1
|
Wu X, Hou S, Ye Y, Gao Z. CXCR2P1 enhances the response of gastric cancer to PD-1 inhibitors through increasing the immune infiltration of tumors. Front Immunol 2025; 16:1545605. [PMID: 40176817 PMCID: PMC11961440 DOI: 10.3389/fimmu.2025.1545605] [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: 12/15/2024] [Accepted: 03/03/2025] [Indexed: 04/04/2025] Open
Abstract
Background Recent years, immunotherapy has emerged as a pivotal approach in cancer treatment. However, the response of gastric cancer to immunotherapy exhibits significant heterogeneity. Therefore, the early identification of gastric cancer patients who are likely to benefit from immunotherapy and the discovery of novel therapeutic targets are of critical importance. Materials and methods We collected data from European Nucleotide Archive (ENA) and Gene Expression Omnibus (GEO) databases. In project PRJEB25780, we performed WGCNA analysis and Lasso regression and chose CXCR2P1 for the subsequent analysis. Then, we compared the expression difference of CXCR2P1 among different groups. Kaplan-Meier curve was used to analyze the prognostic value of CXCR2P1, which was validated by project IMvigor210 and GEO datasets. ESTIMATE and CIBERSORT algorithm were used to evaluate the reshaping effect of CXCR2P1 to immune microenvironment of tumor. Differentially expressed genes (DEG) analysis, enrichGO analysis, Gene Set Enrichment Analysis (GSEA) and co-expression analysis were used to explore the cell biological function and signaling pathway involved in CXCR2P1. Results WGCNA identified CXCR2P1 as a hub gene significantly associated with immune response to PD-1 inhibitors in gastric cancer. CXCR2P1 expression was elevated in responders and correlated with better prognosis. Functional analysis revealed its role in reshaping the tumor immune microenvironment by promoting immune cell infiltration, including M1 macrophages, activated CD4+ T cells, and follicular helper T cells. CXCR2P1 enhanced antigen presentation via the MHC-II complex, influenced key immune pathways, such as Toll-like receptor signaling and T-cell activation, which led to the up-regulation of expression of PD-L1. GSEA showed CXCR2P1 were correlated with microRNAs. Through DEG analysis and expression analysis, MIR215 was identified as a potential direct target of CXCR2P1. Conclusion High expression of CXCR2P1 is correlated with better response to PD-1 inhibitor. It reshapes the immune microenvironment by increasing immune infiltration and changing the fraction of immune cells. In tumor immune microenvironment, CXCR2P1 can promote inflammation, enhance antigen presentation and activate the PD-1/PD-L1-related signaling pathway, which might be achieved by CXCR2P1-MIR215 axis.
Collapse
Affiliation(s)
- Xinchun Wu
- Department of Gastrointestinal Surgery, Peking University People`s Hospital, Beijing, China
- Laboratory of Surgical Oncology, Peking University People`s Hospital, Beijing, China
| | - Sen Hou
- Department of Gastrointestinal Surgery, Peking University People`s Hospital, Beijing, China
- Laboratory of Surgical Oncology, Peking University People`s Hospital, Beijing, China
| | - Yingjiang Ye
- Department of Gastrointestinal Surgery, Peking University People`s Hospital, Beijing, China
- Laboratory of Surgical Oncology, Peking University People`s Hospital, Beijing, China
- Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People’s Hospital, Beijing, China
| | - Zhidong Gao
- Department of Gastrointestinal Surgery, Peking University People`s Hospital, Beijing, China
- Laboratory of Surgical Oncology, Peking University People`s Hospital, Beijing, China
- Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People’s Hospital, Beijing, China
| |
Collapse
|
2
|
Malgundkar SH, Tamimi Y. The pivotal role of long non-coding RNAs as potential biomarkers and modulators of chemoresistance in ovarian cancer (OC). Hum Genet 2024; 143:107-124. [PMID: 38276976 DOI: 10.1007/s00439-023-02635-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 12/14/2023] [Indexed: 01/27/2024]
Abstract
Ovarian cancer (OC) is a fatal gynecological disease that is often diagnosed at later stages due to its asymptomatic nature and the absence of efficient early-stage biomarkers. Previous studies have identified genes with abnormal expression in OC that couldn't be explained by methylation or mutation, indicating alternative mechanisms of gene regulation. Recent advances in human transcriptome studies have led to research on non-coding RNAs (ncRNAs) as regulators of cancer gene expression. Long non-coding RNAs (lncRNAs), a class of ncRNAs with a length greater than 200 nucleotides, have been identified as crucial regulators of physiological processes and human diseases, including cancer. Dysregulated lncRNA expression has also been found to play a crucial role in ovarian carcinogenesis, indicating their potential as novel and non-invasive biomarkers for improving OC management. However, despite the discovery of several thousand lncRNAs, only one has been approved for clinical use as a biomarker in cancer, highlighting the importance of further research in this field. In addition to their potential as biomarkers, lncRNAs have been implicated in modulating chemoresistance, a major problem in OC. Several studies have identified altered lncRNA expression upon drug treatment, further emphasizing their potential to modulate chemoresistance. In this review, we highlight the characteristics of lncRNAs, their function, and their potential to serve as tumor markers in OC. We also discuss a few databases providing detailed information on lncRNAs in various cancer types. Despite the promising potential of lncRNAs, further research is necessary to fully understand their role in cancer and develop effective strategies to combat this devastating disease.
Collapse
Affiliation(s)
- Shika Hanif Malgundkar
- Biochemistry Department, College of Medicine and Health Sciences, Sultan Qaboos University, PC 123, PO Box 35, Muscat, Sultanate of Oman
| | - Yahya Tamimi
- Biochemistry Department, College of Medicine and Health Sciences, Sultan Qaboos University, PC 123, PO Box 35, Muscat, Sultanate of Oman.
| |
Collapse
|
3
|
Ellen JG, Jacob E, Nikolaou N, Markuzon N. Autoencoder-based multimodal prediction of non-small cell lung cancer survival. Sci Rep 2023; 13:15761. [PMID: 37737469 PMCID: PMC10517020 DOI: 10.1038/s41598-023-42365-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 09/09/2023] [Indexed: 09/23/2023] Open
Abstract
The ability to accurately predict non-small cell lung cancer (NSCLC) patient survival is crucial for informing physician decision-making, and the increasing availability of multi-omics data offers the promise of enhancing prognosis predictions. We present a multimodal integration approach that leverages microRNA, mRNA, DNA methylation, long non-coding RNA (lncRNA) and clinical data to predict NSCLC survival and identify patient subtypes, utilizing denoising autoencoders for data compression and integration. Survival performance for patients with lung adenocarcinoma (LUAD) and squamous cell carcinoma (LUSC) was compared across modality combinations and data integration methods. Using The Cancer Genome Atlas data, our results demonstrate that survival prediction models combining multiple modalities outperform single modality models. The highest performance was achieved with a combination of only two modalities, lncRNA and clinical, at concordance indices (C-indices) of 0.69 ± 0.03 for LUAD and 0.62 ± 0.03 for LUSC. Models utilizing all five modalities achieved mean C-indices of 0.67 ± 0.04 and 0.63 ± 0.02 for LUAD and LUSC, respectively, while the best individual modality performance reached C-indices of 0.64 ± 0.03 for LUAD and 0.59 ± 0.03 for LUSC. Analysis of biological differences revealed two distinct survival subtypes with over 900 differentially expressed transcripts.
Collapse
Affiliation(s)
- Jacob G Ellen
- Institute of Health Informatics, University College London, London, UK.
| | - Etai Jacob
- AstraZeneca, Oncology Data Science, Waltham, MA, USA
| | | | | |
Collapse
|
4
|
Gao L, Zhou L, Huang X. Identification of Novel Kinase-Transcription Factor-mRNA-miRNA Regulatory Network in Nasopharyngeal Carcinoma by Bioinformatics Analysis. Int J Gen Med 2021; 14:7453-7469. [PMID: 34744455 PMCID: PMC8566004 DOI: 10.2147/ijgm.s327657] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 10/05/2021] [Indexed: 12/24/2022] Open
Abstract
Purpose Nasopharyngeal carcinoma (NPC) is one of the most common malignant tumors of the head and neck. This study aimed to investigate the crucial genes and regulatory networks involved in the carcinogenesis of NPC using a bioinformatics approach. Methods Five mRNA and two miRNA expression datasets were downloaded from the Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) and miRNAs (DEMs) between NPC and normal samples were analyzed using R software. The WebGestalt tool was used for functional enrichment analysis, and protein-protein interaction (PPI) network analysis of DEGs was performed using STRING database. Transcription factors (TFs) were predicted using TRRUST and Transcriptional Regulatory Element Database (TRED). Kinases were identified using X2Kgui. The miRNAs of DEGs were predicted using miRWalk database. A kinase-TF-mRNA-miRNA integrated network was constructed, and hub nodes were selected. The hub genes were validated using NPC datasets from the GEO and Oncomine databases. Finally, candidate small-molecule agents were predicted using CMap. Results A total of 122 DEGs and 44 DEMs were identified. DEGs were associated with the immune response, leukocyte activation, endoplasmic reticulum stress in GO analysis, and the NF-κB signaling pathway in KEGG analysis. Four significant modules were identified using PPI network analysis. Subsequently, 26 TFs, 73 kinases, and 2499 miRNAs were predicted. The predicted miRNAs were cross-referenced with DEMs, and seven overlapping miRNAs were selected. In the kinase-TF-mRNA-miRNA integrated network, eight genes (PTGS2, FN1, MMP1, PLAU, MMP3, CD19, BMP2, and PIGR) were identified as hub genes. Hub genes were validated with consistent results, indicating the reliability of our findings. Finally, six candidate small-molecule agents (phenoxybenzamine, luteolin, thioguanosine, reserpine, blebbistatin, and camptothecin) were predicted. Conclusion We identified DEGs and an NPC regulatory network involving kinases, TFs, mRNAs, and miRNAs, which might provide promising insight into the pathogenesis, treatment, and prognosis of NPC.
Collapse
Affiliation(s)
- Li Gao
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Lei Zhou
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Zhongshan Hospital Affiliated to Fudan University, Shanghai, People's Republic of China
| | - Xinsheng Huang
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Zhongshan Hospital Affiliated to Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
5
|
Xue Y, Wang P, Jiang F, Yu J, Ding H, Zhang Z, Pei H, Li B. A Newly Identified lncBCAS1-4_1 Associated With Vitamin D Signaling and EMT in Ovarian Cancer Cells. Front Oncol 2021; 11:691500. [PMID: 34422647 PMCID: PMC8377733 DOI: 10.3389/fonc.2021.691500] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 07/14/2021] [Indexed: 11/13/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) were identified rapidly due to their important role in many biological processes and human diseases including cancer. 1α,25-dihydroxyvitamin D3 [1α,25(OH)2D3] and its analogues are widely applied as preventative and therapeutic anticancer agents. However, the expression profile of lncRNAs regulated by 1α,25(OH)2D3 in ovarian cancer remains to be clarified. In the present study, we found 606 lncRNAs and 102 mRNAs that showed differential expression (DE) based on microarray data. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis indicated that the DE genes were mainly enriched in TGF-β, MAPK, Ras, PI3K-Akt, and Hippo signaling pathways, as well as the vitamin D-related pathway. We further assessed the potential lncRNAs that linked vitamin D signaling with EMT, and lncBCAS1-4_1 was identified in the first time. Moreover, we found that the most upregulated lncBCAS1-4_1 showed 75% same transcripts with CYP24A1 (metabolic enzyme of 1α,25(OH)2D3). Finally, the lncBCAS1-4_1 gain-of-function cell model was established, which demonstrated that the knockdown of lncBCAS1-4_1 inhibited the proliferation and migration of ovarian cancer cells. Furthermore, lncBCAS1-4_1 could resist the antitumor effect of 1α,25(OH)2D3, which was associated with upregulated ZEB1. These data provide new evidences that lncRNAs served as a target for the antitumor effect of 1α,25(OH)2D3.
Collapse
Affiliation(s)
- Yaqi Xue
- Deparment of Nutrition and Food Hygiene, Medical College of Soochow University, Suzhou, China.,Department of Clinical Nutrition, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ping Wang
- Deparment of Nutrition and Food Hygiene, Medical College of Soochow University, Suzhou, China
| | - Fei Jiang
- Deparment of Nutrition and Food Hygiene, Medical College of Soochow University, Suzhou, China
| | - Jing Yu
- Deparment of Nutrition and Food Hygiene, Medical College of Soochow University, Suzhou, China
| | - Hongmei Ding
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zengli Zhang
- Deparment of Nutrition and Food Hygiene, Medical College of Soochow University, Suzhou, China
| | - Hailong Pei
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Centre of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Bingyan Li
- Deparment of Nutrition and Food Hygiene, Medical College of Soochow University, Suzhou, China
| |
Collapse
|
6
|
Zhu L, Mei M. Interference of long non-coding RNA HAGLROS inhibits the proliferation and promotes the apoptosis of ovarian cancer cells by targeting miR-26b-5p. Exp Ther Med 2021; 22:879. [PMID: 34194557 PMCID: PMC8237406 DOI: 10.3892/etm.2021.10311] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 02/17/2021] [Indexed: 12/29/2022] Open
Abstract
Ovarian cancer (OV) is the fifth most common type of cancer affecting women worldwide. Long non-coding RNAs (lncRNAs) serve essential roles in the progression of OV. As such, the present study aimed to investigate the specific role of HAGLR opposite strand lncRNA (HAGLROS) in OV and the underlying mechanism of action through which HAGLROS exerts its effects on OV cells. In the present study, the expression of HAGLROS in several OV cell lines was first detected using reverse transcription-quantitative PCR. HAGLROS was then silenced to evaluate cell viability, proliferation and apoptosis, which were determined using Cell Counting Kit-8, colony formation and TUNEL assays, respectively. Additionally, immunofluorescence staining and western blotting were used to confirm the expression profile of proliferation- and apoptosis-related proteins. Moreover, a dual luciferase reporter assay was used to verify the potential interactions between HAGLROS and microRNA (miR)-26b-5p. Subsequently, rescue assays were performed to investigate the effects of HAGLROS and miR-26b-5p on OV progression. The results indicated that HAGLROS was highly expressed in OV cells. Interference of HAGLROS led to a decrease in the proliferation, but an increase in the apoptosis of OV cells, accompanied by downregulated expression levels of Ki67 and Bcl-2, and upregulated expression levels of Bax and cleaved caspase-3. Further study revealed that HAGLROS acted as a sponge for miR-26b-5p and positively regulated its expression. miR-26b-5p inhibitor transfection partially reversed the effects of HAGLROS knockdown on the proliferation and apoptosis of OV cells. In conclusion, the results of the present study suggested that interference of HAGLROS suppressed the proliferation and promoted the apoptosis of OV cells through regulating miR-26b-5p, indicating that HAGLROS may be a promising biomarker in OV diagnosis and treatment.
Collapse
Affiliation(s)
- Lin Zhu
- Department of Gynecology, Beijing Haidian District Maternal and Child Health Hospital, Beijing 100080, P.R. China
| | - Mei Mei
- Department of Obstetrics and Gynecology, Xiangyang No. 1 People's Hospital Affiliated to Hubei Medical College, Xiangyang, Hubei 441000, P.R. China
| |
Collapse
|
7
|
López-Camarillo C, Ruíz-García E, Salinas-Vera YM, Silva-Cázares MB, Hernández-de la Cruz ON, Marchat LA, Gallardo-Rincón D. Deciphering the Long Non-Coding RNAs and MicroRNAs Coregulation Networks in Ovarian Cancer Development: An Overview. Cells 2021; 10:1407. [PMID: 34204094 PMCID: PMC8227049 DOI: 10.3390/cells10061407] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/29/2021] [Accepted: 06/01/2021] [Indexed: 01/17/2023] Open
Abstract
Non-coding RNAs are emergent elements from the genome, which do not encode for proteins but have relevant cellular functions impacting almost all the physiological processes occurring in eukaryotic cells. In particular, microRNAs and long non-coding RNAs (lncRNAs) are a new class of small RNAs transcribed from the genome, which modulate the expression of specific genes at transcriptional and posttranscriptional levels, thus adding a new regulatory layer in the flux of genetic information. In cancer cells, the miRNAs and lncRNAs interactions with its target genes and functional pathways are deregulated as a consequence of epigenetic and genetic alterations occurring during tumorigenesis. In this review, we summarize the actual knowledge on the interplay of lncRNAs with its cognate miRNAs and mRNAs pairs, which interact in coregulatory networks with a particular emphasis on the mechanisms underlying its oncogenic behavior in ovarian cancer. Specifically, we reviewed here the evidences unraveling the relevant roles of lncRNAs/miRNAs pairs in altered regulation of cell migration, angiogenesis, therapy resistance, and Warburg effect. Finally, we also discussed its potential clinical implications in ovarian cancer and related endocrine disease therapies.
Collapse
Affiliation(s)
- César López-Camarillo
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, 03100 CDMX, Mexico;
- Grupo de Investigación en Cáncer de Ovario y Endometrio, Instituto Nacional de Cancerología, 14080 CDMX, Mexico; (E.R.-G.); (D.G.-R.)
| | - Erika Ruíz-García
- Grupo de Investigación en Cáncer de Ovario y Endometrio, Instituto Nacional de Cancerología, 14080 CDMX, Mexico; (E.R.-G.); (D.G.-R.)
- Laboratorio de Medicina Traslacional y Departamento de Tumores Gastrointestinales, Instituto Nacional de Cancerología, 14080 CDMX, Mexico
| | | | - Macrina B. Silva-Cázares
- Coordinación Académica Región Altiplano, Universidad Autónoma de San Luis Potosí, 78700 San Luis Potosí, Mexico;
| | | | - Laurence A. Marchat
- Programa en Biomedicina Molecular y Red de Biotecnología, Instituto Politécnico Nacional, 07340 CDMX, Mexico;
| | - Dolores Gallardo-Rincón
- Grupo de Investigación en Cáncer de Ovario y Endometrio, Instituto Nacional de Cancerología, 14080 CDMX, Mexico; (E.R.-G.); (D.G.-R.)
- Laboratorio de Medicina Traslacional y Departamento de Tumores Gastrointestinales, Instituto Nacional de Cancerología, 14080 CDMX, Mexico
| |
Collapse
|
8
|
PPARD May Play a Protective Role against the Development of Schizophrenia. PPAR Res 2020; 2020:3480412. [PMID: 32831816 PMCID: PMC7428834 DOI: 10.1155/2020/3480412] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 05/29/2020] [Indexed: 11/24/2022] Open
Abstract
PPARD has been suggested to contribute to the etiology of schizophrenia (SCZ) with the underlying mechanisms largely unknown. Here, we first collected and analyzed the PPARD expression profile from three groups: (1) 18 healthy control (HC) subjects, (2) 14 clinical high-risk (CHR) patients, and (3) 19 early onset of SCZ (EOS) patients. After that, we conducted a systematical pathway analysis to explore the potential mechanisms involved in PPARD exerting influence on the pathological development of SCZ. Compared to the HC group, the expression of PPARD was slightly decreased in the EOS group (LFC = −0.34; p = 0.23) and increased in the CHR group (LFC = 0.65; p = 0.20). However, there was a significant difference between the EOS group and the CHR group (LFC = −0.99; p = 0.015), reflecting the amount of variation in PPARD expression before and after the onset of SCZ. Pathway analysis suggested that overexpression of PPARD may regulate ten proteins or molecules to inhibit the pathological development of SCZ, including the deactivation of eight SCZ promoters and stimulation of two SCZ inhibitors. Our results support the association between PPARD and SCZ. The pathways identified may help in the understanding of the potential mechanisms by which PPARD contributes to the etiology of SCZ.
Collapse
|
9
|
Lerebours A, Robson S, Sharpe C, Nagorskaya L, Gudkov D, Haynes-Lovatt C, Smith JT. Transcriptional Changes in the Ovaries of Perch from Chernobyl. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2020; 54:10078-10087. [PMID: 32686935 DOI: 10.1021/acs.est.0c02575] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Fish have been highly exposed to radiation in freshwater systems after the Chernobyl Nuclear Power Plant (NPP) accident in 1986 and in freshwater and marine systems after the more recent Fukushima NPP accident in 2011. In the years after the accident, the radioactivity levels rapidly declined due to radioactive decay and environmental processes, but chronic lower dose exposures persisted. To gain insights into the long-term effects of environmental low dose radiation on fish ovaries development, a high-throughput transcriptomic approach including a de novo assembly was applied to different gonad phenotypes of female perch: developed gonads from reference lakes, developed/irradiated from medium contaminated lake, and both developed/irradiated and undeveloped from more highly contaminated lakes. This is the most comprehensive analysis to date of the gene responses in wildlife reproductive system to radiation. Some gene responses that were modulated in irradiated gonads were found to be involved in biological processes including cell differentiation and proliferation (ggnb2, mod5, rergl), cytoskeleton organization (k1C18, mtpn), gonad development (nell2, tcp4), lipid metabolism (ldah, at11b, nltp), reproduction (cyb5, cyp17A, ovos), DNA damage repair (wdhd1, rad51, hus1), and epigenetic mechanisms (dmap1). Identification of these genes provides a better understanding of the underlying molecular mechanisms underpinning the development of the gonad phenotypes of wild perch and how fish may respond to chronic exposure to radiation in their natural environment, though causal attribution of gene responses remains unclear in the undeveloped gonads.
Collapse
Affiliation(s)
- Adélaïde Lerebours
- School of the Environment, Geography and Geosciences, University of Portsmouth, Portsmouth PO1 3QL, United Kingdom
- School of Biological Sciences, University of Portsmouth, Portsmouth PO1 2DY, United Kingdom
| | - Samuel Robson
- Centre for Enzyme Innovation, University of Portsmouth, Portsmouth PO1 2DT, United Kingdom
| | - Colin Sharpe
- School of Biological Sciences, University of Portsmouth, Portsmouth PO1 2DY, United Kingdom
| | - Liubov Nagorskaya
- Applied Science Center for Bioresources of the National Academy of Sciences of Belarus, Minsk 220072, Belarus
| | - Dmitri Gudkov
- Institute of Hydrobiology of the National Academy of Sciences of Ukraine, Kiev UA-04210, Ukraine
| | | | - Jim T Smith
- School of the Environment, Geography and Geosciences, University of Portsmouth, Portsmouth PO1 3QL, United Kingdom
| |
Collapse
|
10
|
Xia X, Burn MS, Chen Y, Karakaya C, Kallen A. The relationship between H19 and parameters of ovarian reserve. Reprod Biol Endocrinol 2020; 18:46. [PMID: 32404103 PMCID: PMC7218823 DOI: 10.1186/s12958-020-00578-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 02/25/2020] [Indexed: 02/06/2023] Open
Abstract
CONTEXT The H19 long noncoding RNA (lncRNA) belongs to a highly conserved, imprinted gene cluster involved in embryonic development and growth control. We previously described a novel mechanism whereby the Anti-mullerian hormone (Amh) appears to be regulated by H19. However, the relationship between circulating H19 and markers of ovarian reserve including AMH not been investigated. OBJECTIVE To determine whether H19 expression is altered in women with decreased ovarian reserve. DESIGN Experimental study. SETTING Yale School of Medicine (New Haven, USA) and Gazi University School of Medicine (Ankara, Turkey). PATIENTS OR OTHER PARTICIPANTS A total of 141 women undergoing infertility evaluation and treatment. INTERVENTION Collection of discarded blood samples and cumulus cells at the time of baseline infertility evaluation and transvaginal oocyte retrieval, respectively. MAIN OUTCOME MEASURE Serum and cumulus cell H19 expression. RESULTS Women with diminished ovarian reserve (as determined by AMH) had significantly lower serum H19 expression levels as compared to controls (p < 0.01). Serum H19 was moderately positively correlated with serum AMH. H19 expression was increased 3.7-fold in cumulus cells of IVF patients who demonstrated a high response to gonadotropins, compared to low responders (p < 0.05). CONCLUSION In this study, we show that downregulation of H19 in serum and cumulus cells is closely associated with decreased ovarian reserve, as measured by decreased AMH levels and reduced oocyte yield at oocyte retrieval. Further study with expanded sample sizes is necessary to determine whether H19 may be of use as a novel biomarker for diminished ovarian reserve.
Collapse
Affiliation(s)
- Xi Xia
- Reproductive Center, Peking University Shenzhen Hospital, Shenzhen 518000, China
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Yale School of Medicine, Gynecology, and Reproductive Sciences, New Haven, Connecticut 06512 USA
| | - Martina S. Burn
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Yale School of Medicine, Gynecology, and Reproductive Sciences, New Haven, Connecticut 06512 USA
| | - Yong Chen
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Yale School of Medicine, Gynecology, and Reproductive Sciences, New Haven, Connecticut 06512 USA
- Department of Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province 350122 P.R. China
| | - Cengiz Karakaya
- Department of Medical Biochemistry, Gazi University School of Medicine, Ankara, Turkey
| | - Amanda Kallen
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Yale School of Medicine, Gynecology, and Reproductive Sciences, New Haven, Connecticut 06512 USA
| |
Collapse
|
11
|
Salamini-Montemurri M, Lamas-Maceiras M, Barreiro-Alonso A, Vizoso-Vázquez Á, Rodríguez-Belmonte E, Quindós-Varela M, Cerdán ME. The Challenges and Opportunities of LncRNAs in Ovarian Cancer Research and Clinical Use. Cancers (Basel) 2020; 12:E1020. [PMID: 32326249 PMCID: PMC7225988 DOI: 10.3390/cancers12041020] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/15/2020] [Accepted: 04/17/2020] [Indexed: 12/24/2022] Open
Abstract
Ovarian cancer is one of the most lethal gynecological malignancies worldwide because it tends to be detected late, when the disease has already spread, and prognosis is poor. In this review we aim to highlight the importance of long non-coding RNAs (lncRNAs) in diagnosis, prognosis and treatment choice, to make progress towards increasingly personalized medicine in this malignancy. We review the effects of lncRNAs associated with ovarian cancer in the context of cancer hallmarks. We also discuss the molecular mechanisms by which lncRNAs become involved in cellular physiology; the onset, development and progression of ovarian cancer; and lncRNAs' regulatory mechanisms at the transcriptional, post-transcriptional and post-translational stages of gene expression. Finally, we compile a series of online resources useful for the study of lncRNAs, especially in the context of ovarian cancer. Future work required in the field is also discussed along with some concluding remarks.
Collapse
Affiliation(s)
- Martín Salamini-Montemurri
- EXPRELA Group, Centro de Investigacións Científicas Avanzadas (CICA), Departamento de Bioloxía, Facultade de Ciencias, INIBIC-Universidade da Coruña, Campus de A Coruña, 15071 A Coruña, Spain; (M.S.-M.); (M.L.-M.); (A.B.-A.); (E.R.-B.)
| | - Mónica Lamas-Maceiras
- EXPRELA Group, Centro de Investigacións Científicas Avanzadas (CICA), Departamento de Bioloxía, Facultade de Ciencias, INIBIC-Universidade da Coruña, Campus de A Coruña, 15071 A Coruña, Spain; (M.S.-M.); (M.L.-M.); (A.B.-A.); (E.R.-B.)
| | - Aida Barreiro-Alonso
- EXPRELA Group, Centro de Investigacións Científicas Avanzadas (CICA), Departamento de Bioloxía, Facultade de Ciencias, INIBIC-Universidade da Coruña, Campus de A Coruña, 15071 A Coruña, Spain; (M.S.-M.); (M.L.-M.); (A.B.-A.); (E.R.-B.)
| | - Ángel Vizoso-Vázquez
- EXPRELA Group, Centro de Investigacións Científicas Avanzadas (CICA), Departamento de Bioloxía, Facultade de Ciencias, INIBIC-Universidade da Coruña, Campus de A Coruña, 15071 A Coruña, Spain; (M.S.-M.); (M.L.-M.); (A.B.-A.); (E.R.-B.)
| | - Esther Rodríguez-Belmonte
- EXPRELA Group, Centro de Investigacións Científicas Avanzadas (CICA), Departamento de Bioloxía, Facultade de Ciencias, INIBIC-Universidade da Coruña, Campus de A Coruña, 15071 A Coruña, Spain; (M.S.-M.); (M.L.-M.); (A.B.-A.); (E.R.-B.)
| | - María Quindós-Varela
- Translational Cancer Research Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Carretera del Pasaje s/n, 15006 A Coruña, Spain;
| | - María Esperanza Cerdán
- EXPRELA Group, Centro de Investigacións Científicas Avanzadas (CICA), Departamento de Bioloxía, Facultade de Ciencias, INIBIC-Universidade da Coruña, Campus de A Coruña, 15071 A Coruña, Spain; (M.S.-M.); (M.L.-M.); (A.B.-A.); (E.R.-B.)
| |
Collapse
|
12
|
Hu J, Wang L, Zhao W, Huang Y, Wang Z, Shen H. mi-R4435-2HG promotes proliferation and inhibits apoptosis of cancer cells in ovarian carcinoma by upregulating ROCK2. Oncol Lett 2019; 19:1305-1309. [PMID: 31966061 PMCID: PMC6956214 DOI: 10.3892/ol.2019.11204] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 04/24/2019] [Indexed: 11/07/2022] Open
Abstract
The present study aimed to investigate the involvement of long noncoding RNA mi-R4435-2HG in ovarian carcinoma. mi-R4435-2HG and Rho-associated protein kinase 2 (ROCK2) were demonstrated to both be upregulated in ovarian carcinoma. mi-R4435-2HG and ROCK2 were positively correlated in both tumor and healthy tissues. mi-R4435-2HG overexpression mediated the upregulation of ROCK2, while upregulation of ROCK2 did not affect mi-R4435-2HG. Overexpression of mi-R4435-2HG and ROCK2 led to promoted proliferation and inhibited apoptosis of cancer cells, while mi-R4435-2HG and ROCK2 knockdown had the opposite effect. In addition, ROCK2 knockdown attenuated the effects of mi-R4435-2HG overexpression on cancer cell proliferation and apoptosis. Therefore, mi-R4435-2HG promotes proliferation and inhibits apoptosis of cancer cells in ovarian carcinoma by upregulating ROCK2.
Collapse
Affiliation(s)
- Jinghui Hu
- Department of Gynecology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Li Wang
- Department of Gynecology and Οbstetrics, Changzhou Maternal and Child Health Care Hospital Affiliated to Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| | - Wei Zhao
- Department of Gynecology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Yujie Huang
- Department of Gynecology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Zhe Wang
- Department of Gynecology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Heping Shen
- Department of Gynecology, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| |
Collapse
|
13
|
Zhou W, Pan B, Liu L. Integrated bioinformatics analysis revealing independent prognostic long non-coding RNAs DNAH17-AS1 and RP11-400N13.2 and their potential oncogenic roles in colorectal cancer. Oncol Lett 2019; 18:3705-3715. [PMID: 31516583 PMCID: PMC6732947 DOI: 10.3892/ol.2019.10730] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 06/24/2019] [Indexed: 02/06/2023] Open
Abstract
The aberrant expression of long non-coding RNAs (lncRNAs) has been associated with a variety of malignancies, including colorectal cancer (CRC); however, the key lncRNAs associated with patient prognosis and their biological roles in CRC are yet to be determined. The aim of the present study was to determine the key lncRNAs associated with patient prognosis as well as their biological roles in CRC. Therefore, a dataset from The Cancer Genome Atlas containing the lncRNA expression data of 521 CRC and normal colorectal mucosal tissues, as well as the corresponding clinical data, were screened. A total of 1,180 significantly differentially expressed lncRNAs were associated with CRC as determined by t-tests in edgeR. Kaplan-Meier analysis revealed that 56 of the 1,180 lncRNAs were associated with overall survival (OS); 7 of the 56 lncRNAs were identified as key lncRNAs associated with the Tumor-Node-Metastasis stage of CRC by Kruskal-Wallis test. Subsequent univariate and multivariate Cox regression analyses of the 7 lncRNAs revealed 2 lncRNAs, DNAH17-AS1 and RP11-400N13.2, as potential independent prognostic factors for the OS of patients with CRC. Furthermore, the expression levelsof these 2 lncRNAs were significantly upregulated in CRC compared with those in normal tissues, which suggested that they may serve an oncogenic role in CRC. In addition, networks comprising the 2 lncRNAs and their respective co-expressed protein-coding genes (PCGs) were constructed using cor.test in R. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses of these PCGs were conducted; DNAH17-AS1- and RP11-400N13.2-associated PCGs were reported to be involved in G-protein coupling-related functions. Thus, these independent prognostic lncRNAs and their associated functions identified in the present study may provide novel insight into potential prognostic biomarkers and therapeutic targets for the treatment of CRC.
Collapse
Affiliation(s)
- Wen Zhou
- Clinical Laboratory, Tianjin Hospital, Tianjin 300211, P.R. China
| | - Boyu Pan
- Department of Gastrointestinal Cancer Biology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin Clinical Research Center for Cancer, Tianjin 300060, P.R. China.,National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin Clinical Research Center for Cancer, Tianjin 300060, P.R. China
| | - Liren Liu
- Department of Gastrointestinal Cancer Biology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin Clinical Research Center for Cancer, Tianjin 300060, P.R. China.,National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin Clinical Research Center for Cancer, Tianjin 300060, P.R. China
| |
Collapse
|
14
|
Bao W, Cao F, Ni S, Yang J, Li H, Su Z, Zhao B. lncRNA FLVCR1-AS1 regulates cell proliferation, migration and invasion by sponging miR-485-5p in human cholangiocarcinoma. Oncol Lett 2019; 18:2240-2247. [PMID: 31404302 PMCID: PMC6676717 DOI: 10.3892/ol.2019.10577] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Accepted: 04/18/2019] [Indexed: 12/19/2022] Open
Abstract
Long non-coding RNA (lncRNA) FLVCR1 antisense RNA 1 (FLVCR1-AS1) serves a crucial role in many types of cancer; however, to the best of our knowledge, the biological effect of FLVCR1-AS1 in cholangiocarcinoma (CCA) remains unclear. The present study aimed to elucidate the involvement of FLVCR1-AS1 in the regulation of human CCA cell growth, migration and invasion, as well as the mechanisms underlying its effect. The expression levels of FLVCR1-AS1 in CCA tumor tissues, adjacent normal tissues, CCA cell lines and a cholangiocyte cell line were determined by reverse transcription-quantitative polymerase chain reaction. A significantly higher expression level of FLVCR1-AS1 was identified in CCA tumor tissues and the CCA cell lines HuCCT1 and CCLP1 compared with the normal controls. Short hairpin RNA targeting FLVCR1-AS1 (shFLVCR1-AS1) and a control plasmid (shNC) were transfected into CCA cell lines. Cell proliferation, colony formation, migration and invasion of CCA cells transfected with shFLVCR1-AS1 were significantly suppressed compared with the shNC groups. The expression levels of migration and invasion-associated proteins, including Twist, matrix metalloproteinase (MMP)-2 and MMP-9, were also significantly suppressed by shFLVCR1-AS1-treatment. Furthermore, FLVCR1-AS1 knockdown inhibited tumor growth in a xenograft model. Mechanistically, FLVCR1-AS1 was demonstrated to sponge microRNA-485-5p (miR-485-5p) in human CCA. The expression of miR-458-5p was significantly decreased in CCA tissue compared with normal tissue, and Pearson's correlation analysis revealed that FLVCR1-AS1 expression was negatively correlated with miR-485-5p expression in CCA tissues. These results suggested that lncRNA FLVCR1-AS1 may be used as a novel therapeutic target and a potential diagnostic marker for CCA.
Collapse
Affiliation(s)
- Wenqing Bao
- Department of General Surgery, Shanghai Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200135, P.R. China
| | - Feng Cao
- Department of General Surgery, Shanghai Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200135, P.R. China
| | - Shubin Ni
- Department of General Surgery, Shanghai Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200135, P.R. China
| | - Jing Yang
- Department of General Surgery, Shanghai Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200135, P.R. China
| | - Haidong Li
- Department of General Surgery, Shanghai Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200135, P.R. China
| | - Zhenxi Su
- Department of General Surgery, Shanghai Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200135, P.R. China
| | - Bin Zhao
- Department of General Surgery, Shanghai Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200135, P.R. China
| |
Collapse
|
15
|
Zhang Y, Liu YX, Xiao QX, Liu Q, Deng R, Bian J, Deng IB, Al-Hawwas M, Yu FX. Microarray Expression Profiles of lncRNAs and mRNAs in Postoperative Cognitive Dysfunction. Front Neurosci 2018; 12:694. [PMID: 30349449 PMCID: PMC6187303 DOI: 10.3389/fnins.2018.00694] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 09/18/2018] [Indexed: 12/17/2022] Open
Abstract
Postoperative cognitive dysfunction (POCD) is serious disorder in the central nervous system common in aged patients after anesthesia. Although its clinical symptoms are well recognized, however, the molecular etiology of the POCD remains unrevealed. Similarly, neither gold standard molecular diagnosis nor effective treatment is available for POCD until the present. Therefore, we aimed to explore the molecular mechanism of this disorder through investigating lncRNAs and mRNAs associated with POCD human patients and investigate their underlying regulatory pathways. In this study, we recruited 200 patients requiring hip or knee replacement surgery. Their neurological functions were assessed at two time points, 1 day before the surgery and 30 days post-surgery. In parallel, serum samples were collected from the participants to analyze lncRNAs and mRNAs differential expression profile between POCD and non-POCD patients using microarray analysis. To further investigate the role differentially expressed mRNA and lncRNAs, Gene Ontology (GO), pathway analyses on mRNAs and lncRNA-mRNA interaction network were performed. As a result, 68 lncRNAs and 115 mRNAs were dysregulated in the POCD group compared to non-POCD group. Among them, the top 10 upregulated lncRNAs and 10 downregulated lncRNAs were listed for enrichment analysis. Interestingly, we found that these lncRNA and mRNA are involved in biological process, molecular function, and cellular component in addition to various signaling pathways, suggesting that the pathogenesis of POCD involves lncRNAs and mRNAs differential expression. Consequently, the genetic dysregulation between the non-POCD and POCD patients participates in the occurrence and development of POCD, and could be served as diagnostic biomarkers and drug targets for POCD treatment.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Anesthesiology, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Yue-Xin Liu
- Department of Anesthesiology, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Qiu-Xia Xiao
- Department of Anesthesiology, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Qing Liu
- Department of Anesthesiology, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Rui Deng
- Department of Anesthesiology, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Jiang Bian
- Department of Anesthesiology, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Isaac Bul Deng
- School of Pharmacy and Medical Sciences, Sansom Institute, Division of Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Mohammed Al-Hawwas
- School of Pharmacy and Medical Sciences, Sansom Institute, Division of Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Feng-Xu Yu
- Department of Cardiothoracic Surgery, Affiliated Hospital, Southwest Medical University, Luzhou, China
| |
Collapse
|