC-activating guanine exchange factor, specifically in cancer stem cells of transition zone tumors. We identify ELMO1 as a novel target of TGFβ signaling and show that restoration of Tgfbr2 results in a complete block of ELMO1 in vivo. Knocking down Elmo1 impairs metastasis of carcinoma cells to the lung, thereby providing insights into the mechanisms of progression of Tgfbr2-deficient invasive transition zone squamous cell carcinoma.
Collapse
Affiliation(s)
- Heather A McCauley
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, United States
| | - Véronique Chevrier
- Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, F-13009, CNRS, UMR7258, F-13009, Institut Paoli-Calmettes, F-13009, Aix-Marseille University, UM 105, F-13284, Marseille, France
| | - Daniel Birnbaum
- Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, F-13009, CNRS, UMR7258, F-13009, Institut Paoli-Calmettes, F-13009, Aix-Marseille University, UM 105, F-13284, Marseille, France
| | - Géraldine Guasch
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, United States
- Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, F-13009, CNRS, UMR7258, F-13009, Institut Paoli-Calmettes, F-13009, Aix-Marseille University, UM 105, F-13284, Marseille, France
| |
Collapse
|
20
|
Salaga M, Storr M, Martemyanov KA, Fichna J. RGS proteins as targets in the treatment of intestinal inflammation and visceral pain: New insights and future perspectives. Bioessays 2016; 38:344-54. [PMID: 26817719 PMCID: PMC4916644 DOI: 10.1002/bies.201500118] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Regulators of G protein signaling (RGS) proteins provide timely termination of G protein-coupled receptor (GPCR) responses. Serving as a central control point in GPCR signaling cascades, RGS proteins are promising targets for drug development. In this review, we discuss the involvement of RGS proteins in the pathophysiology of the gastrointestinal inflammation and their potential to become a target for anti-inflammatory drugs. Specifically, we evaluate the emerging evidence for modulation of selected receptor families: opioid, cannabinoid and serotonin by RGS proteins. We discuss how the regulation of RGS protein level and activity may modulate immunological pathways involved in the development of intestinal inflammation. Finally, we propose that RGS proteins may serve as a prognostic factor for survival rate in colorectal cancer. The ideas introduced in this review set a novel conceptual framework for the utilization of RGS proteins in the treatment of gastrointestinal inflammation, a growing major concern worldwide.
Collapse
Affiliation(s)
- Maciej Salaga
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Poland
| | - Martin Storr
- Walter Brendel Center of Experimental Medicine, University of Munich, Germany
| | - Kirill A. Martemyanov
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
- Corresponding authors: J.F. Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland, Phone: ++48 42 272 57 07, Fax: ++48 42 272 56 94, . K.A.M., Department of Neuroscience, The Scripps Research Institute, 130 Scripps Way C347, Jupiter, FL 33458, USA, Phone: ++1 561 228 2770,
| | - Jakub Fichna
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Poland
- Corresponding authors: J.F. Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland, Phone: ++48 42 272 57 07, Fax: ++48 42 272 56 94, . K.A.M., Department of Neuroscience, The Scripps Research Institute, 130 Scripps Way C347, Jupiter, FL 33458, USA, Phone: ++1 561 228 2770,
| |
Collapse
|
21
|
Hoshi Y, Endo K, Shirakihara T, Fukagawa A, Miyazawa K, Saitoh M. The potential role of regulator of G-protein signaling 16 in cell motility mediated by δEF1 family proteins. FEBS Lett 2016; 590:270-8. [PMID: 26823172 PMCID: PMC4819697 DOI: 10.1002/1873-3468.12042] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 12/02/2015] [Accepted: 12/09/2015] [Indexed: 01/09/2023]
Abstract
The epithelial–mesenchymal transition (EMT) is associated with tumor progression. We reported previously that expression of the δEF1 family proteins (δEF1/ZEB1 and SIP1/ZEB2), key regulators of the EMT, is positively correlated with EMT phenotypes and aggressiveness of breast cancer. Here, we show that the expression levels of regulator of G‐protein signaling 16 (RGS16) are negatively correlated with those of the δEF1 family proteins. On the basis of the results of gain‐ and loss‐of‐function analyses, we suggest that δEF1 family proteins promote cell motility of breast cancer cells directly or indirectly through repressing expression of RGS16.
