1
|
Zheng S, Gao E, Guo L, Xie L, Zhao B, Hong Q, Li J, Hu X, Tao B. LncRNA MIAT binding to GATA3 activates MAPK signaling pathway and influences bronchopulmonary dysplasia. Int J Biol Macromol 2025; 286:138280. [PMID: 39626812 DOI: 10.1016/j.ijbiomac.2024.138280] [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: 08/01/2024] [Revised: 11/18/2024] [Accepted: 11/30/2024] [Indexed: 12/19/2024]
Abstract
Bronchopulmonary dysplasia (BPD) manifests in premature neonates with aberrant pulmonary function. Numerous long non-coding RNAs (lncRNAs) have been implicated in the pathogenesis of BPD. This study aims to elucidate the impact of the lncRNA myocardial infarction-associated transcript (MIAT) on the initiation and progression of BPD. Initially, BPD murine models were established through hyperoxia induction in newborn mice. Subsequently, MIAT and GATA binding protein 3 (GATA3) expression levels were assessed, and intravenous administration of short hairpin RNAs (shRNAs) targeting MIAT and GATA3 was performed. Pulmonary histological alterations were examined through histological staining. Levels of inflammatory mediators were quantified using enzyme-linked immunosorbent assay (ELISA) kits. The interaction between MIAT and GATA3 was scrutinized through RNA immunoprecipitation, RNA pull-down, and fluorescence in situ hybridization. The downstream mechanisms of GATA3 were explored using bioinformatics analysis. In summary, lncRNA MIAT exhibited elevated expression in the lung tissues of BPD-afflicted mice. MIAT localized to the nucleus and interacted with GATA3, thereby activating the mitogen-activated protein kinase (MAPK) pathway. Knockdown of MIAT or silencing of GATA3 attenuated the inflammatory response, deactivated the MAPK pathway, and ameliorated BPD symptoms in mice, on the other hand, p-Cresyl sulfate potassium can activate the MAPK signaling pathway and attenuates the effects of si-MIAT or si-GAT3. These improvements were characterized by enhanced alveolar differentiation and reduced glycogen and collagen deposition. In conclusion, lncRNA MIAT plays a pivotal role in activating the MAPK pathway and exacerbating hyperoxia-induced BPD in mice through the binding to GATA3. It's an important discovery for the pathogenesis of BPD and may provide some new treatment for infants diagnosed with BPD.
Collapse
Affiliation(s)
- Siqiang Zheng
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Erji Gao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Liang Guo
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Lei Xie
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Bin Zhao
- Department of Pulmonary Nodule Center, Shandong Public Health Clinical Center, Jinan, Shandong 250100, China
| | - Qi Hong
- Department of Thoracic Surgery, Shenyang Tenth People's Hospital, Shenyang Chest Hospital, Shenyang, Liaoning 110044, China
| | - Juanjuan Li
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Xuefei Hu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Bo Tao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China.
| |
Collapse
|
2
|
Long Y, Luo Y, Hu L, Liao H, Liu J. Targeting miR-146b-5p to Regulate KDM6B Expression Aggravates Bronchopulmonary Dysplasia. Mol Biotechnol 2024; 66:2078-2086. [PMID: 37584827 DOI: 10.1007/s12033-023-00849-1] [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: 03/09/2023] [Accepted: 08/02/2023] [Indexed: 08/17/2023]
Abstract
miR-146b-5p has been studied to be highly expressed in bronchopulmonary dysplasia (BPD), but whether it is involved in regulating the process of BPD in premature infants remains unclear. This study was to explore miR-146b-5p in premature BPD and reveal its molecular mechanism. BPD mouse model and high-oxygen MLE-12 cell model were established. HE staining, TUNEL staining, and IF staining were conducted to evaluate the pathological injury and protein expression in mouse lung tissue. LDH assay, MMT assay, and flow cytometry were achieved to evaluate cytotoxicity, cell viability, and apoptosis. ELISA and immunoblotting were performed to evaluate inflammatory cytokines and Wnt pathway proteins in lung tissues and cells. Dual-luciferase reporter assay and RIP assay were needed to examine the targeting relationship between miR-146b-5p and KDM6B. miR-146b-5p was abundantly expressed in BPD and KDM6B was lowly expressed. miR-146b-5p knockdown improved hyperoxia-induced lung epithelial cell inflammation and apoptosis in both models. miR-146b-6p upregulation or KDM6B downregulation aggravated hyperoxia-induced inflammation and apoptosis of lung epithelial cells. This effect of overexpressing miR-146b-5p was rescued by forcing KDM6B. MiR-146b-5p activated Wnt signaling by regulating KDM6B. miR-146b-5p activates the Wnt pathway through targeted regulation of KDM6B, thereby aggravating hyperoxia-induced inflammation and apoptosis of lung epithelial cells.
