1
|
Liu ZZ, Ji FH, Piao Y. Non-coding RNAs participate in interactions between senescence and gastrointestinal cancers. Front Genet 2025; 15:1461404. [PMID: 39831201 PMCID: PMC11739115 DOI: 10.3389/fgene.2024.1461404] [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: 07/08/2024] [Accepted: 12/04/2024] [Indexed: 01/22/2025] Open
Abstract
Relationships between cellular senescence and gastrointestinal cancers have gained prominence in recent years. The currently accepted theory suggests that cellular senescence and cancer occurrence exhibit "double-edged sword" effects. Cellular senescence is related to cancer via four "meta-hallmarks" i.e., genomic instability, epigenetic alterations, chronic inflammation, and dysbiosis, along with two "antagonistic hallmarks" i.e., telomere attrition and stem cell exhaustion. These relationships are characterized by both agonistic and antagonistic elements, but the existence of an intricate dynamic balance remains unknown. Non-coding RNAs (ncRNAs) have vital roles in post-transcriptional regulation, but how they participate in agonistic and antagonistic relationships between cellular senescence and gastrointestinal cancers remains to be fully investigated. In this article, we systematically review how ncRNAs (including microRNAs (miRNAs), long ncRNAs (lncRNAs), and circularRNAs (circRNAs)) participate in interactions between cellular senescence and gastrointestinal cancers. Our aim is to elucidate a triangular relationship between "ncRNAs-senescence-gastrointestinal cancers" which considered these three elements as an equal important standing. We are keen to identify prognostic or therapeutic targets for gastrointestinal cancers from, i.e., aging-related ncRNAs, or discover novel strategies to treat and manage in the elderly. We seek to clarify complex relationships where ncRNAs participate in "senescence-gastrointestinal cancers" interactions.
Collapse
Affiliation(s)
| | | | - Ying Piao
- Department of Oncology, General Hospital of Northern Theater Command, Shenyang, China
| |
Collapse
|
2
|
Nickerson JA, Momen-Heravi F. Long non-coding RNAs: roles in cellular stress responses and epigenetic mechanisms regulating chromatin. Nucleus 2024; 15:2350180. [PMID: 38773934 PMCID: PMC11123517 DOI: 10.1080/19491034.2024.2350180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 04/22/2024] [Indexed: 05/24/2024] Open
Abstract
Most of the genome is transcribed into RNA but only 2% of the sequence codes for proteins. Non-coding RNA transcripts include a very large number of long noncoding RNAs (lncRNAs). A growing number of identified lncRNAs operate in cellular stress responses, for example in response to hypoxia, genotoxic stress, and oxidative stress. Additionally, lncRNA plays important roles in epigenetic mechanisms operating at chromatin and in maintaining chromatin architecture. Here, we address three lncRNA topics that have had significant recent advances. The first is an emerging role for many lncRNAs in cellular stress responses. The second is the development of high throughput screening assays to develop causal relationships between lncRNAs across the genome with cellular functions. Finally, we turn to recent advances in understanding the role of lncRNAs in regulating chromatin architecture and epigenetics, advances that build on some of the earliest work linking RNA to chromatin architecture.
Collapse
Affiliation(s)
- Jeffrey A Nickerson
- Division of Genes & Development, Department of Pediatrics, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Fatemeh Momen-Heravi
- College of Dental Medicine, Columbia University Medical Center, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| |
Collapse
|
3
|
Desiderio A, Pastorino M, Campitelli M, Prevenzano I, De Palma FDE, Spinelli R, Parrillo L, Longo M, Milone M, Miele C, Raciti GA, Beguinot F. Hypomethylation at PANDAR promoter progressively induces senescence in adipocyte precursor cells in subjects with obesity and type 2 diabetes. FASEB J 2024; 38:e70093. [PMID: 39373976 DOI: 10.1096/fj.202401470r] [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: 06/28/2024] [Revised: 09/12/2024] [Accepted: 09/23/2024] [Indexed: 10/08/2024]
Abstract
The risk of developing type 2 diabetes (T2D) is heterogeneous among individuals with obesity. Functional decline of adipocyte precursor cells (APCs) and accumulation of senescent cells in the subcutaneous adipose tissue contributes to the progression toward T2D. LncRNAs regulate cell senescence and may be implicated in determining this abnormality in APCs. Here, we report that APCs from individuals with obesity show a gradual increase in multiple senescence markers, which worsens in parallel with the progression from normal glucose tolerance (NGT) to impaired glucose tolerance (IGT) or T2D. Transcriptomic analysis identified PANDAR as the top-ranked lncRNA differentially expressed in APCs from individuals with obesity and T2D and non-obese subjects. Q-PCR confirmed PANDAR up-regulation in APCs from individuals with obesity, at progressively increased levels in those who developed, respectively, IGT and T2D. Bisulfite sequencing and luciferase assays revealed that, in parallel with glucose tolerance deterioration, the -1317 CpG at the PANDAR promoter became hypo-methylated in obesity, resulting in enhanced PANDAR induction by p53. PANDAR silencing in senescent APCs from individuals with obesity and T2D caused repression of senescence programs and cell cycle re-entry. PANDAR transcription in white blood cells (WBCs) mirrored that in APCs. Also, individuals with obesity exhibited rescue of PANDAR transcription in WBCs following bariatric surgery, accompanied by enhanced methylation at the regulatory PANDAR -1317 CpG. In conclusion, PANDAR dysregulation is a newly identified mechanism determining the early senescence of APCs from individuals with obesity, which worsens along the progression toward T2D. In the future, PANDAR targeting may represent a valuable strategy to delay this progression.
Collapse
Affiliation(s)
- Antonella Desiderio
- Department of Translational Medicine, Federico II University of Naples, Naples, Italy
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Monica Pastorino
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
- Department of Molecular Medicine and Biotechnology, Federico II University of Naples, Naples, Italy
| | - Michele Campitelli
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Immacolata Prevenzano
- Department of Translational Medicine, Federico II University of Naples, Naples, Italy
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | | | - Rosa Spinelli
- Department of Translational Medicine, Federico II University of Naples, Naples, Italy
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Luca Parrillo
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Michele Longo
- Department of Translational Medicine, Federico II University of Naples, Naples, Italy
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Marco Milone
- Department of Clinical Medicine and Surgery, Federico II University of Naples, Naples, Italy
| | - Claudia Miele
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Gregory Alexander Raciti
- Department of Translational Medicine, Federico II University of Naples, Naples, Italy
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Francesco Beguinot
- Department of Translational Medicine, Federico II University of Naples, Naples, Italy
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| |
Collapse
|
4
|
He Y, Huang X, Ma Y, Yang G, Cui Y, Lv X, Zhao R, Jin H, Tong Y, Zhang X, Li J, Peng M. A novel aging-associated lncRNA signature for predicting prognosis in osteosarcoma. Sci Rep 2024; 14:1386. [PMID: 38228673 PMCID: PMC10791644 DOI: 10.1038/s41598-024-51732-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 01/09/2024] [Indexed: 01/18/2024] Open
Abstract
Osteosarcoma (OS) is one of the most prevalent bone tumors in adolescents, and the correlation between aging and OS remains unclear. Currently, few accurate and reliable biomarkers have been determined for OS prognosis. To address this issue, we carried out a detailed bioinformatics analysis based on OS with data from the Cancer Genome Atlas data portal and Human Aging Genomic Resources database, as well as in vitro experiments. A total of 88 OS samples with gene expression profiles and corresponding clinical characteristics were obtained. Through univariate Cox regression analysis and survival analysis, 10 aging-associated survival lncRNAs (AASRs) were identified to be associated with the overall survival of OS patients. Based on the expression levels of the 10 AASRs, the OS patients were classified into two clusters (Cluster A and Cluster B). Cluster A had a worse prognosis, while Cluster B had a better prognosis. Then, 5 AASRs were ultimately included in the signature through least absolute shrinkage and selection operator-Cox regression analysis. Kaplan‒Meier survival analysis verified that the high-risk group exhibited a worse prognosis than the low-risk group. Furthermore, univariate and multivariate Cox regression analyses confirmed that the riskScore was an independent prognostic factor for OS patients. Subsequently, we discovered that the risk signature was correlated with the properties of the tumor microenvironment and immune cell infiltration. Specifically, there was a positive association between the risk model and naïve B cells, resting dendritic cells and gamma delta T cells, while it was negatively related to CD8+ T cells. Finally, in vitro experiments, we found that UNC5B-AS1 inhibited OS cells from undergoing cellular senescence and apoptosis, thereby promoting OS cells proliferation. In conclusion, we constructed and verified a 5 AASR-based signature, that exhibited excellent performance in evaluating the overall survival of OS patients. In addition, we found that UNC5B-AS1 might inhibit the senescence process, thus leading to the development and progression of OS. Our findings may provide novel insights into the treatment of OS patients.
Collapse
Affiliation(s)
- Yi He
- Department of Mini-Invasive Spinal Surgery, The Third People's Hospital of Henan Province, Zhengzhou, 450006, Henan, China
| | - Xiao Huang
- Department of Clinical Laboratory, Luohe Central Hospital, Luohe, 462300, Henan, China
| | - Yajie Ma
- Department of Medical Affair, The Third People's Hospital of Henan Province, Zhengzhou, 450006, Henan, China
| | - Guohui Yang
- Department of Emergency Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yuqing Cui
- General ICU, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Xuefeng Lv
- Department of Clinical Laboratory, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Rongling Zhao
- Department of Clinical Laboratory, The Third People's Hospital of Henan Province, Zhengzhou, 450006, Henan, China
| | - Huifang Jin
- Department of Blood Transfusion, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yalin Tong
- Department of Digestion, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Xinyu Zhang
- Department of Medical Affair, The Third People's Hospital of Henan Province, Zhengzhou, 450006, Henan, China
| | - Jitian Li
- Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital), Henan Provincial Orthopedic Institute, Henan University of Chinese Medicine, 100 Yongping Road, Zhengzhou, 450000, Henan, China.
| | - Mengle Peng
- Department of Clinical Laboratory, The Third People's Hospital of Henan Province, Zhengzhou, 450006, Henan, China.
| |
Collapse
|
5
|
Martinez-Castillo M, M. Elsayed A, López-Berestein G, Amero P, Rodríguez-Aguayo C. An Overview of the Immune Modulatory Properties of Long Non-Coding RNAs and Their Potential Use as Therapeutic Targets in Cancer. Noncoding RNA 2023; 9:70. [PMID: 37987366 PMCID: PMC10660772 DOI: 10.3390/ncrna9060070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 10/25/2023] [Accepted: 11/08/2023] [Indexed: 11/22/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) play pivotal roles in regulating immune responses, immune cell differentiation, activation, and inflammatory processes. In cancer, they are gaining prominence as potential therapeutic targets due to their ability to regulate immune checkpoint molecules and immune-related factors, suggesting avenues for bolstering anti-tumor immune responses. Here, we explore the mechanistic insights into lncRNA-mediated immune modulation, highlighting their impact on immunity. Additionally, we discuss their potential to enhance cancer immunotherapy, augmenting the effectiveness of immune checkpoint inhibitors and adoptive T cell therapies. LncRNAs as therapeutic targets hold the promise of revolutionizing cancer treatments, inspiring further research in this field with substantial clinical implications.
Collapse
Affiliation(s)
- Moises Martinez-Castillo
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA; (M.M.-C.); (G.L.-B.); (P.A.)
- Liver, Pancreas and Motility Laboratory, Unit of Research in Experimental Medicine, School of Medicine, Universidad Nacional Autónoma de México (UNAM), Mexico City 06726, Mexico
| | - Abdelrahman M. Elsayed
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Al-Azhar University, Cairo 11754, Egypt;
- Havener Eye Institute, Department of Ophthalmology and Visual Science, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Gabriel López-Berestein
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA; (M.M.-C.); (G.L.-B.); (P.A.)
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Paola Amero
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA; (M.M.-C.); (G.L.-B.); (P.A.)
| | - Cristian Rodríguez-Aguayo
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA; (M.M.-C.); (G.L.-B.); (P.A.)
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| |
Collapse
|
6
|
Andonegui-Elguera MA, Cáceres-Gutiérrez RE, Oliva-Rico D, Díaz-Chávez J, Herrera LA. LncRNAs-associated to genomic instability: A barrier to cancer therapy effectiveness. Front Genet 2022; 13:984329. [DOI: 10.3389/fgene.2022.984329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 10/31/2022] [Indexed: 11/22/2022] Open
Abstract
Although a large part of the genome is transcribed, only 1.9% has a protein-coding potential; most of the transcripts are non-coding RNAs such as snRNAs, tRNAs, and rRNAs that participate in mRNA processing and translation. In addition, there are small RNAs with a regulatory role, such as siRNAs, miRNAs, and piRNAs. Finally, the long non-coding RNAs (lncRNAs) are transcripts of more than 200 bp that can positively and negatively regulate gene expression (both in cis and trans), serve as a scaffold for protein recruitment, and control nuclear architecture, among other functions. An essential process regulated by lncRNAs is genome stability. LncRNAs regulate genes associated with DNA repair and chromosome segregation; they are also directly involved in the maintenance of telomeres and have recently been associated with the activity of the centromeres. In cancer, many alterations in lncRNAs have been found to promote genomic instability, which is a hallmark of cancer and is associated with resistance to chemotherapy. In this review, we analyze the most recent findings of lncRNA alterations in cancer, their relevance in genomic instability, and their impact on the resistance of tumor cells to anticancer therapy.
Collapse
|
7
|
Interplays between non-coding RNAs and chemokines in digestive system cancers. Biomed Pharmacother 2022; 152:113237. [PMID: 35716438 DOI: 10.1016/j.biopha.2022.113237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 06/01/2022] [Accepted: 06/02/2022] [Indexed: 11/24/2022] Open
Abstract
Within tumors, chemokines and their cognate receptors are expressed by infiltrated leukocytes, cancerous cells, and related cells of stroma, like tumor-associated fibroblasts and tumor-associated macrophages. In malignancies, the altered expression of chemokines/chemokine receptors governs leukocyte infiltration and activation, epithelial-mesenchymal transition (EMT), cancer cell proliferation, angiogenesis, and metastasis. Non-coding RNAs (ncRNAs) contribute to multiple physiological and pathophysiological processes. Some miRNAs can exert anti-tumorigenic activity in digestive system malignancies by repressing the expression of tumor-promoting chemokines/chemokine receptors or by upregulating tumor-suppressing chemokines/chemokine receptors. However, many miRNAs exert pro-tumorigenic activity by suppressing the expression of chemokines/chemokine receptors or by upregulating tumor-promoting chemokines/chemokine receptors. LncRNA and circRNAs also exert pro- and anti-tumorigenic effects by targeting downstream miRNAs influencing the expression of tumor-promoting and tumor-suppressor chemokines/chemokine receptors. On the other side, some chemokines influence the expression of ncRNAs affecting tumor formation. The current review explains the communications between ncRNAs and chemokines/chemokine receptors in certain digestive system malignancies, such as gastric, colorectal, and pancreatic cancers and hepatocellular carcinoma to gain better insights into their basic crosstalk as well as possible therapeutic modalities.
Collapse
|
8
|
Ghafouri-Fard S, Khoshbakht T, Hussen BM, Baniahmad A, Branicki W, Taheri M, Eghbali A. Emerging Role of Non-Coding RNAs in Senescence. Front Cell Dev Biol 2022; 10:869011. [PMID: 35865636 PMCID: PMC9294638 DOI: 10.3389/fcell.2022.869011] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
Senescence is defined as a gradual weakening of functional features of a living organism. Cellular senescence is a process that is principally aimed to remove undesirable cells by prompting tissue remodeling. This process is also regarded as a defense mechanism induced by cellular damage. In the course of oncogenesis, senescence can limit tumor progression. However, senescence participates in the pathoetiology of several disorders such as fibrotic disorders, vascular disorders, diabetes, renal disorders and sarcopenia. Recent studies have revealed contribution of different classes of non-coding RNAs in the cellular senescence. Long non-coding RNAs, microRNAs and circular RNAs are three classes of these transcripts whose contributions in this process have been more investigated. In the current review, we summarize the available literature on the impact of these transcripts in the cellular senescence.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Tayyebeh Khoshbakht
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bashdar Mahmud Hussen
- Department of Pharmacognosy, College of Pharmacy, Hawler Medical University, Erbil, Iraq
- Center of Research and Strategic Studies, Lebanese French University, Erbil, Iraq
| | - Aria Baniahmad
- Institute of Human Genetics, Jena University Hospitals, Jena, Germany
- *Correspondence: Aria Baniahmad, ; Mohammad Taheri, ; Ahmad Eghbali,
| | - Wojciech Branicki
- Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | - Mohammad Taheri
- Institute of Human Genetics, Jena University Hospitals, Jena, Germany
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- *Correspondence: Aria Baniahmad, ; Mohammad Taheri, ; Ahmad Eghbali,
| | - Ahmad Eghbali
- Anesthesiology Research Center, Mofid Children Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- *Correspondence: Aria Baniahmad, ; Mohammad Taheri, ; Ahmad Eghbali,
| |
Collapse
|
9
|
Predictive Potentials of ZEB1-AS1 in Colorectal Cancer Prognosis and Their Correlation with Immunotherapy. JOURNAL OF ONCOLOGY 2022; 2022:1084555. [PMID: 35794981 PMCID: PMC9252708 DOI: 10.1155/2022/1084555] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 05/27/2022] [Accepted: 05/30/2022] [Indexed: 11/29/2022]
Abstract
Background CRC is the third most common cancer globally. The tumor immune microenvironment is closely associated with the overexpressed lncRNA ZEB1-AS1. However, in individuals with CRC, the ZEB1-AS1 gene's ability to predict immune response is a mystery. Materials and Methods The ZEB1-AS1 gene's prognostic potential was thoroughly investigated. We analyzed and included into the TCGA database all ZEB1-AS1 and ZEB1-AS1-related genes using LASSO-Cox regression. Researchers examined the link among ZEB1-AS1 and the tumor immune microenvironment, immune checkpoint, and tumor mutation burden (TMB) in CRC through the TCGA database. Using a predictive model, researchers were able to determine the link between ZEB1-AS1 and NUDT3 and CRC prognosis. Result According to our findings, individuals with reduced ZEB1-AS1 expression had a better prognosis in CRC. Based on the expression of two genes in the TCGA database, patients were divided into two cohorts. The B lymphocytes and macrophages are less likely to be recruited by tissues with a low-risk score. TMB and immunological checkpoints were shown to have a connection. Based on these genes, a predictive nomogram was built and confirmed, with a C-index of 0.78. Conclusion Prognostic models based on ZEB1-AS1 and ZEB1-AS1-related genes are more accurate for CRC patients when it comes to the prognosis and immune checkpoint responsiveness.
Collapse
|
10
|
Eteleeb AM, Thunuguntla PK, Gelev KZ, Tang CY, Rozycki EB, Miller A, Lei JT, Jayasinghe RG, Dang HX, White NM, Reis-Filho JS, Mardis ER, Ellis MJ, Ding L, Silva-Fisher JM, Maher CA. LINC00355 regulates p27 KIP expression by binding to MENIN to induce proliferation in late-stage relapse breast cancer. NPJ Breast Cancer 2022; 8:49. [PMID: 35418131 PMCID: PMC9007952 DOI: 10.1038/s41523-022-00412-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 03/02/2022] [Indexed: 12/13/2022] Open
Abstract
Late-stage relapse (LSR) in patients with breast cancer (BC) occurs more than five years and up to 10 years after initial treatment and has less than 30% 5-year relative survival rate. Long non-coding RNAs (lncRNAs) play important roles in BC yet have not been studied in LSR BC. Here, we identify 1127 lncRNAs differentially expressed in LSR BC via transcriptome sequencing and analysis of 72 early-stage and 24 LSR BC patient tumors. Decreasing expression of the most up-regulated lncRNA, LINC00355, in BC and MCF7 long-term estrogen deprived cell lines decreases cellular invasion and proliferation. Subsequent mechanistic studies show that LINC00355 binds to MENIN and changes occupancy at the CDKN1B promoter to decrease p27Kip. In summary, this is a key study discovering lncRNAs in LSR BC and LINC00355 association with epigenetic regulation and proliferation in BC.
Collapse
Affiliation(s)
- Abdallah M Eteleeb
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Prasanth K Thunuguntla
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Kyla Z Gelev
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Emily B Rozycki
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Alexander Miller
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Reyka G Jayasinghe
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
- The McDonnell Genome Institute, St. Louis, MO, USA
| | - Ha X Dang
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
- The McDonnell Genome Institute, St. Louis, MO, USA
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Nicole M White
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Elaine R Mardis
- Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, USA
| | | | - Li Ding
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
- The McDonnell Genome Institute, St. Louis, MO, USA
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Jessica M Silva-Fisher
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA.
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA.
| | - Christopher A Maher
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA.
- The McDonnell Genome Institute, St. Louis, MO, USA.
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Biomedical Engineering, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
11
|
Heydarnezhad Asl M, Pasban Khelejani F, Bahojb Mahdavi SZ, Emrahi L, Jebelli A, Mokhtarzadeh A. The various regulatory functions of long noncoding RNAs in apoptosis, cell cycle, and cellular senescence. J Cell Biochem 2022; 123:995-1024. [PMID: 35106829 DOI: 10.1002/jcb.30221] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 12/28/2021] [Accepted: 01/11/2022] [Indexed: 12/12/2022]
Abstract
Long noncoding RNAs (lncRNAs) are a group of noncoding cellular RNAs involved in significant biological phenomena such as differentiation, cell development, genomic imprinting, adjusting the enzymatic activity, regulating chromosome conformation, apoptosis, cell cycle, and cellular senescence. The misregulation of lncRNAs interrupting normal biological processes has been implicated in tumor formation and metastasis, resulting in cancer. Apoptosis and cell cycle, two main biological phenomena, are highly conserved and intimately coupled mechanisms. Hence, some cell cycle regulators can influence both programmed cell death and cell division. Apoptosis eliminates defective and unwanted cells, and the cell cycle enables cells to replicate themselves. The improper regulation of apoptosis and cell cycle contributes to numerous disorders such as neurodegenerative and autoimmune diseases, viral infection, anemia, and mainly cancer. Cellular senescence is a tumor-suppressing response initiated by environmental and internal stress factors. This phenomenon has recently attained more attention due to its therapeutic implications in the field of senotherapy. In this review, the regulatory roles of lncRNAs on apoptosis, cell cycle, and senescence will be discussed. First, the role of lncRNAs in mitochondrial dynamics and apoptosis is addressed. Next, the interaction between lncRNAs and caspases, pro/antiapoptotic proteins, and also EGFR/PI3K/PTEN/AKT/mTORC1 signaling pathway will be investigated. Furthermore, the effect of lncRNAs in the cell cycle is surveyed through interaction with cyclins, cdks, p21, and wnt/β-catenin/c-myc pathway. Finally, the function of essential lncRNAs in cellular senescence is mentioned.
Collapse
Affiliation(s)
| | - Faezeh Pasban Khelejani
- Department of Cell and Molecular Biology, Faculty of Basic Sciences, University of Maragheh, Maragheh, Iran
| | | | - Leila Emrahi
- Department of Medical Genetics, Faculty of Medical Science, Tarbiat Modares University, Tehran, Iran
| | - Asiyeh Jebelli
- Department of Biological Science, Faculty of Basic Science, Higher Education Institute of Rab-Rashid, Tabriz, Iran.,Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
12
|
Hosseini MS, Samaei NM, Ghaderian SMH, Dastmalchi R, Rajabi S. The oncogenic role of both lncRNA PANDA and BCL2 gene in glioblastoma. GENE REPORTS 2021. [DOI: 10.1016/j.genrep.2021.101160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
13
|
Ghafouri-Fard S, Gholipour M, Hussen BM, Taheri M. The Impact of Long Non-Coding RNAs in the Pathogenesis of Hepatocellular Carcinoma. Front Oncol 2021; 11:649107. [PMID: 33968749 PMCID: PMC8097102 DOI: 10.3389/fonc.2021.649107] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 03/22/2021] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is among the utmost deadly human malignancies. This type of cancer has been associated with several environmental, viral, and lifestyle risk factors. Among the epigenetic factors which contribute in the pathogenesis of HCC is dysregulation of long non-coding RNAs (lncRNAs). These transcripts modulate expression of several tumor suppressor genes and oncogenes and alter the activity of cancer-related signaling axes. Several lncRNAs such as NEAT1, MALAT1, ANRIL, and SNHG1 have been up-regulated in HCC samples. On the other hand, a number of so-called tumor suppressor lncRNAs namely CASS2 and MEG3 are down-regulated in HCC. The interaction between lncRNAs and miRNAs regulate expression of a number of mRNA coding genes which are involved in the pathogenesis of HCC. H19/miR-15b/CDC42, H19/miR-326/TWIST1, NEAT1/miR-485/STAT3, MALAT1/miR-124-3p/Slug, MALAT1/miR-195/EGFR, MALAT1/miR-22/SNAI1, and ANRIL/miR-144/PBX3 axes are among functional axes in the pathobiology of HCC. Some genetic polymorphisms within non-coding regions of the genome have been associated with risk of HCC in certain populations. In the current paper, we describe the recent finding about the impact of lncRNAs in HCC.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdi Gholipour
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bashdar Mahmud Hussen
- Pharmacognosy Department, College of Pharmacy, Hawler Medical University, Erbil, Iraq
| | - Mohammad Taheri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
LINC00978 promotes hepatocellular carcinoma carcinogenesis partly via activating the MAPK/ERK pathway. Biosci Rep 2021; 40:222177. [PMID: 32077915 PMCID: PMC7064789 DOI: 10.1042/bsr20192790] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 11/20/2019] [Accepted: 12/23/2019] [Indexed: 02/07/2023] Open
Abstract
Objective: To study the role of long non-coding RNA (lncRNA) LINC00978 in hepatocellular carcinoma (HCC) carcinogenesis. Materials and methods: LINC00978 expression level was measured by reverse transcription quantitative real-time PCR (RT-qPCR) in HCC tissues and adjacent healthy liver tissues from 49 HCC patients. MTT assay, colony forming assay, and flow cytometry were performed to evaluate the effects of shRNA-mediated LINC00978 knockdown on HCC cell proliferation, cell cycle progression, and apoptosis in vitro. Xenograft tumor model was performed to determine the effects of LINC00978 knockdown on HCC tumor growth in vivo. Western blot was used to assess the activation of signaling molecules in the apoptosis and mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) pathway. Results: LINC00978 expression was significantly up-regulated in human HCC tissue relative to adjacent normal tissue, and LINC00978 high expression was correlated with poor HCC overall survival. LINC00978 was up-regulated in HCC cell lines. ShRNA-mediated LINC00978 knockdown significantly decreased HCC cell proliferation, and induced HCC cell cycle arrest and apoptosis in vitro. LINC00978 knockdown led to significant decrease in tumor xenograft size in vivo. Western blots revealed LINC00978 inhibition decreased ERK, p38, and c-Jun N-terminal kinase (JNK) phosphorylation in HCC cells. Conclusions: LINC00978 is highly expressed in human HCC tissue and correlates with poor HCC prognosis. LINC00978 promotes HCC cell proliferation, cell cycle progression, and survival, partially by activating the MAPK/ERK pathway. Our findings partially elucidated the roles of LINC00978 in HCC carcinogenesis, and identified a therapeutic target for HCC.
Collapse
|
15
|
Miao T, Si Q, Wei Y, Fan R, Wang J, An X. Identification and validation of seven prognostic long non-coding RNAs in oral squamous cell carcinoma. Oncol Lett 2020; 20:939-946. [PMID: 32566023 DOI: 10.3892/ol.2020.11603] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 11/12/2019] [Indexed: 12/19/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is one of the most common malignancies worldwide, due to poor diagnosis and treatment. There is increasing evidence that demonstrates the involvement of long non-coding RNAs (lncRNAs) in carcinogenesis and cancer progression. Therefore, the aim of the present study was to explore potential lncRNA-associated features of patients with OSCC as a valuable and independent prognostic biomarker. A total of 268 lncRNA expression profiles and clinical patient information on OSCC were downloaded from The Cancer Genome Atlas database. The clinical information was exploited for prescreening, using Cox regression analysis, and differentially expressed lncRNAs (DElncRNAs) were identified using edgeR software. Using the 'caret' package, the datasets were categorized into test datasets and training datasets, respectively. Through bioinformatics, seven prognostic DElncRNAs were selected. Using the regression coefficients, a risk score based on the seven-DElncRNA signature was developed to assess the prognostic function of key DElncRNAs. According to the median risk score, patients were classified into high-risk and low-risk groups in the training and test datasets. Additionally, receiver operating characteristic (ROC) curve analysis was conducted to evaluate the sensitivity and specificity of the prognostic DElncRNAs, and the optimal cut-off point was obtained from ROC analysis. Based on the optimal cut-off point, the patients were also categorized into high-risk and low-risk groups. Notably, the optimal cut-off point was more sensitive than the median risk score, particularly in the test dataset. The Kaplan-Meier survival and log rank test analysis results indicated that the P-value, based on the optimal cut-off, was less than the median risk cut-off. Additionally, stratified analysis results revealed that the seven-DElncRNAs signature was also independent of OSCC age. Furthermore, the findings of the present study suggested that the seven-DElncRNA signature can be used as a potential prognostic indicator and may have important clinical significance in OSCC.
Collapse
Affiliation(s)
- Tingting Miao
- School of Stomatology, Lanzhou University, Gansu, Lanzhou 730000, P.R. China
| | - Qingzong Si
- School of Stomatology, Lanzhou University, Gansu, Lanzhou 730000, P.R. China
| | - Yuan Wei
- School of Stomatology, Lanzhou University, Gansu, Lanzhou 730000, P.R. China
| | - Ruihong Fan
- School of Stomatology, Lanzhou University, Gansu, Lanzhou 730000, P.R. China
| | - Junjie Wang
- School of Stomatology, Lanzhou University, Gansu, Lanzhou 730000, P.R. China
| | - Xiaoli An
- School of Stomatology, Lanzhou University, Gansu, Lanzhou 730000, P.R. China
| |
Collapse
|
16
|
Jin KT, Yao JY, Fang XL, Di H, Ma YY. Roles of lncRNAs in cancer: Focusing on angiogenesis. Life Sci 2020; 252:117647. [PMID: 32275935 DOI: 10.1016/j.lfs.2020.117647] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/31/2020] [Accepted: 04/05/2020] [Indexed: 02/07/2023]
Abstract
Approximately 98% of the human genome consists of non-coding sequences that are classified into two classes by size: small non-coding RNAs (≤200 nucleotides) and long non-coding RNAs (≥200 nucleotides). Long non-coding RNAs (lncRNAs) are involved in various cellular events and act as guides, signals, decoys, and dynamic scaffolds. Due to their oncogenic and tumor suppressive roles, lncRNAs are important in cancer development and growth. LncRNAs play their roles by modulating cancer hallmarks, including DNA damage, metastasis, immune escape, cell stemness, drug resistance, metabolic reprogramming, and angiogenesis. Angiogenesis is vital for solid tumors which guarantees their growth beyond 2 mm3. Tumor angiogenesis is a complex process and is regulated through interaction between pro-angiogenic and anti-angiogenic factors within the tumor microenvironment. There are accumulating evidence that different lncRNAs regulate tumor angiogenesis. In this paper, we described the functions and mechanisms of lncRNAs in tumor angiogenesis.
Collapse
Affiliation(s)
- Ke-Tao Jin
- Department of Colorectal Surgery, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing 312000, Zhejiang Province, PR China
| | - Jia-Yu Yao
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou 310014, Zhejiang Province, PR China; Clinical Research Institute, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou 310014, Zhejiang Province, PR China
| | - Xing-Liang Fang
- Acupuncture and Tuina Clinic, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou 310014, Zhejiang Province, PR China
| | - Hua Di
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou 310014, Zhejiang Province, PR China; Department of Hepatobiliary Surgery, Affiliated Hospital of Shaoxing University (Shaoxing Municipal Hospital), Shaoxing 312000, Zhejiang Province, PR China.
| | - Ying-Yu Ma
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou 310014, Zhejiang Province, PR China; Clinical Research Institute, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou 310014, Zhejiang Province, PR China.
| |
Collapse
|
17
|
Guo C, Li J, Steinauer N, Wong M, Wu B, Dickson A, Kalkum M, Zhang J. Histone deacetylase 3 preferentially binds and collaborates with the transcription factor RUNX1 to repress AML1-ETO-dependent transcription in t(8;21) AML. J Biol Chem 2020; 295:4212-4223. [PMID: 32071087 PMCID: PMC7105303 DOI: 10.1074/jbc.ra119.010707] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 02/11/2020] [Indexed: 01/26/2023] Open
Abstract
In up to 15% of acute myeloid leukemias (AMLs), a recurring chromosomal translocation, termed t(8;21), generates the AML1-eight-twenty-one (ETO) leukemia fusion protein, which contains the DNA-binding domain of Runt-related transcription factor 1 (RUNX1) and almost all of ETO. RUNX1 and the AML1-ETO fusion protein are coexpressed in t(8;21) AML cells and antagonize each other's gene-regulatory functions. AML1-ETO represses transcription of RUNX1 target genes by competitively displacing RUNX1 and recruiting corepressors such as histone deacetylase 3 (HDAC3). Recent studies have shown that AML1-ETO and RUNX1 co-occupy the binding sites of AML1-ETO-activated genes. How this joined binding allows RUNX1 to antagonize AML1-ETO-mediated transcriptional activation is unclear. Here we show that RUNX1 functions as a bona fide repressor of transcription activated by AML1-ETO. Mechanistically, we show that RUNX1 is a component of the HDAC3 corepressor complex and that HDAC3 preferentially binds to RUNX1 rather than to AML1-ETO in t(8;21) AML cells. Studying the regulation of interleukin-8 (IL8), a newly identified AML1-ETO-activated gene, we demonstrate that RUNX1 and HDAC3 collaboratively repress AML1-ETO-dependent transcription, a finding further supported by results of genome-wide analyses of AML1-ETO-activated genes. These and other results from the genome-wide studies also have important implications for the mechanistic understanding of gene-specific coactivator and corepressor functions across the AML1-ETO/RUNX1 cistrome.
Collapse
MESH Headings
- Cell Line, Tumor
- Core Binding Factor Alpha 2 Subunit/genetics
- Gene Expression Regulation, Neoplastic
- Genome, Human/genetics
- Histone Deacetylases/genetics
- Humans
- Interleukin-8/genetics
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Oncogene Proteins, Fusion/genetics
- Promoter Regions, Genetic
- RUNX1 Translocation Partner 1 Protein/genetics
- Transcriptional Activation/genetics
- Translocation, Genetic/genetics
Collapse
Affiliation(s)
- Chun Guo
- Department of Pharmacology and Physiology, Saint Louis University, School of Medicine, St. Louis, Missouri 63104
| | - Jian Li
- Department of Pharmacology and Physiology, Saint Louis University, School of Medicine, St. Louis, Missouri 63104
| | - Nickolas Steinauer
- Department of Pharmacology and Physiology, Saint Louis University, School of Medicine, St. Louis, Missouri 63104
| | - Madeline Wong
- Department of Pharmacology and Physiology, Saint Louis University, School of Medicine, St. Louis, Missouri 63104
| | - Brent Wu
- Department of Pharmacology and Physiology, Saint Louis University, School of Medicine, St. Louis, Missouri 63104
| | - Alexandria Dickson
- Department of Pharmacology and Physiology, Saint Louis University, School of Medicine, St. Louis, Missouri 63104
| | - Markus Kalkum
- Department of Molecular Imaging and Therapy, Beckman Research Institute of the City of Hope, Duarte, California 91010
| | - Jinsong Zhang
- Department of Pharmacology and Physiology, Saint Louis University, School of Medicine, St. Louis, Missouri 63104.
| |
Collapse
|
18
|
Abstract
Despite advanced clinical treatments, mortality in patients with metastatic colorectal cancer (CRC) remains high. Three critical determinants in CRC progression include the epithelial proliferation checkpoints, epithelial-to-mesenchymal transition (EMT) and inflammatory cytokines in the tumour microenvironment. Genes involved in these three processes are regulated at the transcriptional and post-transcriptional level. Recent studies revealed previously unappreciated roles of non-coding ribonucleic acids (ncRNAs) in modulating the proliferation checkpoints, EMT, and inflammatory gene expression in CRC. In this review, we will discuss the mechanisms underlying the roles of ncRNAs in CRC as well as examine future perspectives in this field. Better understanding of ncRNA biology will provide novel targets for future therapeutic development.
Collapse
Affiliation(s)
- Shengyun Ma
- Cellular and Molecular Medicine, University of California , San Diego, USA
| | - Tianyun Long
- Cellular and Molecular Medicine, University of California , San Diego, USA
| | | |
Collapse
|
19
|
lncRNA-Triggered Macrophage Inflammaging Deteriorates Age-Related Diseases. Mediators Inflamm 2019; 2019:4260309. [PMID: 31949425 PMCID: PMC6942909 DOI: 10.1155/2019/4260309] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 11/10/2019] [Accepted: 11/13/2019] [Indexed: 02/05/2023] Open
Abstract
Aging and age-related diseases (ARDs) share basic mechanisms largely involving inflammation. A chronic, low-grade, subclinical inflammation called inflammaging occurs during aging. Autophagy defects, oxidative stresses, senescence-associated secretory phenotypes (SASPs), and DNA damage generally contribute to inflammaging and are largely regulated by numerous lncRNA through two-level vicious cycles disrupting cellular homeostasis: (1) inflammaging and the cellular senescence cascade and (2) autophagy defects, oxidative stress, and the SASP cascade. SASPs and inflammasomes simultaneously cause inflammaging. This review discusses the involvement of macrophage inflammaging in various ARDs and its regulation via lncRNA. Among macrophages, this phenomenon potentially impairs its immunosurveillance and phagocytosis mechanisms, leading to decreased recognition and clearance of malignant and senescent cells. Moreover, SASPs extracellularly manifest to induce paracrine senescence. Macrophage senescence escalates to organ level malfunction, and the organism is more prone to ARDs. By targeting genes and proteins or functioning as competing endogenous RNA (ceRNA), lncRNA regulates different phenomena including inflammaging and ARDs. The detailed mechanism warrants further elucidation to obtain pathological evidence of ARDs and potential treatment approaches.
Collapse
|
20
|
Wang Y, Yang F, Yang Q. The regulatory roles and potential prognosis implications of long non-coding RNAs in gastric cancer. Histol Histopathol 2019; 35:433-442. [PMID: 31793657 DOI: 10.14670/hh-18-188] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Accumulating dysregulated lncRNAs have been demonstrated to execute vital functions in the pathogenesis and progress of gastric cancer (GC) through versatile molecular mechanisms. In this review, we classify the mechanisms of dysregulated lncRNAs in GC into several governing types according to their roles at molecular level. For each regulatory role, we illustrate several instructive examples and introduce significant effects of lncRNAs on cellular biological properties of GC. Besides, we summarize a group of lncRNA-signatures that are potential biomarkers in the prediction of prognosis for GC patients.
Collapse
Affiliation(s)
- Yue Wang
- Department of Pathogenobiology, College of Basic Medical Sciences, Jilin University, Changchun City, Jilin Province, China
| | - Fan Yang
- Department of Pathogenobiology, College of Basic Medical Sciences, Jilin University, Changchun City, Jilin Province, China
| | - Qing Yang
- Department of Pathogenobiology, College of Basic Medical Sciences, Jilin University, Changchun City, Jilin Province, China.
| |
Collapse
|
21
|
LncRNAs Regulatory Networks in Cellular Senescence. Int J Mol Sci 2019; 20:ijms20112615. [PMID: 31141943 PMCID: PMC6600251 DOI: 10.3390/ijms20112615] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/19/2019] [Accepted: 05/06/2019] [Indexed: 02/07/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) are a class of transcripts longer than 200 nucleotides with no open reading frame. They play a key role in the regulation of cellular processes such as genome integrity, chromatin organization, gene expression, translation regulation, and signal transduction. Recent studies indicated that lncRNAs are not only dysregulated in different types of diseases but also function as direct effectors or mediators for many pathological symptoms. This review focuses on the current findings of the lncRNAs and their dysregulated signaling pathways in senescence. Different functional mechanisms of lncRNAs and their downstream signaling pathways are integrated to provide a bird’s-eye view of lncRNA networks in senescence. This review not only highlights the role of lncRNAs in cell fate decision but also discusses how several feedback loops are interconnected to execute persistent senescence response. Finally, the significance of lncRNAs in senescence-associated diseases and their therapeutic and diagnostic potentials are highlighted.
Collapse
|
22
|
Sayad A, Taheri M, Omrani MD, Fallah H, Kholghi Oskooei V, Ghafouri-Fard S. Peripheral expression of long non-coding RNAs in bipolar patients. J Affect Disord 2019; 249:169-174. [PMID: 30772744 DOI: 10.1016/j.jad.2019.02.034] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 01/13/2019] [Accepted: 02/11/2019] [Indexed: 10/27/2022]
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) have established roles in the pathogenesis of diverse human disorders including neuropsychiatric disorders. METHODS In the current study, we evaluated expression levels of six apoptosis-related lncRNAs (CCAT2, TUG1, PANDA, NEAT1, FAS-AS1 and OIP5-AS1) in the peripheral blood of bipolar disorder (BD) patients and healthy subjects to assess their contribution in the pathogenesis of BD. RESULTS CCAT2, TUG1 and PANDA were up-regulated in total BD patients compared with total healthy subjects (P values = 0.006, <0.001 and 0.004 respectively) while OIP5-AS1 was down-regulated (P = 0.001). When expression levels of these genes were compared between patients and sex-matched healthy subjects, CCAT2 and TUG1 expression levels were only different in male subgroups; while PANDA expression was different in both male and female subgroups compared with the corresponding control subgroups. Transcript levels of lncRNAs were not correlated with any of demographic or clinical parameters of BD patients or controls after adjustment for gender. Pairwise correlations between expression levels of lncRNAs followed a disease-dependent manner. Based on receiver operating characteristic curves, among the assessed lncRNAs TUG1 had the highest diagnostic power in BD. Combination of transcript levels of CCAT2, TUG1, PANDA and OIP5-AS1 improved both sensitivity and specificity resulting in diagnostic power of 0.96. CONCLUSION Our data demonstrated a possible role of certain lncRNAs in the pathogenesis of BD and potentiated them as diagnostic markers in this disorder.
Collapse
Affiliation(s)
- Arezou Sayad
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taheri
- Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mir Davood Omrani
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Fallah
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Vahid Kholghi Oskooei
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
23
|
Lin YH, Wu MH, Yeh CT, Lin KH. Long Non-Coding RNAs as Mediators of Tumor Microenvironment and Liver Cancer Cell Communication. Int J Mol Sci 2018; 19:ijms19123742. [PMID: 30477236 PMCID: PMC6321423 DOI: 10.3390/ijms19123742] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 11/21/2018] [Accepted: 11/21/2018] [Indexed: 02/06/2023] Open
Abstract
The tumor microenvironment is an important concept that defines cancer development not only through tumor cells themselves but also the surrounding cellular and non-cellular components, including stromal cells, blood vessels, infiltrating inflammatory cells, cancer stem cells (CSC), cytokines, and growth factors, which act in concert to promote tumor cell survival and metastasis. Hepatocellular carcinoma (HCC) is one of the most common and aggressive human malignancies worldwide. Poor prognosis is largely attributable to the high rate of tumor metastasis, highlighting the importance of identifying patients at risk in advance and developing novel therapeutic targets to facilitate effective intervention. Long non-coding RNAs (lncRNA) are a class of non-protein coding transcripts longer than 200 nucleotides frequently dysregulated in various cancer types, which have multiple functions in widespread biological processes, including proliferation, apoptosis, metastasis, and metabolism. lncRNAs are involved in regulation of the tumor microenvironment and reciprocal signaling between cancer cells. Targeting of components of the tumor microenvironment or cancer cells has become a considerable focus of therapeutic research and establishing the effects of different lncRNAs on this network should aid in the development of effective treatment strategies. The current review provides a summary of the essential properties and functional roles of known lncRNAs associated with the tumor microenvironment in HCC.
Collapse
MESH Headings
- Antineoplastic Agents/therapeutic use
- Biomarkers, Tumor/antagonists & inhibitors
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Cytokines/genetics
- Cytokines/metabolism
- Epigenesis, Genetic
- Gene Expression Regulation, Neoplastic
- Hepatocytes/drug effects
- Hepatocytes/metabolism
- Hepatocytes/pathology
- Humans
- Liver Neoplasms/drug therapy
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Macrophages/drug effects
- Macrophages/metabolism
- Macrophages/pathology
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- RNA, Long Noncoding/antagonists & inhibitors
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Signal Transduction
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/genetics
Collapse
Affiliation(s)
- Yang-Hsiang Lin
- Department of Biochemistry, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan.
- Liver Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan.
| | - Meng-Han Wu
- Department of Biochemistry, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan.
| | - Chau-Ting Yeh
- Liver Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan.
| | - Kwang-Huei Lin
- Department of Biochemistry, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan.
- Liver Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan.
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan.
| |
Collapse
|
24
|
Wang H, Ke J, Guo Q, Barnabo Nampoukime KP, Yang P, Ma K. Long non-coding RNA CRNDE promotes the proliferation, migration and invasion of hepatocellular carcinoma cells through miR-217/MAPK1 axis. J Cell Mol Med 2018; 22:5862-5876. [PMID: 30246921 PMCID: PMC6237590 DOI: 10.1111/jcmm.13856] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Accepted: 07/24/2018] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is an invasive malignant tumour and the second major cause of cancer‐related deaths over the world. CRNDE and miR‐217 are non‐coding RNAs which play critical roles in cell growth, proliferation, migration. Mitogen‐activated protein kinase 1 (MAPK1) also participates in cancer cell process. Hence, this study aimed at investigating the effect of CRNDE on migration and invasion of HCC and figuring out the role of miR‐217 and MAPK1 in this process. The overexpression of CRNDE was demonstrated by a microarray‐based lncRNA profiling study. CRNDE expression in HCC was verified by qRT‐PCR. MTT assay and BrdU staining were applied to detect cell proliferation level. Transwell assay was utilized to examine cell migration and invasiveness abilities. Wound healing assay was performed for further exploration of cell migration capacity. MiR‐217 was predicted by bioinformatics. The dual luciferase reporter assay was performed to corroborate the targeting relationship between CRNDE, miR‐217 and MAPK1. MAPK1, the downstream target of miR‐217, was predicted using bioinformatics and was further confirmed by qRT‐PCR and Western blot. The interaction between CRNDE, miR‐217 and MAPK1 was studied by qRT‐PCR, Western blot, MTT, BrdU, transwell assay and wound healing assay. CRNDE was up‐regulated in HCC tissues and HCC cell lines. The high expression of CRNDE facilitated cell proliferation, migration and invasion, while the inhibited one affected on the contrary. MiR‐217, negatively correlated with CRNDE expression, was the target of CRNDE and was more lowly expressed in HCC. With the high expression of miR‐217, HCC cell proliferation, migration and invasion were suppressed. MAPK1, the possible target of miR‐217, was negatively correlated with miR‐217 but positively correlated with CRNDE and had the same effect in HCC formation process as CRNDE. Long non‐coding RNA CRNDE promotes the proliferation, migration and invasion of HCC cells through miR‐217/MAPK1 axis.
Collapse
Affiliation(s)
- Haihao Wang
- Division of Cardiothoracic and vascular surgery, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ji Ke
- Department of Forensic Science and Criminal Intelligence, Hubei University of Police, Wuhan, Hubei, China
| | - Qiannan Guo
- Division of Cardiothoracic and vascular surgery, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Kan-Paatib Barnabo Nampoukime
- Division of Cardiothoracic and vascular surgery, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Peiwen Yang
- Reproductive Medicine Center, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ke Ma
- Division of Infectious Disease, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
25
|
Mehrad‐Majd H, Akhtari J, Haerian M, Ravanshad Y. Clinicopathological and prognostic value of lncRNA PANDAR expression in solid tumors: Evidence from a systematic review and meta‐analysis. J Cell Physiol 2018; 234:4206-4216. [DOI: 10.1002/jcp.27179] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 07/17/2018] [Indexed: 12/25/2022]
Affiliation(s)
- Hassan Mehrad‐Majd
- Surgical Oncology Research Center, Mashhad University of Medical Sciences Mashhad Iran
- Clinical Research Unit, Mashhad University of Medical Sciences Mashhad Iran
| | - Javad Akhtari
- Immunogenetics Research Center, Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences Sari Iran
| | - Monir‐Sadat Haerian
- Faculty of Medicine, Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Yalda Ravanshad
- Department of Community Medicine Mashhad Branch, Islamic Azad University Mashhad Iran
| |
Collapse
|
26
|
Balas MM, Johnson AM. Exploring the mechanisms behind long noncoding RNAs and cancer. Noncoding RNA Res 2018; 3:108-117. [PMID: 30175284 PMCID: PMC6114262 DOI: 10.1016/j.ncrna.2018.03.001] [Citation(s) in RCA: 209] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 03/23/2018] [Indexed: 02/06/2023] Open
Abstract
Over the past decade, long noncoding RNAs (lncRNAs) have been identified as significant players in gene regulation. They are often differentially expressed and widely-associated with a majority of cancer types. The aberrant expression of these transcripts has been linked to tumorigenesis, metastasis, cancer stage progression and patient survival. Despite their apparent link to cancer, it has been challenging to gain a mechanistic understanding of how they contribute to cancer, partially due the difficulty in discriminating functional RNAs from other noncoding transcription events. However, there are several well-studied lncRNAs where specific mechanisms have been more clearly defined, leading to new discoveries into how these RNAs function. One major observation that has come to light is the context-dependence of lncRNA mechanisms, where they often have unique function in specific cell types and environment. Here, we review the molecular mechanisms of lncRNAs with a focus on cancer pathways, illustrating a few informative examples. Together, this type of detailed insight will lead to a greater understanding of the potential for the application of lncRNAs as targets of cancer therapies and diagnostics.
Collapse
Affiliation(s)
- Maggie M. Balas
- Molecular Biology Program, University of Colorado Denver Anschutz Medical Campus 12801 East 17th Ave., Aurora, CO, United States
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver Anschutz Medical Campus 12801 East 17th Ave., Aurora, CO, United States
- RNA Bioscience Initiative, University of Colorado Denver Anschutz Medical Campus 12801 East 17th Ave., Aurora, CO, United States
| | - Aaron M. Johnson
- Molecular Biology Program, University of Colorado Denver Anschutz Medical Campus 12801 East 17th Ave., Aurora, CO, United States
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver Anschutz Medical Campus 12801 East 17th Ave., Aurora, CO, United States
- RNA Bioscience Initiative, University of Colorado Denver Anschutz Medical Campus 12801 East 17th Ave., Aurora, CO, United States
| |
Collapse
|
27
|
Guo M. Cellular senescence and liver disease: Mechanisms and therapeutic strategies. Biomed Pharmacother 2017; 96:1527-1537. [PMID: 29174037 DOI: 10.1016/j.biopha.2017.11.075] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 11/13/2017] [Accepted: 11/13/2017] [Indexed: 12/12/2022] Open
Abstract
Cellular senescence is a fundamental cell fate caused by several cellular injuries which results in irreversible cell cycle arrest yet remaining metabolically active across all species. Cellular senescence not only can prevent tumor occurrence by inhibiting the proliferation of injured cells, but also can affect the surrounding cells through the senescence-associated secretory phenotype (SASP). Attractively, accumulating evidence shows that cellular senescence is closely related to various liver diseases. Therapeutic opportunities based on targeting senescent cells and the SASP are considered to be potential strategy for liver diseases. However, although research on cell senescence has attracted widespread attention, the overview on detailed mechanism and biological function of cell senescence in liver disease is still largely unknown. The present review summarizes the specific role of cell senescence in various liver diseases, and updates the molecular mechanisms underlying cell senescence. Moreover, the review also explores new strategies for prevention and treatment of liver disease through promoting senescence or counteracting excessive pathological senescence.
Collapse
Affiliation(s)
- Mei Guo
- Department of Pathogenic Biology and Immunology of Medical School, Southeast University, Nanjing, Jiangsu, 210009, China.
| |
Collapse
|