1
|
Gill SK, Gomer RH. Translational Regulators in Pulmonary Fibrosis: MicroRNAs, Long Non-Coding RNAs, and Transcript Modifications. Cells 2025; 14:536. [PMID: 40214489 PMCID: PMC11988943 DOI: 10.3390/cells14070536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/27/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025] Open
Abstract
Fibrosing disorders including idiopathic pulmonary fibrosis (IPF) are progressive irreversible diseases, often with poor prognoses, characterized by the accumulation of excessive scar tissue and extracellular matrix. Translational regulation has emerged as a critical aspect of gene expression control, and the dysregulation of key effectors is associated with disease pathogenesis. This review examines the current literature on translational regulators in IPF, focusing on microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and RNA transcript modifications including alternative polyadenylation and chemical modification. Some of these translational regulators potentiate fibrosis, and some of the regulators inhibit fibrosis. In IPF, some of the profibrotic regulators are upregulated, and some of the antifibrotic regulators are downregulated. Correcting these defects in IPF-associated translational regulators could be an intriguing avenue for therapeutics.
Collapse
Affiliation(s)
| | - Richard H. Gomer
- Department of Biology, Texas A&M University, College Station, TX 77843, USA;
| |
Collapse
|
2
|
Rubini A, Zanotti F, Licastro D, Calogero G, Bettini G, Piccoli C, Rubini G, Lovatti L, Zavan B. Therapeutic Potential of Feline Adipose-Derived Stem Cell Exosomes in the Treatment of Feline Idiopathic Cystitis: A Characterization and Functional Analysis of miRNA Content. Nanotheranostics 2025; 9:38-51. [PMID: 39744099 PMCID: PMC11667565 DOI: 10.7150/ntno.99383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 09/28/2024] [Indexed: 04/05/2025] Open
Abstract
Feline Idiopathic Cystitis (FIC), is a chronic lower urinary tract condition in cats analogous to PBS/IC in women, which presents significant treatment challenges due to its idiopathic nature. Recent advancements in regenerative medicine highlight the potential of Adipose Tissue-Derived Stem Cells (ADSCs), particularly through their secretome, which includes mediators, bioactive molecules, and extracellular vesicles (EVs). Notably, exosomes, a subset of EVs, facilitate cell-to-cell communication and, when derived from ADSCs, exhibit anti-inflammatory properties and contribute to tissue regeneration. In this work, we aim to characterize the content of exosomes derived from feline ADSCs (fADSCs) to elucidate their mechanisms of action on recipient cells and assess their therapeutic potential for FIC. Exosomes were isolated from fADSCs and their microRNA (miRNA) content sequenced using Illumina technology. Our findings demonstrate that fADSC-derived exosomes harbor miRNAs that can induce regenerative processes, such as cell proliferation, immune modulation, angiogenesis, and anti-inflammatory responses. Key miRNAs identified include fca-miR-221, fca-let-7f-5p, fca-miR-337-5p, fca-miR-542-5p, fca-miR-24-3p, fca-miR-205, and fca-miR-23a, which promote proliferative, angiogenic, differentiation, and regenerative mechanisms. Additionally, miRNAs with anti-inflammatory effects, such as fca-miR-193a-5p and fca-miR-127-3p, and those positively regulating the immune system, including fca-let-7a-5p and fca-miR-chrC1_18846-5p, were identified. Of particular interest, fca-miR-219-5p (has-miR-6766-3p) has been reported to suppress liver fibrosis.These results underline the therapeutic potential of fADSC-derived exosomes in treating FIC and suggest innovative strategies for feline veterinary medicine.
Collapse
Affiliation(s)
- Andrea Rubini
- Ultravet Diagnostics, 40017, San Giovanni in Persiceto, Italy
| | - Federica Zanotti
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | - Giulia Calogero
- Ultravet Diagnostics, 40017, San Giovanni in Persiceto, Italy
| | - Gisella Bettini
- Clinica Veterinaria Estense, Via Pianelle, 31, 44123 Francolino (Ferrara), Italy
| | - Cristiana Piccoli
- Clinica Veterinaria Estense, Via Pianelle, 31, 44123 Francolino (Ferrara), Italy
| | | | - Luca Lovatti
- Department of Translational Medicine, University of Ferrara, 44121, Ferrara, Italy
| | - Barbara Zavan
- Department of Translational Medicine, University of Ferrara, 44121, Ferrara, Italy
| |
Collapse
|
3
|
Xia Y, Wang H, Shao M, Liu X, Sun F. MAP3K19 Promotes the Progression of Tuberculosis-Induced Pulmonary Fibrosis Through Activation of the TGF-β/Smad2 Signaling Pathway. Mol Biotechnol 2024; 66:3300-3310. [PMID: 37906388 DOI: 10.1007/s12033-023-00941-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 10/09/2023] [Indexed: 11/02/2023]
Abstract
Tuberculosis-induced pulmonary fibrosis (PF) is a chronic, irreversible interstitial lung disease, which severely affects lung ventilation and air exchange, leading to respiratory distress, impaired lung function, and ultimately death. As previously reported, epithelial-mesenchymal transition (EMT) and fibrosis in type II alveolar epithelial cells (AEC II) are two critical processes that contributes to the initiation and progression of tuberculosis-related PF, but the underlying pathological mechanisms remain unclear. In this study, through performing Real-Time quantitative PCR (RT-qPCR), Western blot, immunohistochemistry, and immunofluorescence staining assay, we confirmed that the expression levels of EMT and fibrosis-related biomarkers were significantly increased in lung tissues with tuberculosis-associated PF in vivo and Mycobacterium bovis Bacillus Calmette-Guérin (BCG) strain-infected AEC II cells in vitro. Besides, we noticed that the mitogen-activated protein kinase 19 (MAP3K19) was aberrantly overexpressed in PF models, and silencing of MAP3K19 significantly reduced the expression levels of fibronectin, collagen type I, and alpha-smooth muscle actin to decrease fibrosis, and upregulated E-cadherin and downregulated vimentin to suppress EMT in BCG-treated AEC II cells. Then, we uncovered the underlying mechanisms and found that BCG synergized with MAP3K19 to activate the pro-inflammatory transforming growth factor-beta (TGF-β)/Smad2 signal pathway in AEC II cells, and BCG-induced EMT process and fibrosis in AEC II cells were all abrogated by co-treating cells with TGF-β/Smad2 signal pathway inhibitor LY2109761. In summary, our results uncovered the underlying mechanisms by which the MAP3K19/TGF-β/Smad2 signaling pathway regulated EMT and fibrotic phenotypes of AEC II cells to facilitate the development of tuberculosis-associated PF, and these findings will provide new ideas and biomarkers to ameliorate tuberculosis-induced PF in clinic.
Collapse
Affiliation(s)
- Yu Xia
- Department of Respiratory Medicine, The First Affiliated Hospital of Xinjiang Medical University, No. 137, South Liyu Shan Road, Urumqi, 830054, China.
| | - Haiyue Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Xinjiang Medical University, No. 137, South Liyu Shan Road, Urumqi, 830054, China
| | - Meihua Shao
- Department of Respiratory Medicine, The First Affiliated Hospital of Xinjiang Medical University, No. 137, South Liyu Shan Road, Urumqi, 830054, China
| | - Xuemei Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Xinjiang Medical University, No. 137, South Liyu Shan Road, Urumqi, 830054, China
| | - Feng Sun
- Department of Respiratory Medicine, The First Affiliated Hospital of Xinjiang Medical University, No. 137, South Liyu Shan Road, Urumqi, 830054, China
| |
Collapse
|
4
|
Jin Z, Shi Y, Zhou L. Transparent sparse graph pathway network for analyzing the internal relationship of lung cancer. Front Genet 2024; 15:1437174. [PMID: 39411374 PMCID: PMC11473316 DOI: 10.3389/fgene.2024.1437174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 09/09/2024] [Indexed: 10/19/2024] Open
Abstract
While it is important to find the key biomarkers and improve the accuracy of disease models, it is equally important to understand their interaction relationships. In this study, a transparent sparse graph pathway network (TSGPN) is proposed based on the structure of graph neural networks. This network simulates the action of genes in vivo, adds to prior knowledge, and improves the model's accuracy. First, the graph connection was constructed according to protein-protein interaction networks and competing endogenous RNA (ceRNA) networks, from which some noise or unimportant connections were spontaneously removed based on the graph attention mechanism and hard concrete estimation. This realized the reconstruction of the ceRNA network representing the influence of other genes in the disease on mRNA. Next, the gene-based interpretation was transformed into a pathway-based interpretation based on the pathway database, and the hidden layer was added to realize the high-dimensional analysis of the pathway. Finally, the experimental results showed that the proposed TSGPN method is superior to other comparison methods in F1 score and AUC, and more importantly, it can effectively display the role of genes. Through data analysis applied to lung cancer prognosis, ten pathways related to LUSC prognosis were found, as well as the key biomarkers closely related to these pathways, such as HOXA10, hsa-mir-182, and LINC02544. The relationship between them was also reconstructed to better explain the internal mechanism of the disease.
Collapse
Affiliation(s)
- Zhibin Jin
- Information Engineering College, Shanghai Maritime University, pudong, China
| | - Yuhu Shi
- Information Engineering College, Shanghai Maritime University, pudong, China
| | - Lili Zhou
- Yangpu District Central Hospital, Shanghai, China
| |
Collapse
|
5
|
Bonet F, Campuzano O, Córdoba-Caballero J, Alcalde M, Sarquella-Brugada G, Braza-Boïls A, Brugada R, Hernández-Torres F, Quezada-Feijoo M, Ramos M, Mangas A, Ranea JAG, Toro R. Role of miRNA-mRNA Interactome in Pathophysiology of Arrhythmogenic Cardiomyopathy. Biomedicines 2024; 12:1807. [PMID: 39200271 PMCID: PMC11351583 DOI: 10.3390/biomedicines12081807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/25/2024] [Accepted: 07/29/2024] [Indexed: 09/02/2024] Open
Abstract
Arrhythmogenic cardiomyopathy is an inherited entity characterized by irregular cell-cell adhesion, cardiomyocyte death and fibro-fatty replacement of ventricular myocytes, leading to malignant ventricular arrythmias, contractile dysfunction and sudden cardiac death. Pathogenic variants in genes that encode desmosome are the predominant cause of arrhythmogenic cardiomyopathy. Moreover, signalling pathways such as Wnt/ß-catenin and transforming growth factor-β have been involved in the disease progression. However, still little is known about the molecular pathophysiological mechanisms that underlie arrhythmogenic cardiomyopathy pathogenesis. We used mRNA and small RNA sequencing to analyse the transcriptome of health and arrhythmogenic cardiomyopathy of autopsied human hearts. Our results showed 697 differentially expressed genes and eight differentially expressed miRNAs. Functional enrichment revealed mitochondrial respiratory-related pathways, impaired response to oxidative stress, apoptotic signalling pathways and inflammatory response-related and extracellular matrix response pathways. Furthermore, analysis of the miRNA-mRNA interactome identified eleven negatively correlated miRNA-target pairs for arrhythmogenic cardiomyopathy. Our finding revealed novel arrhythmogenic cardiomyopathy-related miRNAs with important regulatory function in disease pathogenesis, highlighting their value as potential key targets for therapeutic approaches.
Collapse
Affiliation(s)
- Fernando Bonet
- Research Unit, Biomedical Research and Innovation Institute of Cadiz (INiBICA), Puerta del Mar University Hospital, 11009 Cádiz, Spain; (F.B.); (J.C.-C.); (A.M.)
| | - Oscar Campuzano
- Medical Science Department, School of Medicine, University of Girona, 17003 Girona, Spain; (G.S.-B.); (R.B.)
- Institut d’Investigació Biomèdica de Girona (IDIBGI-CERCA), 17190 Salt, Spain;
- Centro Investigación Biomédica en Red, Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain;
| | - José Córdoba-Caballero
- Research Unit, Biomedical Research and Innovation Institute of Cadiz (INiBICA), Puerta del Mar University Hospital, 11009 Cádiz, Spain; (F.B.); (J.C.-C.); (A.M.)
- Department of Molecular Biology and Biochemistry, University of Málaga, 29071 Málaga, Spain;
| | - Mireia Alcalde
- Institut d’Investigació Biomèdica de Girona (IDIBGI-CERCA), 17190 Salt, Spain;
- Centro Investigación Biomédica en Red, Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain;
| | - Georgia Sarquella-Brugada
- Medical Science Department, School of Medicine, University of Girona, 17003 Girona, Spain; (G.S.-B.); (R.B.)
- Pediatric Arrhythmias, Inherited Cardiac Diseases and Sudden Death Unit, Cardiology Department, Sant Joan de Déu Hospital, 08950 Barcelona, Spain
- Arrítmies Pediàtriques, Cardiologia Genètica i Mort Sobtada, Malalties Cardiovasculars en el Desenvolupament, Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain
| | - Aitana Braza-Boïls
- Centro Investigación Biomédica en Red, Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain;
- Cardiopatías Familiares, Muerte Súbita y Mecanismos de Enfermedad (CAFAMUSME) Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - Ramon Brugada
- Medical Science Department, School of Medicine, University of Girona, 17003 Girona, Spain; (G.S.-B.); (R.B.)
- Institut d’Investigació Biomèdica de Girona (IDIBGI-CERCA), 17190 Salt, Spain;
- Centro Investigación Biomédica en Red, Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain;
- Cardiology Service, Hospital Josep Trueta de Girona, 17007 Girona, Spain
| | - Francisco Hernández-Torres
- Medina Foundation, Technology Park of Health Sciences, 18016 Granada, Spain;
- Department of Biochemistry and Molecular Biology III and Immunology, Faculty of Medicine, University of Granada, 18016 Granada, Spain
| | - Maribel Quezada-Feijoo
- Cardiology Department, Hospital Central de la Cruz Roja, 28003 Madrid, Spain; (M.Q.-F.)
- Medicine School, Alfonso X el Sabio University, 28007 Madrid, Spain
| | - Monica Ramos
- Cardiology Department, Hospital Central de la Cruz Roja, 28003 Madrid, Spain; (M.Q.-F.)
- Medicine School, Alfonso X el Sabio University, 28007 Madrid, Spain
| | - Alipio Mangas
- Research Unit, Biomedical Research and Innovation Institute of Cadiz (INiBICA), Puerta del Mar University Hospital, 11009 Cádiz, Spain; (F.B.); (J.C.-C.); (A.M.)
- Medicine Department, School of Medicine, University of Cadiz, 11003 Cádiz, Spain
- Lipid and Atherosclerotic Unit, Puerta del Mar University Hospital, 11009 Cadiz, Spain
| | - Juan A. G. Ranea
- Department of Molecular Biology and Biochemistry, University of Málaga, 29071 Málaga, Spain;
- Institute of Biomedical Research in Málaga and platform of nanomedicine (IBIMA Plataforma BIONAND), 29071 Málaga, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
- Spanish National Bioinformatics Institute (INB/ELIXIR-ES), Instituto de Salud Carlos III (ISCIII), 28020 Madrid, Spain
| | - Rocío Toro
- Research Unit, Biomedical Research and Innovation Institute of Cadiz (INiBICA), Puerta del Mar University Hospital, 11009 Cádiz, Spain; (F.B.); (J.C.-C.); (A.M.)
- Medicine Department, School of Medicine, University of Cadiz, 11003 Cádiz, Spain
| |
Collapse
|
6
|
Shi Y, Jin Z, Deng J, Zeng W, Zhou L. A Novel High-Dimensional Kernel Joint Non-Negative Matrix Factorization With Multimodal Information for Lung Cancer Study. IEEE J Biomed Health Inform 2024; 28:976-987. [PMID: 38032777 DOI: 10.1109/jbhi.2023.3335950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Judging and identifying biological activities and biomarkers inside tissues from imaging features of diseases is challenging, so correlating pathological image data with genes inside organisms is of great significance for clinical diagnosis. This paper proposes a high-dimensional kernel non-negative matrix factorization (NMF) method based on muti-modal information fusion. This algorithm can project RNA gene expression data and pathological images (WSI) into a common feature space, where the heterogeneous variables with the largest coefficient in the same projection direction form a co-module. In addition, the miRNA-mRNA and miRNA-lncRNA interaction networks in the ceRNA network are added to the algorithm as a priori information to explore the relationship between the images and the internal activities of the gene. Furthermore, the radial basis kernel function is used to calculate the feature proportion between different kinds of genes mapped in the high-dimensional feature space and projected into the common feature space to explore the gene interaction in the high-dimensional situation. The original feature matrix is regularized to improve biological correlation, and the feature factors are sparse by orthogonal constraints to reduce redundancy. Experimental results show that the proposed NMF method is better than the traditional NMF method in stability, decomposition accuracy, and robustness. Through data analysis applied to lung cancer, genes related to tissue morphology are found, such as COL7A1, CENPF and BIRC5. In addition, gene pairs with a correlation degree exceeding 0.8 are found, and potential biomarkers of significant correlation with survival are obtained such as CAPN8. It has potential application value for the clinical diagnosis of lung cancer.
Collapse
|
7
|
Zeitlmayr S, Cami D, Selmani B, Gudermann T, Breit A. A dual role for ERK-1/2 in the regulation of plasmin activity and cell migration in metastatic NSCLC-H1299 cells. Arch Toxicol 2023; 97:3113-3128. [PMID: 37712947 PMCID: PMC10567951 DOI: 10.1007/s00204-023-03600-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 08/30/2023] [Indexed: 09/16/2023]
Abstract
Occupational and environmental exposure of various toxins or cigarette smoke causes non-small cell lung carcinoma (NSCLC); a devastating disease with a very low survival rate after metastasis. Increased activity of plasmin is a hallmark in NSCLC metastasis. It is accepted that metastatic cells exhibit higher plasmin activity than cells from primary tumors. Mechanisms behind this elevation, however, are barely understood. We compared plasmin activity and cell migration of A549 cells derived from a primary lung tumor with metastatic H1299 lung cells isolated from lymph nodes. Surprisingly, we found higher plasmin activity and migration for A549 cells. mRNA levels of the plasminogen activator inhibitor-1 (PAI-1) were higher in H1299 cells and activity of extracellular-regulated kinases-1/2 (ERK-1/2) was increased. An inhibitor of ERK-1/2 decreased PAI-1 mRNA levels and increased plasmin activity or cell migration in H1299 cells. Transforming growth factor-β (TGF-β) decreased plasmin activity and migration in A549 cells but enhanced both in H1299 cells. The cytokine massively increased PAI-1 and decreased urokinase plasminogen activator (uPA) levels in A549 cells but strongly induced uPA and only weakly PAI- 1 expression in H1299 cells. Consequently, TGF-β enhanced plasmin activity and cell migration in H1299. Additionally, TGF-β activated ERK-1/2 stronger in H1299 than in A549 cells. Accordingly, an ERK-1/2 inhibitor completely reversed the effects of TGF-β on uPA expression, plasmin activity and migration in H1299 cells. Hence, we provide first data indicating TGF-β-promoted increased plasmin activity and suggest that blocking TGF-β-promoted ERK-1/2 activity might be a straightforward approach to inhibit NSCLC metastasis.
Collapse
Affiliation(s)
- Sarah Zeitlmayr
- Walther Straub Institute of Pharmacology and Toxicology, Medical Faculty, LMU Munich, Goethestrasse 33, 80336, Munich, Germany
| | - Ditila Cami
- Walther Straub Institute of Pharmacology and Toxicology, Medical Faculty, LMU Munich, Goethestrasse 33, 80336, Munich, Germany
| | - Belinda Selmani
- Walther Straub Institute of Pharmacology and Toxicology, Medical Faculty, LMU Munich, Goethestrasse 33, 80336, Munich, Germany
| | - Thomas Gudermann
- Walther Straub Institute of Pharmacology and Toxicology, Medical Faculty, LMU Munich, Goethestrasse 33, 80336, Munich, Germany
| | - Andreas Breit
- Walther Straub Institute of Pharmacology and Toxicology, Medical Faculty, LMU Munich, Goethestrasse 33, 80336, Munich, Germany.
| |
Collapse
|
8
|
Hussen BM, Saleem SJ, Abdullah SR, Mohamadtahr S, Hidayat HJ, Rasul MF, Taheri M, Kiani A. Current landscape of miRNAs and TGF-β signaling in lung cancer progression and therapeutic targets. Mol Cell Probes 2023; 72:101929. [PMID: 37683829 DOI: 10.1016/j.mcp.2023.101929] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 08/27/2023] [Accepted: 09/04/2023] [Indexed: 09/10/2023]
Abstract
Lung cancer (LC) is the primary reason for cancer-associated fatalities globally. Due to both tumor-suppressing and tumor-promoting activities, the TGF-β family of growth factors is extremely essential to tumorigenesis. A non-coding single-stranded short RNA called microRNA (miRNA), which is made up of about 22 nt and is encoded by endogenous genes, can control normal and pathological pathways in various kinds of cancer, including LC. Recent research demonstrated that the TGF-β signaling directly can affect the synthesis of miRNAs through suppressor of mothers against decapentaplegic (SMAD)-dependent activity or other unidentified pathways, which could generate allostatic feedback as a result of TGF-β signaling stimulation and ultimately affect the destiny of cancer tissues. In this review, we emphasize the critical functions of miRNAs in lung cancer progression and, more critically, how they affect the TGF-β signaling pathway, and explore the role of both the TGF-β signaling pathway and miRNAs as potential therapeutic targets for improving the treatments of LC patients.
Collapse
Affiliation(s)
- Bashdar Mahmud Hussen
- Department of Clinical Analysis, College of Pharmacy, Hawler Medical University, Kurdistan Region, Erbil, Iraq; Department of Biomedical Sciences, Cihan University-Erbil, Erbil, Kurdistan Region, 44001, Iraq
| | - Safeen Jasim Saleem
- Department of Clinical Analysis, College of Pharmacy, Hawler Medical University, Kurdistan Region, Erbil, Iraq
| | - Snur Rasool Abdullah
- Medical Laboratory Science, Lebanese French University, Kurdistan Region, Erbil, Iraq
| | - Sayran Mohamadtahr
- Department of Clinical Analysis, College of Pharmacy, Hawler Medical University, Kurdistan Region, Erbil, Iraq
| | - Hazha Jamal Hidayat
- Department of Biology, College of Education, Salahaddin University-Erbil, Kurdistan Region, Iraq
| | - Mohammed Fatih Rasul
- Department of Pharmaceutical Basic Science, Faculty of Pharmacy, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Mohammad Taheri
- Institute of Human Genetics, Jena University Hospital, Jena, Germany; Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Arda Kiani
- Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Lung Research and Developmental Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Samson JS, Parvathi VD. Prospects of microRNAs as therapeutic biomarkers in non-small cell lung cancer. Med Oncol 2023; 40:345. [PMID: 37922117 DOI: 10.1007/s12032-023-02212-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 10/06/2023] [Indexed: 11/05/2023]
Abstract
Lung Cancer, the second most common cancer worldwide, remains the leading cause of cancer-related deaths, contemporarily. More than 85% of identified lung cancer cases are comprised of non-small-cell lung carcinoma (NSCLC). Despite the best advancements in the realm of NSCLC therapy, the five-year survival period of NSCLC patients remains unchanged. Underlying complex molecular heterogeneity, delay in early detection resulting in progression of the disease to its advanced stage and acquired resistance of NSCLC cells during therapy have posed additional challenges for circumventing the discrepancies in treatment strategy. microRNAs (miRNAs) are a class of non-coding RNAs, identified as molecules playing an indispensable role in tumorigenesis & progression and metastasis of several cancers, including NSCLC, either by possessing tumor suppressor or by oncogenic functions. As observed across several studies, miRNA dysregulation has been recognised as a causative mechanism behind NSCLC tumorigenesis. In this review, we discuss the role of miRNAs in NSCLC tumor progression caused by their dysregulation, thereby stating their potential therapeutic application in NSCLC as therapeutic biomarkers. We have also highlighted the recent findings of some of the most widely studied tumor suppressor (miR-486, miR-7 miR-34), and oncogene miRNAs (miR-21, miR-224, miR-135b) that can be further explored for its therapeutic potentialities in the management of NSCLC.
Collapse
Affiliation(s)
- Jennifer Sally Samson
- Department of Biomedical Sciences, Sri Ramachandra Institute of Higher Education and Research, 1, Mount Poonamallee Road, Sri Ramachandra Nagar, Chennai, Tamil Nadu, 600116, India
| | - Venkatachalam Deepa Parvathi
- Department of Biomedical Sciences, Sri Ramachandra Institute of Higher Education and Research, 1, Mount Poonamallee Road, Sri Ramachandra Nagar, Chennai, Tamil Nadu, 600116, India.
| |
Collapse
|
10
|
Kwantwi LB. The dual and multifaceted role of relaxin-2 in cancer. Clin Transl Oncol 2023; 25:2763-2771. [PMID: 36947362 DOI: 10.1007/s12094-023-03146-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 03/01/2023] [Indexed: 03/23/2023]
Abstract
The continuous increase in cancer-associated deaths despite the substantial improvement in diagnosis and treatment has sparked discussions on the need for novel biomarkers and therapeutic strategies for cancer. Although increasing evidence has demonstrated the pivotal role of relaxin-2 in multiple cancers, their role is a double-edged sword with both protumor and antitumor having been reported in various malignant tumors. Considering this dual role, it appears the biological mechanism underpinning the action of relaxin-2 in cancer is not clear and further studies to elucidate their potential as a preventive factor for cancers are of prime importance. Herein, a summarized up-to-date report on the role of relaxin-2 in human cancer including detailed clinical and experimental evidence supporting their tumor-promoting and inhibitory functions in cancer development and progression has been elucidated. Also, signaling pathways and other factors orchestrating the activities of relaxin-2 in the tumor microenvironment has been discussed. Collectively, the evidence from this review has demonstrated the need for further evaluation of the role of relaxin-2 as a diagnostic and or prognostic biomarker for cancer.
Collapse
Affiliation(s)
- Louis Boafo Kwantwi
- Department of Medical Imaging Sciences, Klintaps College of Health and Allied Sciences, Accra, DTD. TDC, 30A Klagon, Com. 19, Tema, Ghana.
| |
Collapse
|
11
|
Nadal E, Saleh M, Aix SP, Ochoa-de-Olza M, Patel SP, Antonia S, Zhao Y, Gueorguieva I, Man M, Estrem ST, Liu J, Avsar E, Lin WH, Benhadji KA, Gandhi L, Guba SC, Diaz IA. A phase Ib/II study of galunisertib in combination with nivolumab in solid tumors and non-small cell lung cancer. BMC Cancer 2023; 23:708. [PMID: 37507657 PMCID: PMC10386782 DOI: 10.1186/s12885-023-11153-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND In this phase Ib/II open-label study, tumor immune suppression was targeted in patients with advanced refractory solid tumors and patients with recurrent/refractory non-small cell lung cancer (NSCLC) using galunisertib with nivolumab. METHODS Eligible patients were ≥ 18 years old, had an Eastern Cooperative Oncology Group performance status ≤ 1, and were treatment-naive for anti-programmed cell death-1, its ligand, or transforming growth factor β receptor 1 kinase inhibitors. Phase Ib was an open-label, dose-escalation assessment of the safety and tolerability of galunisertib with nivolumab in patients with advanced refractory solid tumors. Phase II evaluated the safety of galunisertib with nivolumab in NSCLC patients who had received prior platinum-based treatment but were immuno-oncology agent-naive. RESULTS This trial was conducted between October 2015 and August 2020. No dose-limiting toxicities were observed in phase I. In the phase II NSCLC cohort (n = 25), patients received 150 mg twice daily galunisertib (14 days on/14 days off dosing schedule for all phases) plus nivolumab at 3 mg/kg (intravenously every 2 weeks). In this phase, the most frequent treatment-related adverse events (AEs) were pruritus (n = 9, 36%), fatigue (n = 8, 32%), and decreased appetite (n = 7, 28%). No grade 4 or 5 treatment-related AEs were observed. Six (24%) patients had confirmed partial response (PR) and 4 (16%) had stable disease; 1 additional patient had confirmed PR after initial pseudo-progression. The median duration of response was 7.43 months (95% confidence interval [CI]: 3.75, NR). Among the 7 responders, including the delayed responder, 1 had high PD-L1 expression (≥ 50%). The median progression-free survival was 5.26 months (95% CI: 1.77, 9.20) and the median overall survival was 11.99 months (95% CI: 8.15, NR). Interferon gamma response genes were induced post-treatment and cell adhesion genes were repressed, although the association of these observations with tumor response and clinical outcomes was not statistically powered due to limited samples available. CONCLUSIONS The study met its primary endpoint as galunisertib combined with nivolumab was well tolerated. Preliminary efficacy was observed in a subset of patients in the Phase 2 NSCLC cohort. TRIAL REGISTRATION Trial registered with ClinicalTrials.gov (NCT02423343; 22.04.2015).
Collapse
Affiliation(s)
- Ernest Nadal
- Department of Medical Oncology, Catalan Institute of Oncology, IDIBELL, L'Hospitalet, Barcelona, Spain.
| | | | | | | | | | - Scott Antonia
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Yumin Zhao
- Eli Lilly and Company, Indianapolis, IN, USA
| | | | - Michael Man
- Eli Lilly and Company, Indianapolis, IN, USA
| | | | | | - Emin Avsar
- Eli Lilly and Company, Indianapolis, IN, USA
- Bristol Myers Squibb, Princeton, NJ, USA
| | - Wen Hong Lin
- Bristol Myers Squibb, Princeton, NJ, USA
- Immune-Onc Therapeutics, Palo Alto, CA, USA
| | | | - Leena Gandhi
- Eli Lilly and Company, Indianapolis, IN, USA
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Inmaculada Ales Diaz
- UGCI Oncología Médica, Hospitales Universitarios Regional Y Virgen de La Victoria, IBIMA, Málaga, Spain
| |
Collapse
|
12
|
Li Z, Fu R, Wen X, Zhang L. Network analysis reveals miRNA crosstalk between periodontitis and oral squamous cell carcinoma. BMC Oral Health 2023; 23:19. [PMID: 36639776 PMCID: PMC9840318 DOI: 10.1186/s12903-022-02704-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 12/28/2022] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) is one of the malignant tumors with a poor prognosis. Periodontitis (PD is considered a high-risk factor for OSCC, but the genetic mechanism is rarely studied. This study aims to link OSCC and PD by identifying common differentially expressed miRNAs (Co-DEmiRNAs), their related genes (Hub genes), transcription factors (TFs), signaling pathways, enrichment functions, and compounds, and searching for genetic commonalities. METHODS The miRNAs expression datasets of OSCC and PD were searched from the GEO database. The miRNA and related crosstalk mechanism between OSCC and PD was obtained through a series of analyses. RESULTS hsa-mir-497, hsa-mir-224, hsa-mir-210, hsa-mir-29c, hsa-mir-486-5p, and hsa-mir-31are the top miRNA nodes in Co-DEmiRNA-Target networks. The most significant candidate miRNA dysregulation genes are ZNF460, FBN1, CDK6, BTG2, and CBX6, while the most important dysregulation TF includes HIF1A, TP53, E2F1, MYCN, and JUN. 5-fluorouracil, Ginsenoside, Rh2, and Formaldehyde are the most correlated compounds. Enrichment analysis revealed cancer-related pathways and so on. CONCLUSIONS The comprehensive analysis reveals the interacting genetic and molecular mechanism between OSCC and PD, linking both and providing a foundation for future basic and clinical research.
Collapse
Affiliation(s)
- Zhengrui Li
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology and National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Rao Fu
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology and National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Xutao Wen
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology and National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Ling Zhang
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China.
- National Center for Stomatology and National Clinical Research Center for Oral Diseases, Shanghai, China.
- Shanghai Key Laboratory of Stomatology, Shanghai, China.
| |
Collapse
|
13
|
Yamazaki M, Hosokawa M, Matsunaga H, Arikawa K, Takamochi K, Suzuki K, Hayashi T, Kambara H, Takeyama H. Integrated spatial analysis of gene mutation and gene expression for understanding tumor diversity in formalin-fixed paraffin-embedded lung adenocarcinoma. Front Oncol 2022; 12:936190. [PMID: 36505794 PMCID: PMC9731154 DOI: 10.3389/fonc.2022.936190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 10/31/2022] [Indexed: 11/26/2022] Open
Abstract
Introduction A deeper understanding of intratumoral heterogeneity is essential for prognosis prediction or accurate treatment plan decisions in clinical practice. However, due to the cross-links and degradation of biomolecules within formalin-fixed paraffin-embedded (FFPE) specimens, it is challenging to analyze them. In this study, we aimed to optimize the simultaneous extraction of mRNA and DNA from microdissected FFPE tissues (φ = 100 µm) and apply the method to analyze tumor diversity in lung adenocarcinoma before and after erlotinib administration. Method Two magnetic beads were used for the simultaneous extraction of mRNA and DNA. The decross-linking conditions were evaluated for gene mutation and gene expression analyses of microdissected FFPE tissues. Lung lymph nodes before treatment and lung adenocarcinoma after erlotinib administration were collected from the same patient and were preserved as FFPE specimens for 4 years. Gene expression and gene mutations between histologically classified regions of lung adenocarcinoma (pre-treatment tumor in lung lymph node biopsies and post-treatment tumor, normal lung, tumor stroma, and remission stroma, in resected lung tissue) were compared in a microdissection-based approach. Results Using the optimized simultaneous extraction of DNA and mRNA and whole-genome amplification, we detected approximately 4,000-10,000 expressed genes and the epidermal growth factor receptor (EGFR) driver gene mutations from microdissected FFPE tissues. We found the differences in the highly expressed cancer-associated genes and the positive rate of EGFR exon 19 deletions among the tumor before and after treatment and tumor stroma, even though they were collected from tumors of the same patient or close regions of the same specimen. Conclusion Our integrated spatial analysis method would be applied to various FFPE pathology specimens providing area-specific gene expression and gene mutation information.
Collapse
Affiliation(s)
- Miki Yamazaki
- Department of Life Science and Medical Bioscience, Waseda University, Tokyo, Japan,Computational Bio Big-Data Open Innovation Laboratory, National Institute of Advanced Industrial Science and Technology, Tokyo, Japan
| | - Masahito Hosokawa
- Department of Life Science and Medical Bioscience, Waseda University, Tokyo, Japan,Computational Bio Big-Data Open Innovation Laboratory, National Institute of Advanced Industrial Science and Technology, Tokyo, Japan,Research Organization for Nano and Life Innovation, Waseda University, Tokyo, Japan,Institute for Advanced Research of Biosystem Dynamics, Waseda Research Institute for Science and Engineering, Waseda University, Tokyo, Japan
| | - Hiroko Matsunaga
- Research Organization for Nano and Life Innovation, Waseda University, Tokyo, Japan
| | - Koji Arikawa
- Research Organization for Nano and Life Innovation, Waseda University, Tokyo, Japan
| | - Kazuya Takamochi
- Department of Thoracic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Kenji Suzuki
- Department of Thoracic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Takuo Hayashi
- Department of Human Pathology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Hideki Kambara
- Research Organization for Nano and Life Innovation, Waseda University, Tokyo, Japan,Frontier BioSystems Inc., Tokyo, Japan
| | - Haruko Takeyama
- Department of Life Science and Medical Bioscience, Waseda University, Tokyo, Japan,Computational Bio Big-Data Open Innovation Laboratory, National Institute of Advanced Industrial Science and Technology, Tokyo, Japan,Research Organization for Nano and Life Innovation, Waseda University, Tokyo, Japan,Institute for Advanced Research of Biosystem Dynamics, Waseda Research Institute for Science and Engineering, Waseda University, Tokyo, Japan,*Correspondence: Haruko Takeyama,
| |
Collapse
|
14
|
Combined miR-486 and GP88 (Progranulin) Serum Levels Are Suggested as Supportive Biomarkers for Therapy Decision in Elderly Prostate Cancer Patients. Life (Basel) 2022; 12:life12050732. [PMID: 35629399 PMCID: PMC9143270 DOI: 10.3390/life12050732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/10/2022] [Accepted: 05/10/2022] [Indexed: 11/16/2022] Open
Abstract
Our study aimed to assess the applicability of miR-486 in combination with soluble GP88 protein as a diagnostic and/or predictive biomarker for prostate cancer (PCa) patients. miR-486 and GP88 levels in serum samples from 136 patients undergoing MRI-guided biopsy of the prostate were assessed by qRT−PCR and ELISA, respectively. Of these, 86 patients received a histologically confirmed diagnosis of PCa. Neither marker showed an association with the diagnosis of cancer. PCa patients were separated based on (i) treatment into patients with active surveillance or patients with any type of curative treatment and (ii) age into elderly (>68 years) patients and younger patients (≤68 years). In elderly patients (N = 41) with the intention of curative treatment at optimized cut-off values, significantly higher GP88 levels (p = 0.018) and lower miR-486 levels (p = 0.014) were observed. The total PSA level and ISUP biopsy grade were used in a baseline model for predicting definitive therapy. The baseline model exhibited an area under the curve (AUC) of 0.783 (p = 0.005). The addition of the serum biomarkers miR-486 and GP88 to the baseline model yielded an improved model with an AUC of 0.808 (p = 0.002). Altogether, combined miR-486 and GP88 serum levels are associated with and are therefore suggested as supportive biomarkers for therapy decisions, particularly in elderly PCa patients.
Collapse
|
15
|
miR-486-5p Restrains Extracellular Matrix Production and Oxidative Damage in Human Trabecular Meshwork Cells by Targeting TGF-β/SMAD2 Pathway. J Ophthalmol 2022; 2022:3584192. [PMID: 35251709 PMCID: PMC8890899 DOI: 10.1155/2022/3584192] [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: 12/03/2021] [Accepted: 01/26/2022] [Indexed: 11/17/2022] Open
Abstract
Background Glaucoma is characterized by elevated intraocular pressure caused by aqueous outflow dysfunction. Trabecular meshwork plays a key role in controlling intraocular pressure by modulating aqueous outflow. This study investigated the protective effects of miR-486-5p in H2O2-stimulated human trabecular meshwork cells (TMCs). Methods TMCs were disposed with 300 μM H2O2 to establish oxidative damage models in vitro. miR-486-5p mimics and its controls were transfected into TMCs, and cell apoptosis and extracellular matrix production (ECM) genes were measured by flow cytometry, western blotting, and immunofluorescence staining. Activities of superoxide dismutase (SOD) and malondialdehyde (MDA) were also assayed. Online tools and luciferase reporter assays were used to uncover the relationship between miR-486-5p and the TGF-β/SMAD2 pathway. Results We found that H2O2-induced oxidative damage in TMCs and miR-486-5p was downregulated in H2O2-stimulated TMCs. Overexpression of miR-486-5p mitigated H2O2-induced oxidative damage by inhibiting apoptosis, reducing cleaved caspase-3/9 expression, reducing MDA levels, and increasing SOD levels as well as downregulating ECM genes. SMAD2 was demonstrated to be targeted by miR-486-5p, and miR-486-5p inhibited TGF-β/SMAD2 signaling in H2O2-stimulated TMCs. Additionally, SMAD2 was upregulated by H2O2, and SMAD2 upregulation abrogated the protective effects of miR-486-5p against H2O2-induced injury. Conclusion miR-486-5p restrains H2O2-induced oxidative damage in TMCs by targeting the TGF-β/SMAD2 pathway.
Collapse
|
16
|
Zhou Y, Zhang Y, Li Y, Liu L, Li Z, Liu Y, Xiao Y. MicroRNA-106a-5p promotes the proliferation, autophagy and migration of lung adenocarcinoma cells by targeting LKB1/AMPK. Exp Ther Med 2021; 22:1422. [PMID: 34707704 PMCID: PMC8543179 DOI: 10.3892/etm.2021.10857] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 07/07/2021] [Indexed: 12/31/2022] Open
Abstract
It has previously been reported that lung cancer has the highest morbidity and mortality rate worldwide; however, the pathogenesis underlying lung cancer has not been fully elucidated. The aim of the present was primarily to assess the influence of microRNA (miR)-106a-5p on the biological behaviors of lung cancer cells. In the present study, bioinformatics analysis was used to analyze the expression characteristics of miR-106a-5p and its relationship with the prognosis of patients with lung adenocarcinoma (LUAD) in The Cancer Genome Atlas. A dual luciferase reporter assay was performed to verify the binding of miR-106a-5p and liver kinase B1 (LKB1). The Cell Counting Kit-8, colony formation and Transwell assays were utilized to detect cell viability, proliferation and migration, respectively. Protein and RNA expression levels were examined by western blotting and reverse transcription-quantitative PCR analysis, respectively. It was observed that miR-106a-5p was highly expressed in LUAD and associated with poor prognosis. miR-106a-5p promoted the proliferation and migration of LUAD cells, and inhibited autophagy. By contrast, LKB1 inhibited cell proliferation and migration, promoted autophagy and blocked the cancer-promoting effects of miR-106a-5p. Overexpression of miR-106a-5p inhibited the phosphorylation of AMP-activated protein kinase (AMPK) and tuberin (TSC2), and promoted the phosphorylation of mTOR. By contrast, overexpression of LKB1 blocked the promotion of mTOR phosphorylation, and the inhibition of AMPK and TSC2 phosphorylation caused by miR-106a-5p. In summary, the results of the present study indicated that miR-106a-5p regulated the phosphorylation of the AMPK pathway by targeting LKB1, and was involved in the proliferation, migration and autophagy of LUAD cells.
Collapse
Affiliation(s)
- Yushan Zhou
- Department of Respiratory and Critical Care Medicine, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650051, P.R. China
| | - Yuxuan Zhang
- Department of Respiratory and Critical Care Medicine, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650051, P.R. China
| | - Yanli Li
- Department of Respiratory and Critical Care Medicine, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650051, P.R. China
| | - Liqiong Liu
- Department of Respiratory and Critical Care Medicine, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650051, P.R. China
| | - Zhidong Li
- Department of Respiratory and Critical Care Medicine, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650051, P.R. China
| | - Yanhong Liu
- Department of Respiratory and Critical Care Medicine, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650051, P.R. China
| | - Yi Xiao
- Department of Respiratory and Critical Care Medicine, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650051, P.R. China
| |
Collapse
|
17
|
Zhao H, Yang H, Geng C, Chen Y, Tang Y, Li Z, Pang J, Shu T, Nie Y, Liu Y, Jia K, Wang J. Elevated IgE promotes cardiac fibrosis by suppressing miR-486a-5p. Theranostics 2021; 11:7600-7615. [PMID: 34158869 PMCID: PMC8210611 DOI: 10.7150/thno.47845] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 05/26/2021] [Indexed: 12/13/2022] Open
Abstract
Rationale: Cardiac fibrosis is an important feature of cardiac remodeling and is a hallmark of heart failure. Recent studies indicate that elevated IgE plays a causal role in pathological cardiac remodeling. However, the underlying mechanism of how IgE promotes cardiac fibrosis has not been fully elucidated. Methods and Results: To explore the function of IgE in cardiac fibrosis, we stimulated mouse primary cardiac fibroblasts (CFs) with IgE and found that both IgE receptor (FcεR1) and fibrosis related proteins were increased after IgE stimulation. Specific deletion of FcεR1 in CFs alleviated angiotensin II (Ang II)-induced cardiac fibrosis in mice. To investigate the mechanisms underlying the IgE-mediated cardiac fibrosis, deep miRNA-seq was performed. Bioinformatics and signaling pathway analysis revealed that IgE upregulated Col1a1 and Col3a1 expression in CFs by repressing miR-486a-5p, with Smad1 participating downstream of miR-486a-5p in this process. Lentivirus-mediated overexpression of miR-486a-5p was found to alleviate Ang II-induced myocardial interstitial fibrosis in mice. Moreover, miR-486-5p serum levels were lower in patients with heart failure than in healthy controls, and were negatively correlated with NT-proBNP levels. Conclusions: Our study demonstrates that elevated IgE promotes pathological cardiac fibrosis by modulating miR-486a-5p and downstream factors, such as Smad1. These findings suggest new targets for pathological cardiac fibrosis intervention.
Collapse
Affiliation(s)
- Hongmei Zhao
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing 100005, China
| | - Hongqin Yang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing 100005, China
| | - Chi Geng
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing 100005, China
| | - Yang Chen
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing 100005, China
| | - Yaqin Tang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing 100005, China
| | - Zhiwei Li
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing 100005, China
| | - Junling Pang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing 100005, China
| | - Ting Shu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing 100005, China
| | - Yu Nie
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 102308, China
| | - Yongshuo Liu
- Biomedical Pioneering Innovation Center (BIOPIC), Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China
- Department of Clinical Laboratory, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China
| | - Kegang Jia
- Department of Clinical Laboratory, TEDA International Cardiovascular Hospital, Tianjin 300457, China
| | - Jing Wang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing 100005, China
| |
Collapse
|
18
|
Abolfathi H, Sheikhpour M, Shahraeini SS, Khatami S, Nojoumi SA. Studies in lung cancer cytokine proteomics: a review. Expert Rev Proteomics 2021; 18:49-64. [PMID: 33612047 DOI: 10.1080/14789450.2021.1892491] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Proteins are molecules that have role in the progression of the diseases. Proteomics is a tool that can play an effective role in identifying diagnostic and therapeutic biomarkers for lung cancer. Cytokines are proteins that play a decisive role in activating body's immune system in lung cancer. They can increase the growth of the tumor (oncogenic cytokines) or limit tumor growth (anti-tumor cytokines) by regulating related signaling pathways such as proliferation, growth, metastasis, and apoptosis. AREAS COVERED In the present study, a total of 223 papers including 196 research papers and 27 review papers, extracted from PubMed and Scopus and published from 1997 to present, are reviewed. The most important involved-cytokines in lung cancer including TNF-α, IFN- γ, TGF-β, VEGF and interleukins such as IL-6, IL-17, IL-8, IL-10, IL-22, IL-1β and IL-18 are introduced. Also, the pathological and biological role of such cytokines in cancer signaling pathways is explained. EXPERT OPINION In lung cancer, the cytokine expression changes under the physiological conditions of the immune system, and inflammatory cytokines are associated with the progression of lung cancer. Therefore, the cytokine expression profile can be used in the diagnosis, prognosis, prediction of therapeutic responses, and survival of patients with lung cancer.
Collapse
Affiliation(s)
- Hanie Abolfathi
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.,Department of Biochemistry, Pasteur Institute of Iran, Tehran, Iran
| | - Mojgan Sheikhpour
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.,Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Sadegh Shahraeini
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.,Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Shohreh Khatami
- Department of Biochemistry, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Ali Nojoumi
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.,Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
19
|
Cai C, Zeng Q, Zhou G, Mu X. Identification of novel transcription factor-microRNA-mRNA co-regulatory networks in pulmonary large-cell neuroendocrine carcinoma. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:133. [PMID: 33569435 PMCID: PMC7867924 DOI: 10.21037/atm-20-7759] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Background Large cell neuroendocrine carcinoma (LCNEC) of the lung is a rare neuroendocrine neoplasm. Previous studies have shown that microRNAs (miRNAs) are widely involved in tumor regulation through targeting critical genes. However, it is unclear which miRNAs play vital roles in the pathogenesis of LCNEC, and how they interact with transcription factors (TFs) to regulate cancer-related genes. Methods To determine the novel TF-miRNA-target gene feed-forward loop (FFL) model of LCNEC, we integrated multi-omics data from Gene Expression Omnibus (GEO), Transcriptional Regulatory Relationships Unraveled by Sentence-Based Text Mining (TRRUST), Transcriptional Regulatory Element Database (TRED), and The experimentally validated microRNA-target interactions database (miRTarBase database). First, expression profile datasets for mRNAs (GSE1037) and miRNAs (GSE19945) were downloaded from the GEO database. Overlapping differentially expressed genes (DEGs) and differentially expressed miRNAs (DEMs) were identified through integrative analysis. The target genes of the FFL were obtained from the miRTarBase database, and the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) functional enrichment analyses were performed on the target genes. Then, we screened for key miRNAs in the FFL and performed gene regulatory network analysis based on key miRNAs. Finally, the TF-miRNA-target gene FFLs were constructed by the hypergeometric test. Results A total of 343 DEGs and 60 DEMs were identified in LCNEC tissues compared to normal tissues, including 210 down-regulated and 133 up-regulated genes, and 29 down-regulated and 31 up-regulated miRNAs. Finally, the regulatory network of TF-miRNA-target gene was established. The key regulatory network modules included ETS1-miR195-CD36, TAOK1-miR7-1-3P-GRIA1, E2F3-miR195-CD36, and TEAD1-miR30A-CTHRC1. Conclusions We constructed the TF-miRNA-target gene regulatory network, which is helpful for understanding the complex LCNEC regulatory mechanisms.
Collapse
Affiliation(s)
- Cunliang Cai
- Department of Respiratory and Critical Care Medicine, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Qianli Zeng
- The South China Center for Innovative Pharmaceuticals, Guangzhou, China
| | - Guiliang Zhou
- The South China Center for Innovative Pharmaceuticals, Guangzhou, China
| | - Xiangdong Mu
- Department of Respiratory and Critical Care Medicine, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| |
Collapse
|
20
|
Misra SK, Moitra P, Kondaiah P, Bhattacharya S. Breaking the Barrier of Polynucleotide Size, Type, and Topology in Smad2 Antisense Therapy Using a Cationic Cholesterol Dimer with Flexible Spacer. ACS APPLIED BIO MATERIALS 2020; 3:7712-7721. [DOI: 10.1021/acsabm.0c00924] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Santosh K. Misra
- Department of Organic Chemistry, Indian Institute of Science, Bangalore 560 012, India
| | - Parikshit Moitra
- Technical Research Centre, Indian Association for the Cultivation of Science, Kolkata 700032, India
- Department of Pediatrics, Center for Blood Oxygen Transport and Hemostasis, School of Medicine, Health Sciences Facility III, University of Maryland Baltimore, 670 W Baltimore St, Baltimore, Maryland 21201, United States
| | - Paturu Kondaiah
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore 560 012, India
| | - Santanu Bhattacharya
- Department of Organic Chemistry, Indian Institute of Science, Bangalore 560 012, India
- Technical Research Centre, Indian Association for the Cultivation of Science, Kolkata 700032, India
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Kolkata 700032, India
| |
Collapse
|
21
|
Liu X, Huang S, Guan Y, Zhang Q. Long noncoding RNA OSER1‑AS1 promotes the malignant properties of non‑small cell lung cancer by sponging microRNA‑433‑3p and thereby increasing Smad2 expression. Oncol Rep 2020; 44:599-610. [PMID: 32627026 PMCID: PMC7336450 DOI: 10.3892/or.2020.7645] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 05/20/2020] [Indexed: 02/07/2023] Open
Abstract
OSER1 antisense RNA 1 (OSER1-AS1), a long noncoding RNA, has been well studied in the context of hepatocellular carcinoma. However, its expression status, specific functions, and tumorigenic mechanism in non-small cell lung cancer (NSCLC) remain uninvestigated. Hence, this study aimed to assess OSER1-AS1 expression, test the malignancy-related biological functions of OSER1-AS1, and illustrate how they affect NSCLC progression. OSER1-AS1 expression in NSCLC was measured by reverse transcription-quantitative polymerase chain reaction. Cell Counting Kit-8 assay, flow cytometry, cell migration and invasion assay, and tumor xenograft assay were performed to analyze the effects of OSER1-AS1 on the malignant phenotypes of NSCLC cells. Bioinformatics prediction with luciferase reporter and RNA immunoprecipitation assays were performed to determine the interaction between OSER1-AS1 and microRNA-433-3p (miR-433-3p). OSER1-AS1 was strongly expressed in NSCLC tissues and cell lines. Enhanced OSER1-AS1 expression was significantly correlated with tumor size, TNM stage, and lymph node metastasis in patients with NSCLC. Patients with NSCLC exhibiting high OSER1-AS1 expression had shorter overall survival than those exhibiting low OSER1-AS1 expression. Functionally, a reduction in OSER1-AS1 expression led to significant decreases in NSCLC cell proliferation, migration, and invasion as well as an increase in cell apoptosis in vivo. OSER1-AS1 knockdown suppressed the tumorigenic ability of NSCLC cells in vivo. Mechanistically, OSER1-AS1 acts as a competing endogenous RNA (ceRNA) in NSCLC cells by sponging miR-433-3p and thereby increasing the expression of mothers against decapentaplegic homolog 2 (Smad2). Finally, restoration experiments revealed that the suppression of miR-433-3p and restoration of Smad2 both counteracted the suppressive effects of OSER1-AS1 depletion in NSCLC cells. Our findings illustrate the biological importance of the OSER1-AS1/miR-433-3p/Smad2 pathway in NSCLC progression and offer a novel perspective regarding the identification of effective therapeutic and diagnostic targets.
Collapse
Affiliation(s)
- Xinmei Liu
- Department of Respiratory Disease, Weifang People's Hospital, Weifang, Shandong 261041, P.R. China
| | - Shasha Huang
- Department of General Surgery, Weifang People's Hospital, Weifang, Shandong 261041, P.R. China
| | - Yun Guan
- Department of Respiratory Disease, Weifang People's Hospital, Weifang, Shandong 261041, P.R. China
| | - Qing Zhang
- Department of Respiratory Disease, Weifang People's Hospital, Weifang, Shandong 261041, P.R. China
| |
Collapse
|
22
|
Zhang Y, Shi K, Liu H, Chen W, Luo Y, Wei X, Wu Z. miR-4458 inhibits the epithelial-mesenchymal transition of hepatocellular carcinoma cells by suppressing the TGF-β signaling pathway via targeting TGFBR1. Acta Biochim Biophys Sin (Shanghai) 2020; 52:554-562. [PMID: 32324847 DOI: 10.1093/abbs/gmaa029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most lethal cancers in the world. MicroRNAs play a pivotal role in the progression of various cancers. To date, very little attention has been paid to miR-4458. Therefore, the aim of our study was to explore the function and underlying molecular mechanism of miR-4458 in HCC. We found that the expression of miR-4458 was reduced in HCC tissues and cell lines. Forced overexpression of miR-4458 inhibited the migration, invasion, and epithelial-mesenchymal transition (EMT) of HCC cells, while downregulation of miR-4458 promoted the aggressive phenotype. Furthermore, transforming growth factor beta receptor 1 (TGFBR1), the modulator of the TGF-β signaling pathway, was verified to be a novel target gene of miR-4458 by dual-luciferase reporter gene assay. Upregulated miR-4458 dramatically abolished TGFBR1 and p-Smad2/3 expression, thus blocking the TGF-β signaling pathway. Moreover, restoration of TGFBR1 partially rescued the miR-4458-mediated suppressive effect on the migration, invasion, and EMT and reactivated the TGF-β signaling pathway in HCC cells. In summary, our findings first demonstrated a mechanism of miR-4458 in HCC cell migration, invasion, and EMT by regulating the TGF-β signaling pathway via directly targeting TGFBR1.
Collapse
Affiliation(s)
- Yuke Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Kun Shi
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Hang Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Wei Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Yunhai Luo
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Xufu Wei
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Zhongjun Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| |
Collapse
|