Collapse
Affiliation(s)
- YunFeng Long
- Department of Neonatology, The First Affiliated Hospital of Shaoyang University, No. 39, Tongheng Street, Shuangqing District, Shaoyang City, 422000, Hunan Province, China
| | - Yong Luo
- Department of Neonatology, The First Affiliated Hospital of Shaoyang University, No. 39, Tongheng Street, Shuangqing District, Shaoyang City, 422000, Hunan Province, China
| | - Liu Hu
- Department of Neonatology, The First Affiliated Hospital of Shaoyang University, No. 39, Tongheng Street, Shuangqing District, Shaoyang City, 422000, Hunan Province, China
| | - Hong Liao
- Department of Neonatology, The First Affiliated Hospital of Shaoyang University, No. 39, Tongheng Street, Shuangqing District, Shaoyang City, 422000, Hunan Province, China
| | - Jin Liu
- Department of Neonatology, The First Affiliated Hospital of Shaoyang University, No. 39, Tongheng Street, Shuangqing District, Shaoyang City, 422000, Hunan Province, China.
| |
Collapse
|
3
|
Maeda H, Li X, Go H, Dennery PA, Yao H. miRNA Signatures in Bronchopulmonary Dysplasia: Implications for Biomarkers, Pathogenesis, and Therapeutic Options. FRONT BIOSCI-LANDMRK 2024; 29:271. [PMID: 39082345 PMCID: PMC11799892 DOI: 10.31083/j.fbl2907271] [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: 04/02/2024] [Revised: 05/21/2024] [Accepted: 05/27/2024] [Indexed: 01/18/2025]
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic lung disease in premature infants characterized by alveolar dysplasia, vascular simplification and dysmorphic vascular development. Supplemental oxygen and mechanical ventilation commonly used as life-saving measures in premature infants may cause BPD. microRNAs (miRNAs), a class of small, non-coding RNAs, regulate target gene expression mainly through post-transcriptional repression. miRNAs play important roles in modulating oxidative stress, proliferation, apoptosis, senescence, inflammatory responses, and angiogenesis. These cellular processes play pivotal roles in the pathogenesis of BPD. Accumulating evidence demonstrates that miRNAs are dysregulated in the lung of premature infants with BPD, and in animal models of this disease, suggesting contributing roles of dysregulated miRNAs in the development of BPD. Therefore, miRNAs are considered promising biomarker candidates and therapeutic agents for this disease. In this review, we discuss how dysregulated miRNAs and their modulation alter cellular processes involved in BPD. We then focus on therapeutic approaches targeting miRNAs for BPD. This review provides an overview of miRNAs as biomarkers, and highlights potential pathogenic roles, and therapeutic strategies for BPD using miRNAs.
Collapse
Affiliation(s)
- Hajime Maeda
- Department of Molecular Biology, Cellular Biology, and Biochemistry, Brown University, Providence, RI 02912, USA
- Department of Pediatrics, Fukushima Medical University School of Medicine, 960-1295 Fukushima, Japan
| | - Xiaoyun Li
- Department of Molecular Biology, Cellular Biology, and Biochemistry, Brown University, Providence, RI 02912, USA
- Providence Veterans Affairs Medical Center, Providence, RI 02908, USA
- Department of Medicine, Warren Alpert School of Medicine of Brown University, Providence, RI 02903, USA
- College of Pharmacy, Jinan University, 510632 Guangzhou, Guangdong, China
| | - Hayato Go
- Department of Pediatrics, Fukushima Medical University School of Medicine, 960-1295 Fukushima, Japan
| | - Phyllis A. Dennery
- Department of Molecular Biology, Cellular Biology, and Biochemistry, Brown University, Providence, RI 02912, USA
- Department of Pediatrics, Warren Alpert School of Medicine of Brown University, Providence, RI 02903, USA
| | - Hongwei Yao
- Department of Molecular Biology, Cellular Biology, and Biochemistry, Brown University, Providence, RI 02912, USA
- Providence Veterans Affairs Medical Center, Providence, RI 02908, USA
- Department of Medicine, Warren Alpert School of Medicine of Brown University, Providence, RI 02903, USA
| |
Collapse
|
4
|
Dini G, Ceccarelli S, Celi F. Strategies for the prevention of bronchopulmonary dysplasia. Front Pediatr 2024; 12:1439265. [PMID: 39114855 PMCID: PMC11303306 DOI: 10.3389/fped.2024.1439265] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 07/16/2024] [Indexed: 08/10/2024] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a common morbidity affecting preterm infants and is associated with substantial long-term disabilities. The pathogenesis of BPD is multifactorial, and the clinical phenotype is variable. Extensive research has improved the current understanding of the factors contributing to BPD pathogenesis. However, effectively preventing and managing BPD remains a challenge. This review aims to provide an overview of the current evidence regarding the prevention of BPD in preterm infants, offering practical insights for clinicians.
Collapse
Affiliation(s)
- Gianluca Dini
- Neonatal Intensive Care Unit, Santa Maria Hospital, Terni, Italy
| | | | | |
Collapse
|
5
|
Yang M, Chen Y, Huang X, Shen F, Meng Y. ETS1 Ameliorates Hyperoxia-Induced Bronchopulmonary Dysplasia in Mice by Activating Nrf2/HO-1 Mediated Ferroptosis. Lung 2023; 201:425-441. [PMID: 37490064 PMCID: PMC10444662 DOI: 10.1007/s00408-023-00639-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 07/13/2023] [Indexed: 07/26/2023]
Abstract
PURPOSE Bronchopulmonary dysplasia (BPD) is associated with hyperoxia-induced oxidative stress-associated ferroptosis. This study examined the effect of E26 oncogene homolog 1 (ETS1) on oxidative stress-associated ferroptosis in BPD. METHODS Hyperoxia-induced A549 cells and neonatal mice were used to establish BPD models. The effects of ETS1 on hyperoxia-induced ferroptosis-like changes in A549 cells were investigated by overexpression of ETS1 plasmid transfection and erastin treatment. Glucose consumption, lactate production, and NADPH levels were assessed by the glucose, lactate, and NADP+/NADPH assay kits, respectively. The potential regulatory relationship between ETS1 and Nrf2/HO-1 was examined by treating hyperoxia-induced A549 cells with the Nrf2 inhibitor ML385. ETS1 effect on the Nrf2 promoter was explored by dual-luciferase reporter and chromatin immunoprecipitation assay. The effect of ETS1 on the symptoms of BPD mice was examined by injecting an adenovirus overexpressing ETS1. RESULTS ETS1 overexpression increased hyperoxia-induced cell viability, glucose consumption, lactate production, and NADPH levels and reduced inflammation and apoptosis in A549 cells. In animal experiments, ETS1 overexpression prevented weight loss, airway enlargement, and reductions in radial alveolar counts in BPD mice, while reducing the mean linear intercept, mean alveolar diameter and inflammation. ETS1 overexpression suppressed PTGS2 and CHAC1 expression, reduced ROS, MDA and ferrous iron (Fe2+) production and increased GSH levels in hyperoxia-induced A549 cells and BPD mice. In addition, ETS1 can bind to the Nrf2 promoter region and thus promote Nrf2 transcription. ETS1 overexpression increased the mRNA and protein levels of Nrf2, HO-1, xCT, and GPX4 in hyperoxia-induced A549 cells and BPD mice. In hyperoxia-induced A549 cells, erastin and ML385 treatment abolished the effect of ETS1 overexpression. CONCLUSION ETS1 is important in oxidative stress-related ferroptosis in a hyperoxia-induced BPD model, and the effect is partially mediated by the Nrf2/HO-1 axis.
Collapse
Affiliation(s)
- Min Yang
- Respiratory Department, Hunan Children's Hospital, Changsha, 410007, China.
| | - Yanping Chen
- Respiratory Department, Hunan Children's Hospital, Changsha, 410007, China
| | | | - Fang Shen
- Research Institute of Children, Hunan Children's Hospital, Changsha, 410007, China
| | - Yanni Meng
- Respiratory Department, Hunan Children's Hospital, Changsha, 410007, China
| |
Collapse
|
6
|
Tong Y, Zhang S, Riddle S, Song R, Yue D. Circular RNAs in the Origin of Developmental Lung Disease: Promising Diagnostic and Therapeutic Biomarkers. Biomolecules 2023; 13:biom13030533. [PMID: 36979468 PMCID: PMC10046088 DOI: 10.3390/biom13030533] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/11/2023] [Accepted: 03/12/2023] [Indexed: 03/17/2023] Open
Abstract
Circular RNA (circRNA) is a newly discovered noncoding RNA that regulates gene transcription, binds to RNA-related proteins, and encodes protein microRNAs (miRNAs). The development of molecular biomarkers such as circRNAs holds great promise in the diagnosis and prognosis of clinical disorders. Importantly, circRNA-mediated maternal-fetus risk factors including environmental (high altitude), maternal (preeclampsia, smoking, and chorioamnionitis), placental, and fetal (preterm birth and low birth weight) factors are the early origins and likely to contribute to the occurrence and progression of developmental and pediatric cardiopulmonary disorders. Although studies of circRNAs in normal cardiopulmonary development and developmental diseases have just begun, some studies have revealed their expression patterns. Here, we provide an overview of circRNAs’ biogenesis and biological functions. Furthermore, this review aims to emphasize the importance of circRNAs in maternal-fetus risk factors. Likewise, the potential biomarker and therapeutic target of circRNAs in developmental and pediatric lung diseases are explored.
Collapse
Affiliation(s)
- Yajie Tong
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Shuqing Zhang
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Suzette Riddle
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Rui Song
- Lawrence D. Longo MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
- Correspondence: (R.S.); (D.Y.); Tel.: +01-909-558-4325 (R.S.); +86-24-9661551125 (D.Y.)
| | - Dongmei Yue
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
- Correspondence: (R.S.); (D.Y.); Tel.: +01-909-558-4325 (R.S.); +86-24-9661551125 (D.Y.)
| |
Collapse
|
7
|
Alva R, Mirza M, Baiton A, Lazuran L, Samokysh L, Bobinski A, Cowan C, Jaimon A, Obioru D, Al Makhoul T, Stuart JA. Oxygen toxicity: cellular mechanisms in normobaric hyperoxia. Cell Biol Toxicol 2022; 39:111-143. [PMID: 36112262 PMCID: PMC9483325 DOI: 10.1007/s10565-022-09773-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 09/07/2022] [Indexed: 12/15/2022]
Abstract
In clinical settings, oxygen therapy is administered to preterm neonates and to adults with acute and chronic conditions such as COVID-19, pulmonary fibrosis, sepsis, cardiac arrest, carbon monoxide poisoning, and acute heart failure. In non-clinical settings, divers and astronauts may also receive supplemental oxygen. In addition, under current standard cell culture practices, cells are maintained in atmospheric oxygen, which is several times higher than what most cells experience in vivo. In all the above scenarios, the elevated oxygen levels (hyperoxia) can lead to increased production of reactive oxygen species from mitochondria, NADPH oxidases, and other sources. This can cause cell dysfunction or death. Acute hyperoxia injury impairs various cellular functions, manifesting ultimately as physiological deficits. Chronic hyperoxia, particularly in the neonate, can disrupt development, leading to permanent deficiencies. In this review, we discuss the cellular activities and pathways affected by hyperoxia, as well as strategies that have been developed to ameliorate injury.
Collapse
Affiliation(s)
- Ricardo Alva
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Maha Mirza
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Adam Baiton
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Lucas Lazuran
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Lyuda Samokysh
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Ava Bobinski
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Cale Cowan
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Alvin Jaimon
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Dede Obioru
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Tala Al Makhoul
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Jeffrey A Stuart
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada.
| |
Collapse
|
8
|
LncRNA Rian reduces cardiomyocyte pyroptosis and alleviates myocardial ischemia-reperfusion injury by regulating by the miR-17-5p/CCND1 axis. Hypertens Res 2022; 45:976-989. [PMID: 35264782 DOI: 10.1038/s41440-022-00884-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 12/20/2021] [Accepted: 01/09/2022] [Indexed: 02/07/2023]
Abstract
Myocardial ischemia-reperfusion injury (MIRI) is a pathological process characterized by cardiomyocyte death. Long noncoding RNAs (lncRNAs) have been shown to be dysregulated in the course of MIRI. Accordingly, the current study investigated the mechanism of lncRNA Rian in MIRI-induced cardiomyocyte pyroptosis. First, a murine model of MIRI was established by using the left anterior descending (LAD) coronary artery ligation method. Cardiac function and myocardial histopathological changes were evaluated by echocardiography and hematoxylin and eosin staining. Then, a cell model of MIRI was established by oxygen-glucose deprivation/reoxygenation (OGD/R), followed by analysis of NLRP3, cleaved caspase-1, and GSDMD-N levels by western blotting. The levels of IL-1β, IL-18, TNF-α, and IL-10 were measured using ELISA. LncRNA Rian, miR-17-5p, and CCND1 expression in myocardial tissues and OGD/R cells were examined using RT-qPCR. Finally, the binding relationships between Rian and miR-17-5p and miR-17-5p and CCND1 were validated with the help of dual-luciferase and RNA pull-down assays. Rian was poorly expressed in MIRI mice and OGD/R cells. LncRNA Rian overexpression reduced cardiomyocyte pyroptosis in vivo and in vitro, as indicated by decreased NLRP3, cleaved caspase-1, GSDMD-N, IL-1β, IL-18, and TNF-α levels and increased IL-10 levels. Furthermore, Rian bound to miR-17-5p and promoted CCND1 transcription. Notably, miR-17-5p overexpression or CCND1 silencing reversed the inhibitory effect of Rian overexpression on cardiomyocyte pyroptosis. Collectively, our findings indicate that Rian overexpression reduces cardiomyocyte pyroptosis and alleviates MIRI through the miR-17-5p/CCND1 axis.
Collapse
|
9
|
Sakaria RP, Dhanireddy R. Pharmacotherapy in Bronchopulmonary Dysplasia: What Is the Evidence? Front Pediatr 2022; 10:820259. [PMID: 35356441 PMCID: PMC8959440 DOI: 10.3389/fped.2022.820259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 01/26/2022] [Indexed: 11/13/2022] Open
Abstract
Bronchopulmonary Dysplasia (BPD) is a multifactorial disease affecting over 35% of extremely preterm infants born each year. Despite the advances made in understanding the pathogenesis of this disease over the last five decades, BPD remains one of the major causes of morbidity and mortality in this population, and the incidence of the disease increases with decreasing gestational age. As inflammation is one of the key drivers in the pathogenesis, it has been targeted by majority of pharmacological and non-pharmacological methods to prevent BPD. Most extremely premature infants receive a myriad of medications during their stay in the neonatal intensive care unit in an effort to prevent or manage BPD, with corticosteroids, caffeine, and diuretics being the most commonly used medications. However, there is no consensus regarding their use and benefits in this population. This review summarizes the available literature regarding these medications and aims to provide neonatologists and neonatal providers with evidence-based recommendations.
Collapse
Affiliation(s)
- Rishika P. Sakaria
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Ramasubbareddy Dhanireddy
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, United States
- Department of Obstetrics and Gynecology, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
10
|
Ashrafizadeh M, Zarrabi A, Mostafavi E, Aref AR, Sethi G, Wang L, Tergaonkar V. Non-coding RNA-based regulation of inflammation. Semin Immunol 2022; 59:101606. [PMID: 35691882 DOI: 10.1016/j.smim.2022.101606] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 05/01/2022] [Accepted: 05/25/2022] [Indexed: 01/15/2023]
Abstract
Inflammation is a multifactorial process and various biological mechanisms and pathways participate in its development. The presence of inflammation is involved in pathogenesis of different diseases such as diabetes mellitus, cardiovascular diseases and even, cancer. Non-coding RNAs (ncRNAs) comprise large part of transcribed genome and their critical function in physiological and pathological conditions has been confirmed. The present review focuses on miRNAs, lncRNAs and circRNAs as ncRNAs and their potential functions in inflammation regulation and resolution. Pro-inflammatory and anti-inflammatory factors are regulated by miRNAs via binding to 3'-UTR or indirectly via affecting other pathways such as SIRT1 and NF-κB. LncRNAs display a similar function and they can also affect miRNAs via sponging in regulating levels of cytokines. CircRNAs mainly affect miRNAs and reduce their expression in regulating cytokine levels. Notably, exosomal ncRNAs have shown capacity in inflammation resolution. In addition to pre-clinical studies, clinical trials have examined role of ncRNAs in inflammation-mediated disease pathogenesis and cytokine regulation. The therapeutic targeting of ncRNAs using drugs and nucleic acids have been analyzed to reduce inflammation in disease therapy. Therefore, ncRNAs can serve as diagnostic, prognostic and therapeutic targets in inflammation-related diseases in pre-clinical and clinical backgrounds.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, 34956 Istanbul, Turkey
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, 34396 Istanbul, Turkey.
| | - Ebrahim Mostafavi
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Translational Sciences, Xsphera Biosciences Inc. 6, Tide Street, Boston, MA 02210, USA
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore.
| | - Lingzhi Wang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Vinay Tergaonkar
- Laboratory of NF-κB Signaling, Institute of Molecular and Cell Biology (IMCB), Singapore, Singapore; Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|