Collapse
Affiliation(s)
- Yuta Hoshi
- Research Training Program for Undergraduates, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan.,Department of Biochemistry, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Kaori Endo
- Department of Biochemistry, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Takuya Shirakihara
- Division of Metastasis & Invasion Signaling, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | - Akihiko Fukagawa
- Research Training Program for Undergraduates, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan.,Department of Biochemistry, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Keiji Miyazawa
- Department of Biochemistry, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Masao Saitoh
- Department of Biochemistry, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan.,Center for Medical Education and Sciences, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan
| |
Collapse
|
22
|
Woodard GE, Jardín I, Berna-Erro A, Salido GM, Rosado JA. Regulators of G-protein-signaling proteins: negative modulators of G-protein-coupled receptor signaling. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2015; 317:97-183. [PMID: 26008785 DOI: 10.1016/bs.ircmb.2015.02.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Regulators of G-protein-signaling (RGS) proteins are a category of intracellular proteins that have an inhibitory effect on the intracellular signaling produced by G-protein-coupled receptors (GPCRs). RGS along with RGS-like proteins switch on through direct contact G-alpha subunits providing a variety of intracellular functions through intracellular signaling. RGS proteins have a common RGS domain that binds to G alpha. RGS proteins accelerate GTPase and thus enhance guanosine triphosphate hydrolysis through the alpha subunit of heterotrimeric G proteins. As a result, they inactivate the G protein and quickly turn off GPCR signaling thus terminating the resulting downstream signals. Activity and subcellular localization of RGS proteins can be changed through covalent molecular changes to the enzyme, differential gene splicing, and processing of the protein. Other roles of RGS proteins have shown them to not be solely committed to being inhibitors but behave more as modulators and integrators of signaling. RGS proteins modulate the duration and kinetics of slow calcium oscillations and rapid phototransduction and ion signaling events. In other cases, RGS proteins integrate G proteins with signaling pathways linked to such diverse cellular responses as cell growth and differentiation, cell motility, and intracellular trafficking. Human and animal studies have revealed that RGS proteins play a vital role in physiology and can be ideal targets for diseases such as those related to addiction where receptor signaling seems continuously switched on.
Collapse
Affiliation(s)
- Geoffrey E Woodard
- Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA; Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Isaac Jardín
- Department of Physiology, University of Extremadura, Caceres, Spain
| | - A Berna-Erro
- Department of Physiology, University of Extremadura, Caceres, Spain
| | - Gines M Salido
- Department of Physiology, University of Extremadura, Caceres, Spain
| | - Juan A Rosado
- Department of Physiology, University of Extremadura, Caceres, Spain
| |
Collapse
|
23
|
Carper MB, Denvir J, Boskovic G, Primerano DA, Claudio PP. RGS16, a novel p53 and pRb cross-talk candidate inhibits migration and invasion of pancreatic cancer cells. Genes Cancer 2015; 5:420-35. [PMID: 25568667 PMCID: PMC4279439 DOI: 10.18632/genesandcancer.43] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 11/26/2014] [Indexed: 12/11/2022] Open
Abstract
Data collected since the discovery of p53 and pRb/RB1 suggests these tumor suppressors cooperate to inhibit tumor progression. Patients who have mutations in both p53 and RB1 genes have increased tumor reoccurrence and decreased survival compared to patients with only one tumor suppressor gene inactivated. It remains unclear how p53 and pRb cooperate toward inhibiting tumorigenesis. Using RNA expression profiling we identified 179 p53 and pRb cross-talk candidates in normal lung fibroblasts (WI38) cells exogenously coexpressing p53 and pRb. Regulator of G protein signaling 16 (RGS16) was among the p53 and pRb cross-talk candidates and has been implicated in inhibiting activation of several oncogenic pathways associated with proliferation, migration, and invasion of cancer cells. RGS16 has been found to be downregulated in pancreatic cancer patients with metastases compared to patients without metastasis. Expression of RGS16 mRNA was decreased in the pancreatic cancer cell lines tested compared to control. Expression of RGS16 inhibited migration of the BxPC-3 and AsPC-1 but not PANC-1 cells and inhibited invasion of BxPC-3 and AsPC-1 cells with no impact on cell viability. We have identified for the first time p53 and pRb cross-talk candidates and a role for RGS16 to inhibit pancreatic cancer migration and invasion.
Collapse
Affiliation(s)
- Miranda B Carper
- McKown Translational Genomic Research Institute, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA ; Department of Biochemistry and Microbiology, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA
| | - James Denvir
- Department of Biochemistry and Microbiology, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA
| | - Goran Boskovic
- Department of Biochemistry and Microbiology, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA
| | - Donald A Primerano
- Department of Biochemistry and Microbiology, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA
| | - Pier Paolo Claudio
- McKown Translational Genomic Research Institute, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA ; Department of Biochemistry and Microbiology, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA ; Department of Surgery, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA
| |
Collapse
|
24
|
A gene expression and pre-mRNA splicing signature that marks the adenoma-adenocarcinoma progression in colorectal cancer. PLoS One 2014; 9:e87761. [PMID: 24516561 PMCID: PMC3916340 DOI: 10.1371/journal.pone.0087761] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 12/30/2013] [Indexed: 12/22/2022] Open
Abstract
It is widely accepted that most colorectal cancers (CRCs) arise from colorectal adenomas (CRAs), but transcriptomic data characterizing the progression from colorectal normal mucosa to adenoma, and then to adenocarcinoma are scarce. These transition steps were investigated using microarrays, both at the level of gene expression and alternative pre-mRNA splicing. Many genes and exons were abnormally expressed in CRAs, even more than in CRCs, as compared to normal mucosae. Known biological pathways involved in CRC were altered in CRA, but several new enriched pathways were also recognized, such as the complement and coagulation cascades. We also identified four intersectional transcriptional signatures that could distinguish CRAs from normal mucosae or CRCs, including a signature of 40 genes differentially deregulated in both CRA and CRC samples. A majority of these genes had been described in different cancers, including FBLN1 or INHBA, but only a few in CRC. Several of these changes were also observed at the protein level. In addition, 20% of these genes (i.e. CFH, CRYAB, DPT, FBLN1, ITIH5, NR3C2, SLIT3 and TIMP1) showed altered pre-mRNA splicing in CRAs. As a global variation occurring since the CRA stage, and maintained in CRC, the expression and splicing changes of this 40-gene set may mark the risk of cancer occurrence from analysis of CRA biopsies.
Collapse
|
25
|
Okano M, Yamamoto H, Ohkuma H, Kano Y, Kim H, Nishikawa S, Konno M, Kawamoto K, Haraguchi N, Takemasa I, Mizushima T, Ikeda M, Yokobori T, Mimori K, Sekimoto M, Doki Y, Mori M, Ishii H. Significance of INHBA expression in human colorectal cancer. Oncol Rep 2013; 30:2903-8. [PMID: 24085226 DOI: 10.3892/or.2013.2761] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 08/06/2013] [Indexed: 11/05/2022] Open
Abstract
Inhibin β A (INHBA) is a member of the transforming growth factor β (TGF-β) superfamily. INHBA expression is associated with several types of human cancers; however, its significance in colorectal cancer (CRC) is not fully understood. INHBA expression was studied in 126 primary CRC samples and 4 CRC cell lines. Cell growth was assessed after inhibition of INHBA expression or after exogenous overexpression of INHBA in CRC tissues. INHBA expression was significantly higher in CRC tissues when compared to that in the corresponding normal tissues (P<0.001). Patients in the high expression group showed a poorer overall survival rate when compared to those in the low expression group (P<0.001); the present study did not evaluate for an independent prognostic factor but showed the significance of lymph node metastasis as an independent prognostic factor. The present study suggests that INHBA is useful as a predictive marker for prognosis in CRC patients.
Collapse
Affiliation(s)
- Miho Okano
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Sethakorn N, Dulin NO. RGS expression in cancer: oncomining the cancer microarray data. J Recept Signal Transduct Res 2013; 33:166-71. [DOI: 10.3109/10799893.2013.773450] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
27
|
Colorectal adenoma to carcinoma progression is accompanied by changes in gene expression associated with ageing, chromosomal instability, and fatty acid metabolism. Cell Oncol (Dordr) 2012; 35:53-63. [PMID: 22278361 PMCID: PMC3308003 DOI: 10.1007/s13402-011-0065-1] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/28/2011] [Indexed: 02/08/2023] Open
Abstract
Background Colorectal cancer develops in a multi-step manner from normal epithelium, through a pre-malignant lesion (so-called adenoma), into a malignant lesion (carcinoma), which invades surrounding tissues and eventually can spread systemically (metastasis). It is estimated that only about 5% of adenomas do progress to a carcinoma. Aim The present study aimed to unravel the biology of adenoma to carcinoma progression by mRNA expression profiling, and to identify candidate biomarkers for adenomas that are truly at high risk of progression. Methods Genome-wide mRNA expression profiles were obtained from a series of 37 colorectal adenomas and 31 colorectal carcinomas using oligonucleotide microarrays. Differentially expressed genes were validated in an independent colorectal gene expression data set. Gene Set Enrichment Analysis (GSEA) was used to identify altered expression of sets of genes associated with specific biological processes, in order to better understand the biology of colorectal adenoma to carcinoma progression. Results mRNA expression of 248 genes was significantly different, of which 96 were upregulated and 152 downregulated in carcinomas compared to adenomas. Classification of adenomas and carcinomas using the expression of these genes showed to be very accurate, also when tested in an independent expression data set. Gene-sets associated with ageing (which is related to senescence) and chromosomal instability were upregulated, and a gene-set associated with fatty acid metabolism was downregulated in carcinomas compared to adenomas. Moreover, gene-sets associated with chromosomal location revealed chromosome 4q22 loss and chromosome 20q gain of gene-set expression as being relevant in this progression. Concluding remark These data are consistent with the notion that adenomas and carcinomas are distinct biological entities. Disruption of specific biological processes like senescence (ageing), maintenance of chromosomal instability and altered metabolism, are key factors in the progression from adenoma to carcinoma. Electronic supplementary material The online version of this article (doi:10.1007/s13402-011-0065-1) contains supplementary material, which is available to authorized users.
Collapse
|
28
|
Hirose H, Ishii H, Mimori K, Tanaka F, Takemasa I, Mizushima T, Ikeda M, Yamamoto H, Sekimoto M, Doki Y, Mori M. The significance of PITX2 overexpression in human colorectal cancer. Ann Surg Oncol 2011; 18:3005-12. [PMID: 21479692 DOI: 10.1245/s10434-011-1653-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2010] [Indexed: 01/27/2023]
Abstract
PURPOSE The paired-like homeodomain transcription factor 2 (PITX2) gene encodes a transcription factor controlled by the WNT/Dvl/CTNNB1 and Hedgehog/TGFB pathways in the pathogenesis of colorectal cancer (CRC). Although PITX2 is reportedly involved in various functions, including tissue development by controlling cell growth, its significance in CRC remains unclear. We report our findings regarding abnormal PITX2 expression in human CRC. METHODS PITX2 expression was evaluated in 5 human CRC cell lines and 92 primary CRC samples. Cell growth was evaluated after inhibition of PITX2 expression or after exogenous introduction of PITX2. RESULTS PITX2 expression was seen in all the five CRC cell lines. The study of tissue samples indicated that PITX2 expression was significantly higher in cancerous tissue than in paired control tissue (P = 0.0471). Patients with lower PITX2 expression showed a poorer overall survival rate than those with higher PITX2 expression (P = 0.0481). Multivariate analysis demonstrated that PITX2 expression was an independent prognostic factor. Experimental knockdown and introduction of PITX2 also demonstrated that the level of PITX2 expression is inversely associated with cell growth and invasion in vitro. CONCLUSIONS PITX2 expression is significantly related to the biological behavior of CRC cells and appears to be correlated with clinical survival. Thus, this study revealed a previously uncharacterized unique role and significance of PITX2 expression in CRC.
Collapse
Affiliation(s)
- Hajime Hirose
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Kim JH, Lee JY, Lee KT, Lee JK, Lee KH, Jang KT, Heo JS, Choi SH, Rhee JC. RGS16 and FosB underexpressed in pancreatic cancer with lymph node metastasis promote tumor progression. Tumour Biol 2010; 31:541-548. [PMID: 20571966 DOI: 10.1007/s13277-010-0067-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2010] [Accepted: 06/08/2010] [Indexed: 12/29/2022] Open
Abstract
Lymph node (LN) metastasis is one of the most important adverse prognostic factors for pancreatic cancer. The aim of this study was to identify novel lymphatic metastasis-associated markers for pancreatic cancer. DNA microarray analysis was used to determine and compare the expression profiles of 17 pancreatic cancer tissues with LN metastasis and 17 pancreatic cancer tissues without LN metastasis. The microarray results were validated by real-time reverse transcription-polymerase chain reaction and immunohistochemistry. Only 58 genes were differentially expressed between the two groups with a difference in signal intensity ratio greater than a 1.5-fold change. Of these genes, 30 were significantly down-regulated in the LN metastasis group. Among five selected down-regulated genes for validation using real-time PCR, the expression of DST, FosB, RGS16, and CXCL12 was significantly lower in the LN metastasis group. Immunohistochemical analysis confirmed RGS16 and FosB underexpression in pancreatic cancer tissues with LN metastasis. RGS16 and FosB underexpression was associated with poor patient survival. Our findings show that RGS16 and FosB are underexpressed in pancreatic cancer with lymph node metastasis and associated with reduced survival, suggesting that RGS16 and FosB might be prognostic markers for pancreatic cancer.
Collapse
Affiliation(s)
- Ji Hyang Kim
- Health Promotion Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | | | | | | | | | | | | | | | | |
Collapse
|