1
|
Xu Y, Wang T, Wan J, Ma D, Zhang H, Cheng D, Yang J, Wang M. Long non-coding RNA NEAT1 promotes multiple myeloma malignant transformation via targeting miR-485-5p/ABCB8. Hematology 2024; 29:2422153. [PMID: 39475764 DOI: 10.1080/16078454.2024.2422153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 10/22/2024] [Indexed: 11/07/2024] Open
Abstract
Multiple myeloma (MM) is the second most common hematological cancer all over the world. Long non-coding RNA (lncRNA) nuclear-enriched autosomal transcript-1 (NEAT1) have been reported to play important roles in the development and progression of multiple human malignancies like MM. However, the functional role and molecular mechanism of NEAT1 in MM progression still needs more support to identify potential targets of MM. In the present study, we focused on the clinical and biological significance of NEAT1 in MM. We demonstrated that NEAT1 was up-regulated in MM tissues and cell line. NEAT1 silencing significantly inhibited cell proliferation and promoted cell apoptosis in vitro. And we illustrated that miR-485-5p was a direct target of NEAT1 and the effect of down-regulated NEAT1 on MM cells was partially reversed by the miR-485-5p antisense oligonucleotide (ASO-miR-485-5p). Further investigation revealed that ABCB8 directly interacted with miR-485-5p. Similarly, in vivo experiments confirmed that down-regulated NEAT1 inhibited tumor growth and ABCB8 expression. Taken together, our results demonstrate for the first time that NEAT1/miR-485-5p/ABCB8 axis may be a key pathway for the development and progression of MM, and they may provide a novel avenue for targeted therapy.
Collapse
Affiliation(s)
- Yuxiu Xu
- Department of Hematology-oncology, The First Affiliated Hospital of Henan University of CM, Zhengzhou, Henan, People's Republic of China
| | - Tao Wang
- Department of Hematology-oncology, The First Affiliated Hospital of Henan University of CM, Zhengzhou, Henan, People's Republic of China
| | - Jiangwei Wan
- Department of Hematology-oncology, The First Affiliated Hospital of Henan University of CM, Zhengzhou, Henan, People's Republic of China
| | - Dongsheng Ma
- Department of Hematology-oncology, The First Affiliated Hospital of Henan University of CM, Zhengzhou, Henan, People's Republic of China
| | - Hongyang Zhang
- Zhumadian Central Hospital, Zhumadian, Henan, People's Republic of China
| | - Dongru Cheng
- Zhumadian Central Hospital, Zhumadian, Henan, People's Republic of China
| | - Jing Yang
- Zhumadian Central Hospital, Zhumadian, Henan, People's Republic of China
| | - Meng Wang
- Zhumadian Central Hospital, Zhumadian, Henan, People's Republic of China
| |
Collapse
|
2
|
Mishra R, Thunuguntla P, Perkin A, Duraiyan D, Bagwill K, Gonzales S, Brizuela V, Daly S, Chang YJ, Abebe M, Rajana Y, Wichmann K, Bolick C, King J, Fiala M, Fortier J, Jayasinghe R, Schroeder M, Ding L, Vij R, Silva-Fisher J. LINC01432 binds to CELF2 in newly diagnosed multiple myeloma promoting short progression-free survival to standard therapy. RESEARCH SQUARE 2024:rs.3.rs-4888379. [PMID: 39483883 PMCID: PMC11527149 DOI: 10.21203/rs.3.rs-4888379/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Multiple Myeloma (MM) is an incurable form of cancer that arises from malignant plasma cells, with over 35,000 new cases diagnosed annually in the United States. While there are a growing number of approved therapies, MM remains incurable and nearly all patients will relapse and exhaust available treatments. Mechanisms for disease progression are unclear and little is known regarding the role of long non-coding RNAs (lncRNA) in mediating disease progression and response to treatment. Here, we used transcriptome sequencing to compare newly diagnosed MM (NDMM) patients who had short progression-free survival (PFS) to standard first-line treatment (PFS < 24 months) to patients who had prolonged PFS (PFS > 24 months). We identified 157 differentially upregulated lncRNAs with short PFS and focused our efforts on characterizing the most upregulated lncRNA, LINC01432. We investigated LINC01432 to show that its overexpression significantly increases cell viability and reduces apoptosis, while knockdown significantly reduces viability and increases apoptosis. Next, we show that LINC01432 directly interacts with the RNA binding protein, CELF2. Lastly, we showed that LINC01432-targeted locked nucleic acid antisense oligonucleotides reduce viability and increases apoptosis. In summary, this fundamental study identified lncRNAs associated with short PFS to standard NDMM treatment and further characterized LINC01432.
Collapse
Affiliation(s)
- Richa Mishra
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
| | - Prasanth Thunuguntla
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
| | - Alani Perkin
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
| | - Dhanusha Duraiyan
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
| | - Katelyn Bagwill
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
| | - Savannah Gonzales
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
| | - Vanessa Brizuela
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
| | - Steve Daly
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
| | - Yoon Jae Chang
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
| | - Mahdote Abebe
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
| | - Yash Rajana
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
| | - Kelly Wichmann
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
| | - Catheryn Bolick
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
| | - Jaiyana King
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
| | - Mark Fiala
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
- Siteman Cancer Center, Washington University in St. Louis, MO, 631102
| | - Julie Fortier
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
| | - Reyka Jayasinghe
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
| | - Mark Schroeder
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
- Siteman Cancer Center, Washington University in St. Louis, MO, 631102
| | - Li Ding
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
- Siteman Cancer Center, Washington University in St. Louis, MO, 631102
| | - Ravi Vij
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
- Siteman Cancer Center, Washington University in St. Louis, MO, 631102
| | - Jessica Silva-Fisher
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
- Siteman Cancer Center, Washington University in St. Louis, MO, 631102
| |
Collapse
|
3
|
Srivastava A, Mishra S, Avadhesh, Shekher A, Rai V, Dhasmana A, Das J, Perenzoni D, Iori R, Gupta SC. Moringin, an isothiocyanate modulates multiple cellular signalling molecules in breast cancer cells. Cell Signal 2024; 119:111181. [PMID: 38643946 DOI: 10.1016/j.cellsig.2024.111181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 03/27/2024] [Accepted: 04/14/2024] [Indexed: 04/23/2024]
Abstract
Prohibitin (PHB) is a pleiotropic molecule with a variety of known functions and subcellular locations. PHB's function in breast cancer is poorly understood. Herein, we report that PHB is expressed in cancer types of diverse origin including breast cancer. The cancer patients with changes in PHB were reported to have significantly reduced 'overall survival' in comparison to the cases without alterations in PHB. The expression of PHB was increased by H2O2 and also by Moringin (MG), which is an isothiocyanate derived from the seeds of Moringa oleifera. MG interacted with PHB, DRP1, and SLP2 and inhibited the growth of MCF-7 and MDAMB-231 cells. The isothiocyanate triggered apoptosis in breast cancer cells as revealed by AO/PI assay, phosphatidylserine externalization, cell cycle analysis and DAPI staining. MG induced proapoptotic proteins expression such as cytochrome c, p53, and cleaved caspase-7. Further, cell survival proteins such as survivin, Bcl-2, and Bcl-xL were suppressed. A depolarization of membrane potential suggested that the apoptosis was triggered through mitochondria. The isothiocyanate suppressed the cancer cell migration and interacted with NF-κB subunits. MG suppressed p65 nuclear translocation induced by TNF-α. The reactive oxygen species generation was also induced by the isothiocyanate in breast cancer cells. MG also modulated the expression of lncRNAs. Collectively, the functions of PHB in breast cancer growth is evident from this study. The activities of MG against breast cancer might result from its ability to modulate multiple cancer-related targets.
Collapse
Affiliation(s)
- Ankit Srivastava
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221 005, India
| | - Shruti Mishra
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221 005, India
| | - Avadhesh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221 005, India
| | - Anusmita Shekher
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221 005, India; Department of General Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Vipin Rai
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221 005, India
| | - Anupam Dhasmana
- Department of Bioscience and Cancer Research Institute, Himalayan Institute of Medical Sciences, Swami Rama Himalayan University, Dehradun 248 016, India; Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Jayanta Das
- Department of Biochemistry, All India Institute of Medical Sciences, Guwahati, India
| | - Daniele Perenzoni
- Department of Food Quality and Nutrition, Research and Innovation Centre, Fondazione Edmund Mach (FEM), San Michele all'Adige, Italy
| | - Renato Iori
- Department of Food Quality and Nutrition, Research and Innovation Centre, Fondazione Edmund Mach (FEM), San Michele all'Adige, Italy
| | - Subash C Gupta
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221 005, India; Department of Biochemistry, All India Institute of Medical Sciences, Guwahati, India.
| |
Collapse
|
4
|
Mishra R, Thunuguntla P, Perkin A, Duraiyan D, Bagwill K, Gonzales S, Brizuela V, Daly S, Chang YJ, Abebe M, Rajana Y, Wichmann K, Bolick C, King J, Fiala M, Fortier J, Jayasinghe R, Schroeder M, Ding L, Vij R, Silva-Fisher J. LINC01432 binds to CELF2 in newly diagnosed multiple myeloma promoting short progression-free survival to standard therapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.27.600975. [PMID: 38979159 PMCID: PMC11230414 DOI: 10.1101/2024.06.27.600975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Multiple Myeloma (MM) is a highly prevalent and incurable form of cancer that arises from malignant plasma cells, with over 35,000 new cases diagnosed annually in the United States. While there are a growing number of approved therapies, MM remains incurable and nearly all patients will relapse and exhaust all available treatment options. Mechanisms for disease progression are unclear and in particular, little is known regarding the role of long non-coding RNAs (lncRNA) in mediating disease progression and response to treatment. In this study, we used transcriptome sequencing to compare newly diagnosed MM patients who had short progression-free survival (PFS) to standard first-line treatment (PFS < 24 months) to patients who had prolonged PFS (PFS > 24 months). We identified 157 differentially upregulated lncRNAs with short PFS and focused our efforts on characterizing the most upregulated lncRNA, LINC01432. We investigated LINC01432 overexpression and CRISPR/Cas9 knockdown in MM cell lines to show that LINC01432 overexpression significantly increases cell viability and reduces apoptosis, while knockdown significantly reduces viability and increases apoptosis, supporting the clinical relevance of this lncRNA. Next, we used individual-nucleotide resolution cross-linking immunoprecipitation with RT-qPCR to show that LINC01432 directly interacts with the RNA binding protein, CELF2. Lastly, we showed that LINC01432-targeted locked nucleic acid antisense oligonucleotides reduce viability and increases apoptosis. In summary, this fundamental study identified lncRNAs associated with short PFS to standard NDMM treatment and further characterized LINC01432, which inhibits apoptosis.
Collapse
|
5
|
Ismail NH, Mussa A, Al-Khreisat MJ, Mohamed Yusoff S, Husin A, Al-Jamal HAN, Johan MF, Islam MA. Dysregulation of Non-Coding RNAs: Roles of miRNAs and lncRNAs in the Pathogenesis of Multiple Myeloma. Noncoding RNA 2023; 9:68. [PMID: 37987364 PMCID: PMC10660696 DOI: 10.3390/ncrna9060068] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 11/22/2023] Open
Abstract
The dysregulation of non-coding RNAs (ncRNAs), specifically microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), leads to the development and advancement of multiple myeloma (MM). miRNAs, in particular, are paramount in post-transcriptional gene regulation, promoting mRNA degradation and translational inhibition. As a result, miRNAs can serve as oncogenes or tumor suppressors depending on the target genes. In MM, miRNA disruption could result in abnormal gene expression responsible for cell growth, apoptosis, and other biological processes pertinent to cancer development. The dysregulated miRNAs inhibit the activity of tumor suppressor genes, contributing to disease progression. Nonetheless, several miRNAs are downregulated in MM and have been identified as gene regulators implicated in extracellular matrix remodeling and cell adhesion. miRNA depletion potentially facilitates the tumor advancement and resistance of therapeutic drugs. Additionally, lncRNAs are key regulators of numerous cellular processes, such as gene expression, chromatin remodeling, protein trafficking, and recently linked MM development. The lncRNAs are uniquely expressed and influence gene expression that supports MM growth, in addition to facilitating cellular proliferation and viability via multiple molecular pathways. miRNA and lncRNA alterations potentially result in anomalous gene expression and interfere with the regular functioning of MM. Thus, this review aims to highlight the dysregulation of these ncRNAs, which engender novel therapeutic modalities for the treatment of MM.
Collapse
Affiliation(s)
- Nor Hayati Ismail
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Ali Mussa
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
- Department of Biology, Faculty of Education, Omdurman Islamic University, Omdurman 11111, Sudan
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, Tamil Nadu, India
| | - Mutaz Jamal Al-Khreisat
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Shafini Mohamed Yusoff
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Azlan Husin
- Department of Internal Medicine, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Hamid Ali Nagi Al-Jamal
- School of Biomedicine, Faculty of Health Sciences, Universiti Sultan Zainal Abidin (UniSZA), Kuala Nerus 21300, Terengganu, Malaysia
| | - Muhammad Farid Johan
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Md Asiful Islam
- WHO Collaborating Centre for Global Women’s Health, Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| |
Collapse
|
6
|
Zhao Y, Jia N, Xie X, Chen Q, Hu T. Whole Transcriptome Analysis of Intervention Effect of Sophora subprostrate Polysaccharide on Inflammation in PCV2 Infected Murine Splenic Lymphocytes. Curr Issues Mol Biol 2023; 45:6067-6084. [PMID: 37504299 PMCID: PMC10377888 DOI: 10.3390/cimb45070383] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 07/13/2023] [Accepted: 07/18/2023] [Indexed: 07/29/2023] Open
Abstract
(1) Background: Sophora subprostrate, is the dried root and rhizome of Sophora tonkinensis Gagnep. Sophora subprostrate polysaccharide (SSP1) was extracted from Sophora subprostrate, which has shown good anti-inflammatory and antioxidant effects. Previous studies showed SSP1 could modulate inflammatory damage induced by porcine circovirus type 2 (PCV2) in murine splenic lymphocytes, but the specific regulatory mechanism is unclear. (2) Methods: Whole transcriptome analysis was used to characterize the differentially expressed mRNA, lncRNA, and miRNA in PCV2-infected cells and SSP1-treated infected cells. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) and other analyses were used to screen for key inflammation-related differentially expressed genes. The sequencing results were verified by RT-qPCR, and western blot was used to verify the key protein in main enriched signal pathways. (3) Results: SSP1 can regulate inflammation-related gene changes induced by PCV2, and its interventional mechanism is mainly involved in the key differential miRNA including miR-7032-y, miR-328-y, and miR-484-z. These inflammation-related genes were mainly enriched in the TNF signal pathway and NF-κB signal pathway, and SSP1 could significantly inhibit the protein expression levels of p-IκB, p-p65, TNF-α, IRF1, GBP2 and p-SAMHD1 to alleviate inflammatory damage. (4) Conclusions: The mechanism of SSP1 regulating PCV2-induced murine splenic lymphocyte inflammation was explored from a whole transcriptome perspective, which provides a theoretical basis for the practical application of SSP1.
Collapse
Affiliation(s)
- Yi Zhao
- College of Animal Science and Technology, Guangxi University, Nanning 530005, China
| | - Nina Jia
- College of Animal Science and Technology, Guangxi University, Nanning 530005, China
| | - Xiaodong Xie
- College of Animal Science and Technology, Guangxi University, Nanning 530005, China
| | - Qi Chen
- College of Animal Science and Technology, Guangxi University, Nanning 530005, China
| | - Tingjun Hu
- College of Animal Science and Technology, Guangxi University, Nanning 530005, China
| |
Collapse
|
7
|
Guo N, Song Y, Zi F, Zheng J, Cheng J. Abnormal expression pattern of lncRNA H19 participates in multiple myeloma bone disease by unbalancing osteogenesis and osteolysis. Int Immunopharmacol 2023; 119:110058. [PMID: 37058751 DOI: 10.1016/j.intimp.2023.110058] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 03/07/2023] [Accepted: 03/16/2023] [Indexed: 04/16/2023]
Abstract
BACKGROUND Accumulating genetic and epigenetic alterations in multiple myeloma (MM) have been demonstrated to be closely associated with osteolytic bone disease, generally characterized as increased osteoclast formation and decreased osteoblast activity. Previously, serum long non-coding RNA (lncRNA) H19 has been proved to be a biomarker for the diagnosis of MM. Whereas, its role in MM-associated bone homeostasis remains largely elusive. METHODS A cohort of 42 MM patients and 40 healthy volunteers were enrolled for evaluating differential expressions of H19 and its downstream effectors. The proliferative capacity of MM cells was monitored by CCK-8 assay. Alkaline phosphatase (ALP) staining and activity detection, either with Alizarin red staining (ARS) were employed to assess osteoblast formation. Osteoblast- or osteoclast-associated gene were detected using qRT-PCR and western blot analysis. Bioinformatics analysis, RNA pull-down, RNA immunoprecipitation (RIP), and chromatin immunoprecipitation (ChIP) were subjected to verify H19/miR-532-3p/E2F7/EZH2 axis, which was accounted for epigenetic suppression of PTEN. The functional role of H19 on MM development through unbalancing osteolysis and osteogenesis was also confirmed in the murine MM model. RESULTS Upregulation of serum H19 was observed in MM patients, suggesting its positive correlation with the poor prognosis of MM patients. Loss of H19 dramatically weakened cell proliferation of MM cells, promoted osteoblastic differentiation, and impaired osteoclast activity. While reinforced H19 exhibited the opposite effects. Akt/mTOR signaling plays an indispensable role in H19-mediated osteoblast formation and osteoclastgenesis. Mechanistically, H19 served as a sponge for miR-532-3p to upregulate E2F7, a transcriptional activator of EZH2, thereby accounting for modulating epigenetic suppression of PTEN. The in vivo experiments further validated that H19 exerted important impacts on tumor growth through breaking the balance between osteogenesis and osteolysis via Akt/mTOR signaling. CONCLUSION Collectively, increased enrichment of H19 in MM cells exhibits an essential role in MM development by disturbing bone homeostasis.
Collapse
Affiliation(s)
- Ninghong Guo
- Department of Hematology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, PR China
| | - Yuan Song
- Department of Hematology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, PR China
| | - Fuming Zi
- Department of Hematology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, PR China
| | - Jifu Zheng
- Department of Hematology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, PR China
| | - Jing Cheng
- Department of Hematology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, PR China.
| |
Collapse
|
8
|
Qin J, Ke B, Liu T, Kong C, Li A, Fu H, Jin C. Aberrantly expressed long noncoding RNAs as potential prognostic biomarkers in newly diagnosed multiple myeloma: A systemic review and meta-analysis. Cancer Med 2023; 12:2199-2218. [PMID: 36057947 PMCID: PMC9939128 DOI: 10.1002/cam4.5135] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 05/17/2022] [Accepted: 08/02/2022] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Numerous studies have manifested long noncoding RNAs (lncRNAs) as biomarkers to determine the prognosis of multiple myeloma (MM) patients. Nevertheless, the prognostic role of lncRNAs in MM is still ambiguous. Herein, we performed a meta-analysis to evaluate the predictive value of aberrantly expressed lncRNAs in MM. METHODS A systemic literature search was performed in PubMed, EMBASE, Cochrane, and Web of Science databases until October 9, 2021, and the protocol was registered in the PROSPERO database (CRD42021284364). Our study extracted the hazard ratios (HRs) and 95% confidence intervals (CIs) of overall survival (OS), progression-free survival (PFS), or event-free survival (EFS). Begg's and Egger's tests were employed to correct publication bias. RESULT Twenty-six individual studies containing 3501 MM patients were enrolled in this study. The results showed that aberrant expression of lncRNAs was associated with poor OS and PFS of MM patients. The pooled HRs for univariate OS and PFS were 1.48 (95% CI = 1.17-1.88, p < 0.001) and 1.30 (95% CI = 1.18-1.43, p < 0.001), respectively, whereas the pooled HRs for multivariate OS and PFS were 1.50 (95% CI = 1.16-1.95, p < 0.001) and 1.59 (95% CI = 1.22-2.07, p < 0.001), respectively. Subgroup analysis suggested that MALAT1, TCF7, NEAT1, and PVT1 upregulation were associated with poor OS (p < 0.05), PVT1, and TCF7 upregulation were implicated with worse PFS (p < 0.05), while only TCF7 overexpression was correlated with reduced EFS (p < 0.05). Moreover, the contour-enhanced funnel plot demonstrated the reliability of our current conclusion, which was not affected by publication bias. CONCLUSION Aberrantly expressed particular lncRNAs are critical prognostic indicators in long-term survival as well as promising biomarkers in progression-free status. However, different cutoff values and dissimilar methods to assess lncRNA expression among studies may lead to heterogeneity.
Collapse
Affiliation(s)
- Jiading Qin
- Medical College of Nanchang UniversityNanchangJiangxi330006China
- Department of HematologyJiangxi Provincial People's HospitalNanchangJiangxi330006China
| | - Bo Ke
- Department of HematologyJiangxi Provincial People's HospitalNanchangJiangxi330006China
- National Clinical Research Center for Hematologic DiseasesThe First Affiliated Hospital of Soochow UniversitySoochowJiangsu215006China
| | - Tingting Liu
- Department of HematologyJiangxi Provincial People's HospitalNanchangJiangxi330006China
| | - Chunfang Kong
- Medical College of Nanchang UniversityNanchangJiangxi330006China
- Department of HematologyJiangxi Provincial People's HospitalNanchangJiangxi330006China
| | - Anna Li
- Department of HematologyJiangxi Provincial People's HospitalNanchangJiangxi330006China
| | - Huan Fu
- Department of HematologyJiangxi Provincial People's HospitalNanchangJiangxi330006China
| | - Chenghao Jin
- Medical College of Nanchang UniversityNanchangJiangxi330006China
- Department of HematologyJiangxi Provincial People's HospitalNanchangJiangxi330006China
- National Clinical Research Center for Hematologic DiseasesThe First Affiliated Hospital of Soochow UniversitySoochowJiangsu215006China
| |
Collapse
|
9
|
Zhong Y, Xu S, Liu Z. The potential of glutamine metabolism-related long non-coding RNAs (lncRNAs) as prognostic biomarkers in multiple myeloma patients. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1362. [PMID: 36660731 PMCID: PMC9843343 DOI: 10.21037/atm-22-6190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 12/19/2022] [Indexed: 01/01/2023]
Abstract
Background Glutamine (Gln) metabolism has been confirmed as an important fuel in cancer metabolism. This study aimed to uncover potential links of Gln with long non-coding RNAs (lncRNAs) and the prognostic value of Gln-associated lncRNAs in multiple myeloma (MM) patients. Methods The RNA-seq expression profile and corresponding clinical data of gastric cancer obtained from Gene Expression Omnibus (GEO) database. Unsupervised consensus clustering was used to cluster MM samples based on Gln-associated lncRNAs. The overall survival (OS), biological pathways, and immune microenvironment were compared in different subtypes. Differential analysis was utilized to identify differentially expressed lncRNAs (DElncRNAs) in different subtypes. A risk model was constructed based on DElncRNAs by using Cox regression, least absolute shrinkage and selection operator (LASSO), and the stepAIC algorithm. Results We screened 50 Gln-associated lncRNAs and identified 3 molecular subtypes (clust1, clust2, and clust3) based on lncRNA expression profiles. Clust3 subtype showed the worst prognosis and highest enrichment of Gln metabolism pathway. Angiogenesis, epithelial-mesenchymal transition (EMT), and cell cycle-related pathways were relatively activated in clust3. Then, we identified 11 prognostic DElncRNAs for constructing the risk model. The MM samples were divided into high- and low-risk groups with distinct prognosis according to the risk score. The risk score was significantly associated with cell cycle and infiltration of many immune cells. Conclusions This study characterized the role of Gln-associated lncRNAs in Gln metabolism contributing for tumor-related pathways and immune microenvironment in MM patients. The 11 lncRNAs in the risk model may serve as potential targets for exploring the mechanism of Gln metabolism or serve as potential biomarkers for MM prognosis.
Collapse
Affiliation(s)
- Yun Zhong
- Department of Lymphohematology and Oncology, Jiangxi Cancer Hospital, Nanchang, China
| | - Shenghua Xu
- Department of Lymphohematology and Oncology, Jiangxi Cancer Hospital, Nanchang, China
| | - Zhe Liu
- Department of Orthopedics, Jiangxi Cancer Hospital, Nanchang, China
| |
Collapse
|
10
|
Li X, Jin X, Wang J, Li X, Zhang H. Dexamethasone attenuates dry eye-induced pyroptosis by regulating the KCNQ1OT1/miR-214 cascade. Steroids 2022; 186:109073. [PMID: 35779698 DOI: 10.1016/j.steroids.2022.109073] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 06/21/2022] [Accepted: 06/23/2022] [Indexed: 10/17/2022]
Abstract
Dry eye disease (DED) is an inflammatory disorder of the ocular surface seriously affecting the quality of life of patients. Topical dexamethasone (Dex) administration protects the cornea from the hyperosmotic stress (HS) induced by tears. Pyroptosis participates in the activation of epithelial inflammation during DED. However, it remains unclear whether Dex attenuates the progression of DED through pyroptosis. In this study, we aimed to investigate the effect of Dex on DED using both cell and animal models and its underlying mechanism. The inflammatory factors contained in tears were detected using a cytokine assay. The pyroptosis in DED mice and human corneal epithelial cells (HCECs) treated with hyperosmotic medium under various treatments was evaluated by immunohistochemical assays (IHC) or western blotting (WB). RNA expression was manipulated with siRNA or agomir microRNAs and measured using a polymerase chain reaction. The scratch assay was used to assess the migration rate of HCECs. Remaining corneal defects were evaluated using fluorescein staining and photographed using a digital camera. Dex could suppress the release of inflammatory factors and notably attenuate pyroptosis, KCNQ1OT1 expression, and NF-κB activation induced by HS injury in vivo and in vitro. KCNQ1OT1 upregulation could activate pyroptosis by sponging miR-214. Furthermore, KCNQ1OT1 knockdown and miR-214 overexpression reversed the effect of HS, promoted the migration of HCECs, and accelerated corneal wound healing. Dex effectively suppressed HS-induced pyroptosis through the KCNQ1OT1/miR-214/caspase-1 signaling axis by inhibiting the NF-κB activation. Our results provide a novel understanding of the mechanism of Dex as an anti-inflammatory drug in DED.
Collapse
Affiliation(s)
- Xuran Li
- Eye Hospital, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Basic and Clinical Research of Heilongjiang Province, Harbin, China
| | - Xin Jin
- Eye Hospital, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Basic and Clinical Research of Heilongjiang Province, Harbin, China
| | - Jingrao Wang
- Eye Hospital, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Basic and Clinical Research of Heilongjiang Province, Harbin, China
| | - Xinyue Li
- Eye Hospital, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Basic and Clinical Research of Heilongjiang Province, Harbin, China
| | - Hong Zhang
- Eye Hospital, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Basic and Clinical Research of Heilongjiang Province, Harbin, China.
| |
Collapse
|
11
|
Chen Y, Wang N, Cao L, Zhang D, Peng H, Xue P. Long non-coding RNA HOXB-AS1 is a prognostic marker and promotes hepatocellular carcinoma cells' proliferation and invasion. Open Life Sci 2022; 17:944-951. [PMID: 36045719 PMCID: PMC9380905 DOI: 10.1515/biol-2022-0040] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 11/12/2021] [Accepted: 01/03/2022] [Indexed: 11/15/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) are broadly transcribed in the genome of human and play critical roles in the progression of multiple diseases. Long non-coding HOXB cluster antisense RNA 1 (HOXB-AS1) is a tumor exciter in various cancers. This study aimed to investigate the involvement of HOXB-AS1 in hepatocellular carcinoma (HCC). In the following study, HOXB-AS1 was unveiled to be highly expressed in HCC tissues as opposed to normal tissues. Silencing of HOXB-AS1 led to the loss of proliferation, migration, and invasiveness of HCC cells, namely Hep3B and Huh7. Moreover, the data showed that expression levels of HOXB-AS1 contribute significantly to the patient's survival rates. Otherwise, HOXB-AS1 levels in the serum of patients proved HOXB-AS1 as a biomarker for analysis and treatment of HCC. In summary, this study highlights HOXB-AS1 as key upregulated lncRNA in HCC which being an oncogene can cause proliferation and metastasis of HCC cells. The results also highlighted HOXB-AS1 as a promising biomarker for early diagnosis and prognosis of patients with HCC.
Collapse
Affiliation(s)
- Yubin Chen
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Guangzhou Medical University, No. 63 Yayun South Road, Guangzhou, 510000, Guangdong Province, China
| | - Na Wang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Guangzhou Medical University, No. 63 Yayun South Road, Guangzhou, 510000, Guangdong Province, China
| | - Liangqi Cao
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Guangzhou Medical University, No. 63 Yayun South Road, Guangzhou, 510000, Guangdong Province, China
| | - Dawei Zhang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Guangzhou Medical University, No. 63 Yayun South Road, Guangzhou, 510000, Guangdong Province, China
| | - Heping Peng
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Guangzhou Medical University, No. 63 Yayun South Road, Guangzhou, 510000, Guangdong Province, China
| | - Ping Xue
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Guangzhou Medical University, No. 63 Yayun South Road, Guangzhou, 510000, Guangdong Province, China
| |
Collapse
|
12
|
Yuan W, Huang J, Hou S, Li H, Bie L, Chen B, Li G, Zhou Y, Chen X. The Antigastric Cancer Effect of Triptolide is Associated With H19/NF-κB/FLIP Axis. Front Pharmacol 2022; 13:918588. [PMID: 36110523 PMCID: PMC9469193 DOI: 10.3389/fphar.2022.918588] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 06/02/2022] [Indexed: 12/11/2022] Open
Abstract
Background and Objective: Triptolide (TP), one of the fat-soluble components extracted from the Chinese medicinal herb Tripterygium wilfordii Hook F. (TWHF), possesses strong antitumor bioactivities, but its dose-dependent side effects restrict its wide application. This study was designed to investigate whether inflammatory factors increased the antitumor effects of the nontoxic dose of TP on gastric cancer cells and tried to explore the possible molecular mechanisms. Method: AGS and MKN45 cells were treated with different doses of TP and TNF-α. Cell viability and apoptosis were detected in vitro. In addition, NF-κB mediated prosurvival signals and cytoprotective proteins, especially FLICE-inhibitory protein (FLIP), were detected to determine their effects on TP/TNF-α–induced apoptosis. Moreover, the function of lncRNA H19/miR-204-5p/NF-κB/FLIP axis was investigated in vitro, and the antigastric cancer effect of TP plus TNF-α was proved in the mice xenograft model. Result:In vitro experimental results showed that TP pretreatment promoted apoptosis in AGS and MKN45 cells upon TNF-α exposure. TP/TNF-α–mediated apoptosis was partly mediated by the inhibitory effect of NF-κB–mediated FLIP expression. Oncogene H19 lying in the upstream pathway of NF-κB played a vital role upon TNF-α exposure, and bioinformatics analysis proved that H19 participated in TP/TNF-α–induced apoptosis via binding of miR-204-5p. Lastly, a low dose of TP and TNF-α inhibited the tumor weight and tumor volume of AGS and MKN45 cells in vivo. Conclusion: TP pretreatment increased apoptosis in TNF-α–stimulated gastric cancer cells, which are dependent on the disruption of the H19/miR-204-5p/NF-κB/FLIP axis. Cotreatment of TP and TNF-α is a better option for enhancing the anticancer effect and lowering the side effect of TP.
Collapse
Affiliation(s)
- Weiwei Yuan
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Jinxi Huang
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Shanshan Hou
- Department of Pharmacy, Zhejiang Pharmaceutical College, Ningbo, China
| | - Huahua Li
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Liangyu Bie
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou University, Zhengzhou, China
| | - Beibei Chen
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou University, Zhengzhou, China
| | - Gaofeng Li
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Yang Zhou
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou University, Zhengzhou, China
- *Correspondence: Xiaobing Chen, ; Yang Zhou,
| | - Xiaobing Chen
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou University, Zhengzhou, China
- *Correspondence: Xiaobing Chen, ; Yang Zhou,
| |
Collapse
|
13
|
Khazamipour A, Gholampour-Faroji N, Zeraati T, Vakilian F, Haddad-Mashadrizeh A, Ghayour Mobarhan M, Pasdar A. A novel causative functional mutation in GATA6 gene is responsible for familial dilated cardiomyopathy as supported by in silico functional analysis. Sci Rep 2022; 12:13752. [PMID: 35962153 PMCID: PMC9374661 DOI: 10.1038/s41598-022-13993-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 05/31/2022] [Indexed: 11/09/2022] Open
Abstract
Dilated cardiomyopathy (DCM), one of the most common types of cardiomyopathies has a heterogeneous nature and can be seen in Mendelian forms. Next Generation Sequencing is a powerful tool for identifying novel variants in monogenic disorders. We used whole-exome sequencing (WES) and Sanger sequencing techniques to identify the causative mutation of DCM in an Iranian pedigree. We found a novel variant in the GATA6 gene, leading to substituting Histidine by Tyrosine at position 329, observed in all affected family members in the pedigree, whereas it was not established in any of the unaffected ones. We hypothesized that the H329Y mutation may be causative for the familial pattern of DCM in this family. The predicted models of GATA6 and H329Y showed the high quality according to PROCHECK and ERRAT. Nonetheless, simulation results revealed that the protein stability decreased after mutation, while the flexibility may have been increased. Hence, the mutation led to the increased compactness of GATA6. Overall, these data indicated that the mutation could affect the protein structure, which may be related to the functional impairment of GATA6 upon H329Y mutation, likewise their involvement in pathologies. Further functional investigations would help elucidating the exact mechanism.
Collapse
Affiliation(s)
- Afrouz Khazamipour
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Nazanin Gholampour-Faroji
- Biotechnology Department, Iranian Research Organization for Science and Technology (IROST), Tehran, Iran
| | - Tina Zeraati
- Medical Genetics Research Centre, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farveh Vakilian
- Atherosclerosis Prevention Research Centre, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Aliakbar Haddad-Mashadrizeh
- Industrial Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Majid Ghayour Mobarhan
- Metabolic Syndrome Research Centre, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Alireza Pasdar
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran. .,Medical Genetics Research Centre, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran. .,Division of Applied Medicine, Medical School, University of Aberdeen, Foresterhill, Aberdeen, UK. .,Bioinformatics Research Centre, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
14
|
KIZILYER A. An optimized protocol for the electroporation of NCI H929 multiple myeloma cells. MEHMET AKIF ERSOY ÜNIVERSITESI VETERINER FAKÜLTESI DERGISI 2022. [DOI: 10.24880/maeuvfd.1126466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Multiple myeloma cell lines are difficult to transfect with non-viral nucleic acid delivery methods. While electroporation is the most efficient tool for the transfection of most hard-to-transfect cells, human multiple myeloma cells differ in their permissiveness and each cell type require different electroporation conditions for an efficient transgene delivery. In this study, various parameters for NCI H929 human multiple myeloma cells are tested to generate an optimized electroporation protocol. Findings from this paper showed that besides the voltage and capacitance settings, cell count, the cell cycle status of cells, the amount of nucleic acid and removal of death cells all impacted the electroporation efficiency and viable cell count. These results are expected to serve as a starting point and a guide for researchers.
Collapse
|
15
|
Arpi MNT, Simpson TI. SFARI genes and where to find them; modelling Autism Spectrum Disorder specific gene expression dysregulation with RNA-seq data. Sci Rep 2022; 12:10158. [PMID: 35710789 PMCID: PMC9203566 DOI: 10.1038/s41598-022-14077-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 06/01/2022] [Indexed: 11/09/2022] Open
Abstract
Autism Spectrum Disorders (ASD) have a strong, yet heterogeneous, genetic component. Among the various methods that are being developed to help reveal the underlying molecular aetiology of the disease one approach that is gaining popularity is the combination of gene expression and clinical genetic data, often using the SFARI-gene database, which comprises lists of curated genes considered to have causative roles in ASD when mutated in patients. We build a gene co-expression network to study the relationship between ASD-specific transcriptomic data and SFARI genes and then analyse it at different levels of granularity. No significant evidence is found of association between SFARI genes and differential gene expression patterns when comparing ASD samples to a control group, nor statistical enrichment of SFARI genes in gene co-expression network modules that have a strong correlation with ASD diagnosis. However, classification models that incorporate topological information from the whole ASD-specific gene co-expression network can predict novel SFARI candidate genes that share features of existing SFARI genes and have support for roles in ASD in the literature. A statistically significant association is also found between the absolute level of gene expression and SFARI's genes and Scores, which can confound the analysis if uncorrected. We propose a novel approach to correct for this that is general enough to be applied to other problems affected by continuous sources of bias. It was found that only co-expression network analyses that integrate information from the whole network are able to reveal signatures linked to ASD diagnosis and novel candidate genes for the study of ASD, which individual gene or module analyses fail to do. It was also found that the influence of SFARI genes permeates not only other ASD scoring systems, but also lists of genes believed to be involved in other neurodevelopmental disorders.
Collapse
Affiliation(s)
| | - T Ian Simpson
- School of Informatics, University of Edinburgh, 10 Crichton Street, Edinburgh, EH8 9AB, UK. .,Simons Initiative for the Developing Brain (SIDB), Centre for Brain Discovery Sciences, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
16
|
Wu B, Zhang Y, Yu Y, Zhong C, Lang Q, Liang Z, Lv C, Xu F, Tian Y. Long Noncoding RNA H19: A Novel Therapeutic Target Emerging in Oncology Via Regulating Oncogenic Signaling Pathways. Front Cell Dev Biol 2021; 9:796740. [PMID: 34977037 PMCID: PMC8716783 DOI: 10.3389/fcell.2021.796740] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 11/24/2021] [Indexed: 12/24/2022] Open
Abstract
Long noncoding RNA H19 (H19) is an imprinting gene with only maternal expression that is involved in regulating different processes in various types of cells. Previous studies have shown that abnormal H19 expression is involved in many pathological processes, such as cancer, mainly through sponging miRNAs, interacting with proteins, or regulating epigenetic modifications. Accumulating evidence has shown that several oncogenic signaling pathways lead to carcinogenesis. Recently, the regulatory relationship between H19 and oncogenic signaling pathways in various types of cancer has been of great interest to many researchers. In this review, we discussed the key roles of H19 in cancer development and progression via its regulatory function in several oncogenic signaling pathways, such as PI3K/Akt, canonical Wnt/β-catenin, canonical NF-κB, MAPK, JAK/STAT and apoptosis. These oncogenic signaling pathways regulated by H19 are involved in cell proliferation, proliferation, migration and invasion, angiogenesis, and apoptosis of various cancer cells. This review suggests that H19 may be a novel therapeutic target for cancers treatment by regulating oncogenic signaling pathways.
Collapse
Affiliation(s)
- Baokang Wu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yizhou Zhang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yang Yu
- Department of Surgery, Jinzhou Medical University, Jinzhou, China
| | - Chongli Zhong
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qi Lang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhiyun Liang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Chao Lv
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Feng Xu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yu Tian
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
17
|
H19- and hsa-miR-338-3p-mediated NRP1 expression is an independent predictor of poor prognosis in glioblastoma. PLoS One 2021; 16:e0260103. [PMID: 34843522 PMCID: PMC8629300 DOI: 10.1371/journal.pone.0260103] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 11/02/2021] [Indexed: 12/19/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and also the most invasive brain cancer. GBM progression is rapid and its prognosis is poor. Therefore, finding molecular targets in GBM is a critical goal that could also play important roles in clinical diagnostics and treatments to improve patient prognosis. We jointly analyzed the GSE103227, GSE103229, and TCGA databases for differentially expressed RNA species, obtaining 52 long non-coding RNAs (lncRNAs), 31 microRNAs (miRNAs), and 186 mRNAs, which were used to build a competing endogenous RNA network. Kaplan–Meier and receiver operating characteristic (ROC) analyses revealed five survival-related lncRNAs: H19, LINC01574, LINC01614, RNF144A-AS1, and OSMR-AS1. With multiple optimization mRNAs, we found the H19-hsa-miR-338-3P-NRP1 regulatory pathway. Additionally, we noted high NRP1 expression in GBM patients, and Kaplan–Meier and ROC analyses showed that NRP1 expression was associated with GBM prognosis. Cox analysis indicated that NRP1 is an independent prognostic factor in GBM patients. In conclusion, H19 and hsa-miR-338-3P regulate NRP1 expression, and this pathway plays an important role in GBM.
Collapse
|
18
|
Mardani M, Rashedi S, Keykhaei M, Farrokhpour H, Azadnajafabad S, Tavolinejad H, Rezaei N. Long non-coding RNAs (lncRNAs) as prognostic and diagnostic biomarkers in multiple myeloma: A systematic review and meta-analysis. Pathol Res Pract 2021; 229:153726. [PMID: 34942515 DOI: 10.1016/j.prp.2021.153726] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 11/10/2021] [Accepted: 11/26/2021] [Indexed: 12/27/2022]
Abstract
BACKGROUND Recently, emerging studies have demonstrated the utility of particular long non-coding RNAs (lncRNAs) as useful biomarkers for the diagnosis and prognosis of multiple myeloma (MM). We systematically reviewed the literature and conducted a meta-analysis to quantify the predictive effectiveness of lncRNAs in the prognosis and diagnosis of MM. METHODS A systematic search was performed in PubMed, Embase, and Web of Science until March 24, 2021. A meta-analysis was conducted to explore the correlation between the expression of lncRNAs and prognostic endpoints, including overall survival (OS), progression-free survival (PFS), and disease-free survival (DFS) or event-free survival (EFS). Moreover, the diagnostic performance of lncRNAs in MM was investigated by calculating accuracy metrics. RESULTS Overall, 43 studies were included in this systematic review, amongst which 36 studies assessed prognostic endpoints (including 5499 participants and 69 lncRNAs), and 11 studies evaluated diagnostic outcomes (with 1723 participants and 11 lncRNAs). The overexpression of CRNDE (hazard ratio (HR)= 1.94, 95% confidence interval (CI) 1.61, 2.34), NEAT1 (HR=1.97, 95%CI 1.36, 2.85), PVT1 (HR=1.92, 95%CI 1.25, 2.97), and TCF7 (HR=1.98, 95%CI 1.42, 2.76) was significantly associated with reduced OS. Furthermore, upregulation of PVT1 was significantly correlated with poor PFS (HR=1.86, 95%CI 1.29, 2.68). The pooled diagnostic performance of lncRNAs was as follows: sensitivity 0.78 (95%CI 0.73, 0.82), specificity 0.88 (95%CI 0.83, 0.92), and area under the curve 0.89 (95%CI 0.86, 0.92). CONCLUSIONS Our results revealed the potential significance of lncRNAs in MM as diagnostic and prognostic markers, which may be the future targets for individualized therapy.
Collapse
Affiliation(s)
- Mahta Mardani
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Sina Rashedi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Mohammad Keykhaei
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Hossein Farrokhpour
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Sina Azadnajafabad
- Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran; Department of Surgery, Tehran University of Medical Sciences, Tehran, Iran.
| | - Hamed Tavolinejad
- Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
19
|
Knockdown of LncRNA SNHG1 Suppresses Corneal Angiogenesis by the Regulation of miR-195-5p/VEGF-A. J Ophthalmol 2021; 2021:6646512. [PMID: 34712495 PMCID: PMC8548121 DOI: 10.1155/2021/6646512] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 08/22/2021] [Accepted: 08/23/2021] [Indexed: 12/13/2022] Open
Abstract
LncRNA SNHG1 (SNHG1) has been widely studied as the causative factor of angiogenesis and proliferative agent in gastric, lung, cervical, and hepatocellular carcinomas. However, its significance of angiogenesis and progression of corneal neovascularization (CRNV) is least understood. This study focuses on the molecular mechanisms followed by SNHG1 to establish CRNV and its angiogenesis. Bioinformatics analysis to identify potential miRNA targets of SNHG1 and vascular endothelial growth factor A (VEGF-A) was conducted using StarBase and was subsequently confirmed by the luciferase reporter assay. Relative quantitative expression of SNHG1 in human umbilical vein endothelial cells (HUVECs) was detected through qRT-PCR and western blot analysis. Cell proliferation was detected through CCK-8 assay, whereas migratory abilities of the cells were determined with transwell assay. A capillary-like tube formation assay was performed to detect the tube formation ability of the cells. Following this, relative expression of miR-195-5p and VEGF-A was determined through qRT-PCR and western blot analysis. Results from the experiments manifested upregulated levels of SNHG1 and VEGF-A in HUVECs and CRNV tissues as compared with the control group, whereas downregulated levels of miR-195-5p were measured in the CRNV tissues and HUVECs, suggesting the negative correlation between lncRNA and miRNA. Overexpressed vascular endothelial growth factor promoted cell proliferation and tube formation; however, its silencing leads to inhibition in angiogenesis and proliferation. Potential binding sites of SNHG1 showed miR-195-5p as its direct target and SNHG1 as a sponge for this miRNA. Knockdown and downregulated levels of SNHG1 showed a notable decrease and inhibition in angiogenesis and migration of CRNV cells. The study showed that SNHG1 inhibition significantly reduced cell proliferation, migration, and tube formation in HUVECs transfect with lncRNA SNHG1. Mechanistic insights into the SNHG1 showed that SNHG1 acts as a sponge for miR-195-5p and upregulates the levels of VEGF-A.
Collapse
|
20
|
CRISPR/Cas9 small promoter deletion in H19 lncRNA is associated with altered cell morphology and proliferation. Sci Rep 2021; 11:18380. [PMID: 34526543 PMCID: PMC8443613 DOI: 10.1038/s41598-021-97058-0] [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: 05/09/2021] [Accepted: 08/11/2021] [Indexed: 02/08/2023] Open
Abstract
The imprinted H19 long non-coding RNA, a knowing oncofetal gene, presents a controversial role during the carcinogenesis process since its tumor suppressor or oncogenic activity is not completely elucidated. Since H19 lncRNA is involved in many biological pathways related to tumorigenesis, we sought to develop a non-cancer lineage with CRISPR-Cas9-mediated H19 knockdown (H19-) and observe the changes in a cellular context. To edit the promoter region of H19, two RNA guides were designed, and the murine C2C12 myoblast cells were transfected. H19 deletion was determined by DNA sequencing and gene expression by qPCR. We observed a small deletion (~ 60 bp) in the promoter region that presented four predicted transcription binding sites. The deletion reduced H19 expression (30%) and resulted in increased proliferative activity, altered morphological patterns including cell size and intracellular granularity, without changes in viability. The increased proliferation rate in the H19- cell seems to facilitate chromosomal abnormalities. The H19- myoblast presented characteristics similar to cancer cells, therefore the H19 lncRNA may be an important gene during the initiation of the tumorigenic process. Due to CRISPR/Cas9 permanent edition, the C2C12 H19- knockdown cells allows functional studies of H19 roles in tumorigenesis, prognosis, metastases, as well as drug resistance and targeted therapy.
Collapse
|
21
|
Yang J, Qi M, Fei X, Wang X, Wang K. LncRNA H19: A novel oncogene in multiple cancers. Int J Biol Sci 2021; 17:3188-3208. [PMID: 34421359 PMCID: PMC8375239 DOI: 10.7150/ijbs.62573] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 07/14/2021] [Indexed: 12/13/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) are a series of non-coding RNAs that lack open reading frameworks. Accumulating evidence suggests important roles for lncRNAs in various diseases, including cancers. Recently, lncRNA H19 (H19) became a research focus due to its ectopic expression in human malignant tumors, where it functioned as an oncogene. Subsequently, H19 was confirmed to be involved in tumorigenesis and malignant progression in many tumors and had been implicated in promoting cell growth, invasion, migration, epithelial-mesenchymal transition, metastasis, and apoptosis. H19 also sequesters some microRNAs, facilitating a multilayer molecular regulatory mechanism. In this review, we summarize the abnormal overexpression of H19 in human cancers, which suggests wide prospects for further research into the diagnosis and treatment of cancers.
Collapse
Affiliation(s)
- Jun Yang
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Manlong Qi
- Department of Clinical Genetics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Xiang Fei
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Xia Wang
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Kefeng Wang
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| |
Collapse
|
22
|
Yao W, Chen Q, Li S, Jia X, Xu L, Wei L. RELT promotes the growth of esophageal squamous cell carcinoma by activating the NF-κB pathway. Cell Cycle 2021; 20:1231-1241. [PMID: 34121605 PMCID: PMC8331000 DOI: 10.1080/15384101.2021.1924451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 04/27/2021] [Indexed: 10/21/2022] Open
Abstract
The members of the tumor necrosis factor receptor (TNFR) family have been demonstrated to play critical roles in various cancers. However, little is known about the function of the Receptor Expressed in Lymphoid Tissues (RELT) in cancers, which is a member of the TNFR family, especially in the esophageal squamous cell carcinoma (ESCC). In this study, we found that RELT expression was increased in ESCC tissues and was consequently associated with poor overall survival of ESCC patients. Moreover, RELT overexpression was found to promote cell growth, cell cycle progression, and suppressed cell apoptosis in vitro; it also decreased the expression of p27 and caspase 3, and increased the expression of survivin. In addition, RELT contributed to the tumorigenesis of ESCC in vivo. Furthermore, we suggest that RELT may function in the pathogenesis of ESCC by activating the nuclear factor κB (NF-κB) pathway. An inhibitor of NF-κB reversed the RELT-induced malignancy in the ESCC cells. Altogether, our findings identified that RELT served as an oncogene in ESCC through the NF-κB pathway, suggesting that RELT may be developed as a novel biomarker for the diagnosis and treatment of the ESCC.
Collapse
Affiliation(s)
- Wenjian Yao
- Department of Thoracic Surgery, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan, China
| | - Qing Chen
- Department of Oncology, Jingjiang People’s Hospital, Jingjiang, Jiangsu, P. R. China
| | - Saisai Li
- Department of Thoracic Surgery, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan, China
| | - Xiangbo Jia
- Department of Thoracic Surgery, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan, China
| | - Lei Xu
- Department of Thoracic Surgery, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan, China
| | - Li Wei
- Department of Thoracic Surgery, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan, China
| |
Collapse
|
23
|
Huang Z, Chu L, Liang J, Tan X, Wang Y, Wen J, Chen J, Wu Y, Liu S, Liao J, Hou R, Ding Z, Zhang Z, Liang H, Song S, Yang C, Zhang J, Guo T, Chen X, Zhang B. H19 Promotes HCC Bone Metastasis Through Reducing Osteoprotegerin Expression in a Protein Phosphatase 1 Catalytic Subunit Alpha/p38 Mitogen-Activated Protein Kinase-Dependent Manner and Sponging microRNA 200b-3p. Hepatology 2021; 74:214-232. [PMID: 33615520 DOI: 10.1002/hep.31673] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 11/06/2020] [Accepted: 11/13/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND AND AIMS Bone is the second most frequent site of metastasis for HCC, which leads to an extremely poor prognosis. HCC bone metastasis is typically osteolytic, involving the activation of osteoclasts. Long noncoding RNA H19 plays an important role in the pathogenesis of human cancers. Nonetheless, the mechanism underlying the participation of H19 in HCC bone metastasis remains unclear. APPROACH AND RESULTS The current study established a mouse HCC bone metastasis model by using serial intracardiac injection and cell isolation to obtain cells with distinct bone metastasis ability. H19 was highly expressed in these cells and in clinical HCC bone metastasis specimens. Both osteoclastogenesis in vitro and HCC bone metastasis in vivo were promoted by H19 overexpression, whereas these processes were suppressed by H19 knockdown. H19 overexpression attenuated p38 phosphorylation and further down-regulated the expression of osteoprotegerin (OPG), also known as osteoclastogenesis inhibitory factor. However, up-regulated OPG expression as well as suppressed osteoclastogenesis caused by H19 knockdown were recovered by p38 interference, indicating that p38 mitogen-activated protein kinase (MAPK)-OPG contributed to H19-promoted HCC bone metastasis. Furthermore, we demonstrated that H19 inhibited the expression of OPG by binding with protein phosphatase 1 catalytic subunit alpha (PPP1CA), which dephosphorylates p38. SB-203580-mediated inactivation of p38MAPK reversed the down-regulation of HCC bone metastasis caused by H19 knockdown in vivo. Additionally, H19 enhanced cell migration and invasion by up-regulating zinc finger E-box binding homeobox 1 through the sequestration of microRNA (miR) 200b-3p. CONCLUSIONS H19 plays a critical role in HCC bone metastasis by reducing OPG expression, which is mediated by the PPP1CA-induced inactivation of the p38MAPK pathway; and H19 also functions as a sponge for miR-200b-3p.
Collapse
Affiliation(s)
- Zhao Huang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Liang Chu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Junnan Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Xiaolong Tan
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Yu Wang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Jingyuan Wen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Jin Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Yu Wu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Sha Liu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Jingyu Liao
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Rui Hou
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zeyang Ding
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Zhanguo Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Huifang Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Shasha Song
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Caihong Yang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinming Zhang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Guo
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China.,Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China.,Key Laboratory of Organ Transplantation, National Health Commission, Wuhan, China.,Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China.,Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China.,Key Laboratory of Organ Transplantation, National Health Commission, Wuhan, China.,Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| |
Collapse
|
24
|
Ding T, Deng R, Huang T. Long non-coding RNA T cell factor 7 is associated with increased disease risk and poor prognosis, and promotes cell proliferation, attenuates cell apoptosis and miR-200c expression in multiple myeloma. Oncol Lett 2021; 21:129. [PMID: 33552250 PMCID: PMC7798040 DOI: 10.3892/ol.2020.12390] [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: 08/20/2019] [Accepted: 04/20/2020] [Indexed: 12/13/2022] Open
Abstract
The aim of the present study was to investigate the association of long non-coding RNA T cell factor 7 (lncRNA TCF7) with disease risk, prognosis and its cellular function in multiple myeloma (MM). A total of 132 de novo symptomatic patients with MM and 50 controls were enrolled. Plasma cells from patients with MM and controls were separated from bone marrow samples to detect lncRNA TCF7 expression using reverse transcription-quantitative PCR. In addition, treatment responses, event-free survival (EFS) and overall survival (OS) were measured. The effects of lncRNA TCF7 on proliferation, apoptosis and microRNA-200c (miR-200c) expression were assessed by gain- and loss-of-function experiments in RPMI-8226 and U-266 cells. The results demonstrated that lncRNA TCF7 expression was upregulated in patients with MM compared with controls, and the receiver operating characteristic curve revealed that lncRNA TCF7 could distinguish patients with MM from controls with an area under the curve of 0.793 (95% CI, 0.725-0.861). In patients with MM, high lncRNA TCF7 expression was associated with higher β2-microglobulin, more advanced International Staging System stage and increased t (14; 16) mutations. Furthermore, it was demonstrated that lncRNA TCF7 was downregulated in patients with complete response (CR) compared with patients without CR. Furthermore, high lncRNA TCF7 expression predicted worse EFS and OS. lncRNA TCF7 also promoted cell proliferation, whereas it reduced cell apoptosis and miR-200c expression in RPMI-8226 and U-266 cells. In conclusion, the present results suggested that lncRNA TCF7 may be used as a potential biomarker and as a treatment target for MM.
Collapse
Affiliation(s)
- Tianling Ding
- Department of Hematology, Huashan Hospital Affiliated to Fudan University, Shanghai 200040, P.R. China
| | - Ruoyu Deng
- Shanghai Qeejen Bio-tech Institution, Shanghai 200434, P.R. China
| | - Ting Huang
- Department of Oncology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| |
Collapse
|
25
|
Chen Y, Zhang C, Xiao CX, Li XD, Hu ZL, He SD, Xiao XJ, Xu F. Dexamethasone can attenuate the pulmonary inflammatory response via regulation of the lncH19/miR-324-3p cascade. JOURNAL OF INFLAMMATION-LONDON 2021; 18:1. [PMID: 33413425 PMCID: PMC7789598 DOI: 10.1186/s12950-020-00266-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 11/26/2020] [Indexed: 12/20/2022]
Abstract
Objective To investigate lncRNAs and their roles in regulating the pulmonary inflammatory response under dexamethasone (Dex) treatment. Methods IL-1β (10 ng/mL) and LPS (1 μg/mL) was used to construct inflammatory cell models with A549 cells; IL-1β performed better against LPS. Different concentrations of Dex were used to attenuate the inflammation induced by IL-1β, and its effect was assessed via RT-PCR to detect inflammatory cytokine-related mRNA levels, including those of IKβ-α, IKKβ, IL-6, IL-8, and TNF-α. Furthermore, ELISA was used to detect the levels of the inflammatory cytokines TNF-α, IL-6, and IL-8. RT-PCR was used to quantify the levels of lncRNAs, including lncMALAT1, lncHotair, lncH19, and lncNeat1. LncH19 was most closely associated with the inflammatory response, which was induced by IL-1β and attenuated by Dex. Among the lncRNAs, the level of lncH19 showed the highest increase following treatment with 1 and 10 μM Dex. Therefore, lncH19 was selected for further functional studies. LncH19 expression was inhibited by shRNA transduced with lentivirus. Cell assays for cell proliferation and apoptosis as well as RT-PCR, western blot, and ELISA for inflammatory genes were conducted to confirm the functions of lncH19. The predicted target miRNAs of lncH19 were hsa-miR-346, hsa-miR-324-3p, hsa-miR-18a-3p, hsa-miR-18b-5p, hsa-miR-146b-3p, hsa-miR-19b-3p, and hsa-miR-19a-3p. Following estimation via RT-PCR, hsa-miR-346, hsa-miR-18a-3p, and hsa-miR-324-3p showed consistent patterns in A549 NC and A549 shlncH19. An miRNA inhibitor was transfected into A549 NC and A549 shlncH19 cells, and the expression levels were determined via RT-PCR. hsa-miR-324-3p was inhibited the most compared with hsa-miR-346 and hsa-miR-18a-3p and was subjected to further functional studies. RT-PCR, ELISA, and western blotting for inflammatory gene detection were conducted to validate the functions of the target hsa-miR-324-3p. Results Treatment with 1 and 10 μM Dex could effectively attenuate the inflammatory response. During this process, lncH19 expression significantly increased (P < 0.05). Therefore, treatment with 1 μM Dex was used for further study. Under IL-1β treatment with or without Dex, lncH19 inhibition led to an increase in cell proliferation; a decrease in cell apoptosis; an increase in the protein levels of inflammatory genes; phosphorylation of P65, ICAM-1, and VCAM-1; and increase inflammatory cytokines. Prediction of the targets of lncH19 and validation via RT-PCR revealed that miR-346, miR-18a-3p, and miR-324-3p negatively correlate with lncH19. Additionally, Dex increased the lncH19 expression but reduced that of the miRNAs. Among the miRNAs, miR-324-3p was the most markedly downregulated miRNA following treatment of miRNA inhibitors. The MTS assay and cell apoptosis assay showed that the miR-324-3p inhibitor inhibited cell proliferation and induced cell apoptosis, thereby significantly attenuating the inflammatory response, which reversed the effect of lncH19 in regulating cell proliferation and the secretion of inflammatory cytokines (P < 0.05). Therefore, lncH19 might regulate miR-324-3p in pulmonary inflammatory response under Dex treatment. Conclusion Dex can attenuate the pulmonary inflammatory response by regulating the lncH19/miR-324-3p cascade.
Collapse
Affiliation(s)
- Ye Chen
- Department of Pediatric, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, China
| | - Chao Zhang
- Department of Pediatric Intensive Care Unit, Children's Hospital of Chongqing Medical University, No. 136 Zhongshan two road Yuzhong district, Chongqing, 400013, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400013, China.,National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400013, China.,China International Science and Technology Cooperation base of Child development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400013, China.,Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, 400013, China
| | - Chang-Xue Xiao
- Department of Pediatric Intensive Care Unit, Children's Hospital of Chongqing Medical University, No. 136 Zhongshan two road Yuzhong district, Chongqing, 400013, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400013, China.,National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400013, China.,China International Science and Technology Cooperation base of Child development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400013, China.,Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, 400013, China
| | - Xiao-Dong Li
- Department of Neonatology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, China
| | - Zhi-Li Hu
- Department of Pediatric, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, China
| | - Shou-di He
- Traditional Chinese Medicine Department of Rheumatism, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, China
| | - Xiao-Jun Xiao
- Department of Medicine, Research Center of Allergy & Immunology, Shenzhen University, Shenzhen, 518055, China
| | - Feng Xu
- Department of Pediatric Intensive Care Unit, Children's Hospital of Chongqing Medical University, No. 136 Zhongshan two road Yuzhong district, Chongqing, 400013, China. .,Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400013, China. .,National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400013, China. .,China International Science and Technology Cooperation base of Child development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400013, China. .,Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, 400013, China.
| |
Collapse
|
26
|
Abstract
NF-κB signaling pathway has important roles in the regulation of growth and development of nervous system. This pathway has also been shown to participate in the pathogenesis of schizophrenia. Meanwhile, activity of NF-κB signaling pathway is regulated by several factors including non-coding RNAs (lncRNAs). In the current study, we evaluated expression of nine NF-κB-related lncRNAs namely DILC, ANRIL, PACER, CHAST, ADINR, DICER1-AS1, HNF1A-AS1, H19 and NKILA as well as two mRNA coding genes namely ATG5 and CEBPA in the peripheral blood of patients with schizophrenia compared with matched healthy subjects. Expressions of these genes were assessed by real time PCR technique. Expression of PACER was lower in patients with schizophrenia compared with controls (Posterior beta = − 0.684, P value = 0.049). On the other hand, expressions of CHAST, CEBPA, H19, HNF1A-AS1 and DICER1-AS1 were higher in patients compared with controls (Posterior beta = 0.39, P value = 0.005; Posterior beta = 0.844, P value < 0.0001; Posterior beta = 0.467, P value < 0.0001; Posterior beta = 1.107, P value = 0.005; Posterior beta = 0.176, P value = 0.044, respectively). We also appraised the diagnostic power of transcript quantities of CHAST, CEBPA, DICER1-AS1, H19 and HNF1A-AS1 in distinguishing between patients with schizophrenia and controls through depicting ROC curves. Based on the area under curve (AUC) values, CEBPA had the best diagnostic power (AUC = 0.948, P < 0.0001), followed by H19 (AUC = 0.815, P < 0.0001). Taken together, our study demonstrated dysregulation of NF-κB-related lncRNAs and genes in the peripheral blood of patients with schizophrenia and their potential as peripheral markers for this psychiatric condition.
Collapse
|
27
|
Safa A, Gholipour M, Dinger ME, Taheri M, Ghafouri-Fard S. The critical roles of lncRNAs in the pathogenesis of melanoma. Exp Mol Pathol 2020; 117:104558. [PMID: 33096077 DOI: 10.1016/j.yexmp.2020.104558] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/12/2020] [Accepted: 10/17/2020] [Indexed: 12/14/2022]
Abstract
Long non-coding RNAs (lncRNAs) embrace a huge fraction of human transcripts and participate in the pathogenesis of human disorders especially malignant conditions. Malignant melanoma, as the most fatal type of cutaneous malignnacies, is associated with dysregulation of several lncRNAs including PVT1, H19, MALAT1, and CCAT1. Moreover, a portion of lncRNAs are exclusively expressed in melanoma cell lines. Expression levels of several lncRNAs are associated with TNM stage, tumor size and progression of melanoma. Thus, these lncRNAs are regarded as biomarkers for this malignancy. Peripheral transcript levels of a number of lncRNAs, such as PVT1, SNHG5 and SPRY4-IT1, could distinguish melanoma patients from unaffected persons with appropriate sensitivity and specificity values. Moreover, expression levels of numerous lncRNAs in tissue biopsies could differentiate malignant samples from benign samples. Based on the results of both cell line and in vivo studies, lncRNAs regulate critical pathways in the carcinogenesis of melanoma, such as the PI3K/Akt and NF-κB signaling pathways, and are involved in the modulation of response to chemotherapeutic agents. Here we review the existing information on the role of lncRNAs in malignant melanoma.
Collapse
Affiliation(s)
- Amin Safa
- Institute of Research and Development, Duy Tan University, Da Nang 550000, Viet Nam
| | - Mahdi Gholipour
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Marcel E Dinger
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, 2052 Sydney, NSW, Australia
| | - Mohammad Taheri
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
28
|
Shek D, Read SA, Akhuba L, Qiao L, Gao B, Nagrial A, Carlino MS, Ahlenstiel G. Non-coding RNA and immune-checkpoint inhibitors: friends or foes? Immunotherapy 2020; 12:513-529. [DOI: 10.2217/imt-2019-0204] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Non-coding RNAs (ncRNAs) are an abundant component of the human transcriptome. Their biological role, however, remains incompletely understood. Nevertheless, ncRNAs are highly associated with cancer development and progression due to their ability to modulate gene expression, protein translation and growth pathways. Immune-checkpoint inhibitors (ICIs) are considered one of the most promising and highly effective therapeutic approaches for cancer treatment. ICIs are monoclonal antibodies targeting immune checkpoints such as CTLA-4, PD-1 and PD-L1 signalling pathways that stimulate T cell cytotoxicity and can result in tumor growth suppression. This Review will summarize existing knowledge regarding ncRNAs and their role in cancer and ICI therapy. In addition, we will discuss potential mechanisms by which ncRNAs may influence ICI treatment outcomes.
Collapse
Affiliation(s)
- Dmitrii Shek
- Blacktown Clinical School & Research Centre, Western Sydney University, Sydney, NSW, Australia
- Accreditation Centre, RUDN University, Moscow, Russia
| | - Scott A Read
- Blacktown Clinical School & Research Centre, Western Sydney University, Sydney, NSW, Australia
- Storr Liver Centre, Westmead Institute for Medical Research, University of Sydney, Sydney, NSW, Australia
- Blacktown Hospital, Sydney, NSW, Australia
| | - Liia Akhuba
- Accreditation Centre, RUDN University, Moscow, Russia
| | - Liang Qiao
- Storr Liver Centre, Westmead Institute for Medical Research, University of Sydney, Sydney, NSW, Australia
- Westmead Hospital & Westmead Clinical School, University of Sydney, Sydney, NSW, Australia
| | - Bo Gao
- Westmead Hospital & Westmead Clinical School, University of Sydney, Sydney, NSW, Australia
- Blacktown Hospital, Sydney, NSW, Australia
| | - Adnan Nagrial
- Westmead Hospital & Westmead Clinical School, University of Sydney, Sydney, NSW, Australia
- Blacktown Hospital, Sydney, NSW, Australia
| | - Matteo S Carlino
- Westmead Hospital & Westmead Clinical School, University of Sydney, Sydney, NSW, Australia
- Melanoma Institute Australia, Sydney, NSW, Australia
- Blacktown Hospital, Sydney, NSW, Australia
| | - Golo Ahlenstiel
- Blacktown Clinical School & Research Centre, Western Sydney University, Sydney, NSW, Australia
- Storr Liver Centre, Westmead Institute for Medical Research, University of Sydney, Sydney, NSW, Australia
- Blacktown Hospital, Sydney, NSW, Australia
| |
Collapse
|
29
|
Li J, Zou J, Wan X, Sun C, Chu Z, Hu Y. Roles of noncoding RNAs in drug resistance in multiple myeloma. J Cell Physiol 2020; 235:7681-7695. [PMID: 32324301 DOI: 10.1002/jcp.29726] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 04/08/2020] [Accepted: 04/10/2020] [Indexed: 12/13/2022]
Abstract
Despite the administration of new effective drugs in recent years, relapse and drug resistance are still the main obstacles in multiple myeloma (MM) treatment, making MM an incurable disease. To overcome drug resistance in MM, it is critical to understand the underlying mechanisms of malfunctioning gene expression and develop novel targeted therapies. During the past few decades, with the discovery and characterization of noncoding RNAs (ncRNAs), the landscape of dysregulated ncRNAs of cancers as well as their biological and pathobiological functions in tumorigenesis and drug resistance have been recognized. Studies about ncRNAs improved the understanding of variations of drug response among individuals at a level distinguished from genetic polymorphism, and provided with new orientations for targeted therapies. In this review, we will summarize the emerging impact and underlying molecular mechanisms of the most relevant classes of ncRNAs in drug resistance of MM, and discuss the potential as well as strategies of treating ncRNAs as therapeutic targets.
Collapse
Affiliation(s)
- Jingwen Li
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Zou
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyue Wan
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chunyan Sun
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan, China
| | - Zhangbo Chu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
30
|
Lu Q, Xu W, Liu L, Zhou X, Ye L, Song D, Zhang L, Huang D. Traumatic compressive stress inhibits osteoblast differentiation through long chain non-coding RNA Dancr. J Periodontol 2020; 91:1532-1540. [PMID: 32160313 DOI: 10.1002/jper.19-0648] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 01/02/2020] [Accepted: 02/16/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND Occlusal trauma is an important local contributing factor aggravating periodontal pocket and alveolar bone absorption in periodontal diseases. Our previous studies have found that occlusal trauma inhibited osteogenic differentiation through nuclear factor (NF)-κB signaling. To further investigate the underlying mechanism, the aim of this study was to explore the role of long chain non-coding differentiation antagonizing non-protein coding RNA (Dancr) in the inhibitory effect of traumatic stress on osteoblast differentiation. METHODS We took the MC3T3-E1 cells as object in vitro research and stimulated cells with simple stress load, Dancr-siRNA + stress load, Dancr overexpression-plasmid + stress load. Quantitative real-time polymerase chain reaction was used to detect the RNA expression levels of Dancr, alkaline phosphatase (Alp) and Runt-related transcription factor 2 (Runx2). The protein expressions of Alp and Runx2 were tested by Western blot and the activity of Alp was qualitatively demonstrated by Alp staining. In addition, Western blot was performed to investigate the role of Dancr in affecting NF-κB signaling pathway. RESULTS Traumatic compressive stress inhibited the expressions of Alp, Runx2, andDancr in MC3T3-E1 cells. Stress-induced inhibition of osteoblast differentiation was promoted after silencing Dancr. Overexpression of Dancr could alleviate the inhibitory effect of traumatic force on osteoblast differentiation to some extent. Furthermore, NF-κB signaling was activated after silencing Dancr, and the activated effect of traumatic force on NF-κB signaling could be alleviated through overexpression of Dancr to some extent. CONCLUSION Traumatic compressive stress can indirectly activate the NF-κB signaling through downregulation of Dancr, thereby inhibiting osteogenic differentiation.
Collapse
Affiliation(s)
- Qian Lu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, Sichuan, China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Weizhe Xu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, Sichuan, China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Linyi Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, Sichuan, China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, Sichuan, China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Ling Ye
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, Sichuan, China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Dongzhe Song
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, Sichuan, China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Lan Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, Sichuan, China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Dingming Huang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, Sichuan, China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
31
|
Long noncoding RNA atlas of the inflammation caused by asthma in mice. Arch Pharm Res 2020; 43:421-432. [PMID: 32222886 DOI: 10.1007/s12272-020-01223-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 02/29/2020] [Indexed: 10/24/2022]
Abstract
There is little evidence regarding the roles of long noncoding RNAs (lncRNAs) in inflammation caused by asthma. In this study, we successfully generated an asthma mouse model that was induced by ovalbumin (OVA). The effects of dexamethasone (Dex) treatment on lung tissue were investigated using pathological and biochemical methods, including Diff-Quik staining, enzyme-linked immunosorbent assay (ELISA), hematoxylin-eosin (H&E) staining, and western blotting (WB). The inflammation was effectively relieved with Dex treatment. High-throughput sequencing revealed that a total of 1490 lncRNAs were detected in lung tissue samples. Differential expression analysis revealed that the Dex group had 20 upregulated and 15 downregulated lncRNAs compared with those in the Model group. Moreover, nine differentially expressed and inflammation-related lncRNAs were verified by quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR). Furthermore, the regulation networks of these nine lncRNAs, their potential binding microRNA (miRNAs), and the putative target genes showed that these lncRNAs play important roles in the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling pathway. We further identified the expression levels of three potential binding miRNAs by qRT-PCR. The results of this study contribute to a better understanding of the functions of lncRNAs in inflammation caused by asthma.
Collapse
|
32
|
Ronchetti D, Todoerti K, Vinci C, Favasuli V, Agnelli L, Manzoni M, Pelizzoni F, Chiaramonte R, Platonova N, Giuliani N, Tassone P, Amodio N, Neri A, Taiana E. Expression Pattern and Biological Significance of the lncRNA ST3GAL6-AS1 in Multiple Myeloma. Cancers (Basel) 2020; 12:cancers12040782. [PMID: 32218309 PMCID: PMC7225964 DOI: 10.3390/cancers12040782] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/16/2020] [Accepted: 03/23/2020] [Indexed: 11/16/2022] Open
Abstract
The biological impact of long non-coding RNAs (lncRNAs) in multiple myeloma (MM) is becoming an important aspect of investigation, which may contribute to the understanding of the complex pathobiology of the disease whilst also providing novel potential therapeutic targets. Herein, we investigated the expression pattern and the biological significance of the lncRNA ST3 beta-galactoside alpha-2,3 sialyltransferase 6 antisense RNA 1 (ST3GAL6-AS1) in MM. We documented a high ST3GAL6-AS1 expression level in MM compared to normal plasma cells (PCs) or other hematological malignancies. Transcriptome analyses of MM PCs from patients included in the CoMMpass database indicated a potential involvement of ST3GAL6-AS1 in MAPK signaling and ubiquitin-mediated proteolysis pathways. ST3GAL6-AS1 silencing by LNA-gapmeR antisense oligonucleotides inhibits cell proliferation and triggers apoptosis in MM cell line. Notably, ST3GAL6-AS1 silencing in vitro displayed the down-regulation of the MAPK pathway and protein ubiquitination. These data suggest that ST3GAL6-AS1 deregulation may play a pathogenetic role in MM by affecting both proliferation pathways and circuits fundamental for PC survival. However, ST3GAL6-AS1 expression levels seem not to be significantly associated with clinical outcome and its targeting appears to exert antagonistic effects with proteasome inhibitors used in MM. These findings strongly urge the need for further studies investigating the relevance of ST3GAL6-AS1 in MM.
Collapse
Affiliation(s)
- Domenica Ronchetti
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (D.R.); (C.V.); (V.F.); (L.A.); (M.M.); (E.T.)
- Hematology, Fondazione Cà Granda IRCCS Policlinico, 20122 Milan, Italy; (K.T.); (F.P.)
| | - Katia Todoerti
- Hematology, Fondazione Cà Granda IRCCS Policlinico, 20122 Milan, Italy; (K.T.); (F.P.)
| | - Cristina Vinci
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (D.R.); (C.V.); (V.F.); (L.A.); (M.M.); (E.T.)
| | - Vanessa Favasuli
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (D.R.); (C.V.); (V.F.); (L.A.); (M.M.); (E.T.)
- Hematology, Fondazione Cà Granda IRCCS Policlinico, 20122 Milan, Italy; (K.T.); (F.P.)
| | - Luca Agnelli
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (D.R.); (C.V.); (V.F.); (L.A.); (M.M.); (E.T.)
| | - Martina Manzoni
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (D.R.); (C.V.); (V.F.); (L.A.); (M.M.); (E.T.)
- Hematology, Fondazione Cà Granda IRCCS Policlinico, 20122 Milan, Italy; (K.T.); (F.P.)
| | - Francesca Pelizzoni
- Hematology, Fondazione Cà Granda IRCCS Policlinico, 20122 Milan, Italy; (K.T.); (F.P.)
| | - Raffaella Chiaramonte
- Department of Health Sciences, University of Milan, 20142 Milan, Italy; (R.C.); (N.P.)
| | - Natalia Platonova
- Department of Health Sciences, University of Milan, 20142 Milan, Italy; (R.C.); (N.P.)
| | - Nicola Giuliani
- Hematology and BMT Unit, Azienda Ospedaliero-Universitaria di Parma, 43125 Parma, Italy;
| | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (P.T.); (N.A.)
| | - Nicola Amodio
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (P.T.); (N.A.)
| | - Antonino Neri
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (D.R.); (C.V.); (V.F.); (L.A.); (M.M.); (E.T.)
- Hematology, Fondazione Cà Granda IRCCS Policlinico, 20122 Milan, Italy; (K.T.); (F.P.)
- Correspondence: ; Tel.: +39-02-5032-0420; Fax: +39-02-5032-0403
| | - Elisa Taiana
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (D.R.); (C.V.); (V.F.); (L.A.); (M.M.); (E.T.)
- Hematology, Fondazione Cà Granda IRCCS Policlinico, 20122 Milan, Italy; (K.T.); (F.P.)
| |
Collapse
|
33
|
The Non-Coding RNA Landscape of Plasma Cell Dyscrasias. Cancers (Basel) 2020; 12:cancers12020320. [PMID: 32019064 PMCID: PMC7072200 DOI: 10.3390/cancers12020320] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 01/22/2020] [Accepted: 01/23/2020] [Indexed: 12/14/2022] Open
Abstract
Despite substantial advancements have been done in the understanding of the pathogenesis of plasma cell (PC) disorders, these malignancies remain hard-to-treat. The discovery and subsequent characterization of non-coding transcripts, which include several members with diverse length and mode of action, has unraveled novel mechanisms of gene expression regulation often malfunctioning in cancer. Increasing evidence indicates that such non-coding molecules also feature in the pathobiology of PC dyscrasias, where they are endowed with strong therapeutic and/or prognostic potential. In this review, we aim to summarize the most relevant findings on the biological and clinical features of the non-coding RNA landscape of malignant PCs, with major focus on multiple myeloma. The most relevant classes of non-coding RNAs will be examined, along with the mechanisms accounting for their dysregulation and the recent strategies used for their targeting in PC dyscrasias. It is hoped these insights may lead to clinical applications of non-coding RNA molecules as biomarkers or therapeutic targets/agents in the near future.
Collapse
|
34
|
Long Noncoding RNA H19 Promotes Tumorigenesis of Multiple Myeloma by Activating BRD4 Signaling by Targeting MicroRNA 152-3p. Mol Cell Biol 2020; 40:MCB.00382-19. [PMID: 31712391 DOI: 10.1128/mcb.00382-19] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 11/02/2019] [Indexed: 12/17/2022] Open
Abstract
Multiple myeloma (MM) accounts for over twenty percent of hematological cancer-related death worldwide. Long noncoding RNA (lncRNA) H19 is associated with multiple tumorigenesis and is increased in MM, but the underlying mechanism of H19 in MM is unclear. In this study, the expression of H19, microRNA 152-3p (miR-152-3p), and BRD4 in MM patients was evaluated by quantitative real-time PCR (qRT-PCR) and Western blotting. Colony formation and flow cytometry analysis were used to determine the effects of H19 and miR-152-3p on MM cell proliferation, apoptosis, and cell cycle. A luciferase reporter assay was conducted to confirm the interaction among H19, miR-152-3p, and BRD4. A nude mouse xenograft model was established, and the cell proliferation and apoptosis were evaluated by immunohistochemistry (IHC) staining and terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick end labeling assay. We found that levels of H19 and BRD4 were upregulated and the expression of miR-152-3p was downregulated in MM patients. Dual luciferase reporter assay showed H19 targeted miR-152-3p to promote BRD4 expression. Knockdown of H19 repressed proliferation and enhanced apoptosis and cell cycle G1 arrest by upregulating miR-152-3p in MM cells. Furthermore, H19 knockdown suppressed the growth of xenograft tumor, reduced Ki-67 and BRD4 levels, and increased cell apoptosis in xenograft tumor tissues. Taking these results together, H19 knockdown suppresses MM tumorigenesis via inhibiting BRD4-mediated cell proliferation through targeting miR-152-3p, implying that H19 is a promising biomarker and drug target for MM.
Collapse
|
35
|
Targeting of IL-6-Relevant Long Noncoding RNA Profiles in Inflammatory and Tumorous Disease. Inflammation 2020; 42:1139-1146. [PMID: 30825076 DOI: 10.1007/s10753-019-00995-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Interleukin-6 (IL-6) is a critical cytokine with a diverse repertoire of physiological functions. Dysregulation of IL-6 signaling is associated with inflammatory disorders as well as cancers. However, blockade of IL-6 activity via antibodies directed against the IL-6 signaling pathway may compromise the efficacy of the immune system; therefore, patients may not have a uniformly satisfactory response to treatment. Long noncoding RNAs (lncRNAs) have been discovered to be evolutionary conserved transcripts of noncoding DNA sequences and have emerged as biomarkers with great predictive and prognostic value, further employed as a targeted anticancer therapy. LncRNAs have been recently implicated in the regulation of IL-6-related signaling and function; they are tightly linked to the development of a range of IL-6 dysregulated diseases. Here, we will highlight those lncRNAs involved in IL-6 signaling, with an emphasis on the mechanisms of lncRNAs that interact with IL-6. Targeting of such lncRNAs related to IL-6 regulation could be, in the near future, a promising therapeutic strategy in the treatment of inflammatory- and tumor-related diseases.
Collapse
|
36
|
Zhu FX, Wang XT, Ye ZZ, Gan ZP, Lai YR. Construction of a prognosis‑associated long noncoding RNA‑mRNA network for multiple myeloma based on microarray and bioinformatics analysis. Mol Med Rep 2020; 21:999-1010. [PMID: 32016443 PMCID: PMC7003030 DOI: 10.3892/mmr.2020.10930] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 04/10/2019] [Indexed: 02/07/2023] Open
Abstract
At present, the association between prognosis-associated long noncoding RNAs (lncRNAs) and mRNAs is yet to be reported in multiple myeloma (MM). The aim of the present study was to construct prognostic models with lncRNAs and mRNAs, and to map the interactions between these lncRNAs and mRNAs in MM. LncRNA and mRNA data from 559 patients with MM were acquired from the Genome Expression Omnibus (dataset GSE24080), and their prognostic values were calculated using the survival package in R. Multivariate Cox analysis was used on the top 20 most significant prognosis-associated mRNAs and lncRNAs to develop prognostic signatures. The performances of these prognostic signatures were tested using the survivalROC package in R, which allows for time-dependent receiver operator characteristic (ROC) curve estimation. Weighted correlation network analysis (WGCNA) was conducted to investigate the associations between lncRNAs and mRNAs, and a lncRNA-mRNA network was constructed using Cytoscape software. Univariate Cox regression analysis identified 39 lncRNAs and 1,445 mRNAs that were significantly associated with event-free survival of MM patients. The top 20 most significant survival-associated lncRNAs and mRNAs were selected as candidates for analyzing independent MM prognostic factors. Both signatures could be used to separate patients into two groups with distinct outcomes. The areas under the ROC curves were 0.739 for the lncRNA signature and 0.732 for the mRNA signature. In the lncRNA-mRNA network, a total of 143 mRNAs were positively or negatively associated with 23 prognosis-associated lncRNAs. NCRNA00201, LOC115110 and RP5-968J1.1 were the most dominant drivers. The present study constructed a model that predicted prognosis in MM and formed a network with the corresponding prognosis-associated mRNAs, providing a novel perspective for the clinical diagnosis and treatment of MM, and suggesting novel directions for interpreting the mechanisms underlying the development of MM.
Collapse
Affiliation(s)
- Fang-Xiao Zhu
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
| | - Xiao-Tao Wang
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
| | - Zhi-Zhong Ye
- Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, Guangdong 518040, P.R. China
| | - Zhao-Ping Gan
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yong-Rong Lai
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
37
|
Ghafouri-Fard S, Esmaeili M, Taheri M. H19 lncRNA: Roles in tumorigenesis. Biomed Pharmacother 2019; 123:109774. [PMID: 31855739 DOI: 10.1016/j.biopha.2019.109774] [Citation(s) in RCA: 179] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 11/23/2019] [Accepted: 12/04/2019] [Indexed: 12/12/2022] Open
Abstract
H19 is a long non-coding RNA [lncRNA] which was firstly described as an oncofetal transcript. The imprinted gene is normally expressed from the maternal allele. However, this pattern of imprinting is dysregulated in several cancers leading to aberrant up-regulation of H19 in malignant tissues. Several studies have utilized this aberrant expression pattern to find specific biomarkers for detection of cancer in tumoral tissues or peripheral blood. Moreover, single nucleotide polymorphisms within H19 have been associated with risk of oral squamous cell carcinoma, hepatocellular carcinoma, breast cancer, bladder cancer, gastric cancer and colorectal cancer. Taken together, H19 is regarded as a biomarker for cancer and a putative therapeutic target in these human disorders.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammadhosein Esmaeili
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Mohammad Taheri
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
38
|
Isodeoxyelephantopin, a Sesquiterpene Lactone Induces ROS Generation, Suppresses NF-κB Activation, Modulates LncRNA Expression and Exhibit Activities Against Breast Cancer. Sci Rep 2019; 9:17980. [PMID: 31784542 PMCID: PMC6884568 DOI: 10.1038/s41598-019-52971-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 10/24/2019] [Indexed: 01/23/2023] Open
Abstract
The sesquiterpene lactones, Isodeoxyelephantopin (IDET) and Deoxyelephantopin (DET) are known to exhibit activities against some cancer types. The activities of these lactones against breast cancer and the molecular bases is not known. We examined the efficacy of lactones in breast cancer preclinical model. Although both lactones exhibited drug like properties, IDET was relatively effective in comparison to DET. IDET suppressed the proliferation of both invasive and non-invasive breast cancer cell lines. IDET also suppressed the colony formation and migration of breast cancer cells. The assays for Acridine Orange (AO)/Propidium Iodide (PI) staining, cell cycle distribution, phosphatidylserine externalization and DNA laddering suggested the apoptosis inducing potential of IDET. The treatment with IDET also induced an accumulation of cells in the sub-G1 and G2/M phases. The exposure of breast cancer cells to the lactone was associated with a depolarization in mitochondrial membrane potential, and cleavage of caspase and PARP. The lactone induced reactive oxygen species (ROS) generation in breast cancer cells. Further, the use of N-acetyl cysteine (NAC) suppressed IDET induced ROS generation and apoptosis. The NF-κB-p65 nuclear translocation induced by okadaic acid (OA) was suppressed by the sesquiterpene. IDET also suppressed the expression of NF-κB regulated tumorigenic proteins, and induced the expression of proapoptotic gene (Bax) in cancer cells. While the expression of oncogenic lncRNAs was suppressed, the tumor suppressor lncRNAs were induced by the sesquiterpene. Collectively, the modulation of multiple cell signaling molecules by IDET may contribute to its activities in breast cancer cells.
Collapse
|
39
|
Gupta SC, Awasthee N, Rai V, Chava S, Gunda V, Challagundla KB. Long non-coding RNAs and nuclear factor-κB crosstalk in cancer and other human diseases. Biochim Biophys Acta Rev Cancer 2019; 1873:188316. [PMID: 31639408 DOI: 10.1016/j.bbcan.2019.188316] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 09/23/2019] [Accepted: 09/23/2019] [Indexed: 12/29/2022]
Abstract
The regulation of the pleiotropic transcription factor, nuclear factor-κB (NF-κB) by miRNAs and proteins is extensively studied. More recently, the NF-κB signaling was also reported to be regulated by several long non-coding RNAs (lncRNAs) that constitute the major portion of the noncoding component of the human genome. The common NF-κB associated lncRNAs include NKILA, HOTAIR, MALAT1, ANRIL, Lethe, MIR31HG, and PACER. The lncRNA and NF-κB signaling crosstalk during cancer and other diseases such as cardiomyopathy, celiac disease, cerebral infarction, chronic kidney disease, diabetes mellitus, Kawasaki disease, pregnancy loss, and rheumatoid arthritis. Some NF-κB related lncRNAs can affect gene expression without modulating NF-κB signaling. Most of the lncRNAs with a potential to modulate NF-κB signaling are regulated by NF-κB itself suggesting a feedback regulation. The discovery of lncRNAs have provided a new type of regulation for the NF-κB signaling and thus could be explored for therapeutic interventions. The manner in which lncRNA and NF-κB crosstalk affects human pathophysiology is discussed in this review. The challenges associated with the therapeutic interventions of this crosstalk are also discussed.
Collapse
Affiliation(s)
- Subash C Gupta
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India.
| | - Nikee Awasthee
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India
| | - Vipin Rai
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India
| | - Srinivas Chava
- Department of Biochemistry & Molecular Biology, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Venugopal Gunda
- Pediatric Oncology Laboratory, Child Health Research Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Kishore B Challagundla
- Department of Biochemistry & Molecular Biology, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
40
|
Huang LJ, Shen Y, Bai J, Wang FX, Feng YD, Chen HL, Peng Y, Zhang R, Li FM, Zhang PH, Lei XR, Xue F, Ma YP, Hu JS, He AL. High Expression Levels of Long Noncoding RNA Small Nucleolar RNA Host Gene 18 and Semaphorin 5A Indicate Poor Prognosis in Multiple Myeloma. Acta Haematol 2019; 143:279-288. [PMID: 31597158 DOI: 10.1159/000502404] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 07/29/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND The aim of this study was to detect the expression of long noncoding RNA small nucleolar RNA host gene 18 (SNHG18) andsemaphorin 5A (SEMA5A) genes in multiple myeloma (MM) patients and to explore the correlation of the expression of these genes with the clinical characteristics and prognosis of MM patients. METHODS Forty-seven newly diagnosed MM, 18 complete remission MM, 13 refractory/relapse MM, and 22 iron deficiency anemia (serving as control) samples were extracted at the Department of Hematology, Second Affiliated Hospital of Xian Jiaotong University between January 2015 and December 2016. The clinical features of the MM patients are summarized. Real-time quantitative PCR was performed to analyze the relative expression levels of the SNHG18 and SEMA5Agenes. The clinical characteristics and overall survival (OS) of the MM patients were statistically analyzed while measuring different levels of SNHG18 and SEMA5Agene expression. At the same time, the correlation between the expression of SNHG18 and SEMA5A was also analyzed. RESULTS The analysis confirmed that SNHG18 and its possible target gene SEMA5A were both highly expressed in newly diagnosed MM patients. After analyzing the clinical significance of SNHG18 and SEMA5A in MM patients, we found that the expression of SNHG18 and SEMA5A was related to the Durie-Salmon (DS), International Staging System (ISS), and Revised International Staging System (R-ISS) classification systems, and the Mayo Clinic Risk Stratification for Multiple Myeloma (mSMART; p < 0.05). Moreover, we observed a significant difference in OS between the SNHG18/SEMA5A high expression group and the low expression group. We found a positive correlation between SNHG18 and SEMA5A expression (r = 0.709, p < 0.01). Surprisingly, the expected median OS times of both the SNHG18 and SEMA5Ahigh expression groups were significantly decreased, which was in contrast to those of both the SNHG18 and SEMA5Alow expression groups and the single-gene high expression group (p < 0.05). CONCLUSION High expression of both SNHG18 and SEMA5A is associated with poor prognosis in patients with MM.
Collapse
Affiliation(s)
- Ling-Juan Huang
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of General Medicine, The First Affiliated Hospital of Xi'an Medical University, Xi'an, China
- School of General Medicine, Xi'an Medical University, Xi'an, China
| | - Ying Shen
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ju Bai
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Fang-Xia Wang
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yuan-Dong Feng
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hong-Li Chen
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yue Peng
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ru Zhang
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Fang-Mei Li
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Pei-Hua Zhang
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiao-Ru Lei
- Institute of Hematology, Xi'an Central Hospital, Xi'an, China
| | - Feng Xue
- Department of Hematology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Yan-Ping Ma
- Department of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Jin-Song Hu
- Department of Genetics and Molecular Biology, Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Ai-Li He
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China,
| |
Collapse
|
41
|
Jiang H, Wu FR, Liu J, Qin XJ, Jiang NN, Li WP. Effect of astragalosides on long non-coding RNA expression profiles in rats with adjuvant-induced arthritis. Int J Mol Med 2019; 44:1344-1356. [PMID: 31364738 PMCID: PMC6713426 DOI: 10.3892/ijmm.2019.4281] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 06/12/2019] [Indexed: 11/06/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease of unknown etiology, which occurs in ~1.0% of the general population. Increasing studies have suggested that long non-coding RNAs (lncRNAs) may serve important roles in various biological processes and may be associated with the pathogenesis of different types of disease, including RA. Astragalosides (AST) has been used as a traditional Chinese medicine for the treatment of RA. However, the mechanism underlying its therapeutic effect has remained unclear to date. Thus, there is an urgent need to elucidate the possible mechanism of AST in the treatment of RA from the perspective of lncRNAs. In the present study, the lncRNAs and mRNAs of a vehicle group, animal model group and AST treatment (control) group were determined by Arraystar Rat lncRNA/mRNA microarray. The differentially expressed genes with a fold change >1.5 and P<0.05 were selected and analyzed. Gene Ontology (GO) and pathway analysis was performed using the Database for Annotation, Visualization and Integration Discovery, and the coding-non-coding gene co-expression network was drawn based on the correlation analysis between the differentially expressed lncRNAs and mRNAs. Based on node degree and the correlation between bioinformatics analysis and RA, the critical differentially expressed lncRNAs were selected, analyzed and verified by reverse transcription-quantitative PCR (RT-qPCR) analysis. The results showed that, following AST treatment, up to 75 lncRNAs and 247 mRNAs were found to be differentially expressed among the three groups. GO and pathway analysis manifested that 135 GO terms and 17 pathways were enriched by differentially expressed genes. Four lncRNAs (MRAK012530, MRAK132628, MRAK003448 and XR_006457) were selected as the critical lncRNAs and their trend in expression showed consistency between the RT-qPCR and microarray data. In conclusion, AST had a regulatory effect on differentially expressed lncRNAs during the development of RA, and four lncRNAs could be selected as critical therapeutic targets of AST.
Collapse
Affiliation(s)
- Hui Jiang
- Experimental Center of Clinical Research, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui 230031, P.R. China
| | - Fu-Rong Wu
- Department of Pharmacy, Anhui Provincial Hospital, Hefei, Anhui 230001, P.R. China
| | - Jian Liu
- Experimental Center of Clinical Research, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui 230031, P.R. China
| | - Xiu-Juan Qin
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui 230031, P.R. China
| | - Nan-Nan Jiang
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui 230031, P.R. China
| | - Wei-Ping Li
- Department of Pharmacology, College of Basic Medicine, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
42
|
Yuan Z, Bian Y, Ma X, Tang Z, Chen N, Shen M. LncRNA H19 Knockdown in Human Amniotic Mesenchymal Stem Cells Suppresses Angiogenesis by Associating with EZH2 and Activating Vasohibin-1. Stem Cells Dev 2019; 28:781-790. [PMID: 30938218 DOI: 10.1089/scd.2019.0014] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Human amniotic mesenchymal stem cells (HAMSCs) are promising seed cells with great advantages in promoting angiogenesis. However, the mechanisms underlying angiogenesis facilitated by HAMSCs are still unclear. Long noncoding RNA H19 is involved in many biological processes, such as enhancing angiogenesis and proliferation, invasion, and migration of cancer cells. In this study, we constructed HAMSCs of stable low-expression H19 (HAMSC-shH19) and the scramble control (HAMSC-shNC) using lentiviral vectors, and in a three-dimensional coculture with human umbilical vein endothelial cells (HUVECs) to investigate the effect of H19 knockdown in HAMSCs on angiogenesis. Our results demonstrated that H19 knockdown significantly inhibited the angiogenic function of HAMSCs at an early stage in vitro and in vivo. The results of CCK8 and transwell assays demonstrated that the conditioned medium secreted by HAMSCs reduced proliferation and migration of HUVECs after downregulating H19. The angiogenesis factors expressed and secreted by HAMSC-shH19 were decreased compared with those secreted by the control, while angiogenesis inhibitors were elevated. Furthermore, we conducted chromatin immunoprecipitation and RNA-binding protein immunoprecipitation assays and found that H19 could interact with the histone methyltransferase Enhancer of Zeste homolog 2 (EZH2) and that H19 knockdown inhibited the ability of EZH2 to recruit methyl groups to the promoter region of the angiogenesis inhibitor gene vasohibin-1 (VASH1), thus increasing VASH1 expression and secretion of HAMSCs, suppressing angiogenesis. In summary, our study identified H19 as an important regulator in HAMSCs for promoting angiogenesis, which would help to construct ideal gene-modified seed cells to enhance angiogenesis in regenerative medicine.
Collapse
Affiliation(s)
- Zhiyao Yuan
- 1 Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yifeng Bian
- 2 Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Xiaojie Ma
- 2 Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Zichun Tang
- 2 Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Ning Chen
- 2 Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,3 The Affiliated Stomatological Hospital of Soochow University, Suzhou Stomatological Hospital, Suzhou, China
| | - Ming Shen
- 2 Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| |
Collapse
|
43
|
Mishra S, Verma SS, Rai V, Awasthee N, Chava S, Hui KM, Kumar AP, Challagundla KB, Sethi G, Gupta SC. Long non-coding RNAs are emerging targets of phytochemicals for cancer and other chronic diseases. Cell Mol Life Sci 2019; 76:1947-1966. [PMID: 30879091 PMCID: PMC7775409 DOI: 10.1007/s00018-019-03053-0] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 02/01/2019] [Accepted: 02/19/2019] [Indexed: 12/18/2022]
Abstract
The long non-coding RNAs (lncRNAs) are the crucial regulators of human chronic diseases. Therefore, approaches such as antisense oligonucleotides, RNAi technology, and small molecule inhibitors have been used for the therapeutic targeting of lncRNAs. During the last decade, phytochemicals and nutraceuticals have been explored for their potential against lncRNAs. The common lncRNAs known to be modulated by phytochemicals include ROR, PVT1, HOTAIR, MALAT1, H19, MEG3, PCAT29, PANDAR, NEAT1, and GAS5. The phytochemicals such as curcumin, resveratrol, sulforaphane, berberine, EGCG, and gambogic acid have been examined against lncRNAs. In some cases, formulation of phytochemicals has also been used. The disease models where phytochemicals have been demonstrated to modulate lncRNAs expression include cancer, rheumatoid arthritis, osteoarthritis, and nonalcoholic fatty liver disease. The regulation of lncRNAs by phytochemicals can affect multi-steps of tumor development. When administered in combination with the conventional drugs, phytochemicals can also produce synergistic effects on lncRNAs leading to the sensitization of cancer cells. Phytochemicals target lncRNAs either directly or indirectly by affecting a wide variety of upstream molecules. However, the potential of phytochemicals against lncRNAs has been demonstrated mostly by preclinical studies in cancer models. How the modulation of lncRNAs by phytochemicals produce therapeutic effects on cancer and other chronic diseases is discussed in this review.
Collapse
Affiliation(s)
- Shruti Mishra
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India
| | - Sumit S Verma
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India
| | - Vipin Rai
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India
| | - Nikee Awasthee
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India
| | - Srinivas Chava
- Department of Biochemistry and Molecular Biology, and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Kam Man Hui
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre, Singapore, 169610, Singapore
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Kishore B Challagundla
- Department of Biochemistry and Molecular Biology, and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
| | - Subash C Gupta
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India.
| |
Collapse
|
44
|
Fu Y, Liu X, Zhang F, Jiang S, Liu J, Luo Y. Bortezomib-inducible long non-coding RNA myocardial infarction associated transcript is an oncogene in multiple myeloma that suppresses miR-29b. Cell Death Dis 2019; 10:319. [PMID: 30967527 PMCID: PMC6456577 DOI: 10.1038/s41419-019-1551-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/23/2019] [Accepted: 03/26/2019] [Indexed: 01/06/2023]
Abstract
Clinical outcomes of patients with multiple myeloma (MM) have almost doubled the overall survival over the last decade owing to the use of proteasome inhibitor such as bortezomib (BTZ). However, some patients with MM develop primary resistance to BTZ, whereas others develop resistance after treatment. In this study, we investigated relationships between BTZ resistance and dysfunction of long non-coding RNAs (lncRNAs) in patients with MM. Bone marrow samples were collected from patients with MM and healthy donors for lncRNA microarray and survival analyses. To investigate functions and underlying mechanisms of lncRNA-mediated BTZ resistance in MM, we performed CCK-8 assays, flow cytometry analyses, dual luciferase report gene assays, and RNA pulldown assays with samples from nude mice carrying tumor xenografts and in clinical samples. Differentially expressed lncRNA myocardial infarction associated transcripts (MIAT) were highly expressed in patients with MM compared with healthy controls, and were predictive of poor survival outcomes. Moreover, MIAT expression was significantly increased in BTZ-resistant patients with MM compared with newly diagnosed patients with MM, and was identified as a BTZ-inducible lncRNA. Specifically, BTZ upregulated MIAT expression through increased stat1 phosphorylation. Silencing of MIAT inhibited MM cell growth and sensitized MM cells to BTZ by negatively regulating miR-29b. Our data demonstrated the utility of MIAT as a tool for overcoming BTZ resistance in patients with MM.
Collapse
Affiliation(s)
- Yunfeng Fu
- The Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Xiao Liu
- The Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Fangrong Zhang
- The Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Siyi Jiang
- The Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Jing Liu
- The Third Xiangya Hospital of Central South University, Changsha, 410013, China.
| | - Yanwei Luo
- The Third Xiangya Hospital of Central South University, Changsha, 410013, China.
| |
Collapse
|
45
|
Pu J, Huang H, Su J, Yuan J, Cong H, Wang X, Ju S. Decreased expression of long noncoding RNA XLOC_013703 promotes cell growth via NF-κB pathway in multiple myeloma. IUBMB Life 2019; 71:1240-1251. [PMID: 30861305 DOI: 10.1002/iub.2029] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 01/25/2019] [Accepted: 02/10/2019] [Indexed: 01/24/2023]
Abstract
Long noncoding RNAs (lncRNAs) are dysregulated in cancer and involved in oncogenic or tumor inhibitory processes. The aim of the study was to investigate the expression pattern of lncRNA XLOC_013703 in multiple myeloma (MM) and to evaluate its biological role and potential significance. We found that XLOC_013703 was significantly decreased in CD138 positive plasma cells and serum of MM patients compared to normal controls, and the decreased XLOC_013703 expression was correlated with β2-MG, serum-free light chain (s-FLC) and revised international staging system. RNA-fluorescence in situ hybridization results revealed that XLOC_013703 was distributed both in the nucleus and in the cytoplasm of MM cells including H929, RPMI8226, and U266. Overexpression of XLOC_013703 inhibited the proliferation of U266 cells and blocked the cell cycle in G1 stage, thus contributing to MM cell apoptosis. By contrast, knockdown of XLOC_013703 promoted the growth of H929 cells. Western blot analysis confirmed that the expression of p-IκBα and nuclear P65 was substantially increased in shRNA transfection groups compared to control groups, whereas overexpression of XLOC_013703 reduced these expressions. In conclusion, we confirmed that the decreased expression of a novel lncRNA, XLOC_013703, in MM. XLOC_013703 was involved in MM cell survival and proliferation via nuclear factor-κB pathway which represents a potential therapeutic target for MM. © 2019 IUBMB Life, 71(9):1240-1251, 2019.
Collapse
Affiliation(s)
- Jiang Pu
- Laboratory Medicine Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Hongming Huang
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Jianyou Su
- Laboratory Medicine Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Jie Yuan
- Laboratory Medicine Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Hui Cong
- Laboratory Medicine Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Xudong Wang
- Laboratory Medicine Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Shaoqing Ju
- Laboratory Medicine Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| |
Collapse
|
46
|
Bai Y, Wang W, Zhang Y, Zhang F, Zhang H. lncRNA MIAT suppression alleviates corneal angiogenesis through regulating miR-1246/ACE. Cell Cycle 2019; 18:661-669. [PMID: 30782069 DOI: 10.1080/15384101.2019.1578143] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Corneal neovascularization (CRNV) is a prevalence eye disorder that affects the transparency and refraction properties of eyes. To explore the correlation between the level of Angiotensin II (Ang II) and corneal angiogenesis, the rat model of CRNV was established using alkali-burn, while the human umbilical vein endothelial cells (HUVECs) were stimulated using VEGF to induce the CRNV cells in vitro. RNA immunoprecipitation (RIP) and RNA pull-down were performed to validate the relationship between MIAT and miR-1246. The expression of MIAT and Ang II was increased, while miR-1246 was decreased in CRNV rat model. VEGF stimulation significantly promoted cell proliferation and migration of HUVECs, knockdown of MIAT dramatically reversed the effects of VEGF, while cells co-transfected with miR-1246 inhibitor obviously abolished the effect of VEGF+si-MIAT, however, enalaprilat abolished the effects of VEGF+si-MIAT+miR-1246 inhibitor. MIAT directly regulated the expression of miR-1246. In conclusion, VEGF stimulation promoted cell proliferation and migration of HUVECs mainly through regulating MIAT/miR-1246/ACE.
Collapse
Affiliation(s)
- Yanhui Bai
- a Department of Ophthalmology , First Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - Weiqun Wang
- a Department of Ophthalmology , First Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - Youmei Zhang
- a Department of Ophthalmology , First Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - Fengyan Zhang
- a Department of Ophthalmology , First Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - Haohao Zhang
- b Division of Endocrinology, Department of Internal Medicine , First Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| |
Collapse
|
47
|
Corrado C, Costa V, Giavaresi G, Calabrese A, Conigliaro A, Alessandro R. Long Non Coding RNA H19: A New Player in Hypoxia-Induced Multiple Myeloma Cell Dissemination. Int J Mol Sci 2019; 20:ijms20040801. [PMID: 30781795 PMCID: PMC6413127 DOI: 10.3390/ijms20040801] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 02/06/2019] [Accepted: 02/11/2019] [Indexed: 12/13/2022] Open
Abstract
The long non-coding RNA H19 (lncH19) is broadly transcribed in the first stage of development and silenced in most cells of an adult organism; it appears again in several tumors where, through different molecular mediators, promotes cell proliferation, motility and metastases. LncH19 has been associated with hypoxia-inducible factor 1-alpha (HIF-1α) activation and, in some tumors, it has proved to be necessary and required to sustain hypoxic responses. Here we propose to investigate a putative role for the lncH19 in hypoxia induced multiple myeloma (MM) progression. Transcriptional analysis of MM cell lines (RPMI and MM1.S) exposed to normoxia or hypoxia (1% O2) was done in order to evaluate lncH19 levels under hypoxic stimulation. Then, to investigate the role of lncH19 in hypoxia mediated MM progression, transcriptional, protein and functional assays have been performed on hypoxia stimulated MM cell lines, silenced or not for lncH19. Our data demonstrated that hypoxic stimulation in MM cell lines induced the overexpression of lncH19, which, in turn, is required for the expression of the hypoxia induced genes involved in MM dissemination, such as C-X-C Motif Chemokine Receptor 4 (CXCR4) and Snail. Moreover, adhesion assays demonstrated that lncH19 silencing abrogates the increased adhesion on stromal cells induced by the hypoxic condition. Finally, Western blot analysis indicated that lncH19 silencing impaired HIF1α nuclear translocation. The LncH19, required for the induction of hypoxic responses in MM cells, could represent a new therapeutic target for MM.
Collapse
Affiliation(s)
- Chiara Corrado
- Department of BioMedicine, Neurosciences and Advanced Diagnostics (Bi.N.D), Via Divisi 83, 90133 Palermo, Italy.
| | - Viviana Costa
- IRCCS ISTITUTO ORTOPEDICO RIZZOLI, 40138 Bologna, Italy.
| | - Gianluca Giavaresi
- IRCCS ISTITUTO ORTOPEDICO RIZZOLI, 40138 Bologna, Italy.
- Laboratory of Preclinical and Surgical Studies, IRCCS ISTITUTO ORTOPEDICO RIZZOLI, 40138 Bologna, Italy.
| | - Annalisa Calabrese
- Department of BioMedicine, Neurosciences and Advanced Diagnostics (Bi.N.D), Via Divisi 83, 90133 Palermo, Italy.
| | - Alice Conigliaro
- Department of BioMedicine, Neurosciences and Advanced Diagnostics (Bi.N.D), Via Divisi 83, 90133 Palermo, Italy.
| | - Riccardo Alessandro
- Department of BioMedicine, Neurosciences and Advanced Diagnostics (Bi.N.D), Via Divisi 83, 90133 Palermo, Italy.
| |
Collapse
|
48
|
Butova R, Vychytilova-Faltejskova P, Souckova A, Sevcikova S, Hajek R. Long Non-Coding RNAs in Multiple Myeloma. Noncoding RNA 2019; 5:E13. [PMID: 30682861 PMCID: PMC6468639 DOI: 10.3390/ncrna5010013] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 01/15/2019] [Accepted: 01/15/2019] [Indexed: 02/07/2023] Open
Abstract
Multiple myeloma (MM) is the second most common hematooncological disease of malignant plasma cells in the bone marrow. While new treatment brought unprecedented increase of survival of patients, MM pathogenesis is yet to be clarified. Increasing evidence of expression of long non-coding RNA molecules (lncRNA) linked to development and progression of many tumors suggested their important role in tumorigenesis. To date, over 15,000 lncRNA molecules characterized by diversity of function and specificity of cell distribution were identified in the human genome. Due to their involvement in proliferation, apoptosis, metabolism, and differentiation, they have a key role in the biological processes and pathogenesis of many diseases, including MM. This review summarizes current knowledge of non-coding RNAs (ncRNA), especially lncRNAs, and their role in MM pathogenesis. Undeniable involvement of lncRNAs in MM development suggests their potential as biomarkers.
Collapse
Affiliation(s)
- Romana Butova
- Babak Myeloma Group, Department of Pathological Physiology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic.
| | | | - Adela Souckova
- Babak Myeloma Group, Department of Pathological Physiology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic.
| | - Sabina Sevcikova
- Babak Myeloma Group, Department of Pathological Physiology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic.
| | - Roman Hajek
- Department of Hematooncology, University Hospital Ostrava and Faculty of Medicine, University Ostrava, 70852 Ostrava, Czech Republic.
| |
Collapse
|
49
|
Awasthee N, Rai V, Verma SS, Sajin Francis K, Nair MS, Gupta SC. Anti-cancer activities of Bharangin against breast cancer: Evidence for the role of NF-κB and lncRNAs. Biochim Biophys Acta Gen Subj 2018; 1862:2738-2749. [DOI: 10.1016/j.bbagen.2018.08.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 08/22/2018] [Accepted: 08/22/2018] [Indexed: 11/27/2022]
|
50
|
High LINC01605 expression predicts poor prognosis and promotes tumor progression via up-regulation of MMP9 in bladder cancer. Biosci Rep 2018; 38:BSR20180562. [PMID: 30054424 PMCID: PMC6123067 DOI: 10.1042/bsr20180562] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 07/02/2018] [Accepted: 07/18/2018] [Indexed: 12/17/2022] Open
Abstract
The advent of high-throughput sequencing methods has facilitated identification of novel long non-coding RNAs (lncRNAs), which have been demonstrated to play an important role in multiple tumors. Moreover, with the assistance of bioinformatics analysis, LINC01605 has been found to be up-regulated in bladder cancer (BC) tissues compared with normal tissues. Hence, the present study was to explore its specific biological role and related mechanism in BC. The relative expression level of LINC01605 was measured in a cohort of BC tissues with matched normal tissues as well as human BC cell lines by quantitative real-time PCR (qRT-PCR). Survival analysis was performed to explore the relationship between LINC01605 expression and the prognosis of BC patients. The biological function of LINC01605 was studied in vitroand in vivo, by means of CCK-8 assay, colony formation assay, transwell assay, and tumor xenografts mice model. LINC01605 was found to be frequently highly expressed in both human BC cells and tissues. Survival analysis indicated that high LINC01605 expression was associated with higher histological grade and clinical stages. In addition, down-regulated LINC01605 in BC cells could significantly inhibit the abilities of proliferation, migration, and invasion in vitro and knockdown of LINC01605 in subcutaneous xenograft tumor model could impede tumorigenesis in vivo. Mechanistically, LINC01605 could activate epithelial–mesenchymal transition (EMT) signaling pathway and promote the expression of matrix metallopeptidase (MMP) 9 (MMP9). In summary, our results shed light on that LINC01605, as a new prognostic biomarker, could promote the proliferation, migration, and invasion of BC cells via activating EMT signaling pathway and up-regulating MMP9 expression.
Collapse